Nanovesículas de Tamaño Controlado para Aplicaciones Biotecnológicas Y Bioanalíticas
Nanovesículas de Tamaño Controlado para Aplicaciones Biotecnológicas Y Bioanalíticas
Nanovesículas de Tamaño Controlado para Aplicaciones Biotecnológicas Y Bioanalíticas
TESIS DOCTORAL
Oviedo, 2019
UNIVERSITY OF OVIEDO
Computational Modelling
Ph.D. THESIS
Oviedo, 2019
RESUMEN DEL CONTENIDO DE TESIS DOCTORAL
2.- Autor
Nombre: Pablo García Manrique DNI/Pasaporte/NIE: -Q
Tras una introducción sobre las generalidades de las nanovesículas, el quinto capítulo de
la tesis presenta una profunda revisión del estado del arte del concepto de exosoma artificial,
así como la revisión crítica de las diferentes aproximaciones metodológicas para su
producción y una nueva clasificación de estos nuevos biomateriales en función de los
mismos. Además, se han expuesto los usos biotecnológicos de dichas partículas, como son
la distribución y liberación controlada de fármacos, su papel en diagnóstico, y su posible
empleo en el desarrollo y estudio de métodos bioanalíticos. Para ello, es necesario procesos
adaptados para la preparación de exosomas puramente artificiales mediante técnicas nano-
biotecnológicas. Esta temática es precisamente sobre la que versa el trabajo experimental
expuesto en los siguientes capítulos.
Nanovesicles, a particular type of organic colloids, is generating a growing interest in the field
of biotechnology, and particularly in the field of bioanalysis. The versatility of its morphological
and functional characteristics makes them a very promising biomaterial. Some of these
aplications are as drug delivery vehicle, multilabel system for immnunoassays or biomietic
membrane or extracellular vesicles. However, these new applications impose a series of
requirements on preparation methods that carry along a technological challenge and have
triggered a very active field of research. Control over the particle size and its monodispersity are
essential characteristics that must be controlled by improving current methods and proposing
new technologies designed for such purpose. Withing this framework, the general objective of
this Doctoral Thesis is the development of efficient methods for controlled size syntesis. The
nanovesicles obtained have been used to mimic exosomes (extracelular vesicles) with the aim
of investigating their use as analytical standard at immunoassays. These nanovesicles are
attracting high interest as novel biomarkers at the biomedical field, and novel approaches are
necessary in order to develop analytical methods.
After an introduction about the general concepts of nanovesicles, the fifth chapter presents
a deep review of the state of the art of the artificial exosome concept, as well as the critical
evaluation of the different approaches for its production. A systematic classification of these
new biomaterials was carried out, with the aim of providing some reference guides for
standardization the field. In addition, the biotechnological application of these particles has been
discussed. This includes the distribution and controlled release of drugs, their role in diagnosis,
and their possible use in the development of bioanalytical methods. For that purpose, novel
methods are required for the preparation of fully artificial exosomes by using nano-
biotechnological techniques. This topic is the core of the experimental work presented in the
following chapters.
Finally, in the nineth chapter the models obtained for niosomes in previous stages have been
used to obtain particles with an average size and monodispersity similar to that of natural
exosomes. In addition, by surface functionalization first with streptavidin and in a second step
with recombinant biotinylated tetraspanin peptides (classical molecular biomarkers of
exosomes), fully artificial exosomes have been developed. These particles have been tested by
ELISA immunoassays as a potential new analytical standard for the development and/or
enhacement of bioanalytical methods based on molecular recognition for the determination of
extracellular vesicles, such as exosomes.
1. Introducción ....................................................................................................................... 27
2. Objetivos/Objetives ............................................................................................................ 37
3.3.2. Bioconjugación....................................................................................................... 68
3.4. Exosomas: ejemplo de sistema vesicular natural con interés biotecnológico ...... 73
4. Experimental ........................................................................................................................ 93
5
4.3. Turbiscan (Formulaction): estabilidad coloidal ....................................................... 98
5.3. Impact of the artificial EVs classification at the design of new therapeutic agents
based on EVs ............................................................................................................................ 108
5.4.2 Obtaining a good substrate for modification: isolation procedures. ............ 111
5.5. Top-down and bottom-up methods for the development of full synthetic EVs122
6
6.2.3 Vesicles preparation ............................................................................................. 149
7.3.4. Effect of surfactant acyl chain length over particles size and monodispersity
................................................................................................................................................ 191
7
7.5. Bibliographic references ............................................................................................ 193
8. Particle size and encapsulation efficiency dependence with cargo molecular weight
and film hydration solution for niosomes formulated with mixture of surfactants .......... 199
9.2.3. Tetraspanins (CD9/63) large extracellular loops (LELs) production .......... 228
8
9.3. Results and discussion............................................................................................... 230
9.3.2. Artificial EVs production using Nio_Str functionalized with tetraspanin LEL
................................................................................................................................................ 233
9
Índice de Figuras
Figura 3.1. Diversidad química y morfológica de las vesículas artificiales creadas por
autoensamblaje de moléculas anfifílicas. Figura adaptada de la referencia 1. SUV: small
unilamelar vesicles; LUV: large unilamelar vesicles; MLV: multilamelar vesicles.............. 45
Figura 3.2. Estructura típica de una molécula anfifílica, en la que se señalan sus dos
porciones principales: la cabeza polar y la cola apolar, entre las cuales se establece la
interfase cuando son puestas en disolución acuosa. ................................................................ 46
Figura 3.3. (A) Representación gráfica del concepto del parámetro de
empaquetamiento crítico o CPP sobre un fosfolípido (superior) y dos surfactantes no
iónicos como son el Span® 60 y Span® 20 (medio e inferior respectivamente) que difieren
únicamente en longitud de cadena apolar. (B) Implicaciones geométricas de los valores de
CPP sobre la morfología del agregado de compuestos anfifílicos, figura adaptada de la
referencia 6. .................................................................................................................................... 48
Figura 3.4. Factores que condicionan la curvatura de la bicapa, como el parámetro de
empaquetado crítico que deriva de la forma de las moléculas anfifílicas, y como éste afecta
al radio de curvatura R durante la etapa de crecimiento de la bicapa discoidal. Figura
compuesta y adapta de la referencia 7. ...................................................................................... 49
Figura 3.5. Simulación informática del remodelado que sufre la micela inicial de
moléculas anfifílicas durante su auto-ensamblado en bicapas para formar vesículas. Estos
cambios en la estructura de la micela se producen como fruto de interacciones cabeza-
cabeza en la superficie y cola-cola en el interior al interactuar las moléculas con las de
agua de la fase contínua. Este reordenamiento lleva a la creación de poros por los que
circula el agua (B1 y B2), y este paso de disolvente acaba por reordenar la estructura hasta
formar un disco plano (D1). En rojo se representan las moléculas de agua, mientras que en
azul la cabeza polar y en negro la cola apolar. Figura tomada de la referencia 11. ............. 50
Figura 3.6. Surfactantes no iónicos más frecuentes en formulaciones de niosomas. Se
representan de arriba abajo en función del valor creciente de HBL, que surge de la
diferente longitud de cadena apolar que poseen los miembros de una misma familia de
surfactante. Figura tomada de la referencia 114. ...................................................................... 54
Figura 3.7. Diversidad morfológica de las vesículas, y su relación con los métodos de
preparación. Figura adaptada de la referencia 9. ..................................................................... 59
11
Figura 3.8. Diferentes estrategias de mejora de las propiedades básicas de las vesículas
artificiales, basadas en la incorporación de biomoléculas bioactivas, para el desarrollo de
biomateriales multifuncionales. Figura adaptada de la referencia 14. .................................. 67
Figura 3.9. Esquema ilustrativo de los grupos funcionales involurados en las
principales estrategias de conjugación basadas en el establecimiento de un enlace químico
entre la superficie de una vesícula y una biomolécula. (a) Entrecruzamiento de aminas
primarias mediante glutaraldehido, (b) enlace amida entre un carboxilo y una amina
primaria, (c) enlace amida mediante reacción de para-nitrofenilcarbonil con una amina
primaria, (d) puente disulfuro, (e) enlace tioester mediante reacción con maleimidetiol, y
(f) enlace hidrazona. Figura adaptada de la referencia 118. .................................................... 68
Figura 3.10. (A) Estrategia de bioconjugación basada en la interacción Estreptavidina-
Biotina, donde la Estreptavidina se conjuga previamente sobre la superficie de la bicapa
mediante alguno de los métodos mostrados en la figura 10, y el otro elemento se
encuentra biotinilado. (B) Conjugación de biomoléculas mediante el método de la
carbodiimida empleando EDC/NHS como reactivos de conjugación. Este método puede
ser empleado para la conjugación de Estreptavidina como se indica en el caso A.............. 70
Figura 3.11. Estrategias de purificación de vesículas para la eliminación del material
no encapsulado o conjugado. (A) Diálisis, (B) (Ultra)centrifugación, y (C) Cromatografía
de Exclusión de Tamaño (SEC) por filtración en gel. ............................................................... 71
Figura 3.12. Representación gráfica de la estructura y componentes de un exosomas
natural. Figura adaptada de la referencia . ................................................................................ 74
Figura 3.13. Ventajas (verde) y desventajas (rojo) de los dos tipos de aproximaciones
metodológicas basadas en bionanotecnología para la producción de exosomas puramente
artificiales con fines teranósticos. ................................................................................................ 76
Figura 4.1. Equipo Zetasizer NANO-ZS, Malvern Instruments. ...................................... 96
Figura 4.2. Microscopio TEM JEOL-2000 ExII, Jeol. ........................................................... 97
Figura 4.3. Equipo Turbiscan LAB Expert con estación de envejecimiento,
Formulaction. ................................................................................................................................. 98
Figura 4.4. Equipo de HPLC HP serie 1100 con módulo de detección UV/Vis HP
G1315A UV/ 281, Hewlett-Packard. ........................................................................................ 100
Figura 4.5. Lector de placas y absorbancia TECAN Genios, Tecan Trading AG. ........ 100
Figura 4.6. Analizador de imágenes de luminiscencia para geles y membranas
LAS4000 mini Image System analyser de, GE Healthcare. ................................................... 101
Figura 4.7. Equipo de DSC 822e, Mettler-Toledo. ............................................................ 102
12
Figure 5.1. Artificial EVs landscape: explored routes up-to-date for the preparation of
artificial EVs for specific purposes.*EBSSNs (ref. 19)............................................................. 107
Figure 6.1. Pareto chart of the standardized effects of independent variables (factors)
on (A) Z-average size and (B) PDI of PC liposomes for the Plackett-Burman fractional
factorial design............................................................................................................................. 151
Figure 6.2. Pareto chart of the standardized effects of independent variables (factors)
on the (A, C) Z-average size and (B,D) PDI of (A,B) PC liposomes and (C,D) S60:Cho
niosomes (1:0.5, w/w) for the 23 full factorial design............................................................ 155
Figure 6.3. Three-dimensional (3D) response surface plots for the factors O/A
(organic/aqueous phase volume ratio), C (lipid or surfactant/stabilizer concentration,
g/L), and A (sonication amplitude, %) for the (A,C) Z-average size and (B,D) PDI of (A,B)
PC liposomes and (C,D) S60:Cho niosomes (1:0.5, w/w)...................................................... 156
Figure 6.4. Contour Plot for the factors O:A (organic:aqueous phase volume ratio), C
(lipid or surfactant/stabilizer concentration, g/L) and A (sonication amplitude, %) on Z-
average size, nm (A) and PDI (C) of PC-liposomes, and Z-average size, nm (B) and PDI
(D) of S60:Cho niosomes (1:0.5, w/w). ..................................................................................... 157
Figure 6.5. Optimization plot and values of individual (d) and composite (D)
desirability provided by the response optimizer (Minitab, version 17) for an example of
size-tuned PC liposome (desired size = 70 nm, with a minimum PDI value). ................... 163
Figure 6.6. Optimization plot and values of individual (d) and composite (D)
desirability provided by the response optimizer (Minitab, version 17) for an example of
size-tuned S60:Cho niosome (1:0.5 w/w) (desired size = 240 nm, with a minimum PDI).
........................................................................................................................................................ 164
Figure 6.7. (A,B) BS profiles and (C,D) TEM micrographs of empty vesicles designed
with a controlled size and PDI values by applying the models obtained from experimental
design: (A,C) PC liposomes and (B,D) S60:Cho niosomes (1:0.5, w/w). ............................ 165
Figure 7.1. Schematic diagram of a continuous flow microreactor based on
hydrodynamic flow focusing for vesicular systems production. The reduction of focused
stream width under laminar flow conditions makes possible the mixing of chemical
species by molecular diffusion, since time for mixing decreases with the square root of
distance. By changing flow rates, the kinetics and extension of mixing can be modified,
and then, the size of particles. Amphiphilic molecules are self-assembled into bilayers
once critical concentration of solvent is reached, and molecules acquired an ordered state
13
to minimize the interaction with water molecules. At a certain size bending modulus
induce planar bilayer to be closed into vesicles. ..................................................................... 175
Figure 7.2. Pictures composition showing the whole setup (central) and detailed
components (sides) used in this work for niosomes production by Hydrodynamic Flow
Focusing with controlled temperature. .................................................................................... 177
Figure 7.3. Calibration plot (A) and temperature stability (B) of the in-house designed
thermostatic chamber for microfluidics chips, fabricated by 3D-printing technology with
PLA filaments. Values represented are the average of three independents measurements.
........................................................................................................................................................ 180
Figure 7.4. Figure 2. Precipitation of Span® 60 (Tm=45 °C) at room temperature (upper
arrow) at the focusing region (left), 0.5 and 1.0 cm downstream (centre and right). At a
temperature above surfactant Tm, focusing is complete and vesicles formation could be
checked by negative staining (Phosphotungstic acid 2%) and Transmission Electron
Microscopy (TEM). ...................................................................................................................... 183
Figure 7.5. Size (nm) and size distribution (PDI, a.u.) measured by DLS in undiluted
samples from niosomes formulated with Span® 60:Cholesterol (1:0.5 molar ratio) at 5mM
(A,C) and 20mM (B,D) in a continuous flow microreactor based on hydrodynamic flow
focusing at controlled temperature (50 °C). Each condition was tested twice, and each
batch was measured by triplicate. ............................................................................................. 184
Figure 7.6. Effect of bilayer components concentration for niosomes formulated with
Span® 60:Cholesterol (1:0.5 molar ratio) and produced under the same flow conditions
(QT and FRR) for size (upper row) and size distribution (lower row). ............................... 186
Figure 7.7. Images (4X) of focusing region and end of the mixing channel evidencing
hydrodynamic flow focussing of a central ethanol stream at different FRR values for
QT=100 μL/min and 50 °C. Yellow colour indicates acid pH (pure EtOH, no mixing),
while blue colour indicates basic pH (complete mixing by codiffusion of solvent and no
solvent). Bromoxylenol blue dye was dissolved in EtOH (acidified with acetic acid), and
PBS was adjusted with NaOH to pH 10. .................................................................................. 187
Figure 7.8. Values of ethanol focused stream (Wf, μm) as flow rate ratio (FRR)
increased for different values of volumetric rates (QT) at constant temperature (50 °C),
and different temperatures at constant QT (100 μL/min). Values represent the average of
two independent measurements, taken at approx. at 100 μm from the end of focussed
region. ........................................................................................................................................... 188
14
Figure 7.9.Size (nm) (left) and size distribution (PDI, a.u.) (right) measured by DLS in
undiluted samples from niosomes formulated with Span® 20:Cholesterol (1:0.5 molar
ratio) at 5mM in a continuous flow microreactor based on hydrodynamic flow focusing at
different controlled temperatures (30, 40, 50, and 60 °C). Each condition was tested twice,
and each batch was measured by triplicate. ............................................................................ 190
Figure 7.10. Influence of acyl chain length (C12 and C18 for Span® 20 and Span® 60
respectively) of two different sorbitan sters used in niosomes formulation (surfactant:
cholesterol 1:0.5 molar ratio, 5 mM), and produced under different conditions by
hydrodynamic flow focusing at controlled temperature (50 °C) and a flow rate QT=100
μl/min. .......................................................................................................................................... 192
Figure 8.1. Chemical structures of the encapsulated compounds ................................. 202
Figure 8.2. (A) Size values of empty vesicles. Tw80: tween® 80; Sp80: span® 80; Dc:
dodecanol. (B) Effect of hydration solution and cargo molecular weight over particle size,
for niosomal formulations Tw80:Sp80 (1:1 molar ratio) and Tw80:Sp80:Dc (1:1:1 molar
ratio) prepared by Thin Film Hydration method (TFH)........................................................ 209
Figure 8.3. Effect of hydration solution and cargo molecular weight over encapsulation
efficiecy, for niosomal formulations (A)Tw80:Sp80 (1:1 molar ratio) and (B) Tw80:Sp80:Dc
(1:1:1 molar ratio) prepared by Thin Film Hydration method (TFH). Tw80: tween® 80;
Sp80: span® 80; Dc: dodecanol. Dialysis (10 MWCO membranes) and gel filtration
(Sepahdex G25 or Seharose CL-4B, depending on the compound) were used as
purification methods................................................................................................................... 212
Figure 8.4. Naparticle Tracking Analysis (NTA) of the three different hydration
solution based niosomes after and before SEC purification. MQ and Gly vesicles were
purified using Sephadex G25 and PEG vesicles by Sepharose CL-4B. ................................ 215
Figure 8.5. DSC curves acquired in heating mode for the formulation without
dodecanol in the three different hydration solutions: ultrapure water (MQ), water:glycerol
60:40 v/v (Gly), and water:PEG-400 55:45 v/ v (PEG). ......................................................... 216
Figure 9.1. Schematic fully artificial exosome produced by bottom-up bio-
nanotechnological methods based on supramolecular chemistry. The different molecular
components are detailed with their functions......................................................................... 226
Figure 9.2. Figure 2. (A) Elution profile of a solution of streptavidin from a Sepharose
CL-4B SEC gravity elution column. Signal quantification of each 0.5Ml fraction was
measured by BCA total protein assay according with manufacturer instruction. Graph
insight shows (left) the first 3.5 mL collected of a suspension of red dye loaded niosomes
15
to allow their visualization, after their elution from the SEC column; (right) the SEC
column after the elution of the 3.5 mL of dyed niosomes. Both elements, niosomes and
free protein, eluted from the column enough separate to allow their separation based on
Sepharose CL-4B gravity elution coloumns. (B) Dot-Blot assay for checking the effectively
of streptavidin bioconjugation to niosomes through carbodiimide method (EDC/NHS).
Standards of different concentrations allows the semiquantification of the process by
comparison of spot intensity. The result shows the 5 different batches. Biotinylated-HRP
(4µg/mL) was used as detecting agent. (C) Size distribution by DLS of bared niosomes (or
NIO) and streptavidin-conjugated niosomes or (NIO_Str). The average size increment
(2nm) shown as peak displacement demonstrate the effective conjugation of the protein.
(D) Size distribution by DLS of the 5 different batches of NIO_Str (152 ± 3 nm), to
demonstrate the reproducibility of the process. ..................................................................... 232
Figure 9.3. (A) Dot-blot assays for revelation of LEL, CD9 or CD63, positive fractions
collected from a Sepharose CL-2B gravity elution SEC column. Mon- and double-
functionalized Nio_Str have been produced. (B) Size distribution measured by NTA
(Nanosight, Malvern Instruments) of previous described fully artificial exosomas
(NIO_LEL). A sample of natural exosomas isolated from mesenchymal triple-negative
breast cancer cell line SUM159 was also measured for comparison purposes. .................. 235
Figure 9.4. Dot-blot assay for the screening and selection of α-tetraspanin antibodies
for their future use in ELISAs for the detection of fully artificial exosomes (NIO_LEL).
Secondary antibodies labelled with HRP were appropriate selected. Different negative
controls were also introduced (bared niosomes, niosomes functionalized with streptavidin
with and without biotin saturation), as the use of isolated exosomas from mesenchymal
triple-negative breast cancer cell line SUM159 as positive control. Values are uniplicate
assays. (PU) monoclonal purified antibody; (biot) Biotynilated monoclonal antibody;
(poli) polyclonal antibody. ......................................................................................................... 236
Figure 9.5. (A,B) Cross-reactive responses for mono-functionalized LEL-tetraspanin
niosomes (fully articial exosomas) for different configuration of capture (c) and detection
(d) anti-tetraspanin antibodies. (C) ELISA assays for the detection of NIO_LEL9 (N9) and
NIO_LEL63 (N63) fully artificial exosomes using different combination of capture (c) and
detection (d) antibodies. Monoclonal α-CD9 or α-CD63 antibodies were used for capture,
whereas polyclonal α-CD9 or α-CD63 antibodies were used for detection. (D) ELISA
assays for the detection of NIO_LEL9/63 using different combinations of capture (c) and
16
detection (d) antibodies. The graphs shows the mean ± SD of 3 independent experiments.
*** p˂0.005, Student’s t-test. ....................................................................................................... 238
Figure 9.6. Dose-response graphs for different types of fully artificial exosomas
detected by ELISA assay using monoclonal and polyclonal antibodies α-tetraspanins CD9
y CD63 for capture and detection respectively. As secondary appropriate α-IgG-HRP was
used. N9 and N9/63 are NIO_LEL9 and NIO_LEL9/63 respectively, which are mono-
and double-functionalized LEL-tetraspanin niosomes. ......................................................... 240
17
Índice de Tablas
Tabla 3.1. Usos de los surfactantes no iónico en función de su valor HLB. ................... 55
Tabla 3.2. Ejemplos de aplicaciones biotecnológicas de los diversos tipos de sistemas
vesiculares según sector industrial y tipo de NVs.................................................................... 58
Tabla 3.3. Ventajas y desventajas de los métodos de purificación de vesículas. ........... 73
Table 5.1. Classification of techniques for the production of artificial EVs, mainly
exosomes, according to type of final product (semi- or fully synthetic) and the principle of
the obtention mechanism ........................................................................................................... 113
Table 5.2. Pre-isolation methods for cargo incorporation into EVs ............................... 115
Table 5.3. Post-isolation methods for cargo incorporation into EVs ..................................... 119
Table 5.4. Summary of the published work about the generation of mimetic EVs
nanovesicles by Top-Down bionanotechnology. Cell source and type of cargo are
encapsulated, and main characteristics are given. ................................................................. 123
Table 5.5. Advantages and disadvantage of most frequently used methods for Small
Unilamellar Vesicles (SUVs) preparation ................................................................................ 127
Table 5.6. Summary of published work about the development of mimetic EVs
nanovesicles by bottom-up bionanotechnology. Formulation of the vesicles, molecules for
the surface functionalization and main physical characteristic (size) are given. ............... 129
Table 6.1. Plackett-Burman Fractional Factorial Design: Responses, Levels, and Factors
........................................................................................................................................................ 148
Table 6.2. Full factorial design (23) with center point repetitions (n=5): factors, levels
and responses............................................................................................................................... 148
Table 6.3. ANOVA results (coded units) for Z-average size of PC-liposomes for the 23
full factorial design; results for S60:Cho niosomes are also given (cursive numbers) ...... 153
Table 6.4. ANOVA results (coded units) for PDI of PC-liposomes for the 23 full
factorial design; results for S60:Cho niosomes are also given (cursive numbers) ............. 154
Table 6.5. COOK's distance and DFITS values obtained for each response in the full
factorial designs ........................................................................................................................... 155
Table 6.6. Estimated coded coefficients for the considered effects on Z-average size
and PDI of PC liposomes and S60:Cho niosomes (1:0.5, w/w) ............................................ 162
Table 7.1. Morphological characteristics of mixing channel for original Solidworks®
CAD 2016 design and 3D-Printed positive moulds (3D-PM) onto VeroClearTM resin with
19
the 3D printer Objet350 ConnexTM (Stratsys). Average and standard deviation values are
given for the parameters............................................................................................................. 181
Table 7.2. Correlation factor between flow focusing parameters (FRR and Wf) and
particle size at different concentration of bilayer components and variable QT (A), and at
fixed concentration and QT for different working temperatures (B). .................................. 189
Table 8.1. Chromatography mediums used for gel filtration based purification of
loaded niosomes .......................................................................................................................... 206
Table 9.1. Some commercial available kits based on ELISA for exosomes quantification
in biological samples. Most of them are based on colorimetric signal quantification based
on HRP substrates, with a typical format of 96 well microtiter plate. ................................. 241
20
Lista de Abreviaturas
A
AA Ácido ascórbico
B
B12 Vitamina B12, conocida también como cobalamina
C
CRY2 Acronimo anglosajón para cryptochrome 2
D
DBCO Acrónimo anglosajón para dibenzocyclooctyne
Dc Dodecanol
21
E
EBSSNs
EDC 1-etil-3-(3-dimetilaminopropil)carbodiimida
EE Eficacia de encapsulación
EXPLORs Acrónimo anglosajón para exosomes for protein loading via optically reversible
protein–protein interactions
F
FRR Acrónimo inglés para flow rate ratio
G
Gly Glicerol, aunque también se refiere a una disolución agua:glicerol 60:40 v/v
H
HEK293 Acrónimo anglosajón para human embryonic kidney cell line 293
I
IDO Acrónimo anglosajón para indoleamine 2,3-dioxygenase
L
LEL Acrónimo anglosajón para large extracelular loop
22
LUV Acrónimo anglosajón para large unilamelar vesicles
M
miRNA Acrónimo anglosajón para micro interference ribonucleic acid
N
(sulfo)NHS Sulfo N-hyidroxyisulfosuccinimida
Nio niosomas
NVs Nanovesículas
O
O/A relación de volúmenes de fase orgánica/acuosa
P
PB Acronimo anglosajón para phosphate buffer
P-B Plackett-Burman
23
PEG Poly(Ethylene) glicol, aunque también se refiere a una disolución de
polietilengligol-400 55:45 v/v.
R
RB Rodamina B
S
SDS-PAGE Acronimo anglosajón para sodium dodecyl sulfate polyacrylamide gel
electrophoresis
Str Estreptavidina
T
TAMEL Acronimo anglosajón para targeted and molecular extracelular vesicles loading
TMB 3,3’,5,5’-Tetrametilbenzidina
U
UC Ultracentrifugación
Uv/vis Ultravioleta/visible
24
V
VE Vesículas extracelulares
Símbolos y unidades
λ Longitud de onda
ζ Potencial zeta
A Amplitud de sonicación (%)
P p-valor (u.a.)
Re Número de Reynolds
T Temperatura
V0 Volumen muerto
25
1. Introducción
Capítulo 1. Introducción
30
parámetros importantes en el bioanálisis de las VE. Por otra parte, su modificación
biotecnológica para incroporar moléculas bioactivas de diversa naturaleza química en
el interior o en superficie, hace necesario la adaptación o diseño de procesos adaptados
a tal fín, con la idea de producir estructuras biomiméticas de una forma adecuada y
asequible. Si bien los liposomas han destacado en estas aplicaciones, este campo en
auge, ofrece elevadas posibilidades para los niosomas también, y su empleo en
bioanalítica de exosomas está ofreciendo los primeros resultados. 35
31
respuestas diferenciadas atribuidas al material, y en cierta forma predecible. En otros
casos, como es su papel como marca en immunoensayos, es una característica casi más
importante que el propio tamaño. Puesto que a mayor tamaño, mayor es su superficie,
la capacidad de portar más elementos de reconocimiento aumenta en proporción, lo
que tiene importantes repercusiones sobre las características analíticas como puede ser
el límite de detección. Distribuciones de tamaños amplias introducen heterogeneidad
en el comportamiento de las partículas en cuanto a capacidades de reconocimiento y
generación de señal, ya que pueden producirse impedimentos estéricos que reduzcan
la señal analítica y generen un aumento en el límite de detección, siendo contrario a la
finalidad de su empleo en esta aplicación, como es una amplificación de la señal
analítica.
Referencias bibliográficas
[1] Willen Norden (editor) (2011). Colloids and interfaces in Life Sciences and
Bionanotechnologies, segunda edición, CRC Press, Taylor & Francis Group, New York, USA.
[2] De S., Kundu R., Biswas A. (2012). Synthesis of gold nanoparticles in niosomes. Journal of
Colloids and Interface Science, 386:9-15.
[3] Gómez-Hens A., Fernández-Romero J.M. (2005). The role of liposomes in analytical
process. Trends in Analytical Chemistry, 24:9-19.
[4] Chen I.A., Walde P. (2010). From self-assembled vesicles to protocells. Cold Spring Harbor
Perspectives in Biology, 2:a002170.
[5] Damera D.P., Krishna Venuganti V.V., Nag A. (2018). Deciphering the role of bilayer of a
noisome towards controlling the entrapment and release of dyes. Chemistry Select, 3:3930-3938.
32
[6] Caddeo C., Diez Sales O, Valentia D., Ruiz Saurí A., Fadda A.M., Manconi M. (2013).
Inhibition of skin inflammation in mice by diclofenac in vesicular carriers: Liposomes,
ethosomes and PEVs. Int. J. Pharm., 443:128-136.
[7] Hamid Reza Ahmadi Ashtiani H.R., Bishe P., Lashgari N.-A., Nilforoushzadeh M.A.,
Zare S. (2016). Liposomes in cosmetics. Journal of Skin and Stem Cell, 3(3):e65815.
[8] Gutiérrez G., Matos M., Barrero P., Pando D., Iglesias O., Pazos C. (2016). Iron-entrapped
niosomes and their potential application for yogurt fortification. Food Science and Technology,
74:550-556.
[9] Gatt S., Bercovier H., Barenholz Y. Use of liposomes for combating oil spills and their
potential application to bioreclamation. In: On-site bioreclamation: processes for xenobiotic and
hydrocaron treatment, editado por Hinchee R.E. and Olfenbuttel R.F. 1991, Battelle Press, USA.
[10] Eroğlu İ., İbrahim M. (2019). Liposome-Ligand Conjugates: A Review on the Current
State of Art. Journal of drug targeting, in press.
[11] Anbumozhi Angayarkanni S., Kampf N., Klein J. (2019). Surface interactions between
boundary layers of poly(ethylene oxide)-liposome complexes: lubrication, bridging and
selective ligation. Langmuir, in press.
[12] Al-Jamal W.T. and Kostarelos K. (2007). Liposome–nanoparticle hybrids for multimodal
diagnostic and therapeutic applications. Nanomedicine, 2(1):85-98.
[13] Carugo D., Bottaro E., Owen J., Stride E., and Nastruzzi C. (2016). Liposome production
by microfluidics: potential and limiting factors. Sci. Rep., 19(6):25876.
[14] Lombardo D., Calandra P., Barreca D., Magazù S., Kiselev M.A. (2016). Soft interaction
in liposome nanocarriers for therapeutic drug delivery. Nanomaterials, 6(7):125.
[15] Ishida T., Ichihara M., Wang X.Y., Yamamoto K., Kimura J., Majima E., Kiwada H.
(2006). Injection of PEGylated liposomes in rats elicits PEG-specific IgM, which is responsible
for rapid elimination of a second dose of PEGylated liposomes. J. Control Release., 112(1):15-25.
[16] Lukyanov A.N., Elbayoumi T.A., Chakilam A.R., Torchilin V.P. (2004). Tumor-targeted
liposomes: doxorubicin-loaded long-circulating liposomes modified with anti-cancer antibody.
J. Control Release, 100(1):135-144.
[17] Sihorkar V., Vyas S.P. (2001). Potential of polysaccharide anchored liposomes in drug
delivery, targeting and immunization. J. Pharm. Pharm. Sci, 4(2):138-158.
33
[18] Kunisawa J. , Masuda T., Katayama K., Yoshikawa T., Tsutsumi Y., Akashi M., Mayumi
T., Nakagawa S. (2005). Fusogenic liposome delivers encapsulated nanoparticles for cytosolic
controlled gene release. J. Control Release., 105(3):344-353.
[19] Kumar Rengan A., Banerjee R., Srivastava R. (2012). Thermosensitive gold-liposome
hybrid nanostructures for photothermal therapy of cancer. Comunicación oral. 12th IEEE
International Conference on Nanotechnology (IEEE-NANO). 20-23 Agosto, Birmingham, Reino
Unido.
[20] Lee J., Lee H., Goh U., Kim J., Jeong M., Lee J., Park J.H.. (2016). Cellular engineering
with membrane fusogenic liposomes to produce functionalized extracellular vesicles. ACS
Applied Materials and Interfaces, 8(11):6790-6795.
[21] Sato Y.T., Umezaki K., Sawada S., Mukai S., Sasaki Y., Harada N., Shiku H., Akiyoshi
K. (2016). Engineering hybrid exosomes by membrane fusion with liposomes. Sci. Rep., 6:21933.
[22] Yáñez-Mó M., Siljander P.R., Andreu Z., Zavec A.B., Borràs F.E., Buzas E.I., Buzas K.,
Casal E., Cappello F., Carvalho J., Colás E., Cordeiro-da Silva A., Fais S., Falcon-Perez J.M.,
Ghobrial I.M., Giebel B., Gimona M., Graner M., Gursel I., Gursel M., Heegaard N.H., Hendrix
A., Kierulf P., Kokubun K., Kosanovic M., Kralj-Iglic V., Krämer-Albers E.M., Laitinen S., Lässer
C., Lener T., Ligeti E., Linē A., Lipps G., Llorente A., Lötvall J., Manček-Keber M., Marcilla A.,
Mittelbrunn M., Nazarenko I., Nolte-'t Hoen E.N., Nyman T.A., O'Driscoll L., Olivan M.,
Oliveira C., Pállinger É., Del Portillo H.A., Reventós J., Rigau M., Rohde E., Sammar M.,
Sánchez-Madrid F., Santarém N., Schallmoser K., Ostenfeld M.S., Stoorvogel W., Stukelj R., Van
der Grein S.G., Vasconcelos M.H., Wauben M.H., De Wever O. (2015). Biological properties of
extracellular vesicles and their physiological functions. Journal of Extracellular Vesicles,
14(4):27066.
[23] Cappello, F., Logozzi, M., Campanella, C., Bavisotto, C. C., Marcilla, A., Properzi, F.,
Fais, S. (2017). Exosome levels in human body fluids: a tumor marker by themselves?. European
Journal of Pharmaceutical Sciences, 96:93-98.
[24] Jung K.O., Jo H., Yu J.H., Gambhir S.S., Pratx G. (2018). Development and MPI tracking
of novel hypoxia-targeted theranostic exosomas. Biomaterials, 177:139-148.
[25] Liu Q., Boyd B.J. (2013) Liposomes in biosensors. Analyst, 138:391-409.
[26] Lee M., Durst R.A., Wong R.B. (1998). Development of flow-injection liposome
immunoanalysis (FILIA) for imazethapyr. Talanta, 46:851-859.
34
[27] Lin B., Liu D., Yan J., Qiao Z., Zhong Y., Yan J., Zhu Z., Ji T., Yang C.J. (2016). Enzyme-
encapsulated liposome-linked immunosorbent assay enabling sensitive personal glucose meter
readout for portable detection of disease biomarkers. ACS Applied Material & Interfaces, 8:6890-7.
[28] Edwards K.A., Korff R., Baeumner A.J. (2017). Liposome-Enhanced Lateral-Flow
Assays for Clinical Analyses. Methods in Molecular Biology., 1571:407-434
[29] Ho J.A., Wu L.-C. , Huang M.R., Lin Y.-J., Baeumner A.J., Durst R.A. (2007) Application
of Ganglioside-Sensitized Liposomes in a Flow Injection Immunoanalytical System for the
Determination of Cholera Toxin. Analytical Chemistry, 79:246-250.
[30] Olguín Y., Villalobos P., Carrascosa L.G., Young M., Valdez E., Lechuga L., Galindo R.
(2013). Detection of flagellin by interaction with human recombinant TLR5 immobilized in
liposomes. Anal. Bioanal. Chem., 405(4):1267-1281.
[31] Shweta Pawar S., Bhattacharya A., Nag A. (2019). Metal-Enhanced Fluorescence Study
in Aqueous Medium by Coupling Gold Nanoparticles and Fluorophores Using a Bilayer Vesicle
Platform. ACS Omega, 4(3):5983-5990.
[32] Mazur F., Bally M., Städler B., Chandrawati R. (2017). Liposomes and lipid bilayers in
biosensors. Advanced in Colloids and Interface Science, 249:88-99.
[33] Chapman R., Lin Y., Burnapp M., Bentham A., Hillier D., Zabron A., Khan S., Tyreman
M., Stevens M.M. (2015). Multivalent nanoparticles networks enable point-of-care detection of
human phospholipase-A2 in serum. ACS Nano, 9:256-73.
[34] Lane R. E., Korbie D., Anderson W., Vaidyanathan R., Trau, M. (2015). Analysis of
exosome purification methods using a model liposome system and tunable-resistive pulse
sensing. Sci. Rep., 5:7639.
[35] Lozano-Andrés E., Libregts S.F., Toribio V., Royo F., Morales S., López-Martín S., Valés-
Gómez M., Reyburn H.T., Falcón-Pérez J.M., Wauben M.H., Soto M., Yáñez-Mó M. (2019).
Tetraspanin-decorated extracellular vesicle-mimetics as a novel adaptable reference material.
Journal of Extracellular Vesicles, 8(1):1573052.
[36] Danaei M., Dehghankhold M., Ataei S., Davarani Hasanzadeh F., Javanmard R.,
Dokhani A., Khorasani S., Mozafari M.R. (2018). Impact of particle size and polidespersity index
on the clinical applications oflipidic nanocarrier systems. Pharmaceutics, 10(57):1-17.
35
[37] Kraft J.C., Freeling, J.P., Wang Z., Ho R.J. (2014). Emerging research and clinical
development trends of liposome and lipid nanoparticle drug delivery systems. J. Pharm. Sci.,
103:29-52.
[38] Blasi P., Giovagnoli S., Schoubben A., Ricci M., Rossi C. (2007). Solid lipid nanoparticles
for targeted brain drug delivery. Adv. Drug. Deliv. Rev., 59:454-477.
[39] Andar A.U., Hood R.R., Vreenland W.N., DeVoe D.L., Swaan P.W. (2014). Microfluidic
preparation of liposomes to determine particle size influence on cellular uptake mechanism.
Pharmaceutical Research, 31:401-413.
36
2. Objetivos/Objetives
Capítulo 2. Objetivos
39
Chapter 2. Objetives
The main objective of this Ph.D. Thesis is the study of methodologies for the
preparation of vesicular systems (liposomes and niosomes), with special emphasis on
the controlled production of particle size for their use in bioanalytical applications.
Specifically, they will be used for the development of a mimetic model of exosomes for
their use as an analytical standard. This main objective has been divided into the
following specific objectives:
41
3. Consideraciones básicas
Capítulo 3. Consideraciones básicas
Las vesículas son un tipo de coloide orgánico artificial, formado por la agregación
supramolecular de moléculas anfifílicas que se disponen en una organización de
bicapa, que puede ser unilamelar o multilamelar. 1 Ésta, separa una cavidad acuosa
interior de la solución en la que se encuentran. La naturaleza química y la disposición y
tamaño de la misma establecen los tipos de vesículas artificiales que se pueden
encontrar en la literatura. La Figura 3.1 representa esta diversidad química y
morfológica que se pueden encontrar en este tipo de sistemas.
Figura 3.1. Diversidad química y morfológica de las vesículas artificiales creadas por autoensamblaje de
moléculas anfifílicas. Figura adaptada de la referencia 1. SUV: small unilamelar vesicles; LUV: large
unilamelar vesicles; MLV: multilamelar vesicles.
45
estructura supramolecular. 2 La característica por la que pueden o no formar bicapa se
comentará más adelante.
Figura 3.2. Estructura típica de una molécula anfifílica, en la que se señalan sus dos porciones
principales: la cabeza polar y la cola apolar, entre las cuales se establece la interfase cuando son puestas en
disolución acuosa.
46
Este punto se conoce generalmente como concentración crítica de agregación o CCA, que
en el caso de la formación de micelas recibe el nombre de concentración micelar crítica o
CMC, valor de extremada importancia en la predicción de fenómenos de interfase en
disoluciones acuosas de tensioactivos.
𝑣𝑣
𝑐𝑐𝑐𝑐𝑐𝑐 = Ecuación 1
𝑎𝑎𝑎𝑎
*1 Término general para referirse a la porción apolar de la molécula anfifílica que actúa como monómero
de la estructura.
47
Figura 3.3. (A) Representación gráfica del concepto del parámetro de empaquetamiento crítico o CPP
sobre un fosfolípido (superior) y dos surfactantes no iónicos como son el Span® 60 y Span® 20 (medio e
inferior respectivamente) que difieren únicamente en longitud de cadena apolar. (B) Implicaciones
geométricas de los valores de CPP sobre la morfología del agregado de compuestos anfifílicos, figura
adaptada de la referencia 6.
48
Figura 3.4. Factores que condicionan la curvatura de la bicapa, como el parámetro de empaquetado crítico
que deriva de la forma de las moléculas anfifílicas, y como éste afecta al radio de curvatura R durante la
etapa de crecimiento de la bicapa discoidal. Figura compuesta y adapta de la referencia 7.
49
Figura 3.5. Simulación informática del remodelado que sufre la micela inicial de moléculas anfifílicas
durante su auto-ensamblado en bicapas para formar vesículas. Estos cambios en la estructura de la micela
se producen como fruto de interacciones cabeza-cabeza en la superficie y cola-cola en el interior al
interactuar las moléculas con las de agua de la fase contínua. Este reordenamiento lleva a la creación de
poros por los que circula el agua (B1 y B2), y este paso de disolvente acaba por reordenar la estructura
hasta formar un disco plano (D1). En rojo se representan las moléculas de agua, mientras que en azul la
cabeza polar y en negro la cola apolar. Figura tomada de la referencia 11.
50
encapsulados. Esa presencia de colesterol inhibe la transición gel-líquido de los
compuestos de membrana, 17 y por lo tanto permite alcanzar rápidamente valores de
módulos de bending que induzcan el cierre de la bicapa con prontitud, por lo que las
vesículas formadas son pequeñas al reducirse la etapa de crecimiento.
Por otra parte, pueden producirse subdominios como son las balsas lipídicas
(reordenamientos de lípidos que se caracterizan por un mayor grado de
empaquetamiento y por ende, una reducida fluidez).2 También se han descrito la
existencia de micro-dominios en estados fluidos. 21 Es comprensible pues que esto
modifique las energías involucradas en el cierre de la vesícula y en la duración de la
etapa de crecimiento. Así mismo, la incorporación de heterogeneidad en la formulación
y la posibilidad de reordenamientos intra-moleculares de los compuestos anfifílicos
con cadenas no rígidas, es la base de la existencia de vesículas estables y altamente
deformables como son los transfersomas. 22
51
componentes libres durante la formación de las vesículas son aspectos metodológicos
que también poseen impacto sobre las características de las vesículas. Este hecho
conforma la base del control del proceso e introduce la posibilidad de obtener sistemas
vesiculares con características deseadas. Para ello, el estudio de procesos y el diseño de
los mismos teniendo en cuenta estos aspectos son un campo de estudio que permite la
ampliación del empleo de estos biomateriales en innovadoras aplicaciones.
Los tipos de moléculas anfifílicas que han sido empleadas en formulaciones para la
preparación de sistemas vesiculares son numerosos, y con naturaleza química muy
diversa: fosfolípidos (neutros o cargados), esfingolípidos, copolímeros, ácidos grasos,
surfactantes no iónicos y/o de amonio cuaternario, o incluso mezclas de dos o más de
estos tipos de compuestos.
Liposomas
Sin lugar a dudas se trata del tipo de vesícula más empleado y sobre el que existe
un mayor número de publicaciones tanto experimentales como de revisión. Parte de
esta popularidad se debe a las ventajas que poseen, como son un alto grado de bio-
compatibilidad, su capacidad de portar tanto moléculas hidrófilas como hidrófobas en
52
su interior o bicapa respectivamente y su carácter biodegradable. 25 Han sido
formulados tanto con composiciones muy sencillas 26 como con composiciones
miméticas de la composición natural de membranas biológicas, 27 y la recreación de
dominios de membrana han sido descritas con éxito. 28 Fosfatidilcolina, fosfatidilserina,
fosfatidiletanolamina, esfingomielina, y colesterol son algunos de los tipos de lípidos
naturales más comunes en liposomas.
Sin embargo, poseen una clara desventaja como es una reducida estabilidad en
suspensiones acuosas, donde los fenómenos de hidrólisis degradan los lípidos y
reducen la vida útil de los liposomas. Esta característica fue la fuerza impulsora para
explorar otros compuestos a la hora de preparar vesículas.
Niosomas
53
Figura 3.6. Surfactantes no iónicos más frecuentes en formulaciones de niosomas. Se representan de
arriba abajo en función del valor creciente de HBL, que surge de la diferente longitud de cadena apolar
que poseen los miembros de una misma familia de surfactante. Figura tomada de la referencia 114.
Estas formulaciones pueden manejarse como las de los liposomas, por lo que los
mismos tipos morfológicos de vesículas pueden obtenerse, ya que los métodos de
preparación empleados son los mismos en ambos tipos de vesículas.
54
Tabla 3.1. Usos de los surfactantes no iónico en función de su valor HLB.
1-3 Antiespumantes
3-6 Emulsiones agua en aceite
7-9 Agentes humectantes y dispersantes
8-18 Emulsiones aceite en agua
13-15 Detergencia
15-18 Solubilización
Transfersomas
Esfingosomas
Otro tipo de vesículas con base lipídica son los esfingosomas, nombre recibido en
alusión al tipo de lípido empleado en su formulación: los esfingolípidos. 35 El empleo de
estos lípidos tiene su origen en la búsqueda de una mayor estabilidad por ausencia de
fenómenos de oxidación en fase acuosa. 36 Como estabilizante de membrana para la
formación de una bicapa estable y con buenos valores de eficacia de encapsulación se
suele emplear colesterol. 37
55
Ufasomas
Otro tipo de vesículas con base mixta lipídica y de surfactantes son los ufasomas, 38
vesículas formuladas a base de ácidos grasos insaturados (oleico y linoleico) e
insaturados (octanoico y decanoico). De nuevo, su composición y propiedades de
membrana los hacen buenos candidatos para la preparación de vehículos de
administración de biocompuestos por vía tópica. 39
Etosomas
Este tipo de vesículas recuerda a los transfersomas, ya que poseen de nuevo una
base lipídica y otro componente encargado de modular la elasticidad mediante
relajación molecular de la bicapa. En este caso, esa función es llevada a cabo por
moléculas de alcoholes como el etanol e isopropanol en elevadas proporciones. 40 De
forma análoga a los casos anteriores, son de gran utilidad en la administración tópica. 41
Quatsomas
Polimersomas
56
3.1.2.1. El papel de los aditivos en las formulaciones
Uno de las moléculas más empleadas para fines estéricos es el polímero hidrofílico
poli(etilenglicol) o PEG. 49 Su incorporación post-preparación o la incorporación en
forma de lípido modificado aumenta considerablemente la vida de la vesícula, ya que
no solo aporta el citado impedimento estérico, si no que les hace “invisibles” al sistema
monocitico-nuclear. 50
Compuestos con carga negativa como dicetilfosfato y ácido fosfatídico, o con carga
positiva como estearilamina y cloruro de cetilpiridinio, son empleados frecuentemente6
para aportar carga superficial neutra diferente de cero (parámetro medido por el
potencial ζ). Esta carga, ayuda a mantener la estabilidad coloidal mediante repulsiones
electrostáticas, siendo ±30 mV un valor de ζ lo suficiente grande como para asegurar
dicha estabilidad. 51
57
A pesar de existir vesículas formuladas con ellos como son los ufasomas, en este caso la
proporción en la que se añaden hace que sean clasificados como aditivos.
Los lípidos y los esteroles son fácilmente modificables de forma química para
introducir nuevos grupos funcionales que permitan ser empleados en reacciones de
bioconjugación de moléculas o a superficies. 54 Son numerosos los ejemplos de su uso
para tal fin con liposomas y niosomas. 55- 57 Estos compuestos presentan un elevado
coste, aunque pueden ser preparados en el laboratorio,30 pero su empleo en relaciones
molares pequeñas respecto a los componentes mayoritarios permite su empleo con
facilidad y de una forma económicamente viable.
Tabla 3.2. Ejemplos de aplicaciones biotecnológicas de los diversos tipos de sistemas vesiculares según
sector industrial y tipo de NVs.
58
3.1.3. Tipos morfológicos de vesículas
Figura 3.7. Diversidad morfológica de las vesículas, y su relación con los métodos de preparación. Figura
adaptada de la referencia 9.
Por un lado tenemos vesículas unilamelares (una única bicapa separa el medio
acuoso de la cavidad interior), que se dividen de acuerdo en el tamaño en pequeñas
(conocidas como SUVs, Small Unilamelar Vesicles), o grandes (conocidad como LUVs,
Large Unilamer Vesicles). Estos tipos se relacionan con métodos de cambio de
polaridad del medio, como la inyección de disolventes, y con métodos que implican
hidratación de películas finas mediante descargas eléctricas o flujos de fase acuosa en
microcanales respectivamente. Estos métodos se comentan en la sección 3.2.
Por otro lado, existen vesículas con más de una bicapa, y éstas se encuentran de
forma concéntrica como si fuesen las capas de una cebolla (vesículas multamelares o
59
MLV). Generalmente presentan un tamaño grande, aproximadamente entorno a media
micra, y este tipo morfológico se asocia al método de hidratación de capa fina, donde la
película se hidrata de forma progresiva liberando bicapas que geman de la película
hasta cerrarse en vesículas.
Por otro lado, existen metodologías para reducir el tamaño o cambiar el tipo
morfológico, como son la extrusión 78 y la sonicación. 79 En el primero, el paso por una
membrana de tamaño de poro reducido hace que las vesículas se rompan en
fragmentos que se cierran en nuevas vesículas con un tamaño similar al del poro. En el
segundo, la aplicación de ultrasonidos en las suspensiones crea fenómenos de
cavitación con igual resultado que la extrusión. En este caso, el tamaño final va en
función de la amplitud de la frecuencia de los ultrasonidos.
60
A continuación se hace una breve descripción de los métodos de preparación
encontrados en la literatura. Se han agrupado en base al principio general en el que se
sustentan.
Inyección de etanol
Inyección de éter
61
formación de las vesículas en la etapa de eliminación del disolvente. 85 Únicamente se
emplea en la encapsulación de compuestos apolares, ya que la encapsulación pasiva de
compuestos hidrófilos no es compatible.
Existe una variedad, donde se evapora el primer disolvente tras la disolución de los
compuestos de membrana, y se forma una capa fina que se re-disuelve en un segundo
disolvente que se mezcla con la fase acuosa, y se procede como en la versión anterior. 86
En ambos casos, se forman LUVs y MLVs, por lo que se requieren etapas de post-
procesado para reducir el tamaño o cambiar el tipo de vesícula.
Eliminación de detergentes
Este método es el más antiguo, con el que se descubrieron los liposomas. Se parte
de una disolución orgánica que contiene los componentes de membrana y que
mediante rotavapor es desecada para forma una película final en las paredes del
matraz. Esta capa fina es hidratada por la fase acuosa a temperatura adecuada a la Tc
del sistema, y una fuerza mecánica en forma de agitación o ultrasonidos asiste en la
homogenización e hidratación. Debido a este proceso, MLVs es el producto
mayoritario, por lo que se requieren de etapas siguientes para obtener SUVs. Es
adecuada a todo tipo de compuestos, polares y apolares, e incluso para la
encapsulación de NPMs.12
Sonicación directa
62
a agitación contínua a medida que se reduce la temperatura. 89 En ocasiones, los
diferentes componentes de membrana son disueltos por separado y puesto en común
en la etapa de agitación. 90
Microfluidización
Proniosomas
lo que provoca un comportamiento híbrido entre líquido y gas, por lo que puede difundir como
un gas y disolver sustancias como un líquido.
63
consumo de disolventes orgánicos y de la toxicidad residual de su uso, (iii) facilidad
del proceso que se lleva a cabo en una única etapa, y (iv) esterilidad de la producción
inherente al empleo de fluidos supercríticos). 95
El fluido más empleado es CO2, por ser inerte, no tóxico, barato y no inflamable.
Este gas se mezcla con el disolvente que contiene los componentes de membrana a una
alta temperatura y presión. Esta mezcla se inyecta sobre la fase acuosa, y en ese
contacto se forman las vesículas mientras se evapora el disolvente al burbujear el gas.
Existen números ejemplos del empleo de estas técnicas para producir niosomas 96 y
liposomas. 97
Electroformación
64
ultrasonidos. Este método suele generar vesícular unilamelares gigantes (más de una
micra).
Mejoras de este método han ido encaminadas al uso de micro-patrones sobre los
electrodos, para conseguir un mayor control de la hidratación de la capa fina
depositada sobre ellos, y así, controlar el tamaño de partícula generada. 103
Similar al anterior, en este método es el flujo de la fase acuosa a través del canal la
fuerza que desprende las bicapas de la capa fina previamente depositada, 104 por lo que
se consiguen vesículas MLVs y altamente polidispersas.
Micro-extrusión
En esta técnica, se deposita una capa fina sobre una membrana de nitruro de silicio
con un tamaño de poro nanométrico (≤ 300 nm), y es impulsada por un flujo de fase
acuosa para atravesando la membrana, fragmentarse y general pequeñas vesículas de
tamaño controlado y monodisperso. 105
65
acuosos, y el empleo de micro pulsos de aire o fluido sobre la misma efectúa su
deformación hasta formar una vesícula que emerge de la bicapa cuando ésta se
deforma en exceso y se fragmenta.
66
Figura 3.8. Diferentes estrategias de mejora de las propiedades básicas de las vesículas artificiales,
basadas en la incorporación de biomoléculas bioactivas, para el desarrollo de biomateriales
multifuncionales. Figura adaptada de la referencia 14.
3.3.1. Encapsulación
En el segundo de los casos el proceso es activo puesto que se realiza una vez se han
creado las vesículas, por lo que es necesario mecanismos físico-químicos que
incorporen las moléculas a dichas estructuras. Estos mecanismos se basan en la
creación de gradientes transmembrana, bien sean de pH o iónicos. 114
67
Por otro lado, la composición de la bicapa condiciona la incorporación de moléculas
en la misma, y el peso molecular del compuesto a encapsular suele ser un parámetro
crucial. 115 Recordemos que las interacciones moleculares en la bicapa es parte de la
estabilidad de la misma, por lo que la llegada de nueva moléculas puede desestabilizar
el sistema. Por último, el método de preparación de vesículas puede jugar un papel
importante, 116 especialmente para la encapsulación pasiva ya que como se ha
mencionado, los mecanismos de formación de las vesículas son complejos y altamente
influenciables por variables metodológicas.
3.3.2. Bioconjugación
Figura 3.9. Esquema ilustrativo de los grupos funcionales involurados en las principales estrategias de
conjugación basadas en el establecimiento de un enlace químico entre la superficie de una vesícula y una
biomolécula. (a) Entrecruzamiento de aminas primarias mediante glutaraldehido, (b) enlace amida entre
un carboxilo y una amina primaria, (c) enlace amida mediante reacción de para-nitrofenilcarbonil con
una amina primaria, (d) puente disulfuro, (e) enlace tioester mediante reacción con maleimidetiol, y (f)
enlace hidrazona. Figura adaptada de la referencia 118.
68
Las diversas estrategias de bioconjugación de vesículas han sido revisadas 118.
Debido a esto, únicamente se comentarán aquellas estrategias que han sido empleadas
en la presente Tesis Doctoral.
69
reacciones cruzas e interacciones inespecíficas, los ésteres que quedan libres pueden ser
bloqueados con una amina primaria como puede ser la hidroxilamina o la glicina. 126
3.3.3. Purificación
Diálisis
Se trata de uno de los métodos más empleados en la literatura 127-129. Este proceso
está basado en los flujos osmóticos que se establecen a favor de gradiente de
concentración. La suspensión de nanovesículas se contiene en un saco o dispositivo
comercial separado del medio por una membrana, cuyo tamaño de poro deja pasar
únicamente las moléculas de compuesto no encapsulado o conjugado (ver Figura
3.11.A). Como inconvenientes de esta técnica destacan: el tiempo requerido para
completar la purificación y la necesidad de optimizarse para cada compuesto de forma
70
individual y la dilución de la muestra por entrada de medio de purificación hacia el
interior del cartucho o bolsa. Como ventajas destacan la capacidad de purificar grandes
lotes y la sencillez de la técnica.
Figura 3.11. Estrategias de purificación de vesículas para la eliminación del material no encapsulado o
conjugado. (A) Diálisis, (B) (Ultra)centrifugación, y (C) Cromatografía de Exclusión de Tamaño (SEC)
por filtración en gel.
Centrifugación/Ultracentrifugación
Se trata de otro método muy frecuente, 130,131 y no solo para sistemas vesiculares.
