0% found this document useful (0 votes)
78 views93 pages

Advanced Drug Delivery Systems

Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
0% found this document useful (0 votes)
78 views93 pages

Advanced Drug Delivery Systems

Copyright
© © All Rights Reserved
We take content rights seriously. If you suspect this is your content, claim it here.
Available Formats
Download as PDF, TXT or read online on Scribd
You are on page 1/ 93

EDITOR

ADVANCED DRUG DELIVERY TECHNOLOGIES


PROF. DR. EMEL ÖYKÜ ÇETİN UYANIKGİL
ADVANCED DRUG DELIVERY
TECHNOLOGIES
ADVANCED
EDITOR:
PROF. DR. EMEL ÖYKÜ ÇETİN UYANIKGİL
DRUG DELIVERY
Prof. Dr. Emel Öykü ÇETİN UYANIKGİL graduated
from Hacettepe University Faculty of Pharmacy and
completed her master's degree at Hacettepe
University Institute of Health Sciences, Department
TECHNOLOGIES
of Pharmaceutical Technology/ Biopharmaceutics
and Pharmacokinetics. She became a research
assistant at Ege University, Faculty of Pharmacy,
Department of Pharmaceutical Technology/
Biopharmaceutics and Pharmacokinetics in 2000
and started her phd. In 2009, he became assistant
professor, in 2014 associated professor and in 2021

PROF. DR. EMEL ÖYKÜ ÇETİN UYANIKGİL


professor. She has publications in national and
international journals, oral and poster presentations
in national and international congresses
/symposiums. She is a researcher and executive in
many projects. She also has project support awards
and a patent in her research field. She has studies in
many areas, mainly formulation development, drug
delivery systems, cancer, wound and burn models, EDITOR
and diabetes.

www.bztturanpublishinghouse.com
/bztturanpublishinghouse
EDITOR

ADVANCED DRUG DELIVERY TECHNOLOGIES


PROF. DR. EMEL ÖYKÜ ÇETİN UYANIKGİL
ADVANCED DRUG DELIVERY
TECHNOLOGIES
ADVANCED
EDITOR:
PROF. DR. EMEL ÖYKÜ ÇETİN UYANIKGİL
DRUG DELIVERY
Prof. Dr. Emel Öykü ÇETİN UYANIKGİL graduated
from Hacettepe University Faculty of Pharmacy and
completed her master's degree at Hacettepe
University Institute of Health Sciences, Department
TECHNOLOGIES
of Pharmaceutical Technology/ Biopharmaceutics
and Pharmacokinetics. She became a research
assistant at Ege University, Faculty of Pharmacy,
Department of Pharmaceutical Technology/
Biopharmaceutics and Pharmacokinetics in 2000
and started her phd. In 2009, he became assistant
professor, in 2014 associated professor and in 2021

PROF. DR. EMEL ÖYKÜ ÇETİN UYANIKGİL


professor. She has publications in national and
international journals, oral and poster presentations
in national and international congresses
/symposiums. She is a researcher and executive in
many projects. She also has project support awards
and a patent in her research field. She has studies in
many areas, mainly formulation development, drug
delivery systems, cancer, wound and burn models, EDITOR
and diabetes.

www.bztturanpublishinghouse.com
/bztturanpublishinghouse
ADVANCED DRUG DELIVERY TECHNOLOGIES

Editor

Emel Öykü Çetin Uyanıkgil


Published by
BZT TURAN PUBLISHINGHOUSE
Certificate Number: 202401
Delaware, United States
and Baku, Azerbaijan

www.bztturanpublishinghouse.com
[email protected]

ADVANCED DRUG DELIVERY TECHNOLOGIES

Emel Öykü Çetin Uyanıkgil

Language: English
Publication Date: December 2024
Cover designed by Kağan SARGI
Print and digital versions typeset by BZT TURAN Media Co. Ltd.

E-ISBN: 9 7 8 - 9 9 5 2 - 8 5 4 1 - 5 - 2
DOI: https://doi.org/10.30546/19023.978-9952-8541-1-4.2024.002.

OPEN ACCESS
Suggested Citiation:
Uyanıkgil, E. (2024). ADVANCED DRUG DELIVERY TECHNOLOGIES. BZT TURAN PUBLISHING
HOUSE.
DOI: https://doi.org/10.30546/19023.978-9952-8541-1-4.2024.002.
Editor

Prof. Dr. Emel Öykü ÇETİN UYANIKGİL graduated from Hacettepe University Faculty
of Pharmacy and completed her master's degree at Hacettepe University Institute of
Health Sciences, Department of Pharmaceutical Technology/ Biopharmaceutics and
Pharmacokinetics. She became a research assistant at Ege University, Faculty of
Pharmacy, Department of Pharmaceutical Technology/ Biopharmaceutics and
Pharmacokinetics in 2000 and started her phd. In 2009, he became assistant professor,
in 2014 associated professor and in 2021 professor. She has publications in national
and international journals, oral and poster presentations in national and international
congresses/symposiums. She is a researcher and executive in many projects. She also
has project support awards and a patent in her research field. She has studies in many
areas, mainly formulation development, drug delivery systems, cancer, wound and
burn models, and diabetes.
Preface Emulgels, microemulgels, and
nanoemulgels represent a fascinating blend
In recent years, the field of pharmaceutical
of emulsions and gels, combining the
sciences has witnessed remarkable
advantages of both systems. These
advancements in drug delivery systems.
formulations offer improved bioavailability,
Among these, the development of
controlled release, and enhanced stability,
emulgels, microemulgels, nanoemulgels,
making them suitable for various
niosomes, and exosomes has opened new
therapeutic applications. In this book, we
horizons for effective and targeted drug
delve into the formulation, characterization,
delivery. This book aims to provide a
and evaluation of these emulgel-based
comprehensive overview of these
systems, providing insights into their
innovative carrier systems, highlighting
unique properties and potential uses.
their potential applications, benefits, and
future prospects. Niosomes, another cutting-edge carrier
system, have garnered significant attention
In the evolving landscape of
due to their ability to encapsulate both
pharmaceutical sciences, the formulation
hydrophilic and lipophilic drugs. As non-
and delivery of poorly water-soluble drugs
ionic surfactant vesicles, niosomes offer
present significant challenges. Self-
advantages such as biocompatibility,
Emulsifying Drug Delivery Systems
improved drug stability, and controlled
(SEDDS) have emerged as a promising
release. We explore the formulation
solution to enhance the bioavailability of
techniques, physicochemical properties,
these drugs. This innovative approach
and therapeutic applications of niosomes,
leverages the unique properties of self-
showcasing their versatility and efficacy.
emulsifying formulations to facilitate the
absorption and efficacy of therapeutic The secretome, particularly in the form of
compounds. This document aims to provide exosomes, has emerged as a promising
a comprehensive overview of SEDDS, cell-free carrier system. Exosomes,
exploring their mechanisms, formulation nanometer-sized extracellular vesicles, play
strategies, and potential applications in a crucial role in intercellular communication
modern medicine. By delving into the and hold immense potential for drug
intricacies of SEDDS, we aspire to shed light delivery and regenerative medicine. This
on their transformative potential and the book provides an in-depth analysis of the
future directions of drug delivery research. secretome, focusing on the isolation,
characterization, and therapeutic
applications of exosomes, highlighting their innovation in the field of drug delivery
role as natural and efficient drug carriers. systems. As we continue to unravel the
potential of self-emulsifying drug delivery
Through a detailed exploration of these
systems, emulgels, niosomes, secrotome
advanced carrier systems, this book aims to
and exosomes, we move closer to realizing
bridge the gap between research and
more effective, targeted, and personalized
clinical practice, offering valuable insights
therapies for a wide range of diseases.
for scientists, researchers, and healthcare
professionals. The chapters presented here Sincerely,
are authored by leading experts in the field,
ensuring a thorough and up-to-date
understanding of the topics covered. İzmir, Türkiye
Prof. Dr. Emel Öykü Çetin Uyanıkgil
We hope this book serves as a valuable
02.07.2024
resource, inspiring further research and
Authors List

Ayşegül Taşkıran
E-mail: taskiranaysegü[email protected]
ORCID: 0000-0001-9780-6948

Derya Erışık
E-mail: [email protected]
ORCID: 0000-0002-8199-6051

Emel Öykü Çetin Uyanıkgil


E-mail: [email protected]; [email protected]
ORCID: 0000-0001-8822-9130

Emine Esin Çalışkan


E-mail:[email protected]
ORCID: 0000-0002-8837-1222

Esra Demirtürk
E-mail: [email protected]
ORCID: 0000-0002-8918-0073

Yalçın Çelik Aydın


E-mail: [email protected]
ORCID: 0000-0001-5549-1087

Yiğit Uyanıkgil
E-mail : [email protected] ; [email protected]
ORCID: 0000-0002-4016-0522
CONTENTS

Chapter 1 ............................................................................................... 1
Evaluation of Emulgel, Microemulgel and Nanoemulgel Formalitions
ESRA DEMİRTÜRK

Chapter 2 .............................................................................................. 21

Exosomes
DERYA ERIŞIK , YİĞİT UYANIKGİL

Chapter 3 ............................................................................................... 34
Niosomes
EMİNE ESİN ÇALIŞKAN , EMEL ÖYKÜ ÇETİN UYANIKGİL

Chapter 4 ............................................................................................... 51
Self-Emulsifying Drug Delivery Systems (SEDDS)
YALÇIN ÇELIK AYDIN, EMEL ÖYKÜ ÇETİN UYANIKGİL

Chapter 5 ............................................................................................... 70
The Secretome: Cell-free carrier system
AYŞEGÜL TAŞKIRAN
EVALUATION OF EMULGEL,
MICROEMULGEL AND NANOEMULGEL
FORMALITONS

Esra Demirtürk
Chapter 1 the dispersion of at least two
immiscible liquids, usually oil and
water, in each other with the help
Evaluation of
of surface active substances

Emulgel, (surfactants). If the emulsion


contains colloidal oil droplets in
Microemulgel and water, this emulsion type is oil
(o/w) and if it contains water
Nanoemulgel
droplets in oil, it can be water
Formalitions (w/o) type. Surfactant addition is
required to ensure stability in
ESRA DEMİRTÜRK1 thermodynamically unstable
emulsion formulations.
Introduction Surfactants are amphiphilic
molecules containing hydrophilic
Emulgel is a versatile topical
and lipophilic ends together, which
formulation that combines the
prevent the coalescence of
properties of both emulsion and
colloidal droplets in the inner
gel. Emulsions are heterogeneous
phase and help them to remain
systems with homogeneous
stable by homogeneously
appearance consisting of colloidal
distributing in the continuous
droplets as the dispersed phase
phase. In this way, they can form
(inner phase) and continuous
stable systems by binding to the
phase (outer phase) as a result of
water molecule with their

1 Çukurova University, Faculty of


Pharmacy, Department of pharmaceutical
Technology, Adana, 01330, Türkiye
hydrophilic ends and to the oil biocompatibility with living tissues,
molecule with their hydrophobic making it well-tolerated by the
ends in the formulation. skin. Its soft consistency and high
swelling degree also contribute to
If the emulsion contains colloidal
its similarity with living tissues,
oil droplets in water, this emulsion
further enhancing its compatibility
type is oil (o/w) and if it contains
with the body. Overall, the topical
water droplets in oil, it can be
usage of emulgel offers significant
water (w/o) type. It offers several
advantages such as improved
advantages, including improved
stability, enhanced drug release,
stability, enhanced drug release,
better skin penetration, sustained
and better skin penetration.
release of active ingredients, and
Emulgel allows for the
excellent biocompatibility with
incorporation of various categories
living tissues.
of drugs such as antibacterials,
antifungals, and antivirals, making Microemulgel and nanoemulgel
it suitable for a wide range of are two types of emulgels that
therapeutic applications. differ in the size of their dispersed
Additionally, emulgel can provide a phase particles. Microemulgel
sustained release of active refers to an emulgel with smaller
ingredients, ensuring prolonged dispersed phase particles, typically
therapeutic effect. This is achieved ranging from 10 to 1000
through the ability of emulgel to nanometers in size. Nanoemulgel,
encapsulate water-soluble on the other hand, has even
compounds and control the smaller dispersed phase particles,
release of drugs. Furthermore, typically ranging from 1 to 100
emulgel exhibits excellent nanometers in size. The main
advantage of microemulgel is that emulsifying agent. It is commonly
it provides increased stability and used in topical preparations for
enhanced drug release properties localized drug delivery, as it offers
compared to traditional emulgels benefits such as enhanced drug
(Sharma et al., 2016). On the penetration and prolonged
other hand, nanoemulgel offers release. Microemulgel, on the
even greater stability and other hand, is a combination of
improved drug release properties, microemulsion and gel. It
thanks to its much smaller particle combines the advantages of both
size. This smaller particle size systems, with the microemulsion
allows for better penetration and providing improved drug solubility
absorption of the active and the gel providing sustained
ingredients into the skin, leading release properties. Nanoemulgel
to more effective drug delivery. takes it a step further by
Additionally, nanoemulgels have a incorporating nanoemulsions into
higher surface area-to-volume the gel matrix (Sharma et al.,
ratio, which can result in faster 2016). It is important to note that
drug release kinetics. further research and studies are
still being conducted to fully
Emulgel, microemulgel, and understand the characteristics and
nanoemulgel are all types of potential applications of emulgel,
emulsions that have been microemulgel, and nanoemulgel.
extensively studied and developed While there is still ongoing
in recent years. Emulgel refers to research, emulgel, microemulgel,
a type of gel that is formed by and nanoemulgel have shown
combining an oil phase and a promise as drug delivery systems
water phase with the help of an
due to their ability to improve drug structure, ingredient interactions,
penetration, provide sustained and bioavailability of lipid-soluble
release, and enhance stability. nutrients in the emulgel
formulation. The tests include
Characterization studies of
evaluating the cosmetic properties
topical dosage forms
of oleuropein in the emulgel
against UVB-induced erythema,
It is necessary to assess the
assessing the protective and
pharmaceutical product to
lenitive effects of the emulgel on
guarantee quality and uniformity
healthy volunteers, and
throughout batches. These tests
conducting sensory analysis to
aid in comprehending the stability
evaluate attributes such as
and behavior of the product. Few
appearance, texture, smoothness,
universal tests, such as those for
color, odor, spreadability, and
description, identification, assay,
overall acceptance of the
and contaminants, exist for every
developed emulgel product (Rai et
specific dosage form, according to
al., 2018). The in vitro
USP. A topical dosage form must
characterization tests conducted
pass a few particular tests, as
on the emulgel formulation
determined by USP on an
involved various aspects such as
individual basis: pH, particle size,
rheological studies, stability
sterility, water content,
testing using centrifugation and
microbiological limits, antioxidant
accelerated ageing tests, and
and antibacterial content, and the
viscosity studies. The rheological
polymorphic character of the API.
studies were performed using
In vitro characterization tests aim
continuous shear and oscillation
to understand the interfacial
experiments at high shear rates. enhance their penetration, while
During the rheological studies, the stabilizing against gravitational
viscosity of the emulgel separation and flocculation. In the
formulation was measured as a preparation of emulsion-based
function of shear rate. The results gels, it is necessary to examine the
of the viscosity studies indicated important process parameters that
the flow behavior of the emulgel have a significant effect on the size
formulation under different shear and formulation stability. In order
rates. The emulgel formulation to accomplish this, we must select
exhibited certain rheological the proper preparation process at
properties, such as shear-thinning the early stages.
behavior, which means that its
viscosity decreased with Some of the commonly used in

increasing shear rate. One vivo characterization tests include

important characteristic of these skin irritation tests, skin

formulations is the droplet size permeation studies, and efficacy

distribution, particularly in evaluations. In skin irritation tests,

nanoemulsions (Kumar et al., the emulgels and microemulgels

2019).The droplet size in are applied to the skin of test

nanoemulsions is typically in the subjects to evaluate their potential

range of 20-200 nm, which allows irritant effects. Skin permeation

for enhanced drug solubilityand studies involve applying

improved bioavailability (Jaiswal et formulations to the skin and

al., 2014).This small droplet size measuring the penetration of

also enables the nanoemulsions to active ingredients into underlying

uniformly deposit on the skin and tissues to assess their ability to


deliver therapeutic effects (Kumar
et al., 2018).Efficacy evaluations rely on extensive in vivo studie
assess the desired effects of the and human clinical trials.
emulgels and microemulgels, such
Table 1 shows the comparativ
as moisturization, wrinkle
permeation of different activ
reduction, or acne treatment (Ma
ingredients through the ski
et al., 2021). These tests are
following application as emulge
essential for assessing the safety,
microemulgel and nanoemulge
effectiveness, and performance of
Since emulsions were used in th
emulgels and microemulgels in
preparation of these formulation
real-life scenarios.In addition to
some studies also included
these in vivo characterization
comparison of microemulsion an
tests, it is also crucial to conduct
microemulgel formulations.
human clinical trials to further
validate the results obtained from
the preliminary in vivo studies. In
the meantime, the knowledge
obtained from in vitro experiments
on model emulsion systems can
provide valuable insights into the
mechanisms of lipid and protein
digestion, aswell as the
bioavailability of lipid-soluble
nutrients. However, it is important
to note that ultimate validation of
emulgels and microemulgels will
Table 1. Clinical study results of different active ingredients

Active Route of Emulgel/ Therapeutic Globule Size Formulation Skin


Ingredient Administration Nanoemulgel/ Outcome Composition Permeability
(References) Microemulgel
Mupirocin Topical Nanoemulgel Antibacterial 1500 nm Oil: Eucalyptus Lag time (h):
(Alhasso et Topical (nanoemulgel) oil , Eucalyptol Nanoemulgel:
al., 2023) infections Surfactant: 1.89
treatment Tween 80 Control: 2.64
Co-surfactant: Cumulative drug
Span 80 permeation 24 h
Gelling agent: (µg/cm2):
Carbopol 940 Nanoemulgel:
1194.07 ± 91.96
Control: 2242.79
± 262.17
The drug flux at
steady state
(flux) (μg/cm2
/h):
Nanoemulgel:
64.92 ± 7.52
Control: 112.42
± 3.75
The permeation
coefficient
(×10−4 cm/h):
Nanoemulgel:
60.67 ± 0.22
Control: 107.14
± 2.73
Enhancement
ratio
Nanoemulgel:
0.53
Control: 1
Tacrolimus Topical Nanoemulgel Treatment of 116.3 ± 20.8 nm Oil: Fish oil (f) The drug flux at
(Mittal et al., psoriasis (nanoemulsion) and Linseed oil steady state
2021) (L) (flux) (μg/cm2
Surfactant: /h):
Tween 80 Nanoemulgel:
Co-surfactant: 14.25
Transcutol-P Marketed
Gelling agent: ointment: 10.42
Carbopol 934 Cumulative
permeation in 24
h (%):
Nanoemulgel:
%54.33
Marketed
ointment: %35.5
The permeation
coefficient
(cm2/h):
Nanoemulgel:
2.850 × 10-2
Marketed
ointment: 2.084
× 10-2
Table 1. Clinical study results of different active ingredients (cont.)

Active Route of Emulgel/ Therapeutic Globule Size Formulation Skin Permeability


Ingredient Administration Nanoemulgel/ Outcome Composition
(References) Microemulgel
Piroxicam Topical Nanoemulgel Anti-inflammatory <200 nm Oil: Oleic acid Cumulative
(Dhawan et (nanoemulsion) Surfactant: amount of drug
al., 2014) Tween 80 permeated
Co-surfactant: (mg/cm2):
Ethanol Drug Solution:
Gelling agent: 0.83±0.02
Carbopol 934 Plain Drug gel:
0.93±0.03
Marketed
formulation:
0.96±0.05
Nanomulsion:
1.01±0.06
Nanoemulgel:
1.0±0.04
Drug permeated
%
Drug Solution:
81.5
Plain Drug gel:
91.3
Marketed
formulation: 94.2
Nanomulsion:
99.1
Nanoemulgel:
98.2
The drug flux at
steady state (flux)
(mg/cm2/h):
Drug Solution:
0.032±0.01
Plain Drug gel:
0.037±0.02
Marketed
formulation:
0.040±0.02
Nanomulsion:
0.044±0.04
Nanoemulgel:
0.042±0.003
Lag Time (h):
Drug Solution:
0.79±0.11
Plain Drug gel:
0.72±0.25
Marketed
formulation:
0.55±0.09
Nanomulsion:
0.57±0.05
Nanoemulgel:
0.41±0.03
Enhancement
ratio
Drug Solution: 1
Plain Drug gel:
1.15
Marketed
formulation: 1.25
Nanomulsion:
1.37
Nanoemulgel:
1.31
Drug Retained
(mg):
Drug Solution:
0.92±0.02
Plain Drug gel:
0.43±0.03
Marketed
formulation:
0.28±0.02
Nanomulsion:
0.04±0.02
Nanoemulgel:
0.09±0.001
Local
accumulation
efficiency
(piroxicam
retained into skin)
Drug Solution:
1.10
Plain Drug gel:
0.46
Marketed
formulation: 0.29
Nanomulsion:
0.03
Nanoemulgel:
0.09
Table 1. Clinical study results of different active ingredients (cont.)

Active Route of Emulgel/ Therapeutic Globule Size Formulation Skin Permeability


Ingredient Administration Nanoemulgel/ Outcome Composition
(References) Microemulgel
Desonide Topical Nanoemulgel Treatment of 38,3 ± 1,47 nm Oil: Cumulative
(Mittal et skin disorders (nanoemulsion) Triacetin,Eucalyptus amount of
al., 2021) oil, oleic acid permeation in 24
Surfactant: Tween h (μg):
80, Span 80, Nanoemulgel:
Poloxamer 407, 133.55 ± 6.47
Co-surfactant: Gel: 96.24 ± 3.94
Transcutol® P Nanoemulsion:
Gelling agent: 198.23 ± 8.72
Carbopol 980 Cream: 47.77 ±
3.05
The drug flux at
steady state
(flux)
(μg/cm2/h):
Nanoemulgel:
6.99 ± 0.70
Gel: 5.43 ± 0.59
Nanoemulsion:
10.92 ± 1.73
Cream: 2.72 ±
0.57
The permeation
coefficient
(cm/h) × 10−3:
Nanoemulgel:
13.98
Gel: 10.86
Nanoemulsion:
21.84
Cream: 5.44
Enhancement
ratio of Kp
Nanoemulgel:
2.57
Gel: 2.00
Nanoemulsion:
4.01
Cream: 1
Curcumin Topical Microemulgel Analgesicand 41,91 ± 0,11 nm Oil: Glycerol Cumulative drug
(Niu et al., anti- (microemulsion) octanoate decanoate permeation 24 h
2023) inflammatory 186,45 ± 0,75 (GTCC) (µg/cm2):
activity nm Surfactant: Pluronic Solution: 1.08 ±
(microemulgel) F68 0.19
Co-surfactant: Microemulsion:
PEG400 4.83 ± 0.45
Gelling agent: Microemulgel:
Chitosan 3.78 ± 0.26
The skin retention
(µg/cm2):
Solution: 0.08 ±
0.01
Microemulsion:
0.31 ± 0.08
Microemulgel:
0.43 ± 0.16
Table 1. Clinical study results of different active ingredients (cont.)

