You are on page 1of 26

Pharmacosomes: the versatile colloidal carriers

Shubhini A. Saraf1, Jovita Kanoujia1, Shailendra K. Saraf2


1
Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, VidyaVihar,
Raebareilly Road, Lucknow, U.P. India-226025
2
Babu Banarasi Das Northern India Institute of Technology, Dr. Akhilesh Das Nagar, Faizabad
Road, Lucknow, U.P. India-227105

Abstract: New effective delivery systems have been developed to reduce the problem of poor
bioavailability. Vesicular systems such as ethosomes, transfersomes, liposomes, niosomes and
pharmacosomes are able to improve upon the biopharmaceutical properties of the drug, resulting in
improved bioavailability. Pharmacosomes, as adaptable vesicle systems, possess inimitable advantages
over various classical vesicular drug delivery systems. Pharmacosomes can be fabricated by simple as
well as novel methods. This chapter discusses the fabrication techniques of pharmacosomes, methods of
physiochemical characterization, applications, mechanism of internalization and future possibility in the
field of drug delivery.

Keywords: Vesicular systems, hollow carriers, drug-phospholipid complex

Introduction

In the past few decades, extensive attention has been focused on the development of new drug delivery
systems. The new drug delivery system should ideally fulfill four fundamental requirements, namely
increased solubility and stability of drugs in vivo, reduced toxic effects, improved drug bioavailability,
and delivery at or near the site of action.1

The word vesicle literally means a membrane enclosed sack which is able to store material within it.
Bangham and coworkers discovered for the first time, that phospholipids have an inherent quality to form
vesicles when they are put in water. They tend to aggregate and form unilamellar or multilamellar
vesicles. These vesicles have donned numerous avatars and have been given various names for each of
2, 3, 4
their modified versions. Vesicles are spherical, closed bilayer shells typically composed of lipids
(phosphatidylcholines5, phosphatidylglycerols5 and cholesterol6) and/or surfactants forming a solvent core
and separating that core from the surrounding solvent.

The vesicular systems such as liposomes7, niosomes8, sphingosomes9, transferosomes10 and


pharmacosomes11 are used to improve the therapeutic index of both, existing and new drug molecules, by
encapsulating an active medicament inside vesicular structure in one such system. Such delivery systems
prolong the existence of the drug in systemic circulation and finally reduce toxicity. (12, 13)

Vaizoglu and Speiser, in 1986, first used the word ‘pharmacosomes’ to describe the amphiphilic
phospholipid complexes prepared from drug-lipid conjugates with or without additional surfactants.14,15

Drug (pharmakon) and carrier (soma) combine together to form a complex known as pharmacosome.
Many drug delivery systems combine a drug and a complex, but the salient features of pharmacosomes
are that they are amphiphilic phospholipid complexes of drugs bearing active hydrogen that binds to the
phospholipids. Pharmacosomes may exist as ultrafine vesicular, micellar or hexagonal aggregate systems.
The forms in which they exist are governed by the chemical structure of the drug lipid complex as shown
in Figure 1.

Any drug having an active hydrogen which can be replaced, like carboxylic, hydroxyl, etc. can be
esterified with lipids resulting in amphiphilic compounds, eg. Aspirin, Valproic acid, Nalidixic acid. 14,16, 17
The drugs containing aldehyde, ketones and esters are also important because their enolate ions are the
key reactive intermediates, eg. Acebutolol, Linezolid, Cycloserine. The enolate ions are Bronsted bases
and they react with Bronsted acids, with the replacement of the more acidic alpha-hydrogen. These
reactions can be exemplified by the Claisen and Aldol reactions, which are not just confined to the
laboratory but are also important in the biological world. Further, the carbanion like nature of many
medicinal agents, aids in the making of the covalent bond needed for the formation of pharmacosomes.
Fig 1. Diagrammatic representation of various forms of pharmacosomes

Advantages13, 16, 18, 19, 20, 21

 Entrapment efficiency is not only high but predetermined, because the drug - phospholipid complex
itself forms vesicles.
 Unlike liposomes, there is no need of following the tedious, time-consuming step for removing the
free, unentrapped drug from the formulation.
 Since the drug is covalently linked, loss due to leakage of drug does not take place. However, loss
may occur by hydrolysis.
 Drug incorporation does not pose any challenges.
 Drug-bilayer interactions do not influence entrapment efficiency in case of pharmacosome. These
factors, on the other hand have great influence on entrapment efficiency in the case of liposomes.
 In pharmacosomes, membrane fluidity depends upon the phase transition temperature of the drug
lipid complex but it does not affect release rate since the drug is covalently bound.
 The drug is released from pharmacosome by hydrolysis (including enzymatic).
 Phospholipid transfer/exchange is reduced, and solublization by HDL (High density lipoprotein) is
low.
 The physicochemical stability of the pharmacosome depends upon the physicochemical properties of
the drug-lipid complex.
 Due to their amphiphilic behavior, such systems allow for a multiple transfer through the lipophilic
membrane system or tissue, through cellular walls by endocytosis and exocytosis, after medication.
 Following absorption, their degradation velocity into active drug molecules depends to a great extent
on the size and functional groups of drug molecule, the chain length of the lipids, and the spacers.
These can be varied precisely for optimized in vivo pharmacokinetics.
 They can be given orally, topically and extra-or intra-vascularly.

Limitations 16

 Pharmacosomes undergoes fusion, aggregation and chemical hydrolysis on storage.


 Synthesis of a drug lipid complex depends upon its amphiphilic nature.
 Bonding of lipids and drugs requires surface and bulk interactions.
 Covalent bonding is required to protect the leakage of drugs.
 Loss of drug may occur due to chemical hydrolysis.

Comparison of different drug delivery systems

Interest in the broad areas of vesicular drug delivery has increased in recent years due to an increased
understanding of enhanced pharmacokinetic and pharmacodynamic parameters as compared to
conventional system. Special characteristic vesicles like liposomes, transfersomes, niosomes etc may be
potential delivery systems for different drug molecules. Comparison of these delivery systems is given in
Table 1.