Este método emplea la mayor densidad de las vesículas, debido a su tamaño, respecto
a los otros elementos en disolución, ya que al someter a una suspensión de vesículas a
la fuerza centrífuga, éstas se irán al fondo del tubo pudiendo decantarse con facilidad
el sobrenadante (ver Figura 3.11.B). Este método permite a su vez una concentración
de la suspensión, ya que el pellet puede reconstituirse en un volumen menor. En
función de la fuerza aplicada en el proceso, se habla de centrifugación o
ultracentrifugación. A menudo este proceso se lleva a cabo a baja temperatura, para
compensar el exceso de temperatura que genera el rotor a tales revoluciones.
71
Filtración por membranas
En este tamiz, los compuestos libres y las vesículas avanzan de forma diferente, ya
que el bajo peso molecular respecto a las vesículas hace que éstos pueden seguir
caminos más largos al ser capaces de penetrar por los poros de las partículas. Al
contrario, las vesículas avanzan por el especio muerto que dejan las partículas entre sí,
por lo que el orden de elución es: primero las vesículas y luego los compuestos libres
en orden de mayor a menor peso molecular (ver Figura 3.11.C).
Los tipos de fase estacionaria más empleados para la purificación de vesículas son
aquellos basados en partículas poliméricas de dextrano o agarosa entrecruzado que
generan mallas de tamaño de poro diferentes. Sephadex® G25, G50 o G75; 135,136 o
Sepharosa CL-2B o CL-4B 137,138 suelen ser los medios más empleados. En función del
peso molecular del compuesto encapsulado/conjugado se emplea uno u otro, siendo
CSephadex® empleado para moléculas de bajo/medio peso molecular y Sepharosa lo
indicado más macromoléculas tipo proteínas o polímeros. En algunos casos, este
método se emplea con fuerza centrífuga para una mayor rapidez y eficacia. 139
72
Tabla 3.3. Ventajas y desventajas de los métodos de purificación de vesículas.
73
Figura 3.12. Representación gráfica de la estructura y componentes de un exosomas natural. Figura
adaptada de la referencia 142.
74
3.4.1. Exosomas como modelo de inspiración de vesículas
mejoradas
Sin embargo, a pesar del desarrollo de estas metodologías, existen una serie de
limitaciones relativas a su uso a nivel clínico, y que incluyen su aislamiento y
purificación a escala de producción con un grado aceptable según estándares clínicos.
Por otro lado, estas desventajas impiden a día de hoy la existencia de un material de
referencia con el que poner a punto y comparar estrategias bioanalíticas que ayuden a
profundizar en su estudio y permitan su plena instauración a nivel clínico. 148
75
Figura 3.13. Ventajas (verde) y desventajas (rojo) de los dos tipos de aproximaciones metodológicas
basadas en bionanotecnología para la producción de exosomas puramente artificiales con fines
teranósticos.
76
Soluciones basadas en métodos Bottom-Up
Es por ello, un campo de trabajo muy interesante, donde la información que nos
aporta el estudio de los exosomas naturales puede ayudar en la creación de nuevos
liposomas mejorados, pero a su vez, éstos nos pueden permitir en el diseño y
desarrollo de nuevos biomateriales complejos que puedan ser empleados en clínica y
bionálisis.
[1] Chen I.A., Walde P. (2010). From self-assembled vesicles to protocells. Cold Spring Harbor
Perspectives in Biology, 2:a002170.
[2] Janney P.A., Kinnunen P.K.J. (2006). Biophysical properties of lipids and dynamic
membranes. Trends in Cell Biology, 16(10):538-546.
[3] Wade L.G. Jr, editor. Organic Chemistry 5th edition. Pearson Prentice Hall, New Jersey,
USA.
[4] Tandford C. (1973). The hydrophobic effect: formation of micelles and biological
membranes. New York, John Wiley and Sons, Inc.
77
[5] Förster S., Zisenis M., Wenz E., Antonietti M. (1996). Micellization of strongly segregated
block copolymers. J. Chem. Phys., 104:9956.
[6] 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.
[7] Antonietti M., Förster S. (2003). Vesicles and liposomes: a self-assembly principle beyond
lipids. Advanced Materials, 15:1323-33.
[8] Lasic D.D. (1982b). A molecular model for vesicle formation. Biochimica et Biophysica Acta,
692:501-502.
[9] Lasic D.D. (1988). The mechanism of vesicle formation. Biochemical Journal, 256:1-11.
[10] Jahn A., Lucas F., Wepf R.A., Dittrich P.S. (2013). Freezing continuous-flow self-
assembly in a microfluidic device: toward imaging of liposome formation. Langmuir, 29:1717-
1723.
[11] Wang Z., He X. (2009). Dynamic of vesicle formation from lipid droplets: mechanism
and controllability. The journal of chemical physics, 130(9):094905.
[12] Kasson P.M., Kelley N.W., Singhal N., Vrljic M., Brunger A.T., Pande V.S. (2006).
Ensemble molecular dynamics yields submillisecond kinetics and intermediates of membrane
fusion. Proceedings of the National Academy of Sciences of the United States of America, 103:11916.
[13] Weiss T.M., Narayanan T., Gradzielski M. (2008). Dynamics of spontaneous vesicle
formation in fluorocarbon and hydrocarbon surfactant mixtures. Langmuir, 24:3759-3766.
[14] Sreekumaria A., Lipowsky R. (2018) Lipids with bulky head groups generate large
membrane curvatures by small compositional asymmetries. Journal of Chemical Physics,
149:084901.
[15] Julicher F., Seifert U., Lipowsky R. (2013). Phase diagrams and shape transformations of
toroidal vesicles. Journal of Physics II, 3:1681-1705.
[16] Manosroi A., Wongtrakul P., Manosroi J., Sakai H., Sugawara F., Yuasa M., Abe M.
(2003). Characerization of vesicles prepared with various non-ionic surfactants mixed with
colesterol. Colloids and Surfaces B: Biointerfaces, 30:129-138.
78
[17] Horiuchi T., Tajima K. (2000). Supramolecular Structure of Functionality Vesicles:
Niosome Formation of Nonionic Amphiphiles and Physico-Chemical Properties in Aqueous
Dispersion. Journal of Japan Oil Chemists' Society 49:1107.
[18] Peltonen L., Hirvonen J., Yliruusi J. (2001). The effect of temperature on sorbitan
surfactant monolayers. Journal of Colloid and Interface Science, 239:134-138.
[19] Leng J., Egelhaaf S.U., Cates M.E. (2003). Kinetics of the Micelle-to-Vesicle Transition:
Aqueous Lecithin-Bile Salt Mixtures. Biophysical Journal, 85:1624-1646.
[20] Zook J.M., Vreeland W.N. (2010). Effects of temperature, acyl chain length, and flow-
rate ratio on liposome formation and size in a microfluidic hydrodynamic focusing device. Soft
Matter, 6:1352-1360.
[21] Hu J., Weikl T., Lipowsky R. (2011). Vesicles with multiple membrane domains. Soft
Matter, 7:6092-6102.
[22] Rai S., Pandey V., Rai G. (2017). Transfersomes as versatile and flexible nano-vesicular
carriers in skin cancer therapy: the state of the art. Nano Reviews & Experiments, 8:1325708.
[23] Bangham A.D., Standish M.M., Watkins J.C. (1965). Diffusion of univalent ions across
the lamellae of swollen phospholipids. J. Mol. Biol., 13:238-252.
[25] Pattni B.B., Chupin V.V., Torchilin V.P. (2015). New developments in liposomal drug
delivery. Chem Rev., 115:10938-10965.
[26] Hong Y.J., Kim J.C. (2011). Egg phosphatidylcholine liposomes incorporating
hydrophobically modified chitosan: pH-sensitive release. J. Nanosci. Nanotechnol., 11(1):204-9.
[27] Lu M., Zhao X., Xing H., Xun Z., Zhu S., Lang L., Yang T., Cai C., Wang D., Ding P.6.
(2018). Comparison of exosome-mimicking liposomes with conventional liposomes for
intracellular delivery of siRNA. Int. J. Pharm., 550(1-2):100-113.
[28] Rinia H.A., de Kruijff B. (2001). Imaging domains in model membranes with atomic
force microscopy. FEBS Letters, 504:194-199.
[29] Handjani-Vila R., Ribier A., Rondot B.A., Vanlerberghie G. (1979). Dispersions of
lamellar phases of non-ionic lipids in cosmetic products, Int. J. Cosmet. Sci., 1(5):303-314.
79
[30] Tavano L., Muzzalupo R., Mauro L., Pellegrino M., Andò S., Picc N. (2013). Transferrin-
conjugated Pluronic niosomes as a new drug delivery system for anticancer therapy.
Langmuir,29(41):12638-12646
[31] Manconi M., Vila A.O., Sinico C., Figueruelo J., Molina F., Fadda A.M. (2006).
Theoretical and experimental evaluation of decypolyglucoside vesicles as potential drug
delivery systems. Journal of Drug Delivery Science and Technology, 16:141-146.
[32] Planas M.E., Gonzalez P., Rodriguez L., Sanchez S., Cevc G. (1992). Noninvasive
percutaneous induction of topical analgesia by a new type of drug carrier, and prolongation of
local pain on sensitivity by anesthetic liposomes. Anesth Analg, 75:615-621.
[33] Caddeo C., Manca M.L., Peris J.E., Usach I., Diez-Sales O., Matos M., Fernàndez-
Busquets X., Fadda A.M., Manconi M. (2018). Tocopherol-loaded transfersomes: In vitro
antioxidant activity and efficacy in skin regeneration. Int. J. Pharma., 551:34-41.
[34] Chaudhary H., Kohli K., Kumar V. (2013). Nano-transfersomes as novel carrier for
transdermal delivery. International Journal of Pharmaceutics, 454:367-380.
[35] Saraf S., Paliwal S., Kaur C.D., Saraf S. (2011). Sphingosomes a novel approach to
vesicular drug delivery. Research Journal of Pharmacy and Technology, 4(5):661-666.
[36] Zhigaltsev I.V., Maurer N., Akhong Q.-F., Leone R., Leng E., Wang J., Semple S.C.,
Cullis P.R. (2005). Journal of Controlled Release, 104:103–111.
[39] Mittal R., Sharma A., Arora S. (2013). Ufasomes mediated cutaneous delivery of
dexamethasone: formulation and evaluation of anti-inflammatory activity by carrageenin-
induced rat paw edema model. Journal of Pharmaceutics, 2003: Article ID 680580.
80
[41] Dayana N., Touitou E. (2000). Carriers for skin delivery of trihexyphenidyl HCl:
ethosomes vs. liposomes. Biomaterials, 21(18):1879-1885.
[42] Ferrer-Tasies L., Moreno-Calvo E., Cano-Sarabia M., Aguilella-Arzo M., Angelova A.,
Lesieur S., Ricart S., Faraudo J., Ventosa N., Veciana J. (2013). Quatsomes: vesicles formed by
Self-Assembly of sterols and quaternary ammonium surfactants. Langmuir, 29:6519–6528.
[43] Tezel U., Pavlostathis S.G. (2015). Quaternary ammonium disinfectants: microbial
adaptation, degradation and ecology. Current Opinion in Biotechnology, 33:296-304.
[44] Taubert A., Napoli A., Meier W. (2004). Self-assembly of reactive amphiphilic block
copolymers as mimetics for biological membranes. Current Opinion in Chemical Biology, 8:598-
603.
[45] Meng F., Zhong Z. (2011). Polymersomes spanning from nano- to microscales:
advanced vehicles for controlled drug delivery and robust vesicles for virus and cell mimicking.
Journal of Physical Chemistry Letters. 2:1533–1539.
[46] Schlaad H., Antonietti M. (2003) Block copolymers with amino acid sequences:
molecular chimeras of polypeptides and synthetic polymers. Eur Phys J E, 10:17-23.
[47] Meier W., Nardin C., Winterhalter M. (2000). Reconstitution of channel proteins in
(polymerized) ABA triblock copolymer membranes. Angew Chem Int Ed Engl, 39:4599-4602.
[48] Napper D.H. (1970). Colloid stability. Ind. Eng. Chem. Prod. Res. Develop., 9(4): 467-
477.
[49] Huang Y., Chen J, Chen X, Gao J, Liang W. (2007). PEGylated synthetic surfactant
vesicles (niosomes): novel carriers for oligomucleotides. J. Mater. Sci. Mater. Med., 19:607-614.
[50] Awasthi V.D., Garcia D., Klipper R., Goins B.A., Phillips W.T. (2004). Neutral and
anionic liposome-encapsulated hemoglobin: effect of postinserted poly(ethylene glycol)-
distearoylphosphatidylethanolamine on distribution and circulation kinetics. Journal of
Pharmacology Experimental and Therapy. 309:241–248.
[51] Bizmark N., Ioannidis M.A. (2015). Effects of Ionic Strength on the Colloidal Stability
and Interfacial Assembly of Hydrophobic Ethyl Cellulose Nanoparticles. Langmuir, 31(34):9282-
9289.
81
[53] Pando D., Matos M., Gutiérrez G., Pazos C. (2015). Formulation of resveratrol
entrapped niosomes for topical use. Colloids Surf. B, 128:398-404.
[54] Iwasakia Y., Tanaka S., Hara M., Ishihara K., Nakabayashia N. (1997). Stabilization of
liposomes attached to polymer surfaces having phosphorylcholine groups. J. Colloid. Interface
Sci., 192(2): 432-439.
[55] Jung. Y.K, Kim T.W., Park H.G., Soh H.T. (2010). Specific Colorimetric Detection of
Proteins Using Bidentate Aptamer‐Conjugated Polydiacetylene (PDA) Liposomes. Adv. Funct.
Mater., 20(18):3092-3097.
[56] Shmeeda H., Liia Mak L., Tzemach D., Astrahan P., Tarshish M., Gabizon A. (2006).
Intracellular uptake and intracavitary targeting of folate-conjugated liposomes in a mouse
lymphoma model with up-regulated folate receptors. Molecular Cancer Therapeutics, 5(4):818-824.
[57] Li K., Chang S., Wang Z., Zhao X., Chen D. (2015). A novel micro-emulsion and micelle
assembling method to prepare DEC205 monoclonal antibody coupled cationic nanoliposomes
for simulating exosomas to target dendritic cells. Int. J. Pharm., 491:105-112.
[58] Lukyanov A.N., Elbayoumi T.A., Chakilam A.R., Torchilin V.P. (2004). Tumor-targeted
liposomes: doxorubicin-loaded long-circulating liposomes modified with anti-cancer antibody.
J. Control Release, 100(1):135-144.
[59] Patel D.M., Jani R.H., Patel C.N. (2011). Ufasomes: A Vesicular Drug Delivery.
Systematic Reviews in Pharmacy, 2(2):72-78.
[60] Mukundan S. Jr., Ghaghada K.B., Badea C.T., Kao C.-Y., Hedlund L.W., Provenzale
J.M., Allan Johnson G.A., Chen E., Bellamkonda R.V., Annapragada A. (2006). A liposomal
nanoscale contrast agent for preclinical CT in Mice. American Journal of Roentgenology, 186: 300-
307.
[61] Lee J., Lee H., Goh U., Kim J., Jeong M., Lee J., Park J.-H.. (2016). Cellular engineering
with membrane fusogenic liposomes to produce functionalized extracellular vesicles. ACS
Applied Materials and Interfaces, 8(11):6790-6795.
[62] Sato Y.T., Umezaki K., Sawada S., Mukai S., Sasaki Y., Harada N., Shiku H., Akiyoshi
K. (2016). Engineering hybrid exosomes by membrane fusion with liposomes. Sci. Rep., 6:21933.
[63] Kakar R., Rao R., Goswami A., Nanda S., Saroha K. (2010). Proniosomes: An Emerging
Vesicular System in Drug Delivery and Cosmetics. Der Pharmacia Lettre, 2(4): 227-239.
82
[64] N. Weiner, L. Lieb, S. Niemiec, C. Ramachandran, Z. Hu , K. Egbaria. (1994).
Liposomes: A Novel Topical Delivery System for Pharmaceutical and Cosmetic Applications.
Journal of Drug Targeting, 2(5):405-410.
[65] Rezvani M., Hesari J., Peighambardoust S.H., Manconi M., Hamishehka H., Escribano-
Ferrer E. (2019). Potential application of nanovesicles (niosomes and liposomes) for fortification
of functional beverages with Isoleucine-Proline-Proline: A comparative study with central
composite design approach. Food Chemistry, 293:368-377.
[66] Gatt S., Bercovier H., Barenholz Y. Use of liposomes for combating oil spills and their
potential application to bioreclamation. In: On-site bioreclamation: processes for xenobiotic and
hydrocaron treatment, editado por Hinchee R.E. and Olfenbuttel R.F. 1991, Battelle Press, USA.
[68] Liu Q., Boyd B.J. (2013) Liposomes in biosensors. Analyst, 138:391-409.
[69] Shweta Pawar S., Bhattacharya A., Nag A. (2019) Metal-Enhanced Fluorescence Study
in Aqueous Medium by Coupling Gold Nanoparticles and Fluorophores Using a Bilayer Vesicle
Platform. ACS Omega, 4(3):5983-5990.
[70] Edwards K.A., Korff R., Baeumner A.J. (2017). Liposome-Enhanced Lateral-Flow
Assays for Clinical Analyses. Methods in Molecular Biology.1571:407-434
[71] Ho J.A., Wu L.-C. , Huang M.R., Lin Y.-J., Baeumner A.J., Durst R.A. (2007) Application
of Ganglioside-Sensitized Liposomes in a Flow Injection Immunoanalytical System for the
Determination of Cholera Toxin. Analytical Chemistry, 79:246-250.
[72] Chapman R., Lin Y., Burnapp M., Bentham A., Hillier D., Zabron A., Khan S., Tyreman
M., Stevens M.M. (2015). Multivalent nanoparticles networks enable point-of-care detection of
human phospholipase-A2 in serum. ACS Nano, 9:256-73.
[73] Bilek G., Kremser L., Blaas D., Kenndler E. (2006). Analysis of liposomes by capillary
electrophoresis and their use as carrier in electrokinetic chromatography. Journal of
Chromatography B, 841(1-2):38-51.
[74] Cipolla D., Wu H., Salentinig S., Boyd B., Rades T., Vanhecke D., Petri-Fink A.,
Rothin-Rutishauser B., Eastman S., Redelmeier T., Gondab I., Chana H.K.. (2016). Formation of
drug nanocrystals under nanoconfinement afforded by liposomes. RSC Advances, 8:6223-6233.
83
[75] Miller J.P., Leo G. Herbette L.G., White R.E. (1996). X-ray Diffraction Analysis of
Cytochrome P450 2B4 Reconstituted into Liposomes. Biochemistry, 35(5):1466-74.
[76] Biswas A., Sarkar S., Bhadra K., De S. (2015). Novel synthesis of biologically active CdS
nanoclusters in cell-mimicking vesicles. Journal of Experimental Nanoscience, 11(9):681-694.
[77] De S., Kundu R., Biswas A. (2012). Synthesis of gold nanoparticles in niosomes. Journal
of Colloids and Interface Science, 386:9-15.
[78] Olsona F., Hunt C.A., Szoka F.C., Vail W.J., Papahadjopoulos D. (1979). Preparation of
liposomes of defined size distribution by extrusion through polycarbonate membranes. Biochim.
Biophys. Acta - Biomembranes, 557:9-23.
[79] Yamaguchi T., Nomura M., Matsuoka T., Koda S. (2009). Effects of frequency and
power of ultrasound on the size reduction of liposome. Chemistry and Physics of lipids, 160:58-62.
[80] Grimaldi N., Andrade F., Segovia N., Ferrer-Tasies L., Sala S., Veciana J., Ventosa N.
(2016). Lipid-based nanovesicles for nanomedicine. Chem. Soc. Rev., 45(23):6520-6545.
[81] Capretto L., Carugo D., Mazzitelli S., Nastruzzi C., and Zhang X. (2013) Microfluidic
and lab-on-a-chip preparation routes for organic nanoparticles and vesicular systems for
nanomedicine applications. Advanced Drug Delivery Reviews, 65:1496-1532
[82] Maestrelli F., González-Rodríguez M.L., Rabasco A.M. Mura P. (2006). Effect of
preparation technique on the properties of liposomes encapsulating ketoprofen-cyclodextrin
complexes aimed for transdermal delivery. Int. J. Pharm., 312(1-2):53-60.
[83] Kremer J.M.H., Esker M.W.J., Pathmamanoharan C., Wiersema P.H. (1977). Vesicles of
variable diameter prepared by a modified injection method. Biochem., 16:3932-3935.
[84] Schieren H., Rudolph S., Finkelstein M., Coleman P., Weissmann G. (1978). Biochim.
Biophys. Acta, 542:137–53.
[85] Abdelkader H., Ismail S., Kamal A., Alany R.G. (2011). Design and evaluation of
controlled-release niosomes and discosomes for naltrexone hydrochloride ocular delivery. J.
Pharm Sci., 100(5):1833-1846.
[86] Szoka F., Papahadjopoulos D. (1978). Procedure for preparation of liposomes with large
internal aqueous space and high capture by reverse-phase evaporation. Proc. Natl. Acad. Sci.
USA, 75:4194–8.
84
[87] Alpes H., Allmann K., Plattner H., Reichert J., Rick R., Schulz S. (1986). Formation of
large unilamellar vesicles using alkyl maltoside detergents. Biochim. Biophys. Acta - Biomembr.,
862:294–302.
[88] Alam M., Zubair S., Farazuddin M., Malik A., Mohammad O. (2013). Development,
characterization and efficacy of niosomal diallyl disulfide in treatment of disseminated murine
candidiasis. Nanomedicine, 9:247-256.
[89] Wallach D.F.H. (1993). In: Liposome Technology. Edited by Gregoriadis G, vol. 2. CRC
Press.
[90] Mozafari S.M., Reed C.J., Rostrom C., Kocum C., Piskin E. (2005). Construction of stable
anionic liposome-plasmid particles using the heating method. A preliminary investigation. Cell.
Mol. Bio. Lett., 7:923-927.
[91] Verma S., Singh S.K., Syan N., Mathur P., Valecha V. (2010). Nanoparticle vesicular
systems: a versatile tool for drug delivery. J. Chem. Pharm. Res., 2(2):496-509.
[92] Duncan R., Florence A.T., Uchegbu I.F., Cociacinch F. (1997). Drug polymer conjugates
encapsulated within niosomes. Patente internacional de aplicación, ref. GB97:00072.
[93] 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. Int. J. Pharm., 361:101-111.
[94] Espirito Santo I., Campardelli R., Cabral Albuquerque E., Vieira de Melo S., Della Porta
G., Reverchon E. (2014). Liposomes preparation using a supercritical fluid assisted continuous
process. Chemical Engineering Journal, 249:153-159.
[95] Kunastitchai S., Pichert L., Sarisuta N., Müller B.W. (2006). Application of aerosol
solvent extraction system (ASES) process for preparation of liposomes in a dry and
reconstitutable form. Int. J. Pharm., 316(1-2):93-101.
[96] Manosroi A., Chutoprapat R., Abe M., Manosroi J. (2008). Characteristics of niosomes
prepared by supercritical carbon dioxide (scCO2) fluid. Int. J. Pharm., 352(1–2):248-255
[97] Trucillo P., Campardelli R., Reverchon E. (2017). Supercritical CO2 assisted liposomes
formation: Optimization of the lipidic layer for an efficient hydrophilic drug loading. Journal of
CO2 Utilization, 18:181-188.
85
[98] Lorenzo Capretto L., Carugo D., Mazzitelli S., Nastruzzi C., Zhang X. (20139.
Microfluidic and lab-on-a-chip preparation routes for organic nanoparticles and vesicular
systems for nanomedicine applications. Adv. Drug Deliv. Rev., 65:1496–1532.
[99] Carugo D., Bottaro E., Owen J., Stride E., and Nastruzzi C. (2016). Liposome production
by microfluidics: potential and limiting factors. Scientific Reports, 19(6):25876.
[100] van Swaay D. and deMello A. (2013). Microfluidic methods for forming liposomes, Lab
Chip, 13:752-767.
[101] Angelova M.I., Dimitrov D.S. (1986). Liposome electroformation. Faraday Discuss.
Chem. Soc., 81:303–311.
[102] Kuribayashi K., Tresset G., Coquet P., Fujita H., Takeuchi S. (2006). Electroformation
of giant liposomes in microfluidic channels. Meas. Sci. Technol., 17:3121.
[103] Le Berre M., Yamada A., Reck L., Chen Y., Baigl D. (2008). Electroformation of giant
phospholipid vesicles on a silicon substrate: advantages of controllable surface properties.
Langmuir, 24:2643–2649
[104] Y.C. Lin,M. Li, Y.T. Wang, T.H. Lai, J.T. Chiang, K.S. Huang, A new method for the
preparation of self-assembled phospholipid microtubes using microfluidic technology, in:
TRANSDUCERS'05 (Ed.), The 13th International Conference on Solid-State Sensors, Actuators
and Microsystems, IEEE, 2005, pp. 1592-1595.
[105] Dittrich P.S., Renaud P., Manz A. (2006). On-chip extrusion of lipid vesicles and tubes
through microsized apertures, Lab Chip, 6:488–493.
[106] Jahn A., Reiner J.E., Vreeland W.N., DeVoe D.L., Locascio L.E., Gaitan M. (2008).
Preparation of nanoparticles by continuous-flow microfluidics, J. Nanoparticle Res., 10:925–934.
[107] Phapal S., Sunthar P. (2013). Influence of micro-mixing on the size of liposomes self-
assembled from miscible liquid phases. Chem. Phys. Lipids, 172–173:20–30.
[108] Kastner E., Verma V., Lowry D., and Perrie Y. (2015). Microfluidic-controlled
manufacture of liposomes for the solubilisation of a poorly water soluble drug. Int. J. Pharm.,
485:122-130.
[109] Seo M., Paquet C., Nie Z., Xu S., Kumacheva E. (2007). Microfluidic consecutive flow-
focusing droplet generators. Soft Matter, 3:986–992.
86
[110] Funakoshi K., Suzuki H., Takeuchi S. (2007). Formation of giant lipid vesicle like
compartments from a planar lipid membrane by a pulsed jet flow, J. Am. Chem. Soc., 129:12608–
12609.
[111] Utada A., Lorenceau E., Link D., Kaplan P., Stone H., Weitz D. (2005). Monodisperse
double emulsions generated from a microcapillary device. Science, 308:537–541
[112] Matos M., Gutiérrez G., Iglesias O., Coca J., Pazos C. (2015). Enhancing encapsulation
efficiency of food-grade double emulsions containing resveratrol or vitamin B12 by membrane
emulsification. J. Food Eng., 166:212-220.
[113] Akbarzadeh A., Rogaie Rezaei-Sadabady R., Davaran S., Woo Joo S., Zarghami N.,
Hanifehpour Y., Samiei M., Kouhi M., Nejati-Koshki K. (2013). Liposome: classification,
preparation, and applications. Nanoscale Research Letters, 8:102.
[115] Damera D.P., Krishna Venuganti V.V., Nag A. (2018). Deciphering the role of bilayer
of a noisome towards controlling the entrapment and release of dyes. ChemistrySelect, 3:3930-
3938.
[116] Ballie A.J., Coombs G.H. (1988). Vesicular systems (Niosome & Liposome) for delivery
of Sodium Stibogluconate in experimental murine visceral leishmaniasis. J. Pharma. Pharmacol.,
40:161-165.
[117] Jeet K., Raines R.T. (2010). Advances in Bioconjugation. Current Organic Chemistry,
14(2):138-147.
[118] Marqués-Gallego P., de Kroon A.I.P.M. (2014). Ligation Strategies for Targeting
Liposomal Nanocarriers. BioMed Research International, 2014: Article ID 129458.
[119] Zhang Yun D.W., Xiang Y., Zhang J.Z.H. (2003). New advance in chemistry: full
quantum mechanical ab initio computation of Streptavidin−Biotin interaction energy. J. Phys.
Chem. B, 107(44):12039-12041.
87
[121] Papadia K., Markoutsa E., Antimisiaris S.G. (2014). A simplified method to attach
antibodies on liposomes by biotin-streptavidin affinity for rapid and economical screening of
targeted liposomes. J. Biomed. Nanotechnol., 10(5):871-6.
[122] Lozano-Andrés, E., Libregts, S. F., Toribio, V., Royo, F., Morales, S., López-Martín, S.,
Valés-Gómez, M., Reyburn, H.T., Falcón-Pérez, J.M., Wauben, M.H., Soto, M., Yáñez-Mó, M.
(2019). Tetraspanin-decorated extracellular vesicle-mimetics as a novel adaptable reference
material. Journal of Extracellular Vesicles, 8(1), 1573052.
[123] Bioconjugate reagents, Chapter 3. En: Bioconjugate techniques, Hermanson G.T. (editor).
Academic Press, Londres, 1-125, 127-228.
[124] Endoh H., Suzuki Y., Hashimoto Y. (1981). Antibody coating of liposomes with 1-
ethyl-3-(3-dimethyl-aminopropyl)carbodiimide and the effect on target specificity. J. Immunol.
Methods, 44:79-85.
[125] Li K., Chang S., Wang Z., Zhao X., Chen D. (2015). A novel micro-emulsion and
micelle assembling method to prepare DEC205 monoclonal antibody coupled cationic
nanoliposomes for simulating exosomas to target dendritic cells. Int. J. Pharm., 491:105-112.
[126] NHS and SULFO-NHS Application note, Thermo Scientific. Disponible en la web.
[127] Gutiérrez G., Matos M., Barrero P., Pando D., Iglesias O., Pazos C. (2016). Iron-
entrapped niosomes and their potential application for yogurt fortification. Food Science and
Technology, 74:550-556.
[128] Pando D., Gutiérrez G., Coca J., Pazos C. (2013). Preparation and characterization of
niosomes containing resveratrol. Journal of Food Engineering, 117(2):227-234.
[129] Pando D., Caddeo C., Manconi M., Fadda A.M., Pazos C. (2013). Nanodesign of olein
vesicles for the topical delivery of the antioxidant resveratrol. J. Pharm. Pharmacol., 65(8):1158-
1167.
[130] Jain S., Chaudhari B.H., Swarnakar N.K. (2011). Preparation and characterization of
niosomal gel for iontophoresis mediated transdermal delivery of isosorbide dinitrate. Drug
Deliv. Transl. Res., 1:309-321.
[131] Zubairu Y., Mohan L., Ngi Zeenat Iqbal N., Talegaonkar S. (2015). Design and
development of novel bioadhesive niosomal formulation for the transcorneal delivery of anti-
infective agent: In-vitro and ex-vivo investigations. Asian Journal of Pharmaceutical Sciences,
10(4):322-330
88
[132] Sánchez-López V., Fernández-Romero J.M., Gómez-Hens A. (2009). Evaluation of
liposome populations using a sucrose density gradient centrifugation approach coupled to a
continuous flow system. Anal. Chim. Acta, 645(1-2):79-85.
[133] Hirai M., Minematsu H., Hiramatsu Y., Kitagawa H., Otani T., Iwashita S., Kudoh T.,
Chen L., Li Y., Okada M., Salomon D.S., Igarashi K., Chikuma M., Seno M. (2010). Novel and
simple loading procedure of cisplatin into liposomes and targeting tumor endothelial cells. Int.
J. Pharm., 391(1-2):274-283.
[135] Ruysschaert T., Audrey Marque A., Duteyrat J.-L., Lesieur S., Winterhalter M.,
Fournier D. (2005). Liposome retention in size exclusion chromatography. BMC Biotechnol., 5:11.
[136] Ehnholm C., Zilversmit D.B. (1973). Exchange of various phospholipids and of
cholesterol between liposomes in the presence of highly purified phospholipid exchange
protein. J. Biol. Chem., 248:1719-1724.
[137] Chen C.S., Yao J., Durst R.A. (2006). Liposome encapsulation of fluorescent
nanoparticles: Quantum dots and silica nanoparticles. Journal of Nanoparticle Research, 8(6):1033-
1038.
[138] Yang X., Zhao X., Phelps M.A., Piao L., Rozewski D.M., Liu Q., Lee L.J., Marcucci G.,
Grever M.R., Byrd J.C., Dalton J.T., Lee R.J. (2009). A novel liposomal formulation of
flavopiridol. Int. J. Pharm., 365(1-2):170-174.
[139] Gupta P.N., Mishra V., Rawat A., Dubey P., Mahor S., Jain S., S., Chatterji D.P., Vyas
S.P. (2005b). Non-invasive vaccine delivery in tranfersomes, niosomes and liposomes: a
comparative study. Int. J. Pharm., 293:73-82.
89
Madrid F., Santarém N. Schallmoser K., Ostenfeld M.S., Stoorvogel W., Stukelj R., Van der
Grein S.G., Vasconcelos M.H., Wauben M.H., De Wever O. (2015). Biological properties of
extracellular vesicles and their physiological functions. Journal of Extracellular Vesicles,
14(4):27066.
[142] https://www.novusbio.com/research-areas/cell-biology. Último acceso 15/08/2019.
[143] Colombo M., Moita C. van Niel G., Kowal J., Vigneron J., Benaroch P., Manel N., Moita
L.F., Théry C., Raposo G. (2013). Analysis of ESCRT functions in exosome biogenesis,
composition and secretion highlights the heterogeneity of extracellular vesicles. J. Cell. Sci.,
126(24):5553-65.
[144] Kowal J., Arras G., Colombo M., Jouve M., Morath J.P., Primdal-Bengtson B., Dingli F.,
Loew D., Tkach M., Théry C. (2016). Proteomic comparison defines novel markers to
characterize heterogeneous populations of extracellular vesicle subtypes. Proc. Natl. Acad. Sci. U
S A., 113(8):E968-77.
[145] Fuhrmann G., Herrmann I.K., Stevens M.M. (2015). Cell-derived vesicles for drug
therapy and diagnosis: opportunities and challenges. Nano Today, 10:397-409.
[146] van der Meel R., Fens M.H.A.M., Vader P., van Solinge W.W., Eniola-Adefeso O.,
Schiffelers R.M. (2014). Extracellular vesicles as drug delivery systems: lessons from the
liposome field. J. Control. Release., 195:72-85.
[147] Johnsen K.B., Gudbergsson J.M., Skov M.N., Pilgaard L., Moos T., Duroux M. (2014). A
comprehensive overview of exosomas as drug delivery vehicles – endogenous nanocarriers for
targeted cancer therapy. Bichim. Biophys. Acta, 1846:75-87.
[148] Valkonen S., van der Pol E., Böing A., Yuana Y., Yliperttula M., R. Nieuwland,
Laitinen S., Siljander P.R.M. (2017). Biological reference materials for extracellular vesicle
studies. Eur. J. Pharm. Sci., 98:4-16.
[149] Jang S.C., Kim O.Y., Yoon C.M., Choi D.S., Roh T.Y., Park J., Nilsson J., Lötvall J., Kim
Y.K., Gho Y.S. (2013). Bioinspired exosome-mimetic nanovesicles for targeted delivery of
chemotherapeutics to malignant tumors. ACS nano, 7:7698-7710.
[150] Hwang D.W., Hongyoon Choi H., Jang S.C., Yoo M.Y., Park J.Y., Choi N.E., Oh H.J.,
Ha S., Yun-Sang Lee, Y.-S., Jeong J.M., Gho Y.S., Lee D.S. (2015). Noninvasive imaging of radio-
labelled exosome-mimetic nanovesicles using 99mTc-HMPAO. Sci Rep., 5:15636.
90
[151] Jo W., Jeong D., Kim J., Cho S., Jang S.C., Han C., Kang J.Y., Gho Y.S., Park J. (2014).
Microfluidic fabrication of cell-derived nanovesicles as endogenous RNA carriers. Lab Chip,
14:1261-1269.
[152] Yoon J., Jo W., Jeong D., Kim J., Jeong H., Park J. (2015). Generation of nanovesicles
with sliced cellular membrane fragments for exogenous material delivery. Biomaterials, 59:12-20.
[154] Shim G., Kim M.-G., Park J.Y., Oh Y.-K. (2013). Application of cationic liposomes for
delivery of nucleic acids. Asian Journal of Pharmaceutical Sciences, 8(2):72-80.
91
4. Experimental
Capítulo 4. Experimental
Para una mejor comprensión de los resultados, tanto los materiales utilizados como
los reactivos y los procedimientos empleados se van a detallar en cada uno de los
capítulos experimentales de manera individual. De esta manera, el apartado
experimental de la Tesis Doctoral recoge de manera general los principios de las
técnicas y los detalles relacionados con la instrumentación empleada en la
caracterización de las nanovesículas producidas a lo largo de la misma.
Emplea un haz de luz láser que se dirige hacia la suspensión de partículas, siendo
la luz reflejada por las mismas en múltiples direcciones. Este reflejo no posee ni una
intensidad ni una direccionalidad fija, si no que debido al movimiento browniano de
las partículas (por choques con moléculas del disolvente), éste fluctúa en el tiempo en
proporción al tamaño de las partículas. Partículas pequeñas se mueven más rápido,
mientras que partículas grandes se mueven despacio, por lo que se observan
fluctuaciones que se producen de acuerdo a ese movimiento.
95
Figura 4.1. Equipo Zetasizer NANO-ZS, Malvern Instruments.
96
4.2. Microscopía Electrónica de Trasmisión (TEM): morfología
Cuando los electrones colisionan con la muestra, en función de su grosor y del tipo
de átomos que la forman, parte de ellos son dispersados selectivamente, es decir, hay
una gradación entre los electrones que la atraviesan directamente y los que son
totalmente desviados. Todos ellos son conducidos y modulados por unas lentes para
formar una imagen final sobre una cámara CCD que puede tener miles de aumentos
con una definición inalcanzable para cualquier otro instrumento. La información que
se obtiene es una imagen con distintas intensidades de gris que se corresponden al
grado de dispersión de los electrones incidentes.
97
En el caso particular de la presente Tesis Doctoral, el microscopio utilizado (ver
Figura 4.2) está especialmente indicado para la nano-caracterización estructural y
analítica de materiales. Posee 200 kV de aceleración lo que permite alcanzar una
resolución de 3,4 Å entre líneas. El equipo dispone además de una cámara CCD de alta
resolución (2048x2048 pixels). Se ha empleado ácido fosfotungstico (2% w/v, en agua
ultrapura).
Figura 4.3. Equipo Turbiscan LAB Expert con estación de envejecimiento, Formulaction.
98
éstos se verán reflejos en un cambio de la señal detectada por ambos detectores y por
consiguiente un cambio en ese patrón a lo largo del tiempo de medida.
99
analíticos cuyas espefificaciones se encuentran detalladas en las secciones
experimentales de cada capítulo donde se ha empleado esta técnica.
Figura 4.4. Equipo de HPLC HP serie 1100 con módulo de detección UV/Vis HP G1315A UV/ 281,
Hewlett-Packard.
Un lector de placas (ver Figura 4.5) es un instrumento que permite detectar eventos
biológicos, químicos o físicos en muestras contenidas en placas de microtitración. Los
métodos comunes de detección para los ensayos de microplacas son: absorbancia,
intensidad de fluorescencia, luminiscencia, fluorescencia resuelta en el tiempo y
polarización de fluorescencia.
Figura 4.5. Lector de placas y absorbancia TECAN Genios, Tecan Trading AG.
100
4.6. Analizador para la medida de Western Blot
Figura 4.6. Analizador de imágenes de luminiscencia para geles y membranas LAS4000 mini Image
System analyser de, GE Healthcare.
101
particular de los sistemas vesiculares, el cambio de estado gel a líquido cristalino de las
bicapas se manifiesta como un pico endotérmico, ya que dicho cambio se produce
gracias a energía en forma de calor. Su posición y forma de pico ofrece información
acerca de la estabilidad y grado de empaquetamiento de los compuestos que forman la
bicapa, y resulta en una técnica muy útil para la caracterización de formulaciones.
102
5. Therapeutic biomaterials based on
extracellular vesicles: classification of bio-
engineering and mimetic preparation routes
Capítulo/Chapter 5
Therapeutic biomaterials based on
extracellular vesicles: classification of bio-
engineering and mimetic preparation routes
Both the structure of the membrane and the formation route are the origin of the
following advantageous aspects: (1) high selective targetability and minimum off-
target effect, thanks to a set of molecules involved in targeting, signalling and receptor-
mediated uptake, complete with all the co-receptors needed for the internalisation
process; (2) capacity of extravasation due to a gel-state core derived from the presence
of hydrated macromolecules (proteins and nucleic acids) combined with a minimum
cytoskeleton that allows deformability while keeping the whole integrity of the vesicle;
(3) Size distribution; (4) great stability in the blood due to the evasion of the innate
immune system; (5) adaptative responses that cause clearance from the blood, with the
corresponding decrease in bioavailability.
105
as vehicles to reach specific target population, sometimes highly protected from
conventional administration routes.
EVs have been modified in the search for broader therapeutic capability.
Sometimes, this included the incorporation of new elements for targeted puporses, in
vitro or in vivo traceability, or the material to be delivered. In other cases, modification
was aimed at the enhancemt of colloidal stability, or change in surface charge to
increase their uptake rate. These new approaches have generate new terms such as
bioengineered exosomes, artificial exosomes, 15 exosome-mimetic nanovesicles, 16
exosome-like nanovesicles, 17,18 and exosome-based semi-synthetic vesicles. 19 These
expressions have been used with different meanings in the literature, but to date, there
has not been yet a clear criterion for their classification. One example is the term
“exosome-like nanovesicles”. In some works, this concept is used to name artificial
vesicles made from cells through different techniques to mimic exosomes.17-18
However, cell-derived vesicles with morphological and biochemical characteristics
similar to exosomes were also named exosome-like nanovesicles by other authors.19-20
Other authors working with non-animal research models used this term to refer to
vesicles with size and flotation density values similar to exosomes. For example,
Regente et al. 21 described the presence of exosome-like vesicles in sunflower plant
fluids, and Prado et al. 22 showed evidence of vesicles quite similar to exosomes during
pollen germination.
106
In order to provide a systematic classification to move around in this new emerging
field, we suggest the nomenclature given in Figure 5.1. This Artificial EVs landscape is
based on the concept behind the term. This way, “artificial EVs” will be used as general
concept to designate all vesicles, modified or manufactured (from natural or synthetic
sources) with the aim to mimic EVs (mainly exosomes) for therapeutic uses. Behind
this term, two categories of artificial EVs could be discerned: “semi-synthetic EVs” and
“fully synthetic” or “EVs mimetic vesicles”, corresponding to modified natural EVs (pre-
or post-isolation) and artificial structures, lab-made or generated from cultured cells.
Type of artificial EVs
ARTIFICIAL EVs
+ + +
Modification pre- or post EVs isolation From cultured cells From individual molecules
Express new target Enhance bioavailabity Exogenous cargo Increasing release Preparation of a well
and functional of poor soluble drug, delivery to cytosol in yields, while keeping known product, with
Purposes
molecules, or make without off-target target cells constant ability to only desirable elements
EVs traceables effects. New physical incorpórate cargo
and chemical
properties
Figure 5.1. Artificial EVs landscape: explored routes up-to-date for the preparation of artificial EVs for
specific purposes.*EBSSNs (ref. 19)
The former generate semi-synthetic products, as they start from a natural substrate,
which can be subsequently modified before or after their isolation. The modification
affects the structures of the outer surface of the vesicles, the membrane or the cargo
that travels within, and could also include hydrophobic molecules at the membrane.
The term fully synthetic, on the other hand, stresses the artificiality of the product.
We have recently briefly commented their potential in therapeutics. 23 These techniques
can be classified on the basis of their manufacturing route: those starting from larger
substrates (cells) that are reduced to units for the creation of small size vesicles (top-
down nanotechnology) or those taking individual molecules (lipids, proteins, etc.) that
self-assemble in higher order structures with tunable composition (bottom-up
107
nanotechnology). Top-down products differ from natural EVs in terms of micro-
structure and biochemical composition, since they are formed from cell fragments: the
characteristical membrane microdomains 24 (lipid rafts) and associated pools of surface
markers (especially tetraspanins) are absent, and the minimal cytoskeleton is not
present.
The preparation route chosen is important for the final purpose of the artificial EVs,
but it could be critical for the clinical trials and subsequent commercialization. The
extense manipulation of EVs during the bio-engineering methods is the reason for their
classification as advanced therapy medicinal products (ATMPs),2 with a particular
regulatory framewok. Following the same criteria, fully artificial EVs produced from
cultured cells (top-down) should be also incorporated into this category.
On the other hand, botton up artificial mimetic EVs are more difficult to assign to
one or another category. To date, a set of proteins has been fixed on lipidic vesicles
with undefined purpose. While there is some evidence that the EVs membrane is
important for the uptake process,1 the role of the artificial membranes is not clear yet.
Comparative studies of the effects over target cell lines with conventional liposomes
and exosome-mimetic nanovesicles would be very useful to clarify this. The work in
this field is reviewed in the section “Bottom-up methodologies: artificial membranes
decorated with functional proteins to mimic EVs functions.”
A critical evaluation of the fully synthetic EVs concept would imply providing an
answer to questions such as: What we are trying to mimic from EVs?; Is the biochemical
composition, the morphology or the whole entity?; Is it worth mimicking a specific function?.
This is still a challenge in the field, since some of these questions are being answered at
the same time for natural EVs. The best approach would involve an extensive
biochemical characterization of natural EVs and a detailed description of their
functions. Regarding functionality, other populations and not only the target cell lines
should be assayed. This could give information about possible side effects. In the same
way, multiple parameters should be registered as output factors, not just the process
targetted by the EVs-based treatment. Proteins, nucleic acids, and lipid composition
from specific types of exosomes are registered in specialized databases such as
108
ExoCarta, 25 EVpedia 26 and Vesiclepedia. 27 But this is not enough: a database with
assays performed with EVs reporting treated cell lines, EVs type as therapeutical
agents, tipe of assay (in vitro or in vivo) and experimental conditions could be of great
interest to the scientific community. The combination of both sources of information
would be the perfect scenario for the design of future artificial EVs-based therapies.
In any case, both types of artificial EVs should meet product specifications related
to “purity, identity, quantity, potency and sterility” in concordance with the
pharmaceutical market regulations. 28 Once more, there are some important differences
in the definition of these parameters depending on the semi- or fully synthetic
character of EVs. These key points will be considered and discussed in the following
sections.
The simplest idea to manufacture EVs would be to use the natural mechanisms for
the formation of vesicles, that is, the cellular machinery itself. It is known that the
composition of the EVs at all levels responds to a high degree of control at very
selective cellular mechanisms. 29 Therefore, the composition of the EVs could be
controlled by intentional alteration of the cellular environment.
This method would lead to the creation of EVs with a composition profile adapted
to a specific purpose. The technological methods used for bioengineering EVs are
explained in the following sections. Two key aspects are the selection of producer cells
(and their in vitro harvesting conditions) and the EVs isolation/enrichment procedures.
Both choices will condition subsequent uses.
Cell lines could potentially release EVs vesicles, but there are great differences in
release rate and biochemical composition and their susceptibility to modifications. 30 It
is also accepted that before translation to clinical use of EVs, limitations regarding
biocompatibility, economic viability, harvesting methods, and immunotolerance must
be overcome. A summary of cell lines used for the production of EVs for clinical
purposes, especially drug delivery, can be found in the literature. 31 Dendritic cells and
cancer cell lines, such as melanoma, are the most used lines for EVs production.
109
Mesenchymal stem cells (MSCs) are one of the most promising sources of EVs,
especially exosomes. 32 Yeo et al. 33 defended their use for mass production of exosomes
with future therapeutic purposes based on some important facts related to their
advantages over other cell lines. Mainly, MSCs are easy to obtain from all human
tissues (even those considered as medical waste), and they have great ex vivo expansion
capacity compatible with immortalization without compromising their therapeutic
efficacy. These two facts are essential to establish a scalable and long-term source of
well-characterised EVs, particularly exosomes.14,28 In addition, their
immunomodulatory effect gives them and their derived EVs important features in
autologous and allogenic therapeutic applications.
Dendritic Cells (DCs) have important roles in immunity (both innate and
adaptative). Some authors have paid attention to this cell line in order to enhance the
production of clinical grade DC-derived exosomes for immunotherapy.33 Properties of
DCs have even been enhanced with different nanoparticles. 34 Exosomes from DCs
modified to express indoleamine 2,3-dioxygenase (IDO), a tryptophan-degrading
enzyme that is important for immune regulation and tolerance maintenance, have been
used in the treatment of rheumatoid arthritis. 35 In another study, DC-derived exosomes
modified to express FasL on the surface, (ligand involved in apoptosis induction), were
tested as inflammatory and autoimmune therapy. 36 Other recent works are related to
the role of tumour-released exosomes to load antigens into DCs for the therapy of
malignant mesothelioma. 37
To overcome the problem of low release rate, some authors lowered the micro-
environmental pH, mimicking the natural cancer mechanism. 38 By culturing HEK293
cell lines at different pH, these authors found that low values of pH were the best to
isolate high amounts of exosomes. In spite of the impact of these results, it would be
necessary to test similar effects in non-cencerous cell lines and to determine how the
change in the harvesting conditions affects EV composition (membrane components
and cargo).
Not only human cell have been explored as EVs source: exosomes isolated from
bovine milk and loaded with different drugs were a promising strategy for mass
production of therapeutic EVs. 39 They can be easily isolated by differential
centrifugation. The biocompatibility of milk-derived exosomes was checked by clinical
110
biochemical analysis in an animal model by oral gavage during 6 h (short-term
toxicity) and 15 days (medium-term toxicity).
In recent years, some attention has been drawn to non-animal (especially plants)
EVs and their potential use in therapy. 40 In particular, fruit-derived exosomes (lemon 41
and grapes 42) have been isolated, characterized and tested as beneficial products.
Perhaps this new source of EVs could be used in the near future for the development of
EBSSNs following modifications to express the desired targeting molecules against
specific cell lines. Evidences about immunotolerance should also be provided in order
to avoid any interference in the results.
In any case, studies involving the encapsulation of the same drug into different cell
lines-derived EVs would be desirable in order to clarify whether the beneficial effect is
due to the drug or the combination of drug/type of carrier.
Since the final destination of artificial EVs would be the administration for
therapeutic purposes, the highest level of standards would be required in order to
preserve patient safety.27,43 A key point in artificial EVs development is the enrichment
from different biological samples, from cell-culture supernatants to several body fluids.
There are different reviews43- 45 and technique-comparative papers 46- 49 about isolation
procedures. They involve ultracentrifugation, filtration, immunoaffinity isolation,
polymeric precipitation and microfluidics techniques, with different degrees of purity
for the final product. 50 In this chapter, we have focussed on scalability, reproducibility
and synthetic EVs potential damage or physical modification.
Since the efficient function of EVs depends on their size distribution, aggregation
and size changes after isolation are important. Lane et al.47 studied these parameters in
four isolation methods: two aggregation kits, a density-based method and
ultracentrifugation. These authors found that some methods kept a constant vesicle
size, but large differences were observed regarding yield of isolation (the two
sedimentation methods gave recovery values two order of magnitude higher than the
other methods). Another reported obstacle is the co-purification of material with
similar physical characteristics to EVs.42,51,52
111
Scalability is also important, since the batch size is correlated- with the homogeneity
of the final product; sometimes low-size batches are more prone to being susceptible to
bias during the process, but on other occasions, higher batches yield more
heterogeneous populations due to microenvironments during procedures.
The scale up step with ultracentrifugation (UC) is limited by the number of rotor
positions and the maximum sample volume. On the other hand, methods such as Size
Exclusion Chromatography (SEC) are easy to scale up by using large columns, but with
the associated longer separation time. Pressure application to reduce processing time
can disturb EVs.43 Other methods, such as immunoaffinity isolations, are only used for
small amounts of original sample because of the high price of the reagents required.
Finally, microfluidic methods45,53 are promising, with possibility to be coupled to
online analysis. 54
Reproducibility is crucial when the product is going to be used in the clinical field.
Comparative results of UC are difficult to obtain due to the high number of models
available in the market, and this could affect the quality of the isolated product.43 The
use of a fixed instrument would keep low variability between batches.47
Welton et al. 55 reported that ready-to-use SEC columns could overcome some
problems related to homemade poured columns,52 thereby avoiding variations from
column to column, and facilitating robust protocols to be used routinely. The main
problem associated with this method is dilution of EVs in the final sample and the
subsequent need for concentration using precipitating agents or UC. This increases
retention time and the possibilities of co-precipitation of other molecules with the same
size and physical properties.
Considerable effort has been made in the field of EV isolation methods, which are
still limiting the expansion of this field. There is not yet a perfect and universal
method, and it is also accepted that selection of the isolation method could impact
downstream steps. 56
112
5.4.3 Strategies for biochemical modification
(III) increasing the amount of molecules into the cellular plasma to be encapsulated
by passive mechanism during MVB formation.
Table 5.1. Classification of techniques for the production of artificial EVs, mainly exosomes, according to
type of final product (semi- or fully synthetic) and the principle of the obtention mechanism
Class I methods (Table 5.2) involve the design of chimeric constructions of proteins
by genetic engineering. In this case, the fusion between the gene of a protein to be
incorporated and the gene of an exosomal-localized protein can be used for the
113
expression of the former on the outer surface of exosomes. This has been referred to in
the literature as Exosome Display technology, 63 and it enables the manipulation of the
protein content of exosomes and the subsequent tailoring of activities. The potential of
the method was successfully demonstrated by the production of specific antibodies
against human leukocyte antigen (HLA), a low immunogenic antigen. 64
Lactadherin C1C2 domain was also used for similar purposes by Zeelemberg et al. 65
and Hartman et al. 66 to induce expression of chicken egg ovalbumin (OVA) peptide
and the human epidermal growth factor receptor 2 antigen (HER2), respectively.
Álvarez-Erviti et al. 67 described a method of inducing surface expression of the central
nervous system–specific rabies viral glycoprotein (RVG) peptide on exosomes isolated
from immature dendritic cells derived from mice. Complementarily, these brain-
targeted exosomes were loaded with siRNA for the first time by electroporation. The
delivery of GAPDH-siRNA specifically to neurons, microglia, and oligodendrocytes in
the brain, resulted in a specific gene knockdown. This was considered the first example
of EV-based genetic therapy. One of the most important facts of this work was the
successful treatment of a highly protected tissue, the brain, in spite of the existence of
brain-blood-barrier selectivity.
114
Table 5.2. Pre-isolation methods for cargo incorporation into EVs
115
More recently, Stickney et al.62 developed another genetic engineering method for
surface expression of proteins in human cells, called surface display technology. In this
case, tetraspanin CD63 was used as a scaffold for the presentation at both
extravesicular and intravesicular orientations.
One example of this strategy is the interaction between specific sequences in RNA
molecules and proteins that are present in the route of exosomes formation.16 Highly
observed sequence motifs into RNA types studied into EVs, called EXOmotifs, was
found in mRNA 70 and miRNA. 71 Exosomes for Protein Loading via Optically
Reversible protein–protein interactions (EXPLORs) and Targeted and Molecular EV
Loading (TAMEL) are technologies based on the molecular interaction between certain
types of proteins or between RNA special structures and specific proteins. Proteins of
interest can also be loaded into the inner compartment of exosomes for their direct
delivery to the cytosol of the target cell as an alternative for therapeutic target
locations. EXPLORs61 has recently presented for that purpose. The system integrates
two elements: one is composed by the genetic fusion of the photoreceptor
cryptochrome 2 (CRY2) to the protein to be loaded, and the other is made by the fusion
of a truncated version of the CRY-interacting basic-helix-loop-helix 1 (CIBN) to
tetraspanin CD9. Both elements can be transitory attached by exposure to blue light,
which induces the interaction between CRY and CIBN, and the interaction can be
stopped once the blue light is not present.
TAMEL is another genetic engineering tool that has recently been published59 for
the active cargo of RNA. This platform is a fusion between an engineered EV-loading
protein and the RNA to be loaded. Engineered EV-loading protein is also a fused
product between an EV-enriched protein and an RNA-binding domain. This
construction is transfected into EV producer cells to make his function. Different
loading degrees can be obtained by selecting the EV-enriched protein. This is related to
the natural expression of different proteins into EVs.
116
Class III methods includes the simplest method: passive loading into vesicles
through their biogenesis. There can be two different approximations to this objective.
First, the overexpression of RNA cargo in the producer cells. The major disadvantage
of this method is the lack of selectivity in the loading process since it is gradient-driven
(the higher the concentration in the cytoplasm, the higher its possibility of being
trapped into exosomes during invagination of MVBs formation). On the other hand,
the great advantage of the method is that by translation of mRNA into receptor cell
cytoplasm, codified proteins can also be passively loaded into EVs.
