You are on page 1of 17

J.

of Supercritical Fluids 165 (2020) 104984

Contents lists available at ScienceDirect

The Journal of Supercritical Fluids


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

Sustainable technologies for liposome preparation


Maja Leitgeb a,b,∗ , Željko Knez a,b , Mateja Primožič a
a
University of Maribor, Faculty of Chemistry and Chemical Engineering, Smetanova Ulica 17, Maribor, 2000, Slovenia
b
University of Maribor, Faculty of Medicine, Taborska Ulica 8, Maribor, 2000, Slovenia

h i g h l i g h t s g r a p h i c a l a b s t r a c t

• Sustainable green technologies for


liposome preparation.
• Production of micro- and nano-sized
liposomes with narrow size distribu-
tion.
• Supercritical fluids assisted methods
improve liposome encapsulation effi-
ciency.

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

Article history: Liposome possesses a great number of advantages and therefore they can be used for a variety of appli-
Received 14 April 2020 cations. Among other things, they can serve as useful drug carriers in preclinical and clinical trials.
Received in revised form 19 June 2020 Supercritical fluids assisted technology is an appropriate method for liposome preparation because of its
Accepted 5 July 2020
nontoxicity to the environment enables particle size manipulation and solvent-free production. Thus, the
Available online 11 July 2020
use of supercritical fluids (SCFs) provides advanced technology for liposome preparation and may become
the dominant technology for their preparation in the future. This review discusses the classification of
Keywords:
liposome and gives a short overview of their applications and conventional methods of preparation.
Liposome
Preparation
Emphasis is placed on the use of various supercritical fluids assisted methods for liposome prepara-
Supercritical fluids tion and their advantages and disadvantages. The reader is also updated about recent developments in
Supercritical fluids assisted methods supercritical fluids assisted liposome production technology.
© 2020 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND
license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Classification of liposomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3. Liposome as a drug delivery system . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
3.1. Commercially available formulations of liposome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4
4. Methods of drug loading in liposome preparation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
5. Conventional techniques for liposome preparation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
5.1. Tools for size reduction and uniform size distribution of liposome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6

∗ Corresponding author at: University of Maribor, Faculty of Chemistry and Chemical Engineering, Smetanova ulica 17, Maribor, 2000, Slovenia.
E-mail addresses: maja.leitgeb@um.si (L. Maja), zeljko.knez@um.si (K. Željko), mateja.primozic@um.si (P. Mateja).

https://doi.org/10.1016/j.supflu.2020.104984
0896-8446/© 2020 The Authors. Published by Elsevier B.V. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.
0/).
2 L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984

6. Supercritical fluids (SCFs) methods for liposome preparation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7


6.1. Depressurization of an expanded liquid organic solution-suspension (DELOS) method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
6.2. Supercritical anti-solvent (SAS) method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
6.3. Rapid expansion of supercritical solutions (RESS) method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
6.4. Supercritical assisted liposome formation (SuperLip) method . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
6.5. Other methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
6.6. Batch or continuous SCF processes for liposome preparation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
7. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13

1. Introduction determined using different analytical methods (UV–vis detection,


HPLC, GC, GC–MS etc.) depending on the active compound which
Liposomes are colloidal, vesicular structures, composed of is incorporated in liposomes. When the incorporated substance is
one or more lipid bilayers surrounding an equal numbers protein, direct and indirect methods for EE determination could be
of aqueous compartments [1]. Liposome can be made from used. While in direct methods, the EE of encapsulation is calculated
natural substances and are therefore nontoxic, biocompati- directly from the loaded protein in the nanoparticles, in the indirect
ble, biodegradable and non-immunogenic. Phosphatidylcholine method, the non-entrapped protein or drug is considered [16]. The
(PC), phosphatidylglycerol (PG), dipalmitoylphosphatidylcholine liposome preparation methods can be categorized as conventional
(DPPC), distearoylphosphatidylcholine (DSPC), cholesterol (CH), and novel methods (e.g. supercritical assisted method for liposome
stearylamine, dicetylphosphat, or a mixture of these are usually preparation). There are already some review articles where SCFs
the composites of the bilayer [2]. technologies for liposome preparation are described [17–20], but
Liposome properties are mostly dependent on lipid composi- in this review an emphasis on the impact of individual SCFs tech-
tion, surface charge, size, and the method of preparation [3]. The nology on the size, types of vesicles and EE of prepared liposomes
rigidity and the charge of the bilayer are conditioned by the choice including lipid/active ingredients of liposome preparation prepared
of the bilayer components. Unsaturated PC species from natural by defined SCFs method is done. It briefly explains the classifica-
sources (egg or soybean PC) give highly permeable and less stable tion of liposome and their potential applications, with a focus on
bilayers, while more rigid and impermeable bilayer structures are novel techniques for liposome formation using supercritical fluid
obtained when lipid saturated phospholipids (PLs) with long acyl assisted technologies.
chains (such as DPPC) are used [3–6]. Liposome possesses a great
number of features and therefore they can be used for a variety of 2. Classification of liposomes
applications. The general application of liposome may be attributed
to their use in respiratory and eye disorders, vaccine adjuvants, Liposome formation enables the amphiphilic character of lipids
brain targeting and anti-infective agents, and their clinical appli- which, with the force of hydrophobic interaction, assemble into
cations include cancer treatment, antimicrobial therapy and gene bilayers. Three groups of liposome exist by their morphology, using
therapy [7–9]. conventional classification: multilamellar vesicle (MLV), small
Liposome can serve as carriers for drugs or other macro- unilamellar vesicle (SUV), large unilamellar vesicle (LUV) and mul-
molecules for delivery into human and animal bodies, since they tivesicular vesicle (MVV) [21] (Fig. 2). These different vesicular
simplify site-specific drug delivery, e.g. to tumor tissues [10] and structures can be achieved from the self-assembly of a certain
are therefore of great interest to the pharmaceutical and cosmetic amphiphilic molecule. Because of their size, amphiphilic character,
industries. Liposomes are also attractive for applications in the food and biocompatibility, liposomes have been widely used as biomem-
and farming industries, since they can be used for encapsulation branes as well as delivery vehicles for various bioactive materials.
of unstable compounds (for example, antimicrobials, antioxidants, However, for drug encapsulation, size and homogeneity of the
flavors and bioactive substances) resulting in better stability of liposomes are more important than the number of lamellars in a
them. Liposome can trap both hydrophobic and hydrophilic com- liposome [14]. Therefore, for vesicles used as drug delivery systems,
pounds, prevent their decomposition, and release the entrapped the membrane composition and supramolecular organization are
compounds at selected targets [11–13]. Therefore, they offer a pow- highly important. For drug delivery applications, the desirable size
erful solubilizing system for a wide range of compounds. of liposome ranges between 50 and 200 nm [22,23].
Liposome exhibit many special biological characteristics,
including (specific) interactions with biological membranes and 3. Liposome as a drug delivery system
various cells, as a result of their physico-chemical properties [14].
Advantages and disadvantages of conventional liposome applica- Drug carriers can provide benefits over traditional drug formu-
tions are depicted in in Fig. 1. Industrial applications of liposomes lations [25]. They protect the entrapped drug from degradation and
are generally focused on drug delivery system and as adjuvants premature metabolization during release in the human body [26].
in vaccination in medicine as well as support matrix for various Liposomes belong to the vesicular delivery systems that can deliver
ingredients and penetration enhancer in cosmetics. Furthermore, hydrophilic or hydrophobic substances. They are spherical, have
liposomes are used as signal enhancers/carriers in medical diag- small particle size and high drug EE, possess an inner aqueous core
nostics and analytical biochemistry, as solubilizers for various and an external double lipidic layer [27] and are biocompatible with
ingredients etc. [14]. human cells [28–30].
Liposomes can be prepared using a wide range of methods which The first liposomes were introduced in 1965 by Alec Bang-
influence the liposome characteristics, such as size, lamellarity ham [31]. As excellent carrier systems because of their similarity
and encapsulation efficiency (EE, which is expressed as the per- to natural cells [19] they are able to overcome biological barri-
centage of drug that is successfully entrapped into the liposome). ers [32]. Research on liposome technology has progressed from
The concentration of incorporated substances in liposomes can be conventional vesicles to “second-generation liposome”, in which
L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984 3

Fig. 1. Advantages and disadvantages of conventional liposome applications [3,15].

Fig. 2. Types of vesicles depending on size and lamellarity. SUV, small unilamellar vesicle (20–100 nm); LUV, large unilamellar vesicle (>100 nm); MLV, multilamellar vesicle
(>500 nm); MVV multivesicular vesicle (>1000 nm) [21,24].

long-circulating liposomes are obtained by modulating the lipid Table 1


Advantages and disadvantages of stealth liposome [37].
composition, size, and charge of the vesicle. Liposomes with mod-
ified surfaces have also been developed using several molecules ADVANTAGES DISADVANTAGES
such as glycolipids or sialic acid [3,33], and modified and unmod- Increase in hydrophilicity PEG dilemma
ified dextrans [34], which afford stealth liposomal properties. A Increase in aqueous solubility Poor cellular uptake
breakthrough in long-circulating liposome production occurred Increase in hydrodynamic volume Ineffective endo/lysosomal escape
when the synthetic polymer poly-(ethylene glycol) (PEG) was Reduction in protein binding ABC phenomenon (multiple injection)
Reduction in RES uptake Obstructed interaction between
incorporated into liposome composition. Consequently, extended
ligands and target
blood-circulation of liposome was detected when the PEG was Disturbed drug release from carriers
presented on the surface of the liposomal carrier. Because of Increased normal tissue exposure
the presence of PEG, mononuclear phagocyte system uptake was
reduced and since they are “invisible” to the bodyś defense system
(reticuloendothelial system (RES)), sterically stabilized liposomes Different strategies for overcoming the disadvantages of PEGy-
are often called stealth liposomes [9,35]. Furthermore, PEG reduces lated liposome have been implemented. To enhance the cellular
the particles’ aggregation by steric stabilization; therefore, the uptake of PEGylated liposome, the ligands (e.g. proteins, vitamins,
PEGylated liposomes’ stability is improved during storage and peptides, antibodies and antibody fragments, nucleic acids etc.) for
application [36]. Although PEGylated liposome exhibits many receptors on the surface of targeted cells on PEGylated carriers can
advantages over earlier varieties, a dilemma (Table 1) regarding be displayed. So-called “active targeting” improves the selectiv-
using PEG for liposome coating appeared, associated with the dif- ity, binding and uptake of the carriers by targeted cells [38,39].
ficulty of cellular absorption of drugs and subsequent endosomal Endo/lysosomal escape of PEGylated carriers can be improved by
escape [37]. de-PEGylation. Intracellular environments with a low pH in endo-
somes/lysosomes (pH-responsive polymeric carriers) [37,40–43]
4 L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984

Fig. 3. Liposome as a drug delivery system and routes of drug administration to the human body.

or the use of enzyme matrix metalloproteinase [44–46] can trigger to enter the market three decades after the discovery of liposome
PEG cleavage. [61,62]. Around twenty liposome-based drugs are now approved
Accelerated blood clearance (ABC) phenomenon, unexpected for clinical use (Table 2) and almost sixty products are in clinical
pharmacokinetic change resulted usually from a second dose of stages of development [60]. Liposome nanotherapeutics for cancer
conventional PEGylated liposomes or after several time repeated treatment have been on the market since the 1990 s, and a variety
injections of PEGylated liposomes was detected [47–49]. To miti- of liposomal drugs for the treatment of other diseases are in various
gate the induction of this phenomenon, different strategies were stages of clinical development (Table 2) [63].
proposed; the administration regimen change [50], reduction in Generally, four basic stages are involved in all methods of
density of PEG on liposome surface [51], alternative polymers liposome preparation; drying down of a mixture of lipids from
usage such as poly(N-vinyl-2-pyrrolidone (PVP) [52] and cleavable an organic solvent, dispersion of lipids in aqueous media, sep-
PEGylation. The most promising alternative is cleavable PEGylation aration and purification of formed liposomes, and analyzing
[37]. PEGylated long-circulating antibody liposomes should not be the final product [3]. Nowadays, novel liposome technologies
overmodified, the quantity of attached antibodies should not com- for delivery of therapeutics such as Stealth liposome technol-
promise the longevity too much, to provide unhindered interaction ogy, Non-PEGylated liposome technology, DepoFoamTM liposome
with target cells [53]. technology and Lysolipid thermally sensitive Liposome (LTSL) tech-
Active molecules can be delivered to the site of action, and at nology are used [8], and by changing the lipid bilayer composition,
present, several formulations of liposome are in clinical use. Drug size, and charge of the vesicle, long-circulating liposome with con-
administration with liposome can be achieved using many routes trolled and gradual release of active substances can be synthesized
(Fig. 3) (intravenous, oral inhalation, local application, and ocular) (Table 2).
and these can be used for the treatment of various diseases [54]. For the purpose of different agents target delivery at a specific
Drugs and other substances can be entrapped either in the internal place in the body (e.g. anticancer agent delivery to the tumors),
aqueous phase of the liposome or in the lipid bilayers, depending functionalization of liposome with a suitable ligand, (i.e. peptides,
on their hydrophilicity [14]. antibodies, aptamers, small molecules, etc.) should be performed
Liposomes, however, suffer from problems of storage stabil- with so called ligand-targeted liposome or targeted liposome.
ity caused by particle aggregation or fusion and PLs degradation, Targeted liposomes do not affect healthy tissues, while the conven-
hydrolysis or oxidation [55,56]. These disadvantages can not only tional approach for e.g. cancer therapy results in low accumulation
be eliminated, but also extended drug delivery route to oral and of anticancer agents at the affected place with associated off-target
inhalable administration can be achieved with the preparation of effects [64]. The main strategies for the therapeutic agent delivery
a solid dry free-flow powder called proliposome [57–59]. They can at affected site involve two approach: passive targeting (PEGylated
form liposomal structures via self-assembly in water. liposome with enhanced permeability and retention effect) and
active targeting (functionalized liposome with targeting ligands)
3.1. Commercially available formulations of liposome [64–66].
The cell penetrating peptide (CPP) assist delivery of therapeu-
Till now many liposomal formulations have been developed for tic agents using liposome as a nanocarriers can be performed: a)
a variety of drugs [8,9,60]. Ambisome® was the first liposomal drug through covalent conjugation of the drug molecules with a CPP, b)
L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984 5

Table 2
Clinically used liposome-based products, their active components, therapeutic applications, structure of lipid bilayer and preparation technology used [8,24].

