Sei sulla pagina 1di 17

Drug Development and Industrial Pharmacy

ISSN: 0363-9045 (Print) 1520-5762 (Online) Journal homepage: https://www.tandfonline.com/loi/iddi20

Review and analysis of FDA approved drugs using


lipid-based formulations

Ronak Savla, Jeff Browne, Vincent Plassat, Kishor M. Wasan & Ellen K. Wasan

To cite this article: Ronak Savla, Jeff Browne, Vincent Plassat, Kishor M. Wasan & Ellen K.
Wasan (2017) Review and analysis of FDA approved drugs using lipid-based formulations, Drug
Development and Industrial Pharmacy, 43:11, 1743-1758, DOI: 10.1080/03639045.2017.1342654

To link to this article: https://doi.org/10.1080/03639045.2017.1342654

© 2017 The Author(s). Published by Informa


UK Limited, trading as Taylor & Francis
Group

View supplementary material

Accepted author version posted online: 04


Jul 2017.
Published online: 06 Jul 2017.

Submit your article to this journal

Article views: 11116

View Crossmark data

Citing articles: 18 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iddi20
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY, 2017
VOL. 43, NO. 11, 1743–1758
https://doi.org/10.1080/03639045.2017.1342654

REVIEW ARTICLE

Review and analysis of FDA approved drugs using lipid-based formulations


Ronak Savlaa , Jeff Browneb, Vincent Plassatc, Kishor M. Wasand and Ellen K. Wasand
a
Catalent Pharma Solutions, Somerset, NJ, USA; bCatalent Pharma Solutions, St. Petersburg, FL, USA; cCatalent Pharma Solutions, Beinheim,
France; dCollege of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, Canada

ABSTRACT ARTICLE HISTORY


Lipid-based drug delivery systems (LBDDS) are one of the most studied bioavailability enhancement tech- Received 27 March 2017
nologies and are utilized in a number of U.S. Food and Drug Administration (FDA) approved drugs. While Revised 12 May 2017
researchers have used several general rules of thumb to predict which compounds are likely to benefit Accepted 6 June 2017
from LBDDS, formulation of lipid systems is primarily an empiric endeavor. One of the challenges is that
KEYWORDS
these rules of thumb focus in different areas and are used independently of each other. The Lipid-based drug delivery
Developability Classification System attempts to link physicochemical characteristics with possible formula- systems; developability
tion strategies. Although it provides a starting point, the formulator still has to empirically develop the for- classification system;
mulation. This article provides a review and quantitative analysis of the molecular properties of these physicochemical properties;
approved drugs formulated as lipid systems and starts to build an approach that provides more directed biopharmaceutics; clinical
guidance on which type of lipid system is likely to be the best for a particular drug molecule. translation

Introduction There are numerous ‘Rules’ and ‘Classification Systems’ avail-


able to formulation scientists for prediction of in vivo perform-
An increasing percentage of drug molecules in pharma develop-
ance. Formulators are working to create in silico tools and using
ment pipelines can be classified according to the
classification systems coupled with in vitro dissolution tests in
Biopharmaceutics Classification System (BCS) as Class II com-
biorelevant media for predicting in vivo outcomes during the
pounds (compounds having good permeability but poor solubil-
formulation design and screening phases of product develop-
ity). Additionally, a considerable percentage of today’s pipeline
ment. While predicting the self-emulsification behavior of LBDDS
molecules are both poorly soluble and poorly permeable (BCS
excipient combinations is reasonable, formulators lack the in
Class IV). These newer drug molecules are discovered and opti-
silico tools to accurately predict how specific drugs will behave
mized through relatively novel technologies such as high-through-
in specific formulations [3]. The aim of this study was to ana-
put screening, fragment-based drug discovery, or computational
lyze the physicochemical characteristics of these drugs and clas-
modeling. While, they demonstrate superior potencies and specif-
sify them according to the different systems to see what, if any,
icities compared to their predecessors, many also possess inherent
correlations exist between physicochemical properties and classi-
challenges in terms of oral delivery [1]. The low solubility and
fication systems or between classification systems. Furthermore,
poor permeability of these compounds translates into suboptimal
this exercise could validate the belief that simple physicochemi-
patient outcomes due to poor oral bioavailability and variable
cal properties are insufficient for de novo LBDDS formulation
pharmacokinetics.
design and highlight key areas for the focus of future studies.
Simple formulation approaches (conventional tablets or pow-
We first examined U.S. Food and Drug Administration (FDA)
der in capsule) are not enough to address these issues.
approved drugs (based on NDA approvals) that have been for-
Bioavailability enhancement technologies (LBDDS, solid amorph-
mulated using LBDDS. We focused only on approved drugs with
ous dispersions, API salt formation, or API particle size reduction)
the rationale that they were robust enough to progress through
have been developed to address the issue of low solubility by
all phases of drug development and receive FDA approval. Next,
improving the dissolution rate and/or the apparent solubility of we classified the drugs using the Lipid Formulation Classification
these drug molecules. A molecule’s dissolution rate can be System (LFCS). We calculated physicochemical properties that
enhanced by increasing the surface area (particle size reduction) may influence the choice of LBDDS used for formulation and
or by stabilizing the amorphous form of the drug in a polymer used these properties to group the drugs into the BCS and
(hot melt extrusion and spray drying). Lipid excipients and sur- Developability Classification System (DCS). Based on the these
factants can be used to present and maintain the drug molecule calculations and classifications, we sought any correlations that
in solution form both pre- and post-administration. Of the bio- would provide guidance as to what types of drug molecules are
availability enhancing technologies, LBDDS are the most well good fit for LBDDS and what specific type of LBDDS. We con-
studied and well established as a formulation approach for clude by proposing therapeutic categories, compound categories,
addressing solubility and/or permeability issues [2]. Therefore, we and other untapped areas that can benefit from the advantages
focus our efforts in this review on LBDDS. offered through the use of LBDDS.

CONTACT Ronak Savla ronak.savla@catalent.com Scientific Affairs Manager, Catalent Pharma Solutions, 14 Schoolhouse Road, Somerset, NJ 08873, USA
Supplemental data for this article can be accessed here.
ß 2017 The Author(s). Published by Informa UK Limited, trading as Taylor & Francis Group
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License (http://creativecommons.org/licenses/by-nc-nd/4.0/),
which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.
1744 R. SAVLA ET AL.

FDA approved orally administered products that use considerations have clear and direct implications on drug absorp-
lipid formulations tion in vivo. The formulator needs to possess knowledge about
physical chemistry, thermodynamics, and gastrointestinal physi-
The increasingly overall water-insoluble nature of drug candidates
ology to ensure that drug precipitation does not occur in the
has been accompanied by the increased use of solubilization tech-
gastrointestinal tract following dispersion and/or digestion of the
nologies for FDA approved drugs. A recent analysis revealed that
formulation. While most LBDSS are designed to solubilize the drug
6% of all approved new molecular entities from 1975 to 2013 uti-
in the formulation prior to administration, endogenously, the drug
lized solubilization technologies [2]. The same analysis showed that
in the formulation matrix experiences environmental transition
LBDDS are the most widely used solubilization technology. An ana-
from a solution state in formulation, to an emulsified state due to
lysis by Strickley in 2007 estimated that LBDDS oral drug products
dispersion upon initial contact with GI fluid, and finally a mixed
account for 2–4% of all commercially available drug products [4].
micellar phase after lipid digestion.
A review of the literature and drug databases (DailyMed,
LBDDS is an umbrella term for numerous delivery systems [oil
DrugBank, and Drugs@FDA) revealed 27 unique drug molecules
solutions, emulsions, dispersions, micelles, self-emulsifying drug
that were FDA approved as 36 different oral LBDDS. (Table 1)
[5–7]. A few of the approved drugs have been discontinued. delivery systems (SEDDS), and self-microemulsifying drug delivery
According to the Federal Register, none have been for safety or systems (SMEDDS)] and can be broadly classified into four types
efficacy reasons. The three HIV protease inhibitor formulations according to the LFCS proposed by Pouton (Table 2) [11,12]. In
were replaced by newer formulations. Saquanavir (FortovaseV) was
R our review, there were 16 drugs that utilized Type I LBDDS, 3
discontinued because a film-coated tablet dosage form with a drugs formulated as Type II, 5 drugs as Type III, and 2 drugs as
lower pill burden was introduced. Ritonavir/lopinavir (KaletraV)
R TYPE IV (Table 1). Type I lipid formulations contain oils that need
was replaced by a solid dispersion formulation, which does not to be digested in order to form mixed micelles. Type I formula-
require refrigeration, had higher drug loading per dosage unit, tions are typically biocompatible and simple (thereby explaining
and reduced daily pill burden. Amprenavir (AgeneraseV) was suc-
R their early use), and often the LFCS formulation type first
ceeded by a pro-drug, fosamprenavir. attempted if the dose of API can be fully solubilized in the fill. The
Analysis of these approved products can offer insights to those vitamin D analogs and retinoids are formulated as Type I LBDDS
physicochemical characteristics of a drug molecule that make LFCS Type II lipid formulations consist of mixtures of lipids and
LBDDS an attractive formulation option. As seen in Table 1 and water- insoluble surfactants (HLB <12). They self-emulsify into
Figure 1, there is a wide diversity, in terms of pharmacological crude oil-in-water (o/w) emulsions usually with energy input. LFCS
class and chemical structures, of approved drugs that have been Type III lipid formulations contain water- soluble surfactants (HLB
formulated using lipid systems. In short, LBDDS are a versatile plat- >12) and co-solvents. These formulations spontaneously self-emul-
form which can benefit a wide range of drug molecules. One rea- sify upon contact with an aqueous environment. They are com-
son for the versatility is the range of excipients used in LBDDS. monly referred to as self-emulsifying drug delivery systems
The major categories of LBDDS excipients are lipids, water insol- (SEDDS) when dispersion droplet size is >200 nm or as self-micro
uble surfactants (HLB <12), water soluble surfactants (HLB >12), emulsifying drug delivery systems (SMEDDS) when dispersion
and hydrophilic cosolvents. The choice and relative percentage of droplet size is <200 nm. Finally, LFCS Type IV lipid formulations do
excipients in LBDDS influence drug solubility, LBDDS dispersability, not contain oil and are based on water-soluble surfactants and co-
and formulation properties. This in part explains its historic and solvents. The fine dispersions formed when in contact with an
current popularity for formulating poorly soluble and/or perme- aqueous environment translates to rapid drug release and absorp-
able drugs. tion. The solvent capacity of these systems for maintaining a
The interest and growth in lipid based systems can be traced poorly soluble compound is usually limited which often results in
to several findings and developments. Unless they are in solution the precipitation of the compound in the GI tract.
following oral administration, poorly soluble drugs have poor
absorption, variable pharmacokinetics, and tend to experience sig-
nificant positive food effect. Lipid formulations partially improve Excipient use in LBDDS
the solubility of drugs by stimulating the same physiologic events, LBDDS include a wide range of excipients. Some considerations
although to a lower magnitude, as administration of drugs with a that a formulator must consider include: regulatory acceptance,
fatty meal [8]. safety, solvent capacity, dispersion characteristics (usually in biore-
The soft capsule has become the most common dosage form levent media), digestability, interactions with efflux and metabol-
for the administration of lipid formulations (Table 1). Soft capsules ism processes (P-gp and CYP), stability, purity and amount/type of
are the utilized dosage form for 25 of 36 lipid formulations, 6 impurities, compatibility with the capsule, and cost. Excipient
products are hard shell capsules, and 5 are oral solutions, The safety is a major consideration since the vast majority of FDA
advantages of soft capsules have been extensively documented approved drugs using a LBDDS are intended for chronic use.
elsewhere [9,10]. The introduction of NeoralV in 1995 is a widely
R