The second approximation explores the active loading of the producer cells, i.e. by
nanocarriers such as fusogenic liposomes. This strategy is based on physico-chemical
properties that govern the type of mechanism (the fusiogenic properties of the two
elements that take place in the method, cells and liposomes). Second, they modify the
whole cell and not only the exosomes.
A similar method was used to modify the composition of EVs, with special focus on
the modification of the properties of the EV membrane.57 Dyes, fluorescent lipids with
different length and saturation grade of acyl chains, and chemotherapeutics were
loaded into cells by means of EVs.
These authors carried out too a membrane surface modification, with the possibility
of conjugation with molecules for targeting purposes. They first prepared MFLs
containing azide-modified lipids which were fusioned with cells. By simple incubation
with dibenzocyclooctyne (DBCO)-modified peptides, a covalent bond was created due
117
to the fast and selective reaction between DBCO and the azide group. 73 This strategy
will allow new possibilities of surface ligand decoration on EVs for targeting purposes
(such as peptides, aptens or antibodies) or for the introduction of molecules with
therapeutic properties via interaction with selective receptors. By the combination of
different head-group modified lipids, several different ligands could be incorporated
into EVs outer membrane, including receptors and co-receptors.
These are the methods that require an external force (chemical or physical) to
incorporate new molecules on previously isolated exosomes (Table 5.3).
The simplest way to incorporate any cargo into cell culture or body fluid isolated
EVs is the co-incubation of both elements. This strategy was explored by Sun et al. 74
who found that curcumin exosome-loaded exhibited better stability and higher
bioavailability in serum in an animal model. For these therapeutic-modified exosomes,
an improvement at in vivo anti-inflammatory and septic-shock was observed.
The most commonly used method for cargo incorporation into EVs after their
release is electroporation. 77 This technique involves the temporary permeabilization of
membranes through the creation of pores due to the application of high-voltage
electricity. Some authors have pointed out that this method is not suitable for siRNA
cargo into EVs due to technical problems, and an overestimated encapsulation into EVs
could be observed. It has been reported that electroporation induces siRNA
aggregation and co-pelletization with EVs during purification by ultracentrifugation,
without dependence on electroporation buffer composition. 78 They also postulated that
slight differences could be found between different EVs attending their cellular origin
(e.g. primary cells).
118
Table 5.3. Post-isolation methods for cargo incorporation into EVs
119
Another relevant problem concerning electroporation is exosome aggregation and
subsequent decrease in functionality. To avoid these problems, Hood et al. 79
electroporated exosomes from mouse B16-F10 melanoma cells by incorporating 5 nm
superparamagnetic iron oxide nanoparticles (SPIONs) as model exogenous cargo.
Other authors compared the loading efficiency of different porphyrins with different
hydrophobicities into EVs with various origins by passive loading (co-incubation), and
by active loading, such as electroporation, extrusion, saponin-assisted drug loading
and hypotonic dialysis.58 The best results were obtained for hydrophobic compounds
and for electroporation. Interestingly, zeta potential (ζ) related to the chemical
composition of EV membranes seems to play a role in loading efficiency, since higher ζ
values led to higher EE. The chemical properties of cargo are also relevant, since their
charge will condition the final outcome. Electroporation did not induce drug
precipitation.
A similar comparative study was carried reported, with the enzyme catalase. 80 For
the preparation of exosomes modified with this oxidative stress-protecting agent used
for the treatment of Parkinson’s disease, these authors selected four methods:
incubation at RT in the presence/absence of saponin, freeze/thaw, sonication, and
extrusion. Sonication yielded the higher EE (26.1 %) and the more stable product. In
contrast, this method also produced the higher increment in size, from 105 nm naïve
exosomes to 183.7 nm in catalase-loaded exosomes. Associated with size increment,
AFM observation also revealed a change in morphology, with a final non-spherical
shape.
120
Despite these promising results regarding the encapsulation of different molecules
into exosomes, standardization in systematic conditions followed by the study of
several cell lines is still necessary to strongly support the use of these methods as
routine practice in the clinical field.
The chemical copper-catalyzed reaction between an alkyne and an azide that forms
a triazole linkage (click-chemistry) has been used for the surface functionalization of
exosomes. 81 These were first chemically modified by the incorporation of alkyne
groups into amine groups from proteins by carbodiimide chemistry. 82 These authors
conjugated azide-Fluor 545 (a fluorescent compound) to activated EVs. Since no
differences in morphological and functional properties were found, it was concluded
that modification by click chemistry does not alter exosome characteristics, and allows
the incorporation of exogenous molecules to the surface of EVs.
Finally, there is another type of cargo modification that has been applied into
artificial vesicular systems (liposomes) with potential applicability to EVs. This method
is based on the ability of some peptides to be incorporated into lipidic membranes
causing disruption. 83,84 By fusion of the peptide D1-7 to an adhesion molecule
expressed in cells, targeted lipidic carriers with therapeutic cargo were produced and
successfully tested in vitro and in vivo. Another interesting application of this strategy
is its ability to insert peptidic cargo into live cell membranes, giving possibilities of
imaging live cells and modifying cell surface. This last property could be explored for
the modification of plasma membrane in EV producer cells.
121
concluded that functional properties could be tuned by modifying the membrane
composition.
Jang et al.16 used a serial extrusion through polycarbonate membrane filters with a
decreasing pore sizes (10 μm, 5 μm, and 1 μm) in a mini-extruder, similar to those
commonly used for the preparation of liposomes. Human monocytes were chosen as
precursors for membrane fragments. The yield production of NVs was 100 fold in
comparison with the production of exosomes by using the same number of cells.
Morphological studies of these NVs by crio-TEM and NTA showed many similarities
with the exosomes, round shape and a peak diameter around 130 nm. Even the
exosomal protein marker profile (CD63, Tsg101, moesin and beta-actin) checked by
Western blot was identical for the NVs and exosomes. The chemotherapeutic drugs,
doxorubicin, 5-FU, gemcitabine and carboplatin were added to the buffer where cells
were resuspended. The encapsulation efficiency in the final purified NVs was found to
be dependent on the initial amount of added drug.
122
Table 5.4. Summary of the published work about the generation of mimetic EVs nanovesicles by Top-
Down bionanotechnology. Cell source and type of cargo are encapsulated, and main characteristics are
given.
123
that of exosome from the same number of cells was achieved. Analysis of the filters by
TEM showed that many cells remained trapped in the structure.
The same device previously cited was employed by Jeon et al.17 to produce
exosome-mimetic NVs from murine embryonic stem cells for the treatment of mice
isolated skin fibroblasts. These authors wanted to explore the potential of mimetic
exosomes to induce proliferation and recovery after injury in an in vitro model.
Genomic and proteical profiles similar to original cells were assessed by PCR and
Western blotting of specific markers, and it was confirmed that successful delivery of
genetic material by NVs was reached.
124
over hydrophilic microchannels. These authors combined the induced shear stress
formation on NVs with the fragmentation of plasma membrane by the blades to obtain
fragments for the generation of exosome-like nanovesicles and the co-encapsulation of
exogenous material, (polystyrene latex beads as a model substance). It was found that
NV diameter increased as the width channel increased. This is because channel
morphology is proportional to Reynolds number (Re). In this particular case, Re is
proportional to the hydraulic diameter and, therefore, to the inertial force, which
directly increases with the channel width. In other words, NVs travelling through wide
channels have higher inertial force when they reach the blades, generating larger sliced
fragments that produce bigger NVs. These have a similar composition to that of
parental cells and naturally released exosomes by those cell lines.
One of the most interesting works in the literature18 describes the encapsulation of
22 nm fluorescent polystyrene latex beads as an exogenous simulated material, adding
these nanoparticles to the media where cells were diluted before slicing. With a final
corrected EE of 30 %, these NVs containing exogenous material were given to
fibroblasts in an in vitro experiment. After a period of time, red dots corresponding to
fluorescent beads were detected in the cytoplasm of fibroblast by confocal microscopy.
The delivery efficiency of encapsulated beads into exosome-like NVs was higher than
that of bead-aggregated NVs, revealing that exogenous material delivery with these
NVs was possible, but the efficiency was still lower than that achieved with parental
cell-components generated NVs.
The third option for obtaining artificial EVs is their construction in a fully synthetic
way by assembling individual molecules (lipids, proteins and cargo) into complex
structures, such as a bilayer structure resembling EVs membranes functionalized with
proteins for mimicking EVs function. This could be achieved by assuming that not all
components in natural exosomes are essential for specific and efficient delivery,13
including the transport of a message through direct contact with target cell receptors.
Another assumption that encourages researchers to explore this route is that, from a
structural and biochemical point of view, exosomes are liposomes with attached
proteins. Therefore, this type of vesicular systems could be an ideal substrate to
125
develop exosome-mimetic structures. The main functional components of exosomes to
be incorporated in mimetic materials have been reviewed.13 The three main
components of exosomes reported were lipids, membrane proteins and therapeutic
cargo.
One of the main advantages of fully artificial EVs over previous strategies is the
production of pure and well-defined biomaterial. In addition, production strategies of
artificial EVs based on liposomes are more sustainble and easier to scaled up.11,89 This
fact is quite important for preclinical and clinical studies and in order to manufacture
products ready to be sent to the market.2
Liposome preparation techniques have been extensively reviewed,89- 101 but not all
the methods yield vesicles suitable to becoming an artificial exosome. It could be
considered that only small unilamellar vesicles (SUVs) are ideal precursors due to their
similarities to natural exosomes (size range and membrane disposition).
126
Table 5.5. Advantages and disadvantage of most frequently used methods for Small Unilamellar Vesicles
(SUVs) preparation
127
determine the crosslinking strategy98-101 which should not compromise the biological
function. Bioconjugation should ideally show ideally be carried out under mild
conditions, aqueous media, chemoselectivity, and with a high yield.
Undecorated liposomes have also been used in the EVs research field, as EV model
for comparing isolation efficacy and physical integrity,47 detection by flow
cytometry, 104 and EV refractive index study. 105 However, their use as scaffold for
artificial EV development could offer new possibilities in basic research about EVs and
theranostic applications. To date, there are been few examples of this approximation
for the development of mimetic exosomes, and no comparative results are available
owing to the great differences between the methods used. A summary of the main
experimental work on mimicking exosome by bottom-up nanotechnology is given at
Table 5.6 and briefly commented below.
The most frequent preparation technique for SUVs as templates for EVs mimicking
is the TFH method combined with extrusion over polycarbonate membranes and
with102 or without15 previous freeze-thaw cycles. Martínez-Lostao et al.102 had a
formulation that included lipids and stoichiometry inspired in natural exosomes. The
introduction of only 5% (w/w) of an iminodiacetic acid derivative or DOGS-NTA
allowed the binding of APO2L/TRAIL-His10 to liposomes in a single step. Its
bioactivity was a higher activity than that of the soluble ligand. Moreover, a treatment
based on these synthetic exosomes achieved 60% of disease improvement in a
rheumatoid arthritis-induced animal model. In another study, liposome-bound
Apo2L/TRAIL overcame the resistance to the soluble ligand exhibited by
chemorresistive tumor cell mutants. 106 The mechanism of action of LUV-TRAIL in
hematologic cells 107 was also studied by using mimetic structures of exosomes.
128
Table 5.6. Summary of published work about the development of mimetic EVs nanovesicles by bottom-up
bionanotechnology. Formulation of the vesicles, molecules for the surface functionalization and main
physical characteristic (size) are given.
129
An innovative methodology for the production of protein encapsulated
nanoliposomes was also reported.103 They produced 82 ± 4 nm antibody-coated
liposomes with aprox. 93 % EE of BSA. The preparation route that combined a micro-
emulsion containing the protein to be encapsulated, with micelles, in order to create a
lipid bilayer formed through layer-by-layer assembling. In this work, authors selected
a Box-Behnken experimental design to optimize (maximize) the EE by adjusting some
formulation parameters. The final optimized formulation is summarised in Table 4.9.
In this particular case, researchers selected mimetic exosomes for the potential
transmission of antigen to DCs by a controlled target delivery using a conjugated
monoclonal antibody anti-DEC205, a highly expressed receptor on the surface of DCs.
The introduction of cholesterol succinate in the outer layer of the liposomes allowed
the bioconjugation of the Ab by EDC/NHS chemistry.
Despite the promising results and the advantages of these methods for the
development of liposome-based artificial EVs, there are some limiting aspects that
hinder the transfer to the clinical field. While technological progress has allowed the
design, development and production of nanomedicines with high pharmaceutical
grade, their clinical impact has been smaller than expected due to a lack of sufficient
information about in vivo interactions and fates inside the human body.11 Specific
challenges13 are related to the functionalization of vesicle surface with a combination of
functional proteins at the same time because actual methodologies are time-consuming
and because of the incorporation of nucleic acids with acceptable efficiencies. The
dependence of vesicle formulation on parameters such as fusion properties and
stability could be another limit of special relevance to immunotolerance. Finally, the
knowledge about key components in exosomes is not yet complete, since they may
vary from one cell line to another. They could even be health-state-dependent and
sensitive to harvesting conditions.
Despite attempts to mimic the exosome natural lipid composition, there is a need
for actually checking whether the formulation is active and involved in the expected
functions or is just a passive element involved only in the scaffolding of true functional
elements. Comparison of the efficiency of the intended integrated component in
differently formulated vesicles could be an interesting experiment to elucidate the role
of membrane components. Parameters of uptake route and incorporation efficiency
could also be measured with different cell lines, in order to assess the role of the target
130
cell. Other compounds as an alternative to lipids, with a high grade of
biocompatibility, could also be used for the formulation of artificial exosome bilayers.
One option is to use non-ionic surfactants 110 for the preparation of niosome-based
artificial exosomes. These compounds offer some advantages 111 over lipids, such as
price, versatility and sustainability. On the other hand, their chemical structure offers
enhanced stability from both a chemical and physical points of view. Niosomes with
size range close to EVs can also be produced.95
It is expected that microfluidic synthesis of nanovesicles will open the path for new
artificial EVs routes, with the required control of size and EE, and minimal
consumption of reagents.
Knowledge about all the biological aspects related with EVs, especially exosomes,
has opened up new frontiers in the clinical field. After an explosion of publications in
recent years about the role of EVs in physiological and pathological conditions, novel
opportunities for the development of enhanced therapeutic biomaterials have arisen.
These observations could help in the production of new materials inspired by natural
vesicles, without the classical inconveniences associated with up-to-date synthetic
alternatives (liposomes, polymersomes, inorganic nanoparticles, etc.). EV-based
therapies include tissue regeneration or immunomodulation, but drug delivery is one
131
of the most promising applications. Production, isolation, modification, and
purification at large-scale clinical grade are the main limitations of EVs becoming a
true clinically settled therapeutic agent.
Fully synthetic vesicles with EVs mimetic properties can be produced by bio-
nanotechnology. Top-down techniques that produce vesicles made of membrane
fragments obtained from the extrusion or slicing of cells, or bottom-up techniques that
take advantage of supra-molecular chemistry (mainly self-assembly) to produce
vesicles from individual molecules, represent the technology developed for that
purpose.
Despite the great potential of artificial EVs, some limitations to their development
as therapeutic tools have been identified. There is not a perfect technique, and,
depending on the final purpose of artificial EVs, combinations of procedures could
offer new insights in the field. Systematic studies with different cellular origins and
target cell lines would expand and consolidate the applications of artificial exosomes.
Comparative work including the encapsulation into different artificial vesicles would
be interesting in order to identify effects due to the carrier.
132
5.7. Referencias bibliográficas
[1] Yáñez-Mó, M., Siljander, P.R.M., Andreu, Z., Bedina Zavec, A., Borràs, F.E., Buzas, E.I., ...
& Colás, E. (2015). Biological properties of extracellular vesicles and their physiological
functions. Journal of extracellular vesicles, 4(1):27066.
[2] Lener T., Gimona M., Aigner L., et al. Applying extracellular vesicles based therapeutics
in clinical trials – an ISEV position paper. J. Extracell. Vesicles 2015;4:30087.
[5] Dalal J, Gandy K, Domen J. Role of mesenchymal stem cell therapy in Crohn’s disease.
Pediatr Res. 2012;71:445-51.
[6] Lamichhane TN, Sokic S, Schardt JS, et al. Emerging roles for extracellular vesicles in
tissue engineering and regenerative medicine. Tissue Eng. Part B Rev. 2015;21:45-54.
[7] Batrakova EV, Kim MS. Using exosomes, naturally-equipped nanocarriers, for drug
delivery. J. Control. Release. 2015;219:396-405.
[8] Tan A, Rajadas J, Seifalian AM. Exosomes as nano-theranostic delivery platforms for
gene therapy. Adv. Drug Deliv. Rev. 2013;65:357-367.
[9] van Dommelen SM, Vader P, Lakhal S, et al. Microvesicles and exosomes: Opportunities
for cell-derived membrane vesicles in drug delivery. J. Control. Release. 2012;161:635-644.
[10] Ha D, Yang N, Nadithe V. Exosomes as therapeutic drug carriers and delivery vehicles
across biological membranes: Current perspectives and future challenges. Acta Pharm. Sin. B.
2016;6:287-296.
[11] van der Meel R, Fens MHAM, Vader P, et al. Extracellular vesicles as drug delivery
systems: Lessons from the liposome field. J. Control. Release. 2014;195:72-85.
[12] El Andaloussi S, Lakhal S, Mäger I, et al. Exosomes for targeted siRNA delivery across
biological barriers. Adv. Drug Deliv. Rev. 2013;65:391-397.
133
[13] Kooijmans SAA, Vader P, van Dommelen SM, et al. Exosome mimetics: A novel class of
drug delivery systems. Int. J. Nanomedicine. 2012;7:1525-1541.
[14 Lakhal S, Wood MJA. Exosome nanotechnology: An emerging paradigm shift in drug
delivery. Bioessays. 2011;33:737-741.
[15] De La Peña H, Madrigal JA, Rusakiewicz S, et al. Artificial exosomes as tools for basic
and clinical immunology. J. Immunol. Methods. 2009;344:121-132.
[16] Jang SC, Kim OY, Yoon CM, et al. Bioinspired exosome-mimetic nanovesicles for
targeted delivery of chemotherapeutics to malignant tumors. ACS Nano. 20013;7:7698-7710.
[17] Jeong D, Jo W, Yoon J, et al. Nanovesicles engineered from ES cells for enhanced cell
proliferation. Biomaterials. 2014;35:9302-9310.
[18] Yoon J, Jo W, Jeong D, et al. Generation of nanovesicles with sliced cellular membrane
fragments for exogenous material delivery. Biomaterials. 2015;59:12-20.
[19] Forterre A, Jalabert A, Berger E, et al. Proteomic analysis of C2C12 myoblast and
myotube exosome-like vesicles: A new paradigm for myoblast-myotube cross talk?. PLoS One.
2014;9:e84153.
[22] Prado N, de Dios Alché J, Casado-Vela J, et al. Nanovesicles are secreted during pollen
germination and pollen tube growth: A possible role in fertilization. Mol. Plant. 2014;7:573-577.
[24] Laulagnier K, Motta C, Hamdi S, et al. Mast cell- and dendritic cell-derived exosomes
display a specific lipid composition and an unusual membrane organization. Biochem. J.
2004;380:161-171.
[25] Simpson RJ, Kalra H, Mathivanan S. ExoCarta as a resource for exosomal research. J.
Extracell. Vesicles 2012;1: 18374.
134
[26] Kim DK, Lee J, Simpson RJ, et al. EVpedia: A community web resource for prokaryotic
and eukaryotic extracellular vesicles research. Semin. Cell Dev. Biol. 2015;40:4-7.
[27] Kalra H, Simpson RJ, Ji H, et al. Vesiclepedia: A compendium for extracellular vesicles
with continuous community annotation. PLoS Biol. 2012;10:e1001450.
[30] Yeo RWY, Lai RC, Zhang B, et al. Mesenchymal stem cell: An efficient mass producer of
exosomes for drug delivery. Adv. Drug Deliv. Rev. 2013;65:336-341.
[32] Lai RC, Yeo RWY, Tan KH, et al. Exosomes for drug delivery—a novel application for
the mesenchymal stem cell. Biotechnol. Adv. 2013;31:543-551.
[33] Lamparski HG, Metha-Damani A, Yao JY, et al. Production and characterization of
clinical grade exosomes derived from dendritic cells. J. Immunol. Methods. 2002;270:211-226.
[35] Bianco NR, Kim SH, Ruffner MA, et al. Therapeutic effect of exosomes from
indoleamine 2,3-dioxygenase–positive dendritic cells in collagen-induced arthritis and delayed-
type hypersensitivity disease models. Arthritis Rheumatol. 2009;60:380-389.
[36] Hee Kim S, Bianco N, Menon R, et al. Exosomes derived from genetically modified DC
expressing FasL are anti-inflammatory and immunosuppressive. Mol. Ther. 2006;13:289-300.
[38] Ban JJ, Lee M, Im W, et al. Low pH increases the yield of exosome isolation. Biochem
Biophys Res Commun. 2015;461:76–79.
135
[39] Munagala R, Aqil F, Jeyabalan J, et al. Bovine milk derived exosomes for drug delivery.
Cancer Lett. 2016;371:48–61.
[41] Raimondo S, Naselli F, Fontana S, et al. Citrus limon derived nanovesicles inhibit
cancer cell proliferation and suppress CML xenograft growth by inducing TRAILmediated cell
death. Oncotarget. 2015;6:19514–19527.
[42] Pérez-Bermúdez P, Blesa J, Soriano JM, et al. Extracellular vesicles in food: experimental
evidence of their secretion in grape fruits. Eur J Pharm Sci. 2017;98:40–50.
[43] Witwer KW, Buzás EI, Bemis LT, et al. Standardization of sample collection, isolation
and analysis methods in extracellular vesicle research. J Extracell Vesicles.
2013;2:20360.
[44] Sunkara V, Woo HK, Cho YK. Emerging techniques in the isolation and
characterization of extracellular vesicles and their roles in cancer diagnostics and prognostics.
Analyst. 2016;141:371–381
[47] Lane RE, Korbie D, Anderson W, et al. Analysis of exosome purification methods using
a model liposome system and tunable-resistive pulse sensing. Sci Rep. 2015;5:1–7.
[48] Tauro BJ, Greening DW, Mathias RA, et al. Comparison of ultracentrifugation, density
gradient separation, and immunoaffinity capture methods for isolating human colon cancer cell
line LIM1863-derived exosomes. Methods. 2012;56:293–304.
[49] Greening DW, Xu R, Ji H, et al. A protocol for exosome isolation and characterization:
evaluation of ultracentrifugation, density-gradient separation, and immunoaffinity capture
methods. In: Posch A, editor. Proteomic profiling: methods and protocols. New York: Springer;
2015. p. 179–209.
[50 Marcus ME, Leonard JN. FedExosomes: engineering therapeutic biological nanoparticles
that truly deliver. Pharmaceuticals. 2013;6:659–680.
136
[51 Yamada T, Inoshima Y, Matsuda T, et al. Comparison of methods for isolating exosomes
from bovine milk. J Vet Med Sci. 2012;74:1523–1525
[52] Böing AN, van der Pol E, Grootemaat AE, et al. Singlestep isolation of extracellular
vesicles by size-exclusion chromatography. J Extracell Vesicles. 2014;3:23430.
[53] Davies RT, Kim J, Jang SC, et al. Microfluidic filtration system to isolate extracellular
vesicles from blood. Lab Chip. 2012;12:5202–5210.
[55] Welton JL, Webber JP, Botos LA, et al. Ready-made chromatography columns for
extracellular vesicle isolation from plasma. J Extracell Vesicles. 2015;4:27269.
[56] Taylor DD, Shah S. Methods of isolating extracellular vesicles impact down-stream
analyses of their cargoes. Methods. 2015;87:3–10.
[57] Lee J, Lee H, Goh U, et al. Cellular engineering with membrane fusogenic liposomes to
produce functionalized extracellular vesicles. ACS Appl Mat Interfaces. 2016;8:6790–6795.
[58] Fuhrmann G, Serio A, Mazo M, et al. Active loading into extracellular vesicles
significantly improves the cellular uptake and photodynamic effect of porphyrins. J Control
Release. 2015;205:35–44.
[59] Hung ME, Leonard JN. A platform for actively loading cargo RNA to elucidate limiting
steps in EV-mediated delivery. J Extracell Vesicles. 2016;5:31027.
[60] Kim MS, Haney MJ, Zhao Y, et al. Development of exosome-encapsulated paclitaxel to
overcome MDR in cancer cells. Nanomed Nanotechnol Biol Med. 2016;12 (3):655–664.
[61] Yim N, Ryu SW, Choi K, et al. Exosome engineering for efficient intracellular delivery
of soluble proteins using optically reversible protein–protein interaction module. Nat Commun.
2016;7:1–9.
[64] Delcayre A inventor, Le PJB inventor; Methods and compounds for the targeting of
protein to exosomes. European patent WO2003016522 A2. 2003.
137
[65] Zeelenberg IS, Ostrowski M, Krumeich S, et al. Targeting tumor antigens to secreted
membrane vesicles in vivo induces efficient antitumor immune responses. Cancer Res.
2008;68:1228–1235.
[66] Hartman ZC, Wei J, Glass OK, et al. Increasing vaccine potency through exosome
antigen targeting. Vaccine. 2011;29:9361–9367.
[67] Alvarez-Erviti L, Seow Y, Yin H, et al. Delivery of siRNA to the mouse brain by
systemic injection of targeted exosomes. Nat Biotechnol. 2011;29:341–345.
[68] Tian Y, Li S, Song J, et al. A doxorubicin delivery platform using engineered natural
membrane vesicle exosomes for targeted tumor therapy. Biomaterials. 2014;35:2383–2390.
[69] Ohno SI, Takanashi M, Sudo K, et al. Systemically injected exosomes targeted to EGFR
deliver antitumor microRNA to breast cancer cells. Mol Ther. 2013;21:185–191.
[70] Bolukbasi MF, Mizrak A, Ozdener GB, et al. miR-1289 and “Zipcode”-like sequence
enrich mRNAs in microvesicles. Mol Ther Nucleic Acids. 2012;1:1–10.
[73] Chang PV, Prescher JA, Sletten EM, et al. Copper-free click chemistry in living animals.
Proc Natl Acad Sci USA. 2010;107:1821–1826.
[74] Sun D, Zhuang X, Xiang X, et al. A novel nanoparticle drug delivery system: the anti-
inflammatory activity of curcumin is enhanced when encapsulated in exosomes. Mol Ther.
2010;18:1606–1614.
[76] Aqil F, Kausar H, Agrawal AK, et al. Exosomal formulation enhances therapeutic
response of celastrol against lung cancer. Exp Mol Pathol. 2016;101:12–21.
138
[77] Weaver JC. Electroporation: A general phenomenon for manipulating cells and tissues.
J Cell Biochem. 1993;51:426–435.
[79] Hood JL, Scott MJ, Wickline SA. Maximizing exosome colloidal stability following
electroporation. Anal Biochem. 2014;448:41–49.
[80] Haney MJ, Klyachko NL, Zhao Y, et al. Exosomes as drug delivery vehicles for
Parkinson’s disease therapy. J Control Release. 2015;207:18–30.
[81] Smyth T, Petrova K, Payton NM, et al. Surface functionalization of exosomes using click
chemistry. Bioconjug Chem. 2014;25:1777–1784.
[82] Hermanson GT. Zero-length crosslinkers. In: Hermanson GT, editor. Bioconjugate
Techniques. 3rd ed. Boston: Academic Press; 2013. p. 259–273.
[83] Sessa G, Freer JH, Colacicco G, et al. Interaction of a lytic polypeptide, melittin, with
lipid membrane systems. J Biol Chem. 1969;244:3575–3582.
[84] Pan H, Myerson JW, Ivashyna O, et al. Lipid membrane editing with peptide cargo
linkers in cells and synthetic nanostructures. Faseb J. 2010;24:2928–2937.
[85] Sato YT, Umezaki K, Sawada S, et al. Engineering hybrid exosomes by membrane
fusion with liposomes. Sci Rep. 2016;6:1–11.
[86] Morris GJ, McGrath JJ. The response of multilamellar liposomes to freezing and
thawing. Cryobiology. 1981;18:390–398.
[89] Wagner A, Vorauer-Uhl K. Liposome technology for industrial purposes. J Drug Deliv.
2011;2011:1–9.
[90] Mozafari MR. Liposomes: an overview of manufacturing techniques. Cell Mol Biol Lett.
2015;10:711–719.
139
[91] Szoka JF, Papahadjopoulos D. Comparative properties and methods of preparation of
lipid vesicles (liposomes). Annu Rev Biophys Bioeng. 1980;9(1):467–508.
[92] Abdus S, Sultana Y, Aqil M. Liposomal drug delivery systems: an update review. Curr
Drug Deliv. 2007;4:297–305.
[93] Antonietti M, Förster S. Vesicles and liposomes: A selfassembly principle beyond lipids.
Adv Mat. 2003;15:1323–1333.
[94] Lasic D.D. (1988). The mechanism of vesicle formation. Biochem J., 256:1–11.
[96] Gregoriadis G, editor. Entrapment of drugs and other materials into liposomes.Volume
II, Liposome Technology. Boca Ratonn: CRC Press; 2007.
[99] Hermanson GT. The reactions of bioconjugation. In: Hermanson GT, editor.
Bioconjugate techniques. 3rd ed. Boston: Academic Press; 2013. p. 229–258.
[100] Nobs L, Buchegger F, Gurny R, et al. (2004). Current methods for attaching targeting
ligands to liposomes and nanoparticles. J Pharm Sci., 93:1980–1992.
[103] Li K, Chang S, Wang Z, et al. A novel micro-emulsion and micelle assembling method
to prepare DEC205 monoclonal antibody coupled cationic nanoliposomes for simulating
exosomes to target dendritic cells. Int J Pharm. 2015;491:105–112.
[104] Chandler W.L, Yeung W., Tait J.F. (2011). A new microparticle size calibration
standard for use in measuring smaller microparticles using a new flow cytometer. J Throm
Haemost., 9:1216–1224.
140
[105] Gardiner C, Shaw M, Hole P, et al. (2014). Measurement of refractive index by
nanoparticle tracking analysis reveals heterogeneity in extracellular vesicles. J Extracell Vesicles,
3:25361.
[108] Pagnan G, Stuart DD, Pastorino F, et al. (2000). Delivery of c-myb antisense
oligodeoxynucleotides to human neuroblastoma cells via disialoganglioside GD2-Targeted
immunoliposomes: antitumor effects. J Natl Cancer Inst., 92:253–261.
[110] Marianecci C, Di Marzio L, Rinaldi F, et al. (2014). Niosomes from 80s to present: the
state of the art. Adv Colloid Interface Sci., 205:187–206.
[114] Bhise NS, Ribas J, Manoharan V, et al. (2014). Organ-on-achip platforms for studying
drug delivery systems. J Control Release., 190:82–93.
[115] Hu CMJ, Fang RH, Wang KC, et al. (2015). Nanoparticle biointerfacing by platelet
membrane cloaking. Nature., 526:118–121.
141
6. Using factorial experimental design to
prepare size-tuned nanovesicles
Capítulo/Chapter 6
Using factorial experimental design to prepare
size-tuned nanovesicles
6.1. Introduction
However, size is one of the most critical properties (together with shape and surface
chemistry) for understanding cell-uptake process and, therefore, bioavailability and
targetability.7 Several studies have focused on the optimization of the drug
encapsulation efficiency, while considering size as just a property for controlling
administration parameters, such as penetration kinetics in topical formulations. For
example, Padamwar et al. 8 studied the encapsulation of Vitamin E in liposomes and
found that the amount of lipids yielded a positive correlation with size, which was, in
turn, negatively correlated with penetration efficiency into the skin. Sometimes, size has
been found to increase with higher amounts of membrane components, such as
cholesterol, whereas it decreased with higher amounts of surfactants (e.g., Tween® 80).
Simultaneously, cholesterol or surfactants could affect encapsulation efficiency (EE).
Optimal situations can be reached as a compromise at intermediate levels of both factors.
In that case, Taha 9 also reported an interaction between membrane components
concentration and size reduction by ultrasounds, making factor optimization an essential
task. In other cases, an opposite effect was observed, and higher concentrations of
membrane components (such as Span® 60 and cholesterol) produced larger sizes and
increased EEs. It is useful to deliver efficient amounts of a selected drug into superficial
143
skin layers without systemic absorption. 10 On this basis, the goal of our work was to set
up a bulk-method for producing nanovesicles of controlled size that could be
subsequently modified for specific applications.
Liposomes were first described by Bangham et al. in 1965, 14 and they are basically
spherical bilayer vesicles formed by the self-assembly of phospholipids. This self-
assembly process is based on the interactions occurring between phospholipids and
water molecules, where the polar head groups of phospholipids are exposed to the
aqueous phases (inner and outer), and the hydrophobic hydrocarbon tails are forced to
face each other in a bilayer. 15 Because of the presence of both lipid and aqueous phases
in liposomes structure, they can be used for encapsulation, delivery, and controlled
release of hydrophilic, lipophilic, and amphiphilic compounds.15,16
On the other hand, niosomes are vesicles formed by the self-assembly of nonionic
surfactants in aqueous media resulting in closed bilayer structures.13,17,18 As liposomes,
their formation process is a consequence of unfavorable interactions between surfactants
and water molecules, and they can also entrap hydrophilic, lipophilic, and amphiphilic
compounds. 19,20
144
More than 20 different methods have been identified for nanovesicle preparation,
and these methods were recently reviewed. 23,24 In this work, a modified ethanol injection
method (EIM) is used, because it offers some advantages over other methods, such as
simplicity, absence of potentially harmful chemicals, and suitability for scaleup. 25
However, there is large number of variables involved in this modified EIM, and
selection of the most important of them (screening) is a crucial step in rationally
preparing vesicles by this versatile method. In this work, Z-average size and
polydispersity index (PDI) were selected as the dependent variables. They are
considered to be of great importance in nanovesicles design because most of the final
applications of these vesicular systems are directly related to these two parameters.
Factorial experimental design and analysis of variance (ANOVA) methodology are
appropriate and efficient statistical tools that permit the effects of several factors that
influence responses to be studied by varying the factors simultaneously in a limited
number of experiments.
In the recent past years, Design of Experiments (DoE) has been extensively used for
the study and optimization of vesicles and other similar organic materials. Different
designs can be applied to reduce the number of factors involved in the preparation
techniques 28 and, therefore, to minimizing the number of experiments without losing
valuable information. Plackett-Burman desing is a type of fractional design involving
relatively few runs, 29 commonly used for the screening of variables.
145
vitamin E,8 hybrid liposomes (with both low- and high-transition-temperature
phospholipids) to improve the encapsulation and delivery of silymarin, 30 and niosomes
for topical delivery applications.10,31 DoE has also been used to enhance transdermal flux
of raloxifene hydrochloride 32 or diclofenac diethylamine 33 loaded transfersomes, and of
other polymeric nanoparticles encapsulating an anticancer drug. 34 Moreover, the
interaction between vesicles and proteins, such as pectin, to improve drug-delivery
properties has been studied by DoE. 35 Nanostructured lipid carriers (NLCs) loaded with
flurbiprofen were also produced under optimal conditions using full factorial design. 36
In this work, an initial fractional factorial design with two levels (Plackett-Burman)
was used to screen the most important factors in vesicles preparation by the EIM. Then,
a 23 two-level full factorial design using center-point replicates was applied to study the
influence of the main factors and their interactions on Z-average size and PDI. Once the
appropriate operating conditions were determined, vesicles stability was studied by
using multiple light scattering technology and by measuring the encapsulation
efficiencies (EE) of different compounds.
6.2.1 Materials
A phosphate buffer (PB) solution (10 mM, pH 7.4) was used in all experiments as the
aqueous phase. The buffer solution was prepared in Milli-Q water by dissolving proper
amounts of sodium dihydrogen phosphate and sodium hydrogen phosphate, supplied
by Panreac (Barcelona, Spain). Sodium chloride from Panreac was added to increase the
ionic strength when it was required according to experiments listed in Table 1. For the
encapsulation experiments, Fat Red Bluish or Sudan Red 7B dye (C24H21N5, MW= 379.46
146
g/mol) and cholecalciferol or Vitamin D3 (C27H44O, MW=384.64 g/mol) were purchased
from Sigma-Aldrich.
Factors that could potentially affect the size of vesicles produced by the EIM were
classified in four groups, according to the different steps involved in this preparation
method: formulation (organic/aqueous phase volume ratio, phospholipid concentration,
and ionic strength), injection (injection flow, temperature, and stirring speed), evaporation
(temperature and rotation speed), and sonication (amplitude and time of sonication).
Table 6.1 list the factors and levels involved in the P-B fractional factorial design
used, where O/A is the organic/aqueous phase volume ratio, C is the concentration of
phospholipid, I is the ionic strength, QV is the injection flow, TI is the injection
temperature, NS is the stirring speed during injection, TE is the evaporation temperature,
NE is the evaporator rotation speed, A is the sonication amplitude, and t is the sonication
time.
In a second step, a 23 full factorial design with center-point repetitions (n=5) was
carried out to study the main effects and interactions between factors previously selected
by the screening design (Table 6.2). All the other factors were fixed at a certain value.
147
Table 6.1. Plackett-Burman Fractional Factorial Design: Responses, Levels, and Factors
Table 6.2. Full factorial design (23) with center point repetitions (n=5): factors, levels and responses
148
In both designs, mean diameter (Z-average size) and PDI were selected as response
variable. MINITAB statistical software (version 17) was used for all data analysis.
Analysis of variance (ANOVA) was used for this purpose.
Once the models were obtained taking into account significant factors and
interactions, a set of selected size-tuned vesicles were prepared and characterized.
Z-average size and PDI of vesicles were determined via Dynamic Light Scattering
(DLS) using a Zetasizer Nano ZS (Malvern Instruments Ltd, Malvern, UK). Three
independent samples were taken from each formulation, and measurements were
performed three times at room temperature without dilution.
149
has been removed with filter paper, the sample was air-dried, and the thin film of
stained and fixed vesicles was observed with the transmission electron microscope.
Samples were placed in cylindrical glass test cells, and backscattered light was
monitored at 30 °C as a function of time and cell height every 2 h for 7 days.
The optical reading head scans the sample in the cell, providing BS data every 40 μm
in percentages relative to standards as a function of the sample height (in millimeters).
These profiles build up a macroscopic fingerprint of the sample at a given time,
providing useful information about changes in size distribution or appearance of a
creaming layer or a clarification front with time.3,37,38
EE also provides useful information related to the stability of the vesicle membrane.
Hydrophilic compounds are entrapped in aqueous compartments between bilayers,
whereas lipophilic compounds are preferentially located within surfactant or lipid
bilayer. 39 Substances as drugs, bioactive compounds, dyes, and nanomaterials
incorporated into vesicles can also affect the morphology and stability of the final
dispersion.
For the purpose of determining EEs, Sudan Red 7B and vitamin D3 (hydrophobic
compounds) were encapsulated in the two different formulations.
150
peak area of filtered sample
EE = x 100
peak area of non-filtered sample
Equation 2
The RP-HPLC system was equipped with a HP G1315A UV/VIS absorbance detector
(Agilent Technologies, Palo Alto, CA, USA). The column was a Zorbax Eclipse Plus C18
with a particle size of 5 µm, 4.6 mm × 150 mm (Agilent Technologies). The mobile phase
consisted of a mixture of (A) 100% Milli-Q-water and (B) 100% methanol with gradient
elution at 0.8 mL/min. The step gradient started with a mobile phase of 80% A, running
100% mobile phase B in minute 5 for 10 minutes. The mobile phase B was fed for 2
minutes after each injection to prepare the column for the next sample. The separation
was carried out at 30°C. Different wavelengths were used for UV/VIS detector, namely,
533 nm for Sudan Red 7B and 270 nm for Vitamin D3.
The responses (Z-average size and PDI) of each batch from P-B design were
measured by DLS. The relative importance of the main effects on Z-average size and PDI
of PC liposomes are shown in the Pareto chart given in Figure 6.1.
Figure 6.1. Pareto chart of the standardized effects of independent variables (factors) on (A) Z-average size
and (B) PDI of PC liposomes for the Plackett-Burman fractional factorial design
151
complex. However, we decided to use the initial Plackett−Burman design only for
screening purposes and selection of the main factors from the Pareto chart, as is usually
accepted. Effects were selected by applying the hierarchical ordering principle, known
sometimes as the sparsity-of-effects principle, where higher-order effects (three- or four-
way interactions) are sacrificed to study lower-order effects (main effects first and two-
way interactions next). This principle suggests that priority should be given to the
estimation of lower-order effects, especially when resources (time and money) are scarce.
This postulate is an empirical principle whose validity has been confirmed by the
analysis of many real experiments.
According to these data, the most important variables on both responses are
organic/aqueous phase volume ratio, the (final aqueous phase) phospholipid
concentration, and the sonication amplitude. These results are in good agreement with
previous studies carried out by Kremer et al., 40 who evaluated the effect of some
preparation variables on the size and polydispersity of liposomes made from two
different natural phosphatidylcholines. Their experimental results showed that the most
important factor in the final size of liposomes was the lipid concentration in the alcohol
injected into the buffer solution. This factor corresponds to the interaction of lipid
amount and flow rate of organic solvent injected, two factors present in the Pareto chart
in Figure 6.. The same explanation was postulated by other authors,8,41,42 confirming that
the lipid concentration clearly affects the liposome size. This factor was found to be the
most relevant one for controlling morphological characteristics of phosphatidylcholine
liposomes. Szoka et al. 43 found that stirring, ionic strength, and temperature of aqueous
phase could also contribute to final size, but the effects of these factors were smaller than
those observed for lipid concentration, organic/aqueous phase ratio, and chemical
nature of the organic solvent (a parameter not included in our study). Therefore,
experimental results in Figure 6.1 confirm the previously reported observations.43
The ethanol injection method is usually chosen because it avoids the sonication step,
which is needed in several other methods of liposome preparation, such as the thin-film
hydration method. Preliminary experiments (data not shown) indicated that sonication
is a crucial step for reducing the size of both liposomes and niosomes. Alternatively,
small vesicles can be produced without sonication by using low concentration of
lipids/surfactants, but with low yield. This is why we decided to include this step as a
factor at the present study.
152
6.3.2. PC liposomes
The first three main effects from the Pareto chart obtained for the P-B design were
selected for the 23 full factorial design. The ANOVA results for Z-average size and PDI
values are listed in Table 6.3 and Table 6.4, respectively, whereas the corresponding
Pareto chart is shown in Figure 6.2. Mean sizes in the range of 55-156 nm with PDI
values between 0.173 and 0.378 were obtained for PC liposomes (with standard
deviations ranging from 0.304 to 4.40 nm for size and from 0.003 to 0.053 for PDI).
Similar size ranges were also obtained using the EIM in other previously reported
studies.22,27,41,43,44
Table 6.3. ANOVA results (coded units) for Z-average size of PC-liposomes for the 23 full factorial design;
results for S60:Cho niosomes are also given (cursive numbers)
ANOVA indicates analysis of variance; DF, degrees of freedom; SS, sum of squares; MS, mean of squares;
F, Fischer’s ratio; P, p-value.
153
Table 6.4. ANOVA results (coded units) for PDI of PC-liposomes for the 23 full factorial design; results
for S60:Cho niosomes are also given (cursive numbers)
ANOVA indicates analysis of variance; DF, degrees of freedom; SS, sum of squares; MS, mean of squares;
F, Fischer’s ratio; P, p-value.
No clear aberrant tendencies were observed, because the residuals tended to form a
line, no typical cornet pattern was observed, and no time-based pattern was detected.
Only some outliers values were detected (Cook's distance and DFITS values are given in
Table 6.5).
154
A 2.18
B 2.18
A A
C C* A
O:A*C O:A*C
O:A O:A*C*A
C*A C
O:A*C*A O:A
O:A*A O:A*A
C 2.18
D 2.18
C* A C
C A
O:A*C*A O:A
A C*A
O:A O:A*A
O:A*A O:A*C*A
O:A*A O:A*C
Figure 6.2. Pareto chart of the standardized effects of independent variables (factors) on the (A, C) Z-
average size and (B,D) PDI of (A,B) PC liposomes and (C,D) S60:Cho niosomes (1:0.5, w/w) for the 23 full
factorial design.
Table 6.5. COOK's distance and DFITS values obtained for each response in the full factorial designs
155
The ANOVA results allowed for an analysis of the contributions of the effects of the
independent variables on the response function (mean size of PC liposomes). In this case,
significant two-way interactions were identified: (O/A)×C and C×A (see Figure 6.3).
Larger sizes are reached when the organic solution has higher lipid concentration (more
than 20 g/L). On the other hand, C×A interaction reveals that the degree of size
reduction upon application of a higher amplitude depends on the total lipid
concentration present in the medium (referred to final volume of dispersion).
Figure 6.3. Three-dimensional (3D) response surface plots for the factors O/A (organic/aqueous phase
volume ratio), C (lipid or surfactant/stabilizer concentration, g/L), and A (sonication amplitude, %) for the
(A,C) Z-average size and (B,D) PDI of (A,B) PC liposomes and (C,D) S60:Cho niosomes (1:0.5, w/w).
All of the main effects are significant (p-value ˂ 0.05), with a positive effect on mean
size (a higher response value with an increase in the factor level) for the lipid total
156
concentration and a negative effect (a lower response value with a decrease in factor
level) for organic/aqueous phase volume ratio and the sonication amplitude.
Figure 6.4. Contour Plot for the factors O:A (organic:aqueous phase volume ratio), C (lipid or
surfactant/stabilizer concentration, g/L) and A (sonication amplitude, %) on Z-average size, nm (A) and
PDI (C) of PC-liposomes, and Z-average size, nm (B) and PDI (D) of S60:Cho niosomes (1:0.5, w/w).
During the injection of ethanol droplets into the aqueous phase, lipids reorganization
inside these dispersed droplets to form bilayers is favored by the fact that lipids
energetically prefer a parallel molecular arrangement.45 These planar structures give rise
157
to closed vesicles when their size induces enough surface tension to close the structure
and to minimize the bending energy.
The size of these intermediates depend directly on the number of lipid molecules
(concentration) and the dispersion degree (solubilization) in the organic phase. It is
obvious from the previous assessment that higher concentration of lipids in the droplets
will form higher membrane fragments, as our experimental results and previous
observations confirm.8,40-42
It is also important to know how easily lipid droplets are dispersed, as well as their
size and homogeneity. Lipids of higher solubility will then form smaller lipid droplets
and, consequently, short membrane fragments (and ultimately tiny vesicles).40 This
explains, in a simplified way, why higher organic/aqueous phase ratio yields smaller
liposomes.
Another aspect to be taken into consideration is the effect of sonication time. It has
been reported by Silva et al.49 that sonication time plays an important role in decreasing
vesicles size, although they observed that this effect reached a plateau at about 21 min.
Our P-B design revealed a positive effect of sonication time on Z-average size (from 15 to
30 min), although it was weaker than the effects of the other variables selected for the 23
full factorial design (especially sonication amplitude). A similar influence is observed for
PDI response, but with a stronger effect. We preferred to select sonication amplitude
instead of sonication time because one of the goals of controlling factors is to obtain a
narrow size distribution.
As the design included a center point with several repetitions (n=5), the presence of
curvature in the response variables could be tested (Tabla 9.). Because curvature seemed
158
to be significant (p-value ˂ 0.05), a term involving center point (Ct Pt) was included in
the equations for its estimation.
With all this information about the effects and their estimated coefficients, an
equation (R2 = 96.69 %) for Z-average size value of PC liposomes (Y1) was generated in
Equation 3.
Equation 3
Different behavior was observed regarding PDI, which was strongly affected by the
sonication amplitude as the only significant main effect and its interaction with total
lipid amount. There is also another interaction (O/A)×C, but with a lower effect on the
PDI response.
To understand the C×A interaction, it is important to take into account the effect of
the sonication amplitude as the main effect. An increase in this factor level leads to less
monodisperse size distribution, that is, higher PDI values. However, according to the
interaction, this response depends highly on the total amount of lipids present in the
sample. At a low level of lipids amount, the reduction in size is more effective (as
previously mentioned), but the size distribution is large. However, at a high level of the
lipids amount, this enlargement of the size distribution is significantly lower.
Curvature in the response was also tested, again revealing a significant presence (p-
value ˂ 0.05). For PDI response (Y2), the following equation with a R2 value of 89.35 %
was obtained in Equation 4.
Equation 4
These equations are formulated with uncoded coefficients, making it easier to use
them to predict selected target size and PDI values.
159
6.3.3. S60:Cho niosomes
To investigate whether the selected factors in the P−B design for PC liposomes (a
reference model for vesicular systems) produced similar effects with other different
formulations, the same 23 full factorial design using center-point replicates was
performed for a typical noisome formulation, in this particular case, S60:Cho niosomes
(1:0.5, w/w). The main variables were the organic/aqueous phase volume ratio (O/A),
the total concentration of surfactant and stabilizer (C), and the sonication amplitude (A).
The ANOVA results for Z-average size and PDI values are listed in Table 6.3, and the
corresponding Pareto chart and three-dimensional surface plot are shown in Figure 6.2
and Figure 6.3, respectively. Mean sizes in the range 224-362 nm with PDI values
between 0.032 and 0.291 were obtained for S60:Cho niosomes (with standard deviation
ranging from 1.05 to 7.28 nm for size and from 0.009 to 0.052 for PDI). Similar size and
PDI ranges were reported for niosomes prepared by EIM using Span 60 as membrane
component.17
The three main effects are significant, but in contrast to the case for liposomes, the
organic/aqueous phase volume ratio (O/A) shows a positive effect on niosomes size.
This behavior could be due to different molecular features of the surfactant and
stabilizer that result in different interactions with the organic phase and, therefore, poor
or not enough solubility.
The other two variables (C, A) have effects similar to those described above for
liposomes. Therefore, the same explanation regarding surfactant concentration and
sonication amplitude can be applied here to justify their effects on niosomes size. In this
160
case, the stronger effect of C is explained by the influence of cholesterol on the final size
of vesicles, as reported by Padamwar et al.8
Once again, curvature was detected for Z-average size response. The following
equation (Equation 5) was obtained to model this case, with an adjusted correlation
coefficient (R2) of 91.27 %.
Y3=236.9– 4.31 (O/A) – 0.012 C – 0.56 A + 0.0461 (O/A)×C + 0.00363 C×A – 0.00114 (O/A)×C×A
+ 44.00 (Ct Pt) (4)
Equation 5
On the other hand, a completely different behavior was observed regarding the PDI
response. Only the three main effects (O/A, C, A) were found to be significant, and no
interactions were found. Two positive effects on the niosome PDI were detected:
surfactant/stabilizer concentration and sonication amplitude. In this case, the total
concentration of membrane components seemed to have an important role in the vesicle
size distribution, as can be seen in the correspondent Pareto chart (Figure 6.2). This
observation once again can be attributed to the solubilization of membrane components
in the organic phase. Higher concentration of these components requiere better
solubilization in dispersed droplets to reach small membrane fragments.
A negative effect was detected for organic/aqueous phase volume ratio (O/A). As
the final concentration of ethanol increased during the injection process, a smaller size
distribution was obtained. As previously mentioned, no interaction between this factor
and the total concentration of membrane components was observed.
The Equation 6 with a R2 value of 84.73 % was obtained for the niosome PDI
model response (Y4):
161
The estimated coded coefficients for the considered effects on the Z-average sizes and
PDIs of PC liposomes and S60:Cho-niosomes are listed in Table 6.6, as a summary of the
factors´influence. Coded coefficients were used to maintain the orthogonality of the
designs and to allow for a direct comparison between coefficients.
Table 6.6. Estimated coded coefficients for the considered effects on Z-average size and PDI of PC
liposomes and S60:Cho niosomes (1:0.5, w/w)
The operating conditions were selected to prepare PC liposomes with a mean size of
70 nm and the minimum PDI value (predicted value of Y1= 67 ± 4, and Y2= 0.317 ± 0.013),
and S60-Cho niosomes with a mean size of 240 nm and the minimum PDI value
(predicted value of Y3= 239 ± 11, and Y4= 0.120 ± 0.025). These sizes and PDI values were
selected only as an example. The factor output values were O/A= 5:50 , C= 2 g/L and
162
A= 55 % for the liposomes and O/A= 5.9:50, C= 2 g/L and A= 55 % for the niosomes.
Figure 6.5 and Figure 6.6 show optimization plots and values of individual and
composite desirability for size-tuned liposomes and size-tuned niosomes, respectively.
The experimental results showed that the models obtained with the experimental
design were accurate, since mean sizes of 69 ± 0.5 nm (PDI= 0.245 ± 0.005) and 233 ± 3
nm (PDI= 0.112 ± 0.004) were obtained for PC liposomes and S60:Cho niosomes,
respectively. The relative error was low for the experimental results regarding mean size
(3% for Y1 and Y3), but higher for the size distributions (22% for Y2 and 7% for Y4).
Figure 6.5. Optimization plot and values of individual (d) and composite (D) desirability provided by the
response optimizer (Minitab, version 17) for an example of size-tuned PC liposome (desired size = 70 nm,
with a minimum PDI value).
163
Figure 6.6. Optimization plot and values of individual (d) and composite (D) desirability provided by the
response optimizer (Minitab, version 17) for an example of size-tuned S60:Cho niosome (1:0.5 w/w)
(desired size = 240 nm, with a minimum PDI).
The sizes and morphologies of the vesicles were investigated by TEM, using a
negative contrast. Figure 6.7 shows black-stained vesicles, as a result of the interactions
of the electron beam with PTA, which produces a selective deposit of metal ions that
enhances morphological details. Themicrographs show spherical structures of
approximately 80 nmfor the liposomes (Figure 6.7.C) and about 250 nm for the niosomes
(Figure 6.7.D). These values agree with the DLS measurements.
164
A B
CA DB
C D
Figure 6.7. (A,B) BS profiles and (C,D) TEM micrographs of empty vesicles designed with a controlled size
and PDI values by applying the models obtained from experimental design: (A,C) PC liposomes and (B,D)
S60:Cho niosomes (1:0.5, w/w).
Figure 6.7.D shows clusters of niosomes that are all similar in size. Aggregation arose
during the drying step prior to TEM measurements, because no flocculation phenomena
were monitored with Turbiscan apparatus.
Slight differences were noticed in zeta potential measurements, exhibiting low values
for both types of vesicles. Niosomes had values of about -16.8 ± 0.7 mV, whereas the
liposomes had values of -6.9 ± 0.3 mV. This small value for liposomes could be due to
neutralization of the negative charge from phosphate groups by sodium cations present
in the medium (from sodium chloride in the PBS buffer).
The formulated vesicles exhibited a high stability after 1 week of monitoring time. BS
profiles obtained for PC liposomes are given in Figure 6.5, where a variation of 4.5% in
the middle part of the cell (from 10 to 30 mm) is noticed. A simultaneous slight
clarification process was observed in the middle and top parts of the cell in the
corresponding transmission profile (results not shown). This was promoted by some
movement of PC liposomes towards the bottom of the cell, resulting in a slight increase
165
in BS (sedimentation). However, this was a reversible process, caused by differences in
concentration, with the sample remaining stable and maintaining its initial properties
(size and PDI). The vesicles were again characterized after gentle agitation of the cell at
the end of the monitoring time with analogous results.
For S60:Cho niosomes (Figure 6.7.B), the BS profile remained nearly constant
(variations of approximately 0.5%) with time, showing high stability. Some variation was
also observed in transmission profile profile all along the cell height, since the sample
was not translucent.
High EE values were obtained for both Sudan Red 7B and Vitamin D3, as expected
taking into account their hydrophobic character. EE values up to 90.1% and 88.0%
corresponded to Sudan Red 7B encapsulated in PC liposomes and S60:Cho niosomes,
respectively. Experiments carried out with Vitamin D3 led to EE values of 99.2% for PC
liposomes and 73.9% for S60:Cho niosomes. These results are in good agreement with
those previous studies, where compounds with similar chemical properties were
encapsulated.12,13,27
6.4. Conclusions
In this work, an adequate approximation using DoE was applied to study the
influence of experimental factors of the EIM on the mean size and size distribution of PC
liposomes and S60:Cho niosomes (1:0.5, w/w).
An initial screening design enabled a reduction of the number of variables. This was
a necessary step before carrying out a full factorial design. Finally, response models were
applied to prepare selected size-tuned nanovesicles, which were characterized from a
stability point of view.
This was achieved with a low number of experiments (58 runs). This methodology
enabled two different formulations (liposomes and niosomes, the most common types of
166
nanovesicles) to be studied in a comparative way. Stable liposomes and niosomes of the
targeted sizes were successfully prepared with the model equations obtained, with
encapsulation efficiencies higher than 73.9 % in all cases for selected hydrophobic
compounds.