Therapeutic application Liposomal preparation Active compound Method of preparation


(brand name) (lipid bilayer)

Cancer therapy DaunoXome® DAUNORUBICIN (DSPC and CH) Conventional technology


DepoCyt® CYTARABINE DepoFoam technology
(DOPC, triolein, DPPG and CH)
Doxil® DOXORUBICIN (PC and CH) Stealth liposome technology
EvacetTM DOXORUBICIN Conventional technology
(egg PC and CH)
Lipodox® DOXORUBICIN (HSPC, DSPE-MPEG-2000 and CH) Stealth liposome technology
Marqibo® VINCRISTINE (sphingomyelin and CH) Conventional technology
Mepact® MIFAMURTIDE Non-PEGylated liposome technology
MTP-PE (POPC and OOPS)
Myocet® DOXORUBICIN Non-PEGylated liposome technology
(egg PC and CH)
OnivydeTM IRINOTECAN Stealth liposome technology
(DSPC, CH and DSPE-MPEG-2000)
®
Fungal infection Abelcet AMPHOTERICIN B (DMPC and DMPG) Non-PEGylated liposome technology
AmBisome® AMPHOTERICIN B (HSPC, CH and DSPG) Conventional technology
Amphocil® AMPHOTERICIN B (sodium cholesteryl sulphate) Non-PEGylated liposome technology
Amphotec® AMPHOTERICIN B (cholesteryl sulfate) Non-PEGylated liposome technology
Analgetics DepoDurTM MORPHINE SULFATE (DOPC, CH, DPPG, tricaprylin, DepoFoam technology
and triolein)
Exparel® BUPIVACAINE DepoFoam technology
(DPPG, tricaprylin and DEPC)
Viral vaccines Epaxal® HEPATITIS A VACCINE (DOPC and DOPE) Detergent removal method
Inflexal® V INACTIVATED HEMAGLUTININE OF INFLUENZA Detergent removal method
VIRUS STRAINS A and B (DOPC and DOPE)
Photodynamic therapy Visudyne® VERTEPORFIN (egg PG and 1-␣-DMPC) Conventional technology

PC - phosphatidylcholine, CH - cholesterol, DSPC - distearoylphosphatidylcholine, DOPC - dioleoyl phosphatidylcholine, DPPG - dipalmitoyl phosphatidylglycerol, PS - phos-
phatidylserine, DSPE-MPEG-2000−1,2-distearoyl-phosphatidyl ethanolamine-methyl-polyethylene glycol conjugate-2000, DMPC - L-␣-dimyristoylphosphatidylcholine,
DMPG - l-␣-dimyristoylphosphatidylglycerol, DSPG – distearoyl phosphatidylglycerol, DEPC - 1,2-dierucoyl-sn-glycero-3-phosphatidylcholine, DOPE - 1,2-dioleoyl-
sn-glycero-3-phosphoethanolamine, POPC - 1-palmitoyl-2-oleoylphosphatidylcholine, OOPS - 1,2-Dioleoyl-sn-glycero-3-phospho-l-serine. HSPC - hydrogenated soy
phosphatidylcholine, DSPE-MPEG-2000−1,2-distearoyl-phosphatidyl ethanolamine-methyl-polyethylene glycol conjugate-2000.

encapsulation of drugs into CPP attached liposome, and c) by phys- equilibration method used for drug loading strongly depends on
ical adsorption of drugs with CPPs using electrostatic complexation lyophilization. Producing a liposomal drug kit product via the pas-
method [64,67]. With functionalization of liposome using targeting sive equilibration method could be performed by coupling a stable,
ligands better transport of liposome in cancer cells is provided [68], lyophilized empty liposome system to hydrophilic drug loading
nonspecific distribution to undesired tissues is decreased [69] and [72]. Mechanical dispersion methods include lipid film hydra-
high specificity and targeting efficiency are reached [64]. tion, microemulsification, sonication, French pressure cell method,
membrane extrusion, dried reconstituted vesicles and the freeze-
thawed method. Solvent dispersion methods include ethanol
4. Methods of drug loading in liposome preparation
injection method, ether infusion method, double emulsification
and reverse-phase evaporation [73]. Lipid-soluble, water-soluble
One of the critical issues in liposome preparation is loading
and amphiphilic materials may be encapsulated in liposome. This
drugs (hydrophilic or hydrophobic). In a multistage preparation,
is a way to decrease their side effects and toxicity [74].
it is important to know when and at what stage the drug is incor-
Using the filming-rehydration method, for example, multifunc-
porated into the system. Methods of liposome preparation can be
tional composite liposomes containing Laurel essential oil (LEO)
in general divided into two major groups; the active loading tech-
and silver nanoparticles (AgNPs) (Lip-LEO-AgNPs) were prepared.
nique with proliposome and lyophilization, where liposomes are
Later they were mixed with chitosan solution to cast on a polyethy-
obtained by transfer of PLs from an organic phase into an aqueous
lene (PE) film (PC-Lip/LEO/AgNPs) for pork preservation [74].
phase, and passive loading techniques with mechanical disper-
Instability of liposomes in the aqueous state during long-term
sion, solvent dispersion and the detergent removal method with
storage may affect the possibility of using them as drug delivery sys-
dialysis where lipid films are first deposited on a substrate and
tems. Liposomes, stored as aqueous dispersions, are instable since
subsequently hydrated to give liposomes. Active loading refers to
the encapsulated drugs tend to leak out of the bilayer structure
methods of incorporating drugs after liposome preparation. Usu-
and the liposomes might aggregate. Therefore, it is generally nec-
ally, these are gradient loading techniques involving buffer or
essary to use them within the first few months of preparation [75].
ammonium sulfate gradients. Passive loading, or passive aqueous
To overcome this problem lyophilization, as a drying technique
capture, is what happens for all techniques where a drug is captured
for thermolabile pharmaceuticals, should be used [75,76]. A typ-
during liposome formation process. Active loading refers to meth-
ical freeze-drying process consists of three main phases: freezing,
ods of incorporating drugs after liposome preparation. Usually,
primary drying and secondary drying, during which stresses of lipo-
these are gradient loading techniques involving buffer or ammo-
somal dispersions may occur [77]. To stabilize the liposomes during
nium sulfate gradients. Trans-membrane gradient is a driving force
lyophilization process different excipients (e.g. sugars, usually dis-
for active loading of a weak base (e.g. ammonium sulfate) into lipo-
accharides) or adjuvants (e.g. organic solvents, such as tert-butyl
somes and using this approaches, high EE can be obtained. For
alcohol) should be used [77].
appropriate loading, a weak base with a pKa≤11 and log P value
When the liposomes are formed on the basis of W1/O/W2 emul-
ranging from −2.5 to 2.0 should be used [70,71].
sions, the organic solvents are usually removed by evaporation at
Lyophilization has a significant effect on some sorts (e.g.
high temperature [78]. The disadvantage of using evaporation to
cholesterol-free, pegylated (CF-PEG)) of liposomes. The passive
6 L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984

Table 3
Advantages and disadvantages of using conventional techniques for liposome formation.

Method Advantages Disadvantages Types of vesicles Reference

Reverse-phase evaporation simple process, use of large quantity of organic MLVs, LUVs [19,89,90,91]
technique suitable EE solvent (unsuitable for peptides
encapsulation),
time consuming method,
sterilisation is needed
Injection techniques simple process difficult removal of organic solvent, SMVs, SUVs [92,93,94]
time consuming method,
possible nozzle blockage (ether
system),
sterilisation is needed
Electroformation methods homogeneous size distribution high cost of electrodes (mainly for GUVs [95,96,97]
platinum and ITO electrodes),
buffer with ionic low strength (a
maximum of 50 mM)
Microfluidic method good particle size control difficult removal of organic solvent, SUVs, LUVs (for MHF), [98,99,100,101,102]
(1. Micro hydrodynamic unsuitable for bulk production GUVs (for MD),
focusing (MHF), 2. GUVs (for PJM)
Microfluidic droplets (MD), Novel methods for liposome
3. Pulsed jet flow preparation
microfluidics (PJM))
Thin-film hydration (Bangham simple process, low EE (specially for water soluble MLVs [31,103]
method) straightforward approach active agents),
difficult removal of organic solvent,
small scale production,
large vesicles without particle size
control,
time consuming method,
sterilisation is needed
Detergent depletion good particle size control, difficult removal of organic solvent, MLVs, LUVs [104,105,106]
simple process, detergent residue,
homogenous product time consuming method,
poor EE,
sterilisation is needed
Membrane extrusion simple and fast process, drawbacks in large-scale MLVs [107,108,109]
good particle size control, processing,
non contamination (e.g. possibility of clogging the pores
organic solvent)
Heating method simple and fast process, high temperature MLVs, SUV [24,110,111,112]
non contamination (e.g.
organic solvent),
no sterilisation is needed

SMVs - small multilamellar vesicles.


GUVs - giant unilamellar vesicles (size >1000 nm) [24].

remove solvent is in possible leakage of the inner aqueous solution with improved morphologies and minimal residual solvent. Dense
as well as agents by diffusion through the fluid oil phase. Fur- gas technology has become an alternative production technique
thermore, the evaporation process might break down the double that is more environmentally acceptable and economical than con-
emulsions and lead to drug loss [79]. ventional methods.

5. Conventional techniques for liposome preparation 5.1. Tools for size reduction and uniform size distribution of
liposome
Different conventional techniques of liposome formation,
such as the reverse-phase evaporation technique, injection tech- Particle size of liposome is a very important parameter for their
niques, hydration methods, electroformation methods, microflu- application in medical and other in vivo applications, since it affects
idic method, thin-film hydration (Bangham method), freeze drying different properties such as stability, encapsulation efficiency, drug
of double emulsions, membrane extrusion, detergent depletion, release, mucoadhesion and the cellular uptake of liposome [113].
heating method etc. are well documented [3,17,80–86]. Most of Therefore, an important step in liposome preparation is also prepa-
them have drawbacks, such as complex procedures, working under ration of uniform-size distributed liposomes in a reproducible
harsh process conditions (denaturation of active components may manner. Liposomes with diameters >0.1 ␮m can be, in compari-
occur) and poor drug EE [87]. Another drawback is the use of organic son to smaller ones, opsonized more rapidly and to a greater extent,
solvents, since they have to be removed when liposomes are used which leads to more rapid removal from the body by the reticuloen-
in pharmaceutical applications, namely, to produce pharmaceuti- dothelial system [109]. Also, biodistribution of liposome is related
cals, besides safety rules also strict regulatory requirements must to particle size [114].
be followed. This leads to an increase in production costs because of The initial hetero-dispersed suspensions of liposomes can be
the need for additional purification and waste disposal steps [88]. reduced in size and size distribution by several different meth-
Advantages and disadvantages of using conventional techniques ods. One of the most commonly used methods, for size reduction
for liposome formation are depicted in Table 3. and distribution, which is suitable also for large-scale production,
Therefore, there is incentive for the use of SCFs technologies as is homogenization. During the process, hetero-dispersed liposome
a technological platform for the production of liposomal systems preparation is cycled pumped under high pressure through a small
L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984 7

Table 4
Advantages and disadvantages of different size-processing methods for uniform particle size liposome.

ADVANTAGES DISADVANTAGES

HOMOGENIZATION suitable for large-scale production liposome size distribution is quite broad and variable
possible metal and oil contamination
SONICATIONORULTRASONIC IRRADIATION suitable for SUVs preparation limited processing capacity
peroxidative damage to the lipids due to the local
overheating during the process
possible contamination with titanium
EXTRUSION several membrane/filter pore sizes are limitation in large-scale production
available for producing liposomes in different membrane/filter clogging
selected size ranges membrane/filter fragility
quite narrow distribution of the liposome due slow flow through the membrane/filter
to several times cycling of the material
through the selected-size membrane/filter

reaction tank, until a desired average size of liposome is achieved. processing parameters [131]. The rate of decompression and the
Sonication or ultrasonic irradiation is another method for reduc- opening diameter of the nozzle control the size of the resulting
ing the size of liposome because of the shear forces present during liposomes.
the process. Another size-processing method for uniform liposome
preparation is extrusion through uniform pore-size membranes. 6.1. Depressurization of an expanded liquid organic
General advantages and disadvantages of different size- solution-suspension (DELOS) method
processing methods for uniform particle size liposome formation
are depicted in Table 4. For DELOS process, the substances are first dissolved in suitable
In a study by Ong et al. [109], evaluation of different organic solvent and then the solution is mixed with SC CO2 in a
size-processing methods (extrusion, ultrasonication, freeze-thaw vessel at desired temperature and pressure. Then the mixture is
sonication (FTS), sonication and homogenization) to obtain nano- depressurized through a nozzle into a vessel to form liposomes.
size liposome was performed. The extrusion method was found to Therefore, the SC CO2 is used as co-solvent to the organic solvent.
be the most efficient, followed by FTS, ultrasonication, sonication The main advantage of this technique in comparison to PGSS is that
and homogenization. With increase in the flow rate of the extrusion the thermo-sensitive material can be handled to prepare fine par-
process, decrease in the size of extruded liposome was deter- ticles without melting it and DELOS process requires mild working
mined. Furthermore, the liposome size and distribution declined pressure (e.g. 10 MPa 35 ◦ C), which is a very important param-
with decrease in membrane pore size. eter from the economical point of view, especially for industrial
application [132,133]. A possible mechanism of liposome forma-
6. Supercritical fluids (SCFs) methods for liposome tion via the SC CO2 method (DELOS) is illustrated in Fig. 4. Four
preparation stages of the process exist. In the first stage, PL molecules are
present primarily in the form of bilayer or curvature organized
Traditional methods of preparing liposome, such as thin film spontaneously in the aqueous medium at ambient conditions. In
hydration or ethanol injection, have many shortcomings, such as the second stage, CO2 is rapidly solubilized in the aqueous phase
poor monodispersion, poor stability, high residual organic solvent, during pressurization. Dissociation equilibrium results with a rela-
and side effects [115]. In addition, organic solvents used for conven- tively small amount of CO2 in carbonic acid form and the remaining
tional liposome preparation may cause environmental pollution. amount in the unhydrated state [134]. In the third stage or depres-
They may also become toxic and degrade the active ingredient, surization, CO2 molecules are rapidly released from PL bilayers to
presenting a possible risk for human health [116]. In the case of pro- temporarily disrupt them into highly dispersed PLs. In the fourth
tein drugs, detergents or organic solvents can cause denaturation stage after formation of the instantaneous solution, temporarily
of proteins and affect the membrane properties [117]. separated PLs and CH reorganize rapidly, as a result of hydrophobic
Supercritical fluids assisted techniques, such as supercriti- and van der Waals interactions and finally pack into drug-loaded
cal anti-solvent (SAS), rapid expansion of supercritical solutions liposome [131].
process (RESS), supercritical assisted atomization (SAA), super- Lutein-loaded liposomes were prepared using the DELOS
critical fluid extraction (SFE) [118–120] or supercritical reverse method [135]. The expansion of CO2 -PL/lutein suspension which
phase evaporation (SCRPE), particles from gas saturated solutions depressurized from high pressure to ambient conditions resulted
(PGSS), depressurization of an expanded liquid organic solution- in liposome formation. Characterization via several techniques
suspension (DELOS) [17,121–125] and supercritical assisted showed that liposomes with superior characteristics were formed.
liposome formation (SuperLip) [126], are being developed as Liposomes prepared with the SC CO2 method had a particle size
successful alternatives to traditional methods for liposome pro- of 147.6 ± 1.9 nm – 195.4 ± 2.3 nm and exhibited 87.1 % of unil-
duction [127,128]. They can be classified as green technologies. amellar, 8.7 % of bivesicular and 4.2 % of bilamellar spherical vesicles
The most widely used supercritical gas is carbon dioxide, since it and EE of 56.7 ± 0.7 %–97.0 ± 0.8 %. At higher pressure (20–30 MPa)
is non-flammable, non-toxic, non-corrosive, inexpensive, environ- and depressurization rate (9–20 MPa/min) a higher encapsulation
mentally acceptable and has easily manageable critical parameters of lutein was obtained. Also, lutein-to-lipid ratio influenced the
of 31.1 ◦ C and 7.38 MPa [129]. It is also suitable for use with thermo- morphology of vesicles along with size distribution and EE. [135].
sensitive materials [130], and a low energy input is required for SC The bilayers vesicles formation most likely occurred because of the
CO2 processing. depressurization and dispersion of the two nearby domains of PLs.
Liposome, produced with the SC CO2 method have enhanced The reorganization of dispersed internal molecules of PLs formed
intactness, sphericity and uniformity compared to liposome, pro- an inner bilayer which is surrounded by the outer one formed by
duced by the thin film hydration method. Dense phase CO2 enables external LPs [135].
the processing of phospholipid (PL) aggregates into nano/micro Utilization of liposome as an encapsulation medium for antho-
particles. Their characteristics can be controlled via tuning the cyanin via DELOS method may provide unique features to regulate
8 L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984