Active Route of Emulgel/ Therapeutic Globule Size Formulation Skin Permeability


Ingredient Administration Nanoemulgel/ Outcome Composition
(References) Microemulgel
Luliconazole Topical Nanoemulgel Treatment of 86,62 nm Oil: Clove oil *Rat Skin
(Kaur et al., fungal infections (nanoemulsion) Surfactant: The drug flux at
2023) Transcutol P, steady state (flux)
Tween 20 (μg/cm2/h):
Co-surfactant: Pure Drug: 36.43
PEG 600 Marketed
Gelling agent: formulation: 69.60
Carbopol 934 Nanoemulgel:
95.59
The permeation
coefficient
(cm/h)103:
Pure Drug: 7.28
Marketed
formulation: 13.92
Nanoemulgel:
19.11
*Mice Skin
The drug flux at
steady state (flux)
(μg/cm2/h):
Pure Drug: 33.19
Marketed
formulation: 66.47
Nanoemulgel:
93.94
The permeation
coefficient
(cm/h)103:
Pure Drug: 6.63
Marketed
formulation: 13.29
Nanoemulgel:
18.78
*Strat-M
membrane
The drug flux at
steady state (flux)
(μg/cm2/h):
Pure Drug: 36.16
Marketed
formulation: 71.55
Nanoemulgel:
100.48
The permeation
coefficient
(cm/h)103:
Pure Drug: 7.43
Marketed
formulation: 14.31
Nanoemulgel:
20.09
Table 1. Clinical study results of different active ingredients (cont.)

Active Route of Emulgel/ Therapeutic Globule Size Formulation Skin Permeability


Ingredient Administration Nanoemulgel/ Outcome Composition
(References) Microemulgel
Calcipotriol Topical Emulgel Treatment of 10 -15 μm Oil: Liquid *dialysis
(Varma et psoriasis (emulgel) Paraffin, membrane
al., 2014) Kollicream3C, The drug flux at
KolliphorCS20 steady state (flux)
Surfactant: (μg/cm2/h):
Propylene glycol Emulgel: 1.548
Co-surfactant: Ointment: 1.067
PEG 400 *male rat skin
Gelling agent: The drug flux at
Carbopol steady state (flux)
(μg/cm2/h):
Emulgel: 1.212
Ointment: 0.816
Lacidipine Topical Microemulgel Treatment of 30,5 nm Oil: Isopropyl The drug flux at
(Gannu et hypertension and (microemulgel) myristate steady state (flux)
al., 2010) atherosclerosis Surfactant: (μg/cm2/h):
Tween 80 Microemulsion:
Co-surfactant: 64,7
Labrasol Microemulgel: 46,1
Gelling agent: Solution: 8,0
HPMC K 4 M Cumulative amount
of permeation in
24 h (μg):
Microemulsion:
7704,5
Microemulgel:
5820,8
Solution: 910,3
Lag Time (h):
Microemulsion:
2.17
Microemulgel: 2.22
Solution: 2.83
Table 1. Clinical study results of different active ingredients (cont.)

Active Route of Emulgel/ Therapeutic Globule Size Formulation Skin Permeability


Ingredient Administration Nanoemulgel/ Outcome Composition
(References) Microemulgel
Clobetasol- Topical Nanoemulgel Treatment of 37,16 ± 21,16 Oil: Oleic acid The drug flux at
17- psoriasis nm Surfactant: steady state (flux)
propionate (nanoemulgel) Tween 80 (μg/cm2/h):
(Devadiga et Co-surfactant: Nanoemulgel:
al., 2024) Ethanol 498,66 ± 14,40
Gelling agent: Marketed
Carbopol 940 formulation: 383,9
± 8,59
The permeation
coefficient
(μg/min):
Nanoemulgel: 1,6 x
10 -4
Marketed
formulation: 1,27 x
10 -4
The skin retention
(%):
Nanoemulgel:
40.86 ± 0.61
Marketed
formulation: 25.90
± 2.33
Clobetasol Topical Microemulgel Treatment of 18,26 nm Oil: Isopropyl Cumulative amount
propionate vitiligo (microemulsion) myristate of permeation in 8
(Patel et al., Surfactant: h (μg/cm-2):
2013) Cremophor EL Microemulsion:
Co-surfactant: 53,6 ± 2,18
Isopropyl Microemulgel:
alcohol 28,43 ± 0,67
Gelling agent: Marketed
Carbopol 934P formulation: 37,73
± 0,77
The skin retention
(%):
Microemulsion:
31.19 ± 3.16
Microemulgel: 49.3
± 4.12
Table 1. Clinical study results of different active ingredients (cont.)

Active Route of Emulgel/ Therapeutic Globule Size Formulation Skin


Ingredient Administration Nanoemulgel/ Outcome Composition Permeability
(References) Microemulgel
Pseudolaric Topical Microemulgel Treatment of 16,08 nm Oil: The drug flux at
acid B fungal (microemulsion) Isopropylmyristate steady state
(Wan, Tao et infections Surfactant: (flux)
al., 2015) Cremphor EL (μg/cm2/h):
Co-surfactant: Microemulgel:
Transcutol P 1,844
Gelling agent: Microemulsion:
Carbopol 2,25
Gel: 0,517
Cumulative
amount of
permeation in24
h (μg/cm2):
Microemulgel:
31,89
Microemulsion:
50,31
Gel: 13,11
Erythromycin Topical Emulgel Treatment of 256,2 nm Oil: Cinnamon Oil The drug flux at
(Abdallah et skin bacterial (transethosomes) Surfactant: Span 80 steady state
al., 2023) infection Co-surfactant: (flux)
Tween 80 (μg/cm2/h):
Gelling agent: Transethosomal
HPMC emulgel: 120.19
Transethosomal
gel: 102.14
Suspension:
50.77
Cumulative
amount of
permeation in 6
h (μg/cm2):
Transethosomal
emulgel: 792,48
± 18,82
Transethosomal
gel: 716,93 ±
33,26
Suspension:
415,13 ± 11,29
Evaluation of Emulgel, Microemulgel and Nanoemulgel Formulations

Table 1. Clinical study results of different active ingredients (cont.)

Active Route of Emulgel/ Therapeutic Globule Size Formulation Skin Permeability


Ingredient Administration Nanoemulgel/ Outcome Composition
(References) Microemulgel
Levamisole Topical Microemulgel Enhancers the 19,3 ± 0,1 nm Oil: Maisine™ Cumulative drug
(Hu, Qing et release of (microemulsion) 35–1 permeation 36 h
al., 2021) cytokines 62,1 ± 2,5 Surfactant: (µg/cm2):
(microemulgel) Cremophor® Liniment: 162.49 ±
RH40 11.74
Co-surfactant: Gel: 140.34 ±
Transcutol® 40.87
HP Microemulsion:
Gelling agent: 729.47 ± 48.04
Alginate- Microemulgel:
boronic acid 655.82 ± 9.42
Ibuprofen Topical Emulgel Treatment of 2-3 µm Oil: Oleyl Cumulative amount
(Jameel et al., pain and (emulsion) alcohol of drug permeated
2019) inflammation Surfactant: at 24 h (μg/cm2):
Polysorbate 60 Emulgel: 1791 ±
Co-surfactant: 148
Span 80 Marketed gel: 880
Gelling agent: ± 181
Carbomer 940 The drug flux at
steady state (flux)
(μg/cm2/h):
Emulgel: 110.4 ±
14.3
Marketed gel:
101.8 ± 5.5
Lag time (h):
Emulgel: 1.6 ± 0.4
Marketed gel: 0.8
± 0.3
The permeation
coefficient
(cm/h)103:
Emulgel: 2.1 ± 0.3
Marketed gel: 2.0
± 0.1
Metronidazole Topical Emulgel Treatment of Oil: Capmul The drug flux at
(Rao et al., topical infections 908 P steady state (flux)
2013) Surfactant: (μg/cm2/h):
Acconon Emulgel: 351.78 ±
Co-surfactant: 2.23
Propylene Gel: 96.27 ± 2.54
glycol The permeation
Gelling agent: coefficient (cm/h)
Xanthan gum × 10−3:
Emulgel: 35.17 ±
2.45
Gel: 9.62 ± 2.64
Cumulative amount
of permeation in
12h (μg/cm2):
Emulgel:
2589.17±4.21
Gel: 1102.86±4.65
Conclusion the formulation development
process were presented. The
There are some primary qualities of a nano-emulgel are
considerations when choosing an mostly determined by the choice
active substance for topical of components and the suitable
application, such as non-polarity, ratios between them. A departure
small molecular size, molecular from this might have an impact on
mass of less than 500 Daltons and the thermodynamic stability and
half-life of less than 10 hours. In the transformation of a nano-
this review, the results of in vitro emulsion into a nano-emulgel.
in vivo characterization studies of Because of its less mobile
emulgel, microemulgel and dispersion phase and reduced
nanoemulgel formulations interfacial tension, the nano-
prepared with different active emulgel is more stable than a
ingredients were collected and nano-emulsion
data that may guide researchers in
.
References

Abdallah, M. H., Elghamry, H. A., Khalifa, N. E., Khojali, W. M., Khafagy, E. S., Shawky, S.,
... & El-Housiny, S. (2023). Development and Optimization of Erythromycin Loaded
Transethosomes Cinnamon Oil Based Emulgel for Antimicrobial Efficiency. Gels, 9(2),
137. https://doi.org/10.3390/gels9020137

Alhasso, B., Ghori, M. U., & Conway, B. R. (2023). Development of a Nanoemulgel for the
Topical Application of Mupirocin. Pharmaceutics, 15(10), 2387.
https://doi.org/10.3390/pharmaceutics15102387

Devadiga, S., Sermasekaran, A., Singh, A. D., Agrawal, S., Sharma, S., & Choudhary, D.
(2024). Quality by design driven systematic development of nanoemulgel of
clobetasol-17-propionate for effective treatment of psoriasis. Journal of Drug
Delivery Science and Technology, 105422.
https://doi.org/10.1016/j.jddst.2024.105422

Dhawan, B., Aggarwal, G., & Harikumar, S. L. (2014). Enhanced transdermal permeability of
piroxicam through novel nanoemulgel formulation. International journal of
pharmaceutical investigation, 4(2), 65. https://doi.org/10.4103/2230-973X.133053

Gannu, R., Palem, C. R., Yamsani, V. V., Yamsani, S. K., & Yamsani, M. R. (2010). Enhanced
bioavailability of lacidipine via microemulsion based transdermal gels: formulation
optimization, ex vivo and in vivo characterization. International journal of
pharmaceutics, 388(1-2), 231-241. https://doi.org/10.1016/j.ijpharm.2009.12.050

Hu, Q., Lin, H., Wang, Y., Wang, X., Yao, J., Fu, X., & Yu, X. (2021). Design, optimization
and evaluation of a microemulsion-based hydrogel with high malleability for
enhanced transdermal delivery of levamisole. International Journal of
Pharmaceutics, 605, 120829. https://doi.org/10.1016/j.ijpharm.2021.120829

Jaiswal, M., Dudhe, R., & Sharma, P. K. (2015). Nanoemulsion: an advanced mode of drug
delivery system. 3 Biotech, 5, 123-127. https://doi.org/10.1007/s13205-014-0214-0
Jameel, B. M., Huynh, A., Chadha, A., Pandey, S., Duncan, J., Chandler, M., & Baki, G.
(2019). Computer-based formulation design and optimization using Hansen solubility
parameters to enhance the delivery of ibuprofen through the skin. International
Journal of Pharmaceutics, 569, 118549.
https://doi.org/10.1016/j.ijpharm.2019.118549

Kaur, M., Shivgotra, R., Thakur, S., Kour, R., Singh, M., Shahtaghi, N. R., ... & Jain, S. K.
(2023). A novel nanoemulgel formulation of Luliconazole with augmented antifungal
efficacy: In-vitro, in-silico, ex-vivo, and in-vivo studies. Journal of Drug Delivery
Science and Technology, 89, 105102. https://doi.org/10.1016/j.jddst.2023.105102.

Kumar, M., Bishnoi, R. S., Shukla, A. K., & Jain, C. P. (2019). Techniques for formulation of
nanoemulsion drug delivery system: a review. Preventive nutrition and food science,
24(3), 225. https://doi.org/10.3746/pnf.2019.24.3.225

Ma, Q., Zhang, J., Lu, B., Lin, H., Sarkar, R., Wu, T., & Li, X. (2021). Nanoemulgel for
improved topical delivery of desonide: Formulation design and characterization.
AAPS PharmSciTech, 22(5), 163. https://doi.org/10.1208/s12249-021-02035-5

Mittal, S., Ali, J., & Baboota, S. (2021). Enhanced anti-psoriatic activity of tacrolimus loaded
nanoemulsion gel via omega 3-Fatty acid (EPA and DHA) rich oils-fish oil and linseed
oil. Journal of Drug Delivery Science and Technology, 63, 102458.
https://doi.org/10.1016/j.jddst.2021.102458.

Niu, J., Yuan, M., Gao, P., Wang, L., Qi, Y., Chen, J., ... & Liu, X. (2023). Microemulsion-
Based Keratin–Chitosan Gel for Improvement of Skin Permeation/Retention and
Activity of Curcumin. Gels, 9(7), 587. https://doi.org/10.3390/gels9070587

Patel, H. K., Barot, B. S., Parejiya, P. B., Shelat, P. K., & Shukla, A. (2013). Topical delivery
of clobetasol propionate loaded microemulsion based gel for effective treatment of
vitiligo: ex vivo permeation and skin irritation studies. Colloids and Surfaces B:
Biointerfaces, 102, 86-94. https://doi.org/10.1016/j.colsurfb.2012.08.011

Rai, V. K., Mishra, N., Yadav, K. S., & Yadav, N. P. (2018). Nanoemulsion as pharmaceutical
carrier for dermal and transdermal drug delivery: Formulation development, stability
issues, basic considerations and applications. Journal of controlled release, 270, 203-
225. https://doi.org/10.1016/j.jconrel.2017.11.049
Rao, M., Sukre, G., Aghav, S., & Kumar, M. (2013). Optimization of metronidazole emulgel.
Journal of pharmaceutics, 2013. https://doi.org/10.1155/2013/501082

Sharma, A., Garg, T., Aman, A., Panchal, K., Sharma, R., Kumar, S., & Markandeywar, T.
(2016). Nanogel—an advanced drug delivery tool: Current and future. Artificial cells,
nanomedicine, and biotechnology, 44(1), 165-177.
https://doi.org/10.3109/21691401.2014.930745

Varma, V. N. S. K., Maheshwari, P. V., Navya, M., Reddy, S. C., Shivakumar, H. G., & Gowda,
D. V. (2014). Calcipotriol delivery into the skin as emulgel for effective permeation.
Saudi Pharmaceutical Journal, 22(6), 591-599.
https://doi.org/10.1016/j.jsps.2014.02.007

Wan, T., Xu, T., Pan, J., Qin, M., Pan, W., Zhang, G., ... & Xu, Y. (2015). Microemulsion
based gel for topical dermal delivery of pseudolaric acid B: In vitro and in vivo
evaluation. International journal of pharmaceutics, 493(1-2), 111-120.
https://doi.org/10.1016/j.ijpharm.2015.07.058
EXOSOMES

Derya Erışık, Yiğit Uyanıkgil


Chapter 2 were initially thought as "Garbage Bags"
that helped cells in eliminating unwanted

Exosomes components, but now they are understood


to be an essential part of drug delivery
DERYA ERIŞIK1, YİĞİT UYANIKGİL1,2,3
systems and intercellular communication
(Das et. al., 2019).
An Overview of Exosomes Exosomes are well-tolerated in body
Therapeutic nanocarriers have been fluids such as blood, urine, amniotic fluids,
used for drug delivery more frequently in breast milk and ascites and are less
recent years. Liposomes, polymers, immunogenic due to their natural origins.
micelles, viruses, and nanoparticles are Exosomes contain many specific proteins,
examples of advanced delivery systems lipids, mRNA and regulatory microRNAs,
that have been used to improve the and these molecules mediate intercellular
solubility, stability, efficacy, and target transmission. Exosomes have the ability to
specificity of therapeutic cargos. Despite transport their cargos across the plasma
the advancements in these systems, clinical membrane to a specific intracellular region
translation of these nanocarriers is still in its in a target-specific manner. They have
early stages because of the target cells' been widely used as vectors for the
immunogenicity, the delivery system's targeted delivery of nucleic acids and
cytotoxicity, the nanocarriers' rapid proteins since they are the natural carriers
clearance from the circulation, and their of these molecules. The exosomes' intrinsic
limited biodistribution. To eliminate the homing capability is one of the most
complications associated with synthetic significant features of the drug delivery
nanocarriers, there was a need to vehicles. Exosomes show characteristics of
investigate new endogenous delivery the cells from which they originate. The
systems. Because endogenous particles cellular source of exosomes determines
have more compatibility and the least level their target selectivity. By appropriately
of cytotoxicity, these vehicles are effective modifying the surfaces of exosomes, they
in vivo. Exosomes are microvesicles that can be engineered for targeted delivery of
have a diameter of 30 to 100 nm. They therapeutic cargos. The RVG peptide was

1 2
Department of Histology and Embryology, Cord Blood, Cell and Tissue Research and
Application Center, Ege University, 35100,
Faculty of Medicine, Ege University, Izmir
Bornova, Izmir, Türkiye.
35100, Türkiye. 3
1
Department of Stem Cell, Institute of Health
Department of Histology and Embryology, Science, Ege University, Izmir 35100, Türkiye.
Faculty of Medicine, Ege University, Izmir 35100,
Türkiye.
overexpressed on the exosomal membrane multivesicular endosomes, are produced by
in an attempt to modify the surface proteins ESEs that mature into late sorting
of exosomes. This caused the exosomes to endosomes (LSEs). MVBs are formed by the
penetrate the blood–brain barrier (BBB) endosomal limiting membrane inwardly
and become concentrated in the brain invaginating (double invagination of the
(Alvarez et. al., 2011; Das et. al., 2019). plasma membrane). MVBs with several ILVs
(future exosomes) form as a result of this
In this section, first, we have
process. MVB can be degraded by fusing
described the structure, origin, and
with lysosomes or autophagosomes, or it
biological roles of exosomes. Later on, we
can fuse with the plasma membrane and
have focused on the essential role of
release the contained ILVs as exosomes
exosomes in the context of drug delivery.
(Van Niel et. al., 2018; Kalluri and LeBleu,
Finally, we have focused on the transition
2020).
of exosome-based drug delivery to the
clinic application for cancer, and discuss the ILVs have parent cell lipids,
challenges and prospects for the future in proteins, and nucleic acids in them. Tumor
this field. susceptibility gene 101 (TSG101), ALG2-
interacting protein X (ALIX), and
The Process of Exosome Biogenesis
endosomal sorting complex required for
and Release
transport (ESCRT) are the proteins that
The generation of intracellular
promote the production of ILVs. These
multivesicular bodies (MVBs) containing
proteins combine ubiquitylated MVB
intraluminal vesicles (ILVs) and double
membrane proteins and cause the MVB
invagination of the plasma membrane are
membrane to budding inward (Van Niel et.
steps in the biogenesis process of
al., 2006).
exosomes. ILVs are secreted as exosomes,
Exosome release is a secretory
which have a diameter ranging from around
mechanism that is either constitutive in
40 to 160 nm, as a result of MVB fusion to
some cell systems or induced in cell
the plasma membrane and exocytosis. A de
activation. Exosomes are released from the
novo early-sorting endosome (ESE) is
cell after the MVB fuses with the plasma
formed as a result, and in some conditions,
membrane. The secretion pathway is
it may merge with a preexisting ESE
regulated at different stages by the
directly. In addition, the endoplasmic
monomeric G proteins Rab27a and b. The
reticulum and the trans-golgi network also
fusion between MVB membrane and the
play a role in the formation and content of
plasma membrane is one of the next steps,
the ESE. MVBs, also known as
an activity that needs fusogenic lipids, like CD63, CD81, CD82, Epithelial cell adhesion
phosphatidic acid. Thus, exosomes are molecule (EpCAM) and Ras-related protein
produced by a secretion process that (Rab5). For the purpose of separation, the
requires viable cells. While most antibodies can be immobilized in various
microvesicles are larger and may be media conditions and then mixed with
pelleted at 10-20,000g, exosomes that magnetic beads, chromatographic matrix,
have been secreted require high speed plates, and microfluidic devices (Vlassov et.
centrifugation to be recovered. Cut-off al., 2012). The method's disadvantage is
filtration devices are an additional option. that non-exosome vesicles containing the
Protein content varies depending on cell antigens also bind to the antibody. This
systems and density, but an average of 1-3 method results in exosomes with a lower
mg exosomes, or 1-3% of the proteins from purity. Exosomes can also be separated
the parent cells, can be recovered from 106 according to size differences using
cells (Record, 2014; Record et. al., 2014). ultrafiltration. In comparison to
ultracentrifugation, this process requires
Characterization and Isolation of
less time and no specialized equipment.
Exosomes
High performance liquid chromatography is
Exosomes are isolation in cell
an additional method for size-based
culture research by a variety of methods.
exosome isolation. Highly pure exosomes
The most popular method of isolation is
are produced from exosomes made using
differential centrifugation. In this method,
this method. However, this method also
centrifugal force between 200 and 100,000
requires expensive equipment (Qin and Xu,
g is used to separate large particles and cell
2014; Ha et.al., 2016).
debris from culture media, and then
The size, protein content, and lipid
exosomes are separated from the
content of exosomes can be used to classify
supernatant by sedimentation at 100,000 g.
them. Exosomes are sphere-shaped
However, this method does not provide
structures that range in size from 40 to 100
definitive results in terms of purification of
nm. They are substantially smaller than
exosomes (Denzer et. al., 2000; Ha et.al.,
other systems, like microvesicles, which
2016).
range in size from 100 to 500 nm.
Another common method for
Exosomes can be characterized using a
isolating exosomes is the monoclonal
variety of techniques, such as mass
antibody-based technique. The separation
spectrometry, flow cytometry, dynamic
process uses antibodies against exosome-
light scattering, nanoparticle tracking
associated antigens, which include CD9,
analysis, western blot, and microscopy
(Batrakova and Kim, 2015). The protein safety are still unknowns (Li et. al., 2015).
composition of exosomes can also be used In these situations, exosomes seem to be a
to identify and mark them. Tetraspanins better option than liposomal or polymeric
and integrins are the two proteins that are drug delivery systems because they have
most commonly found in exosomes. The many of the desirable features of an ideal
other protein markers include of flotillin 1, drug delivery system, including a long
ALG-2 interacting protein X (ALIX), circulating half-life, the intrinsic ability to
TSG101, and cell adhesion molecules. target tissues, biocompatibility, and
Lipids, like proteins, are major components minimal or no inherent toxicity issues
of exosomes and can be used to (Turturici et. al., 2014; Ha et.al., 2016).
characterize them (Ha et.al., 2016).
Exosomes-Based Drug Delivery:
Exosome-Based Drug Delivery Clinical Applications for Cancer