These results offer new insights into the mechanism and effects of the factors
involved in nanovesicles preparation by the EIM, one of the most easily scaled-up
methods for preparing vesicles for several fields of interest.
[1] Capretto L., Carugo D., Mazzitelli S., Nastruzzi C., Zhang X. (2013). Microfluidic and lab-
on-a-chip preparation routes for organic nanoparticles and vesicular systems for nanomedicine
applications. Adv. Drug Deliv. Rev., 65:1496-1532.
[2] Rongen H.A.H., Bult A., van Bennekom W.P. (1997). Liposomes and immunoassays. J.
Immunol. Methods, 204:105-133.
[3] Pando D., Gutiérrez G., Coca J., Pazos C. (2013). Preparation and characterization of
niosomes containing resveratrol. J. Food Eng., 117:227-234.
[4] Gómez-Hens A., Fernández-Romero J.M. (2005). The role of liposomes in analytical
processes. Trac-Trends Anal. Chem., 24:9-19.
[5] Edwards K.A., Bolduc O.R., Baeumner A.J. (2012). Miniaturized bioanalytical systems:
enhanced performance through liposomes. Curr. Opin. Chem. Biol., 16:444-452.
[6] Liu Q., Boyd B.J. (2013) Liposomes in biosensors. Analyst, 138:391-409.
[7] Canton I., Battaglia G. (2012). Endocytosis at the nanoscale. Chem. Soc. Rev., 41:2718-39.
[8] Padamwar M.N., Pokharkar V.B. (2006). Development of vitamin loaded topical liposomal
formulation using factorial design approach: Drug deposition and stability. Int. J. Pharm., 320:37-
44.
167
[9] Taha E.I. (2014). Lipid vesicular systems: formulation optimization and ex vivo
comparative study. J. Mol. Liq., 196:211-216.
[10] Abdelbary A.A., AbouGhaly M.H.H. (2015). Design and optimization of topical
methotrexate loaded niosomes for enhanced management of psoriasis: Application of Box–
Behnken design, in-vitro evaluation and in-vivo skin deposition study. Int. J. Pharm., 485:235-243.
[11] Jadhav S.M., Morey P., Karpe M., Kadam V. (2012). Novel vesicular system: An overview.
J. Appl. Pharm. Sci., 02:193-202.
[12] Rajera R., Nagpal K., Singh S.K., Mishra D.N. (2011). Niosomes: a controlled and novel
drug delivery system. Biol. Pharm. Bull., 34:945-53.
[13] Uchegbu I. F., Vyas S.P. (1998). Non-ionic surfactant based vesicles (niosomes) in drug
delivery. Int. J. Pharm., 172:33-70.
[14] Bangham A.D., Standish M.M., Watkins J.C. (1965). Diffusion of univalent ions across the
lamellae of swollen phospholipids. J. Mol. Biol., 13:238-252.
[15] da Silva Malheiros P., Daroit D.J., Brandelli A. (2010). Food applications of liposome-
encapsulated antimicrobial peptides. Trends Food Sci. Tech., 21:284-292.
[16] du Plessis J., Weiner N., Müller D.G. (1994). The influence of in vivo treatment of skin
with liposomes on the topical absorption of a hydrophilic and a hydrophobic drug in vitro. Int. J.
Pharm., 103:R1-R5.
[17] Manconi M., Sinico C., Valenti D., Loy G., Fadda A.M. (2002). Niosomes as carriers for
tretinoin I: Preparation and properties. Int. J. Pharm., 234:237-248.
[18] Manca M.L., Manconi M., Nacher A., Carbone C., Valenti D., Maccioni A.M., Sinico C.,
Fadda A.M. (2014). Development of novel diolein–niosomes for cutaneous delivery of tretinoin:
Influence of formulation and in vitro assessment. Int. J. Pharm., 477:176-186.
[19] 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. Adv. Colloid Interface
Sci., 183–184:46-54.
168
[21] Mali N., Darandale S., Vavia P. (2013). Niosomes as a vesicular carrier for topical
administration of minoxidil: formulation and in vitro assessment. Drug Deliv. Transl. Res., 3:587-
592.
[22] Fan M., Xu S., Xia S., Zhang X. (2008). Preparation of salidroside nano-liposomes by
ethanol injection method and in vitro release study. Eur. Food Res. Technol., 227:167-174.
[23] 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. Adv. Colloid Interface Sci.,
205:187-206.
[24] Akbarzadeh A., Rezaei-Sadabady R., Davaran S., Joo S.W., Zarghami N., Hanifehpour Y.,
Samiei M., Kouhi M., Nejati-Koshki K. (2013). Liposome: classification, preparation, and
applications. Nanoscale Res. Lett., 8:102.
[25] Justo O.R., Moraes Â.M. (2011). Analysis of process parameters on the characteristics of
liposomes prepared by ethanol injection with a view to process scale-up: Effect of temperature
and batch volume. Chem. Eng. Res. Des., 89:785-792.
[26] Batzri S., Korn E.D. (1973). Single bilayer liposomes prepared without sonication. Biochim.
Biophys. Acta-Biomembr., 298:1015-1019.
[27] Pham T.T., Jaafar-Maalej C., Charcosset C., Fessi H. (2012). Liposome and niosome
preparation using a membrane contactor for scale-up. Colloid Surf. B-Biointerfaces, 94:15-21.
[28] Loukas Y.L. (1998). Experimental studies for screening the factors that influence the
effectiveness of new multicomponent and protective liposomes. Anal. Chim. Acta, 361:241-251.
[29] Shah S.R., Parikh R.H., Chavda J.R., Sheth N.R. (2013). Application of Plackett–Burman
screening design for preparing glibenclamide nanoparticles for dissolution enhancement. Powder
Technol., 235:405-411.
[30] El-Samaligy M.S., Afifi N.N., Mahmoud E.A. (2006). Increasing bioavailability of
silymarin using a buccal liposomal delivery system: Preparation and experimental design
investigation. Int. J. Pharm., 308:140-148.
[31] Shaikh K.S., Chellampillai B., Pawar A.P. (2010). Studies on nonionic surfactant bilayer
vesicles of ciclopirox olamine. Drug. Dev. Ind. Pharm., 36:946-53.
169
[32] Mahmood S., Taher M., Mandal U.K. (2014). Experimental design and optimization of
raloxifene hydrochloride loaded nanotransfersomes for transdermal application. Int. J. Nanomed.,
9:4331-4346.
[33] Chaudhary H., Kohli K., Kumar V. (2013). Nano-transfersomes as a novel carrier for
transdermal delivery. Int. J. Pharm., 454:367-380.
[35] Alund S.J., Smistad G., Hiorth M.A. (2013). A multivariate analysis investigating different
factors important for the interaction between liposomes and pectin. Colloid Surf. A., 420:1-9.
[36] Gonzalez-Mira E., Egea M.A., Garcia M.L., Souto E.B. (2010). Design and ocular tolerance
of flurbiprofen loaded ultrasound-engineered NLC. Colloid Surf. B, 81:412-421.
[37] Pando D., Caddeo C., Manconi M., Fadda A.M., Pazos C. (2013). Nanodesign of olein
vesicles for the topical delivery of the antioxidant resveratrol. J. Pharm. Pharmacol., 65:1158-1167.
[38] Pando D., Matos M., Gutiérrez G., Pazos C. (2015). Formulation of resveratrol entrapped
niosomes for topical use. Colloid Surf. B, 128:398-404.
[39] Devaraj G.N., Parakh S.R., Devraj R., Apte S.S., Rao B.R., Rambhau D. (2002). Release
Studies on Niosomes Containing Fatty Alcohols as Bilayer Stabilizers Instead of Cholesterol. J.
Colloid Interface Sci., 251:360-365.
[40] Kremer J.M.H., Van der Esker M.W., Pathmamanoharan C., Wiersema P.H. (1977).
Vesicles of variable diameter prepared by a modified injection method. Biochemistry, 16:3932-3935.
[41] Pons M., Foradada M., Estelrich J. (1993). Liposomes obtained by the ethanol injection
method. Int. J. Pharm., 95:51-56.
[42] Justo O.R., Moraes A.M. (2005). Kanamycin incorporation in lipid vesicles prepared by
ethanol injection designed for tuberculosis treatment. J. Pharm. Pharmacol., 57:23-30.
[43] Szoka F.C., Jr. (1996). Preparation of liposome and lipid complex compositions. U.S.
Patent 5, 549, 910.
[44] Ghanbarzadeh S., Arami S. (2013). Enhanced Transdermal Delivery of Diclofenac sodium
via conventional liposomes, ethosomes, and transfersomes. Biomed Res. Int., 1-7.
170
[45] Antonietti M., Förster S. (2003). Vesicles and liposomes: a self-assembly principle beyond
lipids. Adv. Mater., 15:1323-1333.
[46] Wang Z., He X. (2009). Dynamics of vesicle formation from lipid droplets: Mechanism
and controllability. J. Chem. Phys., 130:094905.
[47] Lasic, D.D. (1988). The mechanism of vesicle formation. Biochem. J., 256:1-11.
[48] Janmey P.A., Kinnunen P.K.J. (2006). Biophysical properties of lipids and dynamic
membranes. Trends Cell Biol., 16:538-546.
[49] Silva R., Ferreira H., Little C., Cavaco-Paulo A. (2010). Effect of ultrasound parameters for
unilamellar liposome preparation. Ultrason. Sonochem., 17:628-632.
[50] Yamaguchi T., Nomura M., Matsuok T., Koda S. (2009). Effects of frequency and power of
ultrasound on the size reduction of liposome. Chem. Phys. Lipids, 160:58-62.
[51] Lo C.T., Jahn A., Locascio L.E., Vreeland W.N. (2010). Controlled self-assembly of
monodisperse niosomes by microfluidic hydrodynamic focusing. Langmuir, 26:8559-8566.
171
7. Continuous flow production of size-
controllable niosomes using a thermostatic
microreactor
Capítulo/Chapter 7
Continuous flow production of size-
controllable niosomes using a thermostatic
microreactor size-tuned nanovesicles
7.1. Introduction
Niosomes exhibit unique advantages over the other types of vesicular systems due to
their inherent characteristics of non-ionic surfactants. 6,7 These advantages include; (i)
better chemical and physical stability of suspensions due to the absence of oxidation-
related degradation, (ii) easy derivatization to introduce different functional groups for
stability enhancement or bioconjugation, (iii) wide range of surfactant types available
(with single or double acyl chain, with different length or saturation), (iv) high
immunological tolerance, and (v) cost effectiveness. Firstly introduced in the cosmetic
industry by L’Oreal for dermal bioactive compounds delivery, 8 over the last 15 years
their applications have expanded to many fields. Food fortification, 9 diagnostic agents, 10
analytical chemistry, 11 nanomaterial synthesis, 12 and drug delivery 13 are just some of
examples. For all of these applications, a product with specific characteristics,
homogeneity and reproducibility is desired, and in particular, controlled size and
monodispersity are essential.
Effort has been made to the production of niosomes by traditional methods with tight
control over size and size distribution 14 for some specific applications. 15 For example, in
173
our previous work, 16 we have used experimental design to study the influence of
variables in the ethanol injection process, in order to improve particle size tunability.
One of the most popular chemical families for niosome production involves sorbitan
esters (commercially available as Span®). Span family members differ in terms of acyl
chain length and saturation, with a big range of hydrophilic-lipophilic balance values
(HLB), where HLB is an important parameter with implications in drug encapsulation
efficiency and morphological characteristics of particles. This parameter is also related to
the physical state at room temperature (RT), and influences the minimum temperature
(together with gel-to-liquid transition temperature, or Tc) that is required at the very stage
of the particle formation. On the other hand, some of the compounds used in
formulations with great loading capacity, low release rate and stability in solution are
solid at RT. For these reasons, a higher and controlled temperature level is mandatory
for this process.
Microfluidics technology is very promising for precise control over input variables
when mixing chemical species. 17 Other advantages include low consumption of
chemicals (relevant in formulation optimization), scale-up possibilities for industrial
production, on-line coupling to other processes (such as purification steps), and efficient
control over temperature if required. 18 Jahn et al. 19 reported for the first time the
hydrodynamic flow focussing (HFF) technique (Figure 7.1) for liposomes production.
Following that, other researchers have used this method to examine various liposomes
formulations and for encapsulating either hydrophobic or hydrophilic molecules. 20,21
Under laminar flow conditions within the HFF configuration, a stream of lipids in
organic phase is focussed between two aqueous streams in microchannels, allowing the
mixing of chemical species by molecular diffusion. At the two organic/aqueous
interfaces, bilayers can be formed and self-assembled into liposomes once a critical
concentration is reached. By controlling the flow, the extension of mixing and hence the
size of liposomes, could also be controlled. However, the production of niosomes
through microfluidic routes remains less explored, and limited attention has been paid
to using HFF technique. 22-24
174
Figure 7.1. Schematic diagram of a continuous flow microreactor based on hydrodynamic flow focusing for
vesicular systems production. The reduction of focused stream width under laminar flow conditions makes
possible the mixing of chemical species by molecular diffusion, since time for mixing decreases with the
square root of distance. By changing flow rates, the kinetics and extension of mixing can be modified, and
then, the size of particles. Amphiphilic molecules are self-assembled into bilayers once critical concentration
of solvent is reached, and molecules acquired an ordered state to minimize the interaction with water
molecules. At a certain size bending modulus induce planar bilayer to be closed into vesicles.
At present, the high temperature required for the preparation of niosomes has not
been well taken into account in microfluidics routes. For example, the previous work
that firstly explored microfluidics assembly of niosomes faced such temperature related
challenge, thus only included Span® 20 and Span® 80 (Tm = 25 °C and -30 °C,
respectively) in the study.22
Along with the wide application of continuous flow microreactors for organic
colloids preparation18 is the development of microreactor itself, including design and
manufacturing of such microdevices, with simpler and more affordable production
methods. 25 As a result, some traditional fabrications methods which stem from the
photo-electronics field, such as photolithography, 26 are being substituted by new
processes that require less expensive equipment and can be performed in common labs
with no need for clean rooms facilities. 27 Among the techniques explored, additive
manufacturing, especially 3D-printing, has emerged as a promising method for
microfluidic device manufacturing. 28 The rapid development of 3D-printing technology
and the commercialization of desk printers have enabled researchers to explore its utility
175
in microfluidic prototyping and manufacturing, 29-31 that generally use low cost raw
materials and can print objects with desired resolution.
The aim of the present work was to develop a thermostatic microreactor platform for
the continuous flow production of niosomes in a size-controllable manner. The
microfluidic reactor was designed with a hydrodynamic flow focusing configuration,
and fabricated in order to allow visualization of the dynamic process including
molecular diffusion, with the aid of an inverted microscope and a digital image
acquisition system. 3D-printing technology was used for fabricating the microfluidic
device (positive mould) and thermostatic system. The effect of operational parameters
was investigated on the final morphological characteristic of niosomes. Niosomes were
formulated with non-ionic surfactants with different transition temperatures (Tm) with
controlled temperature as a tailoring parameter to tune the size and homogeneity of
particles.
7.2.1. Materials
176
adhesive glue special for plastic materials, bought in a local store. A transparent piece of
plastic was glued to the cap aperture with the same adhesive used with the other piece.
Teflon tape was used to enhance the closure of both elements in a removable way. Holes
for the inlets and outlet pipes of the microfluidic device were manually prepared with a
sharp tool.
External supply of the recirculator was set approximately at a flow rate of 55 % of the
total volume, while peristaltic pump revolution rate was adjusted to remove water from
the chamber at a rate that allowed a continuous and constant flow through it.
Temperature inside the chamber was monitored with a digital temperature probe (Testo
110, Testo SE & Co., Germany). The sensor probe was introduced into the chamber
through a hole placed in one side of plastic window of the cap (see Figure 7.2).
Figure 7.2. Pictures composition showing the whole setup (central) and detailed components (sides) used in
this work for niosomes production by Hydrodynamic Flow Focusing with controlled temperature.
177
7.2.3. Microfluidic devices manufacturing and channel
characterization
Master mould of devices was designed in Solidworks® CAD 2016 software and 3D-
printed onto VeroClearTM resin with the HR-3D printer Objet350 ConnexTM (Stratasys
Ltd., USA). A post-printing process was also needed. First, mould was flushed with (I)
IPA, (II) deionized water, (III) acetone, and finally compressed air. Then, it was cured
overnight at 60 °C, and on the following day a treatment of the inner surface was carried
out with Aquapeel® (to avoid interference of the resin with PDMS curing process).
Three individual moulds were printed.
Once the positive mould was ready, a mixture of degassed PDMS curing agent (1:10
w/w) was poured into it, and left overnight in an oven at 40 °C. For degassing the PDMS
mixture, a bench centrifuge was used at 3000 rpm for 10 min. It should be noted that
pouring into master mould must be done slowly to minimise bubble formation. On the
following day, the replica of the mould was carefully peeled off from the mould, and
inlets and outlet holes were prepared with a 1.5 mm biopsy punch with plunger
(Miltex®, Fischer Scientific, UK).
The mixing channel (23 mm long) on the 3D-printed positive mould was
characterized in terms of morphology, accuracy and reproducibility by mechanical
profilometry (Talysurf-120L, Taylor-Hobson, United Kingdom). Three equidistance
178
measurements were taken (2 mm across the channel, perpendicular to it), and data were
processed with OriginPro 18 (OriginLab Corporation, USA) software.
The whole setup (microfluidic device inside the thermostatic chamber with
respective inlets and outlets) was placed over the stage of an inverted microscope
(IN200TAB series, AmScope, USA) with a digital imaging system to capture images
(5M.P USB CCD camera, AmScope, USA) supported with the software supplied by the
camera manufacturer. The entire experimental setup is illustrated in Figure 7.2.
A total volume of 2.5 mL was collected from the outlet of the device for each
experimental condition in a glass vial. Size (z-average or peak value, depending on the
number of peaks in the size distribution) and homogeneity (PDI) of particles were
measured by Dynamic Light Scattering (DLS) in a Zetasizer NANO-ZS equipment
(Malvern Instruments Ltd, Malvern, UK). Samples were measured undiluted by
triplicate, with the 173° backscatter detector in disposable low volume cuvettes (Malvern
Instruments Ltd, Malvern, UK).
179
NaOH solution. Once focused, a change in colour of the stream from yellow blue
indicated a molar fraction of aqueous phase close to one, and then, completes mixing by
diffusion.
Figure 7.3. Calibration plot (A) and temperature stability (B) of the in-house designed thermostatic
chamber for microfluidics chips, fabricated by 3D-printing technology with PLA filaments. Values
represented are the average of three independents measurements.
180
However, these dimensions were reasonably constant along the mixing channel length,
especially for channel height.
Table 7.1. Morphological characteristics of mixing channel for original Solidworks® CAD 2016 design
and 3D-Printed positive moulds (3D-PM) onto VeroClearTM resin with the 3D printer Objet350
ConnexTM (Stratsys). Average and standard deviation values are given for the parameters.
In our recent work,31 we found that 3D printed channels with the Objet350 Connex3
printer were smoother than channels printed with a conventional desk 3D printer
(Ultimaker 2+). However, for the same dimensions and aspect ratio, accuracy in cross
sectional shape was lower for the HR-3D printer even at large dimensions (1 mm
181
squared channels). It suggested that further studies are needed to understand this effect
with the scale and for different materials in order to inform printing parameters
optimization in terms of element dimensions, geometry, and printing materials. Apart
from the difference between CAD and 3D-PMs, the cross sectional area of Mould 3 was
similar to that previously used by Lo et al.,22 on which the selected operational
parameters of the present work were based.
The use of non-ionic surfactants for the formulation of organic colloids, especially for
NVs preparation, exhibits numerous advantages.4,6 However, a strict control of the
temperature is necessary if Span®60 (Tm = 45 °C), one of the most commonly used
surfactant in niosome formulation) is involved. Figure 7.4 shows its precipitation at RT
in microchannels once reaching the focusing region, highlighting the significance of
temperature effect.
In Figure 7.4 surfactant precipitation was observed at the focussing region and
persists along the channel length when Span® 60 is used at 25 °C. However, at 50 °C a
complete mix of both phases were produced without the presence of any surfactant
precipitation. Moreover, the production of niosomes at this temperature conditions were
observed using Transmission Electron Microspcopy (TEM) and negative staining
protocol.
This technique has been less explored than traditional bulk preparation routes,18 and
with important advantages such as better control over particle preparation and the
subsequent final characteristics (size and monodispersity, i.e.). This is important for
biomedical,1 food 35 and analytical chemistry2 applications. In this regard, the influence of
operational conditions over particles physical properties was tested by analysing the
results of 3 total flow rates (QT), two different concentrations of bilayer components, for
5 different FRR. Particle size (nm) and size distribution (PDI) were measured by DLS as
output variables. All the combinations were conducted at 50 °C, a temperature over
surfactant Tm.
182
Figure 7.4. Figure 2. Precipitation of Span® 60 (Tm=45 °C) at room temperature (upper arrow) at the
focusing region (left), 0.5 and 1.0 cm downstream (centre and right). At a temperature above surfactant
Tm, focusing is complete and vesicles formation could be checked by negative staining (Phosphotungstic
acid 2%) and Transmission Electron Microscopy (TEM).
In general terms, smaller particles were produced as the FRR increased (Figure 7.5.A
and Figure 7.5.B) for both concentrations (5 and 20 mM), and for all the QT levels. At a
concentration of 5 mM (Figure 7.5.A), the particle size decreased from 278, 298 and 358
nm (when FRR = 5) to 155, 129 and 143 nm (when FRR = 50), where QT = 50, 100 and 200
µL/min, respectively. At a concentration of 20 mM (Figure 7.5.B), similarly, the particle
size reduced from 342, 361 and 386 nm (when FRR = 5) to 164, 147 and 151 nm (when
FRR = 50) at the three QT levels of 50, 100 and 200 µL/min, respectively. Size reduction
was rapidly reached with an increment in FRR from 5 to 15, and this reduction became
less pronounced from FRR 15 to 50. It is important to take into account that when FRR
increased the total amount of bilayer components decreased, not only producing vesicle
with smaller size, since particle concentration was also reduced.
No significant effect of different QT was observed, while only some differences were
noticed in some particular combinations of parameters at low surfactant concentration (5
mM), as seen in Figure 7.5.A. These observations were in accordance with previous
studies22 carried out with identical chip configuration for the production of niosomes
formulated with other sorbitan esters (Span®20 and 80), and also for the production of
liposomes.19,20,36,37 At lower QT, also the linear velocity was lower (hence larger residence
time) what can counteract the effect of the bilayer components concentration.
183
Figure 7.5. Size (nm) and size distribution (PDI, a.u.) measured by DLS in undiluted samples from
niosomes formulated with Span® 60:Cholesterol (1:0.5 molar ratio) at 5mM (A,C) and 20mM (B,D) in a
continuous flow microreactor based on hydrodynamic flow focusing at controlled temperature (50 °C). Each
condition was tested twice, and each batch was measured by triplicate.
The increase in FRR, and the subsequent decrease in initial focused width (Wf),
reduced the time needed for a complete mixing between solvent and no-solvent (tmix,),
thus the critical concentration to induce molecules self-assembly was reached faster. This
led to smaller vesicles since the total amount of bilayer components was reduced. 38 On
the other hand, the reduction of solvent introduced in the mixing channel also decreased
the possibility of particle fusion into bigger unities by Ostwald-ripening phenomena.20,39
A reduced tmix also led to complete mixing in limited length channels. In other cases, no
diffused solvent containing amphiphilic molecules self-assembled out of the channel
under entirely different conditions (outlet pipes, with no laminar flow characteristics).
Regarding size distribution of particles (Figure 7.5.C and Figure 7.5.D), PDI value
reduced as FRR increased from 5 to 15, (for 5 mM: from 0.119, 0.141 and 0.163 nm at FRR
= 5 to 0.092, 0.071 and 0.093 nm at FRR = 50; for 20 mM: from 0.080, 0.131 and 0.118 nm
at FRR = 5 to 0.061, 0.057 and 0.071 nm FRR = 50; for both concentration values are
184
indicated for QT = 50, 100 and 200 µL/min respectively), and remained without
significant changes at higher FRR for both concentrations. Some authors19,20,32 reported a
significantly increase in PDI with the increment of FRR for an identical chip
configuration, but for liposomes production instead. However, our observation was in
line with that of Bottaro et al. [32] in a “Y”-shaped device, while Joshi et al.21 described
also a reduction in PDI as FRR increase during liposome formation. No significant
differences on PDI were observed for all QT levels applied.
The use of microreactors with different channel configurations, and the use of static
mixing enhancers, 40 could be the reason of different results among published works.
Some of them have highlighted the influence of channel dimensions and configurations
over mixing efficiency and particle properties.19,22,36 The preparation of solvent mixture
containing bilayer precursors can also influence the extension and homogeneity of
solubilisation, and in consequence, nanoprecipitation process. In the present work,
ethanol was used as solvent for microfluidic-based preparation of niosomes for the first
time, and this limited the possibility for comparison with other studies.
We have noticed that at high FRRs some transitory perturbations of the focused fluid
were recorded, especially at 50 µL/min. The focused stream exhibited a “beating pulse”
like effect that was likely produced by the syringe pump due to its own pumping
mechanism. These pulses created really short increments in the width of the focused
fluid that introduced alteration in solvent exchange kinetics and the subsequent changes
in the local concentration of bilayer precursors and solvent concentration.
Surprisingly, lower PDI values were obtained at 20 mM for all FRRs at the three
different QT. Indeed, these differences were higher at 50 µL/min. At low concentration,
those mentioned instabilities can induce more pronounced local changes in bilayer
precursor’s abundances, with the corresponding effect in particle monodispersity. To
gain insights into these observations further studies are needed.
On the other hand, larger particles were obtained when a higher concentrated
ethanolic solution of bilayer components was used (20 mM vs. 5 mM). This was observed
at all QT and FRR levels (see in Figure 7.6). The same observation was also reported by
other authors when producing liposome using microchannels,37 and in agreement with
the mechanism of vesicle formation under microfluidic flow dynamic mixing.
185
Figure 7.6. Effect of bilayer components concentration for niosomes formulated with Span® 60:Cholesterol
(1:0.5 molar ratio) and produced under the same flow conditions (QT and FRR) for size (upper row) and
size distribution (lower row).
The efficiency of mixing under the assayed working conditions was studied
following a published methodology.32 With this method, mixing efficiency was
measured through the change in colour of a pH indicator dye (bromoxylenol blue), that
changed from yellow (acidic ethanolic solution containing bilayer precursors) to blue
(basic aqueous phase, PBS pH= 10). A shift in focused fluid colour from yellow to blue
indicated that molar fraction of water into the stream was close to 1 and the subsequent
molar fraction of EtOH became close to 0, evidencing a complete mixing by solvent and
aqueous effluents. This change in colour was easily detected in the inverted microscope,
and recorded with the digital camera. As an example, results for QT = 100 µl/min at
several FRRs are shown in Figure 7.7.
186
Figure 7.7. Images (4X) of focusing region and end of the mixing channel evidencing hydrodynamic flow
focussing of a central ethanol stream at different FRR values for QT=100 μL/min and 50 °C. Yellow colour
indicates acid pH (pure EtOH, no mixing), while blue colour indicates basic pH (complete mixing by
codiffusion of solvent and no solvent). Bromoxylenol blue dye was dissolved in EtOH (acidified with acetic
acid), and PBS was adjusted with NaOH to pH 10.
Complete mixing was only reached at high values of FRR (35 and 50) for QT = 50
µl/min and QT = 100 µl/min, and only at the high FRR (50) for QT = 200 µl/min. As QT
increased, residence time of the fluid inside mixing channel reduced (from 0.5 s at 50
µl/min to 0.13 s at 200 µl/min), preventing to stay the necessary time to reach complete
mixing. Only at high FRR value, tmix was short enough to be compatible with low values
of residence time for our channel dimension (tmix = 17 ms and 8 ms for FRR = 35 and 50,
respectively, predicted according to a theoretical model (Equation 7, 41). In this model Wf
represents the width of focused stream, where w is the channel width and D is solvent
diffusion coefficient.
187
Wf2 w2 1 Equation 7
tmix ~ ≈
4D 9D (1+FRR)
Figure 7.8. Values of ethanol focused stream (Wf, μm) as flow rate ratio (FRR) increased for different
values of volumetric rates (QT) at constant temperature (50 °C), and different temperatures at constant QT
(100 μL/min). Values represent the average of two independent measurements, taken at approx. at 100 μm
from the end of focussed region.
188
Table 7.2. Correlation factor between flow focusing parameters (FRR and Wf) and particle size at different
concentration of bilayer components and variable QT (A), and at fixed concentration and QT for different
working temperatures (B).
(A)
(B)
189
Figure 7.9.Size (nm) (left) and size distribution (PDI, a.u.) (right) measured by DLS in undiluted samples
from niosomes formulated with Span® 20:Cholesterol (1:0.5 molar ratio) at 5mM in a continuous flow
microreactor based on hydrodynamic flow focusing at different controlled temperatures (30, 40, 50, and 60
°C). Each condition was tested twice, and each batch was measured by triplicate.
Figure 7.9 depicts the results of particle size at the same FRRs previously used for
Span® 60 niosomes at different working temperatures: 30, 40, 50 and 60 °C. At 30 °C, a
reduction in particle size from FRR=5 to FRR=15 was observed. As FRR increased size
became also larger (even higher that those particles produced at FRR = 5). This
phenomenon could be related to the observation of cholesterol precipitates inside the
mixing channel that were formed immediately after focusing region. The low solubility
of cholesterol in water at nearly room temperature induced its precipitation as crystals.
Those precipitates modified the flow properties and introduced turbulences that induced
micro domains in the fluid with different concentrations of bilayer components, and
particles with different morphologies. Also the depletion of cholesterol could generate
different particles than those produced in their presence. These perturbations were
magnified at higher FRR, since as seen in Figure 7.9.B the width of focused fluid became
smaller with the increment of FRR, and this stream was relatively smaller than the
formed crystals (around 100 µm structures).
For the rest of temperatures, a similar behaviour as for Span® 60 niosomes was
observed. Particles size became smaller with an inverse correlation with FRR. At higher
temperatures, focused ethanol stream was wider, and these differences were reduced
with the increment in FRR. Only slight differences in particle size could be detected
(Figure 7.9.A).
190
Regarding temperature effect some authors reported an increase in particle size as
temperature increased24 which were attributed to the bilayer expansion at higher
temperature. 43 In our case, such increase in particle size was not observed. It is know that
collapse pressure and surface compressional moduli decrease with temperature for all
surfactants, and this implies that Span monolayers are more expanded with increments
in temperature. However, as temperature increases planar bilayer precursors are less
rigid, which could be easily bended to closed structures, and this effect could lead then
to smaller particles.38
Regarding size distribution and temperature, it was observed that the increment of
temperature yielded more monodisperse particles, especially at 50 °C. Complete mixing
can be reached at FRR = 35 and FRR = 50 at any temperature. Only at 50 and 60 °C PDI
values remained nearly constant (after a first reduction from FRR=5 to 15) with the
increment in FRR.
7.3.4. Effect of surfactant acyl chain length over particles size and
monodispersity
191
Figure 7.10. Influence of acyl chain length (C12 and C18 for Span® 20 and Span® 60 respectively) of two
different sorbitan sters used in niosomes formulation (surfactant: cholesterol 1:0.5 molar ratio, 5 mM), and
produced under different conditions by hydrodynamic flow focusing at controlled temperature (50 °C) and
a flow rate QT=100 μl/min.
7.4. Conclusions
This work shows that temperature is an essential parameter that must be taken into
consideration when formulating niosomes with surfactants with Tm over RT. Also it can
be used to modify the properties of particles (size and dispersity) produced with non-
ionic surfactants with Tm above RT.
We have found that flow focussing at controlled temperature follows the same
patterns as for RT, with the ratio between aqueous and solvent streams being the main
parameter to control focused stream width and hence, mixing efficiency and kinetics.
However, total flow rate only has insignificant effect when FRRs are set to low values,
whilst it can influence residence time, and subsequently, mixing efficiency. In general
192
terms, an increase in FRR yields a focused stream being narrower, and then, smaller
particles due to the reduction in residual solvent and the introduction of less amount of
bilayer components. This reduction allows complete mixing, even at high total flow rate,
resulting in the size distribution of generated particles being more homogeneous. The
counterpart is that production yield is reduced, since particles are generated in a less
concentrated suspension. Another variable found to be relevant is the component
concentration in ethanol feeding solution, with a direct effect on particle size and
monodispersity. A more concentrated solution induces an increment in particle size at
any total flow rate, but surprisingly, better size distribution. Complementary, we have
checked the influence of acyl chain length over particles morphology, and the versatility
that introduces this parameter into the properties and functionalities of this type of
biomaterial.
The effect of ethanol stratification due to differences in density was not taken into
account, which need further investigation in for future work, in particular in its
relationship with focusing temperature.
[1] Albanese A., Tang P.S., Chan W.C. (2012). The effect of nanoparticle size, shape, and
surface chemistry on biological systems. Annu. Rev. Biomed. Eng., 14:1-16.
[2] Kelly K.L., Coronado E., Zhao L.L., Schatz G. (2003). The optical properties of metal
nanoparticles: the influence of size, shape, and dielectric environment. J. Phys. Chem. B, 107:668-
677.
[3] Tarun G., Amit K.G. (2014). Liposomes: targeted and controlled delivery system. Drug
Deliv. Lett., 4:62-71.
193
[4] Abdelkader H., Alani A.W.G., Alany R.G. (2013). Recent advances in non-ionic surfactant
vesicles (niosomes): self-assembly, fabrication, characterization, drug delivery applications and
limitations. Drug Deliv., 21:87-100.
[5] Lee J.S., Feijen J. (2012). Polymersomes for drug delivery: design, formation and
characterization. J. Control. Release, 161:473-483.
[6] 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. Adv. Colloid Interface Sci.,
205:187−206.
[8] Handjani-Vila R., Ribier A., Rondot B.A., Vanlerberghie G. (1979). Dispersions of lamellar
phases of non-ionic lipids in cosmetic products. Int. J. Cosmet. Sci., 1979, 1:303-314.
[9] Gutiérrez G., Matos M., Barrero P., Pando D., Iglesias O., Pazos C. (2016). Iron-entrapped
niosomes and their potential application for yogurt fortification. Food Sci. Technol., 74:550-556.
[10] Demir B., Baris B.F., Pinar G.Z., Unak P., Timur S., (2018) Theranostic niosomes as a
promising tool for combined therapy and diagnosis: “all-in-one” approach. Appl. Nano Mater.,
1:2827-2835.
[11] García-Manrique P., Lozano-Andrés E., Estupiñán-Sánchez O.R., Gutiérrez G., Matos M.,
Pazos C., Yañez-Mo M., Blanco-López C. (2016). Biomimetic small extracelular vesicles, 3rd
GEIVEX Simposium, San Sebastian, Spain, 29-30 September, Poster communication.
[12] De S., Kundu R., Biswas A. (2012). Synthesis of gold nanoparticles in niosomes. J. Colloid.
Interface Sci., 386:9-15.
[13] Bartelds R., Hadi N.M., Pols T., Stuart M.C.A., Pardakhty A., Asadikaram G., Poolman B.
(2018). Niosomes, an alternative for liposomal delivery. PLoS ONE, 13:e0194179.
[14] Justo O.R., Moraes A.M. (2011). Analysis of process parameters on the characteristics of
liposomes prepared by ethanol injection with a view to process scale-up: Effect of temperature
and batch volume. Chem. Eng. Res. Des., 89:785−792.
194
[15] Danaei M., Dehghankhold M., Ataei S., Davarani Hasanzadeh F., Javanmard R., Dokhani
A., Khorasani S., Mozafari M.R. (2018). Impact of particle size and polydispersity index on the
clinical applications of lipidic nanocarrier systems. Pharmaceutics, 57:1-17.
[16] García-Manrique P., Matos M., Gutierrez G., Estupiñán O.R., Blanco-López M.C., Pazos
C. (2016). Using factorial experimental design to prepare size-tuned nanovesicles. Ind. Eng. Chem.
Res., 55:9164-9175.
[17] van Swaay D., deMello A. (2013). Microfluidic methods for forming liposomes. Lab Chip,
13:752-767.
[18 Capretto L., Carugo D., Mazzitelli S., Nastruzzi C., Zhang X. (2013). Microfluidic and lab-
on-a-chip preparation routes for organic nanoparticles and vesicular systems for nanomedicine
applications. Adv. Drug Deliv. Rev., 65:1496-1532.
[19] Jahn A., Vreeland W.N., DeVoe D.L., Locascio L.E., Michael G. (2007). Microfluidic
directed formation of liposomes of controlled size. Langmuir, 23:6289-6293.
[20] Kastner E., Verma V., Lowry D., Perrie Y. (2015). Microfluidic-controlled manufacture of
liposomes for the solubilisation of a poorly water soluble drug. Int. J. Pharm., 485:122-130.
[21] Joshi S., Hussain M.T., Carla B.R., Anderluzzi G., Kastner E., Salmaso S., Kirby D.J.,
Perrie Y. (2016). Microfluidics based manufacture of liposomes simultaneously entrapping
hydrophilic and lipophilic drugs. Int. J. Pharm., 514:160-168.
[22] Lo C.T., Jahn A., Locascio L.E., Vreeland W.N. (2010). Controlled self-assembly of
monodisperse niosomes by microfluidic hydrodynamic focusing. Langmuir, 26:8559-8566.
[23] Obeid M.A., Khadra I., Mullen A.B., Tate R.J., Ferro V.A. (2017). The effects of hydration
media on the characteristic of non-ionic surfactant vesicles (NISV) prepared by microfluidics. Int.
J. Pharm, 56:52-60
[24] García-Salinas S., Himawan E., Gracia M., Arruebo M., Sebastian V. (2018). Rapid on-
Chip assembly of niosomes: batch versus continuous flow reactors. Appl. Mater. Interfaces,
10:19197-19207.
[25] Gross B.C., Erkal J.L., Lockwood S.Y., Chen C., Spence D.M. (2014). Evaluation of 3D
printing and its potential impact on biotechnology and the chemical sciences. Anal. Chem.,
86:3240-3253.
195
[26] Dong J., Liu J., Kang G., Xie J., Wang Y. (2014). Pushing the resolution of
photolithography down to 15 nm by surface plasmon interference. Sci Rep, 4:5618.
[27] Carugo D., Lee J.Y., Pora A., Browning R.J., Capretto Lorenzo, Nastruzzi C., Stride E.,
(2016). Facile and cost effective production of microscale PDMS architecture using a combined
micromilling-replica moulding (µMI-REM) technique. Biomed. Microdevices, 18:1-10.
[28] Au A.K., Huynh W., Horowitz L.F., Folch A. (2016). 3D-Prinded microfluidics. Angew.
Chem. Int. Ed., 55:3862-3881.
[29] Comina G., Suska A. Filippini D. (2014). PDMS lab-on-a-chip fabrication using 3D printed
templates. Lab Chip, 14:424-430.
[30] Hwang Y., Paydar O.H., Candler R.N. (2015). 3D Printed molds for non-planar PDMS
microfluidic channels. Sens. Actuator A-Phys., 226:137-142.
[31] Cristaldi D.A., Yanar F., Mosayyebi A., García-Manrique P., Stulz E., Carugo D., Zhang
X. (2018). Easy-to-perform and cost-effective fabrication of continuous-flow reactors for their
application for nanomaterials synthesis. N Biotechnol., 47:1-7.
[32] Bottaro E., Mosayyebi A., Carugo D., Nastruzzi C. (2017). Analysis of the diffusion
process by pH indicator in microfluidic chips for liposomes production. Micromachines, 8:209.
[33] 3D Printing and additive manufacturing-Fifth Edition, Principles and Applications, 2017,
Chee Kai Chua andKah Fai Leong (editors), World Scientific Publishing Co. Pte. Ltd. Singapore.
[34] Gaal G., Mendesa M., de Almeideb T.P., Piazzettad M.H.O., Gobbi Â.L., Riul Jr A.,
Rodrigues V. (2017). Simplified fabrication of integrated microfluidic devices using fused
deposition modeling 3D printing. Sens. Actuators B Chem., 242:35-40.
[36] Carugo D., Bottaro E., Owen J., Stride E., Nastruzzi C. (2016). Liposome production by
microfluidics: potential and limiting factors. Sci Reports, 19:25876.
[37] Hood R.R., DeVoe D.L. (2015). High-throughput continuous flow production of
nanoscale liposomes by microfluidic vertical flow focusing. Small, 11:5790-5799.
196
[38] Antonietti M., Förster S. (2003). Vesicles and Liposomes: a self-assembly principle beyond
lipids. Adv. Mater., 15:1323-1333.
[39] Zhigaltsev I.V., Belliveau N., Hafez I., Leung A.K.K., Huft J., Hansen C., Cullis P.R.
(2012). Bottom-Up design and synthesis of limit size lipid nanoparticle systems with aqueous and
triglyceride cores using millisecond microfluidic mixing. Langmuir, 28:3633-3640.
[40] Kastner E., Kaur R., Lowry D., Moghaddam B., Wilkinson A., Perrie Y. (2015). High-
throughput manufacturing of size-tuned liposomes by a new microfluidics method using
enhanced statistical tools for characterization. Int. J. Pharm., 477:361-368.
[41] Karnik R., Gu F., Basto P., Cannizzaro C., Dean L., Kyei-Manu, Langer R., Farokhzad O.C.
(2018). Microfluidic platform for controlled synthesis of polymeric nanoparticles. Nano Lett.,
8:2906-2912.
[42] Manosroi A., Wongtrakul P., Mnosroim J., Sakai H., Sugawara F., Yuasa M., Abe M.,
(2003). Characterization of vesicles prepared with various non-ionic surfactants mixed with
colesterol. Colloid Surface B, 30:129-138.
[43] Peltonen L., Hirvonen J., Yliruusi J. (2001). The effect of temperature on sorbitan
surfactant monolayers. J. Colloid Interf. Sci., 239:134-138.
[44] Gutiérrez G. Benito J.M., Pazos C., Coca J. (2014). Evaporation of aqueous dispersed
systems and concentrated emulsions formulated with non-ionic surfactants. Int. J. Heat Mass
Transf., 69:117-128.
197
8. Particle size and encapsulation efficiency
dependence with cargo molecular weight and
film hydration solution for niosomes
formulated with mixture of surfactants
Capítulo/Chapter 8
Particle size and encapsulation efficiency
dependence with cargo molecular weight and
film hydration solution for niosomes
formulated with mixture of surfactants
8.1. Introduction
Vesicles are commonly used as drug delivery systems for different active compounds.
Vesicles show special promise as drug carries due to their unique properties such as
nanometric size, high surface-volumen ratio, and ease of drug-release modulation. 1
Niosomes are a specific type of vesicles formed by the self-assembly of non-ionic
surfactants in aqueous media that leads to closed bilayers. 2 This structure enables them to
encapsulate aqueous solutions leading either the encapsulation of hydrophilic and
hydrophobic compounds. 3
Niosomes offer some advantages over other encapsulation technologies such as their
low cost, chemical stability, biocompatibility, among others. 4 Furthermore, non-ionic
surfactants self-assemblies are easily derivatized, which provides functional versatility to
their structure. 5
In recent years, niosomes have been used for encapsulating drugs, 6 nutraceuticals, 7
antioxidants, 8,9 micronutrients, 10 etc. There are still many challenges in the development of
delivery systems that could encapsulate hydrophilic compounds effectively. There are
colors, nutraceuticals and vitamins of industrial interest that need to be protected of
chemical degradation, to inhibit adverse interaction with other components, to mask off-
flavors, or to obtain a particular release profile. For example, some water soluble colors are
susceptible to chemical degradation under certain conditions, e.g., pH, light or
temperatura. 11 Water soluble vitamins are not stored in the body, and could be washed
out during food processing making necessary to replenish them daily. 12
201
In this work, three different bioactive hydrophilic compounds with different molecular
weight and industrial interest were encapsulated (Figure 8.1): ascorbic acid (Vitamin C),
rhodamine B (Fluorescent organic dye) and cobalamin (Vitamin B12). Ascorbic acid, found
in citrus fruits, berries and vegetables, acts mainly as antioxidant, but also promotes the
production of noradrenaline, collagen, bile acids, and increases the intestinal absorption of
non-heme iron.12,13 Rhodamine B is a synthetic and highly soluble molecule used as
pigment in drug and cosmetic formulations due to its absorption and emission
properties. 14,15 Finally, cobalamin belongs to the B-complex vitamins and can be found in
cheese, fish, milk or eggs. It functions as coenzyme and as an important intermediate in
the metabolism of folic acid, a compound really important to prevent congenital disorder
during the first stage of pregnancy.12
On the other hand, it is important to consider the effect of the composition of aqueous
solution used to hydrate the film, over particles morphology (size and PDI) and functional
characteristics such as encapsulation efficiency (EE), with special focus on hydrophilic
compounds encapsulation. The use of different co-solvents in the self-assembly process of
the niosomes, expands their application through the possibilities of tune particle size and
EE values, depending on selected applications. For example, glycerol has been used to
enhance the solubility of bioactive compounds, 16 but also as cryoprotector agent for
202
liophilization, 17 or to enhance the drug carriers penetration for transdermal
administration.16,18 In addition, polyethylene glycol (PEG), a water soluble polymer with
several molecular weight versions, has been used to prepare highly stable niosomes, 19 and
as sterical stabilizer of liposomes, 20 allowing longer circulation times in blood. 21
8.2.1. Materials
For the film hydration solution, polyethylene glycol 400 (PEG-400, MW 380-420 g/mol,
density 1.128 g/cm3, VWR International LLC, BDH PROLABO), glycerol bidistilled 99.5 %
(Gly, MW 92.09 g/mol, density 1.261 g/cm3, VWR International LLC, BDH PROLABO)
and, ultrapure water were used.
Absolute ethanol from J.T. Baker was used for bilayer components stock solutions.
Methanol HPLC grade from VWR International LLC, BDH PROLABO), and acetic acid
solution (49-51 %, HPLC grade) from Sigma Aldrich were used for liquid chromatography
(HPLC) performance.
203
8.2.2 Niosomes preparation
Niosomes were prepared by a modified Thin Film Hydration method (TFH). The
corresponding amount of surfactants and membrane additives were placed into a 100 mL
round bottom flask in an equimolar ratio (from ethanolic stock solutions). The organic
solvent was removed using a rotary evaporator (Buchi Labortechnik AG, Flawil,
Switzerland), until get a homogeneous dried film. This film was then hydrated using
different aqueous-based solutions at 60 ºC, and agitated at 100 rpm during 30 minutes.
Suspension on vesicles were let acquire room temperature prior to purification and/or
characterization.
Niosomes containing the active principle inside, were prepared by the hydration of the
thin films formed after the evaporation with the corresponding aqueous solutions:
ultrapure water (or MQ), water:glycerol (60:40, v/v) or GLY, and water:PEG-400 (55.3:44.7,
v/v) or PEG. Both solutions have the same density.
Two different formulations of niosomes were studied. The first one contained an
equimolar ratio of Tw80 and Sp80, while the second was a mixture of Tw80, Sp80, and Dc
as bilayer stabilizer in a 1:1:1 molar ratio. The total concentration of membrane
components was kept constant at 10 mM (final concentration in vesicles suspension). The
molar ratio between both surfactants were selected to yield an HLB value of 10.
The purification of niosomes suspension was carried out by using two different
methods: dialysis and gel permeation chromatography.
8.2.3.1. Dialysis
204
collected for the external phase, once the concentration on the external media was more
than the 99.8% of the original concentration, it was considered that the dialysis time was
enough for the purification.
HPLC was used to determine the concentration of free compound in the collected
samples.
HPLC was used to determine the absence of free compound in the first collected
fractions from the column, were niosomes eluted, to assess the suitability of the SEC
column.
Mean diameter (z-average, nm) and Polydispersity Index (PDI, a.u.) for the prepared
niosomes were measured by Dynamic Light Scattering (DLS) on a Zetasizer NanoZS
Series (Malvern Instruments Ltd., Malvern, UK). Measurements were performed by
triplicate with undiluted samples, and at 25 °C. Low volume plastic disposable cuvettes
were used during size characterization (Malvern Instruments Ltd., Malvern, UK).
205
Table 8.1. Chromatography mediums used for gel filtration based purification of loaded niosomes
206
[𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐]𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒𝑒 Equation 8
% 𝐸𝐸𝐸𝐸 = × 100
[𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐𝑐]𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖𝑖
Purified niosomes were diluted 1:10 (v/v) using methanol in order to break the
niosomal bilayers and release the encapsulated compounds. The quantification of the
cargo molecules was carried out by reverse phase liquid chromatography, or RP-HPLC
(HP series 1100 chromatograph, Hewlett Packard, Agilent Technologies), with a Zorbax
Eclipse Plus C18 column (4.6 mm x 150 mm, 5 μm, Agilent Technologies, Santa Clara,
California, USA). UV/vis (HP G1315A detector, Agilent Technologies) and fluorescence
(1260 Infinity A detector, from Agilent Technologies), were used as detection coupled to
the chromatographic separation. The following HPLC programs were used:
Ascorbic Acid
A linear gradient was performed with 0.1 % (v/v) acetic acid in MQ (mobile phase A)
and methanol (mobile phase B). The gradient started with 95 % of A, reaching 20 % of A at
min. 15, and kept constant for 5 min. The flow rate was 0.9 mL/min. Retention time for
AA was 2.28 min at λ = 278 nm.
Rhodamine B
A linear gradient was used with MQ (mobile phase A) and methanol (mobile phase B).
The gradient started with 2 % of B, running 100 % of B at min. 21, and kept constant for 5
min. The flow rate 1 mL/min. Retention time for RB was 19 min at λ = 554 nm.
A linear gradient was used with MQ (mobile phase A) and methanol (mobile phase B).
The gradient started with 20 % of B, obtaining 100 % of B at 5 min and kept constant for 10
min. The flow rate was 0.8 mL/min. Retention time for B12 was 4.35 min at λ = 361 nm.
207
8.2.6. Differential Scanning Calorimetry (DSC)
In the present work, the effect of hydration solution composition in the thin film
hydration method for niosomes preparation has been studied. More precisely, glycol
compounds such as glycerol and polyethylene glycol 400 have been tested to prepare
niosomes for the encapsulation of hydrophilic compounds. A mixture of surfactant has
been used in the formulation, with and without a bilayer stabilizer, such as dodecanol.
The influence of dodecanol over particle size has a discrete effect. For example, it
highly influenced niosomes size when encapsulated AA and PEG was used as hydration
solution. On the other hand, Dc seems to reduce particle size for RB and B12 loaded
niosomes when Gly and PEG are used for hydration.
208
Figure 8.2. (A) Size values of empty vesicles. Tw80: tween® 80; Sp80: span® 80; Dc: dodecanol. (B) Effect of
hydration solution and cargo molecular weight over particle size, for niosomal formulations Tw80:Sp80 (1:1
molar ratio) and Tw80:Sp80:Dc (1:1:1 molar ratio) prepared by Thin Film Hydration method (TFH).
Empty vesicles allows to study the effect of the hydration solution used and the
presence of Dc on the final particle size. The presence of Dc increases the drop size
independently of the hydration medium used, this is due to their location at the
membrane layer and their reduction on the surfactant curvature on the presence of
alcohol. In order hand the use of Gly and PEG solutions as hydration media increases
significantly the particle size of the vesicles formed. It is important to point that these
compounds can be attached to the membrane compounds producing higher stability to the
system but also could increase the particle size.16,28
Molecular weight of encapsulated compounds into the vesicles has not a clear effect
over particle size. For vesicles formulated with Tw80 and Sp80 the particle size seems to
increase when larger molecular weight compounds are encapsulated, these effect is
especially noticeable for the B12, probably due to their significantly major molecular
weight value.
The addition of polyethylene glycol with different MW in aqueous solution for vesicles
(especially niosomes preparation has been also studied.19,22-24 PEG-400 and PEG-6000 are
the most popular options, frequently used for surfactant mixture based formulations,
209
where Tween® and Span® (60 and 80) are widely used. The concentration of PEG-400 in
aqueous solutions was reported to influence the niosomal formation in a formulation¡Error!
Marcador no definido. based on Tween® 80. Similar, PEG-6000 concentration influenced the
particle size of niosomes formulated with Tw80:Sp80 1:0.3 mass ratio.¡Error! Marcador no definido.
Similar size values to the ones reported in this work have been found by other authors
when Span® 60 is combined with PEG-400.¡Error! Marcador no definido.
These glycol compounds alter the packing of the bilayer and increase their curvature,
which is manifested as an increment in particle radius. For example, glycerol has the
property to change the dielectric constant of the bilayer interior, and bilayer components
can re-arrange with different interactions.18 PEG molecules can interact by H bonds with
Tween® 80 molecules, so al low concentration can be part of the bilayer, which is less rigid.
However, al high concentration it can decrease the stability of the membrane until the
disruption. In that case, the instability of niosomes could led to fusion of particles and
large vesicles can be obtained. This may be the mechanism that explains our size results
when PEG is used as hydration solution.
Some works16 have checked the influence of chemical composition of the film
hydration solution for niosomal formulation, with special focus on the presence of poly-ol
compounds (alcohols). These authors reported the influence alcohol type and
concentration over particle size, and they found differences. Particularly, they reported
that glycerol concentration in hydration solution has positive effect: as glycerol
concentration increases (up to 40%, like our study) bigger particles were obtained without
any impact over monodispersity.
Some other works18 using aqueous-glycerol solutions for vesicles preparation by direct
hydration with sonication, have reported that empty and loaded vesicles did not differ in
terms of size, however other hydration solutions such as propylene glycol (1:1, v/v), more
similar to PEG, yields bigger particles compared to MQ or GLY solutions. These authors
have attributed this phenomena to the interaction of this compound with bilayer
components.¡Error! Marcador no definido.
210
Surprising the size of encapsulating RB and B12 vesicle is reduced around 30% when
Dc is used as a membrane component and PEG solution as a hydration method. This
phenomena indicates that even encapsulated dompounds are hydrophilic and hence are
encapsulated at the iner part of the vesicle they have some interacions with the membrane
compounds.
Regarding the monodispersity of the suspensions, in all the cases, both empty and
loaded vesicles showed a PDI value from 0.3 ± 0.1 for empty Gly niosomes (with Dc) to 0.7
± 0.1 for AA loaded MQ niosomes (with Dc). Encapsulation seems to not influence the
value, since in some cases loaded vesicles exhibit better monodispersity than loaded
niosomes.
Regarding the EE of the different compounds we observed differences related with the
molecular weight of the cargo, and for both formulations. The best EE values were
obtained for AA when MQ is used as hydration solution, with independence of the
purification method (Figure 8.3). For this compound it seems that hydration solution
composition is not a key factor for encapsulation, since in almost all the cases the EE
values were similar. Only some particular differences are observed in the formulation
where Dc is used as membrane stabilizer (Gly yields the lowest EE value, 68%, using
dialysis as purification method, and MQ the higher EE value, 100%, using gel filtration as
purification technique).
However, RB and B12 seems to have an important dependence with hydration solution
composition, since a great difference in EE values can be observed depending on medium
chemistry. In the case of RB, EE ranges between 8.4% (Gly) and 60% (PEG) when dialysis
is used as purification method, and from 9% (MQ) to 22% (PEG) in the case of Dc
containing formulation with same purification method.
211
Figure 8.3. Effect of hydration solution and cargo molecular weight over encapsulation efficiecy, for niosomal
formulations (A)Tw80:Sp80 (1:1 molar ratio) and (B) Tw80:Sp80:Dc (1:1:1 molar ratio) prepared by Thin
Film Hydration method (TFH). Tw80: tween® 80; Sp80: span® 80; Dc: dodecanol. Dialysis (10 MWCO
membranes) and gel filtration (Sepahdex G25 or Seharose CL-4B, depending on the compound) were used as
purification methods.
It seems that PEG greatly improve RB encapsulation. Besides these good results, when
gel filtration is used EE remains with similar values and in general, offering lower values
212
than dialysis purified vesicles for both types of formulation. A possible explanation is
given into the next section.
A similar pattern of results is described for B12 encapsulation. Again, the use of PEG as
hydration solution yields the higher EE values (67%) for dialysis purified vesicles for both
formulation, but a not clear effect of medium composition can be observed for gel-filtrated
niosomes. Dc addition into the formulation is not an important parameter for his
encapsulation.
Muzzalupo et al.16 reported that 40% of glycerol yields the higher EE% compared to
other alcohols for sulfadiazine, and hydrophilic drug. They attributed this effect to the
presence of multiple –OH groups in the alcohol that could help the drug to be totally
solubilized thanks to H bonds between both molecules. A similar effect could be related to
B12 EE% without Dc, but may be not useful for RB encapsulation. However, PEG increase
EE of both, RB and B12, for both formulations. PEG has been tested to enhance the
solubility of amphiphilic compounds such as ellagic acid,¡Error! Marcador no definido. quercetin, 25
paclitaxel, 26 however no data about hydrophilic compounds have been found.
