Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

Advanced Drug Delivery Reviews 176 (2021) 113851

Contents lists available at ScienceDirect

Advanced Drug Delivery Reviews


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

Liposome composition in drug delivery design, synthesis,


characterization, and clinical application q
Danielle E. Large, Rudolf G. Abdelmessih, Elizabeth A. Fink, Debra T. Auguste ⇑
Department of Chemical Engineering, Northeastern University, 360 Huntington Ave., Boston, MA 02115, USA

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

Article history: Liposomal drug delivery represents a highly adaptable therapeutic platform for treating a wide range of
Received 23 April 2021 diseases. Natural and synthetic lipids, as well as surfactants, are commonly utilized in the synthesis of
Revised 18 June 2021 liposomal drug delivery vehicles. The molecular diversity in the composition of liposomes enables drug
Accepted 22 June 2021
delivery with unique physiological functions, such as pH response, prolonged blood circulation, and
Available online 2 July 2021
reduced systemic toxicity. Herein, we discuss the impact of composition on liposome synthesis, function,
and clinical utility.
Keywords:
Ó 2021 Elsevier B.V. All rights reserved.
Liposomal drug delivery
Liposome synthesis
Liposome functionality

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2. Liposome synthesis & characterization methodology . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.1. Liposomes classification . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2. Methods of liposomes preparation. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2.1. Thin film hydration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2.2. Reverse-phase evaporation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2
2.2.3. Injection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.2.4. Detergent removal . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.2.5. Dehydration-rehydration . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.2.6. pH jumping . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.2.7. Hydration in a packed bed of colloidal particles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.2.8. Freeze-thaw . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.3. Methods of liposome characterization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.3.1. Drug encapsulation efficiency and release . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.3.2. Size, zeta potential, and stability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3
2.3.3. Pharmacokinetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3. Molecular components of liposomal drug delivery vehicles . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.1. Natural lipids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.1.1. Phospholipids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6
3.1.2. Sphingolipids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.1.3. Sterols . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.1.4. Polysaccharides. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.2. Synthetic lipids . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3.3. Surfactants . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
4. Effect of composition on liposome characteristics & functionality . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7

q
This review is part of the Advanced Drug Delivery Reviews theme issue on
‘‘Transl Drug Delivery”.
⇑ Corresponding author.
E-mail address: d.auguste@northeastern.edu (D.T. Auguste).

https://doi.org/10.1016/j.addr.2021.113851
0169-409X/Ó 2021 Elsevier B.V. All rights reserved.
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

4.1. Drug encapsulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7


4.2. Stability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.3. Surface charge . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.4. Pharmacokinetics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.4.1. Clearance rate and half-life . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
4.4.2. Biodistribution . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
5. Liposomal composition imparts unique functionalities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5.1. pH responsive liposomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5.2. Temperature responsive liposomes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
5.3. Theranostic liposomes. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
6. Liposomal drugs in clinical use and preclinical development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
6.1. Anti-cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
6.2. Pain management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
6.3. Anti-bacterial . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
6.4. Vaccines . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
7. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 11

1. Introduction electroformation in microfluidics, pulsed microfluidic jetting, tran-


sient membrane ejection, continuous droplet interface crossing
Since Alec D. Bangham’s discovery of liposomes in 1965, the encapsulation and stationary phase interdiffusion method [6].
lipid vessel has become a widely utilized vehicle to encapsulate Herein, we discuss the most common methods for bench scale
and deliver molecules to treat a variety of diseases. The primary preparation of liposomes [1,5,6].
component of liposomes are lipids and fatty acids that, due to their
natural occurrence in cell membranes, are considered inherently 2.2.1. Thin film hydration
biocompatible and biodegradable [1]. Structurally, liposomes are The most common method employed for liposome synthesis is
defined by the self-assembly of amphipathic molecules into a thin film hydration [9–12]. In this method, lipids and amphiphilic
bilayer sphere, in which the hydrophilic head groups face the exte- molecules are solubilized and mixed in an organic solvent. The
rior aqueous environment and the hydrocarbon chains assemble mixture is then transferred into a round-bottom flask and the sol-
within the hydrophobic interior. The amphiphilic character of lipo- vent is evaporated using a rotary evaporator under vacuum, leav-
somes makes them ideal drug carriers for molecules of differing ing a thin film of lipids. The thin film is then hydrated in a
polarities. Liposomal encapsulation of drugs reduces systemic tox- solution that may contain one or more hydrophilic drugs that are
icity and improves tolerable dose regimens for anti-cancer, anti- desired to be encapsulated. The temperature of the hydration buf-
bacterial, and anti-fungal therapies [2–4]. The lipid chemistry is fer must be above the gel-liquid phase transition temperature (Tm)
critical for optimizing drug encapsulation, stability, and release, of the lipid. The volume of the aqueous solution used to hydrate
and liposome pharmacokinetics and pharmacodynamics. Herein, the lipid film affects the characteristics of the formed liposomes;
we present a review of the literature focused on the rational design large volumes lead to the formation of MLVs while the rate of
of liposomes based on chemical, mechanical, and physiological hydration determines the efficiency of drug encapsulation [5].
properties. The slower the rate of hydration the higher the encapsulation
efficiency.
2. Liposome synthesis & characterization methodology The size and lamellarity of the vesicles may be controlled by
either extrusion through membranes of specific pore sizes [13] or
2.1. Liposomes classification the use of sonicators, where the frequency of the ultrasonic waves
and the duration of the process determine the size distribution of
Two important characteristics of liposomal vesicles that influ- the fabricated liposomes [14]. A jacketed extruder or water bath
ence drug encapsulation efficiency and circulation time are size may be used to maintain the solution temperature above the Tm
and membrane lamellarity [1,5–7]. The method of synthesis deter- of the lipid if necessary. Although sonication is easier and more
mines the type of liposomes produced. Liposomes are classified as convenient for post-synthesis processing to produce SUVs, espe-
unilamellar vesicles (ULVs) with one bilayer membrane, oligo- cially when large volumes are needed, it results in less uniform
lamellar vesicles (OLVs) with 2–5 bilayer membranes, and multil- liposomes with lower drug encapsulation efficiency compared to
amellar vesicles (MLVs) with five or more bilayer membranes. those produced by extrusion [1]. Sonication may also degrade
ULVs are further categorized into small unilamellar vesicles (20– lipids or drugs encapsulated, partly due to the heat generated dur-
100 nm in diameter, SUVs), large unilamellar vesicles (100 nm- ing the process. Furthermore, drug-loaded SUVs made by thin film
1 mm, LUVs), and giant unilamellar vesicles (>1 mm, GUVs). SUVs hydration followed by extrusion are often stable for longer periods
exhibit uniform drug encapsulation and release kinetics along with than their equivalents made by sonication or detergent removal
longer circulation times; therefore, they are the most commonly methods [1].
used as drug delivery vehicles [8].
2.2.2. Reverse-phase evaporation
2.2. Methods of liposomes preparation Reverse-phase evaporation produces a mixture of LUVs and
MLVs entrapping large aqueous volumes, which allows for encap-
Liposome synthesis is a heavily investigated area of research sulation of large molecules, such as proteins and nucleic acids [1].
with many recent and modified techniques, including: heating, In this method, lipids and amphiphilic molecules are first mixed in
curvature tuning, localized IR heating, osmotic shock, dual asym- an organic solvent [15], then an aqueous buffer, which may contain
metric centrifugation, spray-drying, lyophilization, gel-assisted a solubilized drug, is added to the mixture. Afterwards, the organic
hydration, hydration on glass beads, hydration in microfluidics, solvent is evaporated using a rotary evaporator under low
2
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

pressure, leaving the lipid vesicles dispersed in the aqueous solu- fer. Drugs desired to be encapsulated may be added to the hydrat-
tion. If an application requires smaller, more uniform particles, ing buffer.
the size of liposomes may be reduced by extrusion [16]. In this
case, the pore size of the polycarbonate filter and the number of 2.2.8. Freeze-thaw
extrusion cycles will determine the size and polydispersity of the Freeze-thaw cycles are typically incorporated into liposome
synthesized liposomes [16,17]. synthesis to improve lipid packing and formation of unilamellar
vesicles [35]. This technique may be used with any liposome
2.2.3. Injection preparation method. For example, after thin film hydration the
There are several variations of the injection method [6]. In one solution can be sonicated at room temperature, frozen at
technique [18], lipids and amphiphilic molecules are dissolved in 196 °C in liquid nitrogen and left at room temperature to melt.
an organic solvent of a low boiling point (e.g., diethyl ether) then As the liposomal solution thaws, vesicles fuse forming LUVs. Up
the mixture is injected into a warm aqueous solution whose tem- to 10 freeze-thaw cycles may be used to achieve the intended
perature is constant and above the boiling point of the solvent results. Finally, if smaller vesicles are desired, the resulting solu-
used. This allows the organic solvent to evaporate and lipid vesicles tion may be sonicated again at room temperature. The freeze-
to form, producing primarily LUVs. Alternatively, if the organic sol- thaw technique is not optimal when using high concentrations of
vent used has a higher boiling point (e.g., ethanol), the solvent- lipid for synthesis [1]. Using this technique, a drug encapsulation
lipid mixture may be injected into an aqueous solution at room efficiency of 2030% was reported [1,36].
temperature under constant stirring [19], and the organic solvent
can then be removed via dialysis [20] or filtering. This method is 2.3. Methods of liposome characterization
most expedient for preparing large volumes of liposomal formula-
tions; however, the fabricated liposomes usually have higher poly- The key aspects that define the efficacy of a liposome formula-
dispersity indexes (PDI), suggesting a wide distribution of sizes, tion include size, zeta potential, encapsulation efficiency, release,
and in many cases MLVs may be present [1]. stability, and pharmacokinetics. Size and zeta potential are proper-
ties defined by the liposome preparation method and composition,
2.2.4. Detergent removal respectively. Drug encapsulation efficiency and stability are critical
Several techniques of the detergent removal method may be to protect and deliver the drug payload. Inefficient encapsulation
used to synthesize LUVs. In this method, lipids and amphiphilic can lead to significant waste of expensive drugs. Drug release is
molecules are mixed with a surfactant characterized by a high crit- desired in the site of interest; premature drug release may cause
ical micelle concentration (CMC) in an organic solvent which is undesirable ‘‘off-target” effects. The pharmacokinetics of the lipo-
then evaporated under low pressure [5,21]. A solution of mixed some are described by the circulation time and biodistribution of
micelles is obtained by hydrating the lipid film in an aqueous buf- the drug delivery vehicle. Together, encapsulation, stability,
fer. The detergent is then removed by means of dialysis [22–25], release, circulation time, and biodistribution characterize the abil-
size-exclusion chromatography [26–28], adsorption onto ity of a drug delivery vehicle to achieve the goal of delivering the
hydrophobic beads [29] or dilution [30,31]. The sample is then con- active drug to the diseased site.
centrated which allows the lipids to form LUVs.
2.3.1. Drug encapsulation efficiency and release
2.2.5. Dehydration-rehydration The encapsulation efficiency is a measure of the amount of drug
Dehydration-rehydration is used to synthesize LUVs without incorporated into the liposome during formulation. It is defined by
the use of organic solvents or detergents. In this method, liposomes subtracting the free non-incorporated drug from the total drug and
are made by dispersing lipids and amphihilic molecules at low con- dividing by the total drug initially added. This can be determined
centrations directly into an aqueous solution followed by sonica- using different methods, depending on the chemistry of the drug.
tion [32]. The drug to be encapsulated may be added to the The concentration of drug in solution may be determined spec-
aqueous solution and mixed with the formulated vesicles. When trophotometrically, fluorometrically, or using radiologic methods
water is evaporated under nitrogen, liposomes combine, creating [1].
a multilayered film entrapping the drug molecules. When water Characterization of drug release is often performed in vitro
is later added, large vesicles encapsulating the drug molecules using a dialysis method. Liposomes are placed inside pre-wetted
are produced. dialysis bags with a selected molecular weight cut-off to entrap
the liposomes but allow the drug to permeate across the mem-
2.2.6. pH jumping brane. The concentration of drug released is measured at different
pH jumping is a quick liposome preparation method which also time points. This provides a measure of the rate the drug will be
circumvents the use of organic solvents. When a solution of phos- released from liposome formulations [1]. In reality, drug release
phatidic acid in water is subjected for a short time (<2 min) to a 3.5 in vivo may be impacted by dilution in the bloodstream, pH, blood
fold increase in pH (from  3 to 10.5–11), SUVs are formed [33]. plasma proteins, cells, or turbulent flows.
When the same method is applied to a mixture of phosphatidic
acid and phosphatidyl choline, it yields similar results with the 2.3.2. Size, zeta potential, and stability
ratio of phosphatidic acid : phosphatidyl choline controlling the Liposomal stability is an important indicator of its potential effi-
percentage of SUVs versus LUVs obtained (i.e., the higher the ratio cacy and utility in clinical use. Often, the stability of a formulation is
of phosphatidic acid : phosphatidyl choline, the higher the percent- evaluated by performing physical assessments of the liposomes at
age of SUVs obtained [33]. multiple timepoints (e.g., days, week, or months) and assessing
drug leakage and nanoparticle size. Undesirable changes in the
2.2.7. Hydration in a packed bed of colloidal particles physical characteristics of a liposome formulation include aggrega-
This method uses a packed bed of colloids to produce SUVs tion of the particles and physical degradation of the lipid membrane
(<100 nm in diameter) in a single step [34]. In this method, lipo- over time. Liposomal diameter and surface charge can be deter-
somes are obtained when lipids and amphiphilic molecules dis- mined using dynamic light scattering (DLS) and phase analysis light
solved in an organic solvent are dried on asymmetric alumina scattering (PALS), respectively. Liposomes with neutral surface
particles packed in a capillary, then hydrated with an aqueous buf- charge aggregate and are unstable for drug delivery applications.
3
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