Fig. 4. Liposome formation by the DELOS method - Schematic illustration of proposed mechanism where substances are first dissolved in suitable organic solvent and then
the solution is mixed with SC CO2 in a vessel at desired temperature and pressure. Then the mixture is depressurized through a nozzle into a vessel to form liposomes [131].

the characteristics for its protection, oral administration and


delivery purposes. Liposomes were formed as the CO2 -expanded
suspension was depressurized. Particle size of with SC CO2 pre-
pared anthocyanin-loaded liposome was 159 ± 0.2 nm and EE of
50.6 % at 30 MPa, 9 MPa/min and 50 ◦ C [136]. The anthocyanin
release from liposomes was slow in the simulated gastric fluid but
rapid in the simulated intestinal fluid, induced by the degradation
of the vesicles by pancreatin [136]. High concentration of added
anthocyanin leads to imperfect formation because of enhanced
anthocyanin-phosphate interactions and dividing the drug into the
bilayer. On the other hand, the increase in CH concentration has a
relatively small impact on the formation of liposomal bilayers and
shape but leads to a reduced surface charge of liposomes. Using the
DELOS method at 30 MPa and 50 ◦ C, mostly unilamellar, without
any multilamellar or multivesicular vesicles, were formed [136].
Again, in in vitro release study of melatonin liposome showed
that liposomes produced using SC CO2 were resistant to low pH
in simulated gastric conditions [137]. In a simulated intestinal
environment, enteric solubility of these liposomes, which is an Fig. 5. Liposome preparation using SAS method. The SCF is first pumped to the top
important property for controlled release of bioactive ingredients, of the high-pressure vessel until the system reaches a constant temperature and
pressure. Subsequently, active substance solution is sprayed as fine droplets into
was enhanced. Compared to those produced using the thin-film
above SCF bulk phase through an atomization nozzle. Liposomes are formed in a
hydration (TFH) method, SC CO2 -produced liposome toxicity was second step, when the hydration of the processed material is carried out.
much lower. This was confirmed by analysis of the organic solvent
residue in the liposomes using gas chromatography - mass spec-
trometry [137]. Additionally, when the liposomes were formulated because of many advantages. It enables lower residual solvent con-
using the DELOS method, the mean diameter of the liposomes was tent, is a relatively simple process, and is able to process molecules
66.19 nm and the polydispersity index (PdI) was 0.267, while when with poor solubility in the SCF, as the SCF is used as an anti-solvent
the TFH method was used, mean diameter of the liposomes was [139]. The SAS process has often been used for the precipitation
85.62 nm and the PdI was 0.382, but larger mean diameter devi- of compounds difficult to comminute, such as lipids. An organic
ations using the TFM method were detected. When the liposomes solution containing the solute (drug incorporated in liposomes) to
were formulated with the DELOS method, with increase in pres- be precipitated is placed in contact with a SCF, such as SC CO2 . SC
sure from 10 to 14 MPa, the particle size of the liposomes decreased CO2 acts as an anti-solvent for the solute but is completely miscible
from 88.74 to 66.19 nm. With further increase in pressure, the mean with the organic solvent. When the SAS process is used, in the final
diameter of formed liposomes increased. Therefore, for easier con- obtained liposomes no traces of organic solvents are present com-
trol over dimensions and distribution of liposomes, the SCF method pared to liposomes obtained by conventional methods. Liposomes
is more convenient [137]. are formed in a second step, when the hydration of the processed
Overall, this SC CO2 assisted method was demonstrated to material is carried out [140]. They are hydrated at ambient condi-
be superior for the preparation of liposomes over the traditional tions by an aqueous solution (Fig. 5). The SAS process appears to
TFH method, resulting in small size, relatively high uniformity be an efficient and environmentally friendly process for producing
and enhanced stability of spherical shape, while offering modified liposomes.
physicochemical properties of bilayer membranes and the possi- Vitamin D3 (VD3) proliposomes (VDP) were prepared to make
bility of reducing the usage of sterols [138]. VD3-liposomes (VDL) using SAS via hydration of the proliposomes.
VDP can be first produced by SAS and then the liposomes can be
6.2. Supercritical anti-solvent (SAS) method simply formed via hydrating [141,142].
1,2-Distearoyl-sn-glycero-3-phosphocholine(polyethylene gly-
The most commonly reported methods for proliposome prepa- col) (DSPC-PEG) liposomes loaded with albendazole (ABZ) using
ration are SAS methods. This is the most attractive technique SAS were also prepared. ABZ is a colorless, non-ionizable, lipophilic,
L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984 9

Fig. 6. Liposome preparation using RESS method. RESS process is consisting of two
steps: (a) dissolving the solid substance and carrier in a SCF and (b) formation of
particles due to supersaturation.

low molecular weight crystalline compound which is insoluble in


water and has promising anticancer activity when formulated to
increase its cellular uptake. The diameter of unilamellar and spher-
ical liposomes was 167.2 ± 5.3 nm and a controlled release effect on
ABZ release profile was observed The experiments were performed
at 37 ◦ C, 25 MPa. [143].
Using the SAS technique, hydrophilic and hydrophobic com- Fig. 7. SuperLip process [154]. SuperLip process is consisting of two steps: first,
pounds can be entrapped in lipid vesicles. In this process, the water droplets produced by atomization inside a high-pressure vessel, filled with an
compounds to be encapsulated do not need to be dissolved in SC expanded liquid mixture formed by PLs/ethanol/CO2 , containing drugs are obtained
CO2 . and in the second step, a lipidic layer is formed around them.
The SAS process was upgraded to what is called the continuous
anti-solvent (CAS) process. In the SAS process, micronized PLs react 5.2 nm, 0.254 ± 0.010 and −41.7 ± 0.9 mv. Liposome were prepared
spontaneously upon contact with air [144] before the hydration at following condition; 25 MPa, 48 ◦ C, feed ratio of VC against PC
step. CAS affords a solution to this problem because in this process, of 0.25, while the flow rate of the sprayed mixture was about 15
micronization and hydration are performed in the same autoclave g/min [148].
under pressure [119]. Zhang et al. prepared sirolimus liposomes using RESS and the
effects of temperature, pressure, and equilibrium time on the aver-
6.3. Rapid expansion of supercritical solutions (RESS) method age particle size and envelope rate of the liposomes were discussed
in detail [149]. Wen et al. reported a modified technique of RESS
RESS process is consisting of two steps; (a) dissolving the solid to prepare essential oil liposomes [150,151]. The entrapment effi-
substance in a SCF and (b) formation of particles due to supersatura- ciency of the prepared essential oil liposomes was found to be 82.18
tion. In the RESS process, at first SCF is pumped at desired pressure % under optimum conditions (pressure of 30 MPa, temperature of
and temperature to extractor. The SCF percolates and dissolves the 65 ◦ C and ethanol mole fraction in SC CO2 of 15 %) with an average
solid substance(s) in the extractor and then the resulted solution is particle size of 173 nm (spherical shape). Xu et al. successfully syn-
depressurized through a heated nozzle in a low-pressure chamber thesized nanoscaled CoQ10 liposomes (20−40 nm, spherical shape)
(Fig. 6) [2]. RESS process is simple, effective when single nozzle is using RESS under different pressures. Surprisingly, the trapping
used and it minimizes the use of organic solvent and reuses the SCF efficiency of the liposomes can be up to 90 % when the enduring
in continuous process. The main drawback is poor solubility of most pressure was 20–30 MPa [152]. Melatonin-loaded liposomes with
of the pharmaceutical materials (e.g. polymer) in SC CO2 , therefore uniform size distribution and an average diameter of 66 nm were
large amount of fluid is required, consequently the cost of liposome also successfully prepared using RESS [153]. The EE of the lipo-
production using RESS method is increased. Additional, particles somes could reach 82.2 % under optimal conditions (14 MPa and
may aggregate and nozzle block because of cooling during rapid 40 ◦ C), and only 1.03 wt% of ethanol (residual organic solvent) was
expansion of the supercritical solution [145,146]. Drug-loaded lipo- detected in the products, eliminating some potential dangers for
some can also be prepared via RESS; the operating conditions of practical applications [153].
RESS have been found to strongly affect the properties of the pre- Frederiksen et al. reported the preparation of liposomes con-
pared liposomes. The size, uniformity, and EE of the liposomes can taining a water-soluble marker using the RESS method (7% absolute
be varied by changing the pre-expansion pressure, flow rate, and ethanol and carbon dioxide at 25 MPa and 60 ◦ C). The average size
the position of the nozzle in the reactor. Liposomal microcapsules of the liposomes was about 200 nm, and the total amount of ethanol
were produced using SC CO2 without using any organic solvent, used to obtain an EE of 20 % was 15-fold reduced compared to the
CH, or external energy for pumping and mixing the solution. The ethanol injection method [125].
liposomes’ sizes became uniform when the pressure was increased
from 12.4 MPa to 17.2 MPa [147]. 6.4. Supercritical assisted liposome formation (SuperLip) method
Vitamin C (VC) liposomes were prepared by RESS; EE of the pre-
pared VC liposomes was 75.38 ± 1.03 %. The particle size (spherical SuperLip (Fig. 7) consists of an inversion of the traditional lipo-
shape), PdI and Zeta-potential of the prepared samples was 270.4 ± some production steps: first, water droplets containing drugs are
10 L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984

obtained and in the second step, a lipidic layer is formed around Spherical PC liposomes of different sizes and distribution ranging
them. Water droplets are produced by atomization inside a high- between 250 ± 58 nm and 330 ± 82 nm with wrinkled surfaces
pressure vessel, filled with an expanded liquid mixture formed by were successfully produced at 15 MPa and 40 ◦ C. When using a
PLs/ethanol/CO2 . These droplets are rapidly surrounded by a lipid PG/PC mixture, larger liposomes were produced (280 ± 70 nm and
layer, forming a w/CO2 emulsion, and liposomes (w/w emulsion) 350 ± 101 nm). Both PC and PC/PG liposomes were stable over
are formed when they fall in the water pool located at the bot- one month (zeta potential ranging between −20 mV and −30 mV)
tom of the vessel [126]. The vesicles obtained are nanometric and and very high EE for BSA (92–98 %) were obtained. The authors also
sub-micrometric and may entrap proteins, antibodies, antibiotics, found that pressure and temperature influenced the mean diameter
essential oils, agricultural additives, antioxidants, dyes and hol- of liposome. The higher the pressure and the lower the tempera-
low gold nanoparticles [154]. This process offers the possibility ture (i.e. the higher the expanded liquid density), the smaller the
of producing liposomes with a complex architecture of the lipid mean diameter of liposomes.
membrane. The difficulties experienced using the classical lipo- A novel approach to include lipophilic molecules in liposomes
some formation processes, mainly in reproducibility and EE, can using emulsions was developed by Trucillo et al. [161]. Two dif-
be overcome with this method [155]. ferent strategies for the encapsulation of hydrophobic bioactives
SuperLip was used for the liposome production to deliver the within nanoliposomes were attempted. Lipophilic antioxidants
ophthalmic antibiotics ampicillin and ofloxacin, antibacterial drug were entrapped in liposomes using emulsions: a drug was dissolved
used to stop ocular post-surgery infections. The process parameters in the oil phase of an oil in water emulsion, then the aqueous emul-
for liposome production were: water injector diameter of 80 ␮m, sion was entrapped in the liposomes’ inner core using SuperLip.
operative pressure at 10 MPa and temperature at 40 ◦ C, CO2 /ethanol The O/W emulsion method successfully preserved the antioxidant
ratio (gas to liquid ratio (GLR)) was 2.4 w/w, CO2 flow rate was activity of the entrapped molecules. High preservation of antiox-
6.5 g/min, and drug/lipid ratio was 1% w/w. EE for ofloxacin and idant power using emulsion loaded liposome was obtained. The
ampicillin were up to 97 % and 99 %, respectively. Mean diameters reduction of antioxidant power was about 22 % for lipidic layer
of the liposomes, which were ULV, were in the range from 280 ± entrapment but only 2% using the O/W method.
104 nm to 1.76 ± 0.79 ␮m. The liposomes were stable for at least 3
months, with negligible drug leakage [156]. 6.5. Other methods
A polyphenol-rich aqueous extract from olive pomace was
encapsulated in liposomes using SuperLip process. Preservation of A GAS process for liposome preparation is usually used instead
these temperature sensitive compounds demands mild operative of RESS technology when the substances are poorly soluble in SCFs.
conditions which could be achieved using this process. The mean GAS process is a batch process where the precipitator is partially
diameters of liposomes with spherical shape and a slightly smooth filled with the solution of solute and SCF is used as an anti-solvent
surface produced at 13 MPa was smaller than 265 nm, and for lipo- for precipitating a solute (active compound), which is insoluble in
somes produced at 17 MPa, was 168 nm. EEs were about six times SCFs, and that has been dissolved in an organic solvent. The parti-
larger than using conventional methods and were up to 58 % [157]. cles precipitate as the gas concentration in the solution increases
The amphiphilic antioxidant eugenol was entrapped in the inner with pressure. A clear disadvantage of this technique is the lack of
core and in the lipophilic double layer of liposomes using SuperLip control on the particle formation [162]. Water soluble compounds
for their preparation. On the other hand, liposomes with lipophilic of Cordyceps sinensis CS1197 were encapsulated in nano-liposomes
(␣-lipoic acid) antioxidant in the lipidic layer were also produced with a mean diameter of 72.0 ± 2.0 nm using the GAS method,
with the same technique. EEs were about 94 % and 68 % respectively, and much smaller liposomes were formed, compared to the con-
and their antioxidant power was preserved after processing. Lipo- ventional method. GAS mediated liposome production unit can be
somes of nanometric dimensions with spherical shape and smooth operated in fed-batch mode. Reproducibility of the encapsulation
surface were produced and their mean diameters ranged between of both hydrophilic and hydrophobic active compounds is ensured
255 ± 122 nm and 230 ± 96 nm. The operation conditions were: [163].
10 MPa, 35 ◦ C, ethanol solution flow rate was 3.5 mL/min, CO2 flow Nano-liposomes were designed using the GAS method. The GAS
rate was 6.5 g/min (GLR = 2.4 (w/w) and water flow rate was 10 method was compared with Bangham’s method in terms of the
mL/min [158]. mean diameter, coefficient of uniformity, morphology, stability
To manufacture liposomes from PLs using SCF technology, their index and actual energy required for liposome formation. Liposome
dispersibility was determined. The mean diameter of liposomes production using the GAS method under optimized formulation
using the SCF method for DSPC-PEGylated and DOPC-PEGylated conditions (17 MPa, 50 ◦ C, 1.5 % Tween 80 and a depressurization
liposomes was 98.3 ± 3.3 nm and 124.5 ± 4.1 nm (ULV), while using rate of 2.5 MPa/min) yielded nano-liposomes exhibiting a coeffi-
the thin film method it was 153.6 ± 4.5 nm and 131.3 ± 3.4 nm cient of uniformity of 1.10 ± 0.012 with a mean diameter of 63 ±
(ULV), respectively. SCF mediated liposomes were better in parti- 2 nm and with a stability of up to 3 months at different tempera-
cle size and morphology when compared with those produced with tures. The GAS process requires 1.52 times less energy compared
the thin film method. They were also more stable over a period of to Bangham’s method to achieve nano-scale liposome production
3 months compared to those produced with the thin film method. [164].
This green alternative SCF method of liposome preparation is less SCRPE provides a one-step method for the preparation of LUV
laborious, saves time, and is a low energy process [159]. liposomes with a high EE for both water-soluble and oil-soluble
Spherical PC/CH and PC/PE liposomes were produced with nano- substances, allowing aqueous liposome dispersions to be obtained
metric mean diameters down to 200 nm and theophylline was through emulsion formation by introducing a given amount
encapsulated with EE up to 98 % at operation parameters: ethanolic of water into a homogeneous mixture of SC CO2 /lipid/ethanol
solution flow rate of 3.5 mL/min (2.76 g/min), GLR = 2.4 (w/w), 10 [123,165]. Lipids, organic cosolvent and SCFs are combined in a
MPa, 40 ◦ C and water flow rate of 2.14 mL/min. Liposome size dis- stirred reactor at a temperature above the lipid phase transition
tribution, vesicles stability and drug EE were not affected by lipid temperature and an aqueous solution is then slowly added to the
bilayer composition [155]. reactor. Because of the release of SCFs, liposomes are formed.
Campardelli et al. [160] studied the possibility of producing Bile salt-mediated liposomes containing sodium glycocholate
liposomes with a different composition of the bilayer membrane were prepared using an improved supercritical reverse evaporation
(composed of PC or a mixture of PC/PG) using the SuperLip process. (ISCRPE) method which fully eliminates the use of organic solvent
L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984 11