Interestingly, our formulation offers better AA EE% than liposomes found in the
literature, where only 10% was reached. 27 Regarding vitamin B12, PEG niosomes
with/without Dc offer better encapsulation than liposomes, 28 however there is a great
difference in size (2 µm for niosomes vs less than 100 nm for liposomes). On the other
hand, EE% values for RB are unusual, since the papers only report the encapsulation,
without interest in maximize this parameter.
These results evidence that exists and interaction between cargo molecular weight and
hydration solution composition without any clear influence of niosomes formulation, at
least for the introduction of Dc as bilayer stabilizer. Only in the case of MQ and dialysis, a
clear influence of cargo MW is observed, with a reduction in EE as MW increase. The
composition of film hydration solution could be a key parameter for some compounds EE
optimization, with a possible relationship with their molecular weight (medium or high
molecular weight compounds), and particular dependence of chemical nature of the
compounds, since a not general pattern was observed. In our opinion, the enhancement of
213
EE% values using Gly, and especially PEG, is an interaction of solubility enhancement and
the effect of increase particle size, which for hydrophilic compounds is translated into a
bigger capacity of niosomes to carry more cargo molecules due to physical (volume)
capacity.
Regarding the purification strategies, dialysis is the most popular option chosen to
purified vesicles encapsulating hydrophilic and amphiphilic compounds18,19,¡Error! Marcador no
definido.,¡Error! Marcador no definido.. However, if the permeating through the bilayer is not clear
understood for a specific formulation, realease and purification can be overlapped, and
EE% could be underestimated. To assess this for our formulation we selected SEC
chromatography as alternative method, 29 to be able to compare the performace of both
techniques.
As described previously, important differences in EE values are obtained when the two
purification methods are compared, and those discrepancies seems to be magnified with
hydration medium composition. The general trend is to observe a lower EE value for
niosomes suspensions purified by gel filtration.
The greatest differences are in all the cases observed for the use of PEG in both
formulations, and in the majority of them, MQ offers no differences between methods.
Also, a positive interaction with molecular weight is described for GLY and PEG.
To assess if this result relies on physical forces involved in the purification methods,
empty niosomes prepared in the three different aqueous solutions were subjected to gel
filtration. Particle size, size distribution and concentration were measured by Nanoparticle
Tracking Analysis (NTA) before and after the process (Figure 8.4).
214
Figure 8.4. Naparticle Tracking Analysis (NTA) of the three different hydration solution based niosomes
after and before SEC purification. MQ and Gly vesicles were purified using Sephadex G25 and PEG vesicles
by Sepharose CL-4B.
The results showed a reduction in mean size, and particle concentration for PEG based
niosomes suspension. It seems that a retention of vesicles of a mechanical disruption of
particles may occur during their flow through the column. During the process, the bed of
the column experiments a visible compaction due to the high viscosity of the fluid even at
low flow rate recommended by the manufacturer for viscous solutions. This phenomena
could yield to mechanical stress of the vesicles due to shear forces as they pass through the
compacted gel. Subsequently, loaded vesicles could collapse and their content could be
release to the medium where is trapped by the effect of the gel even when is compacted. A
reduction in EE value could be then observed for this case as reported.
Interestingly, particles in GLY solutions seems to keep their integrity, since no changes
in measured parameters have been reported. And also curiously, MQ suspended niosomes
experiment a slight reduction in size and concentration, what could be related to
entrapment phenomena of the smallest and highest niosomes by capture and clogging
with the stationary phase respectively.
Besides being a popular choice for vesicles purification, SEC has some drawbacks that
must be taken into consideration. For example, it has been reported that vesicles can be
retained by stationary phase 30, and this retention is dependent of particles pores and not
by particles size itself. Sepharose CL-40 contains particles with 20 nm pore size, which is
enough to allow the flow of small vesicles through them. However, large flexible particles
215
like PEG-hydrated may clog the pores and disturb the process, leading to underestimation
of EE% by vesicles loss and vesicles disruption into the columns. This fact could be
potentiated by viscous solution, such as 45% PEG in water.
Differential Scanning Calorimetry (DSC) is a useful technique carried out to study the
phase behavior of lipids and surfactant based bilayers, and gives information about
molecular interactions of the structure, which allows getting information about stability
and fluidity of the vesicle bilayer. 31 Also, this technique has been applied to measure the
EE% of hydrophobic compounds, 32 since they are loaded into the structure and then, alter
the cohesion of the bilayer.
Figure 8.5. DSC curves acquired in heating mode for the formulation without dodecanol in the three different
hydration solutions: ultrapure water (MQ), water:glycerol 60:40 v/v (Gly), and water:PEG-400 55:45 v/ v
(PEG).
Figure 8.5 represents the DSC curves obtained for empty vesicles prepared into the
three different hydration solutions. It is clear, that the incorporation of PEG and Gly
changes the stability of the bilayer, since Tc is decreased and the morphology of the peaks
shows a less ordered structure, evidenced for broad peak transition and loss of symmetry.
Gly and PEG transitions from gel-to-liquid seems to be in two steps, showing
216
heterogeneity of the bilayer. As consequence, bilayer becomes more fluid, and bigger
particles could be formed.
8.4. Conclusions
Hydration solution composition has been correlated with particle size for a niosomal
formulation based on surfactant mixture with/without dodecanol as membrane stabilizer.
In addition, a possible interaction with cargo molecular weight could exist, since in
particular cases, particles become slightly bigger as molecular weight increase. This
increment in size is correlated with an increase in EE for these compounds. Since bigger
particles can encapsulated more compound molecules than smaller ones, especially for
hydrophilic compounds where they are loaded into the aqueous inner cavity of vesicles,
the relationship between EE and hydration solution composition seems to be purely due to
vesicles physical aspects (loading volume capacity). DSC curves showed a change in
bilayer structure with the use of glycerol and polyethylene glycol 400, that may interact
with surfactants and create a less organized structure.
Encapsulation efficiency obtained using dialysis method were higher than obtained for
gel filtration for the same samples. In addition, when dialysis was used, encapsulation
efficiency strongly depended of the hydration media while no differences were observed
when gel permeation was used as purification method. It seems that gel filtration is not an
appropriate purification method for suspensions of large vesicles, especially for viscous
solutions. Mechanical stress of particles during separation could lead to unsatisfactory
results in terms of particle integrity and subsequent EE values.
217
8.5. Bibliographic references
[1] Marianecci C., Petralito S., Rinaldi F., Hanieh P.N., Carafa M. (2016). Some recent
advances on liposomal and niosomal vesicular carriers. J. Drug Deliv. Sci. Technol., 32:256–269.
[2] Uchegbu I.F., Florence A.T. (1995). Non-ionic surfactant vesicles (niosomes): Physical and
pharmaceutical chemistry. Adv. Colloid Interface Sci., 58:1–55.
[3] Uchegbu I.F., Vyas S.P. (1998). Non-ionic surfactant based vesicles (niosomes) in drug
delivery. Int. J. Pharm., 172:33–70.
[4] Salim R., Minanikawa M., Sugimura H., Hashim A. (2014). Amphiphilic designer nano-
carriers for controlled release, from drug delivery to diagnostics. Med. Chem. Commun., 5:1602–1618.
[5] 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. Adv. Colloid Interface Sci.,
205:187–206.
[6] Alomrani A.H., Al-Agamy M.H., Badran M.M. (2015). In vitro skin penetration and
antimycotic activity of itraconazole loaded niosomes: Various non-ionic surfactants. J. Drug Deliv.
Sci. Technol., 28:37–45.
[7] Pando D., Beltrán M., Gerone I., Matos M., Pazos C. (2015). Resveratrol entrapped
niosomes as yoghurt additive. Food Chem., 170:281–287.
[8] Tavano L., Muzzalupo R., Picci N., De Cindio B. (2014). Co-encapsulation of antioxidants
into niosomal carriers: Gastrointestinal release studies for nutraceutical applications. Colloids Surf.
B., 114:82–88.
[9] Tavano L., Muzzalupo R., Picci N., De Cindio B., (2014). Co-encapsulation of lipophilic
antioxidants into niosomal carriers : Percutaneous permeation studies for cosmeceutical
applications. Colloids Surf. B., 114:144–149.
[10] Gutierrez G., Matos M., Barrero P., Pando D., Iglesias O., Pazos C. (2016). Iron-entrapped
niosomes and their potential application for yogurt fortification. LWT - Food Sci. Technol., 74:550–
556.
218
[11] McClements D.J. (2015). Encapsulation, protection, and release of hydrophilic active
components: Potential and limitations of colloidal delivery systems. Adv. Colloid Interface Sci.,
219:27–53.
[12] Fathima S.J., Nallamuthu I., Khanum F. (2017). Vitamins and minerals fortification using
nanotechnology: bioavailability and Recommended Daily Allowances. Nutr. Deliv., 457–496.
[13] Du J., Cullen J.J., Buettner G.R. (2012). Ascorbic acid: Chemistry, biology and the
treatment of cancer. Biochim. Biophys. Acta., 1826:443–457.
[14] Kim H.N., Lee M.H., Kim H.J., Kim J.S., Yoon J., (2008). A new trend in rhodamine-based
chemosensors: application of spirolactam ring-opening to sensing ions. Chem. Soc. Rev., 37:1465–72.
[15] Ulusoy H.İ., (2017). A versatile hydrogel including bentonite and gallocyanine for trace
Rhodamine B analysis. Colloids Surf. A, 513:110–116.
[16] Muzzalupo R., Tavano L., Lai F., Picci N. (2014). Niosomes containing hydroxyl additives
as percutaneous penetration enhancers: Effect on the transdermal delivery of sulfadiazine sodium
salt. Colloids Surf. B., 123:207–212.
[17] Stark B., Pabst G., Prassl R. (2010). Long-term stability of sterically stabilized liposomes
by freezing and freeze-drying: Effects of cryoprotectants on structure, Eur. J. Pharm. Sci., 41:546–555.
[18] Vitonyte J., Manca M.L., Caddeo C. Valenti D., Peris J.E., Usach I. Nacher A., Matos M.,
Gutiérrez G., Orrù G., Fernàndez-Busquets X., Fadda A.M., Manconi M. (2017). Bifunctional viscous
nanovesicles co-loaded with resveratrol and gallic acid for skin protection against microbial and
oxidative injuries. Eur. J. Pharm. Biopharm., 114:278-287.
[19] Hua W., Liu T. (2007). Preparation and properties of highly stable innocuous niosome in
Span 80/PEG 400/H2O system. Colloids Surf. A, 302:377-382.
[20] Yongzhuo Huang Y., Chen J., Chen X., Gao J., Liang W. ().PEGylated synthetic surfactant
vesicles (Niosomes): novel carriers for oligonucleotides. J. Mater. Sci.: Mater. Med., 19(2):607-614.
[21] Gianasi E., Cociancich F., Uchegbu I.F., Florence A.T., Duncan R. (1997). Pharmaceutical
and biological characterisation of a doxorubicin-polymer conjugate (PK1) entrapped in sorbitan
monostearate Span 60 niosomes. Int. J. Pharm., 148:139–148.
219
[22] Liu T., Guo R. (2005). Preparation of a highly stable niosome and its hydrotrope-
solubilization action to drugs. Langmuir, 21:11034-11039.
[23] Junyaprasert V.B., Singhsa P., Suksiriworapong J., Chantasart D. (2012). Physicochemical
properties and skin permeation of Span 60/Tween 60 niosomes of ellagic acid. Int. J. Pharm.,
423:303– 311.
[24] Liu T., Guo R. (2005). Preparation of a highly stable niosome and its hydrotrope-
solubilization action to drugs. Langmuir, 21:11034-11039.
[25] Priprem A., Watanatorn J., Sutthiparinyanont S., Phachonpai W., Muchimapura S. (2018).
Anxiety and cognitive effects of quercetin liposomes in rats. Nanomed. Nanotechnol., 4:70-78.
[26] Yang T., Cui F.-D., Choi M.-K., Lin H., Chung S.-J., Shim C.-K., Kim D.-D. (2007).
Liposome formulation of paclitaxel with enhanced solubility and stability. Drug Delivery, 14:301–
308.
[28] Bochicchio S., Barba A.A., Grassi G., Lamberti G. (2016). Vitamin delivery: Carriers based
on nanoliposomes produced via ultrasonic irradiation. LWT - Food Science and Technology, 69:9-16.
[30] Ruysschaert T., Marque A., Duteyrat J.-L., Lesieur S., Winterhalter M., D. Fournier.
(2005). Liposome retention in size exclusion chromatography. BMC Biotechnology, 5(11).
[31] Demetzos C. (2008). Differential Scanning Calorimetry (DSC): A tool to study the thermal
behavior of lipid bilayers and liposomal stability. J. Liposome Res., 18:159–173.
220
9. Selected tetraspanins functionalized
niosomes as potential standards for exosomes
detection based on immunoassays
Capítulo/Chapter 9
Selected tetraspanins functionalized niosomes
as potential standards for exosomes detection
based on immunoassays
9.1. Introduction
223
biosensing of EVs. Regarding the molecules, tetraspanins CD9, CD63 or CD81 are
commonly used as membrane markers, since EVs are enriched in these transmembrane
proteins. 15
During the development and validation of a new analytical tool or method, the use of
standards is essential for the development stage, but also to characterized their
performance, possibilities and potential limitations. Traditionally, for EVs quantification,
suspensions enriched in exosomes isolated from body fluids or cell cultures media have
been used for these purpose. However, the absence of a well characterized and validated
strategy for EVs isolation makes difficult the existence of a robust RM for the evaluation
and performance of methods inter-comparison studies. The co-isolation of other biological
entities (mainly protein aggregates), and the isolation of heterogeneneous EV fractions in
terms of size (i.e., exosomes or microvesicles) or sub-types (i.e., exosome populations from
different cell lines) are some of the most commonly limitations of isolation procedures for
the establishment of EVs-based RM.8
Scientists have focused the attention on mimetic particles and their used in EVs-related
research. 16,17 Synthetic vesicular systems, such as liposomes or niosomes (prepared from
lipids and non-ionic surfactants, respectively) have been postulated as powerful tools due
to their similarities in terms of morphology and chemical behavior to natural EVs. Some
works have explored the applications of synthetic vesicles for the study of EVs biology. 18
On the other hand, some studies have applied these particles for exosomes modifications
for new intended purposes, mainly for diagnostic and therapy. 19
Also, these vesicles have been used in methods comparison studies. For example, Lane
et al. 20 compared different exosome purification methods using a model liposome system,
and they concluded that the studied purification methods (ultracentrifugation, two
sedimentation reagents, a density gradient method, and the ExoSpin exosome purification
system) possessed different efficiencies, but keep constant vesicles size and size
distribution from real sample. Maas et al. 21 used liposomes to compare methods based on
single-particle analysis (nanoparticle tracking analysis or NTA, tunable resistive pulse
sensing or tRPS and high-resolution flow cytometry or hFC) for EVs quantification, and
found absolute quantification differences between techniques and between synthetic
224
counterparts and natural EVs. Interestingly, also some differences were observed for
liposomes with different size. However, the use of this synthetic vesicles for
methodological comparisons is limited to their physical properties and not to functional
characteristics such as the presence of specific molecular markers. This is important, since
some physical properties such as size and monodispersity can influence the outcome of
the analysis by limiting the sensitivity for smaller particle detection and a bias into the
quantification. The introduction of molecular recognition coupled to a proper size, could
create a robust RM that allows to expand the range of techniques to be tested, but also
introduce new possibilities of information acquisition for deep inter-comparison studies.
During the last years, several technologies have raised in order to overcome the
limitations for the use of natural exosomes in biomedical applications, and the so called
artificial exosomes have emerged with a full range of possibilities and capabilities for
diagnosis and therapy. 22,23 However, their use as true standards for analytical purposes
have remained unexplored, and only some probes of concept have been developed. 24,25
The aim of the present work was the design, development and functional
characterization of a potential standard of EVs (RM), more precisely exosomes, for their
application in immunoassay-based methods. Figure 9.1 capture the molecular composition
and structure of our proposal. Recombinant constructions (large extracellular loops, LEL)
of tetraspanins CD63 and CD9 were bioconjugated to the external surface of niosomes
prepared with a size distribution similar to natural exosomes. Mono- (CD9 or CD63) and
double (CD9 and CD63) functionalized particles have been prepared and tested in an
ELISA assay. Their potential as RM for EVs bioanalytics is evaluated, and a versatile
strategy for their customized production is presented.
225
Figure 9.1. Schematic fully artificial exosome produced by bottom-up bio-nanotechnological methods based on
supramolecular chemistry. The different molecular components are detailed with their functions.
HiTrap columns packed with Sephadex G-25 (5 mL bed volume) and Sepharose CL-
2B/CL-4B gel filtration media were acquired from GE Healthcare Life Sciences
(Pittsburgh, USA).
226
Biotinylated and purified monoclonal antibodies against CD9 (VJ1/20) and CD63
(Tea3/18) were acquired from Immunostep (Salamanca, Spain). Polyclonal antibody
against CD9 was from Santa Cruz Biotechnology (Dalas, TX, USA) and for CD63 was
acquired from Sigma Aldrich. Secondary antibodies HRP-conjugated, and streptavidin-
HRP were acquired from Thermo Scientific.
227
a total sample volume of 2.5 mL was reached by addition of ultrapure water. The solution
was kept at constant mechanical agitation in a vortex for 2 h. To quench possible activated
NHS esters, 1 mg of glycine was added to the suspension.
Removal of unconjugated protein was carried out by gravity elution gel filtration in a
PD-10 empty column packed with Sepharose CL-4B (8.6 mL, bed volume) conditioned
with PBS. A total elution volume of 3.5 mL was recovered in a flow cytometer-grade tube
with sealing cap (BD Plastipak, Vaud, Switzerland), and 0.1% sodium azide in PBS was
used as eluent solution. This concentration was checked to keep vesicles without
modification in colloidal state. A protein quantification kit (based on bicinchoninic acid
assay or BCA, according to manufacturer instruction) was used to determine the elution
profile of a solution of streptavidin to check the suitability of chromatography separation
for purification.
The efficiency of streptavidin conjugation was checked by dot blot, using biotinilated-
HRP enzyme (B-HRP) as protein detection probe and insoluble TMB (suitable for
membranes) as substrate. Briefly, 1 µL of samples (fractions from SEC column) and protein
standards were applied over nitrocellulose membranes (GE Healthcare Life Sciences,
Pittsburgh, USA) and air dried at RT. Membranes were blocked in 5% BSA in PBS-0.05%
Tween® 20 (PBS-T) and then incubated in a 4 µg/mL solution of B-HRP in 0.1% BSA in
PBS-T for 45 min. Membranes were washed and incubated with TMB at variable times,
monitoring the signal from the highest concentration standard to avoid signal saturation.
228
streptavidin in order to avoid possible unspecific signal from biotinylated antibodies used
in ELISA assays.
In the case of double functionalized vesicles, 300 µL of LEL_CD63 was added, while
the amount of LEL_CD9 was reduced to 150 µL according to a previous report showing
that their production yield is approximately the double of CD63.25 This was intended to
keep the ratio of LEL types to 1:1 molar ratio.
In order to remove unbound LEL, sepharose CL-2B columns (10 mL bed volume) were
prepared in plastic syringes (BD Plastipak) with a nylon filter to retain the gel into the
column. A 3 way stopcock (BD Plastipak) was attached to column outlet to control the
elution flow. After equilibration of the column with filtered PBS, the total amount of
vesicles suspension plus LEL was added and a total of 20 fractions (0.5 mL) were collected
into glass vials, and stored at 4 °C.
ELISA assays were performed in 96-well plates (Corning, Corning, NY, USA).
Microplate wells were coated at 4 °C overnight with monoclonal antibodies (10 µg/mL, in
borate buffer saline or BBS buffer, 10 mM and pH 8.2), and blocked with BSA 2% in PBS
for 2h at 37 °C. Samples (100 µL/well) were incubated at 4 °C overnight. Detection was
performed using biotinylated monoclonal antibodies (12.5 µg/mL in PBS) and polyclonal
antibodies (1:250 and 1:500 for anti-CD9 and anti-CD63, respectively) incubated for 1h at
37 °C. Streptavidin-HRP (1:2000) and anti-rabbit IgG-HRP (1:3000) was used as secondary
detection probes. The reaction was developed with o-phenylenediamine dihidrochloride
(OPD, Sigma Aldrich) substrate for colorimetric detection, and signal intensity was
measured at 492 nm in a microplate reader (Tecan Genios, Tecan Trading AG,
229
Switzerland) after addition of stop solution. Washing steps were performed with PBS-T
between incubation steps, and PBS prior to the addition of OPD.
The strategies for the development of artificial exosomas have been reviewed in a
previous publication of our group.23 Their biochemical composition should provide them
with similar physical, optical and functional characteristics to natural EVs, providing a
new range of RM for different isolation and detections strategies. In this work, we have
followed a strategy based on bio-nanotechnology and supramolecular chemistry to create
a synthetic bilayer made of non-ionic surfactants and additives (niosomes) that was then
functionalized with proteins typical of exosomes, against specific receptors are directed in
assays based of molecular recognition such as immunoassays.
In a deep study (data not published) we observed that higher sonication amplitude
needed for size reduction really decrease the life time of the probe, and TiNPs could be
observed by TEM in the prepared suspensions. Also, the ageing of the probe introduce not
controlled alterations in the process, and becomes not reproducible as sonication probe
ages. So, it was decided to avoid the use of sonication, and his influence was compensated
by reasonably changing the value of the other two factors. Concentration of membrane
components in the organic phase was reduced to the half, while its volume was kept at 20
230
mL. In order to compensate the dilution of the prepared nanovesicles solution, we used
evaporation under vacuum (50 °C, 45 bar) to reduce the final aqueous suspension down to
25 mL instead of 50 mL.
With these modifications of initial conditions, niosomes with 150 ± 3 nm (PDI 0.060)
were obtained. For bioconjugation purposes, cholesterol-hemisuccinate was added. This
additive not only introduces surface available carboxylic groups as anchor elements, but it
also provides negative charges that enhance the stability of bared niosomes during
storage. The addition of cholesterol-hemisuccinate did not modify the average size of the
vesicles in comparison with formulations lacking cholesterol-hemisuccinate.
Once we got the optimal suspension of niosomes in terms of size, monodispersity and
particle concentration, protein (streptavidin) could be conjugated with the niosomes at the
external surface to create a generic platform for the development of different types of
artificial EVs. The carbodiimide-based bioconjugation strategy is a two-step procedure to
permanently link two biomolecules, or a molecule with a surface or a nanomaterial [ 28].
This method creates a covalent bond between an amine and an activated carboxylic group
by the use of EDC and NHS. In this particular case, between amine groups from basic
aminoacids in the streptavidin molecule and the carboxylic group from cholesterol
hemisuccinate introduce into niosomal formulation as additive.
This strategy has been previously followed for the conjugation of biomolecules with
nanovesicles, and some examples can be found in the literature. 29,30 Interestingly,
carbodiimide method has been applied in the development of artificial exosome for
therapy. 31 The process is carried out in two steps in order to avoid protein aggregation
induced by cross-linked molecules, since proteins exhibit both functional groups, the
amine and the carboxylic. The pH value of each step (acid for the first, neutral to basic for
the second) was carefully adjusted in order to maximize the coupling process. Two
different amounts of protein were tested in order to check variable dependence over
bioconjugation yield. However, no differences were observed, and the lower amount was
selected in order to keep the process cost-effective.
The elution profile from the SEC column of a solution of streptavidin is shown in
Figure 9.2.A. The elution is enough delayed from the death volume, where nanovesicles
231
eluted, checked by passing through the column a suspension of NVs with a loaded red
dye for visual purposes (Figure 9.2.A, detail).
Figure 9.2. Figure 2. (A) Elution profile of a solution of streptavidin from a Sepharose CL-4B SEC gravity
elution column. Signal quantification of each 0.5Ml fraction was measured by BCA total protein assay
according with manufacturer instruction. Graph insight shows (left) the first 3.5 mL collected of a suspension
of red dye loaded niosomes to allow their visualization, after their elution from the SEC column; (right) the
SEC column after the elution of the 3.5 mL of dyed niosomes. Both elements, niosomes and free protein, eluted
from the column enough separate to allow their separation based on Sepharose CL-4B gravity elution
coloumns. (B) Dot-Blot assay for checking the effectively of streptavidin bioconjugation to niosomes through
carbodiimide method (EDC/NHS). Standards of different concentrations allows the semiquantification of the
process by comparison of spot intensity. The result shows the 5 different batches. Biotinylated-HRP (4µg/mL)
was used as detecting agent. (C) Size distribution by DLS of bared niosomes (or NIO) and streptavidin-
conjugated niosomes or (NIO_Str). The average size increment (2nm) shown as peak displacement
demonstrate the effective conjugation of the protein. (D) Size distribution by DLS of the 5 different batches of
NIO_Str (152 ± 3 nm), to demonstrate the reproducibility of the process.
232
All the batches were characterized in terms particle size and size distribution (Figure
9.2.D), showing a good reproducibility, with an average value of 152 ± 2 nm.
After incubation, excess of LELs was removed by gel filtration (Sepharose CL-2B), and
several fractions were collected into glass vials. The fractions were analyzed by dot blot for
immunodetection of the LEL. Figure 9.3.A shows the detection of LEL_CD9 and
LEL_CD63 constructions in both mono- and double-functionalized niosomes, and
confirms the single and co-functionalization with LELs. The first five fractions correspond
to the void volume of the column and no signal was detected. A progressive increment in
the signal was observed in fractions 5/6 to 9/10, which correspond to those that showed
the characteristics pale white color of the vesicles in suspension. Then, a reduction in
signal is observed prior to another increment in the signal corresponding to the elution of
free LEL that is used in excess.
233
Both characteristics (color ad signal) were taken into consideration, and the 5 fractions
showing higher signals were pooled, so the final volume achieved of Nio_LEL was 2.5 mL
for each type of modified niosomes. Signal differences between CD63 and CD9 for double
functionalized vesicles are due to differences in exposure time. It was needed longer
exposures for CD63 detection. Nio_LEL particles were produced in the range of 5.2 x 1011 –
1.0 x 1012 particles/mL, measured by NTA.
Nio_LEL were also characterized by NTA to check particle average size and size
distribution, and a sample of natural exosomes was also measured for comparative
purposes. Besides differences in terms of peaks intensities across the distributions (Figure
9.3.B), average size of Nio_LEL9, Nio_LEL63, Nio_LEL9/63 and natural exosomas are
similar (185, 160, 159 and 162 nm respectively), and the size distribution limits are similar
for all the particles. However, artificial vesicles are more homogeneous (as expected) since
they are lab-made products, while natural exosomas are more heterogeneous regarding
their natural origin. Mono- and double-functionalized particles remains similar between
types of particles.
The next step was to test the recognition of the Nio_LEL by their specific anti-
tetraspanin antibody and the possible unspecific signal from the other anti-tetraspanin.
Negative controls were introduced (bared niosomes, and niosomes functionalized with
Streptavidin with/without biotin saturation), and a sample of natural exosomes
(mesenchymal triple-negative breast cancer cell line SUM159) was used as positive control.
Both polyclonal and monoclonal antibodies (biotinylated or purified) were tested.
Effective molecular recognition was carried out by dot blot assay as performed to check
niosomes-LEL functionalization. This rapid and simple technique is suitable for screening
of antibodies. 33
234
Figure 9.3. (A) Dot-blot assays for revelation of LEL, CD9 or CD63, positive fractions collected from a
Sepharose CL-2B gravity elution SEC column. Mon- and double-functionalized Nio_Str have been produced.
(B) Size distribution measured by NTA (Nanosight, Malvern Instruments) of previous described fully
artificial exosomas (NIO_LEL). A sample of natural exosomas isolated from mesenchymal triple-negative
breast cancer cell line SUM159 was also measured for comparison purposes.
The results of the different assays carried out showed that the best detection was
obtained with the polyclonal antibodies (Figure 9.4), in terms of specificity (referred to
Nio_LEL recognition and discrimination). Those antibodies exhibit some unspecific
recognition for the other tetraspanin, but less intense than the observed for biotinylated
monoclonal antibodies. In addition, the signal from negative controls was more intense for
235
both types of monoclonal antibodies. However, between them, purified ones offered better
results. Based on these observations, capture by purified monoclonal antibodies and
detection using polyclonal antibodies were selected as configuration for a sandwich-based
ELISA experiments to detect artificial, according to a classical configuration for this type of
immunoassays. 34
Figure 9.4. Dot-blot assay for the screening and selection of α-tetraspanin antibodies for their future use in
ELISAs for the detection of fully artificial exosomes (NIO_LEL). Secondary antibodies labelled with HRP
were appropriate selected. Different negative controls were also introduced (bared niosomes, niosomes
functionalized with streptavidin with and without biotin saturation), as the use of isolated exosomas from
mesenchymal triple-negative breast cancer cell line SUM159 as positive control. Values are uniplicate assays.
(PU) monoclonal purified antibody; (biot) Biotynilated monoclonal antibody; (poli) polyclonal antibody.
To test the potential use of monofunctionalized Nio_LEL as RM, ELISA assays were
carried out using different combinations of capture/detection antibodies for both types of
vesicles (Nio_LEL9 and Nio_LEL63). This proposed ELISA assay with colorimetric
detection used monoclonal antibodies for capture and polyclonal antibodies for primary
detection, with the use of adequate HRP-labeled secondary. The combination of the proper
236
detection and capture antibodies is crucial in the development of ELISA assays, especially
in terms of signal-to-noise ratio and specificity. Multiple antibodies combinations were
tested, and their unspecific signal were also studied.
As seen in Figure 9.5.B, a better signal was observed for all the cases were anti-CD9
was used for detection, even when the assayed particles were Nio_LEL63, or in the case of
Nio_LEL9 captured by anti-CD63. These two conditions offer then the unspecific signal for
detection, clearly lower than the specific signal, and in accordance with dot-blot results.
In the case of detection based on anti-CD63, when capture by anti-CD9, this detection
can discern (p˂0.005) between types of Nio_LEL with a really low sensibility, and coupled
to capture also by anti-CD63 is the only combination that cannot differentiate Nio_LEL
type.
237
Figure 9.5. (A,B) Cross-reactive responses for mono-functionalized LEL-tetraspanin niosomes (fully articial
exosomas) for different configuration of capture (c) and detection (d) anti-tetraspanin antibodies. (C) ELISA
assays for the detection of NIO_LEL9 (N9) and NIO_LEL63 (N63) fully artificial exosomes using different
combination of capture (c) and detection (d) antibodies. Monoclonal α-CD9 or α-CD63 antibodies were used
for capture, whereas polyclonal α-CD9 or α-CD63 antibodies were used for detection. (D) ELISA assays for
the detection of NIO_LEL9/63 using different combinations of capture (c) and detection (d) antibodies. The
graphs shows the mean ± SD of 3 independent experiments. *** p˂0.005, Student’s t-test.
238
9.3.3.2. Double tetraspanin functional particles
However, single molecule detection is often used, but the simultaneous detection of
two different tetraspanins (at least one of them must be CD63, being the others CD9 or
CD81) is accepted as a molecular identification of the endosomal origin of the exosomes,
and can be used as molecular markers in exosomes detection based on immunoassays. 35
For this purpose, double functionalized niosomes (Nio_LEL9/63) were produced and
tested by the same antibodies combination in order to identify which one offers better
sensibility.
In a previous work of our group, 36 an ELISA procedure was developed for the
detection of exosomes based on the simultaneous recognition of two tetraspanin (one used
for capture, and the other for detection). For this purpose, melanoma cell line (Ma-Mel-
86c) derived exosomes isolated by ultracentrifugation were used, and different
combination of antibodies recognizing CD9, CD63, and CD81 were tested. Results
revealed that capture by CD81 and detection carried out by CD9 presented the best signal-
to-noise ratio. However, the same exosomes were better detected in a Lateral Flow
Immunoassay (LFIA) using CD9 for capture and CD63 for detection, besides this
combination offered a higher background signal in ELISA compared to CD81/CD9 for
capture/detection respectively.
In this type of analytical platforms, only Nio_LEL9/63 could be tested, since mono-
fucntionalized particles are not suitable due to how it works a LFIA test. However, the
best capture-detection pair described for this systems is in our case the less sensible one, so
our proposal of RM based on artificial exomes is adequate for ELISA and could not be for
LFIA.
239
Finally, only in the combinations where anti-CD9 is used as detection, the sensibility
was enough to allow the visualization of dose-response signals (Figure 9.6). This result is
the manifestation of those observed in figure 34.D, where it can be seen that detection
based on CD63 is not a good option. In all the cases, a linear correlation is observed. When
the signal intensity proportioned by a specific antibody configuration was enough to allow
visualization of dose-response, this response was fitted to a linear equation, demonstrating
that working condition where into the linear range of the typical sigmoidal response
related to a sandwich assay, which confirms that our RM proposal is suitable.
Figure 9.6. Dose-response graphs for different types of fully artificial exosomas detected by ELISA assay
using monoclonal and polyclonal antibodies α-tetraspanins CD9 y CD63 for capture and detection
respectively. As secondary appropriate α-IgG-HRP was used. N9 and N9/63 are NIO_LEL9 and
NIO_LEL9/63 respectively, which are mono- and double-functionalized LEL-tetraspanin niosomes.
The work of Lane et al.20 has highlighted the physical similarities between synthetic
vesicles and EVs, and those particles have been used as reference materials for
methodological comparisons (NTA, tRPS and hFC). However, all those methods are
classified as unspecific concentration determination methods 37 since they rely on general
physical characteristics and not in a specific molecular marker which allows also
phenotyping possibilities. In this scientific challenge we propose fully artificial exosomes22
as a new potential tool to help into the development and validation of new analytical
methods and platforms, as demonstrated with our results. In our opinion, our proposed
RM could be competitive to those in the market up to date.
240
Tabla 9.1. Some commercial available kits based on ELISA for exosomes quantification in biological samples.
Most of them are based on colorimetric signal quantification based on HRP substrates, with a typical format
of 96 well microtiter plate.
ExoQuant Centaur CD9 for detection Sandwich assay using pan- Lyophilized
Genprice Exosome biomarkers (data not Exosomes
specify by the manufacturer
ExoEL- BioVision CD9 for detection Sandwich assay using pan- Exosome
CD81A1 Exosome biomarkers (data not lyophilized
specify by the manufacturer
CD9/CD63 Cosmo BIO CD63 for Sandwich assay using CD9 for CD9/63 Fusion
Exosome CO. Ltd detection capture protein
ELISA Kit
ExoAssayTM CD Creative Not specified by Sandwich assay using CD9 for Lyophilized
®
Diagnostics the manufacturer capture Exosomes
Commercially available kits based on ELISA assays are marketed with different
configurations (Tabla 9.1). Some of them are based on direct capture of exosomes into
plate wells (such as ExoELISA-(Ultra), SBI System Biosciences), while other relies on
exosome-capture mediated by antibodies-coated weels (such as ExoTESTTM,
HansaBioMed). Majority of them, use lyophilized exosomes as standards, and some
241
manufactures let clear that signal can be different depending on the amount of protein per
vesicle between different types of exosomes, with potential bias of extrapolated
concentration. This fact hinders the quantification of these types of analytes, since the
same intensity could be related to a higher concentration of analytes with a reduced
expression level of detection antigen, or a low concentration of analytes carrying a higher
number of detection epitopes. This is a key point to take into account for analytes such as
exosomas, where the expression levels of CD9 and CD63 differs between cell lines, 38 or
even exosomas from cells with different heath status. This is something that clearly
difficult the development of a universal standard for EVs quantification, especially for
exosomes, as mentioned previously.
In this diverse market our proposal can be compatible, since both types of Nio_LEL
(mono- and double-functionalize) could be potentially applied. On the other hand, our
platform is really versatile, since antigen density can be tuned by changing the density of
streptavidin over their surface, and any biotinylated peptide could be used to
functionalize, giving lots of options to create specific types of artificial exosomas.
Complementary studies about measurements of effective concentration of LEL in the NVs
and their stoichiometry in the case of multiple protein functionalization) will re-enforce
the results presented here. Also, the application of calibration curves obtained with this
innovative RM to different real sample will help to consolidate our model.
The results demonstrate the potential use of this new biomaterial as analytical standard
for molecular recognition based assays, such as immunoassays or aptamer-based assays.
The development of artificial exosomes based on tetraspanin (recombinant LEL) surfaced
functionalized niosomes have been described. The production of mono- and double-
functionalized vesicles with CD9 and/or CD63 is possible, and these particles can be
detected and discerned by sandwich ELISAs, using a classical format based on capture
through monoclonal antibodies and detection based on polyclonal antibodies with
secondary enzyme-labeled antibodies. Dose-response of this particles has been checked,
since a linear fitted response is essential for their use as standard for obtaining calibration
plots used for quantification purposes.
242
The methodology proposed in this study allows the preparation with a tunable
functionalization based on changes in density surface functionalization (by variation in
cholesterol hemisuccinate molar ratio) or changes in the stoichiometry of proteins (by
variation in molar ratio of proteins during coupling to streptavidin-coated niosomes). The
exploration of this variables could be an interesting starting point for future works. Also,
further validation studies must be performed, in order to test their usability for different
cell-line derived exosome quantification. Additional proteins, different than classical
tetraspanins, could be used for the development of pathology-specific standards.
[1] Yáñez-Mó M., Siljander PR., Andreu Z., Zavec AB, Borràs FE, Buzas EI, Buzas K, Casal E,
Cappello F, Carvalho J, Colás E, Cordeiro-da Silva A, Fais S, Falcon-Perez JM, Ghobrial IM, Giebel
B, Gimona M, Graner M, Gursel I, Gursel M, Heegaard NH, Hendrix A, Kierulf P, Kokubun K,
Kosanovic M, Kralj-Iglic V, Krämer-Albers EM, Laitinen S, Lässer C, Lener T, Ligeti E, Linē A,
Lipps G, Llorente A, Lötvall J, Manček-Keber M, Marcilla A, Mittelbrunn M, Nazarenko I, Nolte-'t
Hoen EN, Nyman TA, O'Driscoll L, Olivan M, Oliveira C, Pállinger É, Del Portillo HA, Reventós J,
Rigau M, Rohde E, Sammar M, Sánchez-Madrid F, Santarém N, Schallmoser K, Ostenfeld MS,
Stoorvogel W, Stukelj R, Van der Grein SG, Vasconcelos MH, Wauben MH, De Wever O. Biological
properties of extracellular vesicles and their physiological functions. J Extracell Vesicles. 2015 May
14;4:27066
[2] Kalra, H., Drummen, G., Mathivanan, S. (2016). Focus on extracellular vesicles: introducing
the next small big thing. International journal of molecular sciences, 17(2), 170.
243
[3] Azmi, A. S., Bao, B., & Sarkar, F. H. (2013). Exosomes in cancer development, metastasis, and
drug resistance: a comprehensive review. Cancer and Metastasis Reviews, 32(3-4), 623-642.
[4] Corrado, C., Raimondo, S., Chiesi, A., Ciccia, F., De Leo, G., & Alessandro, R. (2013).
Exosomes as intercellular signaling organelles involved in health and disease: basic science and
clinical applications. International journal of molecular sciences, 14(3), 5338-5366.
[5] Azmi A.S., Bao B., Sarkar F.H. (2013). Exosomes in cancer development, metastasis, and
drug resistance: a comprehensive review. Cancer Metastasis Rev., 32(3-4), 623-642.
[6] Cappello, F., Logozzi, M., Campanella, C., Bavisotto, C. C., Marcilla, A., Properzi, F., Fais, S.
(2017). Exosome levels in human body fluids: a tumor marker by themselves?. European Journal of
Pharmaceutical Sciences, 96, 93-98.
[7] van der Meel R., Krawczyk‐Durka M., van Solinge W.W., Schiffelers R.M. (2014). Toward
routine detection of extracellular vesicles in clinical samples. Int J Lab Hematol., 36:244-253.
[8] Boriachek, K., Islam, M. N., Möller, A., Salomon, C., Nguyen, N. T., Hossain, M. S. A.,
Yamauchi, Y., Shiddiky, M. J. (2018). Biological functions and current advances in isolation and
detection strategies for exosome nanovesicles. Small, 14(6), 1702153.
[9] Peterson, M. F., Otoc, N., Sethi, J. K., Gupta, A., & Antes, T. J. (2015). Integrated systems for
exosome investigation. Methods, 87, 31-45.
[10] Van Der Pol, E., Hoekstra, A. G., Sturk, A., Otto, C., Van Leeuwen, T. G., & Nieuwland, R.
(2010). Optical and non‐optical methods for detection and characterization of microparticles and
exosomes. Journal of Thrombosis and Haemostasis, 8(12), 2596-2607.
[11] Rupert, D. L., Lässer, C., Eldh, M., Block, S., Zhdanov, V. P., Lotvall, J. O., Bally, M., Höök,
F. (2014). Determination of exosome concentration in solution using surface plasmon resonance
spectroscopy. Analytical chemistry, 86(12), 5929-5936.
[12] Boriachek, K., Islam, M. N., Gopalan, V., Lam, A. K., Nguyen, N. T., Shiddiky, M. J. (2017).
Quantum dot-based sensitive detection of disease specific exosome in serum. Analyst, 142(12),
2211-2219.
244
[13] Doldán, X., Fagúndez, P., Cayota, A., Laíz, J., Tosar, J. P. (2016). Electrochemical sandwich
immunosensor for determination of exosomes based on surface marker-mediated signal
amplification. Analytical chemistry, 88(21), 10466-10473.
[14] Zhou, Q., Rahimian, A., Son, K., Shin, D. S., Patel, T., & Revzin, A. (2016). Development of
an aptasensor for electrochemical detection of exosomes. Methods, 97, 88-93.
[15] López-Cobo, S., Campos-Silva, C., Moyano, A., Oliveira-Rodríguez, M., Paschen, A., Yáñez-
Mó, M., Blanco-López, M.C., Valés-Gómez, M. (2018). Immunoassays for scarce tumour-antigens in
exosomes: detection of the human NKG2D-Ligand, MICA, in tetraspanin-containing nanovesicles
from melanoma. Journal of nanobiotechnology, 16(1), 47.
[16] Jang SC, Kim OY, Yoon CM, et al. Bioinspired exosome-mimetic nanovesicles for targeted
delivery of chemotherapeutics to malignant tumors. ACS Nano. 2013;7:7698–7710.
[17] Jeong D, Jo W, Yoon J, et al. Nanovesicles engineered from ES cells for enhanced cell
proliferation. Biomaterials. 2014;35:9302–9310.
[18] Hood J.L., Scott M.J., Wickline S.A. (2014). Maximizing exosome colloidal stability
following electroporation. Analytical Bichemistry, 448:41-49.
[19] Lee J., Kim J., Jeong M., Lee H., Goh U., Kim H, Kim B., Park J-H. (2015) Liposome-based
engineering of cells to package hydrophobic compounds in membrane vesicles for tumor
penetration. Nano Letters, 15:2938-2944.
[20] Lane, R. E., Korbie, D., Anderson, W., Vaidyanathan, R., & Trau, M. (2015). Analysis of
exosome purification methods using a model liposome system and tunable-resistive pulse
sensing. Scientific reports, 5, 7639.
[21] Maas, S. L., De Vrij, J., Van Der Vlist, E. J., Geragousian, B., Van Bloois, L., Mastrobattista,
E., Schiffelers, R.M., Wauben, M.H.M., Broekman; M.L.D., Nolte, E. N. (2015). Possibilities and
limitations of current technologies for quantification of biological extracellular vesicles and
synthetic mimics. Journal of Controlled Release, 200, 87-96.
[22] García-Manrique, P., Gutiérrez, G., & Blanco-López, M. C. (2018). Fully artificial exosomes:
towards new theranostic biomaterials. Trends in biotechnology, 36(1), 10-14.
245
[23] García-Manrique, P., Matos, M., Gutiérrez, G., Pazos, C., & Blanco-López, M. C. (2018).
Therapeutic biomaterials based on extracellular vesicles: classification of bio-engineering and
mimetic preparation routes. Journal of extracellular vesicles, 7(1), 1422676.
[24] García-Manrique P., Lozano-Andrés E., Estupiñán-Sánchez O.R., Gutiérrez G., Matos M.,
Pazos C., Yañez-Mo M., and Blanco-López C., Biomimetic small extracelular vesicles, 3rd GEIVEX
Simposium, San Sebastian, Spain, 29-30 September 2016. Poster communication.
[25] Lozano-Andrés, E., Libregts, S. F., Toribio, V., Royo, F., Morales, S., López-Martín, S., Valés-
Gómez, M., Reyburn, H.T., Falcón-Pérez, J.M., Wauben, M.H., Soto, M., Yáñez-Mó, M. (2019).
Tetraspanin-decorated extracellular vesicle-mimetics as a novel adaptable reference
material. Journal of Extracellular Vesicles, 8(1), 1573052.
[26] García-Manrique, P., Matos, M., Gutiérrez, G., Estupiñán, O. R., Blanco-López, M. C., &
Pazos, C. (2016). Using factorial experimental design to prepare size-tuned nanovesicles. Industrial
& Engineering Chemistry Research, 55(34), 9164-9175.
[27] Introduction to Bioconjugation, Chapter 1, and The Chemistry of Reactive Groups, Chapter
2. In Bioconjugate techniques, Edited by Hermanson G.T. Academic press, Londres, 1-125, 127-228.
[29] Endoh H., Suzuki Y., Hashimoto Y. (1981). Antibody coating of liposomes with 1-ethyl-3-(3-
dimethyl-aminopropyl)carbodiimide and the effect on target specificity. Journal of Immunological
Methods, 44:79-85.
[30] Tan D.M-Y, Fu J-Y, Fu-Shun Wong F-S, Er H-M, Chen Y-S, Nesaretnam K. (2017). Tumor
regression and modulation of gene expression via tumor-targeted tocotrienol niosomes.
Nanomedicine, 12(20):2487-2502.
[31] Li K., Chang S., Wang Z., Zhao X., Chen D. (2015). A novel micro-emulsion and micelle
assembling method to prepare DEC205 monoclonal antibody coupled cationic nanoliposomes for
simulating exosomas to target dendritic cells. International journal of Pharmaceutics, 491:105-112.
[32] Jans, H., X. Liu, Austin L, Maes G, Huo Q. (2009). Dynamic Light Scattering as a Powerful
Tool for Gold Nanoparticle Bioconjugation and Biomolecular Binding Studies." Analytical
Chemistry 81(22): 9425-9432.
246
[33] Renart J., Behrens M.M., Fernández-Renart M., Martínez J.L., Immunoblotting Techniques,
Chapter 23, in Immunoassay, edited by Christopoulos T.K. and Diamandis E.P, Academic Press
Inc., San Diego, California, USA.
[34] Christopoulos T.K. and Diamandis E.P., Immunoassay configurations, Chapter 10, in
Immunoassay, edited by Christopoulos T.K. and Diamandis E.P, Academic Press Inc., San Diego,
California, USA.
[35] Kowal, J., Arras, G., Colombo, M., Jouve, M., Morath, J. P., Primdal-Bengtson, B., Florent
Dingli, F., Loew, D., Tkach, M., Théry, C. (2016). Proteomic comparison defines novel markers to
characterize heterogeneous populations of extracellular vesicle subtypes. Proceedings of the
National Academy of Sciences, 113(8), E968-E977.
[36] Oliveira-Rodríguez, M., López-Cobo, S., Reyburn, H. T., Costa-García, A., López-Martín, S.,
Yáñez-Mó, M., Cernuda-Morollón, E., Paschen, A., Valés-Gómez, M., Blanco-López, M. C. (2016).
Development of a rapid lateral flow immunoassay test for detection of exosomes previously
enriched from cell culture medium and body fluids. Journal of extracellular vesicles, 5(1), 31803.
[37] Rupert, D. L., Claudio, V., Lässer, C., & Bally, M. (2017). Methods for the physical
characterization and quantification of extracellular vesicles in biological samples. Biochimica et
Biophysica Acta (BBA)-General Subjects, 1861(1), 3164-3179.
247
10. Conclusiones/Conclusions
Capítulo 10. Conclusiones
251
ordenador. Estas diferencias podrían atribuirse a parámetros operacionales de
las impresoras, y deben ser estudiados más a fondo en futuros trabajos.
252
Por otra parte, se han descrito efectos particulares del peso molecular de los
compuestos a encapsular tanto sobre el tamaño de partícula como sobre la EE
de dichos compuestos en las vesículas formuladas.
253
mediante immunoensayos sándwich de tipo ELISA. A su vez, la señal
analítica responde a la concentración de dichas partículas, lo que en conjunto
las hace candidatas idóneas a ser empleadas como estándares analíticos en
este tipo de métodos de detección basados en reconocimiento molecular.
254
Chapter 10. Conclusions
The general conclusion that can be drawn from the results obtained throughout the
work carried out in this Ph.D. Thesis is the possibility to carry out a correct control
over the production of nanovesicles with controlled size and monodisperse size
distribution. On the other hand, the encapsulation of hydrophilic compounds and their
relation to particle size is improved. In addition, it is demonstrated their potential for
the development of biomimetic models of exosomes, a natural type of extracellular
vesicles with high clinical and biological interest, and its use as an analytical standard.
This conclusion is based on the following points:
• The study of the effect of the variables of a classical method, such as Ethanol
Injection Method or EIM, on the final properties of the particles (liposomes
and niosomes), specifically size and monodispersity was proposed. The
adequate approach of the DoE has allowed reducing from ten initial
methodological variables with potential effect on the responses to only three
(organic/aqueous phase volume ratio, membrane precursors final
concentration in the aqueous phase, and sonication amplitude), which are
those with a higher effect. Mathematical models base don polynomic
equations have been setted for the prediction of particle size and
monodispersity (PDI) according with the methodological parameters values.
255
be attributed to the operational parameters of the printers and should be
further studied in future works.
The results of this study have demonstrated the suitability of these devices for
the preparation of nanovesicles. The development of a temperature control
system adapted to the microfluidic device has allowed the use of non-ionic
surfactants with a high transition temperature (Tm). Those have been
demonstrated as suitables for the formulation of niosomes.
On the other hand, the device design and its coupling to an inverted
microscope have allowed obtaining information on key processes in the
formation of these particles such as molecular diffusion in parallel flow. The
role of temperature on this diffusion and on the principle on which the
microfluidic device is based, the hydrodynamic flow focusing, has also been
reported. In a similar way to the previous study, it has been possible to
establish the importance of the methodological parameters, being the flow rate
ratio of organic:aqueous streams the fundamental parameter to control the
mixing efficiency and the morphological properties of the particles.
256
Using calorimetric techniques such as DSC, it has been possible to verify how
the composition of the hydration medium alters the order and packaging of
the bilayers, which may be responsible for the observations on size and EE in
the study.
Aditional, it has been possible to verify the suitability of dialysis and gel
filtration for the purification of these vesicular systems formulated in viscous
media. A discrepancy between them has been observed for this type of
suspensions, and it has been concluded that very viscous media prevent a
correct chromatographic separation and introduce a size bias in the
population of vesicles.
• Finally, the information obtained from the models for the ethanol injection
method has been used to improve the characteristics of the niosomes
formulated with Span® 60: Cholesterol (1: 0.5 molar ratio). Specifically, the
organic:aqueous phase volume ratio has set high while the concentration of
the membrane components has been reduced. This has allowed obtaining
niosomes with a similar size to that of natural exosomes, with acceptable PDI
values, and all these avoiding sonication processes, since it has been observed
that the aging of the probe introduces uncontrollable variability in the process.
257
However, the design of these particles determines the selection of antibodies
that can be used for their detection and makes necessary the evaluation of
whether such configurations are suitable for biological samples. In addition,
these exosomes must be characterized in depth to know the surface density of
proteins, as well as the real molar ratio between them when they are doubly
functionalized. This parameter is important to explain the difference in
concentration of natural exosomes in a sample that has been obtained by using
a calibration obtained with artificial exosomes and the value obtained by NTA
(Nanoparticle Tracking Analysis). This information will help to improve the
design of this type of biomaterial for analytical purposes, and its adaptation to
the type of natural exosomes that want to mimic since it is known the huge
phenotypic diversity that exists.
258
ANEXO I. Publicaciones derivadas de la Tesis
Doctoral
Article
pubs.acs.org/IECR
closed bilayer structures.13,17,18 As liposomes, their formation However, a large number of variables are involved in this
process is a consequence of unfavorable interactions between modified EIM, and selection of the most important of them
surfactants and water molecules, and they can also entrap (screening) is a crucial step in rationally preparing vesicles by this
hydrophilic, lipophilic, and amphiphilic compounds.19,20 versatile method. In this work, the Z-average size and
Niosomes exhibit a number of advantages over liposomes, polydispersity index (PDI) were selected as the dependent
such as higher stability, easy access to raw materials, lower variables. They are considered to be of great importance in
toxicity, high compatibility with biological systems, non- nanovesicle design because most of the final applications of these
immunogenicity, and versatility for surface modification.20 vesicular systems are directly related to these two parameters.
Cholesterol is commonly used as a membrane additive for Factorial experimental design and the analysis of variance
nanovesicle preparation to improve vesicle stability, entrapment (ANOVA) methodology are appropriate and efficient statistical
efficiency, and release under storage.20 It increases vesicle size tools that permit the effects of several factors that influence
and rigidity, improving encapsulation efficiency, but at high responses to be studied by varying the factors simultaneously in a
concentrations, it can adversely affect the encapsulation rate.21,22 limited number of experiments.