Fig. 1. Liposomes are composed of a diverse array of molecular components such as lipids, sterols, polysaccharides, and surfactants. Representative structures of these
molecules are shown.

Fig. 2. Incorporation of cholesterol and PEG affects liposomal drug encapsulation and release. (A) Incorporation of cholesterol decreases drug encapsulation within liposomes,
as evidenced by decreasing paclitaxel encapsulation with increasing incorporation of cholesterol. Reproduced with permission [49]. Copyright 2007, Taylor & Francis. (B)
PEGylation of liposomes decreases drug release relative to use of free drug or encapsulation within non-PEGylated liposomes. Reproduced with permission [73]. Copyright
2010, Dove Medical Press Limited.

An increase in size is used to assess the uniformity and stability of ensure degradation has not occurred [37]. Lyophilization is utilized
the liposome population over time. Atomic force microscopy (AFM) to prolong shelf life by reducing lipid oxidation. These techniques
is useful for measuring liposome mechanical characteristics, such are implemented at various timepoints over the shelf-life of the
as Young’s modulus. Fourier-transform infrared spectroscopy liposome formulation; large deviations from initial readings may
(FTIR) is used to analyze the makeup of the lipid membrane and indicate instability of a given formulation.
4
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

Fig. 3. Liposomal composition affects stability. (A) Cholesterol enhances liposomal structural stability, enabling maintenance of liposomal size throughout 10 weeks.
Reproduced with permission [49]. Copyright 2007, Taylor & Francis. (B) Lipids with higher gel-liquid phase transition temperature Tm, such as DSPC and DPPC, increase
liposomal stability and cargo retention relative to lipids of lower Tm, DMPC. Reproduced with permission [41]. Copyright 2004, Taylor & Francis.

Fig. 4. Liposomal composition affects surface charge and pharmacokinetics. (A) Cationic lipids DOTMA and DOTAP enable non-viral gene delivery, as demonstrated by
luciferase expression in the lungs (h), spleen ( ), heart ( ), liver (4), and kidneys ( ). Reproduced with permission [85]. Copyright 1997, Mary Ann Liebert, Inc. (B)
PEGylation of doxorubicin encapsulating liposomes (Doxil Ò) increases blood circulation time relative to administration of free drug, as demonstrated by differences in blood
concentrations of doxorubicin 24 h after infusion. Reproduced with permission [105]. Copyright 2003, Springer Nature.

5
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

Fig. 5. Liposomal composition affects stimuli response. (A) Liposomes composed of pH-responsive lipid DOPE become unstable in response to acidic pH (<7), denoted by the
increased release of cargo as pH decreases. Reproduced with permission [108]. Copyright 2001, Elsevier. (B) Temperature-responsive liposomes release doxorubicin when the
environmental temperature is greater than physiological temperature (37 °C), as evidenced by increased dox release at temperatures at or above 42 °C. Reproduced with
permission [110]. Copyright 2010, Elsevier.

2.3.3. Pharmacokinetics 3.1. Natural lipids


Pharmacokinetics describe what happens to the liposome and
drug after injection into the body. Drug pharmacokinetics are influ- Lipids have a diverse repertoire of structures that share a hydro-
enced by encapsulation within a liposome formulation. Liposome philic headgroup and hydrophobic, hydrocarbon tail. The head-
pharmacokinetics are characterized by the rate of removal from group may be negatively charged, positively charged, or
the bloodstream and accumulation in organs. Pharmacokinetic zwitterionic (having both a negative and positive charge, resulting
studies are performed by taking blood draws from subjects in net neutrality). The headgroup may also be chemically modified
injected with the liposome formulation over a period of time. Lipo- to allow conjugation with other molecules. For example, phos-
somes and drugs are often detected by either fluorescent or radio- phatidylethanolamine (PE) is often conjugated to polyethylene gly-
logic labeling. This information is frequently analyzed using the col via the amine group. The charge of the headgroup bestows
pharmacokinetic two-compartment model, in which the body is stability, as charged liposomes electrostatically repel each other.
divided into central and peripheral compartments. The central The hydrophobic tails may vary in acyl chain length, be symmetric
compartment is comprised of plasma and tissues where vesicle or asymmetric, and be saturated or unsaturated. The backbone of
distribution occurs rapidly, while the peripheral compartment is the lipid is often a phosphate, glycerol, or sphingosine group. These
made up of tissues where distribution happens slowly. Mathemat- distinguishing chemical features regulate bilayer assembly defined
ical models are fit to each of these compartments to model and by lipid packing, response to pH, stability, drug encapsulation and
track liposome dispersal and elimination. Because liposomes are release, and other liposome behaviors.
often comprised of components that exist in the human body,
tracking of adsorption, degradation, metabolism, and excretion is
relegated to natural processes. 3.1.1. Phospholipids
Natural lipids, such as phosphatidylcholine (PC) and PE are the
most common phospholipids present in mammalian cell mem-
3. Molecular components of liposomal drug delivery vehicles branes [38]. Glycerophospholipids, i.e. lipids with a glycerol back-
bone, are the predominant phospholipid in eukaryotic cells [39].
The liposome composition may be derived from natural or syn- Egg phosphatidylcholine (EPC) and egg phosphatidylglycerol
thetic lipids, polysaccharides, sterols, or surfactants (Fig. 1). (EPG) are derived from living tissues; they are composed of a mix-
Research in liposome formulation has led to the emergence of ture of lipid components. As a consequence, there may be inconsis-
unique properties, including: enhanced drug encapsulation, stim- tencies in the amount of the components from batch to batch. For
uli responsiveness, tissue-targeting, prolonged blood circulation, example, EPC derived from chicken egg is composed of L-a-
reduced drug toxicity in non-target tissues, and diagnostic capabil- phosphatidylcholine with acyl chain lengths varying from 14 to
ities (Figs. 2–5). Collectively, these features increase therapeutic 22 and 0 to 4 degrees of saturation. Other natural lipids are derived
efficacy relative to administration of free drug in a variety of clin- from living organisms and subsequently processed to yield a nearly
ical applications. pure lipid composition. In addition, lipid composition may be
6
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

defined by the degree of saturation or unsaturation of the acyl with high purity. Commonly utilized synthetic lipids include phos-
chain. Liposomes composed of PCs with long, saturated acyl chains phatidylcholines, phosphatidylethanolamines, and phosphatidyl-
(high Tm) have demonstrated greater stability in vivo compared to glycerols, such as 1,2-dioleoyl-sn-glycero-3-phosphocholine
formulations with PCs with short, unsaturated acyl chains (low Tm) (DOPC), 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE),
[40,41]. Vesicles composed of endogenous lipids are inherently and dioleoyl phosphatidylglycerol (DOPG), respectively. Synthetic
biocompatible and ideal for therapeutic applications as they mimic lipids are often chosen over natural lipids due to their purity, com-
native cell membranes. mercial availability, chemical functionality, and cost effectiveness.