(ethanol). Formulated as notoginsenoside R1 (NGR1) carriers, they liposome preparation. While increased pressure is favorable for the
can be used for oral administration to improve bioavailability [166]. formation of smaller vesicles, liposomes prepared at ≥10 MPa dis-
The mean EE, particle size, and polydispersity index (PDI) of the play a spherical and stable shape, while higher depressurization
optimized liposomal formulation (NGR1@Liposomes) were 49.49 rates resulted in enhanced uniformity [174].
%, 308.3 nm, and 0.229 at optimal conditions 20 MPa and 60 ◦ C. For SFE, followed by RESS and vacuum-driven cargo loading, was
ISCRPE, higher pressure contributed to the formation of smaller used for vitamin C and E hydrophilic and lipophilic compound
vesicles, while a higher depressurization rate boosted the homo- encapsulation in integrated liposomes. Vitamin E acts as the pri-
geneity of the vesicles. Pressure of at least 10 MPa is recommended mary antioxidant and the resultant vitamin E radical then reacts
to achieve a spherical morphology [167]. with vitamin C to regenerate itself [175,176]. This is how they per-
Disadvantages of SCF-based operations involve mechanical, form synergistic action on their antioxidant activity to scavenge
chemical, and biological hazards. Because of the use of high pres- free radicals in human bodies. The average vesicle size was 951.02
sure when flammable and/or explosive fluids are used in the nm with a zeta potential of −51.87 mV. Most of the liposomes
process, mechanical hazards may appear. A possible leakage of formed using the SCF method were LUVs and MVVs. The EE attained
CO2 (if used in the process) presents a health issue and must be was 32.97 % for vitamin C and 99.32 % for vitamin E [177].
prevented using an over dimensioned vent line, ensuring a good The SCF process offers a sustainable and energy-saving tech-
dispersion of the gas in the outside atmosphere [139]. But most of nique for rapid and continuous production of integrated liposomes.
all, the high cost of the equipment is the major drawback using SC These have the capability to microencapsulate a versatile group of
CO2 for the liposome production. bioactive compounds for food and pharmaceutical applications.
Combined methods give additional advantages and are often PL vesicles may also be prepared using high-pressure CO2 /water
used for liposome production. High EE, good stability, low side systems. PL molecules might act as a surfactant at the high-pressure
effects and low residual solvent are achieved when using a com- CO2 and water interface and form a self-assembled membrane in
bined method of thin film hydration and SC CO2 technology [168]. the water phase after decompression of the systems. PL vesicles
A liposomal hydrogel with embedded curcumin (Cur) for treat- were efficiently formed in liquid (fluid)–liquid systems [178].
ment of infected wounds was prepared using a combined method Depressurization of an expanded solution into aqueous media
of thin-film evaporation and SC CO2 technique (TE-SC CO2 ) with process, developed by Meure et al. [121], is a dense gas technique
freeze-drying. Obtained thin lipid film was incubated at 40 ◦ C and in which liposomes can be produced in a single-step process at
18 MPa for an hour in SC CO2 . The organic solvents were removed moderate temperature and pressure. The gas-expanded solution is
after depressurization. The drug is released via phospholipase A2 released through a nozzle into an aqueous medium [87]. The major
hydrolysis of lecithin in liposome with spherical vesicular structure advantage of this process, compared to conventional methods, is
and a size of 188 ± 12 nm [169]. the reduction of the residual solvent content in the product to 2.2
With thin film rehydration TE-SC CO2 , Cur was loaded in % (v/v), which is significantly lower than those reported for con-
a bilayer of liposomes embedded in three-dimensional porous ventional processing [89,92,93,179]. Supercritical fluid extraction
chitosan/␤-GP thermosensitive gels to form thermosensitive (SFE) was used not only to remove the ethanol residue from lipo-
liposome- hydrogels. Sol at room temperature and gel at body some suspensions prepared by ethanol injection but also to affect
temperature can be used for injectable hydrogel which acts as a their size and distribution leading of liposomes. Nanosomes with
drug carrier. In comparison to Cur-liposomes prepared using the a mean diameter of about 180 ± 40 nm with a good storage sta-
FH method, Cur-liposomes prepared with the TE-SC CO2 method bility were obtained at 12 MPa and 38 ◦ C. This technology is easy
had small particle size, good monodispersity and were more stable to scale up to an industrial scale and merge in one step the sol-
[170]. vent extraction, liposome size engineering and an excellent drug
The same method was used for a novel liposome preparation encapsulation in a single operation unit [180].
with photo-responsiveness for controlled drug release. Two
model drugs and a photo-responsive reversible switch, Cur and 6.6. Batch or continuous SCF processes for liposome preparation
4-butylazobenzene-4-hexyloxy-trimethyl-ammoniumtrifluoro-
acetate (BHA) were used in liposomes in which BHA was Different SCF techniques for liposomes preparation, where a
encapsulated in polar groups of PLs in a bilayer of liposomes. SCF is used as a solvent (SCRPE, RESS), as a cosolvent (SuperLip,
Spherical nanoparticles with a typical vesicle structure and good DELOS), as an antisolvent (GAS, SAS, SEDS, ASES) or as a dispersing
monodispersion with an average size of about 140 ± 15 nm were agent (ISCRPE, PGSS) can take place in a batch and/or continu-
obtained when the thin lipid film formed was hydrated with ous manner (Table 5). Long-term, continuous process saves costs,
double distilled water and incubated in SC CO2 at 40 ◦ C and 20 energy and time, when it is properly implemented but on the other
MPa for 1 h. When the BHA liposomes were formed by traditional hand it is more expensive than the batch process. However, when
film hydration, polydispersity was detected; the average liposome comparing the production rate, batch has lower rate than the con-
sizes were 100 ± 20 nm and 830 ± 23 nm, respectively. [171]. tinuous one. The shut-down times in continuous process are rare,
Solution enhanced dispersion by SC CO2 (SEDS) has been proven whereas in a batch process they are often. Continuous operations
to be a prominent method for preparing proliposomes because of yield more consistent product quality, more predictable behavior
its mild operation, product controllable and no need of auxiliary and a better opportunity to maintain and perfect a steady-state
materials [172]. With this method, berberine proliposomes with EE process. Batch processing may also be more often required in phar-
of 90.3 % ± 4.9 % (at 37.8 ◦ C and 18.3 MPa) and nanoscale spherical maceutical industries, while good manufacturing practice (GMP)
particles with mean diameter around 80 nm and neutral zeta poten- is required. However, continuous flow is becoming a more viable
tial were prepared to improve the oral bioavailability of berberine option for industrial application, due to advancements in design
[173]. and technology.
Liposomes can be prepared using a modified supercritical pro- Since liposomes are most commonly used as a drug delivery sys-
cess via depressurization of the liquid phase. This supercritical tem, their production with SCFs techniques should be adapted to
fluids assisted method is based on a modification of the ISCRPE GMP requirements and be implemented on an industrial scale using
and DELOS methods, to combine the advantages of both techniques single step continuous operation. In order to ensure GMP require-
by employing a PL water suspension similar to that in ISCRPE and ments, the equipment should be constituted of stainless steel with
depressurization from the liquid phases in DELOS, for improved a smooth wall, without edges, easily washable and the substrates
12 L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984

Table 5
Different SCF techniques for liposomes preparation using batch or continuous operation.

Method Lipid/active ingredients Operation Particle size EE Reference

PC and CH/ MELATONIN batch process 88.74−66.19 nm (influenced / [137]


DELOS by pressure)
soy lecithin and CH/ batch process 160 ± 2 nm 52.2 ± 2.1 %. [131]
ANTHOCYANIN
POPC and CH/ FLUORESCEIN batch process 200 nm 20 % [125]
isothiocyanate-dextran
(FITC-DEXTRAN) or zinc
phthalocyanine tetrasulfonic
acid (TSZnPc)
HSPC, DSPE-MPEG-2000 and batch process 231.3 ± 6.74 nm (spherical 91.9 ± 3.45 % [181]
CH/ THYMOQUINONE vesicles)
SAS
soy lecithin S75 and CH/ semi- continuous 0.1–10 ␮m (spherical vesicles) 10−20% [120]
CALCEIN process
hydrogenated PC/ VITAMIN D3 batch process 200 nm (spherical shape) 100 % [142]
HSPC, DSPG and CH/ batch process 137 nm (uniform spherical 90 % [182]
AMPHOTERICIN B shape)
PC and CH/ ATRACTYLONE batch process 124 nm (double-layered 87.25 % [151]
hollow spheres)
Tween 61 and CH/ NON batch process 239−969 nm 28 % [183]
RESS
PL and CH/ SIROLIMUS batch process 150 nm 58 % [149]
HSPC, SPC, CH and batch process of 269.2 ± 2.8 nm (spherical, 79.2 ± 4.4 % [184]
DSPE-PEG-2000/ DOCETAXEL SUVs)
PC and CH/ COENZYME Q10 batch process 20 – 40 nm (spherical shape) 90 % (pressure influenced) [152]
soy lecithin and CH/ continuous 400−1200 nm (pressure 31.65 % [185]
d-(+)-GLUCOSE process influenced; LUVs and MVVs)
soybean PC/ BOVINE SERUM continuous process 130 ± 62–294 ± 144 nm 85−90% [126]
ALBUMIN
SuperLip
L-␣-PC/ FARNESOL, LIMONENE, continuous process 116 ± 32 nm - 230 ± 62 nm 74 % for farnesol, 87 % for [161]
LINALOOL (spherical shape) limonene and 54 % for
linalool
PC/CH and PC/PE/ continuous process 200 nm 98 % [155]
THEOPHYLLINE
L-␣-PC/ FLUORESCEIN or continuous process 100−300 nm (spherical shape) 99 % [186]
AMPICILLIN
L-␣-DPPC and L-␣-DOPC/ batch process (at low ethanol concentration - 40 % [187]
SCRPE d-(+)-GLUCOSE MLVs at higher ethanol
concentration LUVs)
L-␣-DPPC and CH/ batch process 0.1–1.2 ␮m (LUVs) 20 % [123,188]
d-(+)-GLUCOSE
DPPC, PEG-DSPE and CH/ batch process 100−2000 nm (spherical 70 % [189]
BOVINE SERUM ALBUMIN shape)
ISCRPE (improved different PLs/ d-(+)-GLUCOSE batch process LUVs 36 % [188]
SCRPE) L-␣-DPPC and chitosan/ batch process 500 nm (LUVs) 17 % [190]
d-(+)-GLUCOSE
SEDS HSPC and soya PC/ SILYMARIN semi-continuous 160.5 nm (LUVs) 91.4 % [191]
process
PGSS Soybean lecithin and CH/ batch process 1.4–25 ␮m (LUVs, MLVs and 6–14.5 % [192]
LAVANDIN ESSENTIAL OIL MVVs)
GAS PC and CH/ AMPHOTERICIN B batch process 3.5–10 ␮m (spherical shape) 20 % [193]
ASES PC, CH and Poloxamer 407/ batch process 2.7–9.4 ␮m (pH influenced) 60 – 100 % [88]
MICONAZOLE

and other components should be of GMP quality. According GMP, temperatures, and mechanical stresses that can degrade labile com-
the procedures should be standardized and monitored, each stage pounds such as vitamins, enzymes, essential oils, and flavors, and
of the preparation has to be validated and automated cleaning pro- enables liposomes dry powder formulations. This green technol-
tocol is mandatory [194,195]. ogy can produce micro- and nano-sized liposomes with narrow
size distribution by altering critical conditions (i.e., temperature
7. Conclusion and pressure). SCFs methods for liposome formulation also enable
high EE [17]. It has also been argued that SCFs assisted methods
Several liposomal formulations are already on the market, and can be scaled-up without much difficulty [17]. Indeed, the super-
new studies are being conducted in the use of advanced liposome critical extraction of liposomes is easy to scale up to an industrial
preparation technologies. Liposome preparation methods have sig- production and merge in one step the solvent extraction [180],
nificant effects on their size, shape and lamellarity, which influence or ISCRPE has a scale-up potential [85] but at RESS scaling up of
the EE. Conventional methods of liposome preparation are not the process is difficult, because of particle aggregation and noz-
suitable for achieving free-solvent liposomes and industrial-scale zle blockage caused by cooling due to the rapid expansion of the
production under current good manufacturing practice conditions supercritical solution [145,146]. Batch and semi-batch technolo-
[18]. They also suffer from low EE of hydrophilic compounds, dif- gies such as RESS, SAS, SAA etc. are quite complex to scale up due to
ficult removal of organic solvents, and poor control of particle size their discontinuous operations. Therefore, we cannot simply gen-
distribution. Liposomes formulation using SCFs avoids the exten- eralize this fact, but further evaluation of each process shall be
sive use of organic solvents (because of their toxicity, they represent done. Several different SCFs assisted methods for liposome prepara-
possible health risks), avoids the use of water, high operating tion have been developed. Because of the non-polar characteristics
L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984 13