Cholesterol also plays a fundamental role in niosome formation In the recent past, design of experiments (DoE) has been
when hydrophilic surfactants are used (hydrophilic/lipophilic extensively used for the study and optimization of vesicles and
balance of ∼10).20 other similar organic materials. Different designs can be applied
More than 20 different methods have been identified for to reduce the number of factors involved in the preparation
nanovesicle preparation, and these methods were recently techniques28 and, therefore, to minimize the number of
reviewed.23,24 In this work, a modified ethanol injection method experiments without losing valuable information. Plackett−
(EIM) is used, because it offers some advantages over other Burman design is a type of fractional design involving relatively
methods, such as simplicity, absence of potentially harmful few runs,29 commonly used for the screening of variables.
chemicals, and suitability for scaleup.25 Another important role of DoE is in the optimization of
The conventional EIM was first described in 1973.26 In this nanovesicle composition for the enhancement of intended
technique, lipids/surfactants and additives are first dissolved in purposes. For instance, it has been applied to the formulation of
an organic solvent, such as diethyl ether or ethanol, and then liposomes (phospholipid and cholesterol ratio) for the topical
injected slowly through a syringe into an aqueous phase delivery of vitamin E,8 hybrid liposomes (with both low- and
containing the compound of interest. Then, the organic solvent high-transition-temperature phospholipids) to improve the
is removed using a vacuum rotary evaporator. When ethanol is encapsulation and delivery of silymarin,30 and niosomes for
used as the organic solvent, the spontaneous formation of topical delivery applications.10,31 DoE has also been used to
vesicles occurs as soon as the organic solution is in contact with enhance the transdermal flux of raloxifene hydrochloride32 and
the aqueous phase,27 but vigorous agitation is needed to obtain diclofenac diethylamine33 loaded transfersomes and of other
narrow size distributions. For this purpose, a final sonication polymeric nanoparticles encapsulating an anticancer drug.34
stage was applied in this study after organic-phase removal by Moreover, the interactions between vesicles and proteins, such as
vacuum evaporation. pectin, to improve drug-delivery properties has been studied by
9165 DOI: 10.1021/acs.iecr.6b01552
Ind. Eng. Chem. Res. 2016, 55, 9164−9175
Industrial & Engineering Chemistry Research Article
DoE.35 Nanostructured lipid carriers (NLCs) loaded with Table 2. Full Factorial Design (23) with Center-Point
flurbiprofen were also produced under optimal conditions Repetitions (n = 5): Factors, Levels, and Responses
using full factorial design.36
response code meaning
In this work, an initial fractional factorial design with two levels
(Plackett−Burman) was used to screen the most important Y1 Z-average size of PC liposomes
factors in vesicle preparation by the EIM. Then, a 23 two-level full Y2 PDI of PC liposomes
factorial design using center-point replicates was applied to study Y3 Z-average size of S60:Cho niosomes
the influence of the main factors and their interactions on the Z- Y4 PDI of S60:Cho niosomes
average size and PDI. Once the appropriate operating conditions factors
were determined, vesicle stability was studied by using multiple level O/A (X1) C (X2) (g/L) A (X3) (%)
light scattering technology and by measuring the encapsulation −1 (low) 5:50 2 30.0
efficiencies (EEs) of different compounds. 0 (medium) 12.5:50 5 42.5
1 (high) 20:50 8 55.0
2. MATERIALS AND METHODS
2.1. Materials. Phosphatidylcholine (PC) (predominant batch X1 X2 X3 Y1 Y2 Y3 Y4
species C42H80NO8P, MW = 775.04 g/mol) from soybean FF1 1 −1 1 65 0.299 305 0.075
(Phospholipon 90G) was a kind gift from Lipoid (Ludwigshafen, FF2 1 1 −1 97 0.249 362 0.136
Germany). Sorbitan monostearate (Span 60, S60) (C24H46O6, FF3 −1 1 −1 149 0.296 294 0.206
MW = 430.62 g/mol) and cholesterol (Cho) (C27H46O, MW = FF4 −1 1 1 88 0.307 262 0.291
386.65 g/mol) were purchased from Sigma-Aldrich (St. Louis, FF5 −1 −1 1 64 0.342 242 0.120
MO). All membrane components were dissolved in absolute FF6 1 1 −1 100 0.257 360 0.143
ethanol (Sigma-Aldrich, St. Louis, MO). FF7 1 1 1 64 0.272 241 0.182
Methanol, acetonitrile, 2-propanol, and acetic acid of high- FF8 −1 −1 −1 90 0.196 235 0.078
performance liquid chromatography (HPLC) grade were FF9 0 0 0 82 0.219 301 0.195
supplied by Sigma-Aldrich (St. Louis, MO). FF10 1 −1 −1 84 0.205 253 0.032
A phosphate buffer (PB) solution (10 mM, pH 7.4) was used FF11 −1 1 1 107 0.297 276 0.235
in all experiments as the aqueous phase. The buffer solution was FF12 −1 1 −1 156 0.308 275 0.145
prepared in Milli-Q water by dissolving proper amounts of FF13 1 −1 1 65 0.378 248 0.066
sodium dihydrogen phosphate and sodium hydrogen phosphate, FF14 1 −1 −1 97 0.246 268 0.045
supplied by Panreac (Barcelona, Spain). Sodium chloride from FF15 −1 −1 −1 84 0.173 239 0.094
Panreac (Barcelona, Spain) was added to increase the ionic FF16 0 0 0 75 0.224 305 0.253
strength when required according to the experiments listed in FF17 0 0 0 84 0.250 317 0.118
Table 1. For the encapsulation experiments, Fat Red Bluish or FF18 1 1 1 55 0.307 224 0.203
Sudan Red 7B dye (C24H21N5, MW = 379.46 g/mol) and FF19 0 0 −1 77 0.242 308 0.241
cholecalciferol or vitamin D3 (C27H44O, MW = 384.64 g/mol) FF20 0 0 0 84 0.251 337 0.171
were purchased from Sigma-Aldrich (St. Louis, MO). FF21 −1 −1 1 69 0.343 233 0.124
2.2. Factorial Design of Experiments. Factors that could
potentially affect the size of vesicles produced by the EIM were
classified in four groups, according to the different steps involved (version 17) was used for all data analysis. Analysis of variance
in this preparation method: formulation (organic/aqueous phase (ANOVA) was used for this purpose.
volume ratio, phospholipid concentration, and ionic strength), Once the models were obtained taking into account significant
injection (injection flow rate, temperature, and stirring speed), factors and interactions, a set of selected size-tuned vesicles were
evaporation (temperature and rotation speed), and sonication prepared and characterized.
(amplitude and time of sonication). 2.3. Vesicle Preparation. For liposome preparation,
To identify the relative effects of variables on the response, a appropriate weighed amounts of PC were dissolved in different
two-level fractional factorial design was used. A Plackett− volumes of absolute ethanol (5−20 mL range). The same
Burman (P−B) resolution III design with n = 16 runs was procedure was applied to niosome preparation by weighing and
proposed for screening of the initial factors. Two levels were dissolving S60 and Cho in a 1:0.5 weight ratio. Then, the organic
selected for each variable. solution was injected, with a syringe pump (KD Scientific,
Table 1 lists the factors and levels involved in the P−B Holliston, MA) at a flow rate of 120 mL/h, into Milli-Q water
fractional factorial design used, where O/A is the organic/ that was kept at 60 °C and stirred at 500 rpm. Once vesicles
aqueous phase volume ratio, C is the concentration of formed, ethanol was removed at 40 °C under reduced pressure
phospholipid, I is the ionic strength, QV is the injection flow (90 kPa) in a rotary evaporator. The resulting vesicular systems
rate, TI is the injection temperature, NS is the stirring speed were further sonicated for 15 min (CY-500 sonicator, Optic
during injection, TE is the evaporation temperature, NE is the Ivymen System, Biotech SL, Barcelona, Spain), using and
evaporator rotation speed, A is the sonication amplitude, and t is amplitude of 30−55%, a power of 500 W, and a frequency of 20
the sonication time. kHz. The sonication probe was placed in a 100 mL glass beaker at
In a second step, a 23 full factorial design with center-point a constant depth, 1.5 cm above the container bottom.
repetitions (n = 5) was carried out to study the main effects and 2.4. Vesicle Characterization. 2.4.1. Vesicle Size. The Z-
interactions between factors previously selected by the screening average size and PDI of vesicles were determined by dynamic
design (Table 2). All other factors were fixed at certain values. light scattering (DSL) using a Zetasizer Nano ZS system
In both designs, mean diameter (Z-average size) and PDI were (Malvern Instruments Ltd., Malvern, U.K.). Three independent
selected as response variables. Minitab statistical software samples were taken from each formulation, and measurements
9166 DOI: 10.1021/acs.iecr.6b01552
Ind. Eng. Chem. Res. 2016, 55, 9164−9175
Industrial & Engineering Chemistry Research Article
Figure 1. Pareto chart of the standardized effects of independent variables (factors) on the (A) Z-average size and (B) PDI of PC liposomes for the
Plackett−Burman fractional factorial design.
were performed three times at room temperature without rupture and to extract the encapsulated compound. EE was
dilution. calculated according to the equation
2.4.2. Vesicle Morphology. Morphological analysis of vesicles
was carried out by negative staining transmission electron peak area of filtered sample
EE (%) = × 100
microscopy (NS-TEM), using a JEOL-2000 Ex II transmission peak area of unfiltered sample (1)
electron microscope (Tokyo, Japan). A sample drop was placed
on a carbon-coated copper grid, and excess sample was removed The RP-HPLC system was equipped with an HP G1315A UV/
with filter paper. Then, a drop of 2% (w/v) phosphotungstic acid vis absorbance detector (Agilent Technologies, Palo Alto, CA).
(PTA) solution was applied to the carbon grid and allowed to The column was a Zorbax Eclipse Plus C18 column with a
stand for 1 min. Once the excess staining agent had been particle size of 5 μm, 4.6 mm × 150 mm (Agilent Technologies,
removed with filter paper, the sample was air-dried, and the thin Palo Alto, CA). The mobile phase consisted of a mixture of (A)
film of stained and fixed vesicles was observed with the 100% Milli-Q-water and (B) 100% methanol with gradient
transmission electron microscope. elution at 0.8 mL/min. The step gradient started with a mobile
2.4.3. Vesicle Stability. Vesicle stability was determined by phase of 80% A, running 100% mobile phase B starting in minute
measuring backscattering (BS) profiles in a Turbiscan Lab Expert 5 for 10 min. Mobile phase B was fed for 2 min after each
apparatus (Formulaction, L’Union, France) provided with an injection to prepare the column for the next sample. The
aging station (Formulaction, L’Union, France). separation was carried out at 30 °C. Different wavelengths were
Samples were placed in cylindrical glass test cells, and used for the UV/vis detector, namely, 533 nm for Sudan Red 7B
backscattered light was monitored at 30 °C as a function of and 270 nm for vitamin D3.
time and cell height every 2 h for 7 days.
The optical reading head scans the sample in the cell, 3. RESULTS AND DISCUSSION
providing BS data every 40 μm in percentages relative to 3.1. Effects of Variables on Morphological Character-
standards as a function of the sample height (in millimeters). istics. The responses (Z-average size and PDI) of each batch
These profiles build up a macroscopic fingerprint of the sample at from P−B design were measured by dynamic light scattering
a given time, providing useful information about changes in the (DLS). The relative importance of the main effects on the Z-
size distribution or appearance of a creaming layer or a average size and PDI of PC liposomes are shown in the Pareto
clarification front with time.3,37,38 chart given in Figure 1.
2.4.4. Encapsulation Efficiency (EE). EE also provides useful Researchers must be aware of the confusion of main effects
information related to the stability of the vesicle membrane. with two-factor interactions in this type of design (resolution III),
Hydrophilic compounds are entrapped in aqueous compart- where the alias structure is too complex. However, we decided to
ments between bilayers, whereas lipophilic compounds are use the initial Plackett−Burman design only for screening
preferentially located within the surfactant or lipid bilayer.39 purposes and selection of the main factors from the Pareto chart,
Substances such as drugs, bioactive compounds, dyes, and as is usually accepted. Effects were selected by applying the
nanomaterials incorporated into vesicles can also affect the hierarchical ordering principle, known sometimes as the sparsity-
morphology and stability of the final dispersion. of-effects principle, where higher-order effects (three- or four-
For the purpose of determining EEs, Sudan Red 7B and way interactions) are sacrificed to study lower-order effects
vitamin D3 (hydrophobic compounds) were encapsulated in the (main effects first and two-way interactions next). This principle
two different formulations. suggests that priority should be given to the estimation of lower-
Each compound was analyzed by reverse-phase high-perform- order effects, especially when resources (time and money) are
ance liquid chromatography (RP-HPLC) (HP series 1100 scarce. This postulate is an empirical principle whose validity has
chromatograph, Hewlett-Packard, Palo Alto, CA). Before RP- been confirmed by the analysis of many real experiments.
HPLC analysis could be performed, the nonencapsulated According to these data, the most important variables for both
compound had to be removed by passing the sample through a responses are the organic/aqueous phase volume ratio, the (final
Sephadex G-25 column (GE Healthcare Life Sciences, aqueous-phase) phospholipid concentration, and the sonication
Wauwatosa, WI). Then, both filtered and nonfiltered samples amplitude. These results are in good agreement with previous
were diluted 1:10 (v/v) with methanol to facilitate vesicle studies carried out by Kremer et al.,40 who evaluated the effects of
9167 DOI: 10.1021/acs.iecr.6b01552
Ind. Eng. Chem. Res. 2016, 55, 9164−9175
Industrial & Engineering Chemistry Research Article
Figure 2. Pareto chart of the standardized effects of independent variables (factors) on the (A,C) Z-average size and (B,D) PDI of (A,B) PC liposomes
and (C,D) S60:Cho niosomes (1:0.5, w/w) for the 23 full factorial design.
some preparation variables on the size and polydispersity of liposomes (with standard deviations ranging from 0.304 to 4.40
liposomes made from two different natural phosphatidylcholines. nm for size and from 0.003 to 0.053 for PDI). Similar size ranges
Their experimental results showed that the most important were also obtained using the EIM in other previously reported
factor in the final size of the liposomes was the lipid studies.22,27,41,43,44
concentration in the alcohol injected into the buffer solution. The normality, variance homogeneity, and randomness
This factor corresponds to the interaction of the lipid amount assumptions were tested with a normal probability plot,
and the flow rate of organic solvent injected, two factors present frequency histogram, and residuals versus fits and residuals
in the Pareto chart in Figure 1. The same explanation was versus order plots, respectively (Supporting Information, Figure
postulated by other authors,8,41,42 confirming that the lipid S2).
concentration clearly affects the liposome size. This factor was No clear aberrant tendencies were observed, because the
found to be the most relevant one for controlling morphological residuals tended to form a line, no typical cornet pattern was
characteristics of phosphatidylcholine liposomes. Szoka43 found observed, and no time-based pattern was detected. Only some
that stirring, ionic strength, and temperature of the aqueous outlier values were detected (Cook’s distance and DFITS values
phase could also contribute to the final size, but the effects of are given in Table S3 of the Supporting Information).
these factors were smaller than those observed for lipid The ANOVA results allowed for an analysis of the
concentration, organic/aqueous phase ratio, and chemical nature contributions of the effects of the independent variables on the
of the organic solvent (a parameter not included in our study).
response function (mean size of PC liposomes). In this case,
Therefore, the experimental results in Figure 1 confirm the
significant two-way interactions were identified: (O/A) × C and
previously reported observations.43
C × A (see Figure 3). Larger sizes are reached when the organic
The ethanol injection method is usually chosen because it
avoids the sonication step, which is needed in several other solution has a higher lipid concentration (more than 20 g/L). On
methods of liposome preparation, such as the thin-film hydration the other hand, the C × A interaction reveals that the degree of
method. Preliminary experiments (data not shown) indicated size reduction upon application of a higher amplitude depends
that sonication is a crucial step for reducing the size of both on the total lipid concentration present in the medium (referred
liposomes and niosomes. Alternatively, small vesicles can be to the final volume of the dispersion).
produced without sonication by using low concentrations of All of the main effects are significant (p < 0.05), with a positive
lipids/surfactants, but with low yield. This is why we decided to effect on the mean size (a higher response value with an increase
include this step as a factor in the present study. in the factor level) for the total lipid concentration and a negative
3.2. PC Liposomes. The first three main effects from the effect (a lower response value with a decrease in factor level) for
Pareto chart obtained for the P−B design were selected for the 23 the organic/aqueous phase volume ratio and the sonication
full factorial design. The ANOVA results for Z-average size and amplitude.
PDI values are listed in Tables S1 and S2 (Supporting These effects can be explained according to a previously
Information), respectively, whereas the corresponding Pareto reported vesicle formation model.45−47 This model relies on the
chart is shown in Figure 2. Mean sizes in the range of 55−156 nm formation of vesicles through intermediate structures, such as
with PDI values between 0.173 and 0.378 were obtained for PC phospholipid bilayer fragments and sheet-like micelles. These
9168 DOI: 10.1021/acs.iecr.6b01552
Ind. Eng. Chem. Res. 2016, 55, 9164−9175
Industrial & Engineering Chemistry Research Article
Figure 3. Three-dimensional (3D) response surface plots for the factors O/A (organic/aqueous phase volume ratio), C (lipid or surfactant/stabilizer
concentration, g/L), and A (sonication amplitude, %) for the (A,C) Z-average size and (B,D) PDI of (A,B) PC liposomes and (C,D) S60:Cho niosomes
(1:0.5, w/w).
intermediates are the result of amphiphilic self-assembly because applied to vesicles during the sonication process as a result of the
of their characteristic physicochemical properties.48 effect of induced cavitation.49,50 The final effect of ultrasounds
During the injection of ethanol droplets into the aqueous can be controlled by varying the input power, ultrasound
phase, lipid reorganization inside these dispersed droplets to frequency, sonication time, and probe depth into the container.
form bilayers is favored by the fact that lipids energetically prefer As frequency increases, liposomes of smaller size are produced as
a parallel molecular arrangement.45 These planar structures give a result of stronger acoustic cavitation events. This assumption
rise to closed vesicles when their size induces enough surface was confirmed by our results, in accordance with previous
tension to close the structure and minimize the bending energy. studies.49,50 It is important to point out that, to minimize the
The sizes of these intermediates depend directly on the effects of variations in the probe depth, this factor was kept
number of lipid molecules (concentration) and the dispersion constant at 1.5 cm above the container bottom.
degree (solubilization) in the organic phase. It is obvious from Another aspect to be taken into consideration is the effect of
the previous assessment that higher concentrations of lipids in sonication time. It was reported by Silva et al.49 that sonication
the droplets will form higher membrane fragments, as our time plays an important role in decreasing vesicle size, although
experimental results and previous observations confirm.8,40−42 they observed that this effect reached a plateau at about 21 min.
It is also important to know how easily lipid droplets are Our P−B design revealed a positive effect of sonication time on
dispersed, as well as their size and homogeneity. Lipids of higher the Z-average size (from 15 to 30 min), although it was weaker
solubility will then form smaller lipid droplets and, consequently, than the effects of the other variables selected for the 23 full
shorter membrane fragments (and ultimately tiny vesicles).40 factorial design (especially sonication amplitude). A similar
This explains, in a simplified way, why higher organic/aqueous influence was observed for the PDI response, but with a stronger
phase ratios yield smaller liposomes. effect. We preferred to select sonication amplitude instead of
The negative effect of the sonication amplitude is explained by sonication time because one of the goals of controlling factors is
vesicle rupture, which takes place when an excess of energy is to obtain a narrow size distribution.
9169 DOI: 10.1021/acs.iecr.6b01552
Ind. Eng. Chem. Res. 2016, 55, 9164−9175
Industrial & Engineering Chemistry Research Article
Table 3. Estimated Coded Coefficients for the Considered Effects on the Z-Average Size and PDI of PC Liposomes and S60:Cho
Niosomes (1:0.5, w/w)
coefficientsa
response constant X1 X2 X3 X1X2 X1X3 X2X3 X1X2X3 R2
Z-Average Size
liposome (Y1) 89.68 −11.14 12.40 −17.50 −11.75 − −5.97 − 96.69
niosome (Y3) 269.82 12.72 16.87 −15.94 − −12.15 −19.92 −16.04 91.27
PDI
liposome (Y2) 0.280 − − 0.038 −0.012 − −0.029 − 89.35
niosome (Y4) 0.136 −0.026 0.057 0.026 − − − − 84.73
a
X1, organic/aqueous phase volume ratio; X2, PC or S60:Cho concentration (g/L); X3, sonication amplitude (%).
As the design included a center point with several repetitions are shown in Figures 2 and 3, respectively. Mean sizes in the
(n = 5), the presence of curvature in the response variables could range of 224−362 nm with PDI values between 0.032 and 0.291
be tested (Figure 3). Because curvature seemed to be significant were obtained for S60:Cho niosomes (with standard deviations
(p < 0.05), a term involving center point (Ct Pt) was included in ranging from 1.05 to 7.28 nm for size and from 0.009 to 0.052 for
the equations for its estimation. PDI). Similar size and PDI ranges were reported for niosomes
With all of this information about the effects and their prepared by the EIM using Span 60 as the membrane
estimated coefficients, the following equation (R2 = 96.69%) for component.17
the Z-average size value of PC liposomes (Y1) was generated Two-way interactions (O/A × A, C × A) and a three-way
interaction (O/A × C × A) were detected, with sonication
Y1 = 62.8 + 2.55(O/A) + 0.449C − 0.185A amplitude (A) as the common factor in these interactions (see
− 0.0185(O/A) × C − 0.00555C × A − 9.26(Ct Pt)
Figure 2C). Therefore, it can be postulated that sonication
amplitude is the key factor in the niosome size response. The
(2)
response depends on both the O/A and C factor levels, with a
Different behavior was observed regarding PDI, which was higher interaction between the sonication amplitude and the
strongly affected by the sonication amplitude as the only total amount of membrane components. Differences in the
significant main effect and its interaction with the total lipid magnitude of the coefficient of this factor between liposomes and
amount. The O/A × C interaction was also detected, but with a niosomes can be attributed to the initial size before sonication
lower effect on the PDI response. (smaller for liposomes) and vesicle stability.50
To understand the C × A interaction, it is important to take The three main effects are significant, but in contrast to the
into account the effect of the sonication amplitude as the main case for liposomes, the organic/aqueous phase volume ratio (O/
effect. An increase in this factor leads to a less monodisperse size A) shows a positive effect on niosome size. This behavior could
distribution, that is, higher PDI values. However, according to be due to different molecular features of the surfactant and
the interaction, this response depends highly on the total amount stabilizer that result in different interactions with the organic
of lipids present in the sample. At a low level of the lipid amount, phase and, therefore, poor or insufficient solubility.
the reduction in size is more effective (as previously mentioned), The other two variables (C, A) have effects similar to those
but the size distribution is large. However, at a high level of the described above for liposomes. Therefore, the same explanation
lipid amount, this enlargement of the size distribution is regarding surfactant concentration and sonication amplitude can
significantly lower. be applied here to justify their effects on niosome size. In this
Curvature in the response was also tested, again revealing a case, the stronger effect of C is explained by the influence of
significant presence (p < 0.05). For the PDI response (Y2), the cholesterol on the final size of vesicles, as reported by Padamwar
following equation with an R2 value of 89.35% was obtained and Pokharkar.8
Once again, curvature was detected for the Z-average size
Y2 = −0.160 + 0.00939A − 0.0000420(O/A) × C response. The following equation was obtained to model this
− 0.0000250C × A − 0.0425(Ct Pt) case, with an adjusted correlation coefficient (R2) of 91.27%
(3)
These equations were formulated with uncoded coefficients, Y3 = 236.9 − 4.31(O/A) − 0.012C − 0.56A
making it easier to use them to predict selected target size and
PDI values. + 0.0461(O/A) × C + 0.00363C × A
3.3. S60:Cho Niosomes. To investigate whether the − 0.00114(O/A) × C × A + 44.00(Ct Pt) (4)
selected factors in the P−B design for PC liposomes (a reference
model for vesicular systems) produced similar effects with other On the other hand, a completely different behavior was observed
different formulations, the same 23 full factorial design using regarding the PDI response. Only the three main effects (O/A, C,
center-point replicates was performed for a typical niosome A) were found to be significant, and no interactions were found.
formulation, in this particular case, S60:Cho niosomes (1:0.5, w/ Two positive effects on the niosome PDI were detected:
w). The main variables were the organic/aqueous phase volume surfactant/stabilizer concentration and sonication amplitude. In
ratio (O/A), the total concentration of surfactant and stabilizer this case, the total concentration of membrane components
(C), and the sonication amplitude (A). seemed to have an important role in the vesicle size distribution,
The ANOVA results for Z-average size and PDI values are as can be seen in the correspondent Pareto chart (Figure 2). This
listed in Table S1 (Supporting Information), and the observation once again can be attributed to the solubilization of
corresponding Pareto chart and three-dimensional surface plot membrane components in the organic phase. Higher concen-
9170 DOI: 10.1021/acs.iecr.6b01552
Ind. Eng. Chem. Res. 2016, 55, 9164−9175
Industrial & Engineering Chemistry Research Article
Figure 4. Optimization plot and values of individual (d) and composite (D) desirability provided by the response optimizer (Minitab, version 17) for an
example of size-tuned PC liposome (desired size = 70 nm, with a minimum PDI value).
trations of these components require better solubilization in The estimated coded coefficients for the considered effects on
dispersed droplets to reach small membrane fragments. the Z-average sizes and PDIs of PC liposomes and S60:Cho
It is important to note that some combinations of factors niosomes are listed in Table 3, as a summary of the factors’
yielded narrow size distributions, namely, PDI ≤ 0.100, a value influence. Coded coefficients were used to maintain the
frequently obtained by other preparation methods, such as orthogonality of the designs and to allow for a direct comparison
microfluidic hydrodynamic focusing51 also using S60:Cho as the between coefficients.
formulation. 3.4. Vesicle Characterization. Size-tuned vesicles were
A negative effect was detected for the organic/aqueous phase prepared under selected operating conditions by applying the
volume ratio (O/A). As the final concentration of ethanol models obtained from the experimental design (eqs 2−5) and
increased during the injection process, a smaller size distribution the assistance of the response optimizer and response predictor
was obtained. As previously mentioned, no interaction between in Minitab statistical software (version 17). These tools can be
this factor and the total concentration of membrane components
applied to the simultaneous optimization of several responses
was observed.
only when the same set of factors are studied separately, because
The following equation with an R2 value of 84.73% was
obtained for the niosome PDI model response (Y4) a common experimental region is needed.
The operating conditions were selected to prepare PC
Y4 = 0.053 − 0.00392(O/A) + 0.000039C + 0.00067A liposomes with a mean size of 70 nm and the minimum PDI
value (predicted values of Y1 = 67 ± 4 and Y2 = 0.317 ± 0.013)
+ 0.0597(Ct Pt) (5) and S60-Cho niosomes with a mean size of 240 nm and the
9171 DOI: 10.1021/acs.iecr.6b01552
Ind. Eng. Chem. Res. 2016, 55, 9164−9175
Industrial & Engineering Chemistry Research Article
Figure 5. Optimization plot and values of individual (d) and composite (D) desirability provided by the response optimizer (Minitab, version 17) for an
example of size-tuned S60:Cho niosome (1:0.5 w/w) (desired size = 240 nm, with a minimum PDI).
minimum PDI value (predicted values of Y3 = 239 ± 11 and Y4 = vesicles, as a result of the interactions of the electron beam with
0.120 ± 0.025). These sizes and PDI values were selected only as PTA, which produces a selective deposit of metal ions that
an example. The factor output values were O/A = 5:50, C = 2 g/ enhances morphological details. The micrographs show spherical
L, and A = 55% for the liposomes and O/A = 5.9:50, C = 2 g/L, structures of approximately 80 nm for the liposomes (Figure 6C)
and A = 55% for the niosomes. Figures 4 and 5 show optimization and about 250 nm for the niosomes (Figure 6D). These values
plots and values of individual and composite desirability for size- agree with the DLS measurements.
tuned liposomes and size-tuned niosomes, respectively. Figure 6D shows clusters of niosomes that are all similar in
The experimental results showed that the models obtained
size. Aggregation arose during the drying step prior to TEM
with the experimental design were accurate, because mean sizes
of 69 ± 0.5 nm (PDI = 0.245 ± 0.005) and 233 ± 3 nm (PDI = measurements, because no flocculation phenomena were
0.112 ± 0.004) were obtained for the PC liposomes and S60:Cho monitored with the Turbiscan apparatus.
niosomes, respectively. The relative error was low for the Slight differences were noticed in the zeta potential measure-
experimental results regarding mean size (3% for Y1 and Y3) but ments, exhibiting low values for both types of vesicles. Niosomes
higher for the size distributions (22% for Y2 and 7% for Y4). had values of about −16.8 ± 0.7 mV, whereas the liposomes had
The sizes and morphologies of the vesicles were investigated values of −6.9 ± 0.3 mV. This small value for the liposomes could
by TEM, using a negative contrast. Figure 6 shows black-stained be due to neutralization of the negative charge from the
9172 DOI: 10.1021/acs.iecr.6b01552
Ind. Eng. Chem. Res. 2016, 55, 9164−9175
Industrial & Engineering Chemistry Research Article
Figure 6. (A,B) BS profiles and (C,D) TEM micrographs of empty vesicles designed with a controlled size and PDI values by applying the models
obtained from experimental design: (A,C) PC liposomes and (B,D) S60:Cho niosomes (1:0.5, w/w).
phosphate groups by sodium cations present in the medium High EE values were obtained for both Sudan Red 7B and
(from sodium chloride in the PB solution). vitamin D3, as expected taking into account their hydrophobic
The formulated vesicles exhibited a high stability after 1 week character. EE values up to 90.1% and 88.0% were obtained for
of monitoring time. BS profiles obtained for the PC liposomes Sudan Red 7B encapsulated in PC liposomes and S60:Cho
are given in Figure 6, where a variation of 4.5% in the middle part niosomes, respectively. Experiments carried out with vitamin D3
of the cell (from 10 to 30 mm) is noticed. A simultaneous slight led to EE values of 99.2% for PC liposomes and 73.9% for
clarification process was observed in the middle and top parts of S60:Cho niosomes. These results are in good agreement with
the cell in the corresponding transmission profile (results not those of previous studies, where compounds with similar
shown). This was promoted by some movement of the PC chemical properties were encapsulated.12,13,27
liposomes toward the bottom of the cell, resulting in a slight
increase in BS (sedimentation). However, this was a reversible 4. CONCLUSIONS
process, caused by differences in concentration, with the sample In this work, an adequate approximation using DoE was applied
remaining stable and maintaining its initial properties (size and to study the influence of experimental factors of the EIM on the
PDI). The vesicles were again characterized after gentle agitation mean size and size distribution of PC liposomes and S60:Cho
of the cell at the end of the monitoring time with analogous niosomes (1:0.5, w/w).
results. An initial screening design enabled a reduction of the number
For the S60:Cho niosomes (Figure 6B), the BS profile of variables. This was a necessary step before carrying out a full
factorial design. Finally, response models were applied to prepare
remained nearly constant (variations of approximately 0.5%)
selected size-tuned nanovesicles, which were characterized from
with time, showing high stability. Some variation was also
a stability point of view.
observed in the transmission profile all along the cell height, This was achieved with a low number of experiments (58
because the sample was not translucent. runs). This methodology enabled two different formulations
3.4.1. Encapsulation Efficiency (EE). Vesicles containing (liposomes and niosomes, the most common types of nano-
Sudan Red 7B and vitamin D3 as model compounds (both vesicles) to be studied in a comparative way. Stable liposomes
lipophilic) were also prepared and characterized. No differences and niosomes of the targeted sizes were successfully prepared
were observed regarding mean size and PDI values or TEM, zeta with the model equations obtained, with encapsulation
potential, or Turbiscan measurements, meaning that the efficiencies higher than 73.9% in all cases for selected
entrapped compounds did not affect the vesicle’s behavior. hydrophobic compounds.
9173 DOI: 10.1021/acs.iecr.6b01552
Ind. Eng. Chem. Res. 2016, 55, 9164−9175
Industrial & Engineering Chemistry Research Article
The most important variables identified by ANOVA were the psoriasis: Application of Box−Behnken design, in-vitro evaluation and
organic/aqueous phase volume ratio, the (final aqueous-phase) in-vivo skin deposition study. Int. J. Pharm. 2015, 485, 235−243.
phospholipid concentration, and the sonication amplitude. (11) Jadhav, S. M.; Morey, P.; Karpe, M.; Kadam, V. Novel vesicular
These results offer new insights into the mechanism and effects system: An overview. J. Appl. Pharm. Sci. 2012, 02, 193−202.
(12) Rajera, R.; Nagpal, K.; Singh, S. K.; Mishra, D. N. Niosomes: a
of the factors involved in nanovesicle preparation by the EIM,
controlled and novel drug delivery system. Biol. Pharm. Bull. 2011, 34,
one of the most easily scaled-up methods for preparing vesicles 945−53.
for several fields of interest.
■
(13) Uchegbu, I. F.; Vyas, S. P. Non-ionic surfactant based vesicles
(niosomes) in drug delivery. Int. J. Pharm. 1998, 172, 33−70.
ASSOCIATED CONTENT (14) Bangham, A. D.; Standish, M. M.; Watkins, J. C. Diffusion of
*
S Supporting Information univalent ions across the lamellae of swollen phospholipids. J. Mol. Biol.
1965, 13, 238−252.
The Supporting Information is available free of charge on the
(15) da Silva Malheiros, P.; Daroit, D. J.; Brandelli, A. Food
ACS Publications website at DOI: 10.1021/acs.iecr.6b01552. applications of liposome-encapsulated antimicrobial peptides. Trends
ANOVA results for Z-average size and PDI of PC Food Sci. Technol. 2010, 21, 284−292.
liposomes for the 23 full factorial design; Cook’s distances (16) du Plessis, J.; Weiner, N.; Müller, D. G. The influence of in vivo
and DFITS values for each response in the full factorial treatment of skin with liposomes on the topical absorption of a
designs; optimization contour plot for the factors studied hydrophilic and a hydrophobic drug in vitro. Int. J. Pharm. 1994, 103,
in the full factorial design for both responses; testing for R1−R5.
(17) Manconi, M.; Sinico, C.; Valenti, D.; Loy, G.; Fadda, A. M.
normality, variance homogeneity, and randomness Niosomes as carriers for tretinoin I: Preparation and properties. Int. J.
assumptions of ANOVA for the full factorial design Pharm. 2002, 234, 237−248.
(PDF) (18) Manca, M. L.; Manconi, M.; Nacher, A.; Carbone, C.; Valenti, D.;
■
Maccioni, A. M.; Sinico, C.; Fadda, A. M. Development of novel
AUTHOR INFORMATION diolein−niosomes for cutaneous delivery of tretinoin: Influence of
formulation and in vitro assessment. Int. J. Pharm. 2014, 477, 176−186.
Corresponding Author (19) Mahale, N. B.; Thakkar, P. D.; Mali, R. G.; Walunj, D. R.;
*Tel.: +34 985103509. E-mail: [email protected]. Chaudhari, S. R. Niosomes: Novel sustained release nonionic stable
Notes vesicular systems An overview. Adv. Colloid Interface Sci. 2012, 183−
The authors declare no competing financial interest. 184, 46−54.
■
(20) Moghassemi, S.; Hadjizadeh, A. Nano-niosomes as nanoscale
drug delivery systems: An illustrated review. J. Controlled Release 2014,
ACKNOWLEDGMENTS 185, 22−36.
This work was supported by the Ministerio de Economiá y (21) Mali, N.; Darandale, S.; Vavia, P. Niosomes as a vesicular carrier
Competitividad (MINECO, Spain), under Grant MINECO-13- for topical administration of minoxidil: formulation and in vitro
CTQ2013-47396-R. This study was also cofinanced by the assessment. Drug Delivery Transl. Res. 2013, 3, 587−592.
Consejeriá de Educación y Ciencia del Principado de Asturias (22) Fan, M.; Xu, S.; Xia, S.; Zhang, X. Preparation of salidroside nano-
(ref FC-04-COF-50-MEC, PCTI Asturias 2006-2009, ref liposomes by ethanol injection method and in vitro release study. Eur.
Food Res. Technol. 2008, 227, 167−174.
EQP06-024, and FC15-GRUPIN14-022). We especially thank
(23) Marianecci, C.; Di Marzio, L.; Rinaldi, F.; Celia, C.; Paolino, D.;
Prof. Antonia Salas (University of Oviedo) for her advice with Alhaique, F.; Esposito, S.; Carafa, M. Niosomes from 80s to present: The
the statistical work.
■
state of the art. Adv. Colloid Interface Sci. 2014, 205, 187−206.
(24) Akbarzadeh, A.; Rezaei-Sadabady, R.; Davaran, S.; Joo, S. W.;
REFERENCES Zarghami, N.; Hanifehpour, Y.; Samiei, M.; Kouhi, M.; Nejati-Koshki, K.
(1) Capretto, L.; Carugo, D.; Mazzitelli, S.; Nastruzzi, C.; Zhang, X. Liposome: classification, preparation, and applications. Nanoscale Res.
Microfluidic and lab-on-a-chip preparation routes for organic nano- Lett. 2013, 8, 102.
particles and vesicular systems for nanomedicine applications. Adv. Drug (25) Justo, O. R.; Moraes, Â . M. Analysis of process parameters on the
Delivery Rev. 2013, 65, 1496−1532. characteristics of liposomes prepared by ethanol injection with a view to
(2) Rongen, H. A. H.; Bult, A.; van Bennekom, W. P. Liposomes and process scale-up: Effect of temperature and batch volume. Chem. Eng.
immunoassays. J. Immunol. Methods 1997, 204, 105−133. Res. Des. 2011, 89, 785−792.
(3) Pando, D.; Gutiérrez, G.; Coca, J.; Pazos, C. Preparation and (26) Batzri, S.; Korn, E. D. Single bilayer liposomes prepared without
characterization of niosomes containing resveratrol. J. Food Eng. 2013, sonication. Biochim. Biophys. Acta, Biomembr. 1973, 298, 1015−1019.
117, 227−234. (27) Pham, T. T.; Jaafar-Maalej, C.; Charcosset, C.; Fessi, H. Liposome
(4) Gómez-Hens, A.; Fernández-Romero, J. M. The role of liposomes and niosome preparation using a membrane contactor for scale-up.
in analytical processes. TrAC, Trends Anal. Chem. 2005, 24, 9−19. Colloid. Colloids Surf., B 2012, 94, 15−21.
(5) Edwards, K. A.; Bolduc, O. R.; Baeumner, A. J. Miniaturized (28) Loukas, Y. L. Experimental studies for screening the factors that
bioanalytical systems: enhanced performance through liposomes. Curr. influence the effectiveness of new multicomponent and protective
Opin. Chem. Biol. 2012, 16, 444−452. liposomes. Anal. Chim. Acta 1998, 361, 241−251.
(6) Liu, Q.; Boyd, B. J. Liposomes in biosensors. Analyst 2013, 138, (29) Shah, S. R.; Parikh, R. H.; Chavda, J. R.; Sheth, N. R. Application
391−409. of Plackett−Burman screening design for preparing glibenclamide
(7) Canton, I.; Battaglia, G. Endocytosis at the nanoscale. Chem. Soc. nanoparticles for dissolution enhancement. Powder Technol. 2013, 235,
Rev. 2012, 41, 2718−39. 405−411.
(8) Padamwar, M. N.; Pokharkar, V. B. Development of vitamin loaded (30) El-Samaligy, M. S.; Afifi, N. N.; Mahmoud, E. A. Increasing
topical liposomal formulation using factorial design approach: Drug bioavailability of silymarin using a buccal liposomal delivery system:
deposition and stability. Int. J. Pharm. 2006, 320, 37−44. Preparation and experimental design investigation. Int. J. Pharm. 2006,
(9) Taha, E. I. Lipid vesicular systems: formulation optimization and ex 308, 140−148.
vivo comparative study. J. Mol. Liq. 2014, 196, 211−216. (31) Shaikh, K. S.; Chellampillai, B.; Pawar, A. P. Studies on nonionic
(10) Abdelbary, A. A.; AbouGhaly, M. H. H. Design and optimization surfactant bilayer vesicles of ciclopirox olamine. Drug Dev. Ind. Pharm.
of topical methotrexate loaded niosomes for enhanced management of 2010, 36, 946−53.
1
Department of Chemical and Environmental Engineering, University of Oviedo,
Julián Clavería 8, 33006 Oviedo, Spain.
2
Department of Physical and Analytical Chemistry, University of Oviedo, Julián
Clavería 8, 33006 Oviedo, Spain.
- S1-
Table S1. ANOVA results (coded units) for Z-average size of PC-liposomes for
the 23 full factorial design; results for S60:Cho niosomes are also given (cursive
numbers)
- S2-
Table S2. ANOVA results (coded units) for PDI of PC-liposomes for the 23 full
factorial design; results for S60:Cho niosomes are also given (cursive numbers)
2-Way
3
Interactions 0.0162 0.0162 0.0054 11.02 0.0010
0.0017 0.0017 0.0006 0.43 0.7370
3-Way
1
Interactions 0.0017 0.0017 0.0017 3.39 0.0900
0.0002 0.0002 0.0002 0.18 0.6830
Total 20 0.0550
0.1042
ANOVA indicates analysis of variance; DF, degrees of freedom; SS, sum of squares; MS,
mean of squares; F, Fischer’s ratio; P, p-value.
- S3-
Table S3. COOK's distance and DFITS values obtained for each response in the
full factorial designs
- S4-
Figure S1. Contour Plot for the factors O:A (organic:aqueous phase volume
ratio), C (lipid or surfactant/stabilizer concentration, g/L) and A (sonication
amplitude, %) on Z-average size, nm (A) and PDI (C) of PC-liposomes, and Z-
average size, nm (B) and PDI (D) of S60:Cho niosomes (1:0.5, w/w).
- S5-
Figure S2. Testing for normality, variance homogeneity and randomness assumptions of ANOVA for the full
factorial design of PC-liposomes and S60:Cho niosomes (1:0.5 w/w).
- S6-
9. Olle, M. and Virsile, A. (2013) The effects of light-emitting
responsive plant cultivars and most effi- diode lighting on greenhouse plant growth and quality.
homeostasis based on cellular communi-
cient light spectra for production of high- Agric. Food Sci. 22, 223–234 cation. Advances in our comprehension
quality products. A potential risk related to 10. Taulavuori, K. et al. (2013) Blue mood for superfood. Nat. of the development and expansion of sev-
Prod. Commun. 8, 791–794
the use of light spectra deviating substan- 11. Kopsell, D.A. and Sams, C.E. (2013) Increases in shoot tissue
eral pathologies are largely due to our
tially from the daylight spectrum is enrich- pigments, glucosinolates, and mineral elements in sprouting increased understanding of the biological
broccoli after exposure to short-duration blue light from light
ment of nontarget phytochemicals, for emitting diodes. J. Am. Soc. Hort. Sci. 138, 31–37
roles of EVs, with a particular focus on
example, alkaloids, which may give pun- 12. Sabzalian, M.R. et al. (2014) High performance of vege- exosomes [1].
gent tastes or act as toxins or allergens. tables, flowers, and medicinal plants in a red-blue LED
incubator for indoor plant production. Agron. Sustain. Dev.
Therefore, composition of nontarget phy- The increasing amount of data on the
34, 879–886
tochemicals should also be monitored 13. Amoozgar, A. et al. (2017) Impact of light-emitting diode
composition, biogenesis, and roles of
when plants are grown under modified irradiation on photosynthesis, phytochemical composition exosomes in physiology and pathology
and mineral element content of lettuce cv. Grizzly. Photo-
light spectra. The new methodologies synthetica 55, 85–95
has created new possibilities in diagnosis
may also face difficulties if public opinion 14. Arena, C. et al. (2016) The effect of light quality on growth, and therapy [2]. Exosomes have unique
photosynthesis, leaf anatomy and volatile isoprenoids of a
questions the use of photonics to modify characteristics that result from their cellu-
monoterpene-emitting herbaceous species (Solanum
the chemical quality of edible plants. lycopersicum L.) and an isoprene-emitting tree (Platanus lar origin which make them valuable as
orientalis L.). Environ. Exp. Bot. 130, 122–132 new biomarkers for diagnosis, stratifica-
These aspects will need open discussion 15. Crozier, A. et al., eds (2007) Plant Secondary Metabolites:
before plant production becomes more Occurrence, Structure and Role in the Human Diet,
tion and planning, and for treatment effi-
extensively dependent on narrow[83_TD$IF]-band- Blackwell cacy evaluation. Their molecular
width LED lighting. composition, in terms of both their mem-
brane and cargo, includes lipids, proteins,
and nucleic acids, providing information
Acknowledgements
This work was supported by UEF spearhead project
Forum about their cells of origin and their status.
Our ability to now acquire this valuable
CABI (JKH, RJ-T) and the European Social Fund (ESF)
grant no. S12530 (JKH, MK).
Fully Artificial cellular information and the fact that it also
Exosomes: Towards comes from hard-to reach tissues, has
resulted in the term ‘liquid biopsy’, a
1
Department of Environmental and Biological Sciences,
University of Eastern Finland, P.O. Box 1627, FI-70211
Kuopio, Finland
New Theranostic new frontier in the clinical field.
2
Department of Environmental and Biological Sciences,
University of Eastern Finland, P.O. Box 111, [82_TD$IF]FI-80101,
Biomaterials In addition, exosomes combine the
Joensuu, Finland
1,2 advantages of nanocarriers (i.e., particles
Pablo García-Manrique,
used to efficiently deliver molecules) and
*Correspondence:
jarmo.holopainen@uef.fi (J.K. Holopainen).
Gemma Gutiérrez,1 and
therapeutic agents. Exosomes are cur-
http://dx.doi.org/10.1016/j.tibtech.2017.08.009 Maria Carmen Blanco-López2,* rently considered among the most prom-
ising drug delivery systems, especially for
References Bionanotechnology routes have
1. Ballare, C.L. (2014) Light regulation of plant defense. Annu. gene therapy in different disorders (such
Rev. Plant Biol. 65, 335–363 been recently developed to pro- as genetic deficiencies or antitumor pro-
2. Junker, R.R. et al. (2017) Covariation and phenotypic duce fully artificial exosomes: bio-
integration in chemical communication displays: biosyn- gression) [3]. Several studies have been
thetic constraints and eco-evolutionary implications. New mimetic particles designed to published during the past 5 years regard-
Phytol. Published online March 3, 2017. http://dx.doi.org/
10.1111/nph.14505
overcome certain limitations in ing the modification of targeting moieties
3. Carvalho, S.D. et al. (2016) Light quality dependent extracellular vesicle (EV) biology and/or the encapsulation of endogenous
changes in morphology, antioxidant capacity, and volatile
and applications. These particles and exogenous material in exosomes, pre
production in sweet basil (Ocimum basilicum). Front. Plant
Sci. 7, 1328 could soon become true therapeu- or post isolation from cell cultures [4].
4. Tholl, D. (2015) Biosynthesis and biological functions of
tic biomaterials. Here, we outline These studies represent the development
terpenoids in plants. Adv. Biochem. Eng. Biotechnol. 148,
of so-called ‘exosome-based semisyn-
63–106 their current preparation techni-
5. Kadomura-Ishikawa, Y. et al. (2013) Phototropin 2 is thetic nanovesicles (NVs)’, a subtype of
involved in blue light-induced anthocyanin accumulation
ques, their explored and future pos-
artificial exosomes that includes all exo-
in Fragaria x ananassa fruits. J. Plant Res. 126, 847–857 sibilities, and their present limits.
6. Han, T. et al. (2017) Improving “color rendering” of LED somes with modifications for specific pur-
lighting for the growth of lettuce. Sci. Rep. 7, 45944 poses. They have even been tested for
7. Colquhoun, T.A. et al. (2013) Light modulation of volatile
Biology and Applications of autologous therapy, using the patient’s
organic compounds from petunia flowers and select fruits.
Postharvest Biol. Technol. 86, 37–44 Extracellular Vesicles own immune cells (cultured and
8. Christie, J.M. et al. (2012) Plant UVR8 photoreceptor The past decade has witnessed a revolu- expanded outside the body) as a source
senses UV-B by tryptophan-mediated disruption of
cross-dimer salt bridges. Science 335, 1492–1496 tion in our understanding of human body of exosomes.
Figure 1. Advantages (Green) and Disadvantages (Red) of Two Approaches for the Bionanotechnological Development of Fully Artificial Exosomes
as Theranostic Agents. Abbreviation: NV, nanovesicles.
Despite the great expansion of techni- membrane fragments that will be used proliferation enhancement [8]; and (ii) spe-
ques for developing semisynthetic exo- to form the vesicles. Different methodolo- cific microfluidic devices. Simple pressur-
somes, the main drawbacks concerning gies based on this approach have been ization over a device based on an array of
their clinical application include produc- developed, mainly for drug delivery, but parallel hydrophilic microchannels has
tion, isolation, modification, and purifica- also for enhancing cell proliferation and been described for the production of
tion on a large scale and at a suitable for generating exosome-mimetic models NVs for the delivery of endogenous
clinical grade. However, these drawbacks for biodistribution analyses (i.e., monitor- RNA to targeted cell cultures [9]. The
have resulted in the design and manufac- ing exosomes after exogenous adminis- fabrication of microchannels on micro-
ture of fully synthetic exosome-mimetic tration) (Table 1). Two of the most relevant blades (fabricated in silicon nitride)
particles using bionanotechnology. Here, strategies involve: (i) extrusion over poly- resulted in a device able to slice living cells
we provide an overview of these techni- carbonate membrane filters. This is a as they flowed through the channels [10].
ques, organized according to two trends common way to reduce the mean size The incorporation of exogenous material
in nanofabrication: top-down and bot- and homogenize the size distribution of in the cell suspension enhanced their
tom-up approaches (Figure 1). colloidal systems. By applying this tech- encapsulation by plasma membrane frag-
nique to cultured cells, NVs have been ments during reassembly and suggested
Top-Down Methodologies: produced with a simple commercial lipo- a method for in vitro exogenous material
Bioengineering Cells as some extruder and diminishing pore size delivery.
Membrane Fragment Precursors filters [5,6]; they have been used to treat
Top-down methodologies rely on the pro- tumors by targeted encapsulated chemo- Both of these methods are suitable for the
duction of nanosized materials by break- therapeutics. A scaled-up version was production of larger amounts (by a factor
ing bigger and more complex units into also developed with a device designed of more than 250) of biologically active
smaller parts. In this case, the production to be used with conventional laboratory particles compared with natural exosome
of artificial exosomes begins with cultured centrifuges [7]. This device was used to release yields. Sizing and biochemical
cells that are then used to produce mass-produce NVs in a study of cell profiling showed that these NVs were
Manual extrusion over Human monocytes (U937) and Exogenous, chemotherapeutic drugs Targeted delivery of [5]
polycarbonate membrane murine mouse macrophages (doxorubicin, 5-FU, gemcitabine, and chemotherapeutics to an in vitro model
filters with a device for (Raw 264.7) carboplatin) (TNFa-treated HUVECs) and in vivo-
liposome preparation induced malignant tumors (CT26
mouse colon adenocarcinoma cells)
Murine mouse macrophages Exogenous, radiolabeling agent 99mTc- In vivo biodistribution of exosomes and [6]
(Raw 264.7) HMPAO artificial counterparts
Centrifugal-induced extrusion Murine mouse embryonic Endogenous, precursor cell Gene delivery to NIH-3T3 fibroblast [7]
over membrane filters in a stem cell line (D3) characteristic RNA (mOct 3/4 and cells
device designed to be used in mNanog)
lab centrifuges
No intention to encapsulate any Enhanced in vitro cell proliferation for [8]
specific compounds regenerative medicine (murine skin
fibroblasts)
Pressurization over hydrophilic Murine mouse embryonic Endogenous, precursor cell Gene delivery to NIH-3T3 fibroblast [9]
microchannels array on a stem cell line (D3) characteristic RNA (mOct 3/4 and cells
microfluidic device mNanog)
Living cells sliced with silicon Murine mouse embryonic Exogenous, polystyrene beads as Material delivery to mouse embryonic [10]
nitride blades in a microfluidic stem cell line (D3) representative exogenous material fibroblasts
device
Classical liposome, PC:Chol Thin film hydration method MHC class I peptide complexes and Ex vivo and in vivo T cell expansion for [12]
(TFHM) and maleimide-based FAB regions against T cell receptors for immunotherapies
bioconjugation strategy adhesion, early and late activation, and
survival
Mimicking exosome lipid TFHM and Ni2+/His-Tag APO2L/TRIAL-His10 recombinant Downregulation of T cell activation in an [13,14]
composition, PC:Chol:SM protein coordination as proteins autoimmune disease animal model
bioconjugation strategy (antigen-induced arthritis);
immunotherapy for apoptosis induction
in hematological tumors
Innovative liposomes, PC: Microemulsion and micelle- Monoclonal antibody against DEC205 Proof of concept of artificial antigen [15]
CpEL:DOPE assembling method for vesicle dendritic cell antigen presentation to dendritic cells
formation
a
Abbreviations: 99mTc-HMPAO, 99mTc-hexamethylpropyleneamineoxime; Chol, cholesterol; CpEL, chemopor EL; DOPE, dioleoyl-phosphoethanolamine; HUVECs,
human umbilical vein endothelial cells; MHC, major histocompatibility complex; PC, phosphatidylcholine; SM, sphingomyelin; TFHM, thin film hydration method.
substantially similar to natural exosomes issue regarding the production of these include those used in the cosmetic and
[4–9]. Their ability to exhibit receptors and mimetic particles is the need for final puri- drug-delivery industries for the prepara-
co-receptors, which is essential to target fication steps identical to those used for tion of liposomes, which are particles
and effectively interact with receptor cell exosome isolation, which require trained formed by a lipid bilayer that resembles
populations, is of special relevance. In personnel and are time-consuming. the plasma membrane. These materials
addition, their natural origin from cells are a promising starting point to design
Bottom-Up Techniques:
renders them immunotolerant [4–9]. and manufacture artificial exosomes. The
Mimicking the Plasma Membrane self-assembled synthetic bilayer can be
However, these methods have some by Preparing Artificial Bilayers then functionalized with selected proteins
drawbacks. Cargo sorting lacks selectiv- By contrast, bottom-up techniques cre- to mimic the desired exosomal functions.
ity due to the passive encapsulation of the ate complex structures from molecular With this purpose in mind, special lipids
surrounding medium during membrane building blocks by using their physical can be incorporated into the liposome
fragment self-assembly. Another relevant and chemical properties. These methods formulation. These can be modified with
To cite this article: Pablo García-Manrique, María Matos, Gemma Gutiérrez, Carmen Pazos &
María Carmen Blanco-López (2018) Therapeutic biomaterials based on extracellular vesicles:
classification of bio-engineering and mimetic preparation routes, Journal of Extracellular Vesicles,
7:1, 1422676, DOI: 10.1080/20013078.2017.1422676
REVIEW ARTICLE
loading methods with scalable properties in clinical In order to provide a systematic classification to
translation has been identified [14]. move around in this new emerging field, we suggest
the nomenclature given in Figure 1. This artificial EVs
landscape is based on the concept behind the term. In
Bio-engineered and mimetic EVs for
this way, “artificial EVs” will be used as a general
nanomedicine: classification of artificial EVs
concept to designate all vesicles, modified or manufac-
EVs have been modified in the search for broader tured (from natural or synthetic sources), with the aim
therapeutic capability. Sometimes, this included the to mimic EVs (mainly exosomes) for therapeutic uses.
incorporation of new elements for targeted puporses, Behind this term, two categories of artificial EVs can be
in vitro or in vivo traceability, or the material to be discerned: “semi-synthetic EVs” and “fully synthetic”
delivered. In other cases, modification was aimed at the or “EVs mimetic vesicles”, corresponding to modified
enhancement of colloidal stability, or change in surface natural EVs (pre- or post-isolation) and artificial struc-
charge to increase their uptake rate. These new tures, lab-made or generated from cultured cells.
approaches have generated new terms such as bio- The former generate semi-synthetic products, as
engineered exosomes, artificial exosomes [15], exo- they start from a natural substrate, which can be sub-
some-mimetic nanovesicles [16], exosome-like nanove- sequently modified before or after their isolation. The
sicles [17,18] and exosome-based semi-synthetic modification affects the structures of the outer surface
vesicles [19]. These expressions have been used with of the vesicles, the membrane or the cargo that travels
different meanings in the literature, but to date, there within, and could also include hydrophobic molecules
has not yet been a clear criterion for their classification. at the membrane.
One example is the term “exosome-like nanovesicles”. The term fully synthetic, on the other hand, stresses
In some works, this concept is used to name artificial the artificiality of the product. We have recently briefly
vesicles made from cells through different techniques commented their potential in therapeutics [23]. These
to mimic exosomes [17,18]. However, cell-derived vesi- techniques can be classified on the basis of their man-
cles with morphological and biochemical characteris- ufacturing route: those starting from larger substrates
tics similar to exosomes were also named exosome-like (cells) that are reduced to units for the creation of
nanovesicles by other authors [19,20]. Other authors small size vesicles (top-down nanotechnology) or
working with non-animal research models used this those taking individual molecules (lipids, proteins,
term to refer to vesicles with size and flotation density etc.) that self-assemble in higher-order structures with
values similar to those of exosomes. For example, tunable composition (bottom-up nanotechnology).
Regente et al. [21] described the presence of exo- Top-down products differ from natural EVs in terms
some-like vesicles in sunflower plant fluids, and of micro-structure and biochemical composition, since
Prado et al. [22] showed evidence of vesicles quite they are formed from cell fragments: the characteristic
similar to exosomes during pollen germination. membrane microdomains [24] (lipid rafts) and
Figure 1. Artificial EV landscape: explored routes to date for the preparation of artificial EVs for specific purposes. *EBSSNs [19].
JOURNAL OF EXTRACELLULAR VESICLES 3
associated pools of surface markers (especially tetraspa- scientific community. The combination of both sources
nins) are absent, and the minimal cytoskeleton is not of information would be the perfect scenario for the
present. design of future artificial EV-based therapies.