Both liposomes and polymeric Many modern cancer treatments


nanoparticles are currently the most still lack specificity for targeting cancer
popular drug delivery systems. A liposome cells, resulting in severe toxicity toward
is a phospholipid-membrane-containing normal cells, despite major breakthroughs
synthetic vesicle that can self-assemble into in anticancer drug technology. High drug
a variety of forms and sizes in an aqueous concentrations are also required since
environment (Sercombe et. al., 2015). Drug ineffective drug absorption reduces the
delivery mechanisms known as polymeric effectiveness of treatment. Exosomes have
nanoparticles help in the entrapment, a significant role in cancer from two
encapsulation, or attachment of drug perspectives: one is related to
molecules (Agrawal et. al., 2014). Different carcinogenesis, and the other is as a
kinds of drugs molecules, such as diagnostic and therapeutic delivery
analgesics, anti-fungal, and anti-cancer strategy. Exosome-based drug delivery has
drugs, have been delivered using both of become a viable approach for delivery of
these delivery systems. The long circulating drugs in cancer treatment in order to
half-life, stability, and ability of an ideal overcome these problems. Exosomes
liposome to evade the host immune system provide safe transport and prolonged
without toxicity remain unclear release of a wide range of cancer
(Raemdonck et. al., 2014; Ha et.al., 2016). therapeutic cargoes, such as proteins,
Although polymeric nanoparticles may be nucleic acids, and anti-cancer small
more stable than liposomal systems, their molecule drugs (Koh et. al., 2023).
biocompatibility and potential long-term
Exosomes are capable of carrying shown remarkable efficacy of exosomal
both large and small molecules, according doxorubicin in suppressing tumor growth
to a variety of studies (Kar et. al., 2023). and prolonging survival without side effects
Paclitaxel, Doxorubicin, and Curcumin are (Hadla et. al., 2016; Gong et. al., 2019;
examples of small molecule anti-cancer Wang et. al., 2021).
drugs. Antineoplastic agent paclitaxel is a Curcumin is a polyphenol compound
member of the taxanes drug class. that prevents the initiation and metastasis
Paclitaxel has an anti-cancer affect that is of various types of cancer. The clinical use
mediated by stabilizing microtubules and of curcumin is limited because it has poor
suppressing cell division, which deaths absorption and rapid metabolism. To
cancer cells (Barbuti and Chen, 2015). It is overcome these current limitations and
used to treat a wide range of cancers, such safely deliver curcumin to target cells, the
as pancreatic, ovarian, lung, and breast use of curcumin-loaded exosomes is
cancers, as well as glioblastoma increasing attention. Exosomes have shown
multiforme. Due to paclitaxel's limited potential in the treatment of a number of
solubility and dose-dependent toxicity, cancers, including lung, glioblastoma, and
there are clinical challenges to its breast cancer, due to their safe delivery and
application in therapy. Studies on enhanced bioavailability through this
paclitaxel's exosome-based drug delivery process (Zhang et. al., 2007; Wu et. al.,
showed anti-tumor effectson a variety of 2016; Trotta et. al., 2019; González-Sarrías
cancer types (Wang et. al., 2019; et. al., 2022).
Kandimalla et. al., 2021). Exosomes are also capable of
Leukemia, Hodgkin lymphoma, and delivering therapeutic nucleic acids.
solid tumors can all be effectively treated Delivering nucleic acids as therapeutic
with doxorubicin as an anti-cancer drug. agents through cell manipulation has made
However, side effects include reduced gene therapy a potentially effective cancer
absorption, cardiotoxicity, and bone treatment strategy. The current treatment
marrow suppression limit the long-term use aims to regulate abnormal gene
of chemotherapeutic drugs. When using expressions resulting in translation of faulty
exosomal doxorubicin, there is a partial mRNA, by genome editing or exogenous
reduction in the amount of doxorubicin gene expression. In gene therapy, viral
delivered to cardiac endothelial cells. vectors have been commonly used to
Therefore, exosomal doxorubicin has the deliver nucleic acids. Unfortunately, this
advantage of reducing cardiac toxicity therapeutic approach presents many
(Sheibani et. al., 2022). Studies have limitations, including potential toxicity and
immune responses (Naldini, 2011). effects (Tsai et. al., 2012; Lou et. al., 2015;
Alternatively, non-viral vectors can be used. Nie et. al. 2020).
Nevertheless, there are challenges to this Through RNA interference, small
strategy, including high particle sizes and interfering RNAs (siRNAs) regulate the
poor transduction efficiency, which may expression of genes. Drug resistance in a
stimulate immune reactions and faster variety of cancers may be overcome with
reticuloendothelial system clearance. For this approach to therapy. However, siRNAs
clinical application of gene transfer vectors, have some challenges due to their low
these limitations need to be overcome. stability and rapid degradation in systemic
Exosomes have emerged as a potentially circulation. The delivery of siRNAs via
useful approach by being developed as exosomes has attracted a lot of interest and
non-immunogenic, tissue-specific and non- shows great potential as approach to these
toxic vectors (Kim et. al., 2013; Koh et. al., challenges. Research on siRNA-loaded
2023). exosomes has demonstrated promising
miRNAs are a subclass of small outcomes in the treatment of cancer
noncoding RNAs that bind to the 3' (Zhang et. al., 2020; Lin et. al., 2021).
untranslated regions of target mRNAs to
Challenges with Exosome-Based
regulate gene expression. This interaction
Drug Delivery and Prospects for the
causes translational repression or mRNA
Future
degradation, which changes the expression
of genes involved in a number of cellular Exosome-based drug delivery
processes that are important for the technology with major benefits like
initiation of cancer. There is a wide variety improved circulatory bioavailability,
of miRNAs that play important roles in targeted delivery capacities, and lower
various aspects of organ physiology and immune reactions, which is a potential
pathology. miRNAs may be used as development in the pharmaceutical
therapeutics for a variety of cancer types, industry. Before entering the clinical
according to numerous research. To application, some challenges must be
determine the therapeutic potential of overcome. In order to overcome the poor
exosomes, they were loaded with various efficiency and heterogeneity of exosome
miRNAs. miRNAs delivered to target cells products, significant advancements have
via exosomes negatively regulate the been made in isolation and purification
expression of their target genes, suppress approaches. Exosomes are easier to
the proliferation and migration of cancer separate from supernatants of cell culture
cells, and contribute to antimetastatic media than they are from bodily fluids
including plasma, saliva, and urine (Lobb techniques of engineering used. As a result,
et. al., 2015). Exosomes derived from cell it is very important to have a suitable cell
supernatants are heterogeneous. They may source for exosomes to be used for clinical
also contain other EVs such as MVs, purposes. Many research use tumor cell-
apoptotic bodies, and non-vesicular derived exosomes or immortalized cell lines
contaminants. The commonly used to enhance cancer cell targeting efficiency.
ultracentrifugation method for exosome However, it is critical to note the potential
isolation is cost-effective and automated. oncogenicity associated with its use
Unfortunately, the method has (Nakamura et. al., 2017; Koh et. al., 2023).
disadvantages, including difficulty Alternatively, nucleic acid-based therapies
separating exosomes from other EVs in the have been delivered via exosomes derived
same size range and unpredictability in from red blood cells. Mature red blood cells
yield efficiency. Immunoaffinity capture can do not have a nucleus. The goal is to reduce
be used to isolate exosomes that contain the possible risks of horizontal gene
specific antigens. However, there is no transfer by using cells without nucleus as a
unique marker for exosomes, and they are source (Usman et. al., 2018). The use of
not ideal for general production in autologous exosomes should be preferred
clinical applications. There are other for therapeutic drug delivery to reduce
methods available for exosome isolation, undesirable immunological side effects.
such as density gradient centrifugation, Circulating exogenous exosomes quickly
ultrafiltration, and polymer-based disappear within 2-30 minutes and are
precipitation. However, no single method is phagocytosed by macrophageslinked with
the gold standard. Therefore, individualized the mononuclear phagocytic system.
methods need to be determined for the Therefore, there have been concerns about
isolation and purification of exosomes. the stability and endurance of exosomes in
Furthermore, there are alternative systemic circulation (Parada et. al., 2021).
approaches to improve exosome Exosomes employed for therapeutic goals
production yield, such as genetic must stay in circulation for a longer period
manipulation and various controls over cell of time in order to be clinically effective. As
culture conditions. Exosomes are not a result, exosome engineering is necessary.
empty carriers. They have a biological Exosomes, for example, have been
function as they deliver internal contents demonstrated to have a longer half-life
such as DNA, RNA and proteins. Exosome when CD47 is overexpressed on their
structure and function also vary depending surface, allowing them to escape circulating
on cell source, culture conditions, and the monocytes (Yang et. al. 2020).
Despite the limitations noted above, a targeted strategy. In the future, current
exosomes represent a potential therapeutic research will develop exosome-based drug
approach for drug delivery. Although this delivery and pave the way for personalized
technology is currently in its early stages, it medicine in the treatment of diseases that
has the potential to treat many diseases in have an important effect on human health.

Abbreviation list

BBB Blood–Brain Barrier


MVBs Multivesicular Bodies
ILVs Intraluminal Vesicles
ESE Early Sorting Endosome
LSEs Late Sorting Endosomes
TSG101 Tumor Susceptibility Gene 101
ALIX ALG2-interacting protein X
ESCRT Endosomal Sorting Complex Required for Transport
EpCAM Epithelial cell adhesion molecule
Rab5 Ras-related protein
siRNAs Small Interfering RNAs
References

Agrawal, U., Sharma, R., Gupta, M., & Vyas, S. P. (2014). Is nanotechnology a boon for oral drug
delivery?. Drug discovery today, 19(10), 1530–1546.
https://doi.org/10.1016/j.drudis.2014.04.011
Alvarez-Erviti, L., Seow, Y., Yin, H., Betts, C., Lakhal, S., & Wood, M. J. (2011). Delivery of siRNA to the
mouse brain by systemic injection of targeted exosomes. Nature biotechnology, 29(4), 341–
345. https://doi.org/10.1038/nbt.1807.
Barbuti, A. M., & Chen, Z. S. (2015). Paclitaxel Through the Ages of Anticancer Therapy: Exploring Its
Role in Chemoresistance and Radiation Therapy. Cancers, 7(4), 2360–2371.
https://doi.org/10.3390/cancers7040897
Batrakova, E. V., & Kim, M. S. (2015). Using exosomes, naturally-equipped nanocarriers, for drug
delivery. Journal of controlled release : official journal of the Controlled Release Society, 219,
396–405. https://doi.org/10.1016/j.jconrel.2015.07.030
Das, C. K., Jena, B. C., Banerjee, I., Das, S., Parekh, A., Bhutia, S. K., & Mandal, M. (2019). Exosome
as a Novel Shuttle for Delivery of Therapeutics across Biological Barriers. Molecular
pharmaceutics, 16(1), 24–40. https://doi.org/10.1021/acs.molpharmaceut.8b00901.
Denzer, K., Kleijmeer, M. J., Heijnen, H. F., Stoorvogel, W., & Geuze, H. J. (2000). Exosome: from
internal vesicle of the multivesicular body to intercellular signaling device. Journal of cell
science, 113 Pt 19, 3365–3374. https://doi.org/10.1242/jcs.113.19.3365
Gong, C., Tian, J., Wang, Z., Gao, Y., Wu, X., Ding, X., Qiang, L., Li, G., Han, Z., Yuan, Y., & Gao, S.
(2019). Functional exosome-mediated co-delivery of doxorubicin and hydrophobically modified
microRNA 159 for triple-negative breast cancer therapy. Journal of nanobiotechnology, 17(1),
93. https://doi.org/10.1186/s12951-019-0526-7

González-Sarrías, A., Iglesias-Aguirre, C. E., Cortés-Martín, A., Vallejo, F., Cattivelli, A., Del Pozo-Acebo,
L., Del Saz, A., López de Las Hazas, M. C., Dávalos, A., & Espín, J. C. (2022). Milk-Derived
Exosomes as Nanocarriers to Deliver Curcumin and Resveratrol in Breast Tissue and Enhance
Their Anticancer Activity. International journal of molecular sciences, 23(5), 2860.
https://doi.org/10.3390/ijms23052860

Ha, D., Yang, N., & Nadithe, V. (2016). Exosomes as therapeutic drug carriers and delivery vehicles
across biological membranes: current perspectives and future challenges. Acta pharmaceutica
Sinica. B, 6(4), 287–296. https://doi.org/10.1016/j.apsb.2016.02.001
Hadla, M., Palazzolo, S., Corona, G., Caligiuri, I., Canzonieri, V., Toffoli, G., & Rizzolio, F. (2016).
Exosomes increase the therapeutic index of doxorubicin in breast and ovarian cancer mouse
models. Nanomedicine (London, England), 11(18), 2431–2441. https://doi.org/10.2217/nnm-
2016-0154
Kalluri, R., & LeBleu, V. S. (2020). The biology, function, and biomedical applications of
exosomes. Science (New York, N.Y.), 367(6478), eaau6977.
https://doi.org/10.1126/science.aau6977
Kar, R., Dhar, R., Mukherjee, S., Nag, S., Gorai, S., Mukerjee, N., Mukherjee, D., Vatsa, R.,
Chandrakanth Jadhav, M., Ghosh, A., Devi, A., Krishnan, A., & Thorat, N. D. (2023). Exosome-
Based Smart Drug Delivery Tool for Cancer Theranostics. ACS biomaterials science &
engineering, 9(2), 577–594. https://doi.org/10.1021/acsbiomaterials.2c01329
Kandimalla, R., Aqil, F., Alhakeem, S. S., Jeyabalan, J., Tyagi, N., Agrawal, A., Yan, J., Spencer, W.,
Bondada, S., & Gupta, R. C. (2021). Targeted Oral Delivery of Paclitaxel Using Colostrum-
Derived Exosomes. Cancers, 13(15), 3700. https://doi.org/10.3390/cancers13153700

Kim, C. S., Duncan, B., Creran, B., & Rotello, V. M. (2013). Triggered Nanoparticles as
Therapeutics. Nano today, 8(4), 439–447. https://doi.org/10.1016/j.nantod.2013.07.004

Koh, H. B., Kim, H. J., Kang, S. W., & Yoo, T. H. (2023). Exosome-Based Drug Delivery: Translation
from Bench to Clinic. Pharmaceutics, 15(8), 2042.
https://doi.org/10.3390/pharmaceutics15082042
Li, C., Zhang, J., Zu, Y. J., Nie, S. F., Cao, J., Wang, Q., Nie, S. P., Deng, Z. Y., Xie, M. Y., & Wang, S.
(2015). Biocompatible and biodegradable nanoparticles for enhancement of anti-cancer
activities of phytochemicals. Chinese journal of natural medicines, 13(9), 641–652.
https://doi.org/10.1016/S1875-5364(15)30061-3
Lin, D., Zhang, H., Liu, R., Deng, T., Ning, T., Bai, M., Yang, Y., Zhu, K., Wang, J., Duan, J., Ge, S.,
Sun, B., Ying, G., & Ba, Y. (2021). iRGD-modified exosomes effectively deliver CPT1A siRNA to
colon cancer cells, reversing oxaliplatin resistance by regulating fatty acid oxidation. Molecular
oncology, 15(12), 3430–3446. https://doi.org/10.1002/1878-0261.13052

Lobb, R. J., Becker, M., Wen, S. W., Wong, C. S., Wiegmans, A. P., Leimgruber, A., & Möller, A. (2015).
Optimized exosome isolation protocol for cell culture supernatant and human plasma. Journal
of extracellular vesicles, 4, 27031. https://doi.org/10.3402/jev.v4.27031

Lou, G., Song, X., Yang, F., Wu, S., Wang, J., Chen, Z., & Liu, Y. (2015). Exosomes derived from miR-
122-modified adipose tissue-derived MSCs increase chemosensitivity of hepatocellular
carcinoma. Journal of hematology & oncology, 8, 122. https://doi.org/10.1186/s13045-015-
0220-7

Nakamura, K., Sawada, K., Kinose, Y., Yoshimura, A., Toda, A., Nakatsuka, E., Hashimoto, K., Mabuchi,
S., Morishige, K. I., Kurachi, H., Lengyel, E., & Kimura, T. (2017). Exosomes Promote Ovarian
Cancer Cell Invasion through Transfer of CD44 to Peritoneal Mesothelial Cells. Molecular cancer
research : MCR, 15(1), 78–92. https://doi.org/10.1158/1541-7786.MCR-16-0191

Naldini L. (2011). Ex vivo gene transfer and correction for cell-based therapies. Nature reviews.
Genetics, 12(5), 301–315. https://doi.org/10.1038/nrg2985

Nie, H., , Xie, X., , Zhang, D., , Zhou, Y., , Li, B., , Li, F., , Li, F., , Cheng, Y., , Mei, H., , Meng, H., , &
Jia, L., (2020). Use of lung-specific exosomes for miRNA-126 delivery in non-small cell lung
cancer. Nanoscale, 12(2), 877–887. https://doi.org/10.1039/c9nr09011h

Parada, N., Romero-Trujillo, A., Georges, N., & Alcayaga-Miranda, F. (2021). Camouflage strategies for
therapeutic exosomes evasion from phagocytosis. Journal of advanced research, 31, 61–74.
https://doi.org/10.1016/j.jare.2021.01.001

Qin, J., & Xu, Q. (2014). Functions and application of exosomes. Acta poloniae pharmaceutica, 71(4),
537–543.
Raemdonck, K., Braeckmans, K., Demeester, J., & De Smedt, S. C. (2014). Merging the best of both
worlds: hybrid lipid-enveloped matrix nanocomposites in drug delivery. Chemical Society
reviews, 43(1), 444–472. https://doi.org/10.1039/c3cs60299k
Record M. (2014). Intercellular communication by exosomes in placenta: a possible role in cell
fusion?. Placenta, 35(5), 297–302. https://doi.org/10.1016/j.placenta.2014.02.009
Record, M., Carayon, K., Poirot, M., & Silvente-Poirot, S. (2014). Exosomes as new vesicular lipid
transporters involved in cell-cell communication and various pathophysiologies. Biochimica et
biophysica acta, 1841(1), 108–120. https://doi.org/10.1016/j.bbalip.2013.10.004
Sercombe, L., Veerati, T., Moheimani, F., Wu, S. Y., Sood, A. K., & Hua, S. (2015). Advances and
Challenges of Liposome Assisted Drug Delivery. Frontiers in pharmacology, 6, 286.
https://doi.org/10.3389/fphar.2015.00286
Sheibani, M., Azizi, Y., Shayan, M., Nezamoleslami, S., Eslami, F., Farjoo, M. H., & Dehpour, A. R.
(2022). Doxorubicin-Induced Cardiotoxicity: An Overview on Pre-clinical Therapeutic
Approaches. Cardiovascular toxicology, 22(4), 292–310. https://doi.org/10.1007/s12012-022-
09721-1

Trotta, T., Panaro, M. A., Prifti, E., & Porro, C. (2019). Modulation of Biological Activities in Glioblastoma
Mediated by Curcumin. Nutrition and cancer, 71(8), 1241–1253.
https://doi.org/10.1080/01635581.2019.1604978

Tsai, W. C., Hsu, S. D., Hsu, C. S., Lai, T. C., Chen, S. J., Shen, R., Huang, Y., Chen, H. C., Lee, C. H.,
Tsai, T. F., Hsu, M. T., Wu, J. C., Huang, H. D., Shiao, M. S., Hsiao, M., & Tsou, A. P. (2012).
MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. The Journal
of clinical investigation, 122(8), 2884–2897. https://doi.org/10.1172/JCI63455

Turturici, G., Tinnirello, R., Sconzo, G., & Geraci, F. (2014). Extracellular membrane vesicles as a
mechanism of cell-to-cell communication: advantages and disadvantages. American journal of
physiology. Cell physiology, 306(7), C621–C633. https://doi.org/10.1152/ajpcell.00228.2013
Usman, W. M., Pham, T. C., Kwok, Y. Y., Vu, L. T., Ma, V., Peng, B., Chan, Y. S., Wei, L., Chin, S. M.,
Azad, A., He, A. B., Leung, A. Y. H., Yang, M., Shyh-Chang, N., Cho, W. C., Shi, J., & Le, M. T.
N. (2018). Efficient RNA drug delivery using red blood cell extracellular vesicles. Nature
communications, 9(1), 2359. https://doi.org/10.1038/s41467-018-04791-8

Van Niel, G., Porto-Carreiro, I., Simoes, S., & Raposo, G. (2006). Exosomes: a common pathway for a
specialized function. Journal of biochemistry, 140(1), 13–21.
https://doi.org/10.1093/jb/mvj128
Van Niel, G., D'Angelo, G., & Raposo, G. (2018). Shedding light on the cell biology of extracellular
vesicles. Nature reviews. Molecular cell biology, 19(4), 213–228.
https://doi.org/10.1038/nrm.2017.125
Vlassov, A. V., Magdaleno, S., Setterquist, R., & Conrad, R. (2012). Exosomes: current knowledge of
their composition, biological functions, and diagnostic and therapeutic potentials. Biochimica et
biophysica acta, 1820(7), 940–948. https://doi.org/10.1016/j.bbagen.2012.03.017
Wang, J., Tang, W., Yang, M., Yin, Y., Li, H., Hu, F., Tang, L., Ma, X., Zhang, Y., & Wang, Y. (2021).
Inflammatory tumor microenvironment responsive neutrophil exosomes-based drug delivery
system for targeted glioma therapy. Biomaterials, 273, 120784.
https://doi.org/10.1016/j.biomaterials.2021.120784

Wang, P., Wang, H., Huang, Q., Peng, C., Yao, L., Chen, H., Qiu, Z., Wu, Y., Wang, L., & Chen, W.
(2019). Exosomes from M1-Polarized Macrophages Enhance Paclitaxel Antitumor Activity by
Activating Macrophages-Mediated Inflammation. Theranostics, 9(6), 1714–1727.
https://doi.org/10.7150/thno.30716
Wu, H., Zhou, J., Zeng, C., Wu, D., Mu, Z., Chen, B., Xie, Y., Ye, Y., & Liu, J. (2016). Curcumin increases
exosomal TCF21 thus suppressing exosome-induced lung cancer. Oncotarget, 7(52), 87081–
87090. https://doi.org/10.18632/oncotarget.13499