3.1.2. Sphingolipids
Sphingolipids are another type of natural lipid found predomi- 3.3. Surfactants
nantly in mammalian cell membranes. Sphingolipids are com-
prised of an 18-carbon amino-alcohol sphingosine back bone; Surfactants are classified as molecules that reduce the surface
they play a critical role in cell membrane structure and as regula- tension of the liquid in which it is incorporated. Surfactants, com-
tory signaling molecules [42]. The incorporation of sphingolipids, monly referred to as edge activators, are useful additives in lipo-
such as sphingomyelin (SM), in liposomal formulations exhibit some formulations. Edge activators are typically single acyl-chain
increased stability in vivo, longer blood circulation, and increased surfactants that serve to destabilize the lipid bilayer of liposomal
therapeutic efficacy [43]. Ceramides are a type of sphingolipid con- nanoparticles, thereby increasing vessel deformability [57]. Edge
sisting of a sphingosine and a single fatty acid. The incorporation of activators were shown to increase the dermal penetrative ability
ceramides in liposomal formulations increases membrane fluidity of liposomes in anti-cancer [58,59], anti-fungal [60], and transder-
and fusion capabilities with dermal cells [44]. mal applications [61]. Frequently utilized edge activators include:
sodium cholate, Span 60, Span 80, Tween 60, and Tween 80. The
3.1.3. Sterols charge of edge activators may also be utilized to increase therapeu-
Sterols are a class of natural lipid molecules present in nearly all tic efficacy. For example, using sodium cholate, with a positive zeta
living organisms [45]. There are three subtypes of sterols: phytos- potential, electrostatically binds with negatively charged compo-
terols zoosterols, and mycosterols, which are found in plants, ani- nents like DNA. Surfactant type and concentration may be lever-
mals, and microorganisms, respectively [45]. Cholesterol is an aged to improve the therapeutic efficiency of liposomal drug and
endogenous amphiphilic zoosterol and is a critical component of gene delivery [62,63].
mammalian cell membranes. Within the cell membrane, choles-
terol is primarily confined to lipid rafts and plays an important role
in processes of regulating membrane integrity and lipid raft func- 4. Effect of composition on liposome characteristics &
tionality [46]. The incorporation of cholesterol within liposomal functionality
formulations has demonstrated increased stability in vivo and
decreased leakiness across the lipid bilayer, resulting in the pro- 4.1. Drug encapsulation
longed and controlled release of cargo [37,47,48]. The incorpora-
tion of 20–50 mol% cholesterol in liposomal formulations Traditional therapeutic compounds, despite in vitro effective-
demonstrated reduced encapsulation efficiency [49] and increased ness, are often limited in vivo by poor pharmacokinetic properties,
stability in vivo [47], relative to controls lacking cholesterol. rapid clearance, low plasma solubility, or poor biodistribution.
Cholesterol rich liposomes remained stable in the blood stream Liposomal drug delivery is designed to circumvent these limita-
for over 6 h, while cholesterol free liposomes were only stable tions by encapsulating the drug of interest inside a vehicle that
for a few minutes [47]. demonstrates favorable in vivo stability, pharmacokinetics, and
biodistribution. Achieving high encapsulation efficiency is another
3.1.4. Polysaccharides critical parameter of liposomal drug delivery success. While there
Polysaccharides are long-chain polymeric carbohydrates con- are various techniques employed to increase encapsulation effi-
sisting of monosaccharides bound by glycosidic linkages. Polysac- ciency, it can be particularly difficult to efficiently load a drug into
charides play an important role in cellular communication and small liposomes (50–150 nm) due to their low entrapment vol-
are present in cell membranes to aid in processes of cell and tissue umes. This challenge is addressed through the use of reverse phase
recognition, as well as certain transport mechanisms [50]. Coating evaporation and freeze-thaw cycling [64,65]. Hydrophobic and
the lipid membrane with oligo- or polysaccharides may target lipo- hydrophilic compounds may be easily loaded into liposomes with
somes to cell receptors or extend circulation [51,52]. Polysaccha- high encapsulation efficiency via association with the lipid bilayer
rides have additional properties that make them well suited for or aqueous core, respectively.
use in drug delivery systems, such as biocompatibility and anti- Active loading is a drug encapsulation method that converts the
viral, anti-bacterial, and anti-tumoral properties [53]. Mucosal sur- drug from membrane permeable to impermeable. For example, a
faces of the body are particularly good targets for polysaccharide trans-membrane gradient is introduced in order to drive drug
recognition, thus nasal, pulmonary, peroral, and gastrointestinal molecules into empty vesicles via a pH gradient. It was found that
epithelium routes are heavily investigated for targeting with generating pH gradients by using ammonium sulfate [66] or citrate
polysaccharide-coated liposomes [53]. Popular polysaccharides buffer [67] improve the encapsulation efficiency of amphiphilic
used in liposomal formulations are chitosan [54] and hyaluronan drugs while maintaining a simple, economical, and safe process
[55,56]. [68]. These processes are used commercially in the drug loading
of many FDA approved liposomal systems, such as DoxilÒ, Myo-
3.2. Synthetic lipids cetTM, and DuanoXomeÓ.
The physicochemical properties of liposomes may affect the
Synthetic lipids, lipids not occurring naturally or derived from encapsulation efficiency during preparation, including: size, sur-
living sources, are commercially synthesized and frequently utilized face charge, composition, and surface modifications [69]. Incorpo-
as components of therapeutic liposomes. Though these materials are ration of long acyl chain lipids were shown to increase
not endogenous, they are biologically and structurally similar to encapsulation efficiency of hydrophobic drugs within the bilayer
natural lipids, exhibit high biocompatibility, and are synthesized and improved drug retention [70]. Incorporation of cholesterol
7
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

can negatively impact the encapsulation efficiency for drugs that of cell membranes, nonspecific cell uptake is reduced compared to
are incorporated into the lipid membrane [71]. that of cationic liposomes. Reduced nonspecific uptake also
The liposome formulation may be tailored to control the drug enables anionic liposomes to circulate in the bloodstream longer
release rate, leading to improved therapeutic efficacy of the drug. than cationic liposomes [80].
Aspects of liposomal formulation that may be optimized to effect Liposomes with positive surface charge can be synthesized via
drug release include: zeta potential, incorporation of cholesterol, the incorporation of positively charged lipids. The most commonly
and the unique characteristics of the constitutive lipids (head utilized lipids in cationic liposomes include: 1,2-dioleoyl-3-trime
groups, acyl chain length, degree of saturation, and phase transi- thylammonium-propane (DOTAP), C-Chol, N-(1-(2,3-dimyristyloxy
tion temperature) [71]. propyl)-N,N-dimethyl-(2-hydroxyethyl) ammonium bromide
Characteristics of the lipids in the liposome formulation affect (DMRIE), 1,2-dimyristyloxypropyl-3-dimethyl-hydoxyethylammo
packing of the lipid bilayer, which may be optimized to achieve nium bromide (DODAC), and 1,2-di-O-octadecenyl-3-trimethylam
sustained drug release. Increasing the degrees of unsaturation of monium propane (DOTMA). Cationic liposomes are noted for their
the acyl chain has demonstrated increased drug release due to usefulness in nonviral gene transfection and siRNA delivery [81–
the leakiness of the lipid bilayer [72]. Liposomes made with 100% 85]. Positively charged liposomes are utilized in the formulation
DOPC exhibited fast drug release, with no further release after of liposomal vaccine adjuvants [86] as they possess inherent
24 h, whereas liposomes made with 100% DSPC showed slow, sus- immunostimulatory properties [87,88]. Augmented immunogenic
tained release that lasted seven days [72]. A formulation made response and increased vaccine efficiency was demonstrated with
with a mixture of the two showed an intermediate release profile, cationic liposomal vaccines [89]. Further, cationic liposomes have
showing how mixtures of lipids can serve to tune drug release. demonstrated enhanced peritoneal retention compared to neutral
Liposomal incorporation of polyethylene glycol (PEG) has also or anionic liposomes, making them ideal drug carriers for the treat-
demonstrated decreased drug release [73]. A higher degree of lipid ment of gastrointestinal and gynecological malignancies [90].
saturation (and higher Tm), affects the fluidity of the lipid mem- However, due to electrostatic interactions between the
brane. Increasing the Tm decreases overall drug release [71]. positively-charged lipid membrane of the vesicle and negatively-
charged cell membranes, cationic liposomes are nonspecifically
4.2. Stability internalized by cells to a greater extent than anionic liposomes.

Liposomal stability may be classified into physical, chemical, 4.4. Pharmacokinetics


and biological. Physical and chemical stability often refer to the
ability of the liposomal formulation to maintain its properties over Differences in liposomal composition can alter pharmacokinetic
time. Phospholipids are prone to several chemical degradation parameters in vivo, as noted by changes in blood clearance rate,
reactions, including hydrolysis at ester bonds and peroxidation of half-life, and biodistribution. Liposomal formulations which pro-
unsaturated acyl chains; these phenomena can affect the long- long blood circulation enable higher accumulation and deposition
term stability of a liposome formulation. Stability may also of therapeutic cargo within target tissues.
describe the ability of a liposome formulation to be reconstituted
from lyophilization. Various excipients are used to balance osmo- 4.4.1. Clearance rate and half-life
lality, pressure of the interior and exterior of the liposome. Biolog- Liposomal blood clearance rate and half-life are greatly
ical stability refers to liposome integrity in the presence of serum impacted by liposomal composition. The mononuclear phagocyte
proteins. Upon entering the bloodstream, liposomes bind serum system (MPS), which consists of tissue resident macrophages and
proteins, a phenomenon known as opsonization, which results in blood monocytes [91], is primarily responsible for the clearance
rapid clearance [74]. The avoidance of liposomal opsonization of liposomes from the blood stream. Circulating neutrophils also
and subsequent increase in blood circulation time can be achieved significantly hinder drug delivery [92]. There is considerable
through the incorporation of PEG, as described in section 4.4. The research devoted to modulating liposomal composition to reduce
addition of cholesterol in liposomal formulations, alone or in con- uptake by the MPS.
junction with PEG, also increases blood circulation time [75,76]. Liposomal opsonization upon systemic administration directs
Overall, stability describes the maintenance of the initial liposome liposomes to interact with receptors on the surface of liver and
properties in response to different stresses. spleen resident macrophages of the MPS, subsequently inducing
internalization and degradation. In a mechanistically analogous
4.3. Surface charge fashion, liposomal opsonization may also direct particles for
uptake in the liver by hepatocytes [93]. The incorporation of PEG
Each phospholipid exhibits a net charge as a function of their in liposome formulations, known as ‘‘stealth” liposomes, has pro-
headgroup chemistry and solution pH. The liposome compositions longed the circulation time and subsequently increased liposomal
in the final formulation impacts the overall zeta potential of the half-life by hindering opsonization or preferentially binding pro-
liposome, which affects the function and efficacy of the vesicles teins that evade uptake [94,95] Liposomal PEGylation imparts a
as a therapeutic delivery system. greater surface hydrophilicity as well as a barrier of steric hinder-
Anionic liposomes, i.e. those with negative surface charge, are ance, both of which affect the binding of serum proteins to the
composed of lipids with negatively charged head groups, such as: liposome surface [96,97]. Further, liposomes composed primarily
1,2-dioleoyl-sn-glycero-3-phospho-L-serine (DOPS), and 1,2- of neutral phospholipids with a small molar percent of negatively
dioleoyl-sn-glycero-3-phospho-(10 -rac-glycerol) (DOPG). Anionic charged glycolipid, principally monosialoganglioside (GM1), have
liposomes are utilized for DNA transfection [77]. The incorporation also demonstrated prolonged circulation in vivo. Mechanistically,
of negatively charged lipids in liposomal formulations also GM1 reduces liposomal opsonization through steric hinderances
enhanced the delivery of cardiotoxic drugs, such as doxorubicin, introduced by the negatively charged sialic acid and the physical
reducing systemic toxicity compared to administration of free drug boundary provided by the carbohydrate chains [98].
[78]. Further, anionic liposomes demonstrated enhanced vascular
extravasation and lower accumulation in the vascular endothe- 4.4.2. Biodistribution
lium, relative to cationic liposomes [79]. Due to the electrostatic Liposomal biodistribution, i.e. the dispersion of liposomes
repulsion of anionic liposomes with the negatively charged surface throughout the body (often the lungs, kidneys, liver, spleen, stom-
8
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