Table 6 [4] S.K. Sahoo, V. Labhasetwar, Nanotech approaches to drug delivery and
Particle size and EE for liposomes prepared using different supercritical assisted imaging, Drug Discov. Today 8 (2003) 1112–1120, http://dx.doi.org/10.
methods. 1016/s1359-6446(03)02903-9.
[5] A. Gabizon, D. Goren, R. Cohen, Y. Barenholz, Development of liposomal
Method Particle size distribution (nm) EE (%) anthracyclines: from basics to clinical applications, J. Control. Release 53
DELOS very narrow (150−200) low (20–50) (1998) 275–279, http://dx.doi.org/10.1016/s0168-3659(97)00261-7.
[6] T.M. Allen, Liposomes. Opportunities in drug delivery, Drugs 54 (Suppl 4)
SAS very wide (100–10000) medium (20−100)
(1997) 8–14, http://dx.doi.org/10.2165/00003495-199700544-00004.
RESS narrow (20−900) medium (20−90)
[7] R. Seema, C. Chanchal, S. Ravi, R. Ankur, K. Dinesh, S. Satish, D. Harish, 10.
SuperLip wide (100–1700) high (60−100) liposomes: preparations and applications, Int. J. Drug. Dev. Res. 4 (2012).
[8] U. Bulbake, S. Doppalapudi, N. Kommineni, W. Khan, Liposomal
formulations in clinical use: an updated review, Pharmaceutics. 9 (2017),
of the most commonly used SCF, SC CO2 , the EE of hydrophilic http://dx.doi.org/10.3390/pharmaceutics9020012.
[9] N. Lamichhane, T.S. Udayakumar, W.D. D’Souza, C.B. Simone, S.R. Raghavan,
compounds in liposomes is still low. A brief comparison between J. Polf, J. Mahmood, Liposomes: clinical applications and potential for
different supercritical assisted methods for liposome formulation image-guided drug delivery, Molecules. 23 (2018), http://dx.doi.org/10.
in terms of particle size distribution and EE is given in Table 6. 3390/molecules23020288.
[10] T.O.B. Olusanya, R.R.H. Ahmad, D.M. Ibegbu, J.R. Smith, A.A. Elkordy,
The SuperLip method has proven to be an advanced method that Liposomal drug delivery systems and anticancer drugs, Molecules: A Journal
can circumvent the problem of low EE, since the water droplets of Synthetic Chemistry and Natural Product Chemistry 23 (2018), http://dx.
are first generated and then they are covered by PLs, leading to doi.org/10.3390/molecules23040907.
[11] O.M. Atrooz, Efects of alkylresorcinolic lipids obtained from acetonic extract
the production of monodispersed, organic solvent free and high EE of jordanian wheat grains on liposome properties, Int. J. Biol. Chem. 5 (2011)
hydrophilic drug liposomes. Another technique which also facili- 314–321, http://dx.doi.org/10.3923/ijbc.2011.314.321.
tates high EE is the SAS technique, followed by RESS. On the other [12] R.-O. Benech, E.E. Kheadr, R. Laridi, C. Lacroix, I. Fliss, Inhibition of Listeria
innocua in cheddar cheese by addition of nisin Z in liposomes or by in situ
hand, DELOS results in the lowest EE, but with this method, very production in mixed culture, Appl. Environ. Microbiol. 68 (2002)
narrow particle size distribution appears, which is not the case 3683–3690, http://dx.doi.org/10.1128/aem.68.8.3683-3690.2002.
when other SCFs methods are used. [13] T. Shehata, K.-I. Ogawara, K. Higaki, T. Kimura, Prolongation of residence
time of liposome by surface-modification with mixture of hydrophilic
Although the use of SCFs for liposome preparation is an
polymers, Int. J. Pharm. 359 (2008) 272–279, http://dx.doi.org/10.1016/j.
advanced and effective method, some disadvantages still need to ijpharm.2008.04.004.
be overcome, such as the use of organic solvents (DELOS, SuperLip), [14] D.D. Lasic, Applications of liposomes, in: Handbook of Biological Physics,
formulation of large liposomes (SCRPE, ISCRPE), wide particle size Elsevier, 1995, pp. 491–519, http://dx.doi.org/10.1016/S1383-
8121(06)80027-8.
distribution (SCRPE), and multistep preparation (SAS) [174]. [15] J.S. Dua, A.C. Rana, A.K. Bhandari, Liposome: methods of preparation and
Additional research in process design and liposome preparation applications, IJPSR. 3 (2012) 14–20.
using supercritical fluids assisted methods on an industrial scale [16] Y. Amini, S. Amel Jamehdar, K. Sadri, S. Zare, D. Musavi, M. Tafaghodi,
Different methods to determine the encapsulation efficiency of protein in
is necessary, because of the low level of continuous production, PLGA nanoparticles, Biomed. Mater. Eng. 28 (2017) 613–620, http://dx.doi.
high investment costs, and disadvantages associated with the use of org/10.3233/BME-171705.
SCFs. However, the influence of each parameter of the supercritical [17] Y.P. Patil, S. Jadhav, Novel methods for liposome preparation, Chem. Phys.
Lipids 177 (2014) 8–18, http://dx.doi.org/10.1016/j.chemphyslip.2013.10.
fluids assisted process must be better investigated before liposome 011.
production is carried out on an industrial scale [196]. [18] J. Zhong, L.C. Dai, Liposomal preparation by supercritical fluids technology,
Combined methods (supercritical assisted and conventional TE- Afr. J. Biotechnol. 10 (2011) 16406–16413, http://dx.doi.org/10.5897/AJB11.
1394.
SC CO2 ) for liposome preparation are recommended. [19] L.A. Meure, N.R. Foster, F. Dehghani, Conventional and dense gas techniques
Author Contributions for the production of liposomes: a review, AAPS PharmSciTech 9 (2008)
ML and MP wrote the manuscript, participated in conceptual- 798–809, http://dx.doi.org/10.1208/s12249-008-9097-x.
[20] U.B. Kompella, K. Koushik, Preparation of drug delivery systems using
ization and review & editing of the manuscript. ML, ŽK and MP
supercritical fluid technology, Crit. Rev. Ther. Drug Carr. Syst. 18 (2001)
performed a literature review. All authors have read and approved 173–199.
the final manuscript. [21] E. Elizondo, E. Moreno, I. Cabrera, A. Córdoba, S. Sala, J. Veciana, N. Ventosa,
Liposomes and other vesicular systems: structural characteristics, methods
of preparation, and use in nanomedicine, Prog. Mol. Biol. Transl. Sci. 104
Declaration of Competing Interest (2011) 1–52, http://dx.doi.org/10.1016/B978-0-12-416020-0.00001-2.
[22] H. Harashima, K. Sakata, K. Funato, H. Kiwada, Enhanced hepatic uptake of
liposomes through complement activation depending on the size of
The authors declare that they have no known competing finan- liposomes, Pharm. Res. 11 (1994) 402–406, http://dx.doi.org/10.1023/
cial interests or personal relationships that could have appeared to a:1018965121222.
influence the work reported in this paper. [23] M.C. Woodle, Sterically stabilized liposome therapeutics, Adv. Drug Deliv.
Rev. 16 (1995) 249–265, http://dx.doi.org/10.1016/0169-409X(95)00028-6.
[24] C.I. Nkanga, A.M. Bapolisi, N.I. Okafor, R.W.M. Krause, General perception of
Acknowledgments liposomes: formation, manufacturing and applications, liposomes -
advances and perspectives, angel catala, IntechOpen (2019), http://dx.doi.
org/10.5772/intechopen.84255.
The authors acknowledge the financial support of the Slovenian [25] R. Langer, New methods of drug delivery, Science 249 (1990) 1527–1533,
Research Agency (research core funding No. P2-0046 - “Separation http://dx.doi.org/10.1126/science.2218494.
[26] J.A. Champion, Y.K. Katare, S. Mitragotri, Particle shape: a new design
Processes and Product Design” and research core funding No. J2- parameter for micro- and nanoscale drug delivery carriers, J. Control.
1725 “Smart materials for bioapplications”). Release 121 (2007) 3–9, http://dx.doi.org/10.1016/j.jconrel.2007.03.022.
[27] M.R. Rafe, Z. Ahmed, Liposomal drug delivery systems have opened a New
Window in pharmaceutical sciences: a literature-based review, Asian J.
References Pharm. 11 (2017) 250–254.
[28] T. Koklic, Perifosine induced release of contents of trans cell-barrier
[1] K. Shashi, K. Satinder, P. Bharat, A complete review on: liposomes, IRJP. 3 transport efficient liposomes, Chem. Phys. Lipids 183 (2014) 50–59, http://
(2012) 10–16. dx.doi.org/10.1016/j.chemphyslip.2014.05.006.
[2] V. Dordevic, B. Balanc, A. Belscak-Cvitanovic, S. Levic, K. Trifkovic, A. [29] D. Zabeo, A. Cvjetkovic, C. Lasser, M. Schorb, J. Lotvall, J.L. Hoog, Exosomes
Kalusevic, I. Kostic, D. Komes, B. Bugarski, V. Nedovic, Trends in purified from a single cell type have diverse morphology, J. Extracell.
encapsulation technologies for delivery of food bioactive compounds, Food Vesicles 6 (2017), http://dx.doi.org/10.1080/20013078.2017.1329476.
Eng. Rev. 7 (2015) 452–490, http://dx.doi.org/10.1007/s12393-014-9106-7. [30] C. Lasser, M. Eldh, J. Lotvall, Isolation and characterization of RNA-Containing
[3] A. Akbarzadeh, R. Rezaei-Sadabady, S. Davaran, S.W. Joo, N. Zarghami, Y. exosomes, J. Vis. Exp. (2012) e3037, http://dx.doi.org/10.3791/3037.
Hanifehpour, M. Samiei, M. Kouhi, K. Nejati-Koshki, Liposome: classification, [31] A.D. Bangham, M.M. Standish, J.C. Watkins, Diffusion of univalent ions
preparation, and applications, Nanoscale Res. Lett. 8 (2013), http://dx.doi. across the lamellae of swollen phospholipids, J. Mol. Biol. 13 (1965)
org/10.1186/1556-276X-8-102, 102. 238–252, http://dx.doi.org/10.1016/s0022-2836(65)80093-6.
14 L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984