Table 1. Comparison of different drug delivery systems


Vesicle system Description Merits Demerits
Niosomes Niosomes (non-ionic Shape, size, composition, Tedious method of
surfactant vesicles) fluidity can be controlled. preparation.
are microscopic Wide variety of drugs can be Unstable.
vesicles consisting of encapsulated.
an aqueous core Sustained release of drugs is
enclosed by a possible.
membrane of non-
ionic surfactants that
form closed bilayer
structures based on
their amphiphilic
nature. 22, 23
Liposomes Liposomes are lipid Delivery of hydrophobic Expensive.
and microscopic hydrophilic and amphipathic, Degradation by
vesicles that fully drugs possible. oxidation.
enclose an aqueous Capable of encapsulating small Lack of purity of natural
volume composed of molecules and also phospholipids as
phospholipids and macromolecules. excipients.
cholesterol. 24, 25 Leakage of drugs
possible.
Less stable.

Transferosomes Transferosomes are Can deform and pass through Expensive


specially optimized, narrow constriction. Chemically unstable
ultradeformable lipid High entrapment efficiency. because of oxidative
vesicles, capable of High elasticity. Degradation.
penetrating easily into Lack of purity of natural
mammalian skin phospholipids as
intact.26, 27 excipients.

Pharmacosomes Pharmacosomes are High Entrapment efficiency. Fusion, aggregation and


No Leakage of drug like
amphiphilic chemical hydrolysis on
liposomes.
phospholipid storage.
Easily Drug incorporation.
complexes prepared
from drug-lipid
conjugates with or
without additional
surfactants.14,15

Formulation aspects of pharmacosomes

The basic components of pharmacosomes are a lipid and a drug.

Drug: Any drug possessing an active hydrogen atom (-COOH, -OH, -NH 2 etc.) can be esterified with the
lipid, with or without spacer chain, leading to amphiphilic complexes. Synthesis of such a compound
may be guided in such a way that it results into a strongly amphiphilic compound (Figure 2), which will
facilitate membrane, tissue, or cell wall transfer, in the organism. 28 The approach has successfully
improved the therapeutic efficacy of a number of drugs, eg. acyclovir 29, pindolol maleate30, bupranolol
hydrochloride30, 3', 5'-dioctanoyl-5-fluoro-2'-deoxyuridine, etc. 31

H + = H
Drug with Phospholipid Bonding of drug
activated
and phospholipid
hydrogen
Fig 2. Drug-phospholipid complex

Lipids: Lipid or lecithin or phosphatidylcholine is the principal molecular building block of cell
membranes. Phospholipids (such as Phosphatidylcholine, Phosphatidylethanolamine,
Phosphatidylinositol, Diphosphatidylglycerol) are the hydrophobic or electrostatic zwitter ionic
molecules, which upon complexation with the drug, yield amphiphilic products which render
phospholipids water soluble and the drug lipid soluble.(19, 28)

Method of preparation

Conventional method

a. Solvent evaporation or hand shaking method: In this method, drug and phospholipid complex are
(32) (33)
dissolved in a volatile organic solvent (e.g. dichloromethane , ethanol ) in a round bottom flask.
The organic solvent is evaporated at 40°C, using a rotary evaporator, which leaves a thin layer of
solid mixture deposited on the wall of the flask. The dried residue is hydrated with aqueous medium
and forms the typical pharmacosomes,34e.g. silybin–phospholipid complex are prepared by solvent
evaporation method as shown in Table 2.

b. Ether injection: Ether injection technique involves an organic solution of the drug-phospholipid
complex which is injected slowly into the hot hydrating solution, leading to formation of
pharmacosomes.34As the drug lipid solution is injected slowly into aqueous phase, the differences in
temperature between the phases causes rapid vaporization of ether, resulting in the spontaneous
formation of pharmacosomes.35

Novel Methods

a. Super critical fluid (SCF) process: Supercritical antisolvent precipitation (SAS)36 is one of the
promising techniques that has been used to produce micron and submicron sized vesicles. Control of
particle size and its distribution is also possible through this process. Two different SCF technologies,
gas antisolvent (GAS) and solution-enhanced dispersion by supercritical fluid (SEDS), are the most
commonly used techniques (Figure 3).

GAS process involves mass transfer by the mechanism of convection and molecular diffusion, based on
supersaturation of liquids for many solutes. Due to obscurity in controlling slow expansion in the GAS
process, it is impossible to achieve high supersaturation levels in the GAS because of the faster process of
nucleation. This process involves highly turbulent flow of solvent and CO 2, which leads to a very fast
mixing or dispersion. It is possible to control the size, shape and morphology of the material of interest
using this technique.37

Li et al., (2008) compared the physicochemical characteristics of the phospholipid complex of puerarin
prepared by traditional methods (solvent evaporation, freeze-drying and micronization) and a
supercritical fluid (SCF) technology and concluded that supercritical fluid technology for the preparation of
puerarin and its phospholipids complex have significant advantages over the solvent evaporation technique
and other conventional methods.38
Super critical fluid (SCF) process

Gas antisolvent Solution-enhanced dispersion


(GAS) by supercritical fluid (SEDS)

Mass transfer through convection & Premixing of fresh liquid


molecular diffusion mechanism solution & supercritical CO2

Homogeneous Supersaturation in nozzle


supersaturated solution mixing chamber

Washing, filteration, product collection & storage


Fig 3. Super critical fluid (SCF) process.

b. Anhydrous co-solvent lyophilisation method: In this method drug and phospholipid complex is
dissolved in organic solvent (like dimethyl sulfoxide) containing glacial acetic acid or other suitable
solvent followed by gentle agitation to form a clear mixture. The resulting homogeneous solution is
then freeze-dried overnight at suitable temperature and vacuum. Insulin-phospholipid complex has
been prepared by utilizing this technique.39 (Table 2)

c. THF injection: A measured amount of drug and phospholipid are dissolved in tetrahydrofuran
contained in a round-bottom flask. Solvent is then evaporated under vacuum at 40°C and the dried
residue is gathered and placed in desiccators overnight, crushed and finally sieved. The resultant
drug–phospholipid complex is transferred into a glass bottle, flushed with nitrogen and stored at room
temperature. Oxymetrine and Didanosine have been reported to be complexed with phospholipid by
THF injection method.40, 41

d. Thin-layer ultrasonication technique: Zhang et al., (2001) prepared 3', 5'-dioctanoyl-5-fluoro-2'-


deoxyuridine pharmacosomes by thin-layer ultrasonication technique. The effects of drug
phosphatidycholine ratio, pluronic F-68 concentration (%, w/v) and glycerol tristearate (%, w/v)
concentration on the mean particle size, entrapment ratio and drug loading were investigated. 42
(Details)