ach, intestines, and brain) can be altered through the incorporation temperatures, such as DSPC, soy PC, and HSPC, and additives such
of different molecular components. as cholesterol and PEG were shown to be successful in maintaining
Liposomes have garnered attention in the field of cancer drug the circulation time of liposomes. Traditionally, PE was utilized in
delivery for the ability to deliver toxic drugs to cancer cells. Accu- the synthesis of pH-sensitive liposomes to stabilize the carriers at
mulation of liposomes within a tumor is facilitated by the physiological pH. More recently, alternative mechanisms such as
enhanced permeability and retention (EPR) effect. Though not fully the incorporation of pH-sensitive and fusogenic molecules were
understood, the EPR effect is hypothesized to result from leaky vas- explored [109].
culature and an impaired lymphatic system present in tumors.
Tumor accumulation via the EPR effect is enhanced by liposomes 5.2. Temperature responsive liposomes
with prolonged blood circulation. Other factors which have
impacted tumor accumulation include: liposome size [99], elastic- Temperature-sensitive liposomes deposit their payload when
ity [100], and surface charge [79,101]. Liposomal elasticity is mod- external heat is applied, but the modes by which the membrane
ified via the incorporation of cholesterol, surfactants, and lipids of is destabilized differ. Traditional temperature-sensitive liposomes
varying acyl chain length and degree of saturation. Liposomes are composed of lipids that have a gel-to-liquid phase transition
exhibiting low to moderate Young’s Moduli (~0.045–28 MPa) accu- temperature or Tm that is higher than the body temperature, typi-
mulated significantly more in tumors than high Young’s modulus cally ~42 °C. Recently, temperature-sensitive polymers or lysoli-
liposomes [100,102]. Modulation of liposomal surface charge pids were used to initiate membrane destabilization, followed by
demonstrated enhanced accumulation of anionic liposomes within payload deposition when heated [110]. Above the Tm, the lipid
tumors, relative to their cationic or neutral counterparts [103]. membrane increases fluidity and permeability to allow for maxi-
Liposomal surface modifications, such as the conjugation of pep- mum drug release. Traditional thermo-sensitive liposomes were
tide or antibodies to enable targeting of liposomes to overex- formulated using a mixture of lipids with different phase transition
pressed receptors on diseased cells, have also been investigated temperatures which increases lipid packing disorder, subsequently
as a method to increase tumor accumulation (reviewed here increasing cargo permeability. However, the high thermal dose
[104]). threshold for thermo-responsive liposomes may cause damage to
Liposomes predominantly accumulate in the organs of the renal healthy tissues during heat application. To address this limitation,
and lymphatic systems, such as the liver, kidneys, and spleen, a new generation of thermo-sensitive liposomes was explored,
respectively. Liposomal accumulation within the liver and spleen using temperature-sensitive polymers and lysolipids to lower the
is modulated by cells of the MPS present within these organs. Renal phase transition temperature, while still enabling rapid drug
accumulation and clearance of liposomes by the kidneys occurs release [111].
through physiological processes of blood filtration and waste
excretion. Accumulation within other organ systems, such as the
5.3. Theranostic liposomes
respiratory, nervous, and cardiovascular system, i.e. the lungs,
brain, and heart, respectively, is generally minimal. Researchers
Theranostic liposomes, incorporating a diagnostic (i.e. imaging
have taken advantage of these pathways to deliver anti-fungal
modality) and therapeutic agent, are multifunctional nanomedici-
agents directly to the kidney (amphoteric B).
nes for disease treatment and monitoring [112]. Theranostic lipo-
somes can be formulated to be amenable to several imaging
5. Liposomal composition imparts unique functionalities techniques, including magnetic resonance (MRI), near-infrared flu-
orescent (NIR), and nuclear imaging [113,114].
The advent of stimuli-responsive liposomes has accelerated the
use of nanoparticles for site-specific therapeutic delivery. Environ- 6. Liposomal drugs in clinical use and preclinical development
mentally specific stimuli, such as pH, can cause destabilization of
the liposomal membrane, enabling local deposition of drug pay- To emphasize the versatility of liposomes as drug carriers, we
load in target tissue. External stimuli have also been explored, will discuss a few illustrative examples of different liposomal drug
including heat and light. By utilizing an external stimulus, the site formulations either approved by the U.S. Food and Drug Adminis-
of release can be controlled as well as the rate of release. Stimuli- tration (F.D.A.) or the European Medicines Agency (E.M.A.) for clin-
responsive liposomes exhibited increased efficacy in anti-tumor ical use, or in the clinical trials stage. Table 1 lists all liposomal
and gene delivery due to the distinctive properties of the tumor drug formulations currently approved for clinical use. Liposomes
microenvironment, including pH and enzymatic activity. By lever- are used as carriers for analgesic medications such as morphine
aging tumor specific environmental cues, the drug is delivered to and fentanyl, and anesthetics such as ropivacaine [115]. They are
the tumor site in a way that is both targeted and efficient [106]. also used as vehicles for drugs that prevent and treat a broad spec-
trum of diseases such as fungal (e.g., amphotericin B and nystatin),
5.1. pH responsive liposomes bacterial (e.g., amikacin and doxycycline) and viral infections, as
well as chemotherapeutics used for treatment of different types
pH-sensitive liposomes were explored for their ability to target of cancers (e.g., camptothecin, cisplatin, daunorubicin, docetaxel,
tumors and regions of inflammation. When pH-responsive lipo- doxorubicin, gemcitabine, oxaliplatin, paclitaxel, and vincristine)
somes encounter acidic conditions, components of the lipid bilayer [115–117].
become protonated. This chemical change subsequently disrupts
the structural integrity of the bilayer, causing the membrane to 6.1. Anti-cancer
rupture. Thus, when pH-responsive liposomes come into contact
with an acidic microenvironment, such as the endosome of a cell, Aroplatin is currently in phase II clinical trials and is being
they release drug payload [107,108]. pH-sensitive liposomes may investigated for the treatment of metastatic colorectal carcinoma
be optimized to maintain necessary characteristics of liposomes, through intrapleural injection [118]. Colorectal carcinoma is a
such as long circulation time, while optimizing their ability to malignant tumor growth in the epithelium that is part of the
destabilize and deposit their payload effectively at a desirable mucosa layer of the large intestine [119]. Aroplatin contains a
pH. The incorporation of lipids with high phase transition cis-bis-neodecanoato-trans-R, R-1,2-diaminocyclohexane platinum
9
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

Table 1
Liposomal drug formulations approved for clinical use.

Name Company Liposomal Composition Drug Encapsulated Drug Type Route of Clinical References
(molar ratio) Administration Approval
Year
Abelcet Leadiant Biosciences, DMPC : DMPG Amphotericin B Antifungal I.V. 1995 [115,117,132,133]
Inc. (2.3 : 1)
Ambisome Fujisawa Healthcare, HSPC : DSPG : Cholesterol : Amphotericin B Antifungal I.V. 1997 [117,158]
Inc. and Gilead Sciences, Amphotericin B
Inc. (5 : 2 : 2.5 : 1)
Amphocil Zeneca Pharmaceuticals Cholesteryl sulphate : Amphotericin B Antifungal I.V. 1993 [115,159]
Amphotericin B
(1 : 1)
Amphotec Sequus Pharmaceuticals Cholesteryl sulphate : Amphotericin B Antifungal I.V. 1996 [116,117,160]
Inc. Amphotericin B
(1 : 1)
Arikayce Insmed, Inc. of DPPC and Cholesterol : Amikacin Antibacterial Oral Inhalation 2018 [142,146,161]
Bridgewater, NJ. Amphotericin B
(0.6–0.79 : 1 wt ratio)
DaunoXome Galen US, Inc. DSPC : Cholesterol Daunorubicin Chemotherapeutic I.V. 1996 [117,162]
(2 : 1)
DepoDur Pacira Pharmaceuticals, DOPC : DPPG : Cholesterol : Morphine sulfate Narcotic Analgesic Epidural 2004 [117,139]
Inc. Tricaprylin and Triolein
(507 : 11 : 76 : 6 : 1)
Doxil Johnson & Johnson HSPC : Cholesterol : DSPE- Doxorubicin Chemotherapeutic I.V. 1995 [117,135]
PEG2000
(11.2 : 7.8 : 1)
Epaxal Johnson & Johnson DOPC : DOPE Hepatitis A virus Vaccine I.M. 1993 [117,163]
(3 : 1) antigen, strain RG-
SB
Exparel Pacira Pharmaceuticals, DEPC : DPPG : Cholesterol : Bupivacaine Anesthetic I.V. 2011 [117,164]
Inc. Tricaprylin
(7.6 : 1 : 10 : 3.5)
Evacet Liposome Company Inc. (Hydro Soy PC, cholesterol Doxorubicin Chemotherapeutic I.V. 1995 [115,133,165,166]
and DSPE-PEG) :
Doxorubicin
(8 : 1)
Inflexal V Johnson & Johnson 70% Lecithin, 20% Cephalin Influenza virus Vaccine I.M. 1997 [117,151]
and 10% Phospholipids antigen, strains A
(DOPC : DOPE, 3 : 1) and B
Lipodox Sun Pharmaceutical DSPC : Cholesterol : DSPE- Doxorubicin Chemotherapeutic I.V. 1995 [167–170]
Industries Ltd. PEG2000
(10.9 : 7.3 : 1)
Marqibo Acrotech Biopharma, Sphingomyelin : Vincristine Chemotherapeutic I.V. 2012 [117,171]
LLC Cholesterol
(1.5 : 1)
Mepact Takeda Pharmaceutical DOPS : POPC Mifamurtide Immunomodulator/ I.V. 2004 [117,172]
Limited (1 : 2.3) Antitumor
Myocet Zeneus Pharma Ltd. EPG : Cholesterol Doxorubicin Chemotherapeutic I.V. 2000 [117,173]
(1.2 : 1)
Onivyde Merrimack DSPC : MPEG-2000 : DSPE Irinotecan Chemotherapeutic I.V. 2015 [117,174,175]
Pharmaceuticals, Inc. (200 : 133.3 : 1)
Visudyne Novartis International Verteporfin : DMPC and Verteporfin Photosensitizer I.V. 2000 [117,176]
AG EPG
(1 : 8)