cited example illustrating how a self-emulsifying system (the first


IIIA LFCS product) can address clinical concerns and result in a Lipids
product providing a better treatment for patients. There were sev- By definition, LBDDS typically contain drug dissolved or suspended
eral approved LBDDS on the market prior to 1995 (NDA Approval in oils (triglycerides or mixed glycerides). Of the 36 FDA approved
Date of Neoral) and many more that followed (Table 1). Early lipid formulations, 8 formulations did not contain oils and were classi-
formulations consisted of pure oils (LFCS Class I and some Class II). fied as Classes III or IV according to the LFCS. Early lipid formu-
lated drugs contain almost exclusively oils. Ritonavir (NDA
approval: 1996) was the first LBDDS formulated drug that did not
Designing lipid formulations
contain oil (Table 1). The biggest advantage of using oil is that
Unlike other formulation approaches, the absorption process of lipophilic drugs are solubilized and dissolution of the drug within
LBDDS is complex and dynamic. The formulation design the GI tract is not required. Once digested, there are higher odds
Table 1. FDA approved drugs utilizing lipid systems.
Biopharmace- Lipid formula-
New drug utics classifica- Developability tion classifica- Oils: triglycerides or Water-insoluble Water-soluble
Molecule/trade application tion system classiciation tion system Drug Dosage form, mixed mono and surfactants surfactants (HLB Hydrophilic
name (NDA) Year (BCS) system (LFCS) category Dose strength diglycerides (HLB <12) >12) cosolvents
Ergocalciferol 1941 3 IIb I Vitamin D Vitamin D Resistant LFHS, 50,000 IU Soybean oil
V R
(Drisdol ) analog Rickets: 12,000 to
500,000 IU units daily.
Hypoparathyroidism:
50,000 to 200,000 IU
units daily concomi-
tantly with calcium lac-
tate 4 g, six times per
day
Calcitriol 1978 2/4 I I Vitamin D 0.25–0.5 lg qd Soft gelatin cap- Fractionated triglycer-
V R
(Rocaltrol ) analog sule, 0.25 and ides of coconut oil
0.5 lg
Valproic acid 1978 1 I I Anti-epileptic 10–15 mg/kg/day Soft gelatin cap- Corn oil
RV
(Depakene ) sule, 250 mg
Isotretinoin 1982 2 IIb I Retinoid 0.5–2 mg/kg in 2 divided Soft gelatin cap- Beeswax, hydrogen-
V R
(Accutane ) doses sule, 10, 20, ated soybean oil
Discontinued 40 mg flakes, hydrogen-
ated vegetable oil,
soybean oil
Cyclosporin A 1983 2 IIb II Calcineurin 2–10 mg/kg/day in two Oral solution, Olive oil polyoxyethylated Ethanol 12.5%
V R
(Sandimmune ) inhibitor divided doses 100 mg/mL oleic
glycerides
Dronabinol 1985 2/4 IIb I Cannabinoid 2.5–20 mg/day Soft gelatin cap- Sesame oil
V R
(Marinol ) sule, 2.5, 5,
10 mg
Clofazimine 1986 2 I Antileprosy 100 mg/day Soft gelatin cap- Beeswax
V R
(Lamprene ) sule, 50 and
100 mg 100 mg
Discontinued
Cyclosporin A 1990 2 IIb II Calcineurin 2–10 mg/kg/day in 2 div- Soft gelatin cap- Corn oil Linoleic Ethanol 12.7%
V R
(Sandimmune ) inhibitor ided doses sule, 25 and macroglycerides
100 mg
Ranitidine 1994 3 H2-receptor 150 mg bid or 300 mg qd Soft gelatin cap- Medium chain Mixed glycerides of
R
V
(Zantac ) antagonist sule, 150 and triglycerides long chain fatty
Discontinued 300 mg acids (Gelucire
33/01)
Cyclosporin A 1995 2 IIb IIIA/IIIB Calcineurin 2–10 mg/kg/day in two Soft gelatin cap- Corn oil mono-di- Polyoxyl 40 Ethanol 11.9%,
VR
(Neoral ) inhibitor divided doses sule, 10, 25, 60, triglycerides hydrogenated glycerol,
100 mg castor oil propylene
glycol
Cyclosporin A 1995 2 IIb IIIA/IIIB Calcineurin 2–10 mg/kg/day in 2 div- Oral solution, Corn oil-mono-di- Polyoxyl 40 Ethanol 11.9%,
VR
(Neoral ) inhibitor ided doses 100 mg/mL triglycerides hydrogenated propylene
castor oil glycol
Tretinoin 1995 IIb I Retinoid 45 mg/m2/day as two Soft gelatin cap- Beeswax, hydrogen-
V R
(Vesanoid ) evenly divided doses sule, 10 mg ated soybean oil
Discontinued flakes, hydrogen-
ated vegetable oil,
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY

soybean oil
R
V
Ritonavir (Norvir ) 1996 4 IIb IIIA Protease 600 mg bid (adults) Soft gelatin cap- Oleic acid Polyoxyl 35 castor Ethanol
inhibitor sule, 100 mg oil
(continued)
1745
Table 1. Continued
1746

Biopharmace- Lipid formula-


New drug utics classifica- Developability tion classifica- Oils: triglycerides or Water-insoluble Water-soluble
Molecule/trade application tion system classiciation tion system Drug Dosage form, mixed mono and surfactants surfactants (HLB Hydrophilic
name (NDA) Year (BCS) system (LFCS) category Dose strength diglycerides (HLB <12) >12) cosolvents
Saquinavir 1997 4 IIb Protease 1200 mg bid without Soft gelatin cap- Medium chain mono-
V R
(Fortovase ) inhibitor ritonavir; 1000 mg bid sule, 200mg and di-glycerides
Discontinued with ritonavir
Progesterone 1998 2 IIb I (susp) Hormone 200 mg qd Soft gelatin cap- Peanut oil
R
R. SAVLA ET AL.

V
(Prometrium ) sule, 100 and
200 mg
Amprenavir 1999 2 IIb IV Protease 1200 mg bid or 1200 mg Soft gelatin cap- Vitamin E TPGS PEG400, propyl-
V R
(Agenerase ) inhibitor qd with ritoanvir sule, 50 mg ene glycol
discontinued
Bexarotene 1999 IIb IV Retinoid 300-750 mg qd Soft gelatin cap- Polysorbate 20 PEG400
V R
(Targretin ) sule, 75 mg