The preparation procedure was as follows. DO-FUdR, lecithin and GTS were dissolved in
dichloromethane and concentrated under vacuum to a constant weight. A thin even lipid film was
formed by rotary evaporation using a round-bottomed flask and vacuum-desiccated at room
temperature overnight to remove residual traces of organic solvents. An appropriate amount of
aqueous solution of Pluronic F-68 was added into the flask and the mixture was sonicated using a
Sonifier Cell Disruptor equipped with a microprobe for 3 min. The DO-FUdR-SLN were obtained
and stored at 4°C.

e. Alternative method: An alternative approach for producing pharmacosomes has been reported in
which hydrophobic drug Adriamycin has been used to synthesize biodegradable micelle forming drug
conjunct with polymers (polyoxyethylene glycol and polyaspartic acid).The advantage of this method
is that the drug may probably not precipitate out, because of the water solubility of the monomeric
drug conjunct.43 (Details)

f. Modified techniques: Muller-Goymann and Hamann produced fenoprofen pharmacosomes using a


modified technique that involved diluting lyotropic liquid crystals of amphiphilic drugs.44 (Details)

Table 2. An overview of different methods of preparation of pharmacosomes with respective


drugs.
Method used for Preparation of Drug(s) References
pharmacosomes incorporated
33
Solvent evaporation or hand shaking method Silybin
35
Ether injection No drug (blank)
38
Super critical fluid process Puerarin
39
Anhydrous co-solvent lyophilisation method Insulin
40
THF injection Oxymatrine
41
Didanosine
42
Thin layer ultrasonication technique 3', 5'-Dioctanoyl-5-
fluoro-2'-
deoxyuridine
43
Alternative method Adriamycin
44
Modified techniques Fenoprofen
Evaluation parameter

There are various techniques which can be used for evaluation of the complex such as 1H rotating frame
overhause effect spectroscopy/two dimensional rotating frame overhause effect spectroscopy (1H/2D
ROESY) nuclear magnetic resonance (NMR) for the structural characterization in organic solvents,
Fourier transform infrared spectroscopy (FT-IR) and X-ray diffraction for the structural characterization
in the solid state and photon correlation spectroscopy (PCS), phosphorus-31 NMR spectroscopy (31P
NMR) and magic-angle spinning nuclear magnetic resonance/ two-dimensional nuclear overhauser effect
spectroscopy (MAS 1H/2D NOESY NMR) for the structural characterization in aqueous media following
hydration. DSC, surface morphology, solubility studies and drug content are the other important
parameters which give potential information regarding drug-lipid complex.

Formation of complex

a. Differential Scanning Calorimetry (DSC):Differential scanning calorimetry (DSC) has been proven
to be a valuable tool for studying the interaction of bioactive compounds with model lipid bilayers. 45
DSC measures thermal changes on the lipid bilayers and has been extensively used in studies of the
molecular interactions of additives with model lipid bilayers.46, 4748

The differential scanning calorimetry can measure the temperature and energy variation involved in
the phase transition, which reflects the degree of crystallinity and stability of the solid stage of
pharmaceutical compounds. The peak size and shape of the DSC curves are useful in determining
crytallinity of the drug and the carrier.When the temperature is increased, phospholipids melt and
drug is dissolved in phospholipids and partly formed phospholipids complexes, which can be
explained through the theory of preparation by melt-out method. 32, 33
b. Photon Correlation Spectroscopy: Photon correlation spectroscopy measurements are performed to
determine the mean particle size (z-Average: an intensity weighted parameter of the diameter of the
particle) and the polydispersity index (PDI value between 0 and 1 where large values resemble broad
distributions) of the complexation product.
c. FTIR spectroscopy: The formation of the complex can be confirmed by IR spectroscopy comparing
the spectrum of the complex with the spectrum of individual components and their physical mixture
(dry 1:1 molar ratio mixture of drug and lipid has been used as the physical mixture). Stability of
pharmacosome can be evaluated by comparing the spectrum of the complex in solid form with the
spectrum of its micro dispersion in water after lyophilization, at different time intervals.21

d. NMR: Nuclear magnetic resonance is used for mobility of molecules inside the solid lipid
nanoparticles. Morel et al., carried out NMR relaxometric investigations of solid lipid nanoparticles.
(49)
Lasonder et al. studied the interactions of vesicles of dipalmitoylphosphatidylcholine with several
anti-inflammatory drugs by Differential Scanning Calorimetry and by the measurement of Nuclear
Overhauser Effects (NOEs), using 1D- and 2D-NMR methods. 2D-NOE spectra for most of the guest
compounds show intermolecular interactions indicating that at least part of the molecules interacting
with the vesicle walls are included in the alkyl chain area of the bilayer, even in case of the inactive
sulindac sulfone.50

ROESY (Rotational Nuclear Overhauser Effect Spectroscopy) is used for structural characterization
in organic solvents by confirming the presence of cation-π interaction between drug and
phospholipids.

NOESY (nuclear overhauser effect spectroscopy), in combination with MAS–NMR has been shown
to be a valuable tool for lipid–lipid and drug–lipid interactions.51The incorporation of drugs within the
lipid bilayer causes chemical shift changes in the lipid peaks and NOESY cross peaks between drug
and lipid resonances.52

Solid-state 31P NMR has been applied to investigate the potential effect of the drug on the lipid bilayer
phase. The resulting spectrum reveals the shape of the chemical shift tensor directly, which is a probe
for the lipid phase.53

Hüsch et al. prepared and studied the structural elucidation of so-called NSAID–PL (diclofenac
sodium, ibuprofen, piroxicam and dipalmitoylphosphatidylcholine) complexes using NMR together
with other techniques applied at different stages of complex preparation.52

Surface Morphology

a. SEM & TEM: Scanning electron microscopy (SEM) or transmission electron microscopy (TEM) are
effective tools to study the surface morphology of pharmacosomes. SEM images the sample surface
by scanning it with electron beams in a raster scan pattern. The electrons interact with the sample
atoms producing signals that contain information about the sample's surface topography, composition
and other properties such as electrical conductivity. Because the electrons in a TEM pass through the
sample, it is commonly used to look at the internal structure of the sample, while SEM looks at the
surface (or the near-surface).54, 55The shape and size of the pharmacosome strongly depends on factors
like purity grades of phospholipid and the process variables such as speed of rotation, vacuum applied
and the method used.