Yang, Z., Shi, J., Xie, J., Wang, Y., Sun, J., Liu, T., Zhao, Y., Zhao, X., Wang, X., Ma, Y., Malkoc, V.,
Chiang, C., Deng, W., Chen, Y., Fu, Y., Kwak, K. J., Fan, Y., Kang, C., Yin, C., Rhee, J., … Lee,
L. J. (2020). Large-scale generation of functional mRNA-encapsulating exosomes via cellular
nanoporation. Nature biomedical engineering, 4(1), 69–83. https://doi.org/10.1038/s41551-
019-0485-1

Zhang, H. G., Kim, H., Liu, C., Yu, S., Wang, J., Grizzle, W. E., Kimberly, R. P., & Barnes, S. (2007).
Curcumin reverses breast tumor exosomes mediated immune suppression of NK cell tumor
cytotoxicity. Biochimica et biophysica acta, 1773(7), 1116–1123.
https://doi.org/10.1016/j.bbamcr.2007.04.015

Zhang, Q., Zhang, H., Ning, T., Liu, D., Deng, T., Liu, R., Bai, M., Zhu, K., Li, J., Fan, Q., Ying, G., & Ba,
Y. (2020). Exosome-Delivered c-Met siRNA Could Reverse Chemoresistance to Cisplatin in
Gastric Cancer. International journal of nanomedicine, 15, 2323–2335.
https://doi.org/10.2147/IJN.S231214
NIOSOMES

Emine Esin Çalışkan, Emel Öykü Çetin Uyanıkgil


characterization and can be modified by
Chapter 3
incorporating various additives to improved

Niosomes stability, enhanced bioavailability and


control drug release (Ge et al., 2019).
EMINE ESIN ÇALIŞKAN1, EMEL ÖYKÜ Additionally, niosomes have been

ÇETIN UYANIKGİL2 extensively studied for their potential in


controlled release and targeted delivery of
drugs for the treatment of various diseases,
Introduction including cancer, viral infections, microbial
diseases, psoriasis, leishmaniasis, and
In the field of pharmaceuticals, the
migraines (Yasamineh et al., 2022).
development of effective drug delivery
Moreover, niosomes have shown promise
systems is crucial for ensuring the
in prolonging the circulation of drugs in the
localization and targeted administration of
body, potentially reducing toxicity and
pharmaceutical compounds within the body
improving drug penetration into target
(Mahale et al., 2012). One promising drug
tissues.In addition to conventional routes
delivery system is the use of niosomes,
of administration (such as oral and
which are microscopic non-ionic surfactant
parenteral), niosomes can be delivered
bilayer vesicles (Witika et al., 2022). These
through ocular, transdermal, vaginal, and
niosomes are formed by hydrating
inhalation routes (Abdelkader et al., 2014).
synthetic nonionic surfactants, often with
the inclusion of cholesterol or lipids. The Niosomes have been studied for their
unique amphiphilic nature of niosomes potential in delivering various types of
allows them to efficiently encapsulate both drugs, including anti-cancer agents, anti-
lipophilic and hydrophilic drugs, making infective drugs, and vaccines. Their
them versatile vehicles for drug delivery capability to enhance the therapeutic
(Azeem et al., 2009). Niosomes offer efficacy of drugs while minimizing their
several advantages over other drug potential side effects makes them a
delivery systems, such as being non-ionic, valuable tool in modern medicine
stable, biodegradable, biocompatible and (Abdelkader et al., 2014).
non-immunogenic. Furthermore, niosomes
exhibit flexibility in their structural

1
Department of Biopharmaceutics and
Pharmacokinetics, Ege University, Faculty of
2
Department of Biopharmaceutics and
Pharmacy, Izmir 35100, Türkiye. Pharmacokinetics, Ege University, Faculty of
Pharmacy, Izmir 35100, Türkiye.
Furthermore, the cost-effectiveness and will play an increasingly pivotal role in
ease of large-scale production of niosomes shaping the future of drug delivery
have also contributed to their growing systems.
popularity in the pharmaceutical industry.
Components of Niosomes
As research in niosome technology
Niosomes are composed of synthetic
continues to advance, the potential for
nonionic surfactants and other components
innovative drug delivery solutions and
such as cholesterol, which form the bilayer
improved patient outcomes becomes
vesicles. The nonionic surfactants play a
increasingly promising (Abdelkader et al.,
crucial role in the formation of niosomes by
2014). Recent research in niosome
providing stability to the vesicles. The
technology has also focused on improving
addition of cholesterol helps in improving
the targeting of specific areas within the
the rigidity and flexibility of the niosomal
body. By modifying the surface properties
membrane, thereby enhancing drug
of niosomes, scientists have been able to
encapsulation and delivery efficiency
design formulations that can selectively
(Kumar & Rajeshwarrao, 2011).
deliver drugs to particular organs or
tissues. This targeted delivery approach The composition of niosomes allows for the
not only increases the effectiveness of the encapsulation of both hydrophilic and
drug but also reduces the risk of side hydrophobic drugs, making them versatile
effects on non-targeted tissues (Yasamineh for a wide range of pharmaceutical
et al., 2022). applications. Additionally, the incorporation
of other components such as targeting
Moreover, the potential applications of
ligands, polymers, or other functional
niosomes extend beyond traditional drug
molecules can further enhance the
delivery. They have shown promise in gene
specificity and efficacy of niosomal drug
delivery, where the encapsulation of
genetic material within niosomes offers delivery systems (Abdelkader et al., 2014).

protection and efficient transport to target Cholesterol


cells (Barani et al., 2019).
Cholesterol is an essential component of
In conclusion, the versatility, cost- niosomes, playing a crucial role in
effectiveness, and potential for targeted improving the rigidity and flexibility of the
delivery make niosomes an exciting area of niosomal membrane. This enhancement
pharmaceutical research. As the contributes to the efficient encapsulation
understanding of niosomes deepens and and delivery of drugs. Additionally, the
technology advances, it is likely that they composition of niosomes allows for the
encapsulation of both hydrophilic and effectively. It influences the interaction
hydrophobic drugs, making them versatile between the surfactant molecules and the
for a wide range of pharmaceutical drug to be encapsulated, thereby impacting
applications (Nasseri, 2005). the overall performance of the niosomal
drug delivery system (Kumar &
In addition to cholesterol, the incorporation
Rajeshwarrao, 2011; Sharma et al., 2015;
of other components such as targeting
Wu et al., 2021; Yaghoobian et al., 2020).
ligands, polymers, or other functional
molecules can further enhance the Understanding the different types of non-
specificity and efficacy of niosomal drug ionic surfactants and their HLB values is
delivery systems. This flexibility in essential for selecting the most suitable
composition is one of the key advantages components to achieve the desired drug
of niosomes in drug delivery (Aparajay & encapsulation and delivery characteristics
Dev, 2022). in niosomes. This knowledge not only
contributes to the optimization of niosomal
Nonionic Surfactants
formulations but also plays a significant
Non-ionic surfactants are a key component role in the design of efficient drug delivery
in the formation of niosomes. These systems for pharmaceutical applications
surfactants are a diverse group that can be (Sharma et al., 2015). Studies have shown
classified based on their hydrophilic- that an increase in HLB (Hydrophilic-
lipophilic balance value, which determines Lipophilic Balance) leads to an increase in
their solubility in water and oil. Some the length of the alkyl chain and vesicle
common types of non-ionic surfactants size. HLB values in the range of 14-17 are
include alkyl polyglucosides, sorbitan esters reported to be unsuitable, while an HLB of
(e.g., Span series), and polysorbates (e.g., 8 results in the highest encapsulation
Tween series). Alkyl polyglucosides are efficiency (EE) (Yaghoobian et al., 2020).
non-ionic surfactants with a high HLB For instance, the EE of a lipophilic drug can
value, making them suitable for forming be enhanced by using a surfactant with a
niosomes with hydrophilic drugs. On the low HLB (Wu et al., 2021). Another factor
other hand, sorbitan esters such as Span influencing EE is the phase transition
60 and polysorbates such as Tween 80 temperature. A biodegradable surfactant
have lower HLB values, which makes them like Span 60, which has a high transition
suitable for forming niosomes with temperature, offers high EE (Kumar &
lipophilic drugs. The HLB of a surfactant is Rajeshwarrao, 2011).
crucial in determining its ability to form
stable niosomes and encapsulate drugs
Charge Induced Types of Niosomes

In addition to the non-ionic surfactants and Niosomes, as drug delivery systems, can be
cholesterol, the introduction of charged classified into different types based on their
components has been found to have size and structure. The most common types
significant effects on the properties of include Small Unilamellar Vesicles, Large
niosomes. Incorporating charged Unilamellar Vesicles, and Multilamellar
molecules into the niosomal membrane can Vesicles (Johnson & Buttress, 1973; Mui et
induce specific interactions with drug al., 1993).
compounds, leading to improved
encapsulation and release behaviors.
Small Unilamellar Vesicles
When charged molecules are included in
SUVs, also known as small unilamellar
the formation of niosomes, the resulting
vesicles, are characterized by their small
vesicles can exhibit enhanced stability and
size and consist of a single phospholipid
controlled release properties. In some
bilayer. They are particularly advantageous
cases, the presence of charged
for targeted drug delivery due to their
components can facilitate the targeted
ability to pass through small capillaries and
delivery of drugs by promoting interactions
enter cells more efficiently. SUVs have a
with specific cell types or tissues within the
diameter typically ranging from 25 to 100
body.
nanometers, making them suitable for
Electrostatic stabilization achieved through
delivering both hydrophilic and
the addition of charge-inducing agents
hydrophobic drugs, which in turn makes
enhances the stability of niosomes by
them versatile in pharmaceutical
preventing aggregation. Inert bulky surface
applications such as cancer treatment or
groups, such as polyelectrolytes and
gene therapy. However, it is important to
polysaccharides, or charged groups,
note that SUVs are thermodynamically
provide steric repulsion between particles,
unstable and tend to aggregate; they also
leading to more stable niosomal
offer low entrapment efficiency for
dispersions. Dicetyl phosphate, stearyl
hydrophilic agents. SUVs have a diameter
amine, and diacylglycerol are the most
typically ranging from 25 to 100
commonly used charge-inducing agents in
nanometers, making them suitable for
niosomal systems (Aditya et al., 2019;
delivering both hydrophilic and
Johnson & Buttress, 1973; Mui et al.,
hydrophobic drugs, which in turn makes
1993).
them versatile in pharmaceutical
applications such as cancer treatment or Effect of Cholesterol and Surfactant
gene therapy (Banke & Thakre, 2020; Concentration
Ganesh Sai Myneni*, 2021; Khoee, 2018).
The incorporation of cholesterol into
niosomal formulations plays a crucial role in
modulating membrane rigidity and fluidity.
Large Unilamellar Vesicles
By adjusting the cholesterol concentration,
LUVs, also known as large unilamellar it is possible to influence the permeability
vesicles, are larger in size and have a and stability of niosomes, thus impacting
diameter ranging from 100 to 1000 nm. their drug delivery properties. Higher levels
They consist of a single phospholipid of cholesterol can lead to a reduction in
bilayer and their larger size provides a membrane permeability and increased
greater capacity for drug encapsulation, stability, while lower levels may result in
making them suitable for the delivery of a higher permeability and decreased
higher drug payload. LUVs are often stability. Therefore, the careful selection of
utilized for the delivery of larger drug cholesterol concentration is essential for
molecules and biomacromolecules such as achieving the desired characteristics in
proteins and nucleic acids due to their size niosomes (Bartelds et al., 2018).
compatibility (Ganesh Sai Myneni*, 2021;
Similarly, the concentration of non-ionic
Moghassemi et al., 2022; Ray et al., 2018).
surfactants in niosome preparation
Multilamellar Vesicles significantly affects the vesicle size,
MLVs, with a typical size range of 100-1000 morphology, and drug entrapment
nm, consist of multiple lipid bilayers and efficiency. The optimization of surfactant
are larger in size compared to SUVs and concentration is essential for achieving the
LUVs. Their multilayered structure allows desired niosome characteristics, such as
for sustained and prolonged release of size distribution, encapsulation efficiency,
drugs, making them ideal for controlled and stability. (Liu et al., 2000; Nasseri,
drug delivery. MLVs are advantageous for 2005; Salih et al., 2013; Uchegbu &
delivering both hydrophilic and Florence, 1995).
hydrophobic drugs, offering versatility in
Furthermore, the surfactant/lipid ratio
pharmaceutical applications (Ganesh Sai
should be considered in niosome
Myneni*, 2021; Khoee, 2018)
formulation, as it can affect the
Factors Influencing Niosome Formation encapsulation efficiency. An increase in the
surfactant/lipid ratio results in higher
encapsulation efficiency and consequently
increases the system's viscosity. The total maintained above the glass transition
range of the surfactant/lipid ratio should be temperature (Tg) of the niosome system
maintained between 10–30 mM (1–2.5%, (Nasseri, 2005; Uchegbu & Vyas, 1998).
w/w). Additionally, incorporating
Effect of pH on Niosome Formation in
cholesterol at a 1:1 M ratio with the
Hydration Environment
surfactant is a common practice in
In addition to the factors previously
formulations (Aditya et al., 2019; Masjedi &
discussed, the pH of the hydration medium
Montahaei, 2021).
also significantly impacts niosome
Effect of Hydration Medium
formation. The pH level can influence the
Temperature
stability and morphology of the niosomes,
Apart from the composition of niosomes ultimately affecting their performance as
and the impact of cholesterol and drug delivery vehicles.
surfactant concentration, the temperature
Studies have demonstrated that varying
of the hydration medium during niosome
the pH of the hydration environment can
formation also plays a significant role in
lead to changes in the surface charge of the
determining the characteristics of the
niosomes, affecting their interaction with
resulting vesicles. The temperature of the
drugs and biological membranes. These
hydration medium can influence the
pH-induced alterations in surface charge
kinetics of niosome formation, as well as
can impact the colloidal stability and
the size, distribution, and stability of the
dispersion of the niosomes, with potential
formed vesicles.
implications for their shelf-life and in vivo
Studies have shown that the hydration behavior.
medium temperature affects the hydration
Moreover, the pH of the hydration medium
and swelling of the lipid film, thereby
can influence the protonation and
impacting the formation of niosomes.
deprotonation of the components involved
Higher temperatures may lead to increased
in niosome formation, such as the non-ionic
fluidity of the lipid bilayers, potentially
surfactants and other lipid constituents.
affecting the size and encapsulation
This, in turn, can affect the vesicle size,
efficiency of the niosomes. Conversely,
membrane fluidity, and drug encapsulation
lower temperatures might result in slower
efficiency. The pH level of the medium
membrane hydration and vesicle
plays a crucial role both in the synthesis
formation, influencing the vesicle size and
process and in drug encapsulation. The pH
homogeneity. It should be noted that the
of the hydration medium is determined by
hydration medium temperature must be
the solubility of the encapsulated drug. It components. Careful adjustment of the
has been found that a pH of 7.4, when injection rate and temperature of the
using a phosphate buffer, results in stable aqueous phase affects the size and stability
vesicles with small particle sizes. of the resulting niosomes (Ge et al., 2019;
(Akbarzadeh et al., 2021; Mokhtar et al., Moghassemi & Hadjizadeh, 2014).
2008).
Reverse Phase Evaporation Method
Niosome Preparation Methods
In this method, a water-in-oil emulsion is
Thin-Film Hydration Method formed by dispersing an aqueous phase
containing the drug within an organic
The thin-film hydration method is a widely
phase consisting of the lipids, cholesterol,
used technique for niosome preparation. In
and non-ionic surfactants. Subsequent
this method, a thin lipid film is formed by
removal of the organic solvent by
evaporating a chloroform solution of the
evaporation results in the formation of
lipid and cholesterol, if applicable, in a
niosomes encapsulating the aqueous drug
round-bottom flask using a rotary
solution. This method allows for the
evaporator. The film is then hydrated with
entrapment of both hydrophilic and
an aqueous solution containing drug under
hydrophobic drugs within the niosomal
constant stirring, resulting in the formation
bilayers, offering versatility in drug delivery
of niosomes. The properties of the resulting
(Amoabediny et al., 2018).
niosomes can be modulated by adjusting
the hydration temperature, pH, and Bubble Method
surfactant concentration (Amoabediny et
Another niosome preparation method is the
al., 2018; Uchegbu & Vyas, 1998).
bubble method, which utilizes the
Ether Injection Method formation of bubbles to create niosomes.
In this method, a lipid and cholesterol
Another method for niosome preparation is
mixture, if applicable, is dispersed in an
the ether injection method. This technique
aqueous phase containing the non-ionic
involves dissolving the lipids and
surfactant. The dispersion is then subjected
cholesterol, if used, in diethyl ether,
to ultrasonication or mechanical agitation,
followed by injection of the lipid solution
leading to the formation of bubbles within
into an aqueous phase containing the non-
the dispersion. These bubbles serve as
ionic surfactant. The rapid injection of the
templates for the formation of niosomes,
lipid solution leads to the formation of
allowing for the spontaneous self-assembly
niosomes due to the spontaneous self-
of lipid and surfactant components around
assembly of the lipid and surfactant
the gas phase. The bubble method offers Particle Size and Polydispersity Index
advantages in terms of simplicity and (PDI):
scalability, making it suitable for large-scale
The size of niosomes is a significant
niosome production. Moreover, the control
parameter influencing their biodistribution,
of the ultrasonication or mechanical
cellular uptake, and release of
agitation parameters such as intensity and
encapsulated drugs. These two parameters
duration can influence the size and
also play an important role in stability and
distribution of the resulting niosomes
provide information about the size
(Bagheri et al., 2014; Masjedi & Montahaei,
distribution of particles in a given
2021).
formulation. Techniques such as light
Characterization of Niosomes microscopy, electron microscopy analysis,
Scanning Electron Microscopy (SEM),
Morphology
Transmission Electron Microscopy (TEM),
Morphology is a crucial parameter that freeze-fracture replication, Dynamic Light
provides information about the shape, size, Scattering (DLS), and zeta-sizer have been
and structure. Various instruments such as used to characterize the size and
Atomic Force Microscopy (AFM), Field morphology of niosomal formulations. PDI
Emission Scanning Electron Microscopy is defined as a measure of the distribution
(FESEM), and Transmission Electron of niosome sizes. Niosomal formulations
Microscopy (TEM) assist in visualizing with PDI values below 0.5 indicate a
complex structures with precise shape and monodisperse sample (Gupta & Trivedi,
scale. Confocal Laser Scanning Microscopy 2018; Jain & Thareja, 2019; Moghassemi et
(CLSM) helps in determining the al., 2017).
hydrophobic and hydrophilic layers in
Zeta Potential
niosomes using suitable dyes and also
assists in identifying the encapsulated Zeta potential governs the charge on the
therapeutic component. In contrast, Leica surface of niosomal particles, providing
microscopy can be used to analyze the ionic repulsion and ensuring the proper
basic design of niosomes, such as the dispersion of particles in the medium,
formation of a ring-like system (Tangri & thereby stabilizing the preparation.
Khurana, 2011). Niosomes with zeta potentials over +30 mV
or lower than − 30 mV have adequate
strength (Barani et al., 2018). Inadequate
zeta potential can lead to unwanted side
effects such as toxicity, lack of specificity in
drug targeting, and aggregation of vesicles. the concentration of the retained drug. The
The charges in niosomes can be detected encapsulation efficiency of drug-loaded
based on zeta potential, which can be niosomes is calculated as the difference
measured using a zeta potential analyzer, between the total amount incorporated and
nano zeta-sizer, microelectrophoresis, pH- the dialyzed amount (Firthouse et al.,
sensitive fluorophores, high-performance 2011).
capillary electrophoresis, and Dynamic
Encapsulation Efficiency
Light Scattering (DLS) instruments Total amount of drug − Amount of drug unentrapped
=
(Moammeri et al., 2023). Total amount of drug
× 100
Encapsulation Efficiency
In-vitro Release Study
This parameter is typically used to
determine the percentage of the drug In-vitro drug release study is a quantitative
incorporated into the formulation. analysis that helps determine the amount
Encapsulation efficiency plays an important of drug released from the formulation at
role in drug delivery and can be further different time intervals. This step is crucial
used to determine the dosage frequency of in understanding the drug release kinetics
the formulation. Increasing the drug load in and the potential for sustained or
the formulation can effectively reduce the controlled release behavior. This study aids
dosage frequency of the therapeutic in understanding the Cmax, Tmax, and
component. Various methods, including AUC values of the drug in systemic
ultracentrifugation, dialysis, and separation circulation. These variables also help in
techniques, are used to determine determining the toxicity profile of the
encapsulation efficiency. The dialysis bag therapeutic component. Among the various
method is most frequently used. In this reported methods, the dialysis bag method
method, the prepared formulation is placed is most commonly used. In this method,
in a dialysis tube, sealed from both sides, the drug-loaded formulation is usually
and then placed in a thermostatic medium immersed in a physiological buffer
at 35±0.5 °C for 24 hours. The (phosphate buffer) and held in a dialysis
unentrapped drug diffuses out through the tube, properly agitated. At specific time
dialysis membrane, and the concentration intervals, samples are taken to determine
is then measured using UV-Visible the amount of drug released, usually
spectroscopy or High-Performance Liquid analyzed using UV-Visible spectroscopy or
Chromatography (HPLC) at a HPLC at a predetermined range (Escudero
predetermined wavelength to determine et al., 2014; Hao et al., 2002).
Stability of application areas within the
pharmaceutical industry. One notable
Stability data help ensure the efficacy of
application is in the delivery of anti-cancer
the developed formulation over a specified
drugs. Studies have demonstrated that
period. Major disadvantages of these
niosomes can enhance the therapeutic
vesicles include storage-related issues such
efficacy of anti-cancer drugs while reducing
as aggregation, fusion, photodegradation,
their toxic effects on healthy tissues.
and leakage of the drug from these
Additionally, niosomes have been explored
vesicles. The stability of a formulation is
for the delivery of antibiotics, anti-
also indicated by the absence of significant
inflammatory agents, and other therapeutic
changes in various parameters such as
compounds. Their ability to encapsulate
shape, charge, size, etc. (Ammar et al.,
and deliver these substances to specific
2011).
target sites in the body makes them a
Lamellarity Determination valuable tool in pharmaceutical research
Vesicular formulations are characterized by and development. Furthermore, their
the number of lamellar, being either stability and ability to protect encapsulated
unilamellar or multilamellar. Lamellarity drugs from degradation make them a
can be determined using Atomic Force promising avenue for the development of
Microscopy (AFM), Nuclear Magnetic advanced drug delivery systems.
Resonance (NMR), Small Angle X-ray Niosomes have also shown potential in the
Scattering (SAXS), SEM, and TEM delivery of vaccines. Research in this area
(Marianecci et al., 2014). The thickness of has indicated that niosomes can improve
bilayer vesicles can be characterized using the stability and immunogenicity of the
in situ energy dispersive X-ray diffraction vaccines, leading to enhanced immune
and electron microscopy with SAXS (Agrati responses. This makes niosomes a
et al., 2011; Di Marzio et al., 2011; promising candidate for the delivery of
Domenici et al., 2009). various types of vaccines, including those
for influenza, hepatitis, and other infectious
diseases.
Application Areas
Moreover, the application of niosomes
Niosomes are novel vesicular systems in
extends to the field of cosmetic and
the field of pharmaceuticals that have
skincare products. Their ability to
gained attention for their potential as drug
encapsulate and deliver active ingredients
delivery systems (Mahale et al., 2012).
to the skin has garnered interest in the
Niosomes have shown promise in a variety
development of niosome-based In conclusion, the diverse applications of
formulations for targeted delivery and niosomes in the pharmaceutical industry,
improved efficacy of skincare products. ranging from targeted drug delivery to
gene therapy, ocular drug delivery, and
In addition to these areas, ongoing
nutritional supplement delivery, underscore
research is exploring the utilization of
their potential as a versatile and valuable
niosomes for gene delivery, ocular drug
drug delivery system. Ongoing research
delivery, and delivery of nutritional
and development in these areas continue
supplements. Their ability to protect and
to expand the horizons of niosome
effectively deliver genetic material to target
applications, offering promising avenues
cells has made them a subject of interest in
for the advancement of pharmaceutical
gene therapy research. The development
science and healthcare. The potential of
of niosome-based gene delivery systems
niosomes as a versatile drug delivery
holds promise for the treatment of genetic
system has led to continued exploration
disorders and other therapeutic
and innovation in the pharmaceutical
applications related to gene modulation.
industry. One area of interest is the
Furthermore, in the area of ocular drug development of niosome-based treatments
delivery, niosomes have been investigated for neurological disorders. Research has
for their ability to enhance the indicated that niosomes can effectively
bioavailability and sustained release of deliver neuroprotective agents and drugs
ophthalmic drugs. The unique lipid across the blood-brain barrier, opening up
composition of niosomes allows for new possibilities for the treatment of
improved penetration of drugs into ocular conditions such as Alzheimer's disease,
tissues, making them a valuable tool in the Parkinson's disease, and brain tumors.
development of effective treatments for
Furthermore, the application of niosomes in
ocular diseases.
the field of personalized medicine is an
Moreover, the delivery of nutritional exciting frontier. The ability of niosomes to
supplements using niosomes is an encapsulate and deliver specific drug
emerging area of interest. Encapsulating combinations tailored to individual patients'
essential nutrients such as vitamins and needs holds great promise for the
minerals within niosomes can enhance advancement of personalized treatment
their stability and bioavailability, offering regimens.
potential benefits for nutritional
Additionally, the use of niosomes in the
supplement formulations.
delivery of hormones and growth factors
for therapeutic purposes is being actively
explored. Their capability to protect and
deliver these sensitive biological molecules
to target tissues presents opportunities for
the development of innovative hormone
replacement therapies and regenerative
medicine modalities.