Doxercalciferol 1999 2/4 I I Vitamin D Determined by target Soft gelatin cap- Coconut oil Alcohol
V R
(Hectorol ) analog intact parathyroid hor- sule, 0.5, 1,
mone level (iPTH) 2.5 mcg
Usually 10 mcg three times
weekly at dialysis
Sirolimus 1999 IIa III mTOR kinase 6 mg loading dose fol- Oral solution, Phosphatidylcholine, Polysorbate 80 1.5–2.5% etha-
V R
(Rapamune ) inhibitor lowed by 2 mg qd 1 mg/mL mono- and di-glyc- nol, propyl-
erides, soy fatty ene glycol
acids, ascorbyl
palmitate
Cyclosporin A 2000 2 IIb IV Calcineurin 1:1 dose conversion from LFHS, 25, 50, Polysorbate 80, Propylene gly-
V R
(Gengraf ) inhibitor Sandimmune 100 mg Polyoxyl 35 col, alcohol
2.5 mg/kg/day in two div- castor oil 12.8% v/v
ided dose (RA and
Psoriasis)
Cyclosporin A 2000 2 IIb IV Calcineurin 1:1 dose conversion from Oral solution, Polyoxyl 40 Propylene
V R
(Gengraf ) inhibitor Sandimmune 100mg/ml hydrogenated glycol
2.5 mg/kg/day in two div- castor oil,
ided dose (RA and Polysorbate 80
Psoriasis)
Ritonavir/lopinavir 2000 4 III Protease 400/100 mg (3 Soft gelatin Soft gelatin cap- Oleic acid Polyoxyl 35 castor Propylene
R
V
(Kaletra ) inhibitors capsule) bid sule, 133.3 mg oil glycol
Discontinued 800/200 mg (6 Soft gelatin lopinavir/
capsule) qd 33.3 mg
ritonavir
Dutasteride 2001 2/4 IIa I 5 alpha-reduc- 0.5 mg qd Soft gelatin cap- Mono-di-glycerides of
V R
(Avodart ) tase sule, 0.5 mg caprylic/capric acid
inhibitor
Isotretinoin 2003 (ANDA) 2 IIb Retinoid 0.5–1 mg/kg/day given in LFHS, 20, 30, Hydrogenated vege- Polysorbate 80
V R
(Claravis ) two divided doses 40 mg table oil, soybean
oil, white wax
Omega-3-acid ethyl 2004 I Fish oils for 4 g qd or 2 g bid Soft gelatin cap- Soybean oil
V R
esters (Lovaza ) lipid sule, 1 g
regulation
Tipranavir 2005 4 IIb IIIA Protease 500 mg with 200 mg Soft gelatin cap- Mono-/di-glycerides of Polyoxyl 35 castor Ethanol, propyl-
V R
(Aptivus ) inhibitor ritonavir qd (adults) sule, 250 mg caprylic/capric acids oil ene glycol
14 mg/kg with 6mg/kg
ritonavir (pediatric)
(continued)
Table 1. Continued
Biopharmace- Lipid formula-
New drug utics classifica- Developability tion classifica- Oils: triglycerides or Water-insoluble Water-soluble
Molecule/trade application tion system classiciation tion system Drug Dosage form, mixed mono and surfactants surfactants (HLB Hydrophilic
name (NDA) Year (BCS) system (LFCS) category Dose strength diglycerides (HLB <12) >12) cosolvents
Tipranavir 2005 4 IIb IV Protease 500 mg with 200 mg Oral solution, Vitamin E TPGS PEG 400,
V R
(Aptivus ) inhibitor ritonavir qd (adults) 100 mg/ml propylene
14 mg/kg with 6mg/kg glycol,
ritonavir (pediatric) water
Paricalcitol 2005 4 IIa I Vitamin D 1 or 2 mcg daily Soft gelatin cap- Medium chain trigly- Alcohol
V R
(Zemplar ) analog 2 or 4 mcg three times a sule, 1 and cerides fractionated
week 2 mcg from coconut oil or
palm kernel oil
Lubiprostone 2006 2/4 I I Chloride chan- 24 mcg bid for chronic Soft gelatin cap- Medium chain
V R
(Amitiza ) nel activator idiopathic constipation sule, 8 and triglycerides
and opioid-induced 24 mcg
constipation
8 mcg bid for irritable
bowel syndrome with
constipation
Fenofibrate 2006 2 IIb III Peroxisome 50 to 150 mg daily HSC, 50 and Gelucire 44/14
V R
(Lipofen ) proliferator 150 mg (lauroyl mac-
activated rogol glyceride
receptor a type 1500)
(PPARa)
acvtivator
Topotecan HCl 2007 1 I I Topoisomerase 2.3 mg/m2/day orally once LFHS, 0.25 and Hydrogenated vege- Glyceryl
V R
(Hycamtin ) inhibitor daily for 5 consecutive 1 mg base table oil monostearate
days repeated every 21
days
Loratadine 2008 2 Antihistamine 10 mg qd Soft gelatin cap- Caprylic/capric Polysorbate 80
R
(ClaritinV) sule, 10 mg glycerides
Isotretinoin 2012 2 IIb Retinoid 0.5–1 mg/kg/day given in LFHS, 10, 20, 25, Soybean oil, stearoyl Sorbitan
V R
(Absorica ) two divided doses 30, 35, 40 mg polyoxylglycerides monooleate
Enzalutamide 2012 2 IIb I Androgen 160 mg qd Soft gelatin cap- Caprylocaproyl
R
V
(Xtandi ) receptor sule, 40 mg polyoxyglycerides
inhibitor
R
V
Nintedanib (Ofev ) 2014 2/4 IIb II Tyrosine kinase 150 mg bid with food Soft gelatin cap- MCTs, hard fat Lecithin
inhibitor (Ofev) sule, 50 and
100 mg
Calcifediol 2016 2/4 I II/III Vitamin D 60 mcg qd at bedtime Soft gelatin cap- Mixture of lipophilic emulsifier with a HLB <7 and an absorption enhancer with
(RayaldeeTM) analog sule, 30 mcg HLB of 13–18
Oily vehicle- mineral oil, liquid paraffins, or squalene
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY
1747
1748 R. SAVLA ET AL.

Figure 1. Multidimensional (2D) scaling of the chemical structures of the dataset reveals high structural diversity. The axes (V1 and V2) represent chemical space in
two directions.

Table 2. Lipid formulation classification system.


Excipients in formulation
Content of formulation (%w/w)
Oils: triglycerides or Water-insoluble Water-soluble Hydrophilic cosolvents
mixed mono and surfactants surfactants (e.g. PEG, proylene glycol,
Characteristics diglycerides (HLB <12) (HLB >12) transcutol)
Type I Pure oils 100 — — —
Limited or no dispersion
Digestion required
Type II SEDDS 40–80 20–60 — —
Moderate dispersion needed to form an emulsion
Likely to require digestion
Type IIIA SMEDDS 40–80 — 20–40 0–40
Rapid dispersion to form micro- or nano-emulsion
May need digestion
Type IIIB SMEDDS <20 — 20–50 20–50
Rapid dispersion to form micro- or nano-emulsion
Digestion likely not needed
Type IV Oil free — 0–20 30–80 0–50
Rapid dispersion results in micellar solution
No digestion needed

that the drug will stay in solution in vivo due to the micellization variation in gastric emptying and absorption [13]. Corn oil was the
oil component of cyclosporine (SandimmuneV) capsules (Table 1).
R
process. Oils can be divided into long chain triglycerides (LCT) and
The improved formulation (NeoralV) substituted corn mono- and
R
medium chain triglycerides (MCT) [9]. Formulations containing
these typically require digestion before absorption. Lipid digestion di-glycerides for corn oil. The clinical result was less food effect
or lipolysis converts triglycerides into mono- and di-glycerides and and improved pharmacokinetic uniformity [14,15].
fatty acids. The incorporation of mixed glycerides (mono- and di-
glycerides and free fatty acids) in a formulation can improve the
solvent capacity of lipid formulation in cases of compounds that Surfactants and co-solvents
have low solubility in triglycerides alone. Another advantage of Surfactants are commonly used in lipid formulations to enhance
using mixed glycerides is that they are similar to lipid digestion drug solubility and self-emulsifying properties of the vehicle itself
products. Undigested triglycerides have poor miscibility in the to minimize dependence on a patient’s digestion factors.
aqueous environment of the gastrointestinal tract, which leads to Generally, nonionic surfactants have lower toxicity compared to
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 1749

ionic surfactants; therefore, LBDDS usually include nonionic surfac- >25 mg/ml; relative low melting point; and good chemical
tants. The marketed protease inhibitors, with the exception of stability.
saquinavir, contain considerable amounts of surfactants. Therefore, based on the need for additional guidelines for for-
Amprenavir capsules and tipranavir oral solution used vitamin E mulator’s considering a LBDDS approach, we sought to retrospect-
TPGS, which can behave as both a solubilizer and permeation ively analyze any commonalities or outliers in the set of FDA
enhancer. Ritonavir, ritonavir/lopinavir, and tipranavir capsules use approved drugs using a LBDDS to address this need. We con-
polyoxyl 35 castor oil as the surfactant. Polyoxyl 35 castor oil is structed box-and-whisker plots for the different physicochemical
used to improve solubilization, but also has been shown to modu- properties to illustrate the range, quartiles, and median values for
late P-glycoprotein (P-gp) activity on the apical membrane of the each parameter. Restricting our analysis to this set of data pro-
intestine and improve absorption of lipophilic drugs [16]. P-gp on vides some degree of assurance that these formulations are robust
the intestinal villi is known to limit oral absorption of peptidomi- and have withstood the rigors of in vitro, animal, and human test-
metics such as protease inhibitors [17]. Therefore, polyoxyl 35 cas- ing. The major drawback to this approach is that the dataset
tor oil can serve a dual purpose in formulation of drugs that are reviewed is somewhat limited in terms of number of drugs.
substrates for P-gp. As an example, early studies showed that
saquinavir is a P-gp substrate and its oral bioavailability may be
limited by P-gp efflux [18]. However, the formulation does not Chemical structure similarity of FDA approved drugs in LBDDS
contain any excipients that have the potential to inhibit drug Using an online program developed by the Girke lab, ChemMine
efflux. More recent studies using digoxin as a probe in healthy Tools, we performed a multidimensional scaling of the drug mole-
subjects have shown that both saquinavir and ritonavir have cules as shown in Figure 1 [24]. The chemical structures of the
inherent potential to inhibit P-gp [19]. In some studies, ritonavir drugs were entered into ChemMine Tools online platform and the
has also been demonstrated to induce P-gp activity [20], suggest- dissimilarity analysis was performed with a similarity cutoff of 0.7.
ing a complex interaction of the drug and transporter which is still This approach clusters compounds by structural similarity. The pro-
not completely understood. gram generates atom pair descriptors that are used to calculate a
Co-solvents such as propylene glycol and ethanol are used to similarity matrix based on common and unique atom pairs among
increase drug solubilization but also serve to enhance the disper- the compound set using the Tanimoto coefficient, which is the
sion rate of lipid formulations and are often included in SEDDS proportion of shared features between two compounds divided
and SMEDDS. However, upon dispersion in the GI fluids, they rap- by the sum of common and unique features. The Tanimoto coeffi-
idly partition into the aqueous phase and reduce the solvent cap- cient can range from 0 to 1 with a higher value means greater
acity of the formulation often leading to drug precipitation. similarity. The distance matrix is calculated using an all-against-all
comparison based on atom pair similarity and translating the simi-
larity scores into distance values. We assessed the chemical struc-
Physicochemical properties of lipid-formulated drugs tures in a two dimensional plane. The axes V1 and V2 are the axes
Choosing an appropriate formulation approach whether it be a defining two directions in a plane. As seen in Figure 1, the mole-
lipid formulation or another type of formulation is typically an cules are quite diverse in structure. The axes represent molecular
empiric endeavor that consumes significant time and resources spaces in two dimensions. The most common drug classes (circled
(including API quantity). Because of limited API quantity at early in Figure 1) are vitamin D analogs, protease inhibitors, and reti-
stages, many scientists forego lipid formulation evaluation at drug noids. These classes occupy different regions of the graph. The
discovery stages and instead utilize more conventional formulation five vitamin D analogs are distinct from any other molecule struc-
approaches such as aqueous suspensions and powder in a capsule tures and located on the left side of the graph. The retinoids are
[21]. The Lipid Formulation Classification System was developed to distinct from the other molecules as well and located on the
be used in conjunction with in vitro tests to create a system for upper right. The protease inhibitors occupy space on the bottom
the standardization of LBDDS which serves as a database correlat- right. Tipranvair is the only non-peptidomimetic protease inhibitor
ing formulation composition to predicted in vitro–in vivo perform- and is evidenced as slightly outside of the cluster of the other pro-
ance [11]. During the development of a lipid formulation such as tease inhibitors. The protease inhibitors are proximate to sirolimus
SEDDS, the formulator must take into account many considera- and cyclosporine. The similarity is likely due to the peptidomimetic
tions regarding the formulation strategy including excipients, solu- nature of protease inhibitors. This mapping illustrates that LBDDS
bility, stability, and scale-up and production [22]. In general, the is applicable to a wide range of molecular structures.
key design criteria for development of LBDDS are drug solubiliza-
tion and prevention of drug precipitation in vivo. Beyond this, Molecular weight of FDA approved drugs in LBDDS
there exists the need for practical guidelines based on a database
of molecules’ physiochemical properties to assist the formulator in The molecules in Table 1 exhibit a wide range of molecular
predicting the most promising formulation pathways for a given weights from 144 Daltons (Da) for valproic acid to 1202.64 Da for
drug compound. There are a couple of general rules based on cyclosporine with the median molecular weight of 416.64 Da
experience. Pouton and Porter [23] state that drugs with a LogP (Figure 2(A)). A large portion of these drugs have ‘high’ molecular
2 and high melting points (numerically undefined) are usually weights; 9 of the 27 drug molecules have molecular weights
poor candidates for lipid system and better suited for nanomilling above 500 Da. This group of high molecular weight drugs is com-
or amorphous formulations. Lipid systems are excellent vehicles prised of the protease inhibitors, cyclosporine A, dutasteride, nin-
for highly lipophilic drugs (LogP >5). However, there are insuffi- tedanib, and sirolimus (Supplementary Table S1).
cient studies, particularly comparative studies, for the large portion
of drugs that fall between these two extremes in LogP (drugs with
Permeability profiles of FDA approved drugs in LBDDS
LogP between 2 and 5). Similarly, Chen et al. [21] proposed the
following drug profile required for lipid formulations: aqueous There is evidence that the components of LBDDS impact intes-
solubility <10 mcg/ml; LogP >5; solubility in oils and lipids tinal permeability by changing the passive permeability and/or
1750 R. SAVLA ET AL.