(II) (NDDP), a molecule structurally analogous to oxaliplatin. Plat- cytarabine. The liposome in this drug formulation is composed of
inum coordination complexes inhibit the replication of DNA in cholesterol, triolein, DOPC and 1,2-dipalmitoyl-sn-glycero-3-
tumor cells by cross-linking the two DNA strands, inducing cell phospho-(10 -rac-glycerol) (DPPG) in a molar ratio of 11:1:7:1
death [117,120–123]. Aroplatin consists of MLVs encapsulating [116]. The encapsulated drug is slowly released from the 3–
NDDP, made of 1,2-dimyristoyl-sn-glycero-3-phosphocholine 30 mm MLVs – with high internal volume to surface area ratio.
(DMPC) and 1,2-dimyristoyl-sn-glycero-3-phospho-(10 -rac- These giant vesicles consist of 96% aqueous foam and 4% lipids
glycerol) (DMPG). The drug-loaded liposomes increase NDDP accu- [117,129]. The liposome encapsulated cytarabine was shown to
mulation in tumor cells, and reduce its side effects compared with be more effective in targeting and destroying tumor cells in the
the unencapsulated form of the drug [117,124]. meninges and cerebrospinal fluid compared with other forms of
Depocyt is a cytarabine liposomal injection that is F.D.A. the drug [117,130]. In 2017, Pacira BioSciences, Inc. discontinued
approved for treatment of neoplastic meningitis through spinal the production of Depocyt due to manufacturing difficulties [131].
injection [116,125–127]. Neoplastic meningitis or ‘leptomeningeal Doxil is the first F.D.A. approved drug containing PEGylated
metastasis’ is inflammation of the brain caused by tumor growth in liposomes [105,117,132–135]. It is prescribed for the treatment
the arachnoid mater and/or the pia mater of the meninges which of different types of cancers including ovarian cancer,
surround the brain and spinal cord [128]. As the disease pro- AIDS-related Kaposi sarcoma, and multiple myeloma [117,136].
gresses, tumor cells spread through the cerebrospinal fluid result- The chemotherapeutic encapsulated in Doxil is doxorubicin
ing in poor prognosis. DepoCyt contains the chemotherapeutic HCl (Dox). The liposomes in Doxil are composed of

10
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

L-a-phosphatidylcholine hydrogenated soy (HSPC), cholesterol and significantly improved immunogenicity and tolerability over other
N-(carbonyl-methoxypolyethyleneglycol 2000)-1,2-distearoyl-sn- types of conventional influenza vaccines [117].
glycero-3-phosphoethanolamine sodium salt (MPEG-DSPE) in a Finally, it should be noted that decades of research into engi-
molar ratio of 56:38:5 [117,137]. The SUVs are 80–100 nm neering and optimizing these drug delivery systems have enabled
[117,135], and are comprised of Dox:lipid with a mass ratio of the record-breaking fast development of two vaccines, by Pfizer-
8:1 [117,138]. This high drug:lipid ratio is achieved by a remote BioNTech and Moderna, against SARS-CoV-2 [156]. These vaccines
loading technique using an ammonium sulfate gradient contain mRNA fragments of viral spike proteins – which enable
[105,117,135]. Doxil showed an increased circulation time with a SARS-CoV-2 to get attached, and gain entry into cells – encapsu-
half-life of 20–35 h and lower cardiotoxicity compared with the lated into lipid nanoparticles. The function of the lipid vesicles is
free form of the drug [105]. to protect the genetic materials from being degraded by enzymes,
and enable their uptake into cells, so the encoded viral proteins can
6.2. Pain management be synthesized by cellular machinery and an immune response is
initiated [157].
DepoDur is a morphine sulfate extended-release liposome
injection F.D.A. approved for treatment of severe pain [139]. The 7. Conclusion
liposomal composition of DepoDur consists of DOPC, DPPG, choles-
terol, tricaprylin and triolein in a mass ratio of 42:9:33:3:1, form- Liposome composition imparts unique characteristics and func-
ing 17–23 mm MLVs; the drug encapsulated is an analgesic, tionality, making liposomes ideal carriers for a wide range of ther-
morphine sulphate [117,140]. Morphine sulphate binds opioid apeutic cargo and clinical applications. Since their discovery nearly
receptors, inhibitory G protein-coupled receptors, in the central 55 years ago, liposomes have become a staple in the field of drug
nervous system and mitigates severe pain [141,142]. Using the delivery. The use of liposomes in clinical applications has dramat-
morphine encapsulated liposomes, clinical trials demonstrated ically reduced the off-target toxicity of various drugs, enabled pro-
consistent release of the drug up to 48 h post injection [143]. longed blood circulation and beneficial drug biodistribution.
Most recently, liposomes have taken center stage in the battle
against SARS-CoV-2. By providing protection of genetic material,
6.3. Anti-bacterial
lipid nanoparticles have enabled the successful translation of
mRNA vaccines and ushered in a new era in gene therapy. Given
In 2018, Arikayce, Amikacin Liposome Inhalation Suspension
the significant efficacy demonstrated by these vaccines, it is evi-
(ALIS), was approved by the F.D.A. for the treatment of a bacterial
dent that liposomal drug delivery is paving the way for future suc-
lung infection, mycobacterium avium complex (MAC) [144–147].
cess in the delivery of genetic materials for a range of therapeutic
Two types of nontuberculous mycobacteria bacteria can cause
needs. As with the development of the SARS-CoV-2 mRNA vacci-
MAC: Mycobacterium avium and Mycobacterium intracellulare,
nes, optimization of lipid composition will play a critical role in
which mainly affect immunocompromised individuals. [148]. Ari-
the development of these gene therapy-based treatments.
kayce contains amikacin [149], a bactericidal aminoglycoside,
effective in vivo against different species of gram-negative bacteria
including Acinetobacter, Enterobacter, Klebsiella Pseudomonas, Pro- References
teus, Serratia and Escherichia coli, and one species of gram- [1] A. Akbarzadeh et al., Liposome: classification, preparation, and applications,
positive bacteria, Staphylococcus [150]. Arikayce is the first liposo- Nanoscale Res. Lett. 8 (1) (2013) 102.
mal formulation approved to be administerd by inhalation. In this [2] Š. Koudelka, J. Turánek, Liposomal paclitaxel formulations, J. Control. Release
163 (3) (2012) 322–334.
formulation, the liposomal vehicle, which protects and stabilizes
[3] R. Mehta et al., Liposomal amphotericin B is toxic to fungal cells but not to
the active drug until it reaches the lungs, is made of a phospholipid, mammalian cells, Biochim. Biophys. Acta (BBA)-Biomembr. 770 (2) (1984)
1,2-dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), and choles- 230–234.
terol in 2:1 mass ratio, while the concentrations of amikacin and [4] Y.-K. Oh, D.E. Nix, R.M. Straubinger, Formulation and efficacy of liposome-
encapsulated antibiotics for therapy of intracellular Mycobacterium avium
total lipids in the formulation are 70 mg/ml and 47 mg/mL, respec- infection, Antimicrob. Agents Chemother. 39 (9) (1995) 2104–2111.
tively [116,147]. The innovative approach of this drug formulation [5] S. Vemuri, C.T. Rhodes, Preparation and characterization of liposomes as
is that it overcame the difficulty of treating intracellular bacterial therapeutic delivery systems: a review, Pharm. Acta Helv. 70 (2) (1995) 95–
111.
infection by delivering the active drug directly into cells via lipo- [6] C. Has, P. Sunthar, A comprehensive review on recent preparation techniques
somes that are taken up by macrophages within the lungs [147]. of liposomes, J. Liposome Res. 30 (4) (2020) 336–365.
In vivo studies in rats demonstrated a 5- to 8-fold increase in the [7] D.D. Lasic, The mechanism of vesicle formation, Biochem. J. 256 (1) (1988) 1–
11.
concentration of amikacin in pulmonary macrophages at 2, 6, [8] M. Alavi, N. Karimi, M. Safaei, Application of various types of liposomes in
and 24 h after treatment, compared with the inhaled free form of drug delivery systems, Adv. Pharm. Bull. 7 (1) (2017) 3–9.
the drug [147]. [9] A.D. Bangham, H.M.W., N.G.A. Miller, Preparation and Use of Liposomes as
Models of Biological Membranes, in: E.D. Korn (Ed.), Methods in Membrane
Biology, Springer, Boston, MA, 1974.
6.4. Vaccines [10] A.D. Bangham, Lipid bilayers and biomembranes, Annu. Rev. Biochem. 41
(1972) 753–776.
[11] A.D. Bangham, R.W. Horne, Negative staining of phospholipids and their
Inflexal V, an inactivated trivalent influenza vaccine, is com- structural modification by surface-active agents as observed in the electron
prised of 150 nm unilamellar virosomes – liposomes whose sur- microscope, J. Mol. Biol. 8 (5) (1964), p. 660-IN10.
[12] A.D. Bangham, M.M. Standish, J.C. Watkins, Diffusion of univalent ions across
faces are decorated with viral antigens, in this case, the lamellae of swollen phospholipids, J. Mol. Biol. 13 (1) (1965), p. 238-IN27.
haemagglutinin and neuraminidase glycoproteins of influenza [13] F. Olson et al., Preparation of liposomes of defined size distribution by
virus strains A and B [117,151–153]. The liposomes are composed extrusion through polycarbonate membranes, Biochim. Biophys. Acta (BBA) –
Biomembr. 557 (1) (1979) 9–23.
of 70% lecithin, 20% cephalin and 10% phospholipids (DOPC:DOPE,
[14] F. Szoka Jr., D. Papahadjopoulos, Comparative properties and methods of
75:25 molar ratio), where cephalin functions as a structural com- preparation of lipid vesicles (liposomes), Annu. Rev. Biophys. Bioeng. 9 (1980)
ponent and a stimulant to B lymphocytes [117,154]. Each dose of 467–508.
Inflexal V contains 15 mg of haemagglutinin-decorated monovalent [15] F. Szoka, Jr., D. Papahadjopoulos, Procedure for preparation of liposomes with
large internal aqueous space and high capture by reverse-phase evaporation.
virosomes from three different pools of influenza viruses (two of Proceedings of the National Academy of Sciences of the United States of
strain A and one of strain B) [151,155,155]. Inflexal V has shown America, 75(9) (1978) 4194-4198.