In the realm of transdermal drug delivery,


niosomes are being investigated for their
potential to enhance the permeation of
drugs across the skin barrier. This research
holds implications for the development of
improved transdermal patches and
localized drug delivery systems.
References
Abdelkader, H., Alani, A. W. G., & Alany, R. G. (2014). Recent advances in non-ionic surfactant
vesicles (niosomes): Self-assembly, fabrication, characterization, drug delivery applications and
limitations. Drug Delivery, 21(2), 87–100. https://doi.org/10.3109/10717544.2013.838077

Aditya, S., Lakhvinder, K., Prevesh, K., Neelkant, P., & Vaibhav, R. (2019). Niosomes: A
Promising Approach in Drug Delivery Systems. Journal of Drug Delivery & Therapeutics, 4(9), 635–642.

Agrati, C., Marianecci, C., Sennato, S., Carafa, M., Bordoni, V., Cimini, E., Tempestilli, M.,
Pucillo, L. P., Turchi, F., Martini, F., Borioni, G., & Bordi, F. (2011). Multicompartment vectors as novel
drug delivery systems: Selective activation of Tγδ lymphocytes after zoledronic acid delivery.
Nanomedicine: Nanotechnology, Biology and Medicine, 7(2), 153–161.
https://doi.org/10.1016/j.nano.2010.10.003

Akbarzadeh, I., Shayan, M., Bourbour, M., Moghtaderi, M., Noorbazargan, H., Eshrati Yeganeh,
F., Saffar, S., & Tahriri, M. (2021). Preparation, Optimization and In-Vitro Evaluation of Curcumin-
Loaded Niosome@calcium Alginate Nanocarrier as a New Approach for Breast Cancer Treatment.
Biology, 10(3), 173. https://doi.org/10.3390/biology10030173
Ammar, H. O., Ghorab, M., El-Nahhas, S. A., & Higazy, I. M. (2011). Proniosomes as a carrier
system for transdermal delivery of tenoxicam. International Journal of Pharmaceutics, 405(1–2), 142–
152. https://doi.org/10.1016/j.ijpharm.2010.11.003

Amoabediny, G., Haghiralsadat, F., Naderinezhad, S., Helder, M. N., Akhoundi Kharanaghi, E.,
Mohammadnejad Arough, J., & Zandieh-Doulabi, B. (2018). Overview of preparation methods of
polymeric and lipid-based (niosome, solid lipid, liposome) nanoparticles: A comprehensive review.
International Journal of Polymeric Materials and Polymeric Biomaterials, 67(6), 383–400.
https://doi.org/10.1080/00914037.2017.1332623

Aparajay, P., & Dev, A. (2022). Functionalized niosomes as a smart delivery device in cancer
and fungal infection. European Journal of Pharmaceutical Sciences, 168, 106052.
https://doi.org/10.1016/j.ejps.2021.106052

Azeem, A., Anwer, Md. K., & Talegaonkar, S. (2009). Niosomes in sustained and targeted drug
delivery: Some recent advances. Journal of Drug Targeting, 17(9), 671–689.
https://doi.org/10.3109/10611860903079454

Bagheri, A., Chu, B.-S., & Yaakob, H. (2014). Niosomal Drug Delivery Systems: Formulation,
Preparation and Applications. 8(32), 1671–1685. https://doi.org/10.5829/idosi.wasj.2014.32.08.848
Banke, B., & Thakre, N. (2020). Design of Gatifloxacin Niosomes by film hydration method.
International Journal of Social Impact, 5(2), 23–30. https://doi.org/10.25215/2455/0502002
Barani, M., Mirzaei, M., Torkzadeh-Mahani, M., & Nematollahi, M. H. (2018). Lawsone-loaded
Niosome and its antitumor activity in MCF-7 breast Cancer cell line: A Nano-herbal treatment for Cancer.
DARU Journal of Pharmaceutical Sciences, 26(1), 11–17. https://doi.org/10.1007/s40199-018-0207-3
Barani, M., Nematollahi, M. H., Zaboli, M., Mirzaei, M., Torkzadeh-Mahani, M., Pardakhty, A., &
Karam, G. A. (2019). In silico and in vitro study of magnetic niosomes for gene delivery: The effect of
ergosterol and cholesterol. Materials Science and Engineering: C, 94, 234–246.
https://doi.org/10.1016/j.msec.2018.09.026

Bartelds, R., Nematollahi, M. H., Pols, T., Stuart, M. C. A., Pardakhty, A., Asadikaram, G., &
Poolman, B. (2018). Niosomes, an alternative for liposomal delivery. PLOS ONE, 13(4), e0194179.
https://doi.org/10.1371/journal.pone.0194179
Di Marzio, L., Marianecci, C., Petrone, M., Rinaldi, F., & Carafa, M. (2011). Novel pH-sensitive
non-ionic surfactant vesicles: Comparison between Tween 21 and Tween 20. Colloids and Surfaces B:
Biointerfaces, 82(1), 18–24. https://doi.org/10.1016/j.colsurfb.2010.08.004
Domenici, F., Panichelli, D., & Castellano, A. C. (2009). Alamethicin–lipid interaction studied by
energy dispersive X-ray diffraction. Colloids and Surfaces B: Biointerfaces, 69(2), 216–220.
https://doi.org/10.1016/j.colsurfb.2008.11.029

Escudero, I., Geanta, R. M., Ruiz, M. O., & Benito, J. M. (2014). Formulation and
characterization of Tween 80/cholestherol niosomes modified with tri-n-octylmethylammonium chloride
(TOMAC) for carboxylic acids entrapment. Colloids and Surfaces A: Physicochemical and Engineering
Aspects, 461, 167–177. https://doi.org/10.1016/j.colsurfa.2014.07.042
Firthouse, P. U. M., Halith, S. M., Wahab, S. U., Sirajudeen, M., & Mohideen, S. K. (2011).
Formulation and Evaluation of Miconazole Niosomes. International Journal of PharmTech Research,
3(2), 1019–1022.
Ganesh Sai Myneni*, G. V. R. (2021). NOVEL VESICULAR DRUG DELIVERY SYSTEMS: A
REVIEW. https://doi.org/10.5281/ZENODO.4772544
Ge, X., Wei, M., He, S., & Yuan, W.-E. (2019). Advances of Non-Ionic Surfactant Vesicles
(Niosomes) and Their Application in Drug Delivery. Pharmaceutics, 11(2), 55.
https://doi.org/10.3390/pharmaceutics11020055

Gupta, V., & Trivedi, P. (2018). In vitro and in vivo characterization of pharmaceutical topical
nanocarriers containing anticancer drugs for skin cancer treatment. In Lipid Nanocarriers for Drug
Targeting (pp. 563–627). Elsevier. https://doi.org/10.1016/B978-0-12-813687-4.00015-3
Hao, Y., Zhao, F., Li, N., Yang, Y., & Li, K. (2002). Studies on a high encapsulation of colchicine
by a niosome system. International Journal of Pharmaceutics, 244(1–2), 73–80.
https://doi.org/10.1016/S0378-5173(02)00301-0

Jain, A. K., & Thareja, S. (2019). In vitro and in vivo characterization of pharmaceutical
nanocarriers used for drug delivery. Artificial Cells, Nanomedicine, and Biotechnology, 47(1), 524–539.
https://doi.org/10.1080/21691401.2018.1561457

Johnson, S. M., & Buttress, N. (1973). The osmotic insensitivity of sonicated liposomes and the
density of phospholipid-cholesterol mixtures. Biochimica et Biophysica Acta (BBA) - Biomembranes,
307(1), 20–26. https://doi.org/10.1016/0005-2736(73)90021-7
Khoee, S. (2018). Chapter 6—Niosomes: A novel approach in modern drug delivery systems.
In Nanostructures for Drug Delivery (pp. 208–231).

Kumar, G. P., & Rajeshwarrao, P. (2011). Nonionic surfactant vesicular systems for effective
drug delivery—An overview. Acta Pharmaceutica Sinica B, 1(4), 208–219.
https://doi.org/10.1016/j.apsb.2011.09.002

Kumara, P., Sarathchandra Prakash2, N. K., P3, L., & Manral4, K. (2015). Viscoelastic Properties
and Rheological Characterization of Carbomers. International Journal of Latest Research in Engineering
and Technology (IJLRET), 1(6), 2454–5031.
Liu, D.-Z., Chen, W.-Y., Tasi, L.-M., & Yang, S.-P. (2000). Microcalorimetric and shear studies
on the effects of cholesterol on the physical stability of lipid vesicles. Colloids and Surfaces A:
Physicochemical and Engineering Aspects, 172(1–3), 57–67. https://doi.org/10.1016/S0927-
7757(00)00560-4
Mahale, N. B., Thakkar, P. D., Mali, R. G., Walunj, D. R., & Chaudhari, S. R. (2012). Niosomes:
Novel sustained release nonionic stable vesicular systems — An overview. Advances in Colloid and
Interface Science, 183–184, 46–54. https://doi.org/10.1016/j.cis.2012.08.002
Marianecci, C., Di Marzio, L., Rinaldi, F., Celia, C., Paolino, D., Alhaique, F., Esposito, S., &
Carafa, M. (2014). Niosomes from 80s to present: The state of the art. Advances in Colloid and Interface
Science, 205, 187–206. https://doi.org/10.1016/j.cis.2013.11.018
Masjedi, M., & Montahaei, T. (2021). An illustrated review on nonionic surfactant vesicles
(niosomes) as an approach in modern drug delivery: Fabrication, characterization, pharmaceutical, and
cosmetic applications. Journal of Drug Delivery Science and Technology, 61, 102234.
https://doi.org/10.1016/j.jddst.2020.102234

Moammeri, A., Chegeni, M. M., Sahrayi, H., Ghafelehbashi, R., Memarzadeh, F., Mansouri, A.,
Akbarzadeh, I., Abtahi, M. S., Hejabi, F., & Ren, Q. (2023). Current advances in niosomes applications
for drug delivery and cancer treatment. Materials Today Bio, 23, 100837.
https://doi.org/10.1016/j.mtbio.2023.100837

Moghassemi, S., Dadashzadeh, A., Azevedo, R. B., & Amorim, C. A. (2022). Nanoemulsion
applications in photodynamic therapy. Journal of Controlled Release, 351, 164–173.
https://doi.org/10.1016/j.jconrel.2022.09.035

Moghassemi, S., & Hadjizadeh, A. (2014). Nano-niosomes as nanoscale drug delivery systems:
An illustrated review. Journal of Controlled Release, 185, 22–36.
https://doi.org/10.1016/j.jconrel.2014.04.015

Moghassemi, S., Hadjizadeh, A., & Omidfar, K. (2017). Formulation and Characterization of
Bovine Serum Albumin-Loaded Niosome. AAPS PharmSciTech, 18(1), 27–33.
https://doi.org/10.1208/s12249-016-0487-1

Mokhtar, M., Sammour, O. A., Hammad, M. A., & Megrab, N. A. (2008). Effect of some
formulation parameters on flurbiprofen encapsulation and release rates of niosomes prepared from
proniosomes. International Journal of Pharmaceutics, 361(1–2), 104–111.
https://doi.org/10.1016/j.ijpharm.2008.05.031

Mui, B. L., Cullis, P. R., Evans, E. A., & Madden, T. D. (1993). Osmotic properties of large
unilamellar vesicles prepared by extrusion. Biophysical Journal, 64(2), 443–453.
https://doi.org/10.1016/S0006-3495(93)81385-7

Nasseri, B. (2005). Effect of cholesterol and temperature on the elastic properties of niosomal
membranes. International Journal of Pharmaceutics, 300(1–2), 95–101.
https://doi.org/10.1016/j.ijpharm.2005.05.009

Podual, K., Doyle, F. J., & Peppas, N. A. (2000). Dynamic behavior of glucose oxidase-
containing microparticles of poly(ethylene glycol)-grafted cationic hydrogels in an environment of
changing pH. Biomaterials, 21(14), 1439–1450. https://doi.org/10.1016/S0142-9612(00)00020-X

Ray, S. K., Bano, N., Shukla, T., Upmanyu, N., Pandey, S. P., & Parkhe, G. (2018). Noisomes:
As novel vesicular drug delivery system. Journal of Drug Delivery and Therapeutics, 8(6), 335–341.
https://doi.org/10.22270/jddt.v8i6.2029

Salih, O. S., Samein, L. H., & Ali, W. K. (2013). FORMULATION AND IN VITRO EVALUATION
OF ROSUVASTATIN CALCIUM NIOSOMES. International Journal of Pharmacy and Pharmaceutical
Sciences, 5(4), 525–535.
Sharma, V., Anandhakumar, S., & Sasidharan, M. (2015). Self-degrading niosomes for
encapsulation of hydrophilic and hydrophobic drugs: An efficient carrier for cancer multi-drug delivery.
Materials Science and Engineering: C, 56, 393–400. https://doi.org/10.1016/j.msec.2015.06.049
Tangri, P., & Khurana, S. (2011). NIOSOMES: FORMULATION AND EVALUATION. International
Journal of Biopharmaceutics, 1(2), 47–53.
Uchegbu, I. F., & Florence, A. T. (1995). Non-ionic surfactant vesicles (niosomes): Physical and
pharmaceutical chemistry. Advances in Colloid and Interface Science, 58(1), 1–55.
https://doi.org/10.1016/0001-8686(95)00242-I

Uchegbu, I. F., & Vyas, S. P. (1998). Non-ionic surfactant based vesicles (niosomes) in drug
delivery. International Journal of Pharmaceutics, 172(1–2), 33–70. https://doi.org/10.1016/S0378-
5173(98)00169-0

Witika, B. A., Bassey, K. E., Demana, P. H., Siwe-Noundou, X., & Poka, M. S. (2022). Current
Advances in Specialised Niosomal Drug Delivery: Manufacture, Characterization and Drug Delivery
Applications. International Journal of Molecular Sciences, 23(17), 9668.
https://doi.org/10.3390/ijms23179668

Wu, X., Yang, X., Liang, Q., Xue, X., Huang, J., Wang, J., Xu, Y., Tong, R., Liu, M., Zhou, Q.,
& Shi, J. (2021). Drugs for the treatment of glaucoma: Targets, structure-activity relationships and
clinical research. European Journal of Medicinal Chemistry, 226, 113842.
https://doi.org/10.1016/j.ejmech.2021.113842

Yaghoobian, M., Haeri, A., Bolourchian, N., Shahhosseni, S., & Dadashzadeh, S. (2020). The
Impact of Surfactant Composition and Surface Charge of Niosomes on the Oral Absorption of
Repaglinide as a BCS II Model Drug. International Journal of Nanomedicine, Volume 15, 8767–8781.
https://doi.org/10.2147/IJN.S261932

Yasamineh, S., Yasamineh, P., Ghafouri Kalajahi, H., Gholizadeh, O., Yekanipour, Z., Afkhami,
H., Eslami, M., Hossein Kheirkhah, A., Taghizadeh, M., Yazdani, Y., & Dadashpour, M. (2022). A state-
of-the-art review on the recent advances of niosomes as a targeted drug delivery system. International
Journal of Pharmaceutics, 624, 121878. https://doi.org/10.1016/j.ijpharm.2022.121878
SELF-EMULSIFIYING DRUG DELIVERY
SYSTEMS(SEDDS)

Yalçın Çelik Aydın, Emel Öykü Çetin Uyanıkgil


form stable emulsions. The choice of
Chapter 4
excipients and their ratios play a crucial
role in determining the self-emulsifying
Self-Emulsifying Drug
properties and overall performance of

Delivery Systems the system (Rahman et al., 2012).


Additionally, SEDDS offer flexibility in
(SEDDS) terms of formulation, allowing for the
incorporation of various drug
YALÇIN ÇELIK AYDIN1, EMEL ÖYKÜ
compounds with diverse
ÇETIN UYANIKGİL2
physicochemical properties.
Introduction
The versatile nature of SEDDS opens up
Self-emulsifying drug delivery systems new possibilities for the development of
ave gained significant attention in the novel drug delivery systems, offering
pharmaceutical industry due to their potential solutions for improving the
ability to improve the solubility and delivery of poorly water-soluble drugs
bioavailability of poorly water-soluble and ultimately enhancing patient
drugs (Buya et al., 2020). The self- outcomes (18th World Drug Delivery
emulsifying nature of these systems Summit 2024, 2023). In this chapter,
allows for the formation of fine oil-in- we will delve deeper into the
water emulsions when in contact with formulation aspects, advantages,
the aqueous environment of the challenges, and future prospects of
gastrointestinal tract. This in turn SEDDS in pharmaceutical research and
facilitates the transport of the drug development.
across the gastrointestinal epithelium,
1. Important Aspects for Pre-
leading to enhanced absorption and
Formulation
therapeutic efficacy (Pandey & Kohli,
One important aspect in the
2018).
formulation of self-emulsifying drug
SEDDS are typically composed of an oil
delivery systems is the choice and ratio
phase, a surfactant, and a co-
of excipients used (Shrestha et al.,
surfactant, which work together to
2014).