b. AFM: Atomic force microscope (AFM) as a novel powerful technique with nano-meter scale
resolution has been developed for different applications since Binnig et al., (1986) invented it. The
measurement can be done at atmospheric pressure and it is not necessary to pre-treat the specimen,
compared with SEM where the specimen has to be gold-coated. 56, 57 Ping et al., (2005) utilized TEM
and AFM techniques for study of configuration and particle size of didanosine pharmacosomes.41

Solubility studies
Solubility studies are done by ultraviolet spectroscopy and HPLC method to determine the concentration
of drug. Kuntal et al. carried solubility analysis of curcumin, curcumin–phospholipid complex and
physical mixture of curcumin and phospholipid by HPLC method.58

Bioavailability enhancement can be achieved by improving drug liposolubility and also improved drug
entrapment efficiency. Improving the liposolubility of water-soluble drugs is an attractive way to enhance
their loading efficiency as far as hydrophobic carriers are concerned. Moreover, it would be very helpful
to increase their uptake through mucous membranes by hydrophobic modification of hydrophilic
peptides.39

Drug content
It can be determined by ultra violet spectrophotometer with suitable dilutions. High percentage of drug
content is correlated with high percentage of drug loading. Complexation of drug and lipid is responsible
for high loading.59

State of phospholipid complex


X-Ray Powder Diffraction technique is most commonly used to investigate the changes in the crystal
morphology due to a polymorphic transition, and to study the solid state. The degree of crystallinity can
be evaluated using relative integrated intensity of reflection peaks in the given range of reflecting angle,
2θ.38

It has been observed that crystalline diffraction peaks of drug disappear in XRPD of the complex, which
in turn confirms the formation of phospholipid complex. The chemical bonding between drug and the
phospholipids, responsible for the development of pharmacosomes, result into significant changes in
XRPD.60

Potential in Targeting

Pharmacosomes are characterized as drug lipid complexes with high degree of selectivity for the target
cells and capable of delivery of various biologically active substances. Basically, three components are
involved in targeting. The first one is the effector; the substance effecting the necessary action on the
target cells, second is the discriminating agent; which discriminates between the target cell and other cells
(e.g. antibodies to the antigen on the cell surface, tissue specific factors, lectins etc.) and the third is the
penetrating factor; securing the penetration of the effectors into the cells. Increase in discriminating agent
also increases the penetration of effector into the target cells. 61Pharmacosomes, for targeted drug delivery,
have been used for a variety of applications which have been discussed below.

Brain targeting: Pharmacosomes are lipid-based drug delivery systems which have been found to
improve absorption due to lipophilic character and are required for brain targeting.Zhang et a.l prepared
3', 5'-dioctanoyl-5-fluoro-2'-deoxyuridine pharmacosomes (DO-FUdR-PS) which showed high
concentration in tested organs. The brain AUC ratio of DO-FUdR-PS to FUdR was 10.97 and showed a
good targeting efficiency in vivo. Pharmacosomes can improve the ability of drug to cross blood-brain
barrier and are a promising drug carrier for the treatment of central nervous system disorders.31

Tumor targeting: Malignant tumor has been a frequently encountered disease to threaten human health.
Pharmacokinetics and tissue distribution of glycerol-skeleton lipid prodrug pharmacosomes have been
studied after single bolus of i.v. administration to normal Kunming mice.1-Octadecyl-2-fluorouracil-N-
acetyl-3-zidovudine phosphoryl glycerol (OFZPG) pharmacosomes were eliminated quickly from blood
circulation to the major tissues, such as liver and lung. The distribution t 1/2 and the elimination t1/2 were
found to be 5.16 min and 105.13 min, respectively. Antitumor activity of OFZPG pharmacosomes was
investigated, which demonstrated the IC50 in the three human colon cancer cells (COLO205 、 HT-
29、HCT-116) to be 4.6, 12.2 and 12.7μmol/L, respectively, which was better than that of 5-Fluorouracil.
The results indicated that the prodrug could degrade to the active compound efficiently, which could play
an important role in pharmacodynamics of the drug. The results showed that the high dose of OFZPG
pharmacosomes (15 mg/kg) had significant inhibition to tumor, and the tumor inhibition ratio was
41.47%, at a dose lower than 1/10th (mol/mol ) of 5-FU, and with comparative therapeutic effect to
that of 5-FU (tumor inhibition ratio : 59.91%). Hence it is exhibited that OFZPG pharmacosomes may
play an important role in antitumor activity.62

Liver targeting: Ping et al., 2005 reported that Didanosine (ddI) pharmacosomes are concentrated in
liver quickly after intravenous administration, and some in lung and spleen. Its elimination from target
tissues was slow, t1/2 of 5′-cholesterylsuccinyl-didanosine (CS-ddI) in liver was 10 days and these
pharmacosomes showed apparent liver targeting and sustained-release effect in the target tissues.41

Internalization63, 64, 65

Cells internalize materials from their surroundings by phagocytosis or pinocytosis.Particles are


internalized and endocytosed, a process in which a small region of the plasma membrane invaginates to
form a new intracellular membrane-limited vesicle of about 0.05 to 0.1 μm in diameter. Phagocytosis and
pinocytosis are the common mechanism of internalization of nanoparticles or colloidal vesicular particles
like pharmacosomes. In receptor-mediated endocytosis, a specific receptor on the cell surface binds
tightly to the extracellular macromolecule (the ligand) that it recognizes; the plasma-membrane region
containing the receptor-ligand complex then undergoes endocytosis, becoming a transport vesicle.
Receptor-ligand complexes are selectively incorporated into the intracellular transport vesicles; most
other plasma-membrane proteins are excluded (Line is not clear). Moreover, the rate at which a ligand is
internalized is limited by the amount of its corresponding receptor on the cell surface.