Figure 2. Quantitative assessment of chemical properties. (A) molecular weight, (B) polar surface area, (C) LogP, (D) logarithm of octanol solubility, and (E) correlation
between octanol solubility and LogP.

by inhibiting efflux transport [25–28]. Medium chain fatty acids, Solubility profiles of FDA approved drugs in LBDDS
lysophospholipids, surfactants, and cosurfactants increase passive
Poorly water soluble drugs represent a diverse set of compounds
permeability via tight junction opening or via increased mem-
with different physicochemical properties. Most scientists believe
brane solubilization and increased membrane fluidity [29]. We
that lipid systems are designed for highly lipophilic drugs (LogP
sought to evaluate drug molecules in our dataset to see which
>2). Broadly speaking, poorly water soluble drugs can be classified
ones had the physicochemical characteristics indicative of poor
as non-lipophilic hydrophobic (brick dust) or lipophilic hydropho-
permeability and if they were formulated with excipients that
bic (grease balls) [34]. ‘Brick dust’ compounds have tight crystal
have been shown to improve permeability. A study of over 1100
GlaxoSmithKline drug candidates revealed that < 10 rotatable lattices (and higher melting temperatures), are not soluble in gly-
bonds and a polar surface area (PSA) < 140 Å display better per- ceride excipients, and require formulation with surfactants or co-
meation rates [30]. Another study showed that in addition to solvents (LFCS Types IIIB and IV). On the other hand, ‘grease balls’
PSA, LogD at pH ¼7.4 and radius of gyration influence Caco-2 have good solubility in lipid excipients and can be formulated into
permeation [31]. In our analysis, we looked at the PSA, which is LBDDS (LFCS Types I, II, and IIIA).
the summation of the surface area of all polar atoms (oxygen Our analysis showed the dataset of approved drugs in Table 1
and nitrogen) and their attached hydrogens. We also calculated primarily consisted of lipophilic compounds with a median LogP
the permeability in human jejunum at pH 6.5 using the ACD The of 4.85 and an interquartile range of 3.66–5.97 (Figure 2(C)). This
median PSA of the dataset is 59.45 Å (Figure 2(B)) indicating that analysis reveals a significantly higher LogP profile than most
overall these molecules have very good permeability properties. approved drugs based on an analysis by Leeson and Springthrope
In general, the protease inhibitors have relatively high PSAs looking at LogP and molecular mass trends for pipeline and
(>100 Å). These molecules also have high molecular weights approved drugs [35]. In their year-by-year analysis of 592 drugs
(> 500 Da). Only three drugs (ritonavir, saquinavir, and sirolimus) launched between 1983 and 2007, the median cLogP for launched
in our dataset have a PSA above 140 Å (Supplementary Table drugs failed to eclipse a cLogP of 4 for any year. With one excep-
S1). Ritonavir (NorvirV) is a LFCS Type III LBDDS and consists of tion, the median cLogP for every year fell between cLogP of 2 and
R

oleic acid, polyoxyl 35 castor oil, and ethanol. Saquinavir 4. There was weak correlation of increasing median cLogP with
(FortovaseV) is a LFCS Type I LBDDS and contains only medium each passing year and the slope of the straight fit line was very
R

chain mono- and di-glycerides. Sirolimus (RapamuneV) was for-


R
small. A commonly held belief is that drugs with a LogP >2 [23],
mulated as a LFCS Type III LBDDS using phosphatidylcholine, are good candidates to be formulated as LBDDS. Based on the
mono- and di-glycerides, soy fatty acids, ascorbyl palmitate, poly- analysis by Leeson and Springthrope, nearly half of drug candi-
sorbate 80, ethanol, and propylene glycol. Polyoxyl 35 castor oil dates should be formulated as LBDDS. The LogP >2 rule of thumb
and polysorbate 80 have been shown to modulate P-glycoprotein is not sufficiently discriminatory and sets the LogP bar too low.
efflux pump and effects on phospholipid membranes [32,33]. Our analysis showed that the median LogP of 4.85 for lipid formu-
Good permeation is a prerequisite for molecules that exert their lated drugs. Amprenavir, ranitidine, and topotecan were the only
pharmacological effect via nuclear receptor binding (retinoids, compounds with a LogP below 2 (Supplementary Table S1) and
vitamin D analogs, and progesterone) or need to cross the are likely outliers. The molecules with the next lowest LogP values
blood-brain barrier (dronabinol or valproic acid). It is no surprise were valproic acid and lubiprostone with values of 2.54 and 2.76,
that these molecules have the lowest PSAs (PSA range: 20.23–60. respectively. This suggests that using a LogP of 2 to filter mole-
69 Å). cules that benefit from lipid formulation may be setting the level
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 1751

too low. A better approach to decide whether a drug is a good 1.4–1.7) in lipid excipients [21]. Our calculations show that 7 drugs
candidate to be formulated as a LBDDS is to look at other factors have octanol solubility below this rule of thumb. Lubiprostone is a
as done with the DCS. LFCS Type I, ranitidine is LFCS Type IV, and the rest fall into LFCS
We set to further analyze the data set’s LogP values. Our goal Type IV. Because lipid formulations are multicomponent systems
was to determine if LogP value is a sufficient parameter to deter- and have complex behavior, it is difficult to create an in-silico pre-
mine whether a compound has suitable solubility in lipid exci- diction for determining if a drug has sufficient solubility in lipid
pients to be formulated in a lipid system. A compound’s LogP is a systems for further formulation development.
partition coefficient and influenced by two variables: its solubility Enzalutamide (LogP 3.75, log[Oct] 0.88) has both poor aqueous
in octanol and its solubility in water. We sought to assess the rela- and octanol solubility, which combine to demonstrate a rather
tive contribution of each variable on the LogP. In other words, are high LogP. Ranitidine, topotecan and amprenavir had the lowest
higher LogP driven by increasing lipid solubility, decreasing water predicted LogP (ALOGPS) at 0.79, 1.84, and 1.85, respectively.
solubility, or both? Using predicted LogP and predicted water Amprenavir (72–74  C) [36] and ranitidine (69–70  C) have low
solubility values, we were able to calculate the octanol solubility melting points. Topotecan has a melting point of 213–218  C
of FDA approved drugs formulated in lipid systems (Figure 2(D)). .Based on the above mentioned general rule, topotecan would be
The calculated octanol solubility served as a surrogate for suffi- better formulated as an amorphous solid dispersion.
cient solubility in lipid excipients/lipid formulation solvent cap-
acity. The water content in lipid formulation is absent or
negligible. Therefore, we sought to assess if drug solubility in octa- Classification according to BCS and DCS
nol has value as a predictor whether a lipid formulation is the The BCS was proposed by Amidon and colleagues to correlate in
best option compared to LogP. While this is true, LogP may be a vitro dissolution and in vivo bioavailability and illustrate that solu-
better measure of consequences of LBDDS in vivo. This approach bility and permeability were important to oral drug absorption
aims to minimize substantial influence of poor water solubility on [37]. Drugs fall within one of four classes based on their perme-
LogP prediction. From Figure 2(E), it is seen that the logarithm of ability and solubility. The majority of lipid formulated drugs fall
the octanol solubility (Log[Oct]) has a linear relationship with the into either BCS Class 2 (good permeability, poor solubility) or Class
predicted LogP. The slope and y-intercept above 1 indicate that 4 (poor solubility and permeability). The four exceptions in our
overall, the compounds in the dataset have a lipophilic tendency. dataset are ergocalciferol (BCS Class 3), valproic acid (BCS Class 1),
Based on our dataset (Figure 3(A)), there is no preference for ranitidine (BCS Class 3), and topotecan (BCS Class 1).
lipid formulation class (LFCS) based on a molecule’s LogP value. While the BCS is a good regulatory system for identifying drugs
Type I formulations, which are pure oils, have been used for drugs that may be candidates for biowaivers based on in vitro dissolution
with a wide range of LogP values. It is surprising that Type I lipid data, it is not very useful for formulators to determine suitable for-
formulation have been developed for hydrophilic molecules. The mulation approaches. Many of the problematic compounds fall
formulation rationale is not apparent. within the BCS Class 2, but there is insufficient knowledge of their
To support further formulation development studies, formula- solubility behavior. The Developability Classification System (DCS)
tors try to target drug solubility of 25–50 mg/ml (log scale: was developed by Butler and Dressman to be used as a formula-
tion tool [38]. The DCS makes a couple of important amendments
to the BCS. The solubility of the highest strength dose unit is
replaced by the highest single dose, the medium for solubility
determination is changed from buffer to fast state simulated intes-
tinal fluid, and the fluid volume is for solubility determination is
increased from 250 to 500 ml to be more representative of in vivo.
The permeability and solubility characteristics (good or poor) for
each of the four classes align between the BCS and DCS. Like Class
1 BCS drugs, DCS Class 1 drugs have good solubility and perme-
ability. Drugs falling into Class 2 DCS have good permeability and
poor solubility. They are subdivided Classes IIa and IIb based on
their solubility limited absorbable dose. The absorption of drugs in
DCS Class IIa is considered to be dissolution rate-limited because
the dissolution rate is too slow for all drug particles to be in solu-
tion at the absorption site. The absorption of Class DCS IIb drugs is
solubility-limited, which means that there is insufficient GI tract
fluid to dissolve the dose. DCS Class III drugs have good solubility
and poor permeability and those in DCS Class IV have poor perme-
ability and solubility.
Classification of compounds in the dataset according to the
DCS resulted in the molecules being placed in the following DCS
Classes: Class I (n ¼ 6), Class IIa (n ¼ 5), IIb (n ¼ 15), III (n ¼ 0), and
IV (n ¼ 1) (Figure 4). The dose/solubility ratio was calculated and
plotted on the x-axis. A higher value indicated poorer solubility in
fasting state simulated intestinal fluid. The calculated effective per-
meability in human jejunum is plotted on the y-axis. As expected,
Figure 3. Integration of different classification systems. (A) applicability of lipid
the majority of drug molecules fall within DCS Class IIb, those
formulation based on compound’s LogP. Markers represent individual LogP and compounds with intrinsic solubility limitations. It is interesting to
line represents median LogP (B) relationship between BCS and DCS. note the relatively high percentage of drugs that were classified
1752 R. SAVLA ET AL.