[32] S. Amrani, M. Tabrizian, Characterization of nanoscale loaded liposomes [53] R.R. Sawant, V.P. Torchilin, Challenges in development of targeted liposomal
produced by 2D hydrodynamic flow focusing, ACS Biomater. Sci. Eng. 4 therapeutics, AAPS J. 14 (2012) 303–315, http://dx.doi.org/10.1208/s12248-
(2018) 502–513, http://dx.doi.org/10.1021/acsbiomaterials.7b00572. 012-9330-0.
[33] M.C. Taira, N.S. Chiaramoni, K.M. Pecuch, S. Alonso-Romanowski, Stability of [54] M. Riaz, Liposomes preparation methods, Pak. J. Pharm. Sci. 9 (1996) 65–77.
liposomal formulations in physiological conditions for oral drug delivery, [55] B. Roy, P. Guha, R. Bhattarai, P. Nahak, G. Karmakar, P. Chettri, A.K. Panda,
Drug Deliv. 11 (2004) 123–128, http://dx.doi.org/10.1080/ Influence of lipid composition, pH, and temperature on physicochemical
10717540490280769. properties of liposomes with curcumin as model drug, J. Oleo Sci. 65 (2016)
[34] S. Sugahara, M. Kajiki, H. Kuriyama, T. Kobayashi, Complete regression of 399–411, http://dx.doi.org/10.5650/jos.ess15229.
xenografted human carcinomas by a paclitaxel-carboxymethyl dextran [56] A. Bochot, E. Fattal, Liposomes for intravitreal drug delivery: a state of the
conjugate (AZ10992), J. Control. Release 117 (2006) 40–50, http://dx.doi. art, J. Control. Release 161 (2012) 628–634, http://dx.doi.org/10.1016/j.
org/10.1016/j.jconrel.2006.10.009. jconrel.2012.01.019.
[35] M. Çağdaş, A.D. Sezer, S. Bucak, Liposomes as potential drug carrier systems [57] N. Payne, P. Timmins, C. Ambrose, M. Ward, F. Ridgway, Proliposomes - a
for drug delivery, application of nanotechnology in drug delivery, Ali demir novel solution to an old problem, J. Pharm. Sci. 75 (1986) 325–329, http://
sezer, IntechOpen (2014), http://dx.doi.org/10.5772/58459. dx.doi.org/10.1002/jps.2600750402.
[36] K. Knop, R. Hoogenboom, D. Fischer, U.S. Schubert, Poly(ethylene glycol) in [58] W. Rojanarat, N. Changsan, E. Tawithong, S. Pinsuwan, H.-K. Chan, T.
drug delivery: pros and cons as well as potential alternatives, Angew. Chem. Srichana, Isoniazid proliposome powders for inhalation-preparation,
Int. Ed. Engl. 49 (2010) 6288–6308, http://dx.doi.org/10.1002/anie. characterization and cell culture studies, Int. J. Mol. Sci. 12 (2011)
200902672. 4414–4434, http://dx.doi.org/10.3390/ijms12074414.
[37] Y. Fang, J. Xue, S. Gao, A. Lu, D. Yang, H. Jiang, Y. He, K. Shi, Cleavable [59] I. Khan, S. Yousaf, S. Subramanian, O. Korale, M.A. Alhnan, W. Ahmed, K.M.G.
PEGylation: a strategy for overcoming the “PEG dilemma” in efficient drug Taylor, A. Elhissi, Proliposome powders prepared using a slurry method for
delivery, Drug Deliv. 24 (2017) 22–32, http://dx.doi.org/10.1080/10717544. the generation of beclometasone dipropionate liposomes, Int. J. Pharm. 496
2017.1388451. (2015) 342–350, http://dx.doi.org/10.1016/j.ijpharm.2015.10.002.
[38] T.M. Allen, Ligand-targeted therapeutics in anticancer therapy, Nat. Rev. [60] B. Kapoor, R. Gupta, M. Gulati, S.K. Singh, R. Khursheed, M. Gupta, The Why,
Cancer 2 (2002) 750–763, http://dx.doi.org/10.1038/nrc903. Who Where, How, and what of the vesicular delivery systems, Adv. Colloid
[39] H. Hatakeyama, H. Akita, H. Harashima, The Polyethyleneglycol Dilemma: Interface Sci. 271 (2019), http://dx.doi.org/10.1016/j.cis.2019.07.006, UNSP
Advantage and Disadvantage of PEGylation of Liposomes for Systemic Genes 101985.
and Nucleic Acids Delivery to Tumors, Biol. Pharm. Bull. 36 (2013) 892–899, [61] R.N. Davidson, L. Di Martino, L. Gradoni, R. Giacchino, R. Russo, G.B. Gaeta, R.
http://dx.doi.org/10.1248/bpb.b13-00059. Pempinello, S. Scott, F. Raimondi, A. Cascio, Liposomal amphotericin B
[40] M. Kanamala, B.D. Palmer, S.M. Jamieson, W.R. Wilson, Z. Wu, Dual (AmBisome) in Mediterranean visceral leishmaniasis: a multi-centre trial, Q.
pH-sensitive liposomes with low pH-triggered sheddable PEG for enhanced J. Med. 87 (1994) 75–81.
tumor-targeted drug delivery, Nanomedicine. 14 (2019) 1971–1989, http:// [62] I.M. Hann, H.G. Prentice, Lipid-based amphotericin B: a review of the last 10
dx.doi.org/10.2217/nnm-2018-0510. years of use, Int. J. Antimicrob. Agents 17 (2001) 161–169, http://dx.doi.org/
[41] N. Murthy, J. Campbell, N. Fausto, A.S. Hoffman, P.S. Stayton, Design and 10.1016/s0924-8579(00)00341-1.
synthesis of pH-responsive polymeric carriers that target uptake and [63] Z.G. Chen, Small-molecule delivery by nanoparticles for anticancer therapy,
enhance the intracellular delivery of oligonucleotides, J. Control. Release 89 Trends Mol. Med. 16 (2010) 594–602, http://dx.doi.org/10.1016/j.molmed.
(2003) 365–374, http://dx.doi.org/10.1016/s0168-3659(03)00099-3. 2010.08.001.
[42] M. Mohamed, A.S. Abu Lila, T. Shimizu, E. Alaaeldin, A. Hussein, H.A. Sarhan, [64] M.K. Riaz, M.A. Riaz, X. Zhang, C. Lin, K.H. Wong, X. Chen, G. Zhang, A. Lu, Z.
J. Szebeni, T. Ishida, PEGylated liposomes: immunological responses, Sci. Yang, Surface functionalization and targeting strategies of liposomes in solid
Technol. Adv. Mater. 20 (2019) 710–724, http://dx.doi.org/10.1080/ tumor therapy: a review, Int. J. Mol. Sci. 19 (2018), http://dx.doi.org/10.
14686996.2019.1627174. 3390/ijms19010195.
[43] M. Chen, F. Song, Y. Liu, J. Tian, C. Liu, R. Li, Q. Zhang, A dual pH-sensitive [65] L. Belfiore, D.N. Saunders, M. Ranson, K.J. Thurecht, G. Storm, K.L. Vine,
liposomal system with charge-reversal and NO generation for overcoming Towards clinical translation of ligand-functionalized liposomes in targeted
multidrug resistance in cancer, Nanoscale 11 (2019) 3814–3826, http://dx. cancer therapy: challenges and opportunities, J. Control. Release 277 (2018)
doi.org/10.1039/c8nr06218h. 1–13, http://dx.doi.org/10.1016/j.jconrel.2018.02.040.
[44] T. Terada, M. Iwai, S. Kawakami, F. Yamashita, M. Hashida, Novel PEG-matrix [66] J.D. Patel, R. O’Carra, J. Jones, J.G. Woodward, R.J. Mumper, Preparation and
metalloproteinase-2 cleavable peptide-lipid containing galactosylated characterization of nickel nanoparticles for binding to his-tag proteins and
liposomes for hepatocellular carcinoma-selective targeting, J. Control. antigens, Pharm. Res. 24 (2007) 343–352, http://dx.doi.org/10.1007/s11095-
Release 111 (2006) 333–342, http://dx.doi.org/10.1016/j.jconrel.2005.12. 006-9154-7.
023. [67] J. Ye, E. Liu, Z. Yu, X. Pei, S. Chen, P. Zhang, M.-C. Shin, J. Gong, H. He, V.C.
[45] P.S. Kulkarni, M.K. Haldar, R.R. Nahire, P. Katti, A.H. Ambre, W.W. Muhonen, Yang, CPP-Assisted Intracellular Drug Delivery, What Is Next? Int. J. Mol. Sci.
J.B. Shabb, S.K.R. Padi, R.K. Singh, P.P. Borowicz, D.K. Shrivastava, K.S. Katti, K. 17 (2016), http://dx.doi.org/10.3390/ijms17111892.
Reindl, B. Guo, S. Mallik, MMP-9 responsive PEG cleavable nanovesicles for [68] S.R. Benhabbour, J.C. Luft, D. Kim, A. Jain, S. Wadhwa, M.C. Parrott, R. Liu, J.M.
efficient delivery of chemotherapeutics to pancreatic Cancer, Mol. Pharm. 11 DeSimone, R.J. Mumper, In vitro and in vivo assessment of targeting
(2014) 2390–2399, http://dx.doi.org/10.1021/mp500108p. lipid-based nanoparticles to the epidermal growth factor-receptor (EGFR)
[46] H. Hatakeyama, H. Akita, K. Kogure, M. Oishi, Y. Nagasaki, Y. Kihira, M. Ueno, using a novel Heptameric ZEGFR domain, J. Control. Release 158 (2012)
H. Kobayashi, H. Kikuchi, H. Harashima, Development of a novel systemic 63–71, http://dx.doi.org/10.1016/j.jconrel.2011.10.013.
gene delivery system for cancer therapy with a tumor-specific cleavable [69] S. Fathi, A.K. Oyelere, Liposomal drug delivery systems for targeted cancer
PEG-lipid, Gene Ther. 14 (2007) 68–77, http://dx.doi.org/10.1038/sj.gt. therapy: is active targeting the best choice? Future Med. Chem. 8 (2016)
3302843. 2091–2112, http://dx.doi.org/10.4155/fmc-2016-0135.
[47] H. Xu, F. Ye, M. Hu, P. Yin, W. Zhang, Y. Li, X. Yu, Y. Deng, Influence of [70] H. Xu, J. Paxton, J. Lim, Y. Li, W. Zhang, L. Duxfield, Z. Wu, Development of
phospholipid types and animal models on the accelerated blood clearance high-content gemcitabine PEGylated liposomes and their cytotoxicity on
phenomenon of PEGylated liposomes upon repeated injection, Drug Deliv. drug-resistant pancreatic tumour cells, Pharm. Res. 31 (2014) 2583–2592,
22 (2015) 598–607, http://dx.doi.org/10.3109/10717544.2014.885998. http://dx.doi.org/10.1007/s11095-014-1353-z.
[48] T. Ishida, M. Harada, X.Y. Wang, M. Ichihara, K. Irimura, H. Kiwada, [71] N. Ding, Y. Wang, W. Chu, T. Yin, J. Gou, H. He, Y. Zhang, X. Tang, X. Wang, Y.
Accelerated blood clearance of PEGylated liposomes following preceding Wang, Improving plasma stability and antitumor effect of gemcitabine via
liposome injection: effects of lipid dose and PEG surface-density and chain PEGylated liposome prepared by active drug loading, J. Drug Deliv. Sci.
length of the first-dose liposomes, J. Control. Release 105 (2005) 305–317, Technol. (2020) 101538, http://dx.doi.org/10.1016/j.jddst.2020.101538.
http://dx.doi.org/10.1016/j.jconrel.2005.04.003. [72] A. Chaudhury, S. Das, R.F.S. Lee, K.-B. Tan, W.-K. Ng, R.B.H. Tan, G.N.C. Chiu,
[49] P.H. Kierstead, H. Okochi, V.J. Venditto, T.C. Chuong, S. Kivimae, J.M.J. Fréchet, Lyophilization of cholesterol-free PEGylated liposomes and its impact on
F.C. Szoka, The effect of polymer backbone chemistry on the induction of the drug loading by passive equilibration, Int. J. Pharm. 430 (2012) 167–175,
accelerated blood clearance in polymer modified liposomes, J. Control. http://dx.doi.org/10.1016/j.ijpharm.2012.04.036.
Release 213 (2015) 1–9, http://dx.doi.org/10.1016/j.jconrel.2015.06.023. [73] C. Tikshdeep, A. Sonia, P. Bharat, C. Abhishek, Liposome drug delivery: a
[50] R. Saadati, S. Dadashzadeh, Z. Abbasian, H. Soleimanjahi, Accelerated blood review, IJPCS. 1 (2012) 1103–1113.
clearance of PEGylated PLGA nanoparticles following repeated injections: [74] Z. Wu, W. Zhou, C. Pang, W. Deng, C. Xu, X. Wang, Multifunctional
effects of polymer dose, PEG coating, and encapsulated anticancer drug, chitosan-based coating with liposomes containing laurel essential oils and
Pharm. Res. 30 (2013) 985–995, http://dx.doi.org/10.1007/s11095-012- nanosilver for pork preservation, Food Chem. 295 (2019) 16–25, http://dx.
0934-y. doi.org/10.1016/j.foodchem.2019.05.114.
[51] Y. Su, W. Tang, Y. Song, C. Wang, Q. Tian, X. Wang, J. Quan, B. Li, S. Wang, Y. [75] S. Ghanbarzadeh, H. Valizadeh, P. Zakeri-Milani, The effects of lyophilization
Deng, Mixed PEGylated surfactant modifying system decrease the on the physico-chemical stability of sirolimus liposomes, Adv. Pharm. Bull. 3
accelerated blood clearance phenomenon of nanoemulsions in rats, Asian J. (2013) 25–29, http://dx.doi.org/10.5681/apb.2013.005.
Pharm. Sci. 12 (2017) 28–36, http://dx.doi.org/10.1016/j.ajps.2016.07.003. [76] K.-I. Izutsu, Applications of freezing and freeze-drying in pharmaceutical
[52] T. Ishihara, T. Maeda, H. Sakamoto, N. Takasaki, M. Shigyo, T. Ishida, H. formulations, Adv. Exp. Med. Biol. 1081 (2018) 371–383, http://dx.doi.org/
Kiwada, Y. Mizushima, T. Mizushima, Evasion of the accelerated blood 10.1007/978-981-13-1244-1 20.
clearance phenomenon by coating of nanoparticles with various hydrophilic [77] S. Franzé, F. Selmin, E. Samaritani, P. Minghetti, F. Cilurzo, Lyophilization of
polymers, Biomacromolecules. 11 (2010) 2700–2706, http://dx.doi.org/10. liposomal formulations: still necessary, still challenging, Pharmaceutics. 10
1021/bm100754e. (2018), http://dx.doi.org/10.3390/pharmaceutics10030139.
L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984 15