11
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

[16] F. Szoka et al., Preparation of unilamellar liposomes of intermediate size (0.1– [46] P. Goluszko, B. Nowicki, Membrane cholesterol: a crucial molecule affecting
0.2 lm) by a combination of reverse phase evaporation and extrusion interactions of microbial pathogens with mammalian cells, Infect. Immun. 73
through polycarbonate membranes, Biochim. Biophys. Acta (BBA)-Biomembr. (12) (2005) 7791–7796.
601 (1980) 559–571. [47] C. Kirby, J. Clarke, G. Gregoriadis, Effect of the cholesterol content of small
[17] N.-Q. Shi, X.-R. Qi, Preparation of Drug Liposomes by Reverse-Phase unilamellar liposomes on their stability in vivo and in vitro, Biochem. J. 186
Evaporation, in Liposome-Based Drug Delivery Systems, in: W.-L. Lu, X.-R. (2) (1980) 591–598.
Qi (Eds.), Springer Berlin Heidelberg: Berlin, Heidelberg, 2017, p. 1-10. [48] G. Gregoriadis, C. Davis, Stability of liposomes invivo and invitro is promoted
[18] D. Deamer, A.D. Bangham, Large volume liposomes by an ether vaporization by their cholesterol content and the presence of blood cells, Biochem.
method, Biochim. Biophys. Acta, Biomembr. 443 (3) (1976) 629–634. Biophys. Res. Commun. 89 (4) (1979) 1287–1293.
[19] S. Batzri, E.D. Korn, Single bilayer liposomes prepared without sonication, [49] T. Yang et al., Liposome formulation of paclitaxel with enhanced solubility
Biochim. Biophys. Acta, Biomembr. 298 (4) (1973) 1015–1019. and stability, Drug Deliv. 14 (5) (2007) 301–308.
[20] Y. Kagawa, E. Racker, Partial Resolution of the Enzymes Catalyzing Oxidative [50] B.K. Brandley, R.L. Schnaar, Cell-surface carbohydrates in cell recognition and
Phosphorylation: XXV. RECONSTITUTION OF VESICLES CATALYZING 32Pi— response, J. Leukoc. Biol. 40 (1) (1986) 97–111.
ADENOSINE TRIPHOSPHATE EXCHANGE, J. Biol. Chem. 246 (17) (1971) 5477– [51] J. Sunamoto, I.K., Protein-coated and polysaccharide-coated liposomes as
5487. drug carriers. Critical Reviews in Therapeutics Drug Carrier Systems, 1986.
[21] W. Jiskoot et al., Preparation of liposomes via detergent removal from mixed [52] T.M. Allen, C. Hansen, J. Rutledge, Liposomes with prolonged circulation
micelles by dilution, Pharmaceutisch Weekblad 8 (5) (1986) 259–265. times: factors affecting uptake by reticuloendothelial and other tissues,
[22] M.H. Milsmann, R.A. Schwendener, H.G. Weder, The preparation of large Biochim. Biophys. Acta (BBA)-Biomembr. 981 (1) (1989) 27–35.
single bilayer liposomes by a fast and controlled dialysis, Biochim. Biophys. [53] C. Lemarchand, R. Gref, P. Couvreur, Polysaccharide-decorated nanoparticles,
Acta, Biomembr. 512 (1) (1978) 147–155. Eur. J. Pharm. Biopharm. 58 (2) (2004) 327–341.
[23] V. Rhoden, S.M. Goldin, Formation of unilamellar lipid vesicles of controllable [54] M.M. Mady et al., Biophysical studies on chitosan-coated liposomes, Eur.
dimensions by detergent dialysis, Biochemistry 18 (19) (1979) 4173–4176. Biophys. J. 38 (8) (2009) 1127–1133.
[24] O. Zumbuehl, H.G. Weder, Liposomes of controllable size in the range of 40 to [55] D. Peer, R. Margalit, Tumor-targeted hyaluronan nanoliposomes increase the
180 nm by defined dialysis of lipid/detergent mixed micelles, Biochim. antitumor activity of liposomal doxorubicin in syngeneic and human
Biophys. Acta, Biomembr. 640 (1) (1981) 252–262. xenograft mouse tumor models, Neoplasia (New York, NY) 6 (4) (2004) 343.
[25] R.A. Schwendener, M. Asanger, H.G. Weder, n-Alkyl-glucosides as detergents [56] D. Peer, R. Margalit, Loading mitomycin C inside long circulating hyaluronan
for the preparation of highly homogeneous bilayer liposomes of variable sizes targeted nano-liposomes increases its antitumor activity in three mice tumor
(60–240 nm phi) applying defined rates of detergent removal by dialysis, models, Int. J. Cancer 108 (5) (2004) 780–789.
Biochem. Biophys. Res. Commun. 100 (3) (1981) 1055–1062. [57] J. Chen et al., Skin permeation behavior of elastic liposomes: role of
[26] J. Brunner, P. Skrabal, H. Hauser, Single bilayer vesicles prepared without formulation ingredients, Expert Opin. Drug Deliv. 10 (6) (2013) 845–856.
sonication, Physico-chemical properties. Biochim. Biophys. Acta 455 (2) [58] M. Dorrani et al., Development of edge-activated liposomes for siRNA
(1976) 322–331. delivery to human basal epidermis for melanoma therapy, J. Control.
[27] H.G. Enoch, P. Strittmatter, Formation and properties of 1000-A-diameter, Release 228 (2016) 150–158.
single-bilayer phospholipid vesicles, Proc. Natl. Acad. Sci. USA 76 (1) (1979) [59] A. Zeb et al., Improved skin permeation of methotrexate via nanosized
145–149. ultradeformable liposomes, Int. J. Nanomed. 11 (2016) 3813.
[28] T.M. Allen et al., Detergent removal during membrane reconstitution, [60] A.P. Perez et al., Topical amphotericin B in ultradeformable liposomes:
Biochim. Biophys. Acta, Biomembr. 601 (2) (1980) 328–342. formulation, skin penetration study, antifungal and antileishmanial activity
[29] M. Ueno, C. Tanford, J.A. Reynolds, Phospholipid vesicle formation using in vitro, Colloids Surf. B 139 (2016) 190–198.
nonionic detergents with low monomer solubility. Kinetic factors determine [61] G.M. El Zaafarany et al., Role of edge activators and surface charge in
vesicle size and permeability, Biochemistry 23 (13) (1984) 3070–3076. developing ultradeformable vesicles with enhanced skin delivery, Int. J.
[30] P. Schurtenberger et al., Preparation of monodisperse vesicles with variable Pharm. 397 (1–2) (2010) 164–172.
size by dilution of mixed micellar solutions of bile salt and [62] G.M.M El Maghraby, A.C.W., B.W. Barry, Interactions of surfactants (edge
phosphatidylcholine, Biochim. Biophys. Acta, Biomembr. 775 (1) (1984) activators) and skin penetration enhancers with liposomes. International
111–114. Journal of Pharmaceutics, (2004) 276.
[31] T.H. Fischer, D.D. Lasic, A detergent depletion technique for the preparation of [63] Eun Hye Lee, A.K., Yu-Kyoung Oh, Chong-Kook Kim, Effect of edge activators
small vesicles, Mol. Cryst. Liq. Cryst. 102 (5) (1984) 141–153. on the formation and transfection efficiency of ultradeformable liposomes.
[32] R.L. Shew, D.W. Deamer, A novel method for encapsulation of Biomaterials (2005) 26.
macromolecules in liposomes, Biochim. Biophys. Acta, Biomembr. 816 (1) [64] X.C. Xu, Antonio; Burgess, J. Diane, Protein Encapsulation in Unilamellar
(1985) 1–8. Liposomes: High Encapsulation Efficiency and A Novel Technique to Assess
[33] H. Hauser, N. Gains, Spontaneous vesiculation of phospholipids: a simple and Lipid-Protein Interaction, Pharmaceutical Research, 29(7) (2012).
quick method of forming unilamellar vesicles, Proc. Natl. Acad. Sci. 79 (6) [65] T. Ohsawa, H. Miura, K. Harada, Improvement of encapsulation efficiency of
(1982) 1683. water-soluble drugs in liposomes formed by the freeze-thawing method,
[34] S.K. Sundar, M.S. Tirumkudulu, Synthesis of sub-100-nm liposomes via Chem. Pharm. Bull. 33 (9) (1985) 3945–3952.
hydration in a packed bed of colloidal particles, Ind. Eng. Chem. Res. 53 (1) [66] G. Haran et al., Transmembrane ammonium sulfate gradients in liposomes
(2014) 198–205. produce efficient and stable entrapment of amphipathic weak bases, Biochim.
[35] L.J. Cruz, et al., Chapter eight - Targeting Nanoparticles to Dendritic Cells for Biophys. Acta (BBA)-Biomembr. 1151 (2) (1993) 201–215.
Immunotherapy, in: N. Düzgünesß (Ed.), Methods in Enzymology, 2012, [67] L. Mayer, M. Bally, P. Cullis, Uptake of adriamycin into large unilamellar
Academic Press, pp. 143-163. vesicles in response to a pH gradient, Biochim. Biophys. Acta (BBA)-
[36] U. Pick, Liposomes with a large trapping capacity prepared by freezing and Biomembr. 857 (1) (1986) 123–126.
thawing of sonicated phospholipid mixtures, Arch. Biochem. Biophys. 212 (1) [68] E.V. Tazina et al., Specific features of drug encapsulation in liposomes (A
(1981) 186–194. review), Pharm. Chem. J. 45 (8) (2011).
[37] M.-L. Briuglia et al., Influence of cholesterol on liposome stability and on [69] H. He et al., Pharmacokinetics and pharmacodynamics modeling and
in vitro drug release, Drug Deliv. Translational Res. 5 (3) (2015) 231–242. simulation systems to support the development and regulation of
[38] J.N. van der Veen, et al., The critical role of phosphatidylcholine and liposomal drugs, Pharmaceutics 11 (3) (2019).
phosphatidylethanolamine metabolism in health and disease, Elsevier, 2017. [70] M.H. Ali et al., The role of lipid geometry in designing liposomes for the
[39] J. Li et al., A review on phospholipids and their main applications in drug solubilisation of poorly water soluble drugs, Int. J. Pharm. 453 (1) (2013) 225–
delivery systems, Asian J. Pharm. Sci. 10 (2) (2015) 81–98. 232.
[40] J. Senior, G. Gregoriadis, Is half-life of circulating liposomes determined by [71] S. Maritim, Boulas, Pierre & Lin, Yiqing, Comprehensive analysis of liposome
changes in their permeability?, FEBS Lett 145 (1) (1982) 109–114. formulation parameters and their influence on encapsulation, stability and
[41] M. Anderson, A. Omri, The effect of different lipid components on the in vitro drug release in glibenclamide liposomes, Int. J. Pharm. 592 (2021) 120051.
stability and release kinetics of liposome formulations, Drug Delivery 11 (1) [72] G.J. Charrois, T.M. Allen, Drug release rate influences the pharmacokinetics,
(2004) 33–39. biodistribution, therapeutic activity, and toxicity of pegylated liposomal
[42] M. Čuperlović-Culf, 2 - Biology – cancer metabolic phenotype, in: M. doxorubicin formulations in murine breast cancer, Biochim. Biophys. Acta
Čuperlović-Culf (Ed.), NMR Metabolomics in Cancer Research, 2013, (BBA)-Biomembr. 1663 (1–2) (2004) 167–177.
Woodhead Publishing, pp. 15-138. [73] P. Panwar et al., Preparation, characterization, and in vitro release study of
[43] M.S. Webb et al., Sphingomyelin-cholesterol liposomes significantly enhance albendazole-encapsulated nanosize liposomes, Int. J. Nanomed. 5 (2010) 101.
the pharmacokinetic and therapeutic properties of vincristine in murine and [74] Y.e.a Av, Stability Aspects of Liposomes, J. Pharm. Res. 45 (2011).
human tumour models, Br. J. Cancer 72 (4) (1995) 896–904. [75] A. Gabizon, R. Shiota, D. Papahadjopoulos, Pharmacokinetics and tissue
[44] Y. Tokudome et al., Preparation and characterization of ceramide-based distribution of doxorubicin encapsulated in stable liposomes with long
liposomes with high fusion activity and high membrane fluidity, Colloids circulation times, JNCI: J. Natl. Cancer Inst. 81 (19) (1989) 1484–1488.
Surf. B 73 (1) (2009) 92–96. [76] S.C. Semple, A. Chonn, P.R. Cullis, Influence of cholesterol on the association of
[45] N.B. Myant, Chapter 3 - The Distribution of Sterols and Related Steroids in plasma proteins with liposomes, Biochemistry 35 (8) (1996) 2521–2525.
Nature, in: N.B. Myant (Ed.), The Biology of Cholesterol and Related Steroids, [77] S.D. Patil, D.G. Rhodes, D.J. Burgess, Anionic liposomal delivery system for
Butterworth-Heinemann, 1981, pp. 123-159. DNA transfection, AAPS Journal 6 (4) (2004) 13–22.