Figure 4. DCS plot of FDA approved drugs formulated using LBDDS with LFCS represented by different markers.

as DCS Class I. Ranitidine was the only drug with poor permeabil- FDA approved molecules in LBDDS beyond rule of five
ity (< 1  104 cm/sec). As seen in Figure 3(B), most BCS Class 2
Lipinski’s Rule of Five (Ro5) is a rule of thumb used during drug
drugs are DCS IIb with several falling into DCS Class I and IIa. The
development to evaluate drug-likeness of a molecule or determine
majority of drugs in BCS Classes 3 or 4 are typically DCS Class IIb
if a molecule is likely to be orally active in humans [41]. The Ro5
compounds with a few in DCS Classes IIa and IV.
describes chemical properties important for pharmacokinetics and
We sought to evaluate whether DCS class could aid formulators
does not predict pharmacodynamics. There are four criteria: 5
to choose the most appropriate lipid formulation type (LFCS) for
hydrogen bond donors (HBD);  10 hydrogen bond acceptors
solubilization of the entire drug dose and prevention drug precipi-
(HBA); molecular weight (MW) < 500 Daltons; and LogP <5. The
tation in vivo. In the DCS plot (Figure 4), we indicated the LFCS
Rule is said to be ‘broken’ when 2 criteria are not met. Adhering
Class using different shapes for each drug. We had expected that
to the strict guideline, most commercial oral drugs do not violate
Type I LBDDS, which consist of pure oils, would be best suited for
the Ro5. We evaluated the list of FDA approved drugs formulated
the most poorly soluble drugs (those with the highest dose/solu-
as lipid systems to see if any drugs violated the Ro5 and found
bility ratio). However, there were several DCS Classes 1 and 2a
5 cases:
drugs formulated as Type 1 LBDDS. There are numerous factors in
addition to excipient type. Fatty acid chain length of lipid exci-
pients is important as it influences lipid digestion and drug solu- Sirolimus (MW 914.17, LogP 7.45, HBA 12)
bilization. Less lipophilic drugs that can be easily solubilized to the Saquinavir (MW 670.84, HBD 6)
aqueous GI environment can be best formulated with medium Tipranavir (MW 602.7, LogP 6.29)
chain glycerides [39]. Highly lipophilic drugs are better formulated Dutasteride (MW 528.50, LogP 5.45)
using long chain glycerides in order that these drugs remain in Cyclosporine (MW 1202.64, HBA 16)
the undigested oil phase or partition into mixed micelles following
in vivo administration [40]. Furthermore to plotting the drugs on It is rather widely accepted that target biology and chemical
the DCS plot, we sought to examine if drugs that fall within a cer- properties influences the physicochemical properties of ligands.
tain region of the DCS plot will be better formulated as a self- These targets tend to have lipophilic or large, flat binding pockets.
emulsifying system (LFCS IIIA or IIIB). From our analysis (Figure 4), An analysis of recently patented molecules, Bergstro €m et al. [42]
there is no clear correlation between DCS Class and LFCS formula- discovered that molecules are almost exclusively lipophilic for cer-
tion type, and in fact the data is in some cases counter-intuitive. tain targets. Approximately 50% of the targets had molecules with
This is the likely due to the limited dataset. As shown is Figure 4, an average LogP 4. With a limited dataset of approved drugs
the majority of molecules falling into DCS class I appear to be for- using LBDDS, we were unable to extend our analysis across a tar-
mulated as Type I lipid formulations. Type I lipid formulations get or drug class. Our analysis revealed that 64% (16 of 25) drug
were used for the DCS classes I, IIa, and IIb (Figure 4). DCS IIb molecules had a LogP >4. Of the molecules violating the Ro5,
compounds, which were the most common, were shown to be three compounds have high LogP values.
formulated using all types of lipid systems with a preference for An analysis by Paul Leeson showed that while increasing lipo-
Type I lipid formulations. philic character of drugs began after 1990 (predicted LogP of 2.5
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 1753

from 1950–1990, predicted LogP 3.3 since 1990), there are been The three selected parameters of approved drugs that were not
a steady increase in molecular weights since the 1950s (MW 300 developed as LBDDS, compared to those that were, had the fol-
in the 1950s to 450 in the 2000s) [43]. In our dataset, molecular lowing median values and p values, respectively: LogP 4.02 and
weight >500 is the most common physicochemical property viola- 3.85, p values of .13888; effective permeability 6.88  104 cm/s
tion for molecule breaking the Ro5. Our review revealed that 9 of and 6.92  104 cm/s, p values of .82588; and median dose 250 mg
25 molecules had molecular weights >500. Protease inhibitors are and 100 mg, p values of .07508. None of the measurements
known for the high molecular weights, but tipranavir is only prote- between the two sets were shown to be statistically different
ase inhibitor with a LogP that violates the Lipinski parameter. This (p < .05); therefore, the two data sets are comparable. The median
may be explained based on chemical structure since tipranavir magnitude of food effect on area under the curve for the oral
does not contain a peptidomimetic hydroxyethylene core and comparator set was þ35% (range –31 to þ400%) and for the
other protease inhibitors do. LBDDS set, the median magnitude was þ16.5% (range –13
to þ100%). However, the difference was not statistically significant
with a p value of .63122. Therefore, it cannot be concluded from
LBDDS to reduce magnitude of food effect this exercise that LBDDS blunt the food effect.
The absorption of poorly water soluble drugs is improved when
administered with food, termed positive food effect [3]. This find- Future directions in oral LBDDS delivery
ing led to the rationalization that formulating these drugs as
LBDDS could improve bioavailability. LBDDS induce, but to a lower From our analysis, it is evident that lipid formulations are versatile
magnitude, the same physiological events seen during meal con- platforms for drugs with a wide range of physicochemical proper-
sumption. Pancreatic and gallbladder secretions initiate lipid diges- ties. These drugs tend to be lipophilic and permeable thus con-
tion and solubilization of digestion products in mixed micelles. firming the widely held belief about ideal candidates to be
Eventual colloidal particles solubilize lipids and coadministered formulated as lipid formulations. Lipid formulations are commonly
drugs. Based on the foundation, LBDDS are an attractive formula- used for certain classes of drugs such as retinoids, vitamin D ana-
tion option for poorly water soluble drugs that experience a posi- logs, and protease inhibitors. There are several unexplored areas
tive food effect and we hypothesized that the pharmacokinetics of where lipid formulations may play a role in the future.
drugs formulated as LBDDS should not have significant food
effects. The general relationship between food effect and BCS
Drugs for cardiovascular disease
Classification states that Class 1 compounds have no food effect,
Class 3 have a negative food effect, and Classes 2 and 4 have a Currently approved drugs using lipid formulation technology cover
positive food effect [44,45]. We reviewed the package inserts to many therapeutic categories, but none are used for cardiovascular
see if there was any mention of food effect or dosing with regard disease treatment. Cardiovascular drugs are one the most com-
to meals. Table 3 summarizes the findings. The labels for 21 drug mon pharmaceutical categories with more than 100 FDA approved
products included language on administration with food. The drugs and represent an enticing area for lipid formulations. Rao
drugs labeled to be taken with or without food show increased, et al. analyzed the pharmacokinetic profiles of cardiovascular
decreased, or unchanged area under the plasma-time curve (AUC) drugs and discovered that over 80% had bioavailability challenges
or Cmax, but the magnitude of change in the pharmacokinetic [46]. The primary reasons for these challenges were extensive first
parameter is not clinically relevant. Of the drugs labeled to be pass metabolism and poor water solubility. Because of the low
taken with food, eight drugs experience positive food effect and and variable bioavailabilities of some of the best selling drugs, the
one (lubiprostone) has negative food effect. Lubiprostone is authors recommended the use of advanced delivery systems to
labeled to be taken with food to maintain a lower Cmax. This may optimize the drugs and presented several case studies demon-
be to minimize side effects caused by spikes in serum strating solubility improvement and enhanced absorption of car-
concentrations. diovascular drugs when formulated into SEDDS. Several
We hypothesize based on our review of approved drugs formu- cardiovascular drugs experience negative food effects (captopril,
lated as LBDDS experience food effect to a lesser degree than moexipril, quinapril, ramipril, losartan, and pravastatin) and may
drugs with similar solubility properties that are no formulated in not be the best candidates for lipid formulations.
lipid-based systems. In support of our hypothesis, we compared
the physicochemical properties of solubility (LogP), permeability
(effective permeability), and dose for the approved drugs listed in Oral delivery of peptides
Table 3 to an analysis of these same parameters (LogP, effective Lipid formulations are used for several high molecular weight mol-
permeability, and dose) in a comparator test set of 15 oral ecules including peptides and peptidomimetics. Peptides are
approved drugs consisting of protein kinase inhibitors, BRAF inhib- becoming increasingly common in clinical practice especially for
itors, and MEK inhibitors that are not formulated as lipid-based the treatment of diabetes. Insulin and glucagon like peptide 1
systems. The rationale for selecting the above three parameters (GLP-1) analogs are typically administered by subcutaneous injec-
(LogP, effective permeability, and dose) is based on previous stud- tions. These peptides have high molecular weights, short half-lives
ies that suggest these three physiochemical properties can be and high clearance. Bioengineering modifications have been used
used to accurately categorize the food effect (positive, negative, or to improve the pharmacokinetics. However, these improved pepti-
neutral) in 80% of a test group of drugs [45]. We wanted to first des must still be injected.
ensure that the two sets of approved drug were comparable in The use of colloidal carriers offers protection against enzymatic
terms of physicochemical properties that influence food effects degradation in the gastrointestinal tract [47]. Microemulsions are
(LogP, effective permeability, and dose). To do so, we calculated more likely than polymeric carriers to be developed into an
the median values and performed the two-tailed Mann-Whitney approved oral drug product, indicated by the number of approvals
U test with a significance level of 0.05 to compare to difference using each approach. For example, peptides are hydrophilic drugs
in median values between the two data sets of approved drugs. that can be classified as BCS Class III compounds. Permeation and
1754 R. SAVLA ET AL.