1
Department of Biopharmaceutics and
Pharmacokinetics, Ege University, Faculty of
2
Department of Biopharmaceutics and
Pharmacy, Izmir 35100, Türkiye. Pharmacokinetics, Ege University, Faculty of
Pharmacy, Izmir 35100, Türkiye.
The selection of excipients is crucial as The versatility of SEDDS in
it directly impacts the stability, accommodating various drugs with
emulsification efficiency, and drug different physicochemical properties
release profile and droplet size of the makes them a promising platform for
SEDDS (Bashir et al., 2023). Various enhancing drug solubility and
oils such as medium-chain triglycerides, bioavailability. By overcoming the
long-chain triglycerides, and limitations of poorly water-soluble
polyethylene glycols can be used to drugs, SEDDS have the potential to
optimize the solubility of the drug and revolutionize drug delivery and improve
promote self-emulsification. the therapeutic outcomes for patients.
Surfactants like Tween, Span, and
2. SEDDS Components
Labrasol are commonly employed to
Optimization
reduce the interfacial tension between
When it comes to optimizing self-
the oil and water phases, facilitating
emulsifying drug delivery systems,
emulsification.
careful consideration of the
Furthermore, the choice of a co-
components is crucial. The selection
surfactant such as PEG, Transcutol, or
and ratio of excipients directly impact
Cremophor can influence the droplet
the stability, emulsification efficiency,
size and overall stability of the emulsion
and drug release profile of the SEDDS.
(Petkova et al., 2012). The ratio of
2.1. Excipients and Their
surfactant to co-surfactant is critical in
Influence
achieving the desired emulsification
2.1.1. Oils
efficiency and preventing phase
separation. In self-emulsifying drug delivery
systems, oils are a key component as
In addition to the composition of
they serve as the solvent for the
excipients, the processing methods also
lipophilic drug. The choice of oil can
contribute to the performance of
affect the solubility of the drug, the
SEDDS. Techniques such as phase
extent of emulsification, and the
titration, homogenization, and spray
absorption and bioavailability of the
drying are employed to ensure uniform
drug. Examples of oils that can be used
distribution of components and the
in SEDDS formulations include evening
formation of stable emulsions (Buya et
primrose oil, fish oil, Jojoba oil, Olive
al., 2020) (Yang et al., 2017).
oil, Soybean oil, Squalene and
Sunflower oil.
These oils provide a lipophilic process and in stabilizing the resultant
environment that can enhance the emulsion. A surfactant, also known as
solubility of drugs that have poor a surface-active agent, reduces the
aqueous solubility. The percentage of interfacial tension between two phases,
oil in a SEDDS formulation can vary which in the case of SEDDS, are the oil
based on the desired drug loading, the and water phases. This allows the oil
solubility of the drug in the oil, and the droplets containing the drug to disperse
required dose. more readily throughout the aqueous
environment, such as the
The effects of oil in SEDDS are
gastrointestinal tract, once
multifaceted:
administered (Atef & Belmonte, 2008)
 They can influence the self- (Jain et al., 2013) (Malkawi et al.,
emulsification efficiency and the 2020).
size of the resulting emulsion
Examples of surfactants commonly
droplets.
used in SEDDS formulations include:
 The composition of the oil can also
 Polysorbates (e.g., Tween series
impact the stability of the emulsion
like Tween 20, Tween 80)
and its interaction with biological
membranes, potentially enhancing  Cremophor (e.g., Cremophor EL,
the absorption of the drug. Cremophor RH40)

 Additionally, the oil component may  Sorbitan esters (e.g., Span series
affect the taste and overall patient like Span 20, Span 80)
acceptability of the formulation.
 Polyethylene glycols (e.g., PEG
By carefully selecting the oil and 400)
adjusting its concentration in the
 Polyoxyl hydrogenated castor oils
SEDDS formula, formulators can
(e.g., Kolliphor RH40)
optimize drug loading, emulsification
The percentage of surfactant in a
properties, and bioavailability (Singh et
SEDDS formulation is usually high
al., 2014) (Gürsoy & Benita, 2004)
because a sufficient amount is required
(Charman et al., 1992).
to maintain the stability of the
2.1.2. Surfactants
emulsion. Typically, surfactants may
Surfactants in SEDDS formulations play constitute about 30-60% of the total
a vital role in the self-emulsification
formulation by weight (Buya et al., value indicates better water
2020) (McClements & Jafari, 2018). solubility, which is advantageous
for emulsifying and stabilizing the
The effects of surfactants in SEDDS
SEDDS in the aqueous phase of the
include:
digestive tract (Gürsoy & Benita,
1. Emulsification Efficiency: A 2004) (Atef & Belmonte, 2008)
surfactant helps to form fine oil-in- (Buya et al., 2020) (Pandey & Kohli,
water emulsions with small droplet 2018).
sizes, leading to an increased
2.1.3. Co-surfactants
surface area for drug absorption.
Co-surfactants are often included in
2. Drug Solubilization: Surfactants can
SEDDS formulations to further reduce
enhance the solubility of poorly
interfacial tension and to improve the
water-soluble drugs within the
emulsification process. They work
emulsion, potentially increasing
alongside the primary surfactant to
bioavailability.
stabilize the emulsion droplets and
3. Stability: The right surfactant can enhance drug solubilization and
improve the physical stability of the absorption. Co-surfactants can also
drug in the formulation, preventing help in adjusting the fluidity of the
drug precipitation. interfacial film, making it more elastic
4. Absorption: Surfactants can and favoring the formation of smaller
interact with biological membranes, droplets during emulsification
altering permeability and (Emulsion, 2022).
potentially improving the drug’s The most common co-surfactants used
intestinal absorption. While in SEDDS formulations include:
surfactants are crucial for the
 Medium-chain alcohols such as
effective design of SEDDS, their
isopropyl alcohol and ethanol.
selection must be carefully
considered, as some surfactants  Propylene glycol.
can cause gastrointestinal irritation
 Polyethylene glycols of various
or other side effects, especially at
molecular weights.
high concentrations. Additionally,
the surfactant's hydrophilic-  Glycerol.

lipophilic balance value is  Sorbitol.


important; typically, a higher HLB
Co-surfactants are typically used at which is particularly important for
lower concentrations than the primary lipophilic drugs.
surfactant, often ranging from 10% to
3. Preparation Methods for SEDDS
30% of the SEDDS formulation by
Preparation methods are important in
weight (Buya et al., 2020). The exact
the development of self-emulsifying
percentage used can vary depending
drug delivery systems. The techniques
on the properties desired in the final
employed during the formulation
emulsion, such as droplet size,
process significantly influence the
viscosity, and drug release profile.
stability, emulsification efficiency, and
The functions of co-surfactants in
overall performance of SEDDS. Several
SEDDS formulations include:
key processing methods are utilized to
1. Facilitating Self-Emulsification: Co- ensure the uniform distribution of
surfactants aid in the emulsification components and the formation of
process by further reducing the stable emulsions.
surface and interfacial tension,
3.1. Simple Mixing:
making the system more dynamic
and capable of self-emulsification. o The oil, surfactant, and co-
surfactant are measured out
2. Droplet Size Reduction: They
in specified ratios based on
contribute to forming small droplet
preliminary studies such as
sizes, which is important for a large
solubility and phase
interfacial area and thus can
diagrams.
enhance the rate and extent of drug
o The active pharmaceutical
absorption.
ingredient is dissolved in the
3. Stability Improvement: Co-
oil phase with gentle
surfactants help stabilize
heating if necessary to
emulsions, preventing the assist in solubilization.
coalescence of droplets, which
o The oil-solubilized drug is
could lead to phase separation.
then blended with the
4. Solubilizing Agent: Some co- surfactant and co-surfactant
surfactants can also act as mixture until a clear and
solubilizers, increasing the solubility homogenous isotropic
of the drug within the oil phase, mixture is obtained.
o Additional excipients, such when drugs are difficult to
as antioxidants or solubilize.
preservatives, can be added
o After dissolving the API in
and mixed if required (Buya
the oil phase, the mixture is
et al., 2020) (Application Of
subjected to high shear
Surfactants In Pharmacy,
mixing using equipment
2021).
such as a high-speed
3.2. Heating and Cooling Method: homogenizer.

o This method may be used o To obtain a uniform SEDDS,


when one or more the high shear mixing
components of the process is followed by high-
formulation are solids at pressure homogenization, if
room temperature or have necessary (Constantinides,
high melting points. 2000).

o The formulation 3.4. Ultrasonication:


components are gently
o Sometimes, to reduce the
heated above their melting
size of the emulsion
points to form a clear liquid.
droplets further,
o The active drug is dissolved ultrasonication is applied
in this melt. after initial mixing.

o Upon cooling, stirring is o The SEDDS mixture is


maintained to ensure exposed to ultrasonic waves
uniformity and prevent the which apply mechanical
separation of components shear, breaking down the oil
as the mixture returns to droplets into much finer
room temperature (Jatto & emulsions (Witika et al.,
Okhamafe, 2002). 2022) (Quanlu et al., 2013).

3.3. High-Shear Mixing and 3.5. Microfluidization:


Homogenization:
o This is a high-pressure
o This method is employed to process that forces the
achieve smaller droplet SEDDS mixture through
sizes and ensure uniformity microchannels resulting in
intense shear forces and systemic exposure, leading to improved
impact, leading to very therapeutic outcomes (Constantinides,
small and uniform droplet 2000) (Buya et al., 2020) (Singh et al.,
sizes. 2014).

o This method is particularly 4.2. Topical and Transdermal


Delivery
useful for achieving
nanoemulsions (Ge et al., SEDDS have also shown promise in

2019) (Yang et al., 2012). topical and transdermal drug delivery


applications. The nanoemulsion
4. SEDDS Application Areas
formation upon contact with skin or
Self-emulsifying drug delivery systems mucosal surfaces allows for improved
have found diverse application areas drug permeation and localized delivery
due to their potential to enhance drug of therapeutics. Additionally, the lipid-
solubility, permeation, and overall based nature of SEDDS facilitates the
therapeutic efficacy. The unique incorporation of drugs with different
properties and versatile nature of lipophilic properties, enabling their
SEDDS enable their utilization in utilization in topical formulations for
various pharmaceutical and therapeutic enhanced skin penetration and
settings. localized drug action (Singh et al.,
2014) (Pandey & Kohli, 2018) (Reza,
4.1. Oral Drug Delivery
2013).
One of the primary application areas of
SEDDS is oral drug delivery. The ability 4.3. Ocular Delivery
of SEDDS to form fine nanoemulsions Ocular SEDDS are specialized
upon exposure to the gastrointestinal formulations designed to enhance the
environment allows for improved drug delivery of drugs to the eyes. They are
solubilization and enhanced absorption an emerging field that adapts the
across the gastrointestinal mucosa. principles of SEDDS for oral
This makes SEDDS particularly administration to applications involving
advantageous for drugs with poor the eye. These systems are aimed to
aqueous solubility, as well as those overcome the challenges associated
exhibiting low oral bioavailability. By with conventional eye drops, such as
leveraging the self-emulsifying poor bioavailability, limited corneal
properties of SEDDS, pharmaceutical penetration, and rapid pre-corneal loss
compounds can achieve better due to tear production and drainage
(Pandey & Kohli, 2018) (Singh et al., It should be noted that the use of
2014) (Karwal et al., 2016). SEDDS for routes other than oral can
be challenging due to the physiological
4.4. Other Routes of Delivery
and pharmacokinetic differences
SEDDS, although mainly formulated for
associated with these routes (Akhtar et
oral administration, have been explored
al., 2020) (Singh et al., 2014).
for other routes of administration
Furthermore, for invasive routes like
beyond oral, ocular, and topical.
parenteral, the formulation must be
4.4.1. Rectal: SEDDS can be used for sterile and free of irritants.
rectal administration in the form of Development of SEDDS for non-oral
suppositories or enemas. This route routes also requires careful
can be particularly useful for patients consideration of the anatomy and
who are unable or unwilling to take physiology of the administration site,
medications orally, and also to avoid including issues of drug absorption,
the first-pass metabolism by the liver local tissue reactions, and possible
(Atef & Belmonte, 2008). irritation due to the components of the

4.4.2. Parenteral: Although less SEDDS.

common due to the requirement for 5. Difficulties Encountered in the


SEDDS to be non-irritating and suitable Formulation and Use of SEDDS
for injection, parenteral routes such as
The formulation and use of self-
intravenous, subcutaneous, or
emulsifying drug delivery systems can
intramuscular injections involve use of
present various challenges, including
very fine emulsions to ensure safety
issues related to stability, drug
and efficacy (Buya et al., 2020).
compatibility, and manufacturing
4.4.3. Intranasal: Nasal delivery of processes. These difficulties can impact
SEDDS can provide a quick absorption the successful development and
through the mucosal lining of the nasal application of SEDDS in pharmaceutical
passages, potentially offering a direct and therapeutic settings. However,
route to the systemic circulation there are effective solutions and
(Pandey & Kohli, 2018) (Buya et al., strategies available to address these
2020). challenges and optimize the

4.4.4. Vaginal: SEDDS may also be performance of SEDDS.

applied vaginally for localized or 5.1. Solubility and Compatibility


systemic delivery of drugs. Challenges
One of the primary difficulties scalability, reproducibility, and
encountered in the formulation of equipment compatibility. Achieving
SEDDS is the solubility and consistent and reproducible
compatibility of active pharmaceutical emulsification and particle size
ingredients with the lipid-based distribution during large-scale
components. Certain drugs may exhibit production is essential for ensuring
limited solubility in the chosen lipid uniformity and efficacy of the final
excipients, leading to challenges in SEDDS formulation. Additionally, the
achieving a homogenous and stable selection of appropriate equipment for
formulation. Additionally, compatibility all processes is crucial for maintaining
issues between the drug and other the integrity and quality of the SEDDS.
excipients can affect the physical and
Employing advanced manufacturing
chemical stability of the SEDDS,
techniques, such as high-pressure
impacting its efficacy and shelf-life.
homogenization and microfluidization,
Conducting comprehensive solubility can improve the scalability and
studies and compatibility assessments reproducibility of SEDDS production.
during the pre-formulation stage is These techniques enable precise
important identifying suitable lipid control over emulsification and particle
excipients and surfactants that can size reduction, ensuring consistent
effectively solubilize the API. Utilizing quality across batch sizes (Buya et al.,
co-solvents or co-surfactants, such as 2020) (Pandey & Kohli, 2018).
polyethylene glycol, can enhance the
5.3. Gastrointestinal Variability
solubility of poorly soluble drugs in the
The variability in gastrointestinal
lipid based SEDDS. Furthermore,
conditions, including pH levels, bile salt
employing lipid-based prodrugs or
concentrations, and motility, can
nanostructured lipid carriers can
impact the performance and stability of
address compatibility issues and
SEDDS following oral administration.
improve the stability of the formulation
Ensuring robustness and reliability of
(Shrestha et al., 2014) (Severino et al.,
the self-emulsifying system in diverse
2012).
gastrointestinal environments is
5.2. Manufacturing Challenges essential for consistent drug
The manufacturing of SEDDS, solubilization and absorption.
particularly at large scales, can present
Formulating SEDDS with
challenges related to process
gastroprotective coatings or pH-
responsive polymers can enhance their Furthermore, the formulation of SEDDS
stability and performance in varying with gastroprotective coatings or pH-
gastrointestinal conditions. These responsive polymers can mitigate the
protective coatings can shield the impact of gastrointestinal variability,
SEDDS from harsh acidic environments ensuring consistent drug solubilization
and facilitate targeted drug release in and absorption across diverse
specific regions of the gastrointestinal gastrointestinal conditions (Gürsoy &
tract, optimizing drug absorption and Benita, 2004) (Karwal et al., 2016)
bioavailability. (Constantinides, 2000) (Fasinu et al.,
2011).
By addressing these difficulties through
comprehensive pre-formulation 6. Types of SEDDS
studies, advanced manufacturing
SEDDS can be categorized into two
techniques, and specialized formulation
main types according to their droplet
strategies, the formulation and use of
size: Self-Microemulsifying Drug
SEDDS can be optimized to maximize
Delivery Systems and Self-
their therapeutic potential across a
Nanoemulsifying Drug Delivery
wide range of pharmaceutical
Systems.
applications.
6.1. SMEDDS
In conclusion, the formulation and use
SMEDDS are characterized by the
of self-emulsifying drug delivery
formation of microemulsions upon
systems offer immense potential for
dispersion in the gastrointestinal fluid.
improving the solubility, stability, and
These systems typically consist of an oil
bioavailability of poorly soluble drugs.
phase, a surfactant, and a co-
However, several challenges need to be
surfactant. The ability to form fine oil-
addressed to optimize their
in-water microemulsions provides
performance. By focusing on advanced
enhanced solubilization of poorly
manufacturing techniques, such as
water-soluble drugs, leading to
high-pressure homogenization and
improved drug absorption and
microfluidization, and leveraging
bioavailability.
specialized processing capabilities, the
scalability and reproducibility of SEDDS When exposed to gastrointestinal

production can be significantly fluids, SMEDDS form microemulsions

enhanced. with droplet sizes typically ranging from


100 nm to a few microns. Their larger
droplet size compared to SNEDDS can due to the lower interfacial tension and
sometimes result in less rapid drug strong surfactant films that resist
release and absorption. Microemulsions droplet coalescence.
formed by SMEDDS are often said to be
Understanding the differences between
only kinetically stable. This means that
SMEDDS and SNEDDS is crucial for
the droplets tend to aggregate or
optimizing the formulation of self-
coalesce over time, although this
emulsifying drug delivery systems
process is generally slow. The kinetic
based on the specific characteristics of
stability results from a balance of
the drug and desired therapeutic
factors including droplet Brownian
outcomes (Buya et al., 2020) (Krstić et
motion, attractive forces between
al., 2018) (Akhtar et al., 2020).
droplets, and the interfacial film
7. Formulation Type and
strength (Ivanov & Kralchevsky, 1997)
Application
(Capek, 2004).
In addition to categorization based on
6.2. SNEDDS
composition, SEDDS can also be
SNEDDS, on the other hand, are
classified according to their formulation
designed to spontaneously form
types, such as liquid SEDDS, solid
nanoemulsions with droplet sizes
SEDDS, and hybrid SEDDS. Each
typically under 100 nm when in contact
formulation type has unique
with the gastrointestinal environment.
advantages and considerations for drug
Nanoemulsions offer advantages such
solubilization, stability, and dosage
as increased surface area for drug
form design. For instance, liquid SEDDS
absorption and potential for enhanced
offer ease of administration and rapid
drug permeation across biological
drug release, while solid SEDDS provide
barriers. The formulation of SNEDDS
improved stability and portability.
involves the use of excipients that
promote the formation of stable nano- The application of SEDDS extends to
sized emulsions. various therapeutic areas, including but
not limited to oncology, cardiovascular
SNEDDS typically form
diseases, infectious diseases, and
thermodynamically stable
central nervous system disorders. The
nanoemulsions, which means that the
versatility of SEDDS in delivering a wide
small droplets do not coalesce over
range of drugs underscores their
time and the system remains stable
potential to address unmet clinical
indefinitely. Thermodynamic stability is
needs and enhance patient outcomes SEDDS can effectively enhance drug
across diverse therapeutic categories. solubility, absorption, and
bioavailability.
The selection of formulation type and
application should be carefully 8.1. Enhanced Drug Solubilization
and Bioavailability
evaluated based on the
The careful selection of oils,
physicochemical properties of the drug,
surfactants, and co-surfactants in
desired pharmacokinetic profile, and
SEDDS formulation leads to the
target patient population. Tailoring
creation of stable emulsions that
SEDDS formulation and application to
significantly enhance the solubilization
specific drug candidates and
of poorly water-soluble drugs. This
therapeutic indications can maximize
improved solubilization, especially in
their therapeutic benefits and clinical
the case of SMEDDS forming
impact.
microemulsions or SNEDDS forming
By understanding and effectively
nanoemulsions, ultimately translates to
categorizing SEDDS based on
enhanced drug absorption and
composition, formulation type, and
increased bioavailability.
application, pharmaceutical scientists
8.2. Flexibility in Drug
and formulators can optimize the
Formulation
design and implementation of SEDDS
SEDDS offer flexibility in formulating
to meet the unique requirements of
various drug compounds with diverse
diverse drug compounds and patient
physicochemical properties. This
populations(Krstić et al., 2018) (Pandey
adaptability allows for the effective
& Kohli, 2018) (Jain et al., 2013).
incorporation of a wide range of drug
8. Advantages of Self-Emulsifying molecules, further expanding the
Drug Delivery Systems potential applications of SEDDS in
pharmaceutical formulations.
Self-emulsifying drug delivery systems
offer numerous advantages in
pharmaceutical research and 8.3. Tailored Drug Delivery
Systems
development. The precise selection and
formulation of excipients play a pivotal The distinct preparation methods of

role in determining the performance SMEDDS and SNEDDS allow for the

and efficacy of SEDDS. By optimizing customization of self-emulsifying drug

the choice and ratio of excipients, delivery systems based on the specific
characteristics of the drug and desired
therapeutic outcomes. By carefully The ease of administration and
selecting excipients and controlling enhanced bioavailability offered by self-
their ratios, SEDDS can be tailored to emulsifying drug delivery systems can
optimize drug delivery and achieve the contribute to improved patient
desired therapeutic effects (Singh et compliance. Patients may experience
al., 2014). the benefits of reduced dosing
frequency, lower pill burden, and
8.4. Enhanced Stability and Shelf
Life potentially improved treatment
One of the key advantages of self- outcomes, leading to better adherence
emulsifying drug delivery systems is to prescribed regimens (Singh, 2021).
their enhanced stability, which
8.7. Potential for Pediatric and
contributes to prolonged shelf life and Geriatric Applications
improved storage conditions. The The adaptability of SEDDS in
formation of stable emulsions or formulating diverse drug compounds
nanoemulsions ensures that the drug and the potential for customized
product remains homogenized and dosage forms make them promising for
maintains its efficacy over an extended pediatric and geriatric applications. By
period, minimizing the risk of addressing challenges related to drug
degradation and ensuring consistent palatability, dosing flexibility, and ease
performance. of administration, SEDDS can help
meet the specific needs of these patient
8.5. Reduced Variability in Drug
Absorption populations, ultimately improving
SEDDS can help mitigate variability in medication adherence and therapeutic
drug absorption by promoting outcomes.
consistent and enhanced solubilization
In conclusion, the advantages of self-
of the drug, regardless of individual
emulsifying drug delivery systems
patient factors or gastrointestinal
encompass enhanced solubilization and
conditions. This reduction in variability
bioavailability, flexibility in drug
can lead to more predictable and
formulation, tailored drug delivery
reliable pharmacokinetic profiles,
systems, stability and shelf life,
enhancing the overall therapeutic
reduced variability in absorption,
outcomes for patients.
improved patient compliance, and
8.6. Improved Patient Compliance potential applications in pediatric and
geriatric populations. These attributes
position SEDDS as a valuable platform additional excipients. Understanding
for addressing the evolving needs of and predicting these interactions to
pharmaceutical research and achieve optimal formulation and
development, clinical practice, and stability require in-depth knowledge
patient care (Cherniakov et al., 2015) and expertise in pharmaceutical
(Constantinides, 2000). sciences, which can pose challenges
during development and scale-up.
9. Disadvantages of Self-
Emulsifying Drug Delivery 9.3 Manufacturing Challenges
Systems SEDDS manufacturing may pose
challenges related to scalability,
While self-emulsifying drug delivery
process optimization, and
systems offer numerous advantages,
reproducibility. Achieving consistent
there are also several disadvantages
product quality, especially for solid
and challenges that need to be
SEDDS, can be demanding and may
considered in their development and
require specialized equipment and
commercialization.
control measures to ensure uniformity
9.1. Variable Performance and and performance.
Stability
One of the primary disadvantages of 9.4. Patient Acceptance and
Compliance
SEDDS is the potential for variable
Patient acceptance and compliance
performance and stability, especially
with SEDDS formulations can also
when exposed to different
present challenges, particularly if the
environmental conditions such as
dosage form or administration method
changes in pH, temperature, and
differs significantly from conventional
agitation. This variability can affect the
formulations. Educating healthcare
consistency and reliability of drug
providers and patients about the
release and absorption, which are
benefits and usage of SEDDS may be
critical factors for ensuring therapeutic
necessary to ensure acceptance and
efficacy (Maderuelo et al., 2011).
adherence.
9.2. Formulation Complexity and
Interactions Addressing these disadvantages and
The formulation of SEDDS can be challenges in the development,
complex and may involve intricate formulation, and commercialization of
interactions between the oils, SEDDS is crucial for realizing their
surfactants, co-surfactants, and potential benefits and expanding their
application across various therapeutic for variations in patient responses,
areas. It requires a comprehensive optimizing treatment outcomes and
understanding of the limitations and improving patient adherence.
proactive mitigation strategies to
SEDDS also offer opportunities for
optimize the performance and
combining multiple therapeutic agents
acceptance of self-emulsifying drug
within a single formulation, enabling
delivery systems (Cherniakov et al.,
synergistic effects and enhanced
2015) (Singh et al., 2014) (Singh,
treatment regimens. This integrated
2021).
approach has the potential to
10. Future Prospects and revolutionize combination therapy and
Conclusion address complex medical conditions
more effectively.
The future of SEDDS holds promising
prospects for the targeted delivery of In conclusion, self-emulsifying drug
drugs to specific sites within the body. delivery systems have notable
With further research and disadvantages and challenges, but
development, SEDDS can be tailored to proactive strategies can address these
release drugs at precise locations, limitations, paving the way for their
enhancing their efficacy while widespread application in
minimizing systemic side effects. pharmaceutical research and clinical
practice. With ongoing advancements
As the field of personalized medicine
and collaborative efforts, SEDDS hold
continues to advance, SEDDS can play
significant promise for enhancing drug
a vital role in tailoring drug
delivery and improving patient
formulations to individual patient
outcomes.
needs. Customized SEDDS can account
References

18th World Drug Delivery Summit 2024. (2023, February 20).


https://drugdelivery.pharmaceuticalconferences.com/events-list/advanced-drug-delivery-systems

Akhtar, N., Mohammed, S A A., Khan, R., Yusuf, M., Singh, V., Mohammed, H A., Al-Omar,
M S., Abdellatif, A A., Naz, M., & Khadri, H. (2020, August 1). Self-Generating nano-emulsification
techniques for alternatively-routed, bioavailability enhanced delivery, especially for anti-cancers, anti-
diabetics, and miscellaneous drugs of natural, and synthetic origins.
https://doi.org/10.1016/j.jddst.2020.101808

Application Of Surfactants In Pharmacy. (2021, February 18).


https://www.njchm.com/solution/application-of-surfactants-in-pharmacy.html

Atef, E., & Belmonte, A A. (2008, November 1). Formulation and in vitro and in vivo
characterization of a phenytoin self-emulsifying drug delivery system (SEDDS).
https://doi.org/10.1016/j.ejps.2008.07.004

Bashir, M. A., Khan, A., Shah, S. I., Ullah, M., Khuda, F., Abbas, M., ... & Ming, L. C. (2023).
Development and Evaluation of Self-Emulsifying Drug-Delivery System–Based Tablets for
Simvastatin, a BCS Class II Drug. Drug design, development and therapy, 261-272.