Small invaginations of the plasma membrane, termed caveolae, due to their shape which is like a small
cave lined with the membrane protein caveolin, contain some receptor proteins and are used for certain
types of receptor-mediated endocytosis. However, receptor-mediated endocytosis generally occurs via
clathrin-coated pits and vesicles. Clathrin is a protein which coats the vesicle after it gains entry into the
invagination of the cell membrane and begins to ready it, for its journey into the cell, by covering it with
clathrin triskeleton. This rigid structure later disassembles, after it has finished its job of internalization.
Internalization could also be a combination of the mechanisms, clathrin as well as caveole mediated.
Conversely, a clathrin and caveole independent mechanism may also occur (Fig 4).
Most cell-surface receptors that undergo endocytosis will repeatedly deposit their ligands within the cell
and then recycle to the plasma membrane, once again to mediate the internalization of ligand molecules,
thus finishing their round-trip and getting ready for the next. Like the very acidic lysosomes, with an
internal pH of 4.5– 5.0, clathrin-coated vesicles and endosomes contain a V-class ATP-dependent proton
pump. These vesicles also contain a Cl− channel, allowing the proton pump to generate a significant H+
concentration gradient, rather than the transmembrane electric potential that would form if only protons
were transferred from the cytosol to the vesicle lumen. The late endosome is the first vesicle encountered
by receptor-ligand complexes with a low pH and hence is the organelle in which these and most other
receptors dissociate from their tightly bound ligands.

Beginning 15 minutes after internalization, ligands are transferred to lysosomes, but the intact receptors
themselves usually are not found in these organelles. Instead, the receptor-rich vesicles that bud from the
late endosomes mediate the recycling of receptors back to the cell surface. The spherical part of the late
endosome eventually buds off from the transport vesicles, and the transport vesicles with their cargo of
ligands, soon fuse with lysosomes. And thus ends the journey of the pharmacosomes, to release their
payload into the cell.
Fig 4: Process of internalization for pharmacosomes.

(Draw clear figure, no scanned figure is allowed)

Applications of pharmacosomes
Oral Delivery: Peroral drug administration is one of the most convenient routes for drug delivery, but the
major problems encountered include poor absorption of poorly soluble drugs through mucosal membrane
and drug instability in the gastrointestinal (GI) tract. Many approaches have been attempted to
circumvent these problems, including the employment of microparticulate carriers such as nanoparticles
or microemulsions.66Complex formation is one of the very popular ways to enhance the oral
bioavailability of drugs as shown in Table 3.

Table 3. Oral delivery of various drugs through pharmacosomes.


Drug Effect on oral bioavailability after incorporation in pharmacosomes Reference
Incorporated
33
Silybin The bioavailability of silybin in rats was increased remarkably after oral
administration of silybin–phospholipid complex comparing to silybin-N-
methylglucamine.
39
Insulin In-vivo evaluation shows that Insulin–Soybean phosphatidylcholine
complex loaded nanoparticles are able to improve the intestinal
absorption of insulin.
67
Silybin Complexation with phosphatidylcholine in IdB 1016 (complex of
silybin) greatly increases the oral bioavailability of silybin.

Bioavailability Enhancement: Pharmacosomes impart better biopharmaceutical properties to the drug,


resulting in improved bioavailability. Role of pharmacosomes to increase the bioavailability has been
reported by authors for the various types of drugs like non-steroidal anti-inflammatory drugs, proteins,
cardiovascular and antineoplastic drugs as shown in Table 4.

Table 4. Some applications of pharmacosomes in bioavailability enhancement.


Drug Effect after incorporation in References
pharmacosomes
32
Aspirin Water solubility was found to be 15.98 µg/ml
for developed aspirin-PC complex and 8.45
µg/ml for aspirin alone.
33
Silybin Solubility of silybin–phospholipid complex in
water and in n-octanol was effectively
enhanced.
38
Puerarin Remarkable enhancement of its solubility and
in vitro dissolution rate was observed. The
best dissolution rate was obtained by SEDS-
prepared complex, while the highest
solubility was obtained for solvent
evaporation method.
40
Oxymatrine The solubility of oxymatrine –phospholipid
complex in n-octanol was effectively
enhanced.
58
Curcumin Curcumin–phospholipid complex has much
higher solubility in water or n- octanol than
curcumin or physical mixture of curcumin
and phospholipid
59
Diclofenac Water solubility of the prepared complex was
found to be 22.1 mg ml –1 as compared to 10.5
mg ml–1 of diclofenac.
60
Acelofenac Aceclofenac phospholipid complex showed
better solubility and dissolution profile.
67
Silybin Complexation with phosphatidylcholine in
IdB 1016 (complex of silybin) greatly
increases the oral bioavailability of silybin.
68
Ibuprofen The water solubility of ibuprofen
pharmacosomes was found to be much better
than free ibuprofen.
69
Geniposide Geniposide pharmacosomes significantly
increased the lipophilicity of drug, and P
value of pharmacosomes containing genipin
in n-octanol and water was about 20
multiples more than that of geniposide.
70
Propranolol A significantly higher bioavailability of
Palmitoyl propranolol was recorded as
compared to the plain drug
71
Naringenin Water solubility of naringenin improved from
43.83 to 79.31 μg/mL in the complex form.
72
Gallic Acid A controlled release pattern was shown by the
complex which showed continuous release up
to 93% of gallic acid at the end of 24 h in
comparison to free gallic acid which showed
81.91% burst release just in the 0.5 h.

Ocular delivery: The amphiphilic prodrug was converted to pharmacosomes on dilution with water. The
pharmacosomes showed greater shelf stability, facilitated transport across the cornea, and a controlled
release profile.73 Taskintunaet al. prepared and stated that acyclovir diphosphatidylmyristoylglycerol had
prolonged antiviral activity against herpes simplex virus-1 retinitis in a rabbit model. This novel lipid
prodrug improved optical clarity and allowed long-acting intra-vitreal treatment of cytomegalovirus
retinitis and other retinal diseases.29

Antiviral activity: The structural modification of drugs is necessary to obtain amphiphilicity. Some
lipophilic derivatives of nucleosides self-assemble in organic solvents based on hydrogen bonding and
some phospholipid nucleoside conjugates form ordered aggregates in aqueous solutions. Jin et al.
prepared amphiphilic conjugates of acyclovir with lipid into self-assembled nanoparticles (SAN) and
showed site-specific distribution in-vivo. It can be predicted that self-assembling amphiphilic prodrugs
system shall be useful for anti-viral, anti-cancer and gene therapy.15