In any case, both types of artificial EVs should meet
product specifications related to “purity, identity,
Impact of the artificial EV classification in the
quantity, potency and sterility” in concordance with
design of new therapeutic agents based on EVs
the pharmaceutical market regulations [28]. Once
The preparation route chosen is important for the final more, there are several important differences in the
purpose of artificial EVs, but it could be critical for the definition of these parameters depending on the semi-
clinical trials and subsequent commercialization. The or fully synthetic character of EVs. These key points
extensive manipulation of EVs during the bio-engi- will be considered and discussed in the following
neering methods is the reason for their classification sections.
as advanced therapy medicinal products (ATMPs) [2],
with a particular regulatory framewok. Following the
Semi-synthetic exosomes: biotechnological
same criteria, fully artificial EVs produced from cul-
modification of naturally released exosomes
tured cells (top-down) should also be incorporated into
this category. The simplest idea to manufacture EVs would be to use
On the other hand, bottom-up artificial mimetic the natural mechanisms for the formation of vesicles,
EVs are more difficult to assign to one or another that is, the cellular machinery itself. It is known that
category. To date, a set of proteins have been fixed the composition of the EVs at all levels responds to a
on lipidic vesicles with an undefined purpose. While high degree of control at very selective cellular
there is some evidence that the EV membrane is mechanisms [29]. Therefore, the composition of the
important for the uptake process [1], the role of the EVs could be controlled by intentional alteration of
artificial membranes is not yet clear. Comparative stu- the cellular environment.
dies of the effects over target cell lines with conven- This method would lead to the creation of EVs with
tional liposomes and exosome-mimetic nanovesicles a composition profile adapted to a specific purpose.
would be very useful to clarify this. The work in this The technological methods used for bioengineering
field is reviewed in the section “Bottom-up methodol- EVs are explained in the following sections. Two key
ogies: artificial membranes decorated with functional aspects are the selection of producer cells (and their in
proteins to mimic EVs functions.” vitro harvesting conditions) and the EV isolation/
A critical evaluation of the fully synthetic EVs con- enrichment procedures. Both choices will condition
cept would imply providing an answer to questions subsequent uses.
such as: “What we are trying to mimic from EVs?”;
“Is the biochemical composition, the morphology or
Selection of EV cellular origin
the whole entity?”; “Is it worth mimicking a specific
function?” This is still a challenge in the field, since Cell lines could potentially release EVs vesicles, but
some of these questions are being answered at the same there are great differences in release rate and biochem-
time for natural EVs. The best approach would involve ical composition and their susceptibility to modifica-
an extensive biochemical characterization of natural tions [30]. It is also accepted that before translation to
EVs and a detailed description of their functions. clinical use of EVs, limitations regarding biocompat-
Regarding functionality, other populations and not ibility, economic viability, harvesting methods and
only the target cell lines should be assayed. This immunotolerance must be overcome. A summary of
could provide information about possible side effects. cell lines used for the production of EVs for clinical
In the same way, multiple parameters should be regis- purposes, especially drug delivery, can be found in the
tered as output factors, not just the process targetted by literature [31]. Dendritic cells and cancer cell lines,
the EV-based treatment. Proteins, nucleic acids and such as melanoma, are the most commonly used lines
lipid composition from specific types of exosomes are for EV production.
registered in specialized databases such as ExoCarta Mesenchymal stem cells (MSCs) are one of the most
[25], EVpedia [26] and Vesiclepedia [27]. But this is promising sources of EVs, especially exosomes [32].
not enough: a database with assays performed with EVs Yeo et al. [33]. defended their use for mass production
reporting treated cell lines, EV type as therapeutical of exosomes with future therapeutic purposes based on
agents, type of assay (in vitro or in vivo) and experi- some important facts related to their advantages over
mental conditions could be of great interest to the other cell lines. Mainly, MSCs are easy to obtain from
4 P. GARCÍA-MANRIQUE ET AL.
all human tissues (even those considered as medical specific cell lines. Evidence about immunotolerance
waste), and they have a high ex vivo expansion capacity should also be provided in order to avoid any inter-
compatible with immortalization without compromis- ference in the results.
ing their therapeutic efficacy. These two facts are essen- In any case, studies involving the encapsulation of
tial to establish a scalable and long-term source of well- the same drug into different cell lines-derived EVs
characterized EVs, particularly exosomes [14,28]. In would be desirable in order to clarify whether the
addition, their immunomodulatory effect gives them beneficial effect is due to the drug or the combination
and their derived EVs important features in autologous of drug/type of carrier.
and allogenic therapeutic applications.
Dendritic cells (DCs) have important roles in immu-
Obtaining a good substrate for modification:
nity (both innate and adaptative). Some authors have
isolation procedures
paid attention to this cell line in order to enhance the
production of clinical grade DC-derived exosomes for Since the final destination of artificial EVs would be the
immunotherapy [33]. Properties of DCs have even administration for therapeutic purposes, the highest
been enhanced with different nanoparticles [34]. level of standards would be required in order to pre-
Exosomes from DCs modified to express indoleamine serve patient safety [27,43]. A key point in artificial
2,3-dioxygenase (IDO), a tryptophan-degrading EVs development is the enrichment from different
enzyme that is important for immune regulation and biological samples, from cell-culture supernatants to
tolerance maintenance, have been used in the treat- several body fluids. There are different reviews [43–
ment of rheumatoid arthritis [35]. In another study, 45] and technique-comparative papers [46–49] about
DC-derived exosomes modified to express FasL on the isolation procedures. They involve ultracentrifugation,
surface (ligand involved in apoptosis induction) were filtration, immunoaffinity isolation, polymeric precipi-
tested as inflammatory and autoimmune therapy [36]. tation and microfluidics techniques, with different
Other recent works are related to the role of tumour- degrees of purity for the final product [50]. In this
released exosomes to load antigens into DCs for the review, we have focussed on scalability, reproducibility
therapy of malignant mesothelioma [37]. and synthetic EV potential damage or physical
To overcome the problem of low release rate, some modification.
authors lowered the micro-environmental pH, mimick- Since the efficient function of EVs depends on their
ing the natural cancer mechanism [38]. By culturing size distribution, aggregation and size changes after
HEK293 cell lines at different pHs, these authors found isolation are important. Lane et al. [47] studied these
that low pH values were best to isolate high amounts of parameters in four isolation methods: two aggregation
exosomes. In spite of the impact of these results, it kits, a density-based method and ultracentrifugation.
would be necessary to test similar effects in non-can- These authors found that some methods kept a con-
cerous cell lines and to determine how the change in stant vesicle size, but large differences were observed
the harvesting conditions affects EV composition regarding yield of isolation (the two sedimentation
(membrane components and cargo). methods gave recovery values two orders of magnitude
Not only human cells have been explored as a source higher than the other methods). Another reported
of EVs: exosomes isolated from bovine milk and loaded obstacle is the co-purification of material with similar
with different drugs were a promising strategy for mass physical characteristics to EVs [42,51,52].
production of therapeutic EVs [39]. They can be easily Scalability is also important, since the batch size is
isolated by differential centrifugation. The biocompat- correlated with the homogeneity of the final product;
ibility of milk-derived exosomes was checked by clin- sometimes small batches are more prone to being sus-
ical biochemical analysis in an animal model by oral ceptible to bias during the process, but on other occa-
gavage during 6 h (short-term toxicity) and 15 days sions, higher batches yield more heterogeneous
(medium-term toxicity). populations due to microenvironments during
In recent years, some attention has been drawn to procedures.
non-animal (especially plant) EVs and their potential The scale-up step with ultracentrifugation (UC) is
use in therapy [40]. In particular, fruit-derived exo- limited by the number of rotor positions and the max-
somes (lemon [41] and grapes [42]) have been isolated, imum sample volume. On the other hand, methods
characterized and tested as beneficial products. Perhaps such as size exclusion chromatography (SEC) are easy
this new source of EVs could be used in the near future to scale up by using large columns, but with the asso-
for the development of EBSSNs following modifica- ciated longer separation time. Pressure application to
tions to express the desired targeting molecules against reduce processing time can disturb EVs [43]. Other
JOURNAL OF EXTRACELLULAR VESICLES 5
methods, such as immunoaffinity isolations, are only Table 1. Classification of techniques for the production of
used for small amounts of original sample because of artificial EVs, mainly exosomes, according to type of final pro-
the high price of the reagents required. Finally, micro- duct (semi- or fully synthetic) and the principle of the obten-
tion mechanism.
fluidic methods [45,53] are promising, with the possi-
Semi-synthetic exosome production: modification of vesicles naturally
bility of being coupled to online analysis [54]. produced by cells
Reproducibility is crucial when the product is going Pre-isolation modification
to be used in the clinical field. Comparative results of Class I Co-localization of cargo and exosomal carrier
moiety thanks to the natural tropism of
UC are difficult to obtain due to the high number of the second
models available in the market, and this could affect Class II Use of sequences (i.e. nucleic acid-based
sequences) for the exosomal biogenesis
the quality of the isolated product [43]. The use of a pathway signalling
fixed instrument would keep low variability between Class III Take advantage of passive loading via
increments of their presence, by genetic
batches [47]. overexpression or active loading of
Welton et al. [55] reported that ready-to-use SEC producer cells
Post-isolation modification
columns could overcome some problems related to Class IV or passive Methods that use passive adsorption of
homemade poured columns [52], thereby avoiding var- methods molecules into external surface of EVs,
iations from column to column, and facilitating robust owing to their hydrophobicity nature
Class V or active methods V.a (Physical methods), based on the creation
protocols to be used routinely. The main problem asso- of transitional alteration in the integrity of
ciated with this method is dilution of EVs in the final EVs that allows cargo to enter the vesicles
by concentration gradient or by passive
sample and the subsequent need for concentration using incorporation during subsequent restoring
precipitating agents or UC. This increases retention of initial status post-stimuli
V.b (Chemical methods), based on induced
time and the possibilities of co-precipitation of other chemical reactions between EVs and cargo
molecules with the same size and physical properties. with or without previous introduction of
functionalization agents into vesicles
Considerable effort has been made in the field of EV Creation of artificial mimetic structures of the natural exosomes
isolation methods, which are still limiting the expan- Type I or top-down bio- Starting from larger substrates (cells) that are
sion of this field. There is not yet a perfect and uni- nanotechnology reduced to units for the creation of
vesicles with reduced size
versal method, and it is also accepted that selection of Type II or bottom-up bio- Starting with individual molecules (lipids,
the isolation method could impact downstream nanotechnology proteins, etc.) that are assembled in a
controlled way for generating complex
steps [56]. structures of higher order
Strategies for biochemical modification In this case, the fusion between the gene of a protein to
Pre-isolation modification using own cellular be incorporated and the gene of an exosomal-localized
machinery protein can be used for the expression of the former on
The advantages and disadvantages of the methods the outer surface of exosomes. This has been referred
applied to incorporate proteins of nucleic acids are to in the literature as Exosome Display technology [63],
shown in Tables 2 and 3. Based on the study of exosome and it enables the manipulation of the protein content
biogenesis-related mechanisms, several methods have of exosomes and the subsequent tailoring of activities.
been developed [50,57–62]. The following criteria have The potential of the method was successfully demon-
been identified for their classification (see Table 1): strated by the production of specific antibodies against
human leukocyte antigen (HLA), a low immunogenic
(I) location of exosomal functional entities, such as antigen [64].
transmembrane proteins and the use of their Lactadherin C1C2 domain was also used for simi-
natural tropism to co-localized the exogenous lar purposes by Zeelemberg et al. [65] and Hartman
element; et al. [66] to induce expression of chicken egg oval-
(II) strategies using molecular mechanisms for the bumin (OVA) peptide and the human epidermal
introduction of exogenous molecules into EVs growth factor receptor 2 antigen (HER2), respec-
for cytosolic delivery; tively. Álvarez-Erviti et al. [67] described a method
(III) increasing the amount of molecules into the cel- of inducing surface expression of the central nervous
lular plasma to be encapsulated by passive system-specific rabies viral glycoprotein (RVG) pep-
mechanism during MVB formation. tide on exosomes isolated from immature dendritic
cells derived from mice. Complementarily, these
Class I methods (Table 2) involve the design of chi- brain-targeted exosomes were loaded with siRNA
meric constructions of proteins by genetic engineering. for the first time by electroporation. The delivery of
6 P. GARCÍA-MANRIQUE ET AL.
GAPDH-siRNA specifically to neurons, microglia moiety was selected by the elevated expression of his
and oligodendrocytes in the brain resulted in a spe- receptor (EGFR) in tumours of epithelial origin.
cific gene knockdown. This was considered the first More recently, Stickney et al. [62] developed another
example of EV-based genetic therapy. One of the genetic engineering method for surface expression of pro-
most important facts of this work was the successful teins in human cells, called surface display technology. In
treatment of a highly protected tissue, the brain, in this case, tetraspanin CD63 was used as a scaffold for the
spite of the existence of brain–blood-barrier presentation in both extravesicular and intravesicular
selectivity. orientations.
Tian et al. [68] used lysosome-associated membrane Class II methods include a heterogeneous group of
glycoprotein 2b to target electroporated doxorubicin- strategies that have in common the use of specific
loaded exosomes (20% of loading efficacy) produced in molecular interactions between two elements and can
dendritic cells. Ohno et al. [69] used platelet-derived be used to transport the complex into the exosomes.
growth factor receptor transmembrane domain to anchor One example of this strategy is the interaction between
GE11 peptide, a ligand of the epidermal growth factor specific sequences in RNA molecules and proteins that are
receptor (EGFR). This construction was transfected to present in the route of exosomes formation [16]. Highly
Human Embryonic Kidney cell line 293 (HEK293) using observed sequence motifs into RNA types studied in EVs,
pDisplay vector and FuGENE HD transfection reagent. called EXOmotifs, were found in mRNA [70] and miRNA
As a model cargo, siRNA let-7 was selected for its ability to [71]. Exosomes for Protein Loading via Optically
alter cell-cycle progression and reduce cell division in Reversible protein–protein interactions (EXPLORs) and
cancer cells. This siRNA was introduced into EV producer Targeted and Molecular EV Loading (TAMEL) are tech-
cells by the lipofection method. Modified exosomes (15– nologies based on the molecular interaction between cer-
21% of total released exosomes) were isolated by centri- tain types of proteins or between RNA special structures
fugation and intravenously injected into an animal model and specific proteins. Proteins of interest can also be loaded
with induced breast cancer. GE11 peptide as the targeting into the inner compartment of exosomes for their direct
JOURNAL OF EXTRACELLULAR VESICLES 7
delivery to the cytosol of the target cell as an alternative for between an EV-enriched protein and an RNA-binding
therapeutic target locations. EXPLORs [61] has recently domain. This construction is transfected into EV pro-
presented for that purpose. The system integrates two ele- ducer cells to make his function. Different loading
ments: one is produced by the genetic fusion of the photo- degrees can be obtained by selecting the EV-enriched
receptor cryptochrome 2 (CRY2) to the protein to be protein. This is related to the natural expression of
loaded, and the other is made by the fusion of a truncated different proteins into EVs.
version of the CRY-interacting basic-helix-loop-helix 1 Class III methods includes the simplest method:
(CIBN) to tetraspanin CD9. Both elements can be transi- passive loading into vesicles through their biogen-
torily attached by exposure to blue light, which induces the esis. There can be two different approximations to
interaction between CRY and CIBN, and the interaction this objective. First, the overexpression of RNA
can be stopped once the blue light is not present. cargo in the producer cells. The major disadvantage
TAMEL is another genetic engineering tool that of this method is the lack of selectivity in the load-
has recently been published [59] for the active cargo ing process, since it is gradient-driven (the higher
of RNA. This platform is a fusion between an engi- the concentration in the cytoplasm, the higher its
neered EV-loading protein and the RNA to be loaded. possibility of being trapped into exosomes during
Engineered EV-loading protein is also a fused product invagination of MVB formation). On the other
8 P. GARCÍA-MANRIQUE ET AL.
hand, the great advantage of the method is that by Physical and chemical post-isolation modifications
translation of mRNA into receptor cell cytoplasm, These are the methods that require an external force
codified proteins can also be passively loaded (chemical or physical) to incorporate new molecules on
into EVs. previously isolated exosomes (Table 3).
The second approximation explores the active load- Passive methods. Incubation of EVs and cargo. The
ing of the producer cells, i.e. by nanocarriers such as simplest way to incorporate any cargo into cell culture
fusogenic liposomes. This strategy is based on phy- or body fluid isolated EVs is the co-incubation of both
sico-chemical properties that govern the type of elements. This strategy was explored by Sun et al. [74],
mechanism (the fusiogenic properties of the two ele- who found that curcumin exosome-loaded exhibited a
ments that take place in the method, cells and lipo- better stability and higher bioavailability in serum in an
somes). Second, they modify the whole cell and not animal model. For these therapeutic-modified exo-
only the exosomes. somes, an improvement in in vivo anti-inflammatory
As an alternative method for the incorporation of and septic shock was observed.
exogenous molecules (specially designed for hydro- In another study [75], two anti-inflammatory com-
phobic compound) into EVs, Lee et al. [72] pre- pounds were loaded into exosomes and microparticles,
sented the use of membrane fusogenic liposomes and they were administrated intranasally, opening up
(MFLs). By the treatment of cells with MFLs loaded new therapeutic possibilities. The effects of solvents
with a hydrophobic compound (DiI) and a hydro- and drug release kinetics from loaded exosomes by
philic molecule (calcein), these authors obtained dialysis have been studied [39,76].
EVs modified with both molecules. Only slight dif- Active methods. Physical methods: electroporation and
ferences in the efficiency of incorporation into EVs other temporary membrane disruptive methods. The
were found, since a hydrophobic compound would most commonly used method for cargo incorporation
remain in the plasma membrane after liposome into EVs after their release is electroporation [77]. This
fusion and in the subsequent formation of EVs technique involves the temporary permeabilization of
membrane through their routes of biogenesis. In membranes through the creation of pores due to the
contrast, a hydrophilic molecule would be released application of high-voltage electricity. Some authors
into the cytosol. Intercellular transport of both have pointed out that this method is not suitable for
molecules mediated by EVs was successfully siRNA cargo into EVs due to technical problems, and
observed in vitro in a multicellular tumour spheroid an overestimated encapsulation into EVs could be
model. observed. It has been reported that electroporation
A similar method was used to modify the composi- induces siRNA aggregation and co-pelletization with
tion of EVs, with a special focus on the modification of EVs during purification by ultracentrifugation, without
the properties of the EV membrane [57]. Dyes, fluor- any dependence on electroporation buffer composition
escent lipids with different lengths and saturation grade [78]. They also postulated that slight differences could
of acyl chains, and chemotherapeutics were loaded into be found between different EVs regarding their cellular
cells by means of EVs. origin (e.g. primary cells).
These authors also carried out a membrane sur- Another relevant problem concerning electropora-
face modification, with the possibility of conjuga- tion is exosome aggregation and subsequent decrease
tion with molecules for targeting purposes. They in functionality. To avoid these problems, Hood et al.
first prepared MFLs containing azide-modified [79] electroporated exosomes from mouse B16-F10
lipids which were fused with cells. By simple incu- melanoma cells by incorporating 5 nm superparamag-
bation with dibenzocyclooctyne (DBCO)-modified netic iron oxide nanoparticles (SPIONs) as model exo-
peptides, a covalent bond was created due to the genous cargo. Other authors compared the loading
fast and selective reaction between DBCO and the efficiency of different porphyrins with different hydro-
azide group [73]. This strategy will allow new pos- phobicities into EVs with various origins by passive
sibilities of surface ligand decoration on EVs for loading (co-incubation), and by active loading, such
targeting purposes (such as peptides, aptens or anti- as electroporation, extrusion, saponin-assisted drug
bodies) or for the introduction of molecules with loading and hypotonic dialysis [58]. The best results
therapeutic properties via interaction with selective were obtained for hydrophobic compounds and for
receptors. By the combination of a different head- electroporation. Interestingly, zeta potential (ζ) related
group modified lipids, several different ligands to the chemical composition of EV membranes seems
could be incorporated into EV outer membranes, to play a role in loading efficiency, since higher ζ values
including receptors and co-receptors. led to higher EE. The chemical properties of cargo are
JOURNAL OF EXTRACELLULAR VESICLES 9
also relevant, since their charge will condition the final and allows the incorporation of exogenous molecules to the
outcome. Electroporation did not induce drug surface of EVs.
precipitation. Finally, there is another type of cargo modification
In contrast, extrusion over polycarbonate mem- that has been applied into artificial vesicular systems
branes altered the morphology of vesicles and, sub- (liposomes) with potential applicability to EVs. This
sequently, their delivery efficacy. Other authors used method is based on the ability of some peptides to be
the sonication of EVs in the presence of drug solu- incorporated into lipidic membranes causing disrup-
tions [60].The loading efficacy was found to follow tion [83,84]. By fusion of the peptide D1-7 to an adhe-
the order: incubation at RT < electroporation ≪ sion molecule expressed in cells, targeted lipidic
mild sonication. A similar trend was observed for carriers with therapeutic cargo were produced and
size changes after the loading procedure. On the successfully tested in vitro and in vivo. Another inter-
other hand, surface charge and protein profile were esting application of this strategy is its ability to insert
similar after loading, evidencing no alteration in peptidic cargo into live cell membranes, giving possi-
exosome stability. These authors explained the bilities of imaging live cells and modifying cell surfaces.
results concerning sonication as a decrease in bilayer This last property could be explored for the modifica-
rigidity after sonication, which allowed a better tion of plasma membrane in EV producer cells.
incorporation of the hydrophobic drug. Therefore, The modification of EV membranes results in
mild sonication should be considered as an enhance- changes in surface charge, fusiogenic properties,
ment of co-incubation. Additionally, loaded exo- immunogenicity decrease and colloidal stability [85].
somes were stable over large periods of time at Engineered hybrid exosomes were prepared by mem-
different temperatures. brane fusion with liposomes formulated with different
A similar comparative study was carried out with types of lipids (i.e. zwitterionic, anionic, cationic and
the enzyme catalase [80]. For the preparation of PEGlilated). Fusion properties with cell-culture-derived
exosomes modified with this oxidative stress-protect- exosomes were studied according to the chemical nat-
ing agent used for the treatment of Parkinson’s dis- ure of liposome lipids [86]. It was found that zwitter-
ease, these authors selected four methods: incubation ioninc and anionic lipids did not alter the uptake rate,
at RT in the presence/absence of saponin, freeze/ while the introduction of cationic lipid greatly
thaw, sonication and extrusion. Sonication yielded decreased the phenomena, and PEGilated lipid
the higher EE (26.1%) and the more stable product. increased it by twofold. Therefore, it can be concluded
In contrast, this method also produced the higher that functional properties could be tuned by modifying
increment in size, from 105 nm naïve exosomes to the membrane composition.
183.7 nm in catalase-loaded exosomes. Associated
with size increment, AFM observation also revealed
a change in morphology, with a final non-spherical Top-down and bottom-up methods for the
shape. development of full synthetic EVs
Despite these promising results regarding the encap- Production of artificial EVs by generation of
sulation of different molecules into exosomes, standar- plasma membrane fragments: a top-down-inspired
dization in systematic conditions followed by the study methodology
of several cell lines is still necessary to strongly support
the use of these methods as routine practice in the Different approaches based on top-down nanotechnol-
clinical field. ogy have been developed for the production of EVs
Chemical methods: click chemistry mediated functionaliza- mimetic nanovesicles using cells as precursors of plasma
tion and other targeted drug-delivery strategies. The che- membrane fragments. Those strategies rely on the prin-
mical copper-catalysed reaction between an alkyne and an ciple of self-assembly of lipids and lipid membranes into
azide that forms a triazole linkage (click chemistry) has been spherical structures and the encapsulation of surround-
used for the surface functionalization of exosomes [81]. ing material into the aqueous cavity of generated nano-
These were first chemically modified by the incorporation vesicles. Current methods include extrusion over
of alkyne groups into amine groups from proteins by membrane filters [16,17,87], hydrophilic microchannels
carbodiimide chemistry [82]. These authors conjugated [88] or cell slicing by SixNy blades [18] (see Table 4).
azide-Fluor 545 (a fluorescent compound) to activated
EVs. Since no differences in morphological and functional Extrusion over polycarbonate membrane filters
properties were found, it was concluded that modification Jang et al. [16] used a serial extrusion through poly-
by click chemistry does not alter exosome characteristics carbonate membrane filters with decreasing pore sizes
10 P. GARCÍA-MANRIQUE ET AL.
Table 4. Summary of the published work about the generation of mimetic EVs nanovesicles by top-down bio-nanotechnology (cell
source and type of cargo are encapsulated, and main characteristics are given).
Type of material
Generation technique Precursor cell type encapsulated Nanovesicles characteristics Reference
Manual extrusion over Monocytes and Exogenous, Mean size and distribution similar to that of exosomes [16]
polycarbonate membrane macrophages chemotherapeutic Exosomal protein profile similar to that of natural exosomes
filters drugs EE of chemotherapeutics dependent on the original amount
used during extrusion
High rate of drug release
100 times more that of nanovesicles than exosomes from the
same number of cells
Results reproducible with different cell types
Pressurization and extrusion Murine embryonic Endogenous, proteins Average size in the exosome range [88]
over hydrophilic parallel stem cells and RNA Similar intracellular and membrane protein and total RNA profile
microchannels in a to the original cells and exosomes
microfluidic device Same ability to deliver RNA content as exosomes
Centrifugal force and Murine embryonic Endogenous, proteins NVs size and morphology similar to exosomes [87]
extrusion over a filter with stem cells and RNA Cargo of RNA, intracellular proteins and plasma membrane
micro-size pores into a proteins similar in types to exosomes
polycarbonate holder Small RNA profile differs in quantity, especially in miRNA with
structure respect to exosomes
Intravesicle contain twice the concentration of natural exosomes
250 times more vesicles than naturally secreted exosomes
Slicing living cell membrane Murine embryonic Exogenous, Generated NVs in the size range of exosomes [18]
with silicon nitride blades stem cells polystyrene latex Nanovesicle production 100 times more productive than natural
in a microfluidic device beads exosome
30% of EE for 22 nm nanoparticles as model of exogenous
material encapsulation
NVs can deliver exogenous encapsulated material
(10 μm, 5 μm and 1 μm) in a mini-extruder, similar to murine embryonic stem cells for the treatment of mice
those commonly used for the preparation of liposomes. isolated skin fibroblasts. These authors wanted to
Human monocytes were chosen as precursors for explore the potential of mimetic exosomes to induce
membrane fragments. The yield production of NVs proliferation and recovery after injury in an in vitro
was 100-fold in comparison with the production of model. Genomic and proteical profiles similar to origi-
exosomes by using the same number of cells. nal cells were assessed by PCR and Western blotting of
Morphological studies of these NVs by crio-TEM and specific markers, and it was confirmed that successful
NTA showed many similarities with the exosomes, delivery of genetic material by NVs was reached.
round shape and a peak diameter around 130 nm.
Even the exosomal protein marker profile (CD63, Pressurization, extrusion and slicing over hydrophilic
Tsg101, moesin and beta-actin) checked by Western parallel microchannels in a microfluidic device
blot was identical for the NVs and exosomes. The Jo et al. [87] produced exosome-mimetic nanovesicles
chemotherapeutic drugs, doxorubicin, 5-FU, gemcita- by extruding cells over hydrophilic microchannels,
bine and carboplatin were added to the buffer where with the aim of delivering endogenous RNA across
cells were resuspended. The encapsulation efficiency in the plasma membrane with high efficiency and low
the final purified NVs was found to be dependent on toxicity. These authors developed a microfluidic device
the initial amount of added drug. made of PDMS by soft lithography. This device had an
Looking for a scaled-up process using extrusion as array of 37 parallel microchannels, with a common
the generation procedure of NVs, Jo et al. [87] devel- inlet and outlet connection for the pumping with a
oped a device that uses centrifugal force to extrude cells syringe pump and the collection of NVs, respectively.
over polycarbonate filters (10 μm and 5 μm pore sizes). The higher amount of nanovesicles generated by extru-
The device has a central piece where filters are located sion over hydrophilic channels with a similar size to
and connected to two syringes where the sample is exosomes was obtained with a length of 200 μm and a
dispensed by the centrifugal force. Uniformly sized width of 5 μm. These results showed that an appro-
100 nm NVs with a yield 250-fold higher than that of priate total shear force induced by the channel has to
exosomes from the same number of cells was achieved. be reached in order to produce NVs with acceptable
Analysis of the filters by TEM showed that many cells results. This force is responsible for NV generation due
remained trapped in the structure. to elongation of the plasma membrane on the micro-
The same device previously cited was employed by channel surface. When the elongated membrane
Jeon et al. [17] to produce exosome-mimetic NVs from reaches a certain value, it breaks into small portions
JOURNAL OF EXTRACELLULAR VESICLES 11
that directly form nanovesicles, thanks to the self- individual molecules (lipids, proteins and cargo) into
assembly property of lipids in aqueous media. complex structures, such as a bilayer structure resem-
With the appropriate channel morphology, these bling EV membranes functionalized with proteins for
authors produced 100 nm nanovesicles similar in com- mimicking EV functions. This could be achieved by
position (proteinical and nucleic acid profile) to natu- assuming that not all components in natural exosomes
rally produced exosomes. The analysis of these NVs are essential for specific and efficient delivery [13],
[88] revealed that the formation of exosome-like NVs including the transport of a message through direct
through hydrophilic channels produced a delivery sys- contact with target cell receptors. Another assumption
tem of endogenous material with identical results to that encourages researchers to explore this route is
those with exosomes. that, from a structural and biochemical point of view,
More recently, Yoon et al. [18] reported the produc- exosomes are liposomes with attached proteins.
tion of exosome-mimetic nanovesicles by the slicing of Therefore, this type of vesicular system could be an
cells through SixNy blades aligned to the flow direction ideal substrate to develop exosome-mimetic structures.
over hydrophilic microchannels. These authors com- The main functional components of exosomes to be
bined the induced shear stress formation on NVs with incorporated in mimetic materials have been reviewed
the fragmentation of plasma membrane by the blades [13]. The three main components of exosomes
to obtain fragments for the generation of exosome-like reported were lipids, membrane proteins and therapeu-
nanovesicles and the co-encapsulation of exogenous tic cargo.
material (polystyrene latex beads as a model sub- One of the main advantages of fully artificial EVs
stance). It was found that NV diameter increased as over previous strategies is the production of pure and
the width channel increased. This is because channel well-defined biomaterial. In addition, production stra-
morphology is proportional to the Reynolds number tegies of artificial EVs based on liposomes are more
(Re). In this particular case, Re is proportional to the sustainble and easier to scale up [11,89]. This fact is
hydraulic diameter and, therefore, to the inertial force, quite important for preclinical and clinical studies and
which directly increases with the channel width. In in order to manufacture products ready to be sent to
other words, NVs travelling through wide channels the market [2].
have a higher inertial force when they reach the blades, Liposome preparation techniques have been exten-
generating larger sliced fragments that produce larger sively reviewed [89–101], but not all the methods yield
NVs. These have a similar composition to that of vesicles suitable for becoming an artificial exosome. It
parental cells and naturally released exosomes by could be considered that only small unilamellar vesicles
those cell lines. (SUVs) are ideal precursors due to their similarities to
One of the most interesting works in the literature natural exosomes (size range and membrane
[18] describes the encapsulation of 22 nm fluorescent disposition).
polystyrene latex beads as an exogenous simulated Methodologies for SUV preparation (Table 5) can
material, adding these nanoparticles to the media be classified according to different criteria [92]. For
where cells were diluted before slicing. With a final example, number of steps to reach SUV. Another
corrected EE of 30%, these NVs containing exogenous classification is based on the physical principle
material were given to fibroblasts in an in vitro experi- applied to prepare vesicles: mechanical processes,
ment. After a period of time, red dots corresponding to organic solvent replacement, detergent removal and
fluorescent beads were detected in the cytoplasm of other techniques as microfluidic-based methods.
fibroblast by confocal microscopy. The delivery effi- Reverse-phase evaporation, ethanol injection method,
ciency of encapsulated beads into exosome-like NVs ether injection method (EtIM), thin-film hydration
was higher than that of bead-aggregated NVs, revealing method (TFH), homogenization techniques, French
that exogenous material delivery with these NVs was press cell extrusion, microfluidization, extrusion over
possible, but the efficiency was still lower than that membranes, ultrasound and membrane contactors are
achieved with parental cell-component generated NVs. some of the techniques developed for SUV
preparation.
All these techniques rely on the self-assembly of
Bottom-up methodologies: artificial membranes
amphiphilic molecules, such as lipids, in ordered struc-
decorated with functional proteins to mimic EV
tures due to their physicochemical behaviour in aqu-
functions
eous media [93,94]. This principle is the basis of
The third option for obtaining artificial EVs is their bottom-up nanotechnology. Vesicles with a size range
construction in a fully synthetic way by assembling similar to that of natural EVs could be obtained [95]
12 P. GARCÍA-MANRIQUE ET AL.
Table 5. Advantages and disadvantage of most frequently used methods for small unilamellar vesicles (SUVs) preparation.
One-step method for Physical method
Method SUVs preparation applied for preparation Advantages Disadvantages
Ether injection Yes Organic solvent Scale-up adapted Not suitable for thermosensitive
method replacement High hydrophobic compound compounds
encapsulation Solvent not suitable for some
No mechanical degradation of biocompounds
compounds
Ethanol Yes Organic solvent Scale-up adapted Ultrasounds are needed when
injection replacement Non-dangerous substances are handled concentrated samples are produced
method High hydrophobic compound Low encapsulation efficacies of low-
encapsulation molecular-weight hydrophilic
No mechanical degradation of compounds
compounds Not suitable for thermosensitive
compounds
Reverse-phase No Emulsification/organic Widely used Frequently used solvents are not suitable
evaporation solvent replacement Suitable for mass production for some biocompounds
Thin-film No Mechanical processes Applied for any type of amphiphilic Difficult to scale up production
hydration molecules Timely and cost-ineffective due to
method High encapsulation of Hydrophilic necessary downsizing techniques
compounds compared to other
methods
Downsizing / Mechanical processes Good reproducibility Product loss associated with clogging of
Techniques Adapted to scale-up requirements membrane by concentrated samples
French press cell
extrusion
Microfluidization
Extrusion over
membranes
Ultrasounds Yes Mechanical processes Simple methodology Degradation of biological compounds
Possibility of being scaled up Scale-up unadapted
when operational variables were oprtimized by design authors have used classical separation methods, such
of experiments. In addition, a wide spectrum of mole- as ultracentrifugation [102] or gel filtration [15,103]
cules with biological activity, independently of their (SEC) with high exclusion limit resins (Sepharose CL-
physicochemical nature (hydrophilic or hydrophobic, 2B, 4B mainly). Dialysis, however, is not used due to
low molecular weight or macromolecules), can be the high molecular weight of biomolecules selected for
incorporated into liposomes, during or after their for- mimicking exosomes.
mation [96]. Undecorated liposomes have also been used in the
Functionalization of liposomes with biomolecules is EV research field as EV models for comparing isolation
possible, owing to the different headgroup-modified efficacy and physical integrity [47], detection by flow
lipids that are available [97]. Headgroup modification cytometry [104] and EV refractive index study [105].
usually includes a molecule of polyethylene glycol as a However, their use as a scaffold for artificial EV devel-
spacer between the functional group and the polar opment could offer new possibilities in basic research
region of the lipid. This avoids the sterical hindrance about EVs and theranostic applications. To date, there
caused by the proximity of biomolecules and liposome have been few examples of this approximation for the
surface. The chemical modification includes the intro- development of mimetic exosomes, and no compara-
duction of different types of functional groups, such as tive results are available owing to the great differences
biotin, amine, maleimide, carboxylic acid, folate, cya- between the methods used. A summary of the main
nur, DBCO, azide and succinyl groups. These groups experimental work on mimicking exosomes by bottom-
determine the crosslinking strategy [98–101] which up nanotechnology is given in Table 6 and briefly
should not compromise the biological function. commented on below.
Bioconjugation should ideally be carried out under The most frequent preparation technique for SUVs
mild conditions, aqueous media and chemoselectivity, as templates for EVs mimicking is the TFH method
and with a high yield. combined with extrusion over polycarbonate mem-
Successful conjugation of peptides/proteins with branes and with [102] or without [15] previous
liposomes can be checked using conventional molecu- freeze–thaw cycles. Martínez-Lostao et al. [102] had a
lar biology techniques such as dot-blot [15], SDS- formulation that included lipids and stoichiometry
PAGE [102] or even flow cytometry [102]. A prelimin- inspired in natural exosomes. The introduction of
ary purification step is required in order to remove only 5% (w/w) of an iminodiacetic acid derivative or
unconjugated biomolecules. For this purpose, the DOGS-NTA allowed the binding of APO2L/TRAIL-
JOURNAL OF EXTRACELLULAR VESICLES 13
Table 6. Summary of published work about the development of mimetic EVs nanovesicles by bottom-up bio-nanotechnology,
showing formulation of the vesicles, molecules for the surface functionalization and main physical characteristic (size).
Formulation Preparation method Conjugation strategy Size Protein for functionalization Reference
PC:SM:Cho:DOGS-NTA Thin-film hydration Ni2+-NTA headgroup 150–200 nm APO2L/TRAIL-His10 [102]
(55:30:10:5) weight ratio method (KCl 100 mM, functionalized lipid +
For fluorescent labelling, 0.25% HEPES 10 mM pH 7.0, histidine-tagged
mole/mole DSPE-RhodB EDTA 0.1 mM; KHE recombinant
buffer) peptides
Filtered and degassed + 37°C, 30 min
extrusion over 200 nm
membranes
PC:Cho:DSPE-PEG:DSPE-PEG-MAL Thin-film hydration Maleimide headgroup 100 nm MHC class I peptide complexes and [15]
(1:0.5:0.04:0.01) Molar ratio method (Hepes 25mM, functionalized lipid + FAB regions against T-cell
For fluorescent labelling, 1% of NaCl 140mM; pH 7.4) Traut’s reagent protein receptors (adhesion, early and
PC amount of DSPE-RhodB Filtered and degassed + activation late activation and survival)
extrusion over 100 nm 1h RT 20/1 ratio
membranes
Micro-emulsion phase Micro-emulsion and Carboxilic group from 82 nm Monoclonal antibody against [103]
PC:CpEL micelle combining ChoS and amine DEC205 antigen expressed on
(7:3, w/w) method + sonication group from protein dendritic cells
Micelle phase step for 3min EDC/NHS
In 10:1 v/v DE:A 4°C for 12 h
DOPE:DC-Cho
(4:1, w/w)
In 1:2 v/v EtOH:DW
His10 to liposomes in a single step. Its bioactivity was a Box–Behnken experimental design to optimize (max-
higher activity than that of the soluble ligand. imize) the EE by adjusting some formulation para-
Moreover, a treatment based on these synthetic exo- meters. The final optimized formulation is
somes achieved 60% of disease improvement in a rheu- summarized in Table 6. In this particular case,
matoid arthritis-induced animal model. In another researchers selected mimetic exosomes for the potential
study, liposome-bound Apo2L/TRAIL overcame the transmission of antigen to DCs by a controlled target
resistance to the soluble ligand exhibited by chemorre- delivery using a conjugated monoclonal antibody anti-
sistive tumour cell mutants [106]. The mechanism of DEC205, a highly expressed receptor on the surface of
action of LUV-TRAIL in haematologic cells [107] was DCs. The introduction of cholesterol succinate in the
also studied using mimetic structures of exosomes. outer layer of the liposomes allowed the bioconjugation
Another approximation to artificial EVs (exosomes) of the Ab by EDC/NHS chemistry.
for therapeutic purposes was carried out by De la Peña Despite the promising results and the advantages of
et al. [15] using a reported formulation [108,109]. The these methods for the development of liposome-based
main components were phosphatidylcholine and cho- artificial EVs, there are several limiting aspects that
lesterol, and headgroup-modified lipids such as DSPE- hinder the transfer to the clinical field. While techno-
PEG DSPE-PEG-MAL. In order to make traceable logical progress has allowed the design, development
NVs, both in vivo and in vitro, a fluorescent lipid was and production of nanomedicines with high pharma-
included in the formulation, and magnetic nanoparti- ceutical grade, their clinical impact has been smaller
cles (SPIONs) were encapsulated during a thin-film than expected due to a lack of sufficient information
hydration step. After optimization of chemical-acti- about in vivo interactions and fates inside the human
vated ligands binding, mimetic SNVs simulating DCs body [11]. Specific challenges [13] are related to the
derived exosomes were successfully tested as new tools functionalization of vesicle surface with a combina-
in basic and clinical immunology. A T-cell expansion tion of functional proteins at the same time because
rate higher than that with previously reported experi- actual methodologies are time-consuming and
ments using conventional methods was achieved. because of the incorporation of nucleic acids with
An innovative methodology for the production of acceptable efficiencies. The dependence of vesicle for-
protein encapsulated nanoliposomes was also reported mulation on parameters such as fusion properties and
[103]. This produced 82 ± 4 nm antibody-coated lipo- stability could be another limit of special relevance to
somes with approximately 93% EE of BSA. The pre- immunotolerance. Finally, the knowledge about key
paration route that combined a micro-emulsion components in exosomes is not yet complete, since
contained the protein to be encapsulated, with micelles, they may vary from one cell line to another. They
in order to create a lipid bilayer formed through layer- could even be health-state-dependent and sensitive
by-layer assembly. In this work, the authors selected a to harvesting conditions.
14 P. GARCÍA-MANRIQUE ET AL.
Despite attempts to mimic the exosome natural lipid pathological conditions, novel opportunities for the
composition, there is a need for actually checking development of enhanced therapeutic biomaterials have
whether the formulation is active and involved in the arisen. These observations could help in the production
expected functions or is just a passive element involved of new materials inspired by natural vesicles, without the
only in the scaffolding of true functional elements. classical inconveniences associated with up-to-date syn-
Comparison of the efficiency of the intended integrated thetic alternatives (liposomes, polymersomes, inorganic
component in differently formulated vesicles could be nanoparticles, etc.). EV-based therapies include tissue
an interesting experiment to elucidate the role of mem- regeneration or immunomodulation, but drug delivery
brane components. Parameters of uptake route and is one of the most promising applications. Production,
incorporation efficiency could also be measured with isolation, modification and purification at a large-scale
different cell lines, in order to assess the role of the clinical grade are the main limitations of EVs becoming a
target cell. Other compounds as an alternative to lipids, true clinically settled therapeutic agent.
with a high grade of biocompatibility, could also be These limitations have promoted the development
used for the formulation of artificial exosome bilayers. of mimetic materials inspired by EVs, the so-called
One option is to use non-ionic surfactants [110] for the artificial EVs. In this article, we have introduced a
preparation of niosome-based artificial exosomes. systematic classification of the types of artificial EVs
These compounds offer several advantages [111] over according to their preparation routes. Two well-defined
lipids, such as price, versatility and sustainability. On strategies have been developed: semi-synthetic or fully
the other hand, their chemical structure offers synthetic products. The first strategy uses natural exo-
enhanced stability from both a chemical and physical somes as precursors that are modified at the moment
point of view. Niosomes with a size range close to that of their biogenesis (pre-isolation modifications),
of EVs can also be produced [95]. whereas the second strategy modifies the vesicles after
In recent years, microfluidics has been playing an their release by cells and their isolation from cultured
important role in the development of enhanced vesi- media or biological fluids. Genetic engineering-based
cular systems, enabling robust and highly controlled modifications, active loading platforms, specific signal-
preparation routes of vesicles [112] and allowing ling sequences for selective sorting or precursor cell
rapid characterization of products [113]. Another modifications with nanomaterials are some of the
important aspect in the development of exosome- methods developed for exosome-based semi-synthetic
based therapy, regarding any preparation route, is the nanovesicle production.
creation of reduced systems for the study of traffic and Fully synthetic vesicles with EVs mimetic properties
delivery into in vivo microenvironments [19]. Again, can be produced by bio-nanotechnology. Top-down
microfluidic-based systems are opening up new possi- techniques that produce vesicles made of membrane
bilities by the development of organ-on-a-chip plat- fragments obtained from the extrusion or slicing of
forms that enable the study of these processes in an cells, or bottom-up techniques that take advantage of
innovative and highly efficient way [114]. supra-molecular chemistry (mainly self-assembly) to
It is expected that microfluidic synthesis of nanove- produce vesicles from individual molecules, represent
sicles will open the path for new artificial EV routes, the technology developed for that purpose.
with the required control of size and EE, and minimal Despite the great potential of artificial EVs, some
consumption of reagents. limitations to their development as therapeutic tools
Other recently explored drug-delivery systems have have been identified. There is no perfect technique,
developed platelet-mimetic nanoparticles by also using and, depending on the final purpose of artificial EVs,
bottom-up technology. These authors have produced combinations of procedures could offer new insights in
unilamellar polymeric nanoparticles functionalized the field. Systematic studies with different cellular ori-
with immunomodulatory and adhesion antigens, and gins and target cell lines would expand and consolidate
they have tested them as another approach to disease- the applications of artificial exosomes. Comparative
targeted delivery [115]. work including the encapsulation into different artifi-
cial vesicles would be interesting in order to identify
effects due to the carrier.
Conclusions and future perspectives
Multidisciplinary teams with complementary actions
Knowledge about all the biological aspects related with in the fields of applied biology, pharmacology, chemi-
EVs, especially exosomes, has opened up new frontiers in cal engineering, material sciences and medicine would
the clinical field. After an explosion of publications in allow the definitive consolidation of these therapeutic
recent years about the role of EVs in physiological and biomaterials in clinical routines.
JOURNAL OF EXTRACELLULAR VESICLES 15
associated with potential exosome marker proteins. [38] Ban JJ, Lee M, Im W, et al. Low pH increases the yield
FEBS Lett. 2009;583:3363–3366. of exosome isolation. Biochem Biophys Res Commun.
[22] Prado N, de Dios Alché J, Casado-Vela J, et al. 2015;461:76–79.
Nanovesicles are secreted during pollen germination [39] Munagala R, Aqil F, Jeyabalan J, et al. Bovine milk-
and pollen tube growth: A possible role in fertilization. derived exosomes for drug delivery. Cancer Lett.
Mol Plant. 2014;7:573–577. 2016;371:48–61.
[23] García-Manrique P, Gutiérrez G, Blanco-López MC. [40] Zhang M, Viennois E, Xu C, et al. Plant derived edible
Fully Artificial Exosomes: towards New Theranostic nanoparticles as a new therapeutic approach against
Biomaterials. Trends Biotechnol. Forthcoming. diseases. Tissue Barriers. 2016;4(2):e1134415.
[24] Laulagnier K, Motta C, Hamdi S, et al. Mast cell- and [41] Raimondo S, Naselli F, Fontana S, et al. Citrus limon-
dendritic cell-derived exosomes display a specific lipid derived nanovesicles inhibit cancer cell proliferation and
composition and an unusual membrane organization. suppress CML xenograft growth by inducing TRAIL-
Biochem J. 2004;380:161–171. mediated cell death. Oncotarget. 2015;6:19514–19527.
[25] Simpson RJ, Kalra H, Mathivanan S. ExoCarta as a [42] Pérez-Bermúdez P, Blesa J, Soriano JM, et al.
resource for exosomal research. J Extracell Vesicles. Extracellular vesicles in food: experimental evidence
2012;1:18374. of their secretion in grape fruits. Eur J Pharm Sci.
[26] Kim DK, Lee J, Simpson RJ, et al. EVpedia: A com- 2017;98:40–50.
munity web resource for prokaryotic and eukaryotic [43] Witwer KW, Buzás EI, Bemis LT, et al. Standardization
extracellular vesicles research. Semin Cell Dev Biol. of sample collection, isolation and analysis methods in
2015;40:4–7. extracellular vesicle research. J Extracell Vesicles.
[27] Kalra H, Simpson RJ, Ji H, et al. Vesiclepedia: A 2013;2:20360.
compendium for extracellular vesicles with continuous [44] Sunkara V, Woo HK, Cho YK. Emerging techniques in
community annotation. PLoS Biol. 2012;10:e1001450. the isolation and characterization of extracellular vesi-
[28] Gimona M, Pachler K, Laner-Plamberger S, et al. cles and their roles in cancer diagnostics and prognos-
Manufacturing of human extracellular vesicle-based tics. Analyst. 2016;141:371–381.
therapeutics for clinical use. Int J Mol Sci. [45] Liga A, Vliegenthart ADB, Oosthuyzen W, et al.
2017;18:1190. Exosome isolation: A microfluidic road-map. Lab
[29] Villarroya-Beltri C, Baixauli F, Gutiérrez-Vázquez C, Chip. 2015;15:2388–2394.
et al. Sorting it out: regulation of exosome loading. [46] Andreu Z, Rivas E, Sanguino-Pascual A, et al.
Sem Cancer Biol. 2014;28:3–13. Comparative analysis of EV isolation procedures for
[30] Yeo RWY, Lai RC, Zhang B, et al. Mesenchymal stem miRNAs detection in serum samples. J Extracell
cell: an efficient mass producer of exosomes for drug Vesicles. 2016;5:31655.
delivery. Adv Drug Deliv Rev. 2013;65:336–341. [47] Lane RE, Korbie D, Anderson W, et al. Analysis of
[31] Johnsen KB, Gudbergsson JM, Skov MN, et al. A exosome purification methods using a model liposome
comprehensive overview of exosomes as drug delivery system and tunable-resistive pulse sensing. Sci Rep.
vehicles—endogenous nanocarriers for targeted cancer 2015;5:1–7.
therapy. Biochim Biophys Acta Reviews Cancer. [48] Tauro BJ, Greening DW, Mathias RA, et al.
2014;1846:75–87. Comparison of ultracentrifugation, density gradient
[32] Lai RC, Yeo RWY, Tan KH, et al. Exosomes for drug separation, and immunoaffinity capture methods for
delivery—a novel application for the mesenchymal isolating human colon cancer cell line LIM1863-
stem cell. Biotechnol Adv. 2013;31:543–551. derived exosomes. Methods. 2012;56:293–304.
[33] Lamparski HG, Metha-Damani A, Yao JY, et al. [49] Greening DW, Xu R, Ji H, et al. A protocol for exo-
Production and characterization of clinical grade exo- some isolation and characterization: evaluation of
somes derived from dendritic cells. J Immunol ultracentrifugation, density-gradient separation, and
Methods. 2002;270:211–226. immunoaffinity capture methods. In: Posch A, editor.
[34] Klippstein R, Pozo D. Nanotechnology-based manip- Proteomic profiling: methods and protocols. New
ulation of dendritic cells for enhanced immunotherapy York: Springer; 2015. p. 179–209.
strategies. Nanomed Nanotechnol Biol Med. [50] Marcus ME, Leonard JN. FedExosomes: engineering
2010;6:523–529. therapeutic biological nanoparticles that truly deliver.
[35] Bianco NR, Kim SH, Ruffner MA, et al. Therapeutic Pharmaceuticals. 2013;6:659–680.
effect of exosomes from indoleamine 2,3-dioxygenase– [51] Yamada T, Inoshima Y, Matsuda T, et al. Comparison
positive dendritic cells in collagen-induced arthritis of methods for isolating exosomes from bovine milk. J
and delayed-type hypersensitivity disease models. Vet Med Sci. 2012;74:1523–1525.
Arthritis Rheumatol. 2009;60:380–389. [52] Böing AN, van der Pol E, Grootemaat AE, et al. Single-
[36] Hee Kim S, Bianco N, Menon R, et al. Exosomes step isolation of extracellular vesicles by size-exclusion
derived from genetically modified DC expressing chromatography. J Extracell Vesicles. 2014;3:23430.
FasL are anti-inflammatory and immunosuppressive. [53] Davies RT, Kim J, Jang SC, et al. Microfluidic filtration
Mol Ther. 2006;13:289–300. system to isolate extracellular vesicles from blood. Lab
[37] Mahaweni N, Lambers M, Dekkers J, et al. Tumour- Chip. 2012;12:5202–5210.
derived exosomes as antigen delivery carriers in den- [54] He M, Crow J, Roth M, et al. Integrated immunoisola-
dritic cell-based immunotherapy for malignant tion and protein analysis of circulating exosomes using
mesothelioma. J Extracell Vesicles. 2013;2:22492. microfluidic technology. Lab Chip. 2014;14:3773–3780.
JOURNAL OF EXTRACELLULAR VESICLES 17
[55] Welton JL, Webber JP, Botos LA, et al. Ready-made [72] Lee J, Kim J, Jeong M, et al. Liposome-based engineer-
chromatography columns for extracellular vesicle iso- ing of cells to package hydrophobic compounds in
lation from plasma. J Extracell Vesicles. 2015;4:27269. membrane vesicles for tumor penetration. Nano Lett.
[56] Taylor DD, Shah S. Methods of isolating extracellular 2015;15:2938–2944.
vesicles impact down-stream analyses of their cargoes. [73] Chang PV, Prescher JA, Sletten EM, et al. Copper-free
Methods. 2015;87:3–10. click chemistry in living animals. Proc Natl Acad Sci
[57] Lee J, Lee H, Goh U, et al. Cellular engineering with USA. 2010;107:1821–1826.
membrane fusogenic liposomes to produce functiona- [74] Sun D, Zhuang X, Xiang X, et al. A novel nanoparticle
lized extracellular vesicles. ACS Appl Mat Interfaces. drug delivery system: the anti-inflammatory activity of
2016;8:6790–6795. curcumin is enhanced when encapsulated in exosomes.
[58] Fuhrmann G, Serio A, Mazo M, et al. Active loading Mol Ther. 2010;18:1606–1614.
into extracellular vesicles significantly improves the [75] Zhuang X, Xiang X, Grizzle W, et al. Treatment of
cellular uptake and photodynamic effect of porphyrins. brain inflammatory diseases by delivering exosome
J Control Release. 2015;205:35–44. encapsulated anti-inflammatory drugs from the nasal
[59] Hung ME, Leonard JN. A platform for actively loading region to the brain. Mol Ther. 2011;19:1769–1779.
cargo RNA to elucidate limiting steps in EV-mediated [76] Aqil F, Kausar H, Agrawal AK, et al. Exosomal for-
delivery. J Extracell Vesicles. 2016;5:31027. mulation enhances therapeutic response of celastrol
[60] Kim MS, Haney MJ, Zhao Y, et al. Development of against lung cancer. Exp Mol Pathol. 2016;101:12–21.
exosome-encapsulated paclitaxel to overcome MDR in [77] Weaver JC. Electroporation: A general phenomenon
cancer cells. Nanomed Nanotechnol Biol Med. 2016;12 for manipulating cells and tissues. J Cell Biochem.
(3):655–664. 1993;51:426–435.
[61] Yim N, Ryu SW, Choi K, et al. Exosome engineering [78] Kooijmans SAA, Stremersch S, Braeckmans K, et al.
for efficient intracellular delivery of soluble proteins Electroporation-induced siRNA precipitation obscures
using optically reversible protein–protein interaction the efficiency of siRNA loading into extracellular vesi-
module. Nat Commun. 2016;7:1–9. cles. J Control Release. 2013;172:229–238.
[62] Stickney Z, Losacco J, McDevitt S, et al. Development [79] Hood JL, Scott MJ, Wickline SA. Maximizing exosome
of exosome surface display technology in living colloidal stability following electroporation. Anal
human cells. Biochem Biophys Res Commun. Biochem. 2014;448:41–49.
2016;472(1):53–59. [80] Haney MJ, Klyachko NL, Zhao Y, et al. Exosomes as
[63] Delcayre A, Estelles A, Sperinde J, et al. Exosome dis- drug delivery vehicles for Parkinson’s disease therapy.
play technology: applications to the development of J Control Release. 2015;207:18–30.
new diagnostics and therapeutics. Blood Cells, Mol [81] Smyth T, Petrova K, Payton NM, et al. Surface func-
Dis. 2005;35:158–168. tionalization of exosomes using click chemistry.
[64] Delcayre A inventor, Le PJB inventor; Methods and Bioconjug Chem. 2014;25:1777–1784.
compounds for the targeting of protein to exosomes. [82] Hermanson GT. Zero-length crosslinkers. In:
European patent WO2003016522 A2. 2003. Hermanson GT, editor. Bioconjugate Techniques. 3rd
[65] Zeelenberg IS, Ostrowski M, Krumeich S, et al. ed. Boston: Academic Press; 2013. p. 259–273.
Targeting tumor antigens to secreted membrane vesi- [83] Sessa G, Freer JH, Colacicco G, et al. Interaction of a
cles in vivo induces efficient antitumor immune lytic polypeptide, melittin, with lipid membrane sys-
responses. Cancer Res. 2008;68:1228–1235. tems. J Biol Chem. 1969;244:3575–3582.
[66] Hartman ZC, Wei J, Glass OK, et al. Increasing vaccine [84] Pan H, Myerson JW, Ivashyna O, et al. Lipid mem-
potency through exosome antigen targeting. Vaccine. brane editing with peptide cargo linkers in cells and
2011;29:9361–9367. synthetic nanostructures. Faseb J. 2010;24:2928–2937.
[67] Alvarez-Erviti L, Seow Y, Yin H, et al. Delivery of [85] Sato YT, Umezaki K, Sawada S, et al. Engineering
siRNA to the mouse brain by systemic injection of hybrid exosomes by membrane fusion with liposomes.
targeted exosomes. Nat Biotechnol. 2011;29:341–345. Sci Rep. 2016;6:1–11.
[68] Tian Y, Li S, Song J, et al. A doxorubicin delivery [86] Morris GJ, McGrath JJ. The response of multilamellar
platform using engineered natural membrane vesicle liposomes to freezing and thawing. Cryobiology.
exosomes for targeted tumor therapy. Biomaterials. 1981;18:390–398.
2014;35:2383–2390. [87] Jo W, Kim J, Yoon J, et al. Large-scale generation of cell-
[69] Ohno SI, Takanashi M, Sudo K, et al. Systemically derived nanovesicles. Nanoscale. 2014;6:12056–12064.
injected exosomes targeted to EGFR deliver antitumor [88] Jo W, Jeong D, Kim J, et al. Microfluidic fabrication of
microRNA to breast cancer cells. Mol Ther. cell-derived nanovesicles as endogenous RNA carriers.
2013;21:185–191. Lab Chip. 2014;14:1261–1269.