12
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

[78] E.A. Forssen, Z. Tökès, Use of anionic liposomes for the reduction of chronic [112] M. Seleci et al., Theranostic liposome–nanoparticle hybrids for drug delivery
doxorubicin-induced cardiotoxicity, Proc. Natl. Acad. Sci. 78 (3) (1981) 1873– and bioimaging, Int. J. Mol. Sci. 18 (7) (2017) 1415.
1877. [113] Shihong Li, B.G., Lujun Xhang, Ande Bao, Novel Multifunctional Theranostic
[79] S. Krasnici et al., Effect of the surface charge of liposomes on their uptake by Liposome Drug Delivery System: Construction, Characterization, and
angiogenic tumor vessels, Int. J. Cancer 105 (4) (2003) 561–567. Multimodality MR, Near-Infrared Fluorescent, and Nuclear Imaging.
[80] Y. Kuang et al., Cholesterol-based anionic long-circulating cisplatin liposomes Bioconjugate Chemistry, (2012) 23.
with reduced renal toxicity, Biomaterials 33 (5) (2012) 1596–1606. [114] M.S. Muthu, S.-S. Feng, Theranostic liposomes for cancer diagnosis and
[81] P.R. Clark et al., Studies of direct intratumoral gene transfer using cationic treatment: current development and pre-clinical success, Expert Opin. Drug
lipid-complexed plasmid DNA, Cancer Gene Ther. 7 (6) (2000) 853–860. Deliv. 10 (2) (2013) 151–155.
[82] A.D. Miller, Cationic liposomes for gene therapy, Angew. Chem. Int. Ed. 37 [115] G. Bozzuto, A. Molinari, Liposomes as nanomedical devices, Int. J. Nanomed.
(13–14) (1998) 1768–1785. 10 (2015) 975–999.
[83] P. Guo et al., Inhibiting metastatic breast cancer cell migration via the synergy [116] H.I. Chang, M.K. Yeh, Clinical development of liposome-based drugs:
of targeted, pH-triggered siRNA delivery and chemokine axis blockade, Mol. formulation, characterization, and therapeutic efficacy, Int. J. Nanomed. 7
Pharm. 11 (3) (2014) 755–765. (2012) 49–60.
[84] Y. Xu, F.C. Szoka, Mechanism of DNA release from cationic liposome/DNA [117] U. Bulbake et al., Liposomal formulations in clinical use: an updated review,
complexes used in cell transfection, Biochemistry 35 (18) (1996) 5616–5623. Pharmaceutics 9 (2) (2017).
[85] Y.K. Song et al., Characterization of cationic liposome-mediated gene transfer [118] T. Dragovich et al., A Phase 2 trial of the liposomal DACH platinum L-NDDP in
in vivo by intravenous administration, Hum. Gene Ther. 8 (13) (1997) 1585– patients with therapy-refractory advanced colorectal cancer, Cancer
1594. Chemother. Pharmacol. 58 (6) (2006) 759–764.
[86] D. Christensen et al., Cationic liposomes as vaccine adjuvants, Expert Rev. [119] M. Fleming et al., Colorectal carcinoma: pathologic aspects, J. Gastrointestinal
Vaccines 6 (5) (2007) 785–796. Oncol. 3 (3) (2012) 153–173.
[87] D.P. Vangasseri et al., Immunostimulation of dendritic cells by cationic [120] B.W. Harper et al., Advances in platinum chemotherapeutics, Chemistry 16
liposomes, Mol. Membr. Biol. 23 (5) (2006) 385–395. (24) (2010) 7064–7077.
[88] Y. Inoh et al., Effects of lipid composition in cationic liposomes on [121] M.L. Immordino, F. Dosio, L. Cattel, Stealth liposomes: review of the basic
suppression of mast cell activation, Chem. Phys. Lipids 231 (2020) 104948. science, rationale, and clinical applications, existing and potential, Int. J.
[89] Y. Zhuang et al., PEGylated cationic liposomes robustly augment vaccine- Nanomed. 1 (3) (2006) 297–315.
induced immune responses: Role of lymphatic trafficking and biodistribution, [122] M. Hacker, Chapter 13 - Adverse Drug Reactions, in: M. Hacker, W. Messer, K.
J. Control. Release 159 (1) (2012) 135–142. Bachmann (Eds.), Pharmacology, Academic Press, San Diego, 2009, pp. 327–
[90] S. Dadashzadeh et al., Peritoneal retention of liposomes: effects of lipid 352.
composition, PEG coating and liposome charge, J. Control. Release 148 (2) [123] T.C. Johnstone, P.M. Pil, S.J. Lippard, Cisplatin and Related Drugsq, Reference
(2010) 177–186. Module in Biomedical Sciences, Elsevier, 2015.
[91] D.A. Hume, The mononuclear phagocyte system, Curr. Opin. Immunol. 18 (1) [124] N, P.F., Platinum formulations as anticancer drugs clinical and pre-clinical
(2006) 49–53. studies. Curr Top Med Chem 11(21) (2011) 2623-31.
[92] J.L. Betker et al., Nanoparticle uptake by circulating leukocytes: a major [125] https://www.drugs.com/pro/depocyt.html. (Accessed 3 December 2020)..
barrier to tumor delivery, J. Control. Release 286 (2018) 85–93. [126] K.A. Jaeckle et al., Intrathecal treatment of neoplastic meningitis due to breast
[93] G.L. Scherphof, J.A. Kamps, The role of hepatocytes in the clearance of cancer with a slow-release formulation of cytarabine, Br. J. Cancer (2001),
liposomes from the blood circulation, Prog. Lipid Res. 40 (3) (2001) 149–166. https://doi.org/10.1054/bjoc.2000.1574.
[94] V. Torchilinl, M. Papisov, Why do polyethylene glycol-coated liposomes [127] L. Bomgaars et al., Phase I trial of intrathecal liposomal cytarabine in children
circulate so long?: Molecular mechanism of liposome steric protection with with neoplastic meningitis, J. Clin. Oncol. (2004), https://doi.org/10.1200/
polyethylene glycol: Role of polymer chain flexibility, J. Liposome Res. 4 (1) JCO.2004.01.046.
(1994) 725–739. [128] A. Pellerino et al., Neoplastic meningitis in solid tumors: from diagnosis to
[95] S. Schöttler et al., Protein adsorption is required for stealth effect of poly personalized treatments, Therapeutic Adv. Neurol. Disorders 11 (2018), p.
(ethylene glycol)-and poly (phosphoester)-coated nanocarriers, Nat. 1756286418759618-1756286418759618.
Nanotechnol. 11 (4) (2016) 372–377. [129] D.J. Murry, S.M. Blaney, Clinical pharmacology of encapsulated sustained-
[96] M.J. Ernsting et al., Factors controlling the pharmacokinetics, biodistribution release cytarabine, Ann. Pharmacother. 34 (10) (2000) 1173–1178.
and intratumoral penetration of nanoparticles, J. Control. Release 172 (3) [130] M.J. Glantz et al., A randomized controlled trial comparing intrathecal
(2013) 782–794. sustained-release cytarabine (DepoCyt) to intrathecal methotrexate in
[97] G. Storm et al., Surface modification of nanoparticles to oppose uptake by the patients with neoplastic meningitis from solid tumors, Clin. Cancer Res. 5
mononuclear phagocyte system, Adv. Drug Deliv. Rev. 17 (1) (1995) 31–48. (11) (1999) 3394–3402.
[98] T. Allen, The use of glycolipids and hydrophilic polymers in avoiding rapid [131] Pacira BioSciences, I. PACIRA PHARMACEUTICALS, INC. REPORTS SECOND
uptake of liposomes by the mononuclear phagocyte system, Adv. Drug Deliv. QUARTER 2017 FINANCIAL RESULTS. 2017 [cited 2021 March 06];
Rev. 13 (3) (1994) 285–309. Available from: https://investor.pacira.com/news-releases/news-release-
[99] A. Nagayasu, K. Uchiyama, H. Kiwada, The size of liposomes: a factor which details/pacira-pharmaceuticals-inc-reports-second-quarter-2017-0.
affects their targeting efficiency to tumors and therapeutic activity of [132] V. Wagner et al., The emerging nanomedicine landscape, Nat. Biotechnol. 24
liposomal antitumor drugs, Adv. Drug Deliv. Rev. 40 (1–2) (1999) 75–87. (10) (2006) 1211–1217.
[100] P. Guo et al., Nanoparticle elasticity directs tumor uptake, Nat. Commun. 9 (1) [133] A.H. Faraji, P. Wipf, Nanoparticles in cellular drug delivery, Bioorg. Med.
(2018) 1–9. Chem. 17 (8) (2009) 2950–2962.
[101] R.B. Campbell et al., Cationic charge determines the distribution of liposomes [134] A. Bao et al., Direct 99mTc labeling of pegylated liposomal doxorubicin
between the vascular and extravascular compartments of tumors, Cancer (Doxil) for pharmacokinetic and non-invasive imaging studies, J. Pharmacol.
Res. 62 (23) (2002) 6831–6836. Exp. Ther. 308 (2) (2004) 419–425.
[102] H. Wu et al., Cholesterol-tuned liposomal membrane rigidity directs tumor [135] Y. Barenholz, DoxilÒ–the first FDA-approved nano-drug: lessons learned, J.
penetration and anti-tumor effect, Acta Pharm. Sinica B 9 (4) (2019) 858–870. Control. Release 160 (2) (2012) 117–134.
[103] H.-X. Wang et al., Surface charge critically affects tumor penetration and [136] Institute, N.C. Doxil. [cited 2021 March 06]; Available from: https://
therapeutic efficacy of cancer nanomedicines, Nano Today 11 (2) (2016) www.cancer.gov/publications/dictionaries/cancer-terms/def/doxil.
133–144. [137] P.K. Working, A.D. Dayan, Pharmacological-toxicological expert report.
[104] D.E. Large et al., Advances in receptor-mediated, tumor-targeted drug CAELYX. (Stealth liposomal doxorubicin HCl), Hum. Exp. Toxicol. 15 (9)
delivery, Adv. Therap. 2 (1) (2019) 1800091. (1996) 751–785.
[105] A. Gabizon, H. Shmeeda, Y. Barenholz, Pharmacokinetics of pegylated [138] A. Gabizon, H. Shmeeda, Y. Barenholz, Pharmacokinetics of pegylated
liposomal doxorubicin, Clin. Pharmacokinet. 42 (5) (2003) 419–436. liposomal Doxorubicin: review of animal and human studies, Clin.
[106] Y. Lee, D.H. Thompson, Stimuli-responsive liposomes for drug delivery, Wiley Pharmacokinet. 42 (5) (2003) 419–436.
Interdiscip. Rev. Nanomed. Nanobiotechnol. 9 (5) (2017). [139] B. Carvalho et al., Single-dose, extended-release epidural morphine
[107] E. Heidarli, S. Dadashzadeh, A. Haeri, State of the art of stimuli-responsive (DepoDur) compared to conventional epidural morphine for post-cesarean
liposomes for cancer therapy, Iran J. Pharm. Res. 16 (4) (2017) 1273–1304. pain, Anesth. Analg. 105 (1) (2007) 176–183.
[108] Simoes et al., On the mechanisms of internalization and intracellular delivery [140] M. Alam, C.T. Hartrick, Extended-release epidural morphine (DepoDur): an
mediated by pH-sensitive liposomes., 1. Simoes, S., et al., On the mechanisms of old drug with a new profile, Pain Pract 5 (4) (2005) 349–353.
internalization and intracellular delivery mediated by pH-sensitive [141] H. Pathan, J. Williams, Basic opioid pharmacology: an update, Br. J. Pain
liposomes. Biochimica et Biophysica Acta (BBA)-Biomembranes (2001), (2012), https://doi.org/10.1177/2049463712438493.
https://doi.org/10.1016/s0005-2736(01)00389-3. [142] R. Al-Hasani, M.R. Bruchas, Molecular mechanisms of opioid receptor-
[109] S. Simoes et al., On the formulation of pH-sensitive liposomes with long dependent signaling and behavior, Anesthesiology (2011), https://doi.org/
circulation times, Adv. Drug Deliv. Rev. 56 (7) (2004) 947–965. 10.1097/ALN.0b013e318238bba6.
[110] K. Kono et al., Highly temperature-sensitive liposomes based on a [143] C.T. Hartrick, K.A. Hartrick, Extended-release epidural morphine (DepoDur):
thermosensitive block copolymer for tumor-specific chemotherapy, review and safety analysis, Expert Rev. Neurother. 8 (11) (2008) 1641–1648.
Biomaterials 31 (27) (2010) 7096–7105. [144] Incorporated, I. Arikayce. 2020 [cited 2021 January 05]; Available from:
[111] T. Ta, T.M. Porter, Thermosensitive liposomes for localized delivery and https://www.arikayce.com/.
triggered release of chemotherapy, J. Control. Release 169 (1–2) (2013) [145] Administration, U.S.F.a.D. FDA approves a new antibacterial drug to treat a
112–125. serious lung disease using a novel pathway to spur innovation. 2018