Table 3. Food effects and pharmacokinetics of approve drugs.


With With or
Molecule/trade name BCS/DCS food without food Increased Decreased No change
Isotretinoin 2/IIb X Cmax and AUC more than doubled fol-
(AccutaneV)
R
lowing high fat meal
Ritonavir (NorvirV) Absorption þ13% with a meal
R
4/IIb X
Clofazimine 2 X Bioavailability þ62% with food, higher
(LampreneV)
R
peak concentration, faster Tmax
Saquinavir (FortovaseV)
R
4/IIb x 12-h AUC after single dose was
increased from when given with
breakfast
Bexarotene (TargretinV) AUC þ35% and Cmax þ48%
R
?/IIb X
Ritonavir/lopinavir X Moderate fat meal: AUC þ48% and
(KaletraV) Cmax þ23%
R

High fat meal: AUC þ97% and Cmax


þ43%
Lubiprostone 2,4/I X Cmax decreased AUC was unchanged
(AmitizaV)
R
by55% when administered
with high fat meal
Fenofibrate (LipofenV) AUC þ58% and þ25% when adminis-
R
2/IIb X
tered with high fat and low fat
meals
Nintedanib (OfevV)
R
2,4/IIb X AUC increased by 20% and Tmax
delayed
V
R
Tipranavir (Aptivus ) 4/IIb X X No clinically significant
changes
V
R
Cyclosporin A (Neoral ) 2/IIb X Decreased AUC by 13%
and Cmax by 33%
Progesterone 2/IIb X Increased bioavailability with food
(PrometriumV)
R

Amprenavir 2/IIb X Not taken with high fat meal Cmax and AUC
(AgeneraseV)
R
decreased and Tmax
prolonged
Sirolimus (RapamuneV) AUC þ23–35%
R
?/IIa X Effect on CMAX was
inconsistent
V
R
Dutasteride (Avodart ) 2,4/IIa X Cmax decreased by
10–15%
Loratadine (ClaritinV)
R
2 X No clinically significant
changes
Omega-3-acid ethyl X
esters (LovazaV)
R

Paricalcitol (ZemplarV)
R
4/IIa X AUC and Cmax were
unchanged. Tmax
was delayed by 2 h
Topotecan HCl 1/I X AUC similar. Tmax was
(HycamtinV)
R
delayed
Isotretinoin (AbsoricaV) AUC and Cmax þ50% and þ26%,
R
2/IIb X
respectively. Tmax was delayed
V
R
Enzalutamide (Xtandi ) 2/IIb X Similar AUC when fast-
ing or fed

stability in the GI tract are the primary challenges to the oral of these drugs have poor water solubility and significant food
absorption of peptides. As a result of their hydrophilic nature, pep- effects. More specifically, the majority of these agents have a posi-
tides can reside in the aqueous phase of water-in-oil (w/o) micro- tive food effect and are labeled to be taken on an empty stomach,
emulsions. The oil phase can minimize exposure to water and 2 h before or 1 h after a meal [51,52]. The increase in AUC and
protect against degradation. Surfactants, co-surfactants, and Cmax due to food can result in supratherapeutic and toxic concen-
medium chain diacylglycerols in the formulation may improve trations of the anticancer drugs, a number of which carry FDA black
membrane permeability and therefore enhance peptide drug bio- box warnings for potentially life-threatening risks associated with
availability. Furthermore, there is potential to reduce peptide deg- the drug [53]. One of the most common and serious side effects
radation in the acidic stomach and the enzymatically active would be QTc prolongation, potentially creating dangerous cardiac
regions of the gastrointestinal tract given the protection afforded arrhythmias. Considering that patients with chronic disease typically
by a w/o microemulsion [48–50]. take multiple medications for multiple conditions, they may not be
able to adhere to a strict guideline of taking a medication on an
empty stomach. Lipid formulations are known to mimic, to a certain
Oral anticancer drugs extent, the high fat content of a meal and increase solubilization of
Protein kinase inhibitors have led the way in the development of the drug molecule [54]. Therefore, lipid formulations of anticancer
oral anticancer agents. Compared with traditional chemotherapeu- agents have the potential to improve the absorption and minimize
tic agents, these newer drugs are targeted, have better side effect the food effect of anticancer agents thereby eliminating safety con-
profiles, and are administered orally and chronically. The majority cerns arising from lack of patient adherence.
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 1755

Table 4. Oral amphotericin formulations in development.


Relative oral bioavailabilitya
compared to oral FungizoneV
R
Formulation Type of DDS Preclinical efficacy
Deoxycholate/phytantriol/poloxamer 407 Nanostructured liquid-crystalline 2.85 nd
[60] particles (cubosomes) (rats, 10mg/kg)
Copaiba oil, phosphatidylcholine, surfactants Nanoemulsified carrier system 10.5 nd
[77] (rats, 10 mg/kg)
Mono- and diglycerides, Vitamin E-TPGS SEDDS nd Visceral leishmaniasis (mice): 10 mg/kg
[78,79] Fabs 6.25%b bid 5 d: 99% inhibition liver
parasites
Glyceryl monooleate [70,73] Lipid suspension 2.21 Aspergillus fumagatus (rats)
(rats, 50mg/kg) 50mg/kg 1 dose: 95% reduction in
organ CFUs
Phosphatidylserine, calcium [80] Cochleate nd Aspergillus fumagatus (immunocom-
promised mice) 40 mg/kg/day: 70%
survival on day 14
Glyceryl dilaurate, phosphatidylcholine and Solid lipid nanoparticles Fabs 105%
polyethylene glycol (15)-hydroxystearate
[62]
a
AUC oral formulation/AUC oral Fungizone, at the same dose.
b
Oral bioavailability of 10 mg/kg compared to IV Fungizone 0.8 mg/kg over 48 h.

Amphotericin B order to achieve not only improved oral absorption but also the
desired biodistribution as discussed above, by accessing the
Amphotericin B is a polyene macrolide antibiotic used in the treat-
lymphatic transport pathways utilized by dietary lipids [63–67].
ment of serious systemic fungal infections, acting by binding to
When suspended in PeceolV (glyceryl monooleate), amphotericin B
R

fungal cell wall sterols. The drug has amphoteric features as well
(AmpB)’s bioavailability is significantly improved [68–70]. AmpB in
as hydrophobic and hydrophilic portions of this relatively large,
a Peceol/Gelucire 44/14 (lauroyl macrogol-32 glycerides)-based for-
small-molecule drug (MW¼ 924). It has pKa’s of 2, 5.5 and 10.0. At
mulation (iCo -010) has demonstrated efficacy in several animal
28  C, its water solubility is only 0.75 mg/mL and it is also practic- models of systemic fungal infections [71] as well as visceral leish-
ally insoluble in ethanol [55]. maniasis [72,73]. Organ distribution of amphotericin B is shifted
To understand the goals and limitations of developing the away from the kidney, the site of dose-limiting toxicity, with a cor-
highly desirable oral formulation of Amphotericin B, it is important responding increased splenic and hepatic uptake when the drug is
to understand the history and biophysical behavior of LBDDS administered as LBDDS [67].
intravenous formulations. The deoxycholate formulation An alternative LBDDS is the multi-layered, spiral-shaped cochle-
(FungizoneV injection) was developed in the 1970 s to address the
R