Cosmetics: In the area of cosmoceuticals, the complexes of different vegetal derivatives such as
flavonoids, terpenes and saponins showed anti-inflammatory and vasokinetic properties higher than those
observed after administration of the same amount of these substances in free form.Phospholipids exhibit
a marked affinity for some classes of flavonoids. The favorable results (Table 5) obtained with new
complexes highlights the importance of the in-vitro reconstitution of complexes between natural products
and phospholipids which, in all likelihood, take place in the cytoplasm of living cells in order to modify
tissue reactivity.74, 75

Table 5. Applications of complexes between phospholipid and some natural substances in


cosmoceuticals.
Phospholipid-Polyphenol Activity Effect after complex formation
Complexes
Phospholipid-glycyrrhetinic acid Anti-inflammatory activity in Larger and longer lasting activity
complex animals on croton oil-induced of the complexes compared with
dermatitis. the same compounds in their free
forms.
Silymarin-DSPC Anti-inflammatory activity by Silymarin DSPC complex
(Disteroylphosphatidylcholine) polymorphonuclear leucocyte showed an inhibitory activity of
complex aggregation at the site of about 35% both at 12 and at 24 h,
inflammation. while silymarin alone was devoid
of effect.
18- [Beta] -glycyrrhetinic acid Anti-edematous activity. 80% reduction of the edema.
DSPC complex
Ginkgo biloba-DF complex Microcirculation. Improved microcirculation of the
skin

Future perspectives

It has been found that drug solubility is the most essential feature in formulation development and also in
therapeutic efficacy. Vesicles are found to be vehicles of choice to improve the bioavailability of a drug.
Existing vesicular drug delivery systems can be modified to fulfill the requirement of clinical need.
Pharmacosomes offer an efficient way for delivery of various drugs (NSAIDs, proteins, cardiovascular,
chemotherapeutic agents and antineoplastic agent). In this context, more clinical studies are necessary to
provide further information and insight into the different drug delivery systems. The system yet requires
greater efforts to investigate the mechanism of action and to overcome the limitations like chemical
hydrolysis, fusion, loss of drug, in order to exploit the advantages of this system to the fullest.