Buya, A B., Beloqui, A., Memvanga, P B., & Préat, V. (2020, December 9). Self-Nano-
Emulsifying Drug-Delivery Systems: From the Development to the Current Applications and
Challenges in Oral Drug Delivery. https://doi.org/10.3390/pharmaceutics12121194

Capek, I. (2004, March 1). Degradation of kinetically-stable o/w emulsions.


https://doi.org/10.1016/s0001-8686(03)00115-5

Charman, S. A., Charman, W. N., Rogge, M. C., Wilson, T. D., Dutko, F. J., & Pouton, C. W.
(1992). Self-emulsifying drug delivery systems: formulation and biopharmaceutic evaluation of an
investigational lipophilic compound. Pharmaceutical research, 9, 87-93.

Cherniakov, I., Domb, A J., & Hoffman, A. (2015, January 5). Self-nano-emulsifying drug
delivery systems: an update of the biopharmaceutical aspects.
https://doi.org/10.1517/17425247.2015.999038

Constantinides, P. (2000, May 15). Self-Emulsifying Drug Delivery Formulations in the 21st
Century: Challenges and Opportunities. https://pubs.acs.org/doi/abs/10.1021/bk-2000-0752.ch028

Emulsion. (2022, February 4). https://www.newworldencyclopedia.org/entry/Emulsion

Fasinu, P S., Pillay, V., Ndesendo, V M K., Toit, L C D., & Choonara, Y E. (2011, April 7).
Diverse approaches for the enhancement of oral drug bioavailability.
https://doi.org/10.1002/bdd.750

Ge, X., Wei, M., He, S., & Yuan, W. (2019, January 29). Advances of Non-Ionic Surfactant
Vesicles (Niosomes) and Their Application in Drug Delivery.
https://doi.org/10.3390/pharmaceutics11020055

Gürsoy, R N., & Benita, S. (2004, April 1). Self-emulsifying drug delivery systems (SEDDS)
for improved oral delivery of lipophilic drugs. https://doi.org/10.1016/j.biopha.2004.02.001

Ivanov, I., & Kralchevsky, P A. (1997, August 1). Stability of emulsions under equilibrium and
dynamic conditions. https://doi.org/10.1016/s0927-7757(96)03903-9
Jain, S., Jain, A., Pohekar, M., & Thanki, K. (2013, December 1). Novel self-emulsifying
formulation of quercetin for improved in vivo antioxidant potential: Implications for drug-induced
cardiotoxicity and nephrotoxicity. https://doi.org/10.1016/j.freeradbiomed.2013.05.041

Jatto, E., & Okhamafe, A O. (2002, December 1). Review Article - An Overview of
Pharmaceutical Validation and Process Controls in Drug Development.
https://tspace.library.utoronto.ca/bitstream/1807/1485/1/pr02016.pdf

Karwal, R., Garg, T., Rath, G., & Markandeywar, T S. (2016, January 1). Current Trends in
Self-Emulsifying Drug Delivery Systems (SEDDSs) to Enhance the Bioavailability of Poorly Water-
Soluble Drugs. https://doi.org/10.1615/critrevtherdrugcarriersyst.v33.i1.20

Krstić, M., Medarević, D., Đuriš, J., & Ibrić, S. (2018, January 1). Self-nanoemulsifying drug
delivery systems (SNEDDS) and self-microemulsifying drug delivery systems (SMEDDS) as lipid
nanocarriers for improving dissolution rate and bioavailability of poorly soluble drugs.
https://doi.org/10.1016/b978-0-12-813687-4.00012-8

Maderuelo, C., Zarzuelo, A., & Lanao, J M. (2011, August 1). Critical factors in the release of
drugs from sustained release hydrophilic matrices. https://doi.org/10.1016/j.jconrel.2011.04.002

Malkawi, A., Jalil, A., Nazir, I., Matuszczak, B., Kennedy, R A., & Bernkop‐Schnürch, A. (2020,
August 25). Self-Emulsifying Drug Delivery Systems: Hydrophobic Drug Polymer Complexes Provide
a Sustained Release in Vitro. https://doi.org/10.1021/acs.molpharmaceut.0c00389

McClements, D J., & Jafari, S M. (2018, January 1). Improving emulsion formation, stability
and performance using mixed emulsifiers: A review. https://doi.org/10.1016/j.cis.2017.12.001

Pandey, V., & Kohli, S. (2018, January 1). Lipids and Surfactants: The Inside Story of Lipid-
Based Drug Delivery Systems. https://doi.org/10.1615/critrevtherdrugcarriersyst.2018016710

Petkova, R., Tcholakova, S., & Denkov, N D. (2012, March 8). Foaming and Foam Stability
for Mixed Polymer–Surfactant Solutions: Effects of Surfactant Type and Polymer Charge.
https://doi.org/10.1021/la3003096

Quanlu, L., Yinhong, Z., & Jing, W. (2013). Researches and applications of the ultrasonic
emulsifications and dispersions. The Journal of the Acoustical Society of America, 134(5), 3546-3550.

Rahman, M A., Hussain, A., Hussain, M S., Mirza, M A., & Iqbal, Z. (2012, February 29). Role
of excipients in successful development of self-emulsifying/microemulsifying drug delivery system
(SEDDS/SMEDDS). https://doi.org/10.3109/03639045.2012.660949

Reza, J. (2013, June 24). Self-Emulsifying Drug Delivery Systems: A Review.


https://doi.org/10.3329/ijpls.v2i2.15453

Severino, P., Andreani, T., Macedo, A S., Fangueiro, J F., Santana, M H A., Silva, A M., &
Souto, E B. (2012, November 24). Current State-of-Art and New Trends on Lipid Nanoparticles (SLN
and NLC) for Oral Drug Delivery. https://doi.org/10.1155/2012/750891

Shrestha, H., Bala, R., & Arora, S. (2014, May 19). Lipid-Based Drug Delivery Systems.
https://doi.org/10.1155/2014/801820

Singh, B., Beg, S., Khurana, R K., Sandhu, P S., Kaur, R., & Katare, O P. (2014, January 1).
Recent Advances in Self-Emulsifying Drug Delivery Systems (SEDDS).
https://doi.org/10.1615/critrevtherdrugcarriersyst.2014008502

Singh, D. (2021, June 1). Self-nanoemulsifying Drug Delivery System: A Versatile Carrier for
Lipophilic Drugs. https://doi.org/10.2174/2211738509666210422124023
Witika, B A., Bassey, K E., Demana, P H., Siwe‐Noundou, X., & Poka, M S. (2022, August 26).
Current Advances in Specialised Niosomal Drug Delivery: Manufacture, Characterization and Drug
Delivery Applications. https://doi.org/10.3390/ijms23179668

Yang, Y., Fang, Z., Chen, X., Zhang, W., Xie, Y., Chen, Y., Liu, Z., & Yuan, W. (2017, May
23). An Overview of Pickering Emulsions: Solid-Particle Materials, Classification, Morphology, and
Applications. https://doi.org/10.3389/fphar.2017.00287

Yang, Y., Marshall-Breton, C., Leser, M E., Sher, A., & McClements, D J. (2012, December 1).
Fabrication of ultrafine edible emulsions: Comparison of high-energy and low-energy homogenization
methods. https://doi.org/10.1016/j.foodhyd.2012.04.009
THE SECRETOME: CELL-FREE CARRIER SYSTEM

Ayşegül Taşkıran
Chapter 5 factors that mimic the therapeutic effects of
the cells, marking a shift towards cell-free

The Secretome: Cell-free therapeutic strategies. These strategies


promise not only to overcome the
carrier system limitations of direct cell therapies, such as
scalability and safety concerns, but also to
AYSEGÜL TAŞKIRAN1
provide more accessible and potentially
more effective treatments. As researchers
explore the MSC secretome and its
1. Introduction
extracellular vesicles, they uncover
The secretome of a cell, a rich tapestry opportunities to harness these bioactive
of soluble substances, including proteins, molecules in clinical settings, potentially
nucleic acids, and lipids, represents a revolutionizing how we approach
frontier in regenerative medicine. Central to regenerative medicine and patient care.
this domain are extracellular vesicles like This chapter delves into the science behind
exosomes and microvesicles, which the MSC secretome, its clinical implications,
encapsulate and convey these potent and the future it heralds for medical
molecules. This chapter explores the therapies.
innovative use of conditioned media to
2. The secretome and its content
capture the secretome from cells cultured
in controlled environments. Such media, The secretome represents the
enriched with cellular exudates like growth substances secreted by cells into the
factors, cytokines, and enzymes, offers a extracellular space, including proteins,
window into the cellular communication lipids, nucleic acids, and small molecules.
essential for tissue repair and regeneration. These components are crucial in
intercellular communication, immune
The burgeoning field of mesenchymal
responses, and tissue homeostasis. Cells
stem/stromal cell (MSC) therapy has paved
release these soluble molecules that might
the way for groundbreaking advances in
directly influence intracellular pathways in
treating various diseases. Yet, the full
damaged cells or indirectly stimulate
potential of MSCs may lie not in the cells
nearby tissues to produce active
themselves but in their secretome—soluble

1
Department of Histology and Embryology,
Faculty of Medicine, Ege University, Izmir 35100,
Türkiye.
substances (Kim et al., 2021a). The cell- (ESCRT) machinery or pathways that do
secretome comprises all substances not rely on ESCRT and consist of
actively or passively secreted from cells, tetraspanins (CD9, CD63, CD81), heat
such as soluble proteins (including shock proteins, miRNAs, and mRNAs. They
cytokines, chemokines, and growth engage in antigen presentation, facilitate
factors), lipids, free nucleic acids, and tumor metastasis, and facilitate horizontal
extracellular vesicles (EV). These molecules gene transfer (Kim et al., 2021b).
are released through various secretion
Microvesicles (100 to 1000 nm) are
mechanisms involving exocytosis, protein
bigger than exosomes and are created by
translocation, and vesicle or exosome
outward budding and fission of the plasma
encapsulation. Apoptotic bodies,
membrane. Vesicles include cytoplasmic
microvesicles, and exosomes are the
components such as proteins, lipids, and
primary categories into which EVs can be
nucleic acids. They have important blood
classified based on their size, density,
clotting, immune response, and cell
surface indicators, and origin (Jenner &
communication functions. Their
Ribitsch, 2022; Y. J. Liu & Wang, 2023).
development entails the reorganization of
Exosomes, in particular, have garnered
the cytoskeleton and the redistribution of
significant attention in studies due to their
membrane phospholipids. This process
ability to connect with target cells and alter
involves several proteins, including
cell signaling. Understanding the secretome
integrins, selectins, signaling molecules,
components and the carrier systems
and RNA species. Microvesicles have a role
involved in its transport is vital for
in blood clot formation, cancer
comprehending cellular functions and
advancement, and immune system
developing therapeutic strategies (Arbade
regulation (Manzoor et al., 2023).
et al., 2024).
Apoptotic bodies (500 to 2000 nm) are
Exosomes are tiny extracellular vesicles
rather large vesicles produced during the
(30 to 150 nm) originating from endosomes
latter phases of programmed cell death,
and are released into the extracellular
also known as apoptosis. The contents of
environment when multivesicular bodies
these structures include cellular debris,
(MVBs) merge with the plasma membrane.
organelles, nuclear fragments, and proteins
Exosomes transport proteins, lipids, and
enclosed by membranes. These contents
nucleic acids, enabling cell communication
are then engulfed by nearby or specialized
and influencing immunological responses.
immune cells called macrophages. This
They are triggered by either the endosomal
procedure facilitates the removal of dying
sorting complexes required for transport
cells and ensures the balance of tissue are then incubated in this conditioning
stability. Apoptotic bodies aid in preventing medium for a specific period, typically 24 to
autoimmunity and maintaining cellular 72 hours, to allow for the secretion of
function by easing the elimination of bioactive molecules. After the incubation
apoptotic cells and containing potentially period, the conditioned medium containing
hazardous intracellular substances (Tang et the secretome is collected. The collected
al., 2022). medium undergoes centrifugation at a low
speed to remove cellular debris, followed by
Cell-free regenerative medicine
a higher-speed centrifugation. The
treatments provide greater advantages
supernatant is then filtered through a 0.22
than traditional stem cell-based therapies.
μm filter to ensure the removal of any
Secretomes can prevent immunological
remaining cells or large particles (Cases-
compatibility, tumorigenicity, and transfer
Perera et al., 2022; Gwam et al., 2021).
of infections in stem cell treatments due to
Depending on the intended application, the
their cell-free carrier systems. The
secretome can be concentrated using
secretome has the potential to significantly
ultrafiltration techniques, which involve
decrease the expenses and time involved in
passing the conditioned medium through a
growing and sustaining cell lines. This is
filter with a specific molecular weight cut-
because the secretome may be created in
off. This step helps concentrate the
advance in large amounts and can be
bioactive molecules. The concentrated
readily used for therapy when required.
secretome is then aliquoted into sterile
Obtaining the secretome from cells in tubes and stored at -80°C to preserve its
vitro involves a series of carefully controlled integrity, with precautions to avoid
steps to ensure the collection of the repeated freeze-thaw cycles. In some
bioactive molecules secreted by the cells. cases, further characterization of the
The process begins with the cell culture in secretome might be necessary. Techniques
a suitable medium containing essential such as ELISA, mass spectrometry, or
nutrients and growth factors. The cells can proteomics can be used to identify and
adhere and proliferate until they reach the quantify the bioactive components. By
desired confluency, usually around 70- following these steps, researchers can
80%. At this point, the growth medium is obtain a purified and concentrated cell
replaced with a serum-free or low-serum secretome in vitro, which can be utilized for
medium. This is a crucial step as it ensures various therapeutic and research
that the secretome collected is free from applications. This method provides a
contamination by serum proteins. The cells reliable way to harness the therapeutic
potential of mesenchymal stem cell-derived preserving their ability to differentiate into
secretomes, opening new possibilities for several cell types throughout several
non-invasive cell-free treatments in generations. Due to their significant worth
regenerative medicine and emergencies in therapeutic settings, stem cells are
such as heart attacks, stroke, or severe extensively used in regenerative medicine
injuries (Cases-Perera et al., 2022). to restore or substitute impaired tissues.
Moreover, their ability to modulate the
immune system makes them valuable in
3. Mesenchymal stem cell and its treating autoimmune illnesses and
secretome inflammatory conditions. MSCs are utilized

The secretome can be derived from any in tissue engineering to create novel tissues

cell: stem cells, stromal cells, embryonic and organs, as well as to function as

cells, adult cells, cancer cells, etc., one of research models for investigating disease

which is the mesenchymal stem cells causes and evaluating novel

(MSCs). MSCs are adult stem cells that can pharmaceuticals (Kahrizi et al., 2023;

differentiate into various cell types, Margiana et al., 2022; Moghadasi et al.,

including adipocytes, osteoblasts, and 2021).

chondrocytes. These cells, which can The current studies are focused on
regenerate themselves, are predominantly comprehending the mechanisms of MSC
in bone marrow, muscle, and adipose differentiation, enhancing their therapeutic
tissue. However, these cells may also be capacity, and creating novel clinical uses.
found in Wharton's jelly, dental pulp, Research is investigating their potential in
peripheral blood, skin, lungs, chorionic villi, treating osteoarthritis, cardiovascular
menstrual blood, and breast milk. They are diseases, and neurological disorders(T.
characterized by distinct surface markers, Zhou et al., 2021). Although MSCs show
such as the positive biomarkers CD105, great potential, there are still obstacles to
CD73, and CD90, and the negative overcome, such as the requirement for
biomarkers CD45, CD34, CD14/CD11b, standardized isolation techniques and
CD79α/CD19, and HLA-DR. When culture MSCs to guarantee uniformity in
cultivated in vitro, these cells adhere to research and therapeutic uses. Although
plastic surfaces and have a spindle-shaped MSCs avoid the ethical concerns related to
morphology (Eleuteri & Fierabracci, 2019). embryonic stem cells, it is still necessary to

MSCs can be easily isolated and carefully address the ethical aspects of their

cultured, allowing for their expansion while acquisition and obtaining authorization for
their use (Wang et al., 2022; Zhuang et al., wound healing through several
2021). mechanisms, including increasing cell
migration, proliferation, and angiogenesis
MSCs are highly esteemed not only for
(Ahangar et al., 2020). Growth factors
their ability to differentiate into several cell
(PDGF, IGF-1, EGF, FGF, VEGF, etc.),
types and ease of extraction but also for
inflammatory proteins (interleukins, tumor
their secretomes, encompassing a diverse
necrosis factor-alpha, etc.), extracellular
array of biologically active substances such
matrix proteins (matrix metalloproteinases,
as cytokines, growth factors, and
ICAM, collagens, decorin, and laminin), and
extracellular vesicles. These released
angiogenic factors are among the
substances serve vital roles in modifying
secretome components that are pertinent
the cellular surroundings, stimulating tissue
to different stages of wound healing
healing, and producing effects that regulate
(Ibrahim et al., 2022). Park et al. showed
the immune system. The secretome of
that human adipose tissue-derived MSC
MSCs has been discovered to promote the
secretome enhances the activities of
growth of new blood vessels
dermal fibroblasts, keratinocytes, and
(angiogenesis), decrease inflammation,
endothelial cells, which are critical for tissue
and aid in repairing injured tissues (Chouw
repair. In vitro, secretome improved the
et al., 2022). MSCs are desirable for
migration and proliferation of skin cells by
therapeutic purposes since their positive
activating PI3K/Akt or FAK/ERK1/2
benefits may be utilized without directly
pathways, leading to faster wound closure.
transplanting cells, minimizing possible
Secretome treatment accelerated wound
difficulties linked to stem cell treatment.
healing, reduced inflammation, and
Ongoing research extensively investigates
enhanced collagen deposition and re-
the therapeutic possibilities of secretomes
epithelialization in vivo (Park et al., 2018).
produced from MSCs, intending to create
During the healing process of skin lesions
innovative and non-invasive “cell-free”
caused by radiation, the MSC-secretome
therapies for various diseases such as
facilitated the growth of HUVEC cells, the
cardiovascular, neurological, and
regeneration of sebaceous glands, and the
autoimmune disorders.
formation of new blood vessels, resulting in
either no scar formation or fewer scars (Sun
4. The use of MSC secretome in
et al., 2019). The therapeutic potential of
regenerative medicine
secretome is significant, particularly for
MSC secretome plays a crucial role in chronic wounds, burns, diabetic foot ulcers,
tissue repair and regeneration. It promotes and other skin injuries. Clinical studies have
indicated that secretome can be a hypoxia-preconditioned bone marrow MSCs
promising alternative to traditional wound increases growth factors and cytokines,
healing therapies, providing a more promoting cell proliferation, migration, and
efficient and targeted approach to skin angiogenesis (Antebi et al., 2018; Chen et
regeneration. Also, the MSC secretome is al., 2014; Ferreira et al., 2018). Hypoxia-
widely used in central nervous system preconditioned umbilical cord MSCs
pathologies such as spinal cord injuries (W. upregulate angiogenesis genes and reduce
Liu et al., 2019; Noh et al., 2016; inflammation in ischemic models (Han et
Rosenzweig et al., 2018; Tsai et al., 2019; al., 2016). For neurological diseases,
X. Zhou et al., 2019), ischemic stroke (W. hypoxia enhances neuroprotective
Huang et al., 2015; Lee et al., 2016; Xin et proteins. Additionally, hypoxia
al., 2013), Parkinson’s disease (D’angelo et preconditioning can reduce oxidative stress
al., 2020; Oh et al., 2017; Teixeira et al., in hepatic cells and improve osteogenic
2017), Alzheimer’s disease (Giovannelli et differentiation through epigenetic changes
al., 2023; Kuo et al., 2021; Santamaria et (Gwam et al., 2021; Hong et al., 2019).
al., 2021), etc (Pinho et al., 2020).
Stem cell secretome-based therapy is a
Stem cells can be prepared for a specific highly promising approach for treating
purpose by subjecting them to physical various conditions. While extensive analysis
stimulation, such as hypoxia, stress, and of the bioactive components within the
heat shock, or inducing biochemical secretome is crucial, research is also
changes, such as depriving them of serum focused on developing effective delivery
or adding anti-inflammatory chemicals. platforms tailored to specific diseases. The
These preparations can enhance the stem most common method is systemic
cell's ability to prevent cell death, increase administration, particularly intravenous
blood vessel formation, and reduce delivery. However, this can lead to
inflammation (Li et al., 2022). MSCs are excessive accumulation of the secretome in
usually cultured under normoxia (21% the liver and spleen, necessitating multiple
oxygen), but tissue oxygen levels are injections at higher doses to achieve
typically hypoxic (1-7% in bone marrow therapeutic efficacy. On the other hand,
and 10-15% in adipose tissue) (Trigo et al., localized delivery using various biomaterials
2024). Studies have shown that hypoxia- (hydrogels, microneedles, nanoparticles,
preconditioned MSCs secretome enhances scaffold patches) can enhance the targeted
their therapeutic potential. For cutaneous delivery and retention of the secretome in
wound healing, the secretome from specific tissues. This approach requires
lower dosages, improves stability, and diabetic wound environments. The results
enhances the functional effectiveness of demonstrated that the MSC-secretome-
the stem cell secretome (Arifka et al., 2022; loaded hydrogel significantly enhanced
Y. Huang et al., 2022; Li et al., 2022). An both hyperglycemic fibroblasts' proliferative
innovative therapeutic approach involves and migratory activities, indicating its
using engineered nanoparticles to potential effectiveness in improving wound
encapsulate the secretome of MSCs to treat healing for diabetic patients (Sears et al.,
chronic osteoporosis. The secretome, when 2021). Adipose tissue-derived
combined with nanoparticles, enables mesenchymal stem cell secretome
precise distribution to bone tissue, incorporated into a collagen hydrogel
especially in cases of osteoporosis. This enhanced skin-derived cell proliferation,
focused strategy amplifies the localized improved angiogenesis in endothelial cells,
therapeutic impacts, substantially and exhibited antimicrobial properties
enhancing animal models' bone mineral (Klimczak et al., 2021).
density, trabecular volume, and thickness.
On the other hand, there are also some
The secretome promotes the growth and
limitations to using MSC secretome. The
specialization of MSCs and osteoblasts. The
variability in stem cell batches and sources
nanoparticles containing secretome hinder
leads to diverse secretomes, complicating
the development of osteoclasts. This
clinical standardization. Culture conditions
inhibitory effect aids in reducing bone
also impact the secretome composition,
resorption, a prominent issue in
necessitating standardized cell culture
osteoporosis (Zhang et al., 2022). The
methods. Given the complex interactions
researchers developed a crosslinked
within the secretome, advanced
hyaluronic acid hydrogel as a delivery
technologies like proteomic analysis are
vehicle for the MSC-secretome, creating a
needed to identify specific therapeutic
sustained release system that enhances the
effectors. Novel tools such as label-free
repair of endometrial tissue for patients
microfluidic electrochemical biosensors
with Asherman's Syndrome. This therapy
have been developed for continuous
restored endometrial damage and
measurement of secreted biomarkers,
facilitated successful pregnancies in vivo (F.
aiding in understanding the therapeutic
Liu et al., 2019). Sears et al. evaluated the
mechanisms of the stem cell secretome.
effects of a hydrogel infused with MSC-
secretome on the proliferation and 5. Conclusion