[78] S. Matsumoto, M. Kohda, S.-I. Murata, Preparation of lipid vesicles on the [103] A.D. Bangham, M.M. Standish, G. Weissmann, The action of steroids and
basis of a technique for providing W/O/W emulsions, J. Colloid Interface Sci. streptolysin S on the permeability of phospholipid structures to cations, J.
62 (1977) 149–157, http://dx.doi.org/10.1016/0021-9797(77)90076-5. Mol, Biol. 13 (1965), http://dx.doi.org/10.1016/S0022-2836(65)80094-8,
[79] T. Wang, Y. Deng, Y. Geng, Z. Gao, J. Zou, Z. Wang, Preparation of submicron 253-IN28.
unilamellar liposomes by freeze-drying double emulsions, BBA - [104] J. Brunner, P. Skrabal, H. Hauser, Single bilayer vesicles prepared without
Biomembranes 1758 (2006) 222–231, http://dx.doi.org/10.1016/j.bbamem. sonication, Physico-chemical properties, Biochim. Biophys. Acta. 455 (1976)
2006.01.023. 322–331, http://dx.doi.org/10.1016/0005-2736(76)90308-4.
[80] S. Vemuri, C.T. Rhodes, Preparation and characterization of liposomes as [105] M. Winterhalter, D.D. Lasic, Liposome stability and formation: experimental
therapeutic delivery systems: a review, Pharm. Acta Helv. 70 (1995) 95–111, parameters and theories on the size distribution, Chem. Phys. Lipids 64
http://dx.doi.org/10.1016/0031-6865(95)00010-7. (1993) 35–43, http://dx.doi.org/10.1016/0009-3084(93)90056-9.
[81] A. Sharma, U.S. Sharma, Liposomes in drug delivery: progress and [106] W. Jiskoot, T. Teerlink, E.C. Beuvery, D.J.A. Crommelin, Preparation of
limitations, Int. J. Pharm. 154 (1997) 123–140, http://dx.doi.org/10.1016/ liposomes via detergent removal from mixed micelles by dilution, Pharm.
S0378-5173(97)00135-X. Weekbl. Sci. 8 (1986) 259–265, http://dx.doi.org/10.1007/BF01960070.
[82] A. Jesorka, O. Orwar, Liposomes: technologies and analytical applications, [107] C. Jaafar-Maalej, C. Charcosset, H. Fessi, A new method for liposome
Annu. Rev. Anal. Chem. (Palo Alto Calif). 1 (2008) 801–832, http://dx.doi.org/ preparation using a membrane contactor, J. Liposome Res. 21 (2011)
10.1146/annurev.anchem.1.031207.112747. 213–220, http://dx.doi.org/10.3109/08982104.2010.517537.
[83] C. Has, P. Sunthar, A comprehensive review on recent preparation [108] A. Laouini, C. Charcosset, H. Fessi, R.G. Holdich, G.T. Vladisavljević,
techniques of liposomes, J. Liposome Res. (2019) 1–30, http://dx.doi.org/10. Preparation of liposomes: a novel application of microengineered
1080/08982104.2019.1668010. membranes - investigation of the process parameters and application to the
[84] A. Wagner, K. Vorauer-Uhl, Liposome technology for industrial purposes, j encapsulation of vitamin E, RSC Adv. 3 (2013) 4985–4994, http://dx.doi.org/
drug, Deliv. 2011 (2011) 9, http://dx.doi.org/10.1155/2011/591325. 10.1039/C3RA23411H.
[85] B. Maherani, E. Arab-Tehrany, M.R. Mozafari, C. Gaiani, M. Linder, [109] S.G.M. Ong, M. Chitneni, K.S. Lee, L.C. Ming, K.H. Yuen, Evaluation of
Liposomes: A Review of Manufacturing Techniques and Targeting Strategies, extrusion technique for nanosizing liposomes, Pharmaceutics 8 (2016),
Curr. Nanosci. 7 (2011) 436–452, http://dx.doi.org/10.2174/ http://dx.doi.org/10.3390/pharmaceutics8040036.
157341311795542453. [110] S.M. Mortazavi, M.R. Mohammadabadi, K. Khosravi-Darani, M.R. Mozafari,
[86] C. Webb, S. Khadke, S.T. Schmidt, C.B. Roces, N. Forbes, G. Berrie, Y. Perrie, Preparation of liposomal gene therapy vectors by a scalable method without
The Impact of Solvent Selection: Strategies to Guide the Manufacturing of using volatile solvents or detergents, J. Biotechnol. 129 (2007) 604–613,
Liposomes Using Microfluidics, Pharmaceutics. 11 (2019) 653, http://dx.doi. http://dx.doi.org/10.1016/j.jbiotec.2007.02.005.
org/10.3390/pharmaceutics11120653. [111] M.R. Mozafari, Liposomes: an overview of manufacturing techniques, Cell.
[87] C.C. Beh, R. Mammucari, N.R. Foster, Formation of nanocarrier systems by Mol. Biol. Lett. 10 (2005) 711–719.
dense gas processing, Langmuir 30 (2014) 11046–11054, http://dx.doi.org/ [112] M.R. Mozafari, C.J. Reed, C. Rostron, Development of non-toxic liposomal
10.1021/la502594k. formulations for gene and drug delivery to the lung, Technology and Health
[88] S. Kunastitchai, L. Pichert, N. Sarisuta, B.W. Mueller, Application of aerosol Care: Official Journal of the European Society for Engineering and Medicine.
solvent extraction system (ASES) process for preparation of liposomes in a 10 (2002) 342–344.
dry and reconstitutable form, Int. J. Pharm. 316 (2006) 93–101, http://dx. [113] M. Danaei, M. Dehghankhold, S. Ataei, F. Hasanzadeh Davarani, R.
doi.org/10.1016/j.ijpharm.2006.02.051. Javanmard, A. Dokhani, S. Khorasani, M.R. Mozafari, Impact of particle size
[89] F. Szoka, D. Papahadjopoulos, Procedure for preparation of liposomes with and polydispersity index on the clinical applications of lipidic nanocarrier
large internal aqueous space and high capture by reverse-phase systems, Pharmaceutics. 10 (2018) 57, http://dx.doi.org/10.3390/
evaporation, Proc. Natl. Acad. Sci. U S A. 75 (1978) 4194–4198. pharmaceutics10020057.
[90] S.M. Gruner, R.P. Lenk, A.S. Janoff, M.J. Ostro, Novel multilayered lipid [114] R. Fanciullino, S. Mollard, F. Correard, S. Giacometti, C. Serdjebi, A. Iliadis, J.
vesicles: comparison of physical characteristics of multilamellar liposomes Ciccolini, Biodistribution, tumor uptake and efficacy of 5-FU-loaded
and stable plurilamellar vesicles, Biochemistry. 24 (1985) 2833–2842, liposomes: why size matters, Pharm. Res. 31 (2014) 2677–2684, http://dx.
http://dx.doi.org/10.1021/bi00333a004. doi.org/10.1007/s11095-014-1364-9.
[91] C. Pidgeon, S. McNeely, T. Schmidt, J.E. Johnson, Multilayered vesicles [115] D.R. Khan, E.M. Rezler, J. Lauer-Fields, G.B. Fields, Effects of drug
prepared by reverse-phase evaporation: liposome structure and optimum hydrophobicity on liposomal stability, Chem. Biol. Drug Des. 71 (2008) 3–7,
solute entrapment, Biochemistry. 26 (1987) 17–29, http://dx.doi.org/10. http://dx.doi.org/10.1111/j.1747-0285.2007.00610.x.
1021/bi00375a004. [116] D.T. Birnbaum, J.D. Kosmala, D.B. Henthorn, L. Brannon-Peppas, Controlled
[92] D. Deamer, A.D. Bangham, Large volume liposomes by an ether vaporization release of beta-estradiol from PLAGA microparticles: the effect of organic
method, Biochim. Biophys. Acta 443 (1976) 629–634, http://dx.doi.org/10. phase solvent on encapsulation and release, J. Control. Release 65 (2000)
1016/0005-2736(76)90483-1. 375–387, http://dx.doi.org/10.1016/s0168-3659(99)00219-9.
[93] D.W. Deamer, Preparation and properties of ether-injection liposomes, Ann. [117] A.M. Seddon, P. Curnow, P.J. Booth, Membrane proteins, lipids and
N. Y. Acad. Sci. 308 (1978) 250–258, http://dx.doi.org/10.1111/j.1749-6632. detergents: not just a soap opera, Biochim. Biophys. Acta 1666 (2004)
1978.tb22027.x. 105–117, http://dx.doi.org/10.1016/j.bbamem.2004.04.011.
[94] C. Jaafar-Maalej, R. Diab, V. Andrieu, A. Elaissari, H. Fessi, Ethanol injection [118] M. Stievano, N. Elvassore, High-pressure density and vapor-liquid
method for hydrophilic and lipophilic drug-loaded liposome preparation, J. equilibrium for the binary systems carbon dioxide-ethanol, carbon
Liposome Res. 20 (2010) 228–243, http://dx.doi.org/10.3109/ dioxide-acetone and carbon dioxide-dichloromethane, J. Supercrit. Fluids 33
08982100903347923. (2005) 7–14, http://dx.doi.org/10.1016/j.supflu.2004.04.003.
[95] V. Pereno, D. Carugo, L. Bau, E. Sezgin, J. Bernardino de la Serna, C. Eggeling, [119] L. Lesoin, C. Crampon, O. Boutin, E. Badens, Development of a continuous
E. Stride, Electroformation of giant unilamellar vesicles on stainless steel dense gas process for the production of liposomes, J. Supercrit. Fluids 60
electrodes, ACS Omega 2 (2017) 994–1002, http://dx.doi.org/10.1021/ (2011) 51–62, http://dx.doi.org/10.1016/j.supflu.2011.04.018.
acsomega.6b00395. [120] L. Lesoin, C. Crampon, O. Boutin, E. Badens, Preparation of liposomes using
[96] M. Alavi, N. Karimi, M. Safaei, Application of various types of liposomes in the supercritical anti-solvent (SAS) process and comparison with a
drug delivery systems, Adv. Pharm. Bull. 7 (2017) 3–9, http://dx.doi.org/10. conventional method, J. Supercrit. Fluids 57 (2011) 162–174, http://dx.doi.
15171/apb.2017.002. org/10.1016/j.supflu.2011.01.006.
[97] L.A. Bagatolli, T. Parasassi, E. Gratton, Giant phospholipid vesicles: [121] L.A. Meure, R. Knott, N.R. Foster, F. Dehghani, The depressurization of an
comparison among the whole lipid sample characteristics using different expanded solution into aqueous media for the bulk production of liposomes,
preparation methods: A two photon fluorescence microscopy study, Chem. Langmuir 25 (2009) 326–337, http://dx.doi.org/10.1021/la802511a.
Phys. Lipids 105 (2000) 135–147, http://dx.doi.org/10.1016/S0009- [122] L. Zhao, Y. Wei, W. Li, Y. Liu, Y. Wang, X. Zhong, Y. Yu, Solid dispersion and
3084(00)00118-3. effervescent techniques used to prepare docetaxel liposomes for
[98] A. Jahn, W.N. Vreeland, D.L. DeVoe, L.E. Locascio, M. Gaitan, Microfluidic lung-targeted delivery system: in vitro and in vivo evaluation, J. Drug
directed formation of liposomes of controlled size, Langmuir. 23 (2007) Target. 19 (2011) 171–178, http://dx.doi.org/10.3109/
6289–6293, http://dx.doi.org/10.1021/la070051a. 10611861003801859.
[99] R.R. Hood, C. Shao, D.M. Omiatek, W.N. Vreeland, D.L. DeVoe, Microfluidic [123] K. Otake, T. Imura, H. Sakai, M. Abe, Development of a new preparation
synthesis of PEG- and folate-conjugated liposomes for one-step formation of method of liposomes using supercritical carbon dioxide, Langmuir 17 (2001)
targeted stealth nanocarriers, Pharm. Res. 30 (2013) 1597–1607, http://dx. 3898–3901, http://dx.doi.org/10.1021/la010122k.
doi.org/10.1007/s11095-013-0998-3. [124] E. Badens, C. Magnan, G. Charbit, Microparticles of soy lecithin formed by
[100] S.M. Phapal, P. Sunthar, Influence of micro-mixing on the size of liposomes supercritical processes, Biotechnol. Bioeng. 72 (2001) 194–204, http://dx.
self-assembled from miscible liquid phases, Chem. Phys. Lipids 172 (2013) doi.org/10.1002/1097-0290(20000120)72:2<194::AID-BIT8>3.0.CO;2-L.
20–30, http://dx.doi.org/10.1016/j.chemphyslip.2013.04.006. [125] L. Frederiksen, K. Anton, P. vanHoogevest, H.R. Keller, H. Leuenberger,
[101] S. Sugiura, T. Kuroiwa, T. Kagota, M. Nakajima, S. Sato, S. Mukataka, P. Walde, Preparation of liposomes encapsulating water-soluble compounds using
S. Ichikawa, Novel method for obtaining homogeneous giant vesicles from a supercritical carbon dioxide, J. Pharm. Sci. 86 (1997) 921–928, http://dx.doi.
monodisperse water-in-oil emulsion prepared with a microfluidic device, org/10.1021/js960403q.
Langmuir. 24 (2008) 4581–4588, http://dx.doi.org/10.1021/la703509r. [126] I.E. Santo, R. Campardelli, E.C. Albuquerque, S.V. de Melo, G. Della Porta, E.
[102] J.C. Stachowiak, D.L. Richmond, T.H. Li, F. Brochard-Wyart, D.A. Fletcher, Reverchon, Liposomes preparation using a supercritical fluid assisted
Inkjet formation of unilamellar lipid vesicles for cell-like encapsulation, Lab continuous process, Chem. Eng. J. 249 (2014) 153–159, http://dx.doi.org/10.
Chip 9 (2009) 2003–2009, http://dx.doi.org/10.1039/b904984c. 1016/j.cej.2014.03.099.
16 L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984