ate formulation [74]. Precipitates of phosphatidylserine and cal-


solubility problems; however, its use is limited by infusion-related cium incorporating amphotericin B were prepared by Sesana et al.
reactions and dose-related nephrotoxicity. To reduce renal toxicity [75] and demonstrated to have in vitro activity against Leishmania
and increase the therapeutic efficacy of Amphotericin B, intraven- chagasi. However, further development of amphotericin B lipid
ous lipid formulations of Amphotericin B including Ablecet (1995), cochleates has been limited, perhaps in part due to the low bio-
Amphotec (1996), and Ambisome (1997) were approved [56]. availability of the cochleate formulation [76]. There are additional
Abelcet (Amphotericin B lipid complex injection) is comprised of LBDDS in preclinical testing that have promising improvements in
5 mg of Amphotericin B per dose with the lipid excipients L- bioavailability as well, indicated in Table 4. It remains to be seen
a-dimyristoylphosphatidylcholine (DMPC, 3.4 mg) and L-a-dimyris- which oral formulation is eventually developed and accepted. This
toylphosphatidylglycerol (DMPG, 1.5 mg) [57]. The complex forms will depend not only differential efficacy and toxicity in humans,
a stable ribbon-like structure with Amphotericin B with a hetero- but also the ease of manufacturing scale-up and market
geneous 2–5 lm diameter size range [58,59], which is hypothe- competition.
sized to be responsible for its altered biodistribution and toxicity
profile compared to the desoxycholate formation. An alternative
LBDDS was Amphotec/Amphocil, which is comprised of disc-like Conclusion
structures containing cholesterol sulfate that are significantly LBDDS are the most studied bioavailability enhancing technology
smaller, at around 120 nm [60]. Ambisome is comprised of unila- and have been utilized in dozens of FDA approved drugs. Our pri-
mellar phospholipid liposomes <100 nm in mean diameter, com- mary focus was to analyze approved drug molecules that bene-
prised of hydrogenated soy phosphatidylcholine and distearoyl fited from being formulated in LBDDS. We examined the
phosphatidylglycerol. The size and composition of the structures is physicochemical characteristics of these drugs, and we classified
important because it determines not only stability in circulation them according to different classification systems (BCS, DCS, and
but also organ biodistribution, with a significant portion of the LFCS) to see if there were any correlations between physicochemi-
dose going to organs of the reticuloendothelial system. cal characteristics and classification systems or between classifica-
While these lipid-formulations have been very successful, the tion systems. Our analysis of the properties of FDA approved
major limitations of their use are that they must be administered drugs showed a diverse set of drug molecules including a range
intravenously and they are very expensive. Hyperkalemia and of molecular weights, solubilities, PSA, and chemical structures. As
other electrolyte disturbances can be a problem with liposomal expected, the majority of these molecules have poor solubility and
Amphotericin B [61]. The oral bioavailability of Amphotericin B good permeability. We introduced a new physicochemical param-
is <5% and for the deoxycholate form is only 2% [62]. Barriers to eter of octanol solubility, as a surrogate for drug solubility in lipid
oral absorption are considerable due to its lack of both solubility systems/solvent capacity, and demonstrated its potential value for
and lipophilicity. However, LBDDS are ideal for Amphotericin B in guiding lipid formulation. Specifically, logarithm of the octanol
1756 R. SAVLA ET AL.

solubility (Log[Oct]) has a linear relationship with the predicted [11] Pouton CW. Formulation of poorly water-soluble drugs for
LogP not related to simply poor water solubility. A Log[Oct] val- oral administration: physicochemical and physiological
ue >2.5 may be a more useful predictor of suitability in LBDDS issues and the lipid formulation classification system. Eur J
than LogP alone. Although the analysis revealed some clear Pharm Sci. 2006;29:278–287.
emerging trends with respect to choosing the best lipid formula- [12] Griffin B. Advances in lipid-based formulations: overcoming
tion type based on DCS class, e.g. DCS class I molecules are typic- the challenge of low bioavailability for poorly water soluble
ally formulated as Type I lipid formulations, the dataset would drug compounds. Am Pharm Rev. March 2012. Available
benefit from expansion in order to make a firm recommendation. from: http://www.americanpharmaceuticalreview.com/
It is hoped that future studies will expand this dataset with an Featured-Articles/39299-Advances-in-Lipid-based-
integrated assessment of the various physicochemical properties Formulations-Overcoming-the-Challenge-of-Low-
to give the best guidance for new oral drug formulation design. Bioavailability-for-Poorly-Water-Soluble-Drug-Compounds/
[13] MacGregor KJ, Embleton JK, Lacy JE, et al. Influence of lip-
olysis on drug absorption from the gastro-intestinal tract.
Disclosure statement Adv Drug Deliv Rev. 1997;25:33–46.
[14] Kahan BD, Dunn J, Fitts C, et al. Reduced inter- and intra-
No potential conflict of interest was reported by the authors. subject variability in cyclosporine pharmacokinetics in renal
RS, JB, and VP are employees of Catalent Pharma Solutions.
transplant recipients treated with a microemulsion formula-
tion in conjunction with fasting, low-fat meals, or high-fat
ORCID meals. Transplantation 1995;59:505–511.
[15] Mendez R, Abboud H, Burdick J, et al. Reduced intrapatient
Ronak Savla http://orcid.org/0000-0002-0907-4714 variability of cyclosporine pharmacokinetics in renal trans-
plant recipients switched from oral sandimmune to neoral.
Clin. Ther.. 1999;21:160–171.
[16] Katneni K, Charman SA, Porter CJ. Impact of cremophor-EL
References and polysorbate-80 on digoxin permeability across rat
[1] Dahan A, Hoffman A. Rationalizing the selection of oral lipid jejunum: delineation of thermodynamic and transporter
based drug delivery systems by an in vitro dynamic lipolysis related events using the reciprocal permeability approach.
model for improved oral bioavailability of poorly water sol- J Pharm Sci. 2007;96:280–293.
[17] Wacher VJ, Silverman JA, Zhang Y, et al. Role of P-glycopro-
uble drugs. J Control Release. 2008;129:1–10.
tein and cytochrome P450 3A in limiting oral absorption of
[2] Crew M. Bioavailability enhancement - analysis of
peptides and peptidomimetics. J Pharm Sci. 1998;87:
the Historical Use of Solubilization Technologies. Drug
1322–1330.
Development & Delivery. 2014. Available from: http://www.
[18] Kim AE, Dintaman JM, Waddell DS, et al. Saquinavir, an
drug-dev.com/Main/Back-Issues/BIOAVAILABILITY-
HIV protease inhibitor, is transported by P-glycoprotein.
ENHANCEMENT-Analysis-of-the-Histor-657.aspx#sthash.
J Pharmacol Exp Ther. 1998;286:1439–1445.
xOZ5heIQ.dpuf
[19] Schmitt C, Kaeser B, Riek M, et al. Effect of saquinavir/
[3] Feeney OM, Crum MF, McEvoy CL, et al. 50years of oral
ritonavir on P-glycoprotein activity in healthy volunteers
lipid-based formulations: Provenance, progress and future
using digoxin as a probe. CP. 2010;48:192–199.
perspectives. Adv Drug Deliv Rev. 2016;101:167–194. [20] Perloff MD, Von Moltke LL, Marchand JE, et al. Ritonavir
[4] Strickley RG. Currently marketed oral lipid-based dosage induces P-glycoprotein expression, multidrug resistance-
forms: drug products and excipients. In: Hauss D, editor. associated protein (MRP1) expression, and drug transporter-
Oral lipid based formulations. 1st ed. New York: Informa mediated activity in a human intestinal cell line. J Pharm
Healthcare; 2007. Sci. 2001;90:1829–1837.
[5] Wishart DS, Knox C, Guo AC, et al. DrugBank: a comprehen- [21] Chen XQ, Gudmundsson OS, Hageman MJ. Application of
sive resource for in silico drug discovery and exploration. lipid-based formulations in drug discovery. J Med Chem.
Nucleic Acids Res. 2006;34:D668–D672. 2012;55:7945–7956.
[6] Strickley RG. Solubilizing excipients in oral and injectable [22] Gao P. MW. Case studies: development of self-emulsifying
formulations. Pharm Res. 2004;21:201–230. formulations for improving the oral bioavailability of poorly
[7] Agrawal SGT, Tripathi DK, Alexander A. A review on novel soluble, lipophilic drugs. In: DJ H, editor. Oral lipid-based
therapeutic strategies for the enhancement of solubility for formulations enhancing the bioavailability of poorly water
hydrophobic drugs through lipid and surfactant based self soluble drugs. 1st ed. New York: Informa Healthcare; 2007.
micro emulsifying drug delivery system: a novel approach. [23] Pouton CW, Porter CJ. Formulation of lipid-based delivery
Am J Drug Discov Dev. 2012;2:143–183. systems for oral administration: materials, methods and
[8] Porter CJH. Oral lipid-based formulations: Using pre-clinical strategies. Adv Drug Deliv Rev. 2008;60:625–637.
data to dictate formulation strategies for poorly water sol- [24] Backman TW, Cao Y, Girke T. ChemMine tools: an online
uble drugs. In: Hauss D, editor. Oral lipid-based formula- service for analyzing and clustering small molecules.
tions: enhancing the bioavailability of poorly water-soluble Nucleic Acids Res. 2011;39:W486–W491.
drugs. New York Informa Healthcare; 2007. p. 185–205. [25] Constantinides PP, Wasan KM. Lipid formulation strategies
[9] Gullapalli RP. Soft gelatin capsules (softgels). J Pharm Sci. for enhancing intestinal transport and absorption of
2010;99:4107–4148. P-glycoprotein (P-gp) substrate drugs: in vitro/in vivo case
[10] Jones WJ, 3rd, Francis JJ. Softgels: consumer perceptions studies. J Pharm Sci. 2007;96:235–248.
and market impact relative to other oral dosage forms. Adv [26] Aungst BJ. Absorption enhancers: applications and advan-
Ther. 2000;17:213–221. ces. AAPS J. 2012;14:10–18.
DRUG DEVELOPMENT AND INDUSTRIAL PHARMACY 1757