migration of fibroblasts under In conclusion, the cell secretome is a

hyperglycemic conditions, which mimic dynamic and multifaceted system


encompassing many proteins, lipids, vesicles and the MSC secretome, the
nucleic acids, and small molecules. The immense therapeutic promise of these
secretome ensures efficient and regulated entities becomes increasingly apparent.
transport of these bioactive components They replicate the regenerative effects of
through the coordinated actions of carrier MSCs and provide a scalable, cost-effective
systems such as exosomes, microvesicles, solution amenable to clinical
apoptotic bodies, and secretory lysosomes. standardization. This cell-free approach
Exploring the complexities of the secretome addresses direct stem cell therapies'
and its carriers unveils deep insights into logistical and ethical challenges. It
cellular communication and disease promises new treatment options for various
pathways, paving the way for innovative diseases that currently lack effective
therapeutic approaches. interventions. The MSC-derived secretome
Moreover, the secretome, brimming may become a foundational element in the
with bioactive substances, stands at the emerging landscape of medical treatments,
forefront of regenerative medicine, offering new hope for enhanced
potentially surpassing the capabilities of therapeutic outcomes across numerous
traditional stem cell therapies. As research debilitating conditions.
deepens our understanding of extracellular

Abbreviation list

EV extracellular vesicles

MVBs multivesicular bodies

ESCRT endosomal sorting complexes are required for the transport

MSCs mesenchymal stem cells


References
Ahangar, P., Mills, S. J., & Cowin, A. J. (2020). Mesenchymal stem cell secretome as an emerging
cell-free alternative for improving wound repair. In International Journal of Molecular Sciences (Vol. 21,
Issue 19, pp. 1–15). MDPI AG. https://doi.org/10.3390/ijms21197038

Antebi, B., Rodriguez, L. A., Walker, K. P., Asher, A. M., Kamucheka, R. M., Alvarado, L.,
Mohammadipoor, A., & Cancio, L. C. (2018). Short-term physiological hypoxia potentiates the
therapeutic function of mesenchymal stem cells. Stem Cell Research & Therapy, 9(1).
https://doi.org/10.1186/S13287-018-1007-X

Arbade, G., Varghese Jose, J., Gulbake, A., Kadam, S., Kashte, S. B., Arbade, G., Jose, J. V,
Gulbake, A., Kadam, S., & Kashte, S. B. (2024). From stem cells to extracellular vesicles: a new horizon
in tissue engineering and regenerative medicine. Cytotechnology 2024, 1–39.
https://doi.org/10.1007/S10616-024-00631-4

Arifka, M., Wilar, G., Elamin, K. M., & Wathoni, N. (2022). Polymeric Hydrogels as Mesenchymal
Stem Cell Secretome Delivery System in Biomedical Applications. In Polymers (Vol. 14, Issue 6). MDPI.
https://doi.org/10.3390/polym14061218

Cases-Perera, O., Blanco-Elices, C., Chato-Astrain, J., Miranda-Fernández, C., Campos, F., Crespo,
P. V., Sánchez-Montesinos, I., Alaminos, M., Martín-Piedra, M. A., & Garzón, I. (2022). Development of
secretome-based strategies to improve cell culture protocols in tissue engineering. Scientific Reports,
12(1). https://doi.org/10.1038/s41598-022-14115-y

Chen, L., Xu, Y., Zhao, J., Zhang, Z., Yang, R., Xie, J., Liu, X., & Qi, S. (2014). Conditioned Medium
from Hypoxic Bone Marrow-Derived Mesenchymal Stem Cells Enhances Wound Healing in Mice. PLoS
ONE, 9(4), 96161. https://doi.org/10.1371/JOURNAL.PONE.0096161
Chouw, A., Facicilia, G., Sartika, C. R., Faried, A., & Milanda, T. (2022). Factors Influencing the
Therapeutic Potential of the MSC-derived Secretome. In Regenerative Engineering and Translational
Medicine (Vol. 8, Issue 3, pp. 384–393). Springer Science and Business Media Deutschland GmbH.
https://doi.org/10.1007/s40883-021-00242-x

D’angelo, M., Cimini, A., & Castelli, V. (2020). Insights into the effects of mesenchymal stem cell-
derived secretome in Parkinson’s disease. In International Journal of Molecular Sciences (Vol. 21, Issue
15, pp. 1–14). MDPI AG. https://doi.org/10.3390/ijms21155241

Eleuteri, S., & Fierabracci, A. (2019). Insights into the secretome of mesenchymal stem cells and
its potential applications. In International Journal of Molecular Sciences (Vol. 20, Issue 18). MDPI AG.
https://doi.org/10.3390/ijms20184597

Ferreira, J. R., Teixeira, G. Q., Santos, S. G., Barbosa, M. A., Almeida-Porada, G., & Gonçalves, R.
M. (2018). Mesenchymal stromal cell secretome: Influencing therapeutic potential by cellular pre-
conditioning. In Frontiers in Immunology (Vol. 9). Frontiers Media S.A.
https://doi.org/10.3389/fimmu.2018.02837

Giovannelli, L., Bari, E., Jommi, C., Tartara, F., Armocida, D., Garbossa, D., Cofano, F., Torre, M.
L., & Segale, L. (2023). Mesenchymal stem cell secretome and extracellular vesicles for
neurodegenerative diseases: Risk-benefit profile and next steps for the market access. Bioactive
Materials, 29, 16–35. https://doi.org/10.1016/J.BIOACTMAT.2023.06.013
Gwam, C., Mohammed, N., & Ma, X. (2021). Stem cell secretome, regeneration, and clinical
translation: a narrative review. Annals of Translational Medicine, 9(1), 70–70.
https://doi.org/10.21037/ATM-20-5030

Han, K. H., Kim, A. K., Kim, M. H., Kim, D. H., Go, H. N., & Kim, D. I. (2016). Enhancement of
angiogenic effects by hypoxia-preconditioned human umbilical cord-derived mesenchymal stem cells in
a mouse model of hindlimb ischemia. Cell Biology International, 40(1), 27–35.
https://doi.org/10.1002/CBIN.10519

Hong, H. E., Kim, O. H., Kwak, B. J., Choi, H. J., Kim, K. H., Ahn, J., & Kim, S. J. (2019). The
antioxidant action of hypoxic conditioned media from adipose-derived stem cells in the hepatic injury of
expressing higher reactive oxygen species. Annals of Surgical Treatment and Research, 97(4), 159–167.
https://doi.org/10.4174/astr.2019.97.4.159

Huang, W., Lv, B., Zeng, H., Shi, D., Liu, Y., Chen, F., Li, F., Liu, X., Zhu, R., Yu, L., & Jiang, X.
(2015). Paracrine Factors Secreted by MSCs Promote Astrocyte Survival Associated With GFAP
Downregulation After Ischemic Stroke via p38 MAPK and JNK. Journal of Cellular Physiology, 230(10),
2461–2475. https://doi.org/10.1002/JCP.24981

Huang, Y., Li, X., & Yang, L. (2022). Hydrogel Encapsulation: Taking the Therapy of Mesenchymal
Stem Cells and Their Derived Secretome to the Next Level. In Frontiers in Bioengineering and
Biotechnology (Vol. 10). Frontiers Media S.A. https://doi.org/10.3389/fbioe.2022.859927
Ibrahim, R., Mndlovu, H., Kumar, P., Adeyemi, S. A., & Choonara, Y. E. (2022). Cell Secretome
Strategies for Controlled Drug Delivery and Wound-Healing Applications. In Polymers (Vol. 14, Issue
14). MDPI. https://doi.org/10.3390/polym14142929

Jenner, F., & Ribitsch, I. (2022). Secretome, Extracellular Vesicles, Exosomes. Orthobiologics:
Injectable Therapies for the Musculoskeletal System, 155–166. https://doi.org/10.1007/978-3-030-
84744-9_12

Kahrizi, M. S., Mousavi, E., Khosravi, A., Rahnama, S., Salehi, A., Nasrabadi, N., Ebrahimzadeh,
F., & Jamali, S. (2023). Recent advances in pre-conditioned mesenchymal stem/stromal cell (MSCs)
therapy in organ failure; a comprehensive review of preclinical studies. Stem Cell Research & Therapy
2023 14:1, 14(1), 1–18. https://doi.org/10.1186/S13287-023-03374-9
Kim, H. J., Kim, G., Lee, J., Lee, Y., & Kim, J. H. (2021a). Secretome of Stem Cells: Roles of
Extracellular Vesicles in Diseases, Stemness, Differentiation, and Reprogramming. Tissue Engineering
and Regenerative Medicine 2021 19:1, 19(1), 19–33. https://doi.org/10.1007/S13770-021-00406-4
Kim, H. J., Kim, G., Lee, J., Lee, Y., & Kim, J. H. (2021b). Secretome of Stem Cells: Roles of
Extracellular Vesicles in Diseases, Stemness, Differentiation, and Reprogramming. Tissue Engineering
and Regenerative Medicine 2021 19:1, 19(1), 19–33. https://doi.org/10.1007/S13770-021-00406-4
Klimczak, A., Hinc, P., Krawczenko, A., Bielawska‐pohl, A., Paprocka, M., Witkowska, D., Isa, I. L.
M., Pandit, A., & Kraskiewicz, H. (2021). Hatmsc secreted factors in the hydrogel as a potential treatment
for chronic wounds—in vitro study. International Journal of Molecular Sciences, 22(22).
https://doi.org/10.3390/ijms222212241

Kuo, S. C., Chio, C. C., Yeh, C. H., Ma, J. T., Liu, W. P., Lin, M. T., Lin, K. C., & Chang, C. P. (2021).
Mesenchymal stem cell-conditioned medium attenuates the retinal pathology in amyloid-β-induced rat
model of Alzheimer’s disease: Underlying mechanisms. Aging Cell, 20(5).
https://doi.org/10.1111/ACEL.13340
Lee, J. Y., Kim, E., Choi, S. M., Kim, D. W., Kim, K. P., Lee, I., & Kim, H. S. (2016). Microvesicles
from brain-extract-treated mesenchymal stem cells improve neurological functions in a rat model of
ischemic stroke. Scientific Reports, 6. https://doi.org/10.1038/srep33038

Li, F., Zhang, J., Yi, K., Wang, H., Wei, H., Chan, H. F., Tao, Y., & Li, M. (2022). Delivery of Stem
Cell Secretome for Therapeutic Applications. In ACS Applied Bio Materials (Vol. 5, Issue 5, pp. 2009–
2030). American Chemical Society. https://doi.org/10.1021/acsabm.1c01312

Liu, F., Hu, S., Yang, H., Li, Z., Huang, K., Su, T., Wang, S., & Cheng, K. (2019). Hyaluronic Acid
Hydrogel Integrated with Mesenchymal Stem Cell-Secretome to Treat Endometrial Injury in a Rat Model
of Asherman’s Syndrome. Advanced Healthcare Materials, 8(14).
https://doi.org/10.1002/adhm.201900411

Liu, W., Wang, Y., Gong, F., Rong, Y., Luo, Y., Tang, P., Zhou, Z., Zhou, Z., Xu, T., Jiang, T., Yang,
S., Yin, G., Chen, J., Fan, J., & Cai, W. (2019). Exosomes derived from bone mesenchymal stem cells
repair traumatic spinal cord injury by suppressing the activation of a1 neurotoxic reactive astrocytes.
Journal of Neurotrauma, 36(3), 469–484.
https://doi.org/10.1089/NEU.2018.5835/ASSET/IMAGES/LARGE/FIGURE10.JPEG

Liu, Y. J., & Wang, C. (2023). A review of the regulatory mechanisms of extracellular vesicles-
mediated intercellular communication. Cell Communication and Signaling 2023 21:1, 21(1), 1–12.
https://doi.org/10.1186/S12964-023-01103-6

Manzoor, T., Saleem, A., Farooq, N., Dar, L. A., Nazir, J., Saleem, S., Ismail, S., Gugjoo, M. B.,
Shiekh, P. A., & Ahmad, S. M. (2023). Extracellular vesicles derived from mesenchymal stem cells — a
novel therapeutic tool in infectious diseases. Inflammation and Regeneration 2023 43:1, 43(1), 1–17.
https://doi.org/10.1186/S41232-023-00266-6

Margiana, R., Markov, A., Zekiy, A. O., Hamza, M. U., Al-Dabbagh, K. A., Al-Zubaidi, S. H., Hameed,
N. M., Ahmad, I., Sivaraman, R., Kzar, H. H., Al-Gazally, M. E., Mustafa, Y. F., & Siahmansouri, H.
(2022). Clinical application of mesenchymal stem cell in regenerative medicine: a narrative review. Stem
Cell Research & Therapy 2022 13:1, 13(1), 1–22. https://doi.org/10.1186/S13287-022-03054-0
Moghadasi, S., Elveny, M., Rahman, H. S., Suksatan, W., Jalil, A. T., Abdelbasset, W. K.,
Yumashev, A. V., Shariatzadeh, S., Motavalli, R., Behzad, F., Marofi, F., Hassanzadeh, A., Pathak, Y., &
Jarahian, M. (2021). A paradigm shift in cell-free approach: the emerging role of MSCs-derived
exosomes in regenerative medicine. Journal of Translational Medicine 2021 19:1, 19(1), 1–21.
https://doi.org/10.1186/S12967-021-02980-6

Noh, M. Y., Lim, S. M., Oh, K.-W., Cho, K.-A., Park, J., Kim, K.-S., Lee, S.-J., Kwon, M.-S., & Kim,
S. H. (2016). Mesenchymal Stem Cells Modulate the Functional Properties of Microglia via TGF-β
Secretion. Stem Cells Translational Medicine, 5(11), 1538–1549. https://doi.org/10.5966/SCTM.2015-
0217/-/DC1

Oh, S. H., Kim, H. N., Park, H. J., Shin, J. Y., Kim, D. Y., & Lee, P. H. (2017). The Cleavage Effect
of Mesenchymal Stem Cell and Its Derived Matrix Metalloproteinase-2 on Extracellular α-Synuclein
Aggregates in Parkinsonian Models. Stem Cells Translational Medicine, 6(3), 949–961.
https://doi.org/10.5966/SCTM.2016-0111

Park, S. R., Kim, J. W., Jun, H. S., Roh, J. Y., Lee, H. Y., & Hong, I. S. (2018). Stem Cell Secretome
and Its Effect on Cellular Mechanisms Relevant to Wound Healing. Molecular Therapy, 26(2), 606–617.
https://doi.org/10.1016/j.ymthe.2017.09.023
Pinho, A. G., Cibrão, J. R., Silva, N. A., Monteiro, S., & Salgado, A. J. (2020). Cell secretome: Basic
insights and therapeutic opportunities for CNS disorders. In Pharmaceuticals (Vol. 13, Issue 2). MDPI
AG. https://doi.org/10.3390/ph13020031

Rosenzweig, E. S., Brock, J. H., Lu, P., Kumamaru, H., Salegio, E. A., Kadoya, K., Weber, J. L.,
Liang, J. J., Moseanko, R., Hawbecker, S., Huie, J. R., Havton, L. A., Nout-Lomas, Y. S., Ferguson, A.
R., Beattie, M. S., Bresnahan, J. C., & Tuszynski, M. H. (2018). Restorative Effects of Human Neural
Stem Cell Grafts to the Primate Spinal Cord. Nature Medicine, 24(4), 484.
https://doi.org/10.1038/NM.4502

Santamaria, G., Brandi, E., Vitola, P. La, Grandi, F., Ferrara, G., Pischiutta, F., Vegliante, G., Zanier,
E. R., Re, F., Uccelli, A., Forloni, G., de Rosbo, N. K., & Balducci, C. (2021). Intranasal delivery of
mesenchymal stem cell secretome repairs the brain of Alzheimer’s mice. Cell Death and Differentiation,
28(1), 203–218. https://doi.org/10.1038/S41418-020-0592-2
Sears, V., Danaoui, Y., & Ghosh, G. (2021). Impact of mesenchymal stem cell-secretome-loaded
hydrogel on proliferative and migratory activities of hyperglycemic fibroblasts. Materials Today
Communications, 27. https://doi.org/10.1016/j.mtcomm.2021.102285
Sun, J. Y., Zhang, Y. F., Song, X. J., Zhu, J., & Zhu, Q. S. (2019). The Healing Effects of Conditioned
Medium Derived from Mesenchymal Stem Cells on Radiation-Induced Skin Wounds in Rats. Cell
Transplantation, 28(1), 105–115. https://doi.org/10.1177/0963689718807410
Tang, H., Luo, H., Zhang, Z., & Yang, D. (2022). Mesenchymal Stem Cell-Derived Apoptotic Bodies:
Biological Functions and Therapeutic Potential. Cells 2022, Vol. 11, Page 3879, 11(23), 3879.
https://doi.org/10.3390/CELLS11233879

Teixeira, F. G., Carvalho, M. M., Panchalingam, K. M., Rodrigues, A. J., Mendes-Pinheiro, B., Anjo,
S., Manadas, B., Behie, L. A., Sousa, N., & Salgado, A. J. (2017). Impact of the Secretome of Human
Mesenchymal Stem Cells on Brain Structure and Animal Behavior in a Rat Model of Parkinson’s Disease.
Stem Cells Translational Medicine, 6(2), 634–646. https://doi.org/10.5966/SCTM.2016-0071
Trigo, C. M., Rodrigues, J. S., Camões, S. P., Solá, S., & Miranda, J. P. (2024). Mesenchymal stem
cell secretome for regenerative medicine: Where do we stand? Journal of Advanced Research.
https://doi.org/10.1016/j.jare.2024.05.004

Tsai, M. J., Liou, D. Y., Lin, Y. R., Weng, C. F., Huang, M. C., Huang, W. C., Tseng, F. W., & Cheng,
H. (2019). Attenuating Spinal Cord Injury by Conditioned Medium from Bone Marrow Mesenchymal Stem
Cells. Journal of Clinical Medicine, 8(1). https://doi.org/10.3390/JCM8010023

Wang, Y., Qi, Z., Yan, Z., Ji, N., Yang, X., Gao, D., Hu, L., Lv, H., Zhang, J., & Li, M. (2022).
Mesenchymal Stem Cell Immunomodulation: A Novel Intervention Mechanism in Cardiovascular Disease.
In Frontiers in Cell and Developmental Biology (Vol. 9). Frontiers Media S.A.
https://doi.org/10.3389/fcell.2021.742088

Xin, H., Li, Y., Cui, Y., Yang, J. J., Zhang, Z. G., & Chopp, M. (2013). Systemic administration of
exosomes released from mesenchymal stromal cells promote functional recovery and neurovascular
plasticity after stroke in rats. Journal of Cerebral Blood Flow & Metabolism, 33(11), 1711.
https://doi.org/10.1038/JCBFM.2013.152

Zhang, C., Zhang, W., Zhu, D., Li, Z., Wang, Z., Li, J., Mei, X., Xu, W., Cheng, K., & Zhong, B.
(2022). Nanoparticles functionalized with stem cell secretome and CXCR4-overexpressing endothelial
membrane for targeted osteoporosis therapy. Journal of Nanobiotechnology, 20(1).
https://doi.org/10.1186/s12951-021-01231-6
Zhou, T., Yuan, Z., Weng, J., Pei, D., Du, X., He, C., & Lai, P. (2021). Challenges and advances in
clinical applications of mesenchymal stromal cells. In Journal of Hematology and Oncology (Vol. 14,
Issue 1). BioMed Central Ltd. https://doi.org/10.1186/s13045-021-01037-x

Zhou, X., Chu, X., Yuan, H., Qiu, J., Zhao, C., Xin, D., Li, T., Ma, W., Wang, H., Wang, Z., & Wang,
D. (2019). Mesenchymal stem cell derived EVs mediate neuroprotection after spinal cord injury in rats
via the microRNA-21-5p/FasL gene axis. Biomedicine & Pharmacotherapy, 115, 108818.
https://doi.org/10.1016/J.BIOPHA.2019.108818

Zhuang, W. Z., Lin, Y. H., Su, L. J., Wu, M. S., Jeng, H. Y., Chang, H. C., Huang, Y. H., & Ling, T. Y.
(2021). Mesenchymal stem/stromal cell-based therapy: mechanism, systemic safety and biodistribution for
precision clinical applications. In Journal of Biomedical Science (Vol. 28, Issue 1). BioMed Central Ltd.
https://doi.org/10.1186/s12929-021-00725-7

You might also like