[127] P. Trucillo, R. Campardelli, M. Scognamiglio, E. Reverchon, Control of [151] Z. Wen, X. You, B. Liu, Z. Zheng, Y. Pu, L. Jiang, Q. Li, Formation of atractylone
liposomes diameter at micrometric and nanometric level using a liposomes by rapid expansion from supercritical to surfactant solution,
supercritical assisted technique, J. CO2 Util. 32 (2019) 119–127, http://dx. Asia-Pac, J. Chem. Eng. 6 (2011) 624–630, http://dx.doi.org/10.1002/apj.462.
doi.org/10.1016/j.jcou.2019.04.014. [152] S. Xu, B. Zhao, D. He, Synthesis of highly dispersed nanoscaled CoQ(10)
[128] P. Chakravarty, A. Famili, K. Nagapudi, M.A. Al-Sayah, Using supercritical liposome by supercritical fluid, Mater. Lett. 142 (2015) 283–286, http://dx.
fluid technology as a green alternative during the preparation of drug doi.org/10.1016/j.matlet.2014.12.070.
delivery systems, Pharmaceutics. 11 (2019) 629, http://dx.doi.org/10.3390/ [153] Q. Zhang, C. Ou, S. Ye, X. Song, S. Luo, Construction of nanoscale liposomes
pharmaceutics11120629. loaded with melatonin via supercritical fluid technology, J. Microencapsul.
[129] Ž. Knez, M. Leitgeb, M. Primožič, Enzymatic Reactions in Supercritical Fluids, 34 (2017) 687–698, http://dx.doi.org/10.1080/02652048.2017.1376001.
in: High Pressure Fluid Technology for Green Food Processing, Springer, [154] P. Trucillo, R. Campardelli, E. Reverchon, A versatile supercritical assisted
Cham, 2015, pp. 185–215, http://dx.doi.org/10.1007/978-3-319-10611-3 6. process for the one-shot production of liposomes, J. Supercrit. Fluids 146
[130] M. Primožič, M. Čolnik, Ž. Knez, M. Leitgeb, Advantages and disadvantages of (2019) 136–143, http://dx.doi.org/10.1016/j.supflu.2019.01.015.
using SC CO2 for enzyme release from halophilic fungi, J. Supercrit. Fluids [155] P. Trucillo, R. Campardelli, E. Reverchon, Supercritical CO2 assisted
143 (2019) 286–293, http://dx.doi.org/10.1016/j.supflu.2018.09.001. liposomes formation: optimization of the lipidic layer for an efficient
[131] L. Zhao, F. Temelli, Preparation of anthocyanin-loaded liposomes using an hydrophilic drug loading, J. CO2 Util. 18 (2017) 181–188, http://dx.doi.org/
improved supercritical carbon dioxide method, Innov. Food Sci. Emerg. 10.1016/j.jcou.2017.02.001.
Technol. 39 (2017) 119–128, http://dx.doi.org/10.1016/j.ifset.2016.11.013. [156] R. Campardelli, P. Trucillo, E. Reverchon, Supercritical assisted process for
[132] N. Ventosa, S. Sala, J. Veciana, DELOS process: a crystallization technique the efficient production of liposomes containing antibiotics for ocular
using compressed fluids: 1. Comparison to the GAS crystallization method, J. delivery, J. CO2 Util. 25 (2018) 235–241, http://dx.doi.org/10.1016/j.jcou.
Supercrit. Fluids 26 (2003) 33–45, http://dx.doi.org/10.1016/S0896- 2018.04.006.
8446(02)00189-4. [157] P. Trucillo, R. Campardelli, B. Aliakbarian, P. Perego, E. Reverchon,
[133] M. Cano-Sarabia, N. Ventosa, S. Sala, C. Patiño, R. Arranz, J. Veciana, Supercritical assisted process for the encapsulation of olive pomace extract
Preparation of uniform rich cholesterol unilamellar nanovesicles using into liposomes, J. Supercrit. Fluids 135 (2018) 152–159, http://dx.doi.org/10.
CO2-Expanded solvents, Langmuir. 24 (2008) 2433–2437, http://dx.doi.org/ 1016/j.supflu.2018.01.018.
10.1021/la7032109. [158] P. Trucillo, R. Campardelli, E. Reverchon, Production of liposomes loaded
[134] O. Erkmen, Effects of dense phase carbon dioxide on vegetative cells, in: with antioxidants using a supercritical CO2 assisted process, Powder
Dense Phase Carbon Dioxide, John Wiley & Sons, Ltd, 2012, pp. 67–97, Technol. 323 (2018) 155–162, http://dx.doi.org/10.1016/j.powtec.2017.10.
http://dx.doi.org/10.1002/9781118243350.ch4. 007.
[135] L. Zhao, F. Temelli, J.M. Curtis, L. Chen, Encapsulation of lutein in liposomes [159] F. Maqbool, P.M. Moyle, K.J. Thurecht, J.R. Falconer, Dispersibility of
using supercritical carbon dioxide, Food Res. Int. 100 (2017) 168–179, phospholipids and their optimization for the efficient production of
http://dx.doi.org/10.1016/j.foodres.2017.06.055. liposomes using supercritical fluid technology, Int. J. Pharm. 563 (2019)
[136] L. Zhao, F. Temelli, L. Chen, Encapsulation of anthocyanin in liposomes using 174–183, http://dx.doi.org/10.1016/j.ijpharm.2019.03.053.
supercritical carbon dioxide: effects of anthocyanin and sterol [160] R. Campardelli, I.E. Santo, E.C. Albuquerque, S.V. de Melo, G. Della Porta, E.
concentrations, J. Funct. Food. 34 (2017) 159–167, http://dx.doi.org/10. Reverchon, Efficient encapsulation of proteins in submicro liposomes using
1016/j.jff.2017.04.021. a supercritical fluid assisted continuous process, J. Supercrit. Fluids 107
[137] W. Situ, X. Song, S. Luo, Y. Liang, A nano-delivery system for bioactive (2016) 163–169, http://dx.doi.org/10.1016/j.supflu.2015.09.007.
ingredients using supercritical carbon dioxide and its release behaviors, [161] P. Trucillo, R. Campardelli, E. Reverchon, Antioxidant loaded emulsions
Food Chem. 228 (2017) 219–225, http://dx.doi.org/10.1016/j.foodchem. entrapped in liposomes produced using a supercritical assisted technique, J.
2017.01.053. Supercrit. Fluids 154 (2019) 104626, http://dx.doi.org/10.1016/j.supflu.
[138] L. Zhao, F. Temelli, J.M. Curtis, L. Chen, Preparation of liposomes using 2019.104626.
supercritical carbon dioxide technology: effects of phospholipids and [162] R. Parhi, P. Suresh, Supercritical fluid technology: a review, JAPST 1 (2013)
sterols, Food Res. Int. 77 (2015) 63–72, http://dx.doi.org/10.1016/j.foodres. 13, http://dx.doi.org/10.14302/issn.2328-0182.japst-12-145.
2015.07.006. [163] G.M. Shashidhar, B. Manohar, Nanocharacterization of liposomes for the
[139] I. Pasquali, R. Bettini, Are pharmaceutics really going supercritical? Int. J. encapsulation of water soluble compounds from Cordyceps sinensis CS1197
Pharm. 364 (2008) 176–187, http://dx.doi.org/10.1016/j.ijpharm.2008.05. by a supercritical gas anti-solvent technique, RSC Adv. 8 (2018)
014. 34634–34649, http://dx.doi.org/10.1039/c8ra07601d.
[140] L. Lesoin, C. Crampon, O. Boutin, E. Badens, Preparation of liposomes using [164] G.M. Shashidhar, G.V. Pravin, B. Manohar, Nano-engineering of liposomes
the supercritical anti-solvent (SAS) process and comparison with a using a supercritical CO2 mediated gas anti-solvent method, RSC Adv. 6
conventional method, J. Supercrit. Fluids 57 (2011) 162–174, http://dx.doi. (2016) S7739–S7750, http://dx.doi.org/10.1039/c6ra09530e.
org/10.1016/j.supflu.2011.01.006. [165] R. Koynova, B. Tenchov, Recent progress in liposome production, relevance
[141] F. Xia, D. Hu, H. Jin, Y. Zhao, J. Liang, Preparation of lutein proliposomes by to drug delivery and nanomedicine, recent pat, Nanotechnology. 9 (2015)
supercritical anti-solvent technique, Food Hydrocoll. 26 (2012) 456–463, 86–93, http://dx.doi.org/10.2174/187221050902150819151721.
http://dx.doi.org/10.1016/j.foodhyd.2010.11.014. [166] Q. Fan, Y. Zhang, X. Hou, Z. Li, K. Zhang, Q. Shao, N. Feng, Improved oral
[142] X. Fei, J. Heyang, Z. Yaping, G. Xinqiu, Supercritical antisolvent-based bioavailability of notoginsenoside R1 with sodium glycocholate-mediated
technology for preparation of vitamin D-3 proliposome and its liposomes: preparation by supercritical fluid technology and evaluation
characteristics, Chin. J. Chem. Eng. 19 (2011) 1039–1046. in vitro and in vivo, Int. J. Pharm. 552 (2018) 360–370, http://dx.doi.org/10.
[143] F. Macibool, P.M. Moyle, M.S.A. Tan, K.J. Thurecht, J.R. Falconer, Preparation 1016/j.ijpharm.2018.10.005.
of albendazole-loaded liposomes by supercritical carbon dioxide processing, [167] L. Zhao, F. Temelli, Preparation of liposomes using supercritical carbon
Artif. Cell. Nanomed. Biotechnol. 46 (2018) S1186–S1192, http://dx.doi.org/ dioxide via depressurization of the supercritical phase, J. Food Eng. 158
10.1080/21691401.2018.1536059. (2015) 104–112, http://dx.doi.org/10.1016/j.jfoodeng.2015.03.004.
[144] C. Magnan, E. Badens, N. Commenges, G. Charbit, Soy lecithin micronization [168] L. Li, X. An, A novel combined method of thin-film evaporation and a
by precipitation with a compressed fluid antisolvent — influence of process supercritical carbon dioxide technique to prepare a fluorescent
parameters, J. Supercrit. Fluids 19 (2000) 69–77, http://dx.doi.org/10.1016/ siRNA-liposome, RSC Adv. 6 (2016) 92115–92119, http://dx.doi.org/10.
S0896-8446(00)00076-0. 1039/c6ra19751e.
[145] A.Z. Hezave, F. Esmaeilzadeh, Micronization of drug particles via RESS [169] D. Li, X. An, Y. Mu, A liposomal hydrogel with enzyme triggered release for
process, J. Supercrit. Fluids 52 (2010) 84–98, http://dx.doi.org/10.1016/j. infected wound, Chem. Phys. Lipids 223 (2019), http://dx.doi.org/10.1016/j.
supflu.2009.09.006. chemphyslip.2019.104783, UNSP 104783.
[146] null York, Strategies for particle design using supercritical fluid [170] S. Xu, X. An, Preparation, microstructure and function for injectable
technologies, Pharm. Sci. Technol. Today 2 (1999) 430–440, http://dx.doi. liposome-hydrogels, Colloid Surf, A-Physicochem. Eng. Asp. 560 (2019)
org/10.1016/s1461-5347(99)00209-6. 20–25, http://dx.doi.org/10.1016/j.colsurfa.2018.09.037.
[147] F. Sharifi, R. Zhou, C. Lim, A. Jash, A. Abbaspourrad, S.S.H. Rizvi, Generation of [171] Y. Liu, X. An, Preparation, microstructure and function of liposome with light
liposomes using a supercritical carbon dioxide eductor vacuum system: responsive switch, Colloid Surf. B-Biointerfaces. 178 (2019) 238–244, http://
optimization of process variables, J. CO2 Util. 29 (2019) 163–171, http://dx. dx.doi.org/10.1016/j.colsurfb.2018.10.068.
doi.org/10.1016/j.jcou.2018.12.011. [172] J. Jia, N. Song, Y. Gai, L. Zhang, Y. Zhao, Release-controlled curcumin
[148] Z. Jiao, X. Wang, S. Han, X. Zha, J. Xia, Preparation of vitamin C liposomes by proliposome produced by ultrasound-assisted supercritical antisolvent
rapid expansion of supercritical solution process: experiments and method, J. Supercrit. Fluids 113 (2016) 150–157, http://dx.doi.org/10.1016/j.
optimization, J. Drug Deliv. Sci. Technol. 51 (2019) 1–6, http://dx.doi.org/10. supflu.2016.03.026.
1016/j.jddst.2019.02.015. [173] J. Jia, K. Zhang, X. Zhou, J. Ma, X. Liu, A. Xiang, F. Ge, Berberine-loaded solid
[149] W. Zhang, Y. Sun, Y. Li, R. Shen, H. Ni, D. Hu, Preparation and influencing proliposomes prepared using solution enhanced dispersion by supercritical
factors of sirolimus liposome by supercritical fluid, Artif. Cells Blood Substit. CO2: sustained release and bioavailability enhancement, J. Drug Deliv. Sci.
Biotechnol. 40 (2012) 62–65, http://dx.doi.org/10.3109/10731199.2011. Technol. 51 (2019) 356–363, http://dx.doi.org/10.1016/j.jddst.2019.03.
585618. 021.
[150] Z. Wen, B. Liu, Z. Zheng, X. You, Y. Pu, Q. Li, Preparation of liposomes [174] L. Zhao, F. Temelli, Preparation of liposomes using a modified supercritical
entrapping essential oil from Atractylodes macrocephala Koidz by modified process via depressurization of liquid phase, J. Supercrit. Fluids 100 (2015)
RESS technique, Chem. Eng. Res. Des. 88 (2010) 1102–1107, http://dx.doi. 110–120, http://dx.doi.org/10.1016/j.supflu.2015.02.022.
org/10.1016/j.cherd.2010.01.020.
L. Maja, K. Željko and P. Mateja / J. of Supercritical Fluids 165 (2020) 104984 17

[175] J.E. Packer, T.F. Slater, R.L. Willson, Direct observation of a free radical [187] T. Imura, T. Gotoh, K. Otake, S. Yoda, Y. Takebayashi, S. Yokoyama, H.
interaction between vitamin E and vitamin C, Nature. 278 (1979) 737–738, Takebayashi, H. Sakai, M. Yuasa, M. Abe, Control of physicochemical
http://dx.doi.org/10.1038/278737a0. properties of liposomes using a supercritical reverse phase evaporation
[176] A.L. Tappel, Will antioxidant nutrients slow aging processes? Geriatrics. 23 method, Langmuir 19 (2003) 2021–2025, http://dx.doi.org/10.1021/
(1968) 97–105. la020589a.
[177] W.-C. Tsai, S.S.H. Rizvi, Simultaneous microencapsulation of hydrophilic and [188] K. Otake, T. Shimomura, T. Goto, T. Imura, T. Furuya, S. Yoda, Y. Takebayashi,
lipophilic bioactives in liposomes produced by an ecofriendly supercritical H. Sakai, M. Abe, Preparation of liposomes using an improved supercritical
fluid process, Food Res. Int. 99 (2017) 256–262, http://dx.doi.org/10.1016/j. reverse phase evaporation method, Langmuir 22 (2006) 2543–2550, http://
foodres.2017.05.029. dx.doi.org/10.1021/la051654u.
[178] H. Nakamura, S. Taguchi, K. Suga, K. Hayashi, H.-S. Jung, H. Umakoshi, [189] K. Aburai, N. Yagi, Y. Yokoyama, H. Okuno, K. Sakai, H. Sakai, K. Sakamoto, M.
Characterization of the physicochemical properties of phospholipid vesicles Abe, Preparation of liposomes modified with lipopeptides using a
prepared in CO2/water systems at high pressure, Biointerphases. 10 (2015), supercritical carbon dioxide reverse-phase evaporation method, J. Oleo Sci.
031005, http://dx.doi.org/10.1116/1.4928722. 60 (2011) 209–215, http://dx.doi.org/10.5650/jos.60.209.
[179] V. Torchilin, V. Weissig (Eds.), Liposomes: A Practical Approach, second [190] K. Otake, T. Shimomura, T. Goto, T. Imura, T. Furuya, S. Yoda, Y. Takebayashi,
edition, Oxford University Press, Oxford, New York, 2003. H. Sakai, M. Abe, One-step preparation of chitosan-coated cationic
[180] I.E. Santo, R. Campardelli, E.C. Albuquerque, S.A.B. Vieira De Melo, E. liposomes by an improved supercritical reverse-phase evaporation method,
Reverchon, G. Della Porta, Liposomes size engineering by combination of Langmuir 22 (2006) 4054–4059, http://dx.doi.org/10.1021/la051662a.
ethanol injection and supercritical processing, J. Pharm. Sci. 104 (2015) [191] G. Yang, Y. Zhao, Y. Zhang, B. Dang, Y. Liu, N. Feng, Enhanced oral
3842–3850, http://dx.doi.org/10.1002/jps.24595. bioavailability of silymarin using liposomes containing a bile salt:
[181] I. Ahmad, S. Akhter, M. Anwar, S. Zafar, R.K. Sharma, A. Ali, F.J. Ahmad, preparation by supercritical fluid technology and evaluation in vitro and in
Supercritical anti-solvent technique assisted synthesis of thymoquinone vivo, Int. J. Nanomed. Nanosurg. 10 (2015), http://dx.doi.org/10.2147/IJN.
liposomes for radioprotection: formulation optimization, in-vitro and S92665.
in-vivo studies, Int. J. Pharm. 523 (2017) 398–409, http://dx.doi.org/10. [192] S. Varona, A. Martin, M. Jose Cocero, Liposomal incorporation of lavandin
1016/j.ijpharm.2017.03.052. essential oil by a thin-film hydration method and by particles from
[182] C. Lim, S.M. Abuzar, P.R. Karn, W. Cho, H.J. Park, C.-W. Cho, S.-J. Hwang, gas-saturated solutions, Ind. Eng. Chem. Res. 50 (2011) 2088–2097, http://
Preparation, Characterization, and In Vivo Pharmacokinetic Study of the dx.doi.org/10.1021/ie102016r.
Supercritical Fluid-Processed Liposomal Amphotericin B, Pharmaceutics. 11 [193] U.S. Kadimi, D.R. Balasubramanian, U.R. Ganni, M. Balaraman, V.
(2019) 589, http://dx.doi.org/10.3390/pharmaceutics11110589. Govindarajulu, In vitro studies on liposomal amphotericin B obtained by
[183] M.E. Wagner, S.S.H. Rizvi, Novel method of niosome generation using supercritical carbon dioxide-mediated process, Nanomed.-Nanotechnol.
supercritical carbon dioxide part I: process mechanics, J. Liposome Res. 25 Biol. Med. 3 (2007) 273–280, http://dx.doi.org/10.1016/j.nano.2007.08.003.
(2015) 334–346, http://dx.doi.org/10.3109/08982104.2015.1039032. [194] R.H. Bridson, R.C.D. Santos, B. Al-Duri, S.M. McAllister, J. Robertson, H.O.
[184] S. Naik, D. Patel, N. Surti, A. Misra, Preparation of PEGylated liposomes of Alpar, The preparation of liposomes using compressed carbon dioxide:
docetaxel using supercritical fluid technology, J. Supercrit. Fluids 54 (2010) strategies, important considerations and comparison with conventional
110–119, http://dx.doi.org/10.1016/j.supflu.2010.02.005. techniques, J. Pharm. Pharmacol. 58 (2006) 775–785, http://dx.doi.org/10.
[185] W.-C. Tsai, S.S.H. Rizvi, Microencapsulation and characterization of 1211/jpp.58.6.0008.
liposomal vesicles using a supercritical fluid process coupled with [195] B. William, P. Noémie, E. Brigitte, P. Géraldine, Supercritical fluid methods:
vacuum-driven cargo loading, Food Res. Int. 96 (2017) 94–102, http://dx.doi. an alternative to conventional methods to prepare liposomes, Chem. Eng. J.
org/10.1016/j.foodres.2017.03.027. 383 (2020) 123106, http://dx.doi.org/10.1016/j.cej.2019.123106.
[186] P. Trucillo, R. Campardelli, E. Reverchon, Operative parameters optimization [196] P. Girotra, S.K. Singh, K. Nagpal, Supercritical fluid technology: a promising
production of liposomes for the encapsulation of hydrophilic compounds approach in pharmaceutical research, Pharm. Dev. Technol. 18 (2013)
using a new supercritical process, Chem. Eng. Trans. 57 (2017) 799–804, 22–38, http://dx.doi.org/10.3109/10837450.2012.726998.
http://dx.doi.org/10.3303/CET1757134.

You might also like