References
1. Zasadzinski, J. A. (1997). Novel approaches to lipid based drug delivery, Current Opinion in
Solid State & Materials Science, 2, 346-349.
2. Bangham, A.D. (1974). Preparation and use of liposomes as models of biological membranes,
Methods in membrane biology, 1, 1-68
3. Bangham, A. D., Standish, M. M., Watkins, J. C. (1965). Diffusion of univalent ions across the
lamellae of swollen phospholipids, Journal of Molecular Biology, 13, 238–252.
4. Bangham, A. D., Standish, M. M., Weissmann, G. (1965). The action of steroids and streptolysin
S on the permeability of phospholipid structures to cations, Journal of Molecular Biology, 13,
253–259.
5. Lentz, B. R., Alford, D. R., Hoechli, M., Dombrose, F. A. (1982). Phase behavior of mixed
phosphatidylglycerol/phosphatidylcholinemultilamellar and unilamellar vesicles, Biochemistry,
21, 4212-9.
6. Ahrendt, S. A., Fox-Talbot, M. K., Kaufman, H. S., Lillemoe, K. D., Lipsett, P. A., Pitt, H. A.,
Surg A. (1994). Characterization of a small vesicular cholesterol carrier in human gallbladder bile,
Annals of Surgery, 220, 635–643.
7. Torchilin, V. P. (2005). Recent Advances with liposomes as pharmaceutical carriers, Nature
Review. Drug Discovery, 4,145-60.
8. Rajera, R., Nagpal, K., Singh, S. K., Mishra, D. N. (2011). Niosomes: a controlled and novel drug
delivery system, Biological and Pharmaceutical Bulletin, 34, 945-53.
9. Bansal, S., Kashyap, C. P., Aggarwal, G., Harikumar, S. (2012). A comparative review on
vesicular drug delivery system and stability issues, International Journal Of Research in
Pharmacy and Chemistry, 2, 704-713.
10. Jain, S., Jain, P., Umamaheshwari, R., Jain, N. K. (2003). Transfersomes - a novel vesicular
carrier for enhanced transdermal delivery: development, characterization, and performance
evaluation, Drug Development and Industrial Pharmacy, 29, 1013-26.
11. Semalty, A., Semalty, M., Rawat, B. S., Singh, D., Rawat, M. S. (2009). Pharmacosomes: the
lipid-based new drug delivery system, Expert Opinion on Drug Delivery, 6, 599-612.
12. Kumar, R., Kumar, S., Jha, S. S., Jha, A. K. (2011). vesicular system-carrier for drug delivery,
Der Pharmacia Sinica, 2, 192-202.
13. Biju, S. S., Talegaonkar, S., Mishra, P. R., Khar, R. K. (2006). Vesicular systems: An overview,
Indian Journal of Pharmaceutical Sciences, 68, 141-153.
14. Vaizoglu, M. O., Speiser, P. P. (1986). Pharmacosomes-a novel drug delivery system,
Acta PharmaceuticaSuecica, 23, 163-172
15. Jin, Y., Tongc, L., Ai, P., Li, M., Hou, X. (2006). Self-assembled drug delivery systems 1.
Properties and in vitro/in vivo behavior of acyclovir self-assembled nanoparticles (SAN),
International Journal of Pharmaceutics, 309, 199–207.
16. Gangwar, M., Singh, R., Goel, R. K., Nath, G. (2012). Recent advances in various emerging
vescicular systems: An overview, Asian Pacific Journal of Tropical Biomedicine, S1176-S1188.
17. Kaur, H., Rana, A.C., Seth, N., Kaur, A. (2012). Pharmacosomes – Targeted Drug Delivery
System, International Journal of Natural Product Science,1, 17.
18. Nilesh. (2007). Pharmacosomes: An Overview, Retrieved April 7, 2013,
from http://www.farmavita.net/content/view/515/84/
19. Balasubramanian, J., Narayanan, N. (2013). Pharmacosomes: A novel method of Colloidal
dispersions in Drug Delivery, Discovery Pharmacy, 4, 3-4.
20. Kavitha, D., Naga, S. J., Shanker, P. (2010). Pharmacosomes: an emerging vesicular system,
International Journal of Pharmaceutical Sciences Review and Research,5, 168-171.
21. Patel, J.L., Bharadia, P.D. (2012). A Review on: Pharmacosomes as a Novel Vesicular Drug
Delivery System, World Journal of Pharmaceutical research, 1, 456-469.
22. Uchegbu, I. J., Vyas, S. P. (1998). Non-ionic surfactant based vesicles (niosomes) in drug
delivery, International Journal of Pharmaceutics,172, 33–70.
23. Pando, D., Gutiérrez, G.,Coca, J., Pazos, C. (2013). Preparation and characterization of niosomes
containing resveratrol, Journal of Food Engineering,117, 227–234.
24. Shipton, E. A. (2012). Advances in delivery systems and routes for local anaesthetics, Trends in
Anaesthesia and Critical Care, 2, 228-233.
25. Elsayed, M. M. A., Abdallah, O. Y., Naggar, V. F., Khalafallah, N. M. (2007). Lipid vesicles for
skin delivery of drugs: reviewing three decades of research, International Journal of
Pharmaceutics, 332, 1–16.
26. Cevc, G. (2004). Lipid vesicles and other colloids as drug carriers on the skin, Advanced Drug
Delivery Reviews, 56, 675 – 711.
27. Kumar, A., Pathak, K., bali, V. (2012). Ultra-adaptable nanovesicular systems: a carrier for
systemic delivery of therapeutic agents, Drug Discovery Today, 17, 21-22.
28. Goyal, T., Rawat, A.S., Chauhan, M. (2012). Pharmacosomes: opening new doors for drug
delivery, International Journal of Pharmacy and Pharmaceutical Sciences, 4, 25-29.
29. Taskintuna, I., Banker, A. S., Flores-Aguilar, M., Bergeron-Lynn, G., Aldern, K. A., Hostetler, K.
Y., Freeman, W. R. (1997). Evaluation of a novel lipid prodrug for intraocular drug delivery:
effect of acyclovir diphosphatedimyristoylglycerol in a rabbit model with herpes simplex virus-1
retinitis. Retina, 17, 57-64.
30. Vaizoglu, O., Speiser, P., (1992). The pharmacosomes drug delivery approach, European Journal
of Pharmaceutics and Biopharmaceutics, 38, 1-6.
31. Zhang, Z. R., Wang, J. X. (2001). Study on brain targeting 3',5'-dioctanoyl-5-fluoro-2'
deoxyuridine pharmacosomes, Yao XueXueBao, 36, 771-6.
32. Semalty, A., Semalty, M.,Singh, D., Rawat, M. S. (2010). Development and characterization of
aspirin-phospholipid complexfor improved drug delivery, International Journal of
Pharmaceutical Sciences and Nanotechnology, 3, 940-947.
33. Yanyu, X., Yunmei, S., Zhipeng, C., Qineng, P. (2006). The preparation of silybin–phospholipid
complex and the study on its pharmacokinetics in rats, International Journal of Pharmaceutics,
307, 77–82.
34. Gupta, S., Singh, R. P., Lokwani, P., Yadav, S., Gupta, S. K. (2011). Vesicular System As Targeted
Drug Delivery System: An Overview, International Journal of Pharmacy & Technology, 3, 987-
1021.
35. Srinivas, S., Kumar, Y. A., Hemanth, A., Anitha, M. (2010). Preparation and Evaluation of
Niosomes containing Aceclofenac, Digest Journal of Nanomaterials and Biostructures, 5, 249 –
254.
36. Reverchon, E. (1999). Supercritical antisolvent precipitation of micro- and nano-particles, The
Journal of Supercritical Fluids, 15, 1–21.
37. Amaro-González, D., Mabe, G., Zabaloy, M., Brignole, E. A. (2000). Gas antisolvent
crystallization of organic salts from aqueous solution, The Journal of Supercritical Fluids, 17,
249–258.
38. Li, Y., Yang, D., Chen, S.,Chen, S., Chan, A.S. (2007). Comparative physicochemical
characterization of phospholipids complex of puerarin formulated by conventional and
supercritical methods, Pharmaceutical Research, 25, 563-77.
39. Cui, F., Shi, K., Zhang, L., Tao, A., Kawashima, Y. (2006). Biodegradable nanoparticles loaded
with insulin–phospholipid complex for oral delivery: Preparation, in-vitro characterization and in