13
D.E. Large, R.G. Abdelmessih, E.A. Fink et al. Advanced Drug Delivery Reviews 176 (2021) 113851

September 28, 2018 [cited 2021 March 16]; Available from: https://www. [161] Incorporated, I. ARIKAYCE (amikacin liposome inhalation suspension), for
fda.gov/news-events/press-announcements/fda-approves-new- oral inhalation use. October 2020 [cited 2021 May 27]; Available from:
antibacterial-drug-treat-serious-lung-disease-using-novel-pathway-spur- https://www.arikayce.com/pdf/full_prescribing_information.pdf.
innovation#:~:text=The%20U.S.%20Food%20and%20Drug,not%20respond% [162] A. Fassas, A. Anagnostopoulos, The use of liposomal daunorubicin
20to%20conventional%20treatment%20( (DaunoXome) in acute myeloid leukemia, Leuk. Lymphoma 46 (6) (2005)
[146] O. Khan, N. Chaudary, The use of amikacin liposome inhalation suspension 795–802.
(arikayce) in the treatment of refractory nontuberculous mycobacterial lung [163] V. Usonis et al., Antibody titres after primary and booster vaccination of
disease in adults, Drug Des., Develop. Therapy 14 (2020) 2287–2294. infants and young children with a virosomal hepatitis A vaccine (EpaxalÒ),
[147] J. Zhang et al., Amikacin Liposome Inhalation Suspension (ALIS) Penetrates Vaccine 21 (31) (2003) 4588–4592.
Non-tuberculous Mycobacterial Biofilms and Enhances Amikacin Uptake Into [164] Pacira Pharmaceuticals, I. EXPAREL (bupivacaine liposome injectable
Macrophages, Front. Microbiol. 9 (2018). suspension). [cited 2021 March 13]; Available from: https://www.
[148] Sciences, N.C.f.A.T. and G.a.R.D.I. Center. Mycobacterium Avium Complex accessdata.fda.gov/drugsatfda_docs/label/2018/022496s9lbl.pdf.
infections. [cited 2021 January 05]; Available from: https://rarediseases.info. [165] F.M. Muggia, Liposomal encapsulated anthracyclines: new therapeutic
nih.gov/diseases/7123/mycobacterium-avium-complex-infections. horizons, Curr. Oncol. Rep. 3 (2) (2001) 156–162.
[149] Incorporated, I., Safety and Tolerability Study of 2 Dose Level of ArikayceTM in [166] G. Pillai, Chapter 9 - Nanotechnology Toward Treating Cancer:
Patients With Bronchiectasis and Chronic Infection Due to Pseudomonas A Comprehensive Review, in: S.S. Mohapatra, et al. (Eds.), Applications of
Aeruginosa. 2008: https://clinicaltrials.gov/ct2/show/NCT00775138. Targeted Nano Drugs and Delivery Systems, Elsevier, 2019, pp. 221-256.
[150] MEDICINE, U.S.N.L.O. AMIKACIN SULFATE Injection, Solution. 2019 April 10, [167] H.H. Chou et al., Pegylated liposomal doxorubicin (Lipo-Dox) for platinum-
2019 [cited 2021 March 17]; Available from: https://dailymed.nlm. resistant or refractory epithelial ovarian carcinoma: a Taiwanese gynecologic
nih.gov/dailymed/drugInfo.cfm?setid=c0f57839-1c9b-49e5-8c7a- oncology group study with long-term follow-up, Gynecol. Oncol. 101 (3)
708e2d16495d. (2006) 423–428.
[151] R. Mischler, I.C. Metcalfe, Inflexal V a trivalent virosome subunit influenza [168] V. Burade et al., LipodoxÒ (generic doxorubicin hydrochloride liposome
vaccine: production, Vaccine 20 (Suppl. 5) (2002) B17–B23. injection): in vivo efficacy and bioequivalence versus CaelyxÒ (doxorubicin
[152] C. Herzog et al., Eleven years of InflexalÒ V—a virosomal adjuvanted influenza hydrochloride liposome injection) in human mammary carcinoma (MX-1)
vaccine, Vaccine 27 (33) (2009) 4381–4387. xenograft and syngeneic fibrosarcoma (WEHI 164) mouse models, BMC
[153] T. Daemen, et al., CHAPTER 2.6 - Liposomes and virosomes as cancer 17 (1) (2017) 405.
immunoadjuvant and antigen-carrier systems in vaccine formulations, in: [169] J.I. Griffin et al., Revealing Dynamics of Accumulation of Systemically Injected
D.D. Lasic, D. Papahadjopoulos (Eds.), Medical Applications of Liposomes, Liposomes in the Skin by Intravital Microscopy, ACS Nano 11 (11) (2017)
Elsevier Science B.V.: Amsterdam, 1998, p. 117-143. 11584–11593.
[154] R. Glück, I.C. Metcalfe, New technology platforms in the development of [170] H.-H. Chou et al., Pegylated liposomal doxorubicin (Lipo-DoxÒ) for platinum-
vaccines for the future, Vaccine 20 (Suppl. 5) (2002) B10–B16. resistant or refractory epithelial ovarian carcinoma: a Taiwanese gynecologic
[155] S.J. Cryz Jr., R. Glück, Immunopotentiating reconstituted influenza virosomes oncology group study with long-term follow-up, Gynecol. Oncol. 101 (3)
as a novel antigen delivery system, Dev. Biol. Stand. 92 (1998) 219–223. (2006) 423–428.
[156] R. Cross, Without these lipid shells, there would be no mRNA vaccines for [171] M.H. Rahimi-Rad, E. Alizadeh, R. Samarei, Aquatic leech as a rare cause of
COVID-19. 2021 MARCH 6, 2021 [cited 2021 March 17]; Available from: respiratory distress and hemoptysis, Pneumologia 60 (2) (2011) 85–86.
https://cen.acs.org/pharmaceuticals/drug-delivery/Without-lipid-shells- [172] L. Kager, U. Pötschger, S. Bielack, Review of mifamurtide in the treatment of
mRNA-vaccines/99/i8. patients with osteosarcoma, Ther. Clin. Risk Manag. 6 (2010) 279–286.
[157] N. Pardi et al., mRNA vaccines — a new era in vaccinology, Nat. Rev. Drug [173] K. Gardikis et al., New chimeric advanced Drug Delivery nano Systems (chi-
Discovery 17 (4) (2018) 261–279. aDDnSs) as doxorubicin carriers, Int. J. Pharm. 402 (1–2) (2010)
[158] K.V. Clemons, D.A. Stevens, Comparative efficacies of four amphotericin B 231–237.
formulations–Fungizone, amphotec (Amphocil), Am Bisome, and Abelcet– [174] Merrimack Pharmaceuticals, I. ONIVYDE (irinotecan liposome injection).
against systemic murine aspergillosis, Antimicrob. Agents Chemother. 48 (3) 2015 October 2015 [cited 2021 June 14]; Available from: https://www.
(2004) 1047–1050. accessdata.fda.gov/drugsatfda_docs/label/2015/207793lbl.pdf.
[159] J. Tollemar, O. Ringdén, Lipid formulations of amphotericin B. Less toxicity [175] K.-I. Fujita et al., Irinotecan, a key chemotherapeutic drug for metastatic
but at what economic cost?, Drug Saf 13 (4) (1995) 207–218. colorectal cancer, World J. Gastroenterol. 21 (43) (2015) 12234–12248.
[160] D.W. Denning et al., NIAID mycoses study group multicenter trial of oral [176] Guidelines for using verteporfin (Visudyne) in photodynamic therapy for
itraconazole therapy for invasive aspergillosis, Am. J. Med. 97 (2) (1994) choroidal neovascularization due to age-related macular degeneration and
135–144. other causes: update. Retina, 25(2) (2005) 119-34.

14

You might also like