[27] Akhtar N, Ahad A, Khar RK, et al. The emerging role of P- [46] Rao S, Tan A, Thomas N, et al. Perspective and potential of
glycoprotein inhibitors in drug delivery: a patent review. oral lipid-based delivery to optimize pharmacological thera-
Expert Opin Ther Pat. 2011;21:561–576. pies against cardiovascular diseases. J Control Release.
[28] Kayser O, Olbrich C, Yardley V, et al. Formulation of ampho- 2014;193:174–187.
tericin B as nanosuspension for oral administration. Int J [47] Martins S, Sarmento B, Ferreira DC, et al. Lipid-based col-
Pharm. 2003;254:73–75. loidal carriers for peptide and protein delivery – liposomes
[29] Lindmark T, Kimura Y, Artursson P. Absorption enhance- versus lipid nanoparticles. Int J Nanomed. 2007;2:595–607.
ment through intracellular regulation of tight junction per- [48] Hintzen F, Perera G, Hauptstein S, et al. In vivo evaluation
meability by medium chain fatty acids in Caco-2 cells. of an oral self-microemulsifying drug delivery system
J Pharmacol Exp Ther. 1998;284:362–369. (SMEDDS) for leuprorelin. Int J Pharm. 2014;472:20–26.
[30] Veber DF, Johnson SR, Cheng HY, et al. Molecular proper- [49] Liu D, Kobayashi T, Russo S, et al. In vitro and in vivo evalu-
ties that influence the oral bioavailability of drug candi- ation of a water-in-oil microemulsion system for enhanced
dates. J Med Chem. 2002;45:2615–2623. peptide intestinal delivery. AAPS J. 2013;15:288–298.
[31] Hou TJ, Zhang W, Xia K, et al. ADME evaluation in drug dis- [50] Guo L, Ma E, Zhao H, et al. Preliminary evaluation of a
covery. 5. Correlation of Caco-2 permeation with simple novel oral delivery system for rhPTH1-34: in vitro and in
molecular properties. J Chem Inf Comput Sci. 2004;44: vivo. Int J Pharm. 2011;420:172–179.
1585–1600. [51] Ratain MJ. Flushing oral oncology drugs down the toilet.
[32] Flaten GE, Luthman K, Vasskog T, et al. Drug permeability J Clin Oncol. 2011;29:3958–3959.
across a phospholipid vesicle-based barrier 4. The effect of [52] Kang SP, Ratain MJ. Inconsistent labeling of food effect for
tensides, co-solvents and pH changes on barrier integrity and oral agents across therapeutic areas: differences between
on drug permeability. Eur J Pharm Sci. 2008;34:173–180. oncology and non-oncology products. Clin Cancer Res.
[33] Cornaire G, Woodley JF, Saivin S, et al. Effect of polyoxyl 35 2010;16:4446–4451.
castor oil and Polysorbate 80 on the intestinal absorption [53] Food and Drug Administration. Drug Watch. [cited 2016
of digoxin in vitro. Arzneimittelforschung 2000;50:576–579. Nov 4]. Available at: https://www.drugwatch.com/2012/01/
[34] Mullertz A, Ogbonna A, Ren S, et al. New perspectives on 18/fda-black-box-warnings/
lipid and surfactant based drug delivery systems for oral [54] Cerpnjak K, Zvonar A, Gasperlin M, et al. Lipid-based sys-
delivery of poorly soluble drugs. J Pharm Pharmacol. tems as a promising approach for enhancing the bioavail-
2010;62:1622–1636. ability of poorly water-soluble drugs. Acta Pharmaceutica
[35] Leeson PD, Springthorpe B. The influence of drug-like con- (Zagreb, Croatia) 2013;63:427–445.
cepts on decision-making in medicinal chemistry. Nat Rev [55] Yalkowsky SH. Handbook of Aqueous Solubility Data: An
Drug Discov. 2007;6:881–890. Extensive Compilation of Aqueous Solubility Data for
[36] Adkins JC, Faulds D. Amprenavir. Drugs 1998;55:837–842. Organic Compounds Extracted from the AQUASOL
discussion 43-4. dATAbASE. Boca Raton, Florida: CRC Press LLC; 2003.
[37] Amidon GL, Lennernas H, Shah VP, et al. A theoretical basis p. 1263.
for a biopharmaceutic drug classification: the correlation of [56] Food and Drug Administration. Drugs. [cited 2016 Nov 4].
in vitro drug product dissolution and in vivo bioavailability. Available from: http://www.fda.gov/Drugs/default.htm
AbelcetV [package insert]. Gaithersburg, MD: Sigma-Tau
R
Pharm Res. 1995;12:413–420. [57]
[38] Butler JM, Dressman JB. The developability classification sys- Pharmaceuticals Inc; 2013.
tem: application of biopharmaceutics concepts to formula- [58] Janoff AS, Boni LT, Popescu MC, et al. Unusual lipid struc-
tion development. J Pharm Sci. 2010;99:4940–4954. tures selectively reduce the toxicity of amphotericin B. Proc
[39] Dahan A, Hoffman A. The effect of different lipid based for- Natl Acad Sci USA. 1988;85:6122–6126.
mulations on the oral absorption of lipophilic drugs: the [59] Mistro S, Maciel Ide M, de Menezes RG, et al. Does lipid
ability of in vitro lipolysis and consecutive ex vivo intestinal emulsion reduce amphotericin B nephrotoxicity? A system-
permeability data to predict in vivo bioavailability in rats. atic review and meta-analysis. Clin Infect Dis. 2012;54:
Eur J Pharm Biopharm. 2007;67:96–105. 1774–1777.
[40] Porter CJ, Kaukonen AM, Taillardat-Bertschinger A, et al. [60] Guo LS. Amphotericin B colloidal dispersion: an improved
Use of in vitro lipid digestion data to explain the in vivo antifungal therapy. Adv Drug Deliv Rev. 2001;47:149–163.
performance of triglyceride-based oral lipid formulations [61] Johansen HK, Gotzsche PC. Amphotericin B lipid soluble for-
of poorly water-soluble drugs: studies with halofantrine. mulations versus amphotericin B in cancer patients with
J Pharm Sci. 2004;93:1110–1121. neutropenia. Cochrane Database Syst Rev
[41] Lipinski CA, Lombardo F, Dominy BW, et al. approaches to 2014;(9):CD000969.
estimate solubility and permeability in drug discovery and [62] Chen Y-C, Su C-Y, Jhan H-J, et al. Physical characterization
development settings. Adv Drug Deliv Rev. 2001;46:3–26. and in vivo pharmacokinetic study of self-assembling
[42] Bergstrom CA, Charman WN, Porter CJ. Computational pre- amphotericin B-loaded lecithin-based mixed polymeric
diction of formulation strategies for beyond-rule-of-5 com- micelles. Int J Nanomed. 2015;10:7265–7274.
pounds. Adv Drug Deliv Rev. 2016;101:6–21. [63] Porter CJ, Trevaskis NL, Charman WN. Lipids and lipid-based
[43] Leeson PD. Molecular inflation, attrition and the rule of five. formulations: optimizing the oral delivery of lipophilic
Adv Drug Deliv Rev. 2016;101:22–33. drugs. Nat Rev Drug Discov. 2007;6:231–248.
[44] Fleisher D, Li C, Zhou Y, et al. Drug, meal and formulation [64] Trevaskis NL, Charman WN, Porter CJ. Lipid-based delivery
interactions influencing drug absorption after oral adminis- systems and intestinal lymphatic drug transport: a mechan-
tration. Clin Pharmacokinet. 1999;36:233–254. istic update. Adv Drug Deliv Rev. 2008;60:702–716.
[45] Gu CH, Li H, Levons J, et al. Predicting effect of food on [65] Yanez JA, Wang SW, Knemeyer IW, et al. Intestinal lymph-
extent of drug absorption based on physicochemical prop- atic transport for drug delivery. Adv Drug Deliv Rev.
erties. Pharm Res. 2007;24:1118–1130. 2011;63:923–942.
1758 R. SAVLA ET AL.

[66] Zgair A, Wong JC, Lee JB, et al. Dietary fats and pharma- B following administration to mice. J Antimicrob
ceutical lipid excipients increase systemic exposure to orally Chemother. 2010;65:535–537.
administered cannabis and cannabis-based medicines. Am J [73] Wasan EK, Gershkovich P, Zhao J, et al. A novel tropically
Trans Res. 2016;8:3448–3459. stable oral amphotericin B formulation (iCo-010) exhibits
[67] Sachs-Barrable K, Lee SD, Wasan EK, et al. Enhancing drug efficacy against visceral Leishmaniasis in a murine model.
absorption using lipids: a case study presenting the devel- PLoS Negl Trop Dis. 2010;4:e913.
opment and pharmacological evaluation of a novel lipid- [74] Perlin DS. Amphotericin B cochleates: a vehicle for oral
based oral amphotericin B formulation for the treatment of delivery. Curr Opin Investig Drugs. 2004;5:198–201.
systemic fungal infections. Adv Drug Deliv Rev. 2008;60: [75] Sesana AM, Monti-Rocha R, Vinhas SA, et al. In vitro activity
692–701. of amphotericin B cochleates against Leishmania chagasi.
[68] Risovic V, Boyd M, Choo E, et al. Effects of lipid-based oral Mem Inst Oswaldo Cruz. 2011;106:251–253.
formulations on plasma and tissue amphotericin B concen- [76] Lemke A, Kiderlen AF, Kayser O. Amphotericin B. Appl
trations and renal toxicity in male rats. Antimicrob Agents Microbiol Biotechnol. 2005;68:151–162.
Chemother. 2003;47:3339–3342. [77] Gupta PK, Jaiswal AK, Asthana S, et al. Synergistic enhance-
[69] Gershkovich P, Wasan EK, Lin M, et al. Pharmacokinetics ment of parasiticidal activity of amphotericin B using
and biodistribution of amphotericin B in rats following copaiba oil in nanoemulsified carrier for oral delivery: an
oral administration in a novel lipid-based formulation. approach for non-toxic chemotherapy. Br J Pharmacol.
J Antimicrob Chemother. 2009;64:101–108. 2015;172:3596–3610.
[70] Sivak O, Gershkovich P, Lin M, et al. Tropically stable novel [78] Yang Z, Chen M, Yang M, et al. Evaluating the potential of
oral lipid formulation of amphotericin B (iCo-010): biodistri- cubosomal nanoparticles for oral delivery of amphotericin B
bution and toxicity in a mouse model. Lipids Health Dis. in treating fungal infection. Int J Nanomed. 2014;9:327–336.
2011;10:135. Epub 2011/08/10. [79] Yang Z, Tan Y, Chen M, et al. Development of amphotericin
[71] Risovic V, Rosland M, Sivak O, et al. Assessing the antifungal B-loaded cubosomes through the SolEmuls technology for
activity of a new oral lipid-based amphotericin B formula- enhancing the oral bioavailability. AAPS PharmSciTech.
tion following administration to rats infected with 2012;13:1483–1491.
Aspergillus fumigatus. Drug Dev Ind Pharm. 2007;33: [80] Delmas G, Park S, Chen ZW, et al. Efficacy of orally delivered
703–707. cochleates containing amphotericin B in a murine model of
[72] Gershkovich P, Wasan EK, Sivak O, et al. Visceral leishmania- Aspergillosis. Antimicrob Agents Chemother.
sis affects liver and spleen concentrations of amphotericin 2002;46:2704–2707.

Potrebbero piacerti anche