vivo evaluation, Journal of Controlled Release, 114, 242–250.
40. Yue, P-F., Yuan, H-L., Yang, M., Zhu, W-F. (2009). Preparation, Characterization and
Pharmacokinetics in-vivo of Oxymatrine–Phospholipid Complex, Journal of Bioequivalence &
Bioavailability, 1, 99-102.
41. Ai, P., Jin, Y., Chen, D. (2005). Preparation and in-vivo behavior of didanosine pharmacosomes in
rats, Chinese Journal of Pharmaceutics, 3, 227-235.
42. Zhang, Z. R., Wang, J. X. (2001). Study on brain targeting 3',5'-dioctanoyl-5-fluoro-2'
deoxyuridine pharmacosomes, Yao XueXueBao, 36, 771-6.
43. Lawrence, M. J. (1994). Surfactant Systems: Their Use in Drug Delivery, Chemical Society
Reviews, 23, 417–424.
44. Muller-Goymann, C. C., Hamann, H. J., (1991). Pharmacosomes: multilamellar vesicles
consisting of pure drug, European Journal of Pharmrmaceutics and Biopharmaceutics, 37, 113–
117.
45. Mavromoustakos, T., Theodoropoulou, E., Papahatjis, D., Kourouli, T., De-Ping, Y., Trumbore,
M., Makriyannis, A., (1996). Studies on the thermotropic effects of cannabinoids on
phosphatidylcholine bilayers using differential scanning calorimetry and small angle-X-
diffraction. BiochimicaetBiophysica Acta, 1281, 235–244.
46. Hata, T., Matsuki, H., Kaneshina, S., (2000). Effect of local anesthetics on the bilayer membrane
of dipalmitoylphospatidylcholine: interdigitation of lipid bilayer and vesicle-micelle transition.
Biophysical Chemistry, 87, 25–36.
47. Ambrosini, A., Bossi, G., Dante, S., Dubini, B., Gobbi, L., Leone, L., Bossi, M.G.P., Zolese, G.,
(1998). Lipid–drug interactions: thermodynamic and structural effects of antimicotic fluconazole
on DPPC liposomes,Chemistry and Physics of Lipids, 95, 37–47.
48. Gardikis, K., Hatziantoniou, S., Viras, K., Wagner, M., Demetzos, C. (2006). A DSC and Raman
spectroscopy study on the effect of PAMAM dendrimer on DPPC model lipid membranes,
International Journal of Pharmaceutics, 318, 118–123.
49. Morel, S., Terreno, E., Ugazio, E., Aime, S., Gasco, M.R. (1998). NMR relaxometric
investigations of solid lipid nanoparticles (SLN) containing gadolinium (III) complexes.
European Journal of Pharmaceutics and Biopharmaceutics, 45, 157-163.
50. Lasonder, E., Weringa, W.D. (1990). An NMR and DSC study of the interaction of phospholipid
vesicles with some anti-inflammatory agents, Journal of Colloid and Interface Science, 139, 469-
478.
51. Huster, D., Arnold, K., Gawrisch, K., (1999). Investigation of lipid organization in biological
membranes by two-dimensional nuclear overhauser enhancement spectroscopy. Journal of
Physical Chemistry B, 103, 243–251.
52. Husch, J., Dutagaci, B., Glaubitz, C., Geppert, T., Schneider, G., Harms, M., Müller-Goymann, C.
C., Fink, L., Schmidt, M. U., Setzer, C., Zirkel, J., Rebmann, H., Schubert-Zsilavecz, M., Abdel-
Tawab, M. (2011), Structural properties of so-called NSAID–phospholipid-complexes, European
Journal of Pharmaceutical Sciences 44, 103–116.
53. Jurgen, S., Matthias, M., Beate, F., Klaus, A., Daniel, H. (2007). 31P NMR spectroscopy of
phospholipids: from micelles to membranes. Current Analytical Chemistry. 3, 283–301.
54. Zhou, W. L., Wang, Z. L. (2006). Scanning Microscopy for Nanotechnology: Techniques and
Applications. New York, NY: Springer.
55. Liu, F., Wu, J., Chen, K., Xue, D. (2010). Morphology Study by Using Scanning Electron
Microscopy, Microscopy: Science, Technology, Applications and Education, 1781-1792.
56. Yang, H., Wang, Y., Lai, S., An., H, Li., Y, Chen, F. (2007). Application of atomic force
microscopy as a nanotechnology tool in food science, Journal of Food Science,72, 65-75.
57. Binnig, G., Quated, C. F., Gerber, C. H. (1986). Atomic force microscope, Physical Review
letters, 56, 930.
58. Maiti, K., Mukherjee, K., Gantait, A., Saha, B.P., Mukherjee, P.K. (2007). Curcumin–
phospholipid complex: Preparation, therapeutic evaluation and pharmacokinetic study in rats,
International Journal of Pharmaceutics,330, 155–163.
59. Semalty, A., Semalty, M., Singh, D., Rawat, M. S. M. (2009). Development and physicochemical
evaluation of pharmacosomes of diclofenac, ActaPharmaceutica, 59,335–344.
60. Semalty, A., Semalty, M., Rawat,B., Singh, S.D., Rawat, M. S. M. (2010). Development and
Evaluation of Pharmacosomes of Aceclofenac, Indian Journal of Pharmaceutical Sciences, 72,
576–581.
61. Raikhman, L. M., Moshkauskii, Y. S., Piruzyan, L. A. (1978). Pharmacosomes concept-A new
approach to drug preparation, FarmlservticheskiiZhurnal, 12, 18-22.
62. Fang Y, (2011). Study on the Glycerol-Skeleton Antitumor Lipid Prodrug Pharmacosomes,
Master's thesis, Henan University.
63. Goldstein, J.L., Anderson, R. G., Brown, M. S. (1979). Coated pits, coated vesicles, and receptor-
mediated endocytosis, Nature, 279, 679–685.
64. Wileman, T., Harding, C., Stahl, P. (1985). Receptor-mediated endocytosis, Biochem Journal,232,
1–14.
65. Lodish, H., Berk, A., Kaiser, C. A., Krieger, M., Scott, M. P., Bretscher, A., Ploegh, H.,
Matsudaira, P. (2008). Molecular cell biology (6th ed.) W.H. Freeman.
66. Yoo, H.S., Choi, H., Park, T.G. (2000). Protein-Fatty Acid Complex for Enhanced Loading and
Stability within Biodegradable Nanoparticles, Journal of Pharmaceutical Sciences, 90, 194-201.
67. Barzaghi N, Crema F, Gatti G, Pifferi G, Perucca E. (1990). Pharmacokinetic studies on IdB
1016, A silybin- phosphatidylcholine complex, in healthy human subjects, European Journal of
Drug Metabolism and Pharmacokinetics, 15, 333-8.
68. De, A., De, P. K. (2011). Preparation and evaluation of pharmacosomes of ibuprofen, Retrieved
April 10, 2013, from http:// www.scientificipca.org/paper/2011/09/15/201109151734270A.docx
69. Yue,P., ZhengQ., Wu, B., Yang, M., Wang, M., Zhang, H., Hu, P., Wu, Z. (2012). Process
optimization by response surface design and characterization study on geniposide
pharmacosomes, Pharmaceutical Development and Technology, 17, 94-102.
70. Vyas, S.P., Jaitely, V., Kanaujia, P. (1999). Synthesis and characterisation of palymitoylpropanolol
hydrochloride auto-lymphotrophs for oral administration, International Journal of
Pharmaceutics, 186, 177–189.
71. Semalty, A., Semalty, M.,Singh, D., Rawat, M. S. M. (2010). Preparation and characterization of
phospholipid complexes of naringenin for effective drug delivery, Journal of Inclusion
Phenomena and Macrocyclic Chemistry, 67, 253-260.
72. Singh, D., Singh, M., Rawat M., S., Semalty, A., Semalty, M. (2011). Gallic Acid-Phospholipid
Complex: Drug Incorporation and Physicochemical Characterization. Letters in Drug Design &
Discovery, 8, 284-291(8).
73. Kaur, I.P., Kanwar, M. (2002). Ocular Preparations: The Formulation Approach, Drug
Development and Industrial Pharmacy, 28, 473–493.
74. Bombardelli, E. (1991). Phospholipid-polyphenol complexes: a new concept in skin care
ingredients, Cosmetics and Toiletries, 106, 69-76.
75. Bombardelli, E., Curri, S. B., Loggia, D. R., Del, N. P., Tubaro, A., Gariboldi, P. (1989).
Complexes between phospholipids and vegetal derivatives of biological interest, Fitoterapia, 60,
1-9.

You might also like