[70] Bolukbasi MF, Mizrak A, Ozdener GB, et al. miR-1289 [89] Wagner A, Vorauer-Uhl K. Liposome technology for
and “Zipcode”-like sequence enrich mRNAs in micro- industrial purposes. J Drug Deliv. 2011;2011:1–9.
vesicles. Mol Ther Nucleic Acids. 2012;1:1–10. [90] Mozafari MR. Liposomes: an overview of manufactur-
[71] Villarroya-Beltri C, Gutiérrez-Vázquez C, Sánchez- ing techniques. Cell Mol Biol Lett. 2015;10:711–719.
Cabo F, et al. Sumoylated hnRNPA2B1 controls the [91] Szoka JF, Papahadjopoulos D. Comparative properties
sorting of miRNAs into exosomes through binding to and methods of preparation of lipid vesicles (lipo-
specific motifs. Nat Commun. 2013;4:1–10. somes). Annu Rev Biophys Bioeng. 1980;9(1):467–508.
18 P. GARCÍA-MANRIQUE ET AL.
[92] Abdus S, Sultana Y, Aqil M. Liposomal drug delivery [105] Gardiner C, Shaw M, Hole P, et al. Measurement of
systems: an update review. Curr Drug Deliv. refractive index by nanoparticle tracking analysis
2007;4:297–305. reveals heterogeneity in extracellular vesicles. J
[93] Antonietti M, Förster S. Vesicles and liposomes: A self- Extracell Vesicles. 2014;3:25361.
assembly principle beyond lipids. Adv Mat. [106] De Miguel D, Basáñez G, Sánchez D, et al. Liposomes
2003;15:1323–1333. decorated with Apo2L/TRAIL overcome chemoresis-
[94] Lasic DD. The mechanism of vesicle formation. tance of human hematologic tumor cells. Mol Pharm.
Biochem J. 1988;256:1–11. 2013;10:893–904.
[95] García-Manrique P, Matos M, Gutiérrez G, et al. Using [107] de Miguel D, Gallego-Lleyda A, Galan-Malo P, et al.
factorial experimental design to prepare size-tuned Immunotherapy with liposome-bound TRAIL over-
nanovesicles. Ind Eng Chem Res. 2016;55:9164–9175. comes partial protection to soluble TRAIL-induced
[96] Gregoriadis G, editor. Entrapment of drugs and other apoptosis offered by down-regulation of Bim in leuke-
materials into liposomes.Volume II, Liposome mic cells. Clin Trans Oncol. 2015;17:657–667.
Technology. Boca Ratonn: CRC Press; 2007. [108] Pagnan G, Stuart DD, Pastorino F, et al. Delivery of
[97] Avanti Polar Lipids products. Available from: http:// c-myb antisense oligodeoxynucleotides to human neu-
www.avantilipids.com roblastoma cells via disialoganglioside GD2-Targeted
[98] Sullivan SM, Connor J, Huang L. Immunoliposomes: immunoliposomes: antitumor effects. J Natl Cancer
preparation, properties, and applications. Med Res Inst. 2000;92:253–261.
Rev. 1986;6:171–195. [109] Pastorino F, Brignole C, Marimpietri D, et al.
[99] Hermanson GT. The reactions of bioconjugation. In: Doxorubicin-loaded Fab′ fragments of anti-disialogan-
Hermanson GT, editor. Bioconjugate techniques. 3rd glioside immunoliposomes selectively inhibit the
ed. Boston: Academic Press; 2013. p. 229–258. growth and dissemination of human neuroblastoma
[100] Nobs L, Buchegger F, Gurny R, et al. Current methods in nude mice. Cancer Res. 2003;63:86–92.
for attaching targeting ligands to liposomes and nano- [110] Marianecci C, Di Marzio L, Rinaldi F, et al. Niosomes
particles. J Pharm Sci. 2004;93:1980–1992. from 80s to present: the state of the art. Adv Colloid
[101] Schuber F. Chemistry of ligand-coupling to liposomes. Interface Sci. 2014;205:187–206.
In: Schuber F, Philippot JR, editors. Liposomes as tools [111] Moghassemi S, Hadjizadeh A. Nano-niosomes as
in basic research and industry. Boca Raton: CRC Press; nanoscale drug delivery systems: an illustrated review.
1995. p. 21–39. J.Control Release. 2014;185:22–36.
[102] Martinez-Lostao L, García-Alvarez FC, Basáñez G, [112] Capretto L, Carugo D, Mazzitelli S, et al. Microfluidic
et al. Liposome-bound APO2L/TRAIL is an effective and lab-on-a-chip preparation routes for organic
treatment in a rabbit model of rheumatoid arthritis. nanoparticles and vesicular systems for nanomedicine
Arthritis Rheum. 2010;62:2272–2282. applications. Adv Drug Deliv Rev. 2013;65:1496–1532.
[103] Li K, Chang S, Wang Z, et al. A novel micro-emulsion [113] Birnbaumer G, Kupcu S, Jungreuthmayer C, et al.
and micelle assembling method to prepare DEC205 Rapid liposome quality assessment using a lab-on-a-
monoclonal antibody coupled cationic nanoliposomes chip. Lab Chip. 2011;11:2753–2762.
for simulating exosomes to target dendritic cells. Int J [114] Bhise NS, Ribas J, Manoharan V, et al. Organ-on-a-
Pharm. 2015;491:105–112. chip platforms for studying drug delivery systems. J
[104] Chandler WL, Yeung W, Tait JF. A new microparticle Control Release. 2014;190:82–93.
size calibration standard for use in measuring smaller [115] Hu CMJ, Fang RH, Wang KC, et al. Nanoparticle
microparticles using a new flow cytometer. J Throm biointerfacing by platelet membrane cloaking. Nature.
Haemost. 2011;9:1216–1224. 2015;526:118–121.
Colloids and Surfaces B: Biointerfaces 182 (2019) 110378
A R T I C LE I N FO A B S T R A C T
Keywords: The new roles of vesicular systems in advanced biomedical, analytical and food science applications demand
Organic colloids novel preparation processes designed to reach the new standards. Particle size and monodispersity have become
Niosomes essential properties to control. In this work, key parameters, involved in a microfluidic reactor with hydro-
Size control dynamic flow focusing, were investigated in order to quantify their effects on niosomes morphology. Particular
Hydrodynamic flow-focussing
attention was given to temperature, which is both a requirement to handle non-ionic surfactants with phase
Microreactor
transition temperature above RT, and a tailoring variable for size and monodispersity control. With this aim,
3D-printing
niosomes with two different sorbitan esters and cholesterol as stabilizer were formulated. High resolution and
conventional 3D-printing technologies were employed for the fabrication of microfluidic reactor and thermo-
static systems, since this additive technology has been essential for microfluidics development in terms of cost-
effective and rapid prototyping. A customised device to control temperature and facilitate visualization of the
process was developed, which can be easily coupled with commercial inverted microscopes. The results de-
monstrated the capability of microfluidic production of niosomes within the full range of non-ionic surfactants
and membrane stabilizers.
⁎
Corresponding authors.
E-mail addresses: [email protected] (X. Zhang), [email protected] (M.C. Blanco-López).
https://doi.org/10.1016/j.colsurfb.2019.110378
Received 14 March 2019; Received in revised form 16 June 2019; Accepted 17 July 2019
Available online 18 July 2019
0927-7765/ © 2019 Elsevier B.V. All rights reserved.
P. García-Manrique, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110378
members differ in terms of acyl chain length and saturation, with a big facilities [27]. Among the techniques explored, additive manufacturing,
range of hydrophilic-lipophilic balance values (HLB), where HLB is an especially 3D-printing, has emerged as a promising method for micro-
important parameter with implications in drug encapsulation efficiency fluidic device manufacturing [28]. The rapid development of 3D-
and morphological characteristics of particles. This parameter is also printing technology and the commercialization of desk printers have
related to the physical state at room temperature (RT), and influences enabled researchers to explore its utility in microfluidic prototyping
the minimum temperature (together with gel-to-liquid transition tem- and manufacturing [29–31], that generally use low cost raw materials
perature, or Tc) that is required at the very stage of the particle for- and can print objects with desired resolution.
mation. On the other hand, some of the compounds used in formula- The aim of the present work was to develop a thermostatic micro-
tions with great loading capacity, low release rate and stability in reactor platform for the continuous flow production of niosomes in a
solution are solid at RT. For these reasons, a higher and controlled size-controllable manner. The microfluidic reactor was designed with a
temperature level is mandatory for this process. hydrodynamic flow focusing configuration, and fabricated in order to
Microfluidics technology is very promising for precise control over allow visualization of the dynamic process including molecular diffu-
input variables when mixing chemical species [17]. Other advantages sion, with the aid of an inverted microscope and a digital image ac-
include low consumption of chemicals (relevant in formulation opti- quisition system. 3D-printing technology was used for fabricating the
mization), scale-up possibilities for industrial production, on-line cou- microfluidic device (positive mould) and thermostatic system. The ef-
pling to other processes (such as purification steps), and efficient con- fect of operational parameters was investigated on the final morpho-
trol over temperature if required [18]. Jahn et al. [19] reported for the logical characteristic of niosomes. Niosomes were formulated with non-
first time the hydrodynamic flow focussing (HFF) technique (Fig. 1) for ionic surfactants with different transition temperatures (Tm) with con-
liposomes production. Following that, other researchers have used this trolled temperature as a tailoring parameter to tune the size and
method to examine various liposomes formulations and for en- homogeneity of particles.
capsulating either hydrophobic or hydrophilic molecules [20,21].
Under laminar flow conditions within the HFF configuration, a stream 2. Materials and methods
of lipids in organic phase is focussed between two aqueous streams in
microchannels, allowing the mixing of chemical species by molecular 2.1. Materials
diffusion. At the two organic/aqueous interfaces, bilayers can be
formed and self-assembled into liposomes once a critical concentration Sorbitan monostearate or Span® 60 (Sigma-Aldrich), sorbitan
is reached. By controlling the flow, the extension of mixing and hence monolaureate or Span® 20 (Sigma-Aldrich), cholesterol from lamb wool
the size of liposomes, could also be controlled. However, the production (Akros Organics), Phosphate Buffer Saline (10 mM, pH 7.4) prepared
of niosomes through microfluidic routes remains less explored, and from tablets according to manufacture instructions (Sigma-Aldrich),
limited attention has been paid to using HFF technique [22–24]. Bromoxylenol blue (Sigma-Aldrich), and technical grade solvents such
At present, the high temperature required for the preparation of as ethanol absolute, 2-propanol (or isopropyl alcohol, IPA), and acetone
niosomes has not been well taken into account in microfluidics routes. (all from J.T. Baker, Avantor, USA) were used in this work. Ultrapure
For example, the previous work that firstly explored microfluidics as- water was used for all experiments. Poly(dimethylsiloxane) monomer
sembly of niosomes faced such temperature related challenge, thus only Sylgard® 184 or PDMS was purchased from Dow Corning Corporation
included Span® 20 and Span® 80 (Tm =25 °C and -30 °C, respectively) in (Auburn, AL, USA). Other materials used for devices fabrication are
the study. [22] specified in the following respective sections.
Along with the wide application of continuous flow microreactors
for organic colloids preparation [18] is the development of micro-
reactor itself, including design and manufacturing of such micro- 2.2. Thermostatic system fabrication
devices, with simpler and more affordable production methods [25]. As
a result, some traditional fabrications methods which stem from the Thermostatic chamber was design in Autodesk® Inventor® and 3D-
photo-electronics field, such as photolithography [26], are being sub- printed with PLA filaments using a special printer for fused deposition
stituted by new processes that require less expensive equipment and modelling (Ultimaker 2 + 3D printer, Ultimaker B.V., The
can be performed in common labs with no need for clean rooms Netherlands). Main chamber and cap of the device were produced se-
parately. A microscope glass slide of 50 x 70 mm (Corning® microscope
2
P. García-Manrique, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110378
slides, Sigma-Aldrich, Gillingham, UK) was sealed to the chamber with 2.4. Niosomes production and morphological characterization
a 2-phase adhesive glue special for plastic materials, bought in a local
store. A transparent piece of plastic was glued to the cap aperture with Working solutions of 5 and 20 mM of Span® 60:cholesterol and
the same adhesive used with the other piece. Teflon tape was used to Span® 20:cholesterol (1:0.5 molar ratio) were prepared by dilution from
enhance the closure of both elements in a removable way. Holes for the a 50 mM stock solution. Ethanol absolute was used as organic solvent,
inlets and outlet pipes of the microfluidic device were manually pre- since it is miscible in aqueous buffer (PBS, 10 mM pH 7.4). Aqueous and
pared with a sharp tool. organic phases were pumped into microfluidic device once appropriate
The previously described chamber was connected to a temperature- temperature was reached. Three different total flow rates (QT) were
controllable recirculation system (F12-MC, Julabo GmbH, Germany) studied (50, 100 and 200 μL/min), and aqueous:organic flow rates were
through the inlet, and a peristaltic pump (MasterFlex®, Cole-Parmer adjusted to five different flow rates ratios (FRR) (5, 15, 25, 35 and 50).
Instruments Company, USA) through the outlet. The plastic pipes were Span® 20:cholesterol formulation was injected at 30, 40, 50 and 60 °C;
those from the recirculator, and connections to the chamber were made while Span® 60: cholesterol was only injected at 50 °C. All the combi-
with common plastic adapters (see supplementary material). nation of membrane components concentration, QT, FRR, and tem-
External supply of the recirculator was set approximately at a flow perature was conducted by duplicate.
rate of 55% of the total volume, while peristaltic pump revolution rate A total volume of 2.5 mL was collected from the outlet of the device
was adjusted to remove water from the chamber at a rate that allowed a for each experimental condition in a glass vial. Size (z-average or peak
continuous and constant flow through it. Temperature inside the value, depending on the number of peaks in the size distribution) and
chamber was monitored with a digital temperature probe (Testo 110, homogeneity (PDI) of particles were measured by Dynamic Light
Testo SE & Co., Germany). The sensor probe was introduced into the Scattering (DLS) in a Zetasizer NANO-ZS equipment (Malvern
chamber through a hole placed in one side of plastic window of the cap Instruments Ltd, Malvern, UK). Samples were measured undiluted by
(see Fig. S2). triplicate, with the 173° backscatter detector in disposable low volume
cuvettes (Malvern Instruments Ltd, Malvern, UK).
2.3. Microfluidic devices manufacturing and channel characterization
2.5. Mixing efficiency visualization
Master mould of devices was designed in Solidworks® CAD 2016
software and 3D-printed onto VeroClear™ resin with the HR-3D printer Solvent and no-solvent diffusion by hydrodynamic flow focusing
Objet350 Connex™ (Stratasys Ltd., USA). A post-printing process was was monitored by an adaption of a previous published methodology
also needed. First, mould was flushed with (I) IPA, (II) deionized water, [32]. Briefly, a change in colour of a pH indicator dye (bromoxylenol
(III) acetone, and finally compressed air. Then, it was cured overnight blue) was used, since this dye exhibits a strong yellowish colour at pH
at 60 °C, and on the following day a treatment of the inner surface was below 6.0 and blue at pH above 7.6. A saturated solution of dye in
carried out with Aquapeel® (to avoid interference of the resin with absolute ethanol acidified with acetic acid was focused by PBS adjusted
PDMS curing process). Three individual moulds were printed. to pH 10.0 with 2 M NaOH solution. Once focused, a change in colour of
Once the positive mould was ready, a mixture of degassed PDMS the stream from yellow to blue indicated a molar fraction of aqueous
curing agent (1:10 w/w) was poured into it, and left overnight in an phase close to one, and then, completes mixing by diffusion.
oven at 40 °C. For degassing the PDMS mixture, a bench centrifuge was
used at 3000 rpm for 10 min. It should be noted that pouring into 3. Results and discussion
master mould must be done slowly to minimise bubble formation. On
the following day, the replica of the mould was carefully peeled off With the microreactor platform developed, systematic character-
from the mould, and inlets and outlet holes were prepared with a isation and operation were conducted in terms of 3D printing outcomes
1.5 mm biopsy punch with plunger (Miltex®, Fischer Scientific, UK). and nanoproduction, as detailed below. (The performance and opti-
Oxygen plasma (PVA-TePla 300 plasma cleaner, Wettenberg, mization of the thermostatic system are described in the Supplementary
Germany) treatment was applied to bond a microscope glass slide material, Fig. S1).
(50 x 70 mm; Corning® microscope slides, Sigma-Aldrich, Gillingham,
UK) to the PDMS replica to complete the microfluidic channel. Four 3.1. High resolution 3D-printing of master moulds for microfluidic devices
pieces of thermic resistant plastic (Ø 8 mm and 3 mm height) were fabrication
glued in each corner at the bottom of the glass slide, to elevate the
device allowing a flow of hot water under the channels. As a key element of the device, mixing channel morphology was
Polytetrafluoroethylene (PFTE, 0.5 mm I.D.) pipes (Cole-Parmer, characterized by mechanical profilometry onto 3D-printed positive
UK) were inserted into the holes, and the other end was attached to a moulds. A considerable difference in nominal dimensions between
syringe needle to create a connection for introduction of the fluids from Computer Aided Design (CAD) and printed object was observed (Table
syringe pumps (NE-300, NEW ERA Pump Systems Inc., USA). Luer lock S1). With an original squared cross sectional geometry of 100 μm width
syringes (Becton, Dickinson and Company, UK) of 1, 10 or 20 mL were and 100 μm height, printed features onto VeroClear® resin showed a
used depending on the selected Flow Rate Ratio (FRR), i.e. volumetric curved morphology five times wider and approximately half of the
flow rate of total aqueous phase/volumetric flow rate of organic phase. height. At the same time, variations in width and height of the mixing
The mixing channel (23 mm long) on the 3D-printed positive mould channel were found between the three 3D-printed positive moulds (see
was characterized in terms of morphology, accuracy and reproduci- Table S1) even following the same fabrication procedure. However,
bility by mechanical profilometry (Talysurf-120 L, Taylor-Hobson, these dimensions were reasonably constant along the mixing channel
United Kingdom). Three equidistance measurements were taken (2 mm length, especially for channel height.
across the channel, perpendicular to it), and data were processed with A possible explanation for these variations in channel dimensions
OriginPro 18 (OriginLab Corporation, USA) software. could be related to printer operational parameters. Objet350 Connex3
The whole setup (microfluidic device inside the thermostatic printer used Polyjet™ inkjet-head patented technology for a layer-by-
chamber with respective inlets and outlets) was placed over the stage of layer process based on Stereolithography [33]. The jetting head dis-
an inverted microscope (IN200TAB series, AmScope, USA) with a di- pensed a proper amount of a photopolymer resin onto a build tray and
gital imaging system to capture images (5 M.P USB CCD camera, instantly cured them with UV light. The process took place in XY-axes
AmScope, USA) supported with the software supplied by the camera to create a 2D sheet (down to 16 microns thickness), and by lowering
manufacturer. The entire experimental setup is illustrated in Fig. S2. the build tray, another layer was created over the previous one. The
3
P. García-Manrique, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110378
cycle was repeated until the whole design was completed. With a re- preparation routes [18], and with important advantages such as better
solution of 600 × 600 × 1600 dpi (X-Y-Z-axes respectively) and an control over particle preparation and the subsequent final character-
accuracy of 20–85 microns for features below 50 mm (up to 200 mi- istics (size and monodispersity, i.e.). This is important for biomedical
crons for full model size), the final features depended on geometry [1], food [35] and analytical chemistry [2] applications. In this regard,
(proximity between elements), build parameters (exposure time, the influence of operational conditions over particles physical proper-
printing speed) and model orientation [29]. Comina et al. [29] reported ties was tested by analysing the results of 3 total flow rates (QT), two
the successful printing of positive moulds for microfluidics devices with different concentrations of bilayer components, for 5 different FRR.
elements from 50 μm to 2 mm, however, some artefacts were described Particle size (nm) and size distribution (PDI) were measured by DLS as
between close elements with 50 μm in dimension differences, though output variables. All the combinations were conducted at 50 °C, a
working with optimized parameters. Unfortunately, no details about temperature over surfactant Tm.
cross section geometry were given for these channels. Some other au- In general terms, smaller particles were produced as the FRR in-
thors [34] have reported differences between CAD and printed designs creased (Fig. 3A and B) for both concentrations (5 and 20 mM), and for
with efforts in resin formulation optimization. all the QT levels. At a concentration of 5 mM (Fig. 3A), the particle size
In our recent work [31], we found that 3D printed channels with the decreased from 278, 298 and 358 nm (when FRR = 5) to 155, 129 and
Objet350 Connex3 printer were smoother than channels printed with a 143 nm (when FRR = 50), where QT = 50, 100 and 200 μL/min, re-
conventional desk 3D printer (Ultimaker 2+). However, for the same spectively. At a concentration of 20 mM (Fig. 3B), similarly, the particle
dimensions and aspect ratio, accuracy in cross sectional shape was size reduced from 342, 361 and 386 nm (when FRR = 5) to 164, 147
lower for the HR-3D printer even at large dimensions (1 mm squared and 151 nm (when FRR = 50) at the three QT levels of 50, 100 and
channels). It suggested that further studies are needed to understand 200 μL/min, respectively. Size reduction was rapidly reached with an
this effect with the scale and for different materials in order to inform increment in FRR from 5 to 15, and this reduction became less pro-
printing parameters optimization in terms of element dimensions, nounced from FRR 15 to 50. It is important to take into account that
geometry, and printing materials. Apart from the difference between when FRR increased the total amount of bilayer components decreased,
CAD and 3D-PMs, the cross sectional area of Mould 3 was similar to that not only producing vesicle with smaller size, since particle concentra-
previously used by Lo et al. [22], on which the selected operational tion was also reduced.
parameters of the present work were based. No significant effect of different QT was observed, while only some
differences were noticed in some particular combinations of parameters
at low surfactant concentration (5 mM), as seen in Fig. 3A. These ob-
3.2. Production of nanoparticles with temperature control for formulations servations were in accordance with previous studies [22] carried out
with high Tm non-ionic surfactants with identical chip configuration for the production of niosomes for-
mulated with other sorbitan esters (Span® 20 and 80), and also for the
The use of non-ionic surfactants for the formulation of organic production of liposomes [19,20,36,37]. At lower QT, also the linear
colloids, especially for NVs preparation, exhibits numerous advantages velocity was lower (hence larger residence time) what can counteract
[4,6]. However, a strict control of the temperature is necessary if Span® the effect of the bilayer components concentration.
60 (Tm = 45 °C), one of the most commonly used surfactant in niosome The increase in FRR, and the subsequent decrease in initial focused
formulation) is involved. Fig. 2 shows its precipitation at RT in mi- width (Wf), reduced the time needed for a complete mixing between
crochannels once reaching the focusing region, highlighting the sig- solvent and no-solvent (tmix), thus the critical concentration to induce
nificance of temperature effect. molecules self-assembly was reached faster. This led to smaller vesicles
In Fig. 2 surfactant precipitation was observed at the focussing re- since the total amount of bilayer components was reduced [38]. On the
gion and persists along the channel length when Span® 60 is used at other hand, the reduction of solvent introduced in the mixing channel
25 °C. However, at 50 °C a complete mix of both phases were produced also decreased the possibility of particle fusion into bigger unities by
without the presence of any surfactant precipitation. Moreover, the Ostwald-ripening phenomena [20,39]. A reduced tmix also led to com-
production of niosomes at this temperature conditions were observed plete mixing in limited length channels. In other cases, no diffused
using Transmission Electron Microspcopy (TEM) and negative staining solvent containing amphiphilic molecules self-assembled out of the
protocol. channel under entirely different conditions (outlet pipes, with no
This technique has been less explored than traditional bulk
Fig. 2. Precipitation of Span® 60 (Tm = 45 °C) at room temperature (upper arrow) at the focusing region (left), 0.5 and 1.0 cm downstream (centre and right). At a
temperature above surfactant Tm, focusing is complete and vesicles formation could be checked by negative staining (Phosphotungstic acid 2%) and Transmission
Electron Microscopy (TEM).
4
P. García-Manrique, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110378
Fig. 3. Size (nm) and size distribution (PDI, a.u.) measured by DLS in undiluted samples from niosomes formulated with Span® 60:Cholesterol (1:0.5 molar ratio) at
5 mM (A,C) and 20 mM (B,D) in a continuous flow microreactor based on hydrodynamic flow focusing at controlled temperature (50 °C). Each condition was tested
twice, and each batch was measured by triplicate.
laminar flow characteristics). stream exhibited a “beating pulse” like effect that was likely produced
Regarding size distribution of particles (Fig. 3C and D), PDI value by the syringe pump due to its own pumping mechanism. These pulses
reduced as FRR increased from 5 to 15, (for 5 mM: from 0.119, 0.141 created really short increments in the width of the focused fluid that
and 0.163 at FRR = 5 to 0.092, 0.071 and 0.093 at FRR = 15; for introduced alteration in solvent exchange kinetics and the subsequent
20 mM: from 0.080, 0.131 and 0.118 at FRR = 5 to 0.061, 0.057 and changes in the local concentration of bilayer precursors and solvent
0.071 at FRR = 15; for both concentration values are indicated for concentration.
QT = 50, 100 and 200 μL/min respectively). and remained without Surprisingly, lower PDI values were obtained at 20 mM for all FRRs
significant changes at higher FRR for both concentrations. Some au- at the three different QT. Indeed, these differences were higher at
thors [19,20,32] reported a significantly increase in PDI with the in- 50 μL/min. At low concentration, those mentioned instabilities can in-
crement of FRR for an identical chip configuration, but for liposomes duce more pronounced local changes in bilayer precursor’s abundances,
production instead. However, our observation was in line with that of with the corresponding effect in particle monodispersity. To gain in-
Bottaro et al. [32] in a “Y”-shaped device, while Joshi et al. [21] de- sights into these observations further studies are needed.
scribed also a reduction in PDI as FRR increase during liposome for- On the other hand, larger particles were obtained when a higher
mation. No significant differences on PDI were observed for all QT le- concentrated ethanolic solution of bilayer components was used
vels applied. (20 mM vs. 5 mM). This was observed at all QT and FRR levels (see
The use of microreactors with different channel configurations, and Supplementary material, Fig. S4). The same observation was also re-
the use of static mixing enhancers [40], could be the reason of different ported by other authors when producing liposome using microchannels
results among published works. Some of them have highlighted the [38], and in agreement with the mechanism of vesicle formation under
influence of channel dimensions and configurations over mixing effi- microfluidic flow dynamic mixing.
ciency and particle properties [19,22,38]. The preparation of solvent The efficiency of mixing under the assayed working conditions was
mixture containing bilayer precursors can also influence the extension studied following a published methodology [32]. With this method,
and homogeneity of solubilisation, and in consequence, nanoprecipi- mixing efficiency was measured through the change in colour of a pH
tation process. In the present work, ethanol was used as solvent for indicator dye (bromoxylenol blue), that changed from yellow (acidic
microfluidic-based preparation of niosomes for the first time, and this ethanolic solution containing bilayer precursors) to blue (basic aqueous
limited the possibility for comparison with other studies. phase, PBS pH = 10). A shift in focused fluid colour from yellow to blue
We have noticed that at high FRRs some transitory perturbations of indicated that molar fraction of water into the stream was close to 1 and
the focused fluid were recorded, especially at 50 μL/min. The focused the subsequent molar fraction of EtOH became close to 0, evidencing a
5
P. García-Manrique, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110378
of Wf with QT was observed at lower FRR values (Fig. 5A). It was also
observed that a lower QT generated wider focused streams probably due
to the lower pressure exercised by the lateral aqueous flows to the
middle solvent flow, but these differences became less pronounced at
higher FRRs. A similar trend was observed by Bottaro et al. [32] in an
identic channels configuration, but contrary to Jahn et al. [19] who
reported a non-variation in Wf with modifications in QT.
Moreover, an intense inverse correlation (potential) between FRR
and Wf was observed at all the QT levels (Table S2). However, a strong
negative correlation (linear) between particle size and Wf was observed
at the two different concentrations studied. These correlations reflect
that particle size is governed by focusing parameters. It is clear that
niosomes size can be tuned with the selection of the appropriate FRR
and QT values, which are key parameters for Wf and residence time.
6
P. García-Manrique, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110378
Fig. 5. Values of ethanol focused stream (Wf, μm) as flow rate ratio (FRR) increased for different values of volumetric rates (QT) at constant temperature (50 °C), and
different temperatures at constant QT (100 μL/min). Values represent the average of two independent measurements, taken at approx. at 100 μm from the end of
focussed region.
Fig. 6. Size (nm) (left) and size distribution (PDI, a.u.) (right) measured by DLS in undiluted samples from niosomes formulated with Span® 20:Cholesterol (1:0.5
molar ratio) at 5 mM in a continuous flow microreactor based on hydrodynamic flow focusing at different controlled temperatures (30, 40, 50, and 60 °C). Each
condition was tested twice, and each batch was measured by triplicate.
7
P. García-Manrique, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110378
Fig. 7. Influence of acyl chain length (C12 and C18 for Span® 20 and Span® 60 respectively) of two different sorbitan sters used in niosomes formulation (surfac-
tant:cholesterol 1:0.5 molar ratio, 5 mM), and produced under different conditions by hydrodynamic flow focusing at controlled temperature (50 °C) and a flow rate
QT = 100 μl/min.
general terms, an increase in FRR yields a focused stream being nar- Appendix A. Supplementary data
rower, and then, smaller particles due to the reduction in residual
solvent and the introduction of less amount of bilayer components. This Supplementary material related to this article can be found, in the
reduction allows complete mixing, even at high total flow rate, re- online version, at doi:https://doi.org/10.1016/j.colsurfb.2019.110378.
sulting in the size distribution of generated particles being more
homogeneous. The counterpart is that production yield is reduced, References
since particles are generated in a less concentrated suspension. Another
variable found to be relevant is the component concentration in ethanol [1] A. Albanese, P.S. Tang, W.C. Chan, The effect of nanoparticle size, shape, and
feeding solution, with a direct effect on particle size and mono- surface chemistry on biological systems, Annu. Rev. Biomed. Eng. 14 (2012) 1–16.
[2] K.L. Kelly, E. Coronado, L.L. Zhao, G. Schatz, The optical properties of metal na-
dispersity. A more concentrated solution induces an increment in par- noparticles: the influence of size, shape, and dielectric environment, J. Phys. Chem.
ticle size at any total flow rate, but surprisingly, better size distribution. B 107 (3) (2003) 668–677.
Complementary, we have checked the influence of acyl chain length [3] G. Tarun, K.G. Amit, Liposomes: targeted and controlled delivery system, Durg
Deliv. Lett. 4 (1) (2014) 62–71.
over particles morphology, and the versatility that introduces this [4] H. Abdelkader, A.W.G. Alani, R.G. Alany, Recent advances in non-ionic surfactant
parameter into the properties and functionalities of this type of bio- vesicles (niosomes): self-assembly, fabrication, characterization, drug delivery ap-
material. plications and limitations, Drug Deliv. 21 (2) (2013) 87–100.
[5] J.S. Lee, J. Feijen, Polymersomes for drug delivery: design, formation and char-
The effect of ethanol stratification due to differences in density was
acterization, J. Control. Release 161 (2) (2012) 473–483.
not taken into account, which need further investigation in for future [6] C. Marianecci, L. Di Marzio, F. Rinaldi, C. Celia, D. Paolino, F. Alhaique, S. Esposito,
work, in particular in its relationship with focusing temperature. M. Carafa, Niosomes from 80 s to present: the state of the art, Adv. Colloid Interface
Sci. 205 (2014) 187–206.
The findings in this works provide valuable information about mi-
[7] S. De, M.R. Prasad, Self-assembled cell-mimicking vesicles composed of amphiphilic
crofluidics-based production of niosomes at different operational con- molecules: structure and applications, first edition, in: Hiroyuki Ohshima (Ed.),
ditions, and are expected to support the expansion of this technique for Encyclopedia of Biocolloids and Biointerface Science, vol. 1, John Wiley & Sons,
the preparation of a wider range of organic colloids with important 2016.
[8] R. Handjani-Vila, A. Ribier, B.A. Rondot, G. Vanlerberghie, Dispersions of lamellar
characteristics for related industries with growing interest in different phases of non-ionic lipids in cosmetic products, Int. J. Cosmet. Sci. 1 (5) (1979)
application fields. 303–314.
[9] G. Gutiérrez, M. Matos, P. Barrero, D. Pando, O. Iglesias, C. Pazos, Iron-entrapped
niosomes and their potential application for yogurt fortification, Food Sci. Technol.
74 (2016) 550–556.
Declaration of Competing Interest [10] B. Demir, B.F. Baris, G.Z. Pinar, P. Unak, S. Timur, Theranostic niosomes as a
promising tool for combined therapy and diagnosis: “all-in-one” approach, Appl.
Nano Mater. 1 (6) (2018) 2827–2835.
The authors declare no conflicts of interest in this work. [11] P. García-Manrique, E. Lozano-Andrés, O.R. Estupiñán-Sánchez, G. Gutiérrez,
M. Matos, C. Pazos, M. Yañez-Mo, C. Blanco-López, Biomimetic small extracelular
vesicles, 3rd GEIVEX Simposium, San Sebastian, Spain, 29-30 September, (2016)
Poster communication.
Acknowledgements [12] S. De, R. Kundu, A. Biswas, Synthesis of gold nanoparticles in niosomes, J. Colloid
Interface Sci. 386 (2012) 9–15.
This work was supported by the Ministerio de Economía y [13] R. Bartelds, N.M. Hadi, T. Pols, M.C.A. Stuart, A. Pardakhty, G. Asadikaram,
B. Poolman, Niosomes, an alternative for liposomal delivery, PLoS One 13 (4)
Competitividad (MINECO, Spain), under the Grant CTQ2013-47396-R
(2018) e0194179.
and MAT2017-84959-C2-1-R. This study was also financed by the [14] O.R. Justo, A..M. Moraes, Analysis of process parameters on the characteristics of
̂
Consejería de Economía y Empleo del Principado de Asturias (Plan de liposomes prepared by ethanol injection with a view to process scale-up: effect of
Ciencia, Tecnología e Innovación 2013-2017), under the Grant temperature and batch volume, Chem. Eng. Res. Des. 89 (2011) 785–792.
[15] M. Danaei, M. Dehghankhold, S. Ataei, F. Davarani Hasanzadeh, R. Javanmard,
GRUPIN14-022 and IDI/2018/000185. Support from the European A. Dokhani, S. Khorasani, M.R. Mozafari, Impact of particle size and polydispersity
Regional Development Fund (ERDF) is gratefully acknowledged. Pablo index on the clinical applications of lipidic nanocarrier systems, Pharmaceutics 57
García-Manrique is especially grateful to Campus de Excelencia de la (10) (2018) 1–17.
[16] P. García-Manrique, M. Matos, G. Gutierrez, O.R. Estupiñán, M.C. Blanco-López,
Universidad de Oviedo and Banco Santander for his mobility fellowship C. Pazos, Using factorial experimental design to prepare size-tuned nanovesicles,
for the stay at University of Southampton. Ind. Eng. Chem. Res. 55 (34) (2016) 9164–9175.
[17] D. van Swaay, A. deMello, Microfluidic methods for forming liposomes, Lab Chip 13
(2013) 752–767.
8
P. García-Manrique, et al. Colloids and Surfaces B: Biointerfaces 182 (2019) 110378
[18] L. Capretto, D. Carugo, S. Mazzitelli, C. Nastruzzi, X. Zhang, Microfluidic and lab- for their application for nanomaterials synthesis, N. Biotechnol. 47 (2018) 1–7.
on-a-chip preparation routes for organic nanoparticles and vesicular systems for [32] E. Bottaro, A. Mosayyebi, D. Carugo, C. Nastruzzi, Analysis of the diffusion process
nanomedicine applications, Adv. Drug Deliv. Rev. 65 (2013) 1496–1532. by pH indicator in microfluidic chips for liposomes production, Micromachines 8
[19] A. Jahn, W.N. Vreeland, D.L. DeVoe, L.E. Locascio, G. Michael, Microfluidic di- (2017) 209.
rected formation of liposomes of controlled size, Langmuir 23 (11) (2007) [33] Chee Kai Chua, Kah Fai Leong (Eds.), 3D Printing and Additive Manufacturing-Fifth
6289–6293. Edition, Principles and Applications, World Scientific Publishing Co. Pte. Ltd.,
[20] E. Kastner, V. Verma, D. Lowry, Y. Perrie, Microfluidic-controlled manufacture of Singapore, 2017.
liposomes for the solubilisation of a poorly water soluble drug, Int. J. Pharm. 485 [34] G. Gaal, M. Mendesa, T.P. de Almeideb, M.H.O. Piazzettad, Â.L. Gobbi, A. Riul Jr,
(2015) 122–130. V. Rodrigues, Simplified fabrication of integrated microfluidic devices using fused
[21] S. Joshi, M.T. Hussain, B.R. Carla, G. Anderluzzi, E. Kastner, S. Salmaso, D.J. Kirby, deposition modeling 3D printing, Sens. Actuators B Chem. 242 (2017) 35–40.
Y. Perrie, Microfluidics based manufacture of liposomes simultaneously entrapping [35] P.N. Ezhilarasi, P. Karthik, N. Chhanwal, C. Anandharamakrishnan,
hydrophilic and lipophilic drugs, Int. J. Pharm. 514 (2016) 160–168. Nanoencapsulation techniques for food bioactive components: a review, Food
[22] C.T. Lo, A. Jahn, L.E. Locascio, W.N. Vreeland, Controlled self-assembly of mono- Bioprocess Technol. 6 (2013) 628–647.
disperse niosomes by microfluidic hydrodynamic focusing, Langmuir 26 (11) [36] D. Carugo, E. Bottaro, J. Owen, E. Stride, C. Nastruzzi, Liposome production by
(2010) 8559–8566. microfluidics: potential and limiting factors, Sci. Rep. 19 (6) (2016) 25876.
[23] M.A. Obeid, I. Khadra, A.B. Mullen, R.J. Tate, V.A. Ferro, The effects of hydration [37] R.R. Hood, D.L. DeVoe, High-throughput continuous flow production of nanoscale
media on the characteristic of non-ionic surfactant vesicles (NISV) prepared by liposomes by microfluidic vertical flow focusing, Small 11 (2015) 5790–5799.
microfluidics, Int. J. Pharm. 56 (2017) 52–60. [38] M. Antonietti, S. Förster, Vesicles and liposomes: a self-assembly principle beyond
[24] S. García-Salinas, E. Himawan, M. Gracia, M. Arruebo, V. Sebastian, Rapid on-Chip lipids, Adv. Mater. 15 (2003) 1323–1333.
assembly of niosomes: batch versus continuous flow reactors, Appl. Mater. [39] I.V. Zhigaltsev, N. Belliveau, I. Hafez, A.K.K. Leung, J. Huft, C. Hansen, P.R. Cullis,
Interfaces 10 (2018) 19197–19207. Bottom-up design and synthesis of limit size lipid nanoparticle systems with aqu-
[25] B.C. Gross, J.L. Erkal, S.Y. Lockwood, C. Chen, D.M. Spence, Evaluation of 3D eous and triglyceride cores using millisecond microfluidic mixing, Langmuir 28
printing and its potential impact on biotechnology and the chemical sciences, Anal. (2012) 3633–3640.
Chem. 86 (2014) 3240–3253. [40] E. Kastner, R. Kaur, D. Lowry, B. Moghaddam, A. Wilkinson, Y. Perrie, High-
[26] J. Dong, J. Liu, G. Kang, J. Xie, Y. Wang, Pushing the resolution of photolithography throughput manufacturing of size-tuned liposomes by a new microfluidics method
down to 15 nm by surface plasmon interference, Sci. Rep. 4 (2014) 5618. using enhanced statistical tools for characterization, Int. J. Pharm. 477 (2015)
[27] D. Carugo, J.Y. Lee, A. Pora, R.J. Browning, Capretto Lorenzo, C. Nastruzzi, 361–368.
E. Stride, Facile and cost effective production of microscale PDMS architecture [41] R. Karnik, F. Gu, P. Basto, C. Cannizzaro, L. Dean, Kyei-Manu, R. Langer,
using a combined micromilling-replica moulding (μMI-REM) technique, Biomed. O.C. Farokhzad, Microfluidic platform for controlled synthesis of polymeric nano-
Microdevices 18 (2016) 1–10. particles, Nano Lett. 8 (9) (2018) 2906–2912.
[28] A.K. Au, W. Huynh, L.F. Horowitz, A. Folch, 3D-prinded microfluidics, Angew. [42] A. Manosroi, P. Wongtrakul, J. Mnosroim, H. Sakai, F. Sugawara, M. Yuasa, M. Abe,
Chem. Int. Ed. 55 (2016) 3862–3881. Characterization of vesicles prepared with various non-ionic surfactants mixed with
[29] G. Comina, A. Suska, D. Filippini, PDMS lab-on-a-chip fabrication using 3D printed cholesterol, Colloids Surf. B Biointerfaces 30 (2003) 129–138.
templates, Lab Chip 14 (2014) 424–430. [43] L. Peltonen, J. Hirvonen, J. Yliruusi, The effect of temperature on sorbitan surfac-
[30] Y. Hwang, O.H. Paydar, R.N. Candler, 3D Printed molds for non-planar PDMS mi- tant monolayers, J. Colloid Interface Sci. 239 (2001) 134–138.
crofluidic channels, Sens. Actuators A 226 (2015) 137–142. [44] G. Gutiérrez, J.M. Benito, C. Pazos, J. Coca, Evaporation of aqueous dispersed
[31] D.A. Cristaldi, F. Yanar, A. Mosayyebi, P. García-Manrique, E. Stulz, D. Carugo, systems and concentrated emulsions formulated with non-ionic surfactants, Int. J.
X. Zhang, Easy-to-perform and cost-effective fabrication of continuous-flow reactors Head Mass Transf. 69 (2014) 117–128.
9
Supplementary Material-
Pablo García-Manrique1,2, Gemma Gutiérrez2, María Matos2, Andrea Cristaldi3, Ali Mosayyebi3,
Dario Carugo4, Xunli Zhang3*, Mª Carmen Blanco1*
1 Department of Physical and Analytical Chemistry, University of Oviedo; Spain
2 Department of Chemical Engineering and Environmental Technology, University of Oviedo; Spain
3Bioengineering Sciences group, Faculty of Engineering and the Environment, Institute for Life Sciences (IfLS),
1
Table S1. Morphological characteristics of mixing channel for original Solidworks® CAD 2016
design and 3D-Printed positive moulds (3D-PM) onto VeroClearTM resin with the 3D printer
Objet350 ConnexTM (Stratsys). Average and standard deviation values are given for the
parameters.
2
Figure S1. Calibration plot (A) and temperature stability (B) of the in-house designed
thermostatic chamber for microfluidics chips, fabricated by 3D-printing technology with PLA
filaments. Values represented are the average of three independents measurements.
3
Figure S2. Pictures composition showing the whole setup (central) and detailed components
(sides) used in this work for niosomes production by Hydrodynamic Flow Focusing with controlled
temperature.
4
Table S2. Correlation factor between flow focusing parameters (FRR and Wf) and particle size
at different concentration of bilayer components and variable QT (A), and at fixed
concentration and QT for different working temperatures (B).
(A)
(*)
FRR vs Wf 0.99 0.97 0.92
(**)
Wf vs Particle size 0.86 (5 mM) 0.98 (5 mM) 0.97 (5 mM)
linear correlation
(B)
(*)
FRR vs Wf 1.00 0.99 0.97 0.98
(**)
FRR vs Particle size 0.24 0.56 0.91 0.65
FRR, Flow Rate Ratio; QT, Total volumetric rate; Wf, initial width of focused stream; (*) potential correlation; (**)
linear correlation
5
Figure S3. Effect of bilayer components concentration for niosomes formulated with Span®
60:Cholesterol (1:0.5 molar ratio) and produced under the same flow conditions (QT and FRR)
for size (upper row) and size distribution (lower row).
6
ANEXO II. Publicaciones relacionadas con la
Tesis Doctoral
New BIOTECHNOLOGY 47 (2018) 1–7
New BIOTECHNOLOGY
journal homepage: www.elsevier.com/locate/nbt
A R T I C L E I N F O A B S T R A C T
Keywords: The translation of continuous-flow microreactor technology to the industrial environment has been limited by
3D printing cost and complexity of the fabrication procedures and the requirement for specialised infrastructure. In the
Lab-on-a-chip present study, we have developed a significantly more cost-effective and easy-to-perform fabrication method for
Soft lithography the generation of optically transparent, continuous-flow reactors. The method combines 3D printing of master
Nanoparticles
moulds with sealing of the PDMS channels’ replica using a pressure-sensitive adhesive tape. Morphological
Liposomes
Continuous-flow reactors.
characterisation of the 3D printed moulds was performed and reactors were fabricated with an approximately
square-shaped cross-section of 1 mm2. Notably, they were tested for operation over a wide range of volumetric
flow rates, up to 20 ml/min. Moreover, the fabrication time (i.e., from design to the finished product) was <
1 day, at an average material cost of ∼£5. The flow reactors have been applied to the production of both
inorganic nanoparticles (silver nanospheres) and organic vesicular systems (liposomes), and their performance
compared with reactors produced using more laborious fabrication methods. Numerical simulations were per-
formed to characterise the transport of fluids and chemical species within the devices. The developed fabrication
method is suitable for scaled-up fabrication of continuous-flow reactors, with potential for application in bio-
technology and nanomedicine.
Introduction polymethyl methacrylate, PMMA). This procedure does not require the
fabrication of photomasks via photolithography, which is typically
Photolithography has been widely used to manufacture continuous- performed in a cleanroom environment. However, the method requires
flow reactors at high spatial resolution, in terms of both size and shape the use of micromilling machines, as well as oxygen plasma bonding for
of the channels’ architecture [1]. However, this process involves nu- the sealing of the polydimethylsiloxane (PDMS) channels onto a glass
merous steps and generally requires specialised cleanroom facilities, layer. Alternatively, positive moulds can be fabricated in a single step
and expensive materials or instrumentation. In addition, the whole using high-resolution 3D printing (with channel width down to 50 μm),
process (i.e., from the design of the device architecture to the end as demonstrated by Comina et al. [4]. Similarly, three-dimensional (3D)
product) is highly time-consuming. The combination of these factors PDMS microfluidic reactors can be fabricated via UV-activated 3D
has hindered the widespread adoption of this technology by industries printing, as described by Chan et al. [5]. Although 3D printing of resin
and researchers, particularly in the non-specialised or less resourced moulds may be less complex to perform compared to μMi-REM, it re-
laboratories. quires specific treatments of the moulds prior to PDMS casting, due to
In the last decade, efforts have been made to develop more cost- the inhibition effect of the resin on the PDMS cross-linker [6,4]. In
effective and user friendly manufacturing approaches [2]. For instance, addition, both methods rely on the use of oxygen plasma during the
the micromilling-replica moulding (μMi-REM) technique recently de- bonding process. On the other hand, microfluidic devices could be
veloped by Carugo et al. [3] involves the fabrication of positive epoxy entirely 3D printed, as demonstrated by Kitson et al. [7]. This method is
masters obtained from negative micromilled moulds (made of cost-effective and easy-to-perform; however, devices are not optically
⁎
Corresponding author.
⁎⁎
Co-corresponding authors.
E-mail addresses: [email protected] (E. Stulz), [email protected] (D. Carugo), [email protected] (X. Zhang).
https://doi.org/10.1016/j.nbt.2018.02.002
Received 25 July 2017; Received in revised form 2 February 2018; Accepted 4 February 2018
Available online 06 February 2018
1871-6784/ © 2018 Elsevier B.V. All rights reserved.
D.A. Cristaldi et al. New BIOTECHNOLOGY 47 (2018) 1–7
transparent, thus limiting the ability to optically monitor flow and moulds. The following printing settings were adopted: bottom/top
mixing processes. The optical transparency of 3D printed channels was thickness = 0.5 mm, fill density = 100%, print speed = 50 mm/s, and
recently improved by Gaal et al. [8], using a custom built 3D printer to nozzle size = 0.4 mm. PDMS replicas were prepared by pouring a 10.2/
create polylactic acid/polydimethylsiloxane (PLA/PDMS) architectures. 0.8 (w/w) ratio of degassed PDMS precursor and curing agent mixture
However, careful adjustments of the 3D printer settings were needed (Sylgard® 184, Dow Corning Corporation, Michigan, USA) over the
during the fabrication process, and the cross-section of the produced mould (Scheme 1, Step 2). Degassing was carried out by prior cen-
channels differed significantly from the original computer-aided design trifugation at 3000 rpm for 15 min, using the Eppendorf Centrifuge
(CAD). 5804 and Corning Centristar™ tubes (50 ml). After pouring, the liquid
Developments have also been made in terms of the microchannel PDMS was left at room temperature for 4 h and any formed gas bubble
dimensions obtainable using 3D printing, with recent studies re- removed using a sharp tool (every 1 h). This step could be significantly
searching novel resin formulations for stereolithgraphy (SL) and 3D accelerated by placing the mould under vacuum. PDMS curing was
printing with Digital Light Processing (DLP) [9]. With this technique, performed in the oven at 40 °C overnight. The cured PDMS replica
Gong et al. demonstrated the generation of remarkably small micro- channel was peeled off from the master mould, and a 1.5 mm diameter
channels (i.e., with cross section as small as 18 × 20 μm) [10]. Sig- biopsy punch with plunger (Miltex®, Fischer Scientific, UK) was used for
nificant efforts have also been devoted to development of simple creating inlets/outlets. Sealing of the PDMS layer was performed on
bonding techniques for microfluidic devices. For example, Serra et al. ThermalSeal RT™ tape purchased from Excel Scientific (USA) (Scheme
[11] recently demonstrated the use of a commercially available sealing 1, Step 3). To characterise the morphology of the 3D printed moulds, a
tape (Thermalseal RTS™) for bonding of various substrates. The PDMS non-contact Alicona Infinite Focus 3D optical profilometer was em-
channel architectures in this study were fabricated from micromilled ployed (5 × magnification lens, vertical resolution: 410 nm, lateral re-
brass masters. solution: 6.59 μm). In order to evaluate the method’s repeatability,
In the present study, we developed a fast, cost-effective and facile three identical devices were printed and two-dimensional (2D) images
manufacturing process to fabricate optically transparent flow reactors, (at 2.5 × magnification) were acquired to measure (i) the width of the
with milli- or sub-millimetre scale flow channels (i.e., with 0.5–1.0 mm mixing channel at five equidistant locations along the channel (se-
channel width). A combination of techniques was employed including paration distance between measurements = 2 mm) starting from the
3D printing of positive moulds followed by PDMS casting (3D printed junction, and (ii) the radius of curvature of both inlet channels. Mea-
mould casting, 3DPM-C), and direct sealing of the PDMS layer using a surements were performed using Image-J software (NIH, USA).
pressure-sensitive adhesive tape. The 3D printed moulds were char- The manufacture of μMi-REM devices was performed following a
acterized in terms of surface roughness and cross-sectional shape, and protocol previously reported [3]. The PVA-TePla 300 plasma cleaner
reactors were tested over a wide range of operational conditions. To was employed to assist the bonding of the PDMS layer with a
demonstrate the usability of the developed fabrication technique, re- 50 × 70 mm glass sheet (Corning® microscope slides, Sigma Aldrich,
actors were applied to the production of silver nanospheres (SNSs) and Gillingham, UK).
liposomes, as examples of inorganic and organic synthesis, respectively. The two fabrication methods employed in this study are herein
Silver nanospheres (SNSs) have been employed in many research defined as 3DPM-C/Tape and μMi-REM*Glass, where ‘/’ and ‘*’ indicate
fields ranging from photocatalysis [12] to optoelectronics [13], as well adhesive tape and plasma bonding procedures, respectively.
as for biological applications due to their antibacterial properties [14].
SNSs synthesis typically needs a carefully balanced stoichiometry, in Synthesis of inorganic and organic nanoparticles
order to obtain the desired particle size and/or shape [15]. Recently,
Barber et al. demonstrated a coaxial glass reactor for continuous-flow The same experimental set-up was used for the production of both
production of SNSs [16], with superior control over the fluidic en- silver nanospheres and liposomes. Syringe pumps (AL-1010) were
vironment and properties of the produced SNSs, when compared to purchased from World Precision Instruments (UK). Luer Lock syringes
bulk methods. Liposomes, spherical vesicles comprising an aqueous (20 ml) (BD Plastipak) were purchased from BD (Becton, Dickinson and
core surrounded by a lipid bilayer, are employed as vehicles for Company, UK). Polytetrafluoroethylene (PTFE) tubing and connectors
transporting and administering pharmaceutical actives [17]. Produc- (Cole-Palmer, UK) were employed for interfacing the device inlets and
tion of liposomes via solvent exchange mechanism in continuous-flow outlet with syringes and collection vials, respectively. The length of the
reactors has emerged as a promising technique, offering a higher degree tube from the outlet to the collection vial was 26.70 cm.
of control over the physical and dimensional properties of the end Silver nanoparticles were synthesized using silver nitrate 99.9999%
product, compared to batch methods. (AgNO3), tri sodium citrate dihydrate ≥ 99.0% (TSCD), poly-
In this study, we demonstrate continuous-flow production of both vinylpyrrolidone (PVP), and sodium borohydride 99% (NaBH4), which
types of nanoscale particles using cost-effective and easy-to-operate were purchased from Sigma Aldrich UK (Gillingham, UK). Propan-2-ol
reactors. (or isopropyl alcohol, IPA) laboratory reagent grade was purchased
from Fisher Chemical (UK). Milli-Q water was collected using the Q-
Materials and methods Gard purification filter, connected to the Milli-Q Gradient A10 system
(Merck Millipore, USA).
Design, fabrication and morphological characterization of flow reactors Scheme 2 shows the experimental set-up employed for the synthesis
of both silver nanoparticles and liposomes. Syringes were spatially ar-
Fabrication of the 3D printed mould casting (3DPM-C) channels is ranged in a way that allowed performing the same experiment for both
illustrated in Scheme 1. Solidworks® CAD 2016 software was used for 3DPM-C/Tape and μMi-REM*Glass devices, by simply changing the
designing the master mould. The flow reactor architecture has two reactor.
semi-circular inlet channels of 0.50 mm × 1.00 mm (width × height) For SNSs production, a 20 ml syringe was primed with a Milli-Q
and 1.50 mm radius. Inlets converge in to a straight channel of water solution containing AgNO3 (1.02 mM), TSCD (15.02 mM), and
1.00 mm × 1.00 mm × 60.00 mm (width × height × length). The PVP (0.45 M). The second syringe was primed with 15 ml of an IPA/
channel architecture was positioned at the bottom surface of a box Milli-Q solution (9:1 v/v) of NaBH4 (5.28 mM). The total flow rate
structure having a 7.00 mm high edge, which acted as a container for (TFR = FRA + FRB) was kept at the constant value of 1 ml/min,
uncured PDMS (see Step 1 in Scheme 1). The Ultimaker 2+ 3D printer, whereas the flow rate ratio (FRR = FRA/FRB) was varied (5, 7, 9 and
loaded with PLA filaments, was employed as a representative fused 11). Each sample was separately collected in a 1.5 ml Eppendorf tube,
deposition modelling (FDM) tool for the production of the master and 1.5 ml were collected in a waste vial in between each experimental
2
D.A. Cristaldi et al. New BIOTECHNOLOGY 47 (2018) 1–7
Scheme 1. Graphical representation of the manufacturing steps for the 3DPM-C/Tape reactor: 1) Low-cost 3D printing of the positive mould; 2) PDMS casting; and 3) sealing of the cured
PDMS layer onto adhesive tape.
run. In order to evaluate the robustness of the reaction, experiments measure the mean diameter (z-average), the polydispersity index (PDI),
were repeated in triplicate at selected TFR and FRR values. and zeta potential of all liposomal formulations, produced with both
For the synthesis of liposomes, Phospholipon® 90G (lipids) and 3DPM-C/Tape and μMi-REM*Glass reactors. Liposome dimensional
purified phosphatidylcholine from soybean lecithin, were kindly pro- stability was assessed by measuring the mean diameter of samples
vided by Lipoid GmbH (Germany). Pure ethanol (99.9%) was purchased stored at both 4 °C and 25 °C, every 5 days for a total 30 days. All
from Sigma-Aldrich Company Ltd. (UK). A 100 mM lipid solution in measurements were performed using the Zetasizer Nano ZS Malvern,
ethanol was prepared, and the lipid concentration selected to produce UK.
liposomes of a clinically relevant size and mass [18–20]. Milli-Q water
was injected into one inlet (Syringe A) and ethanol-containing lipids
was injected into a second inlet (Syringe B), for each reactor (Scheme Results and discussion
2).
Experiments were carried out maintaining a constant FRR of 25, at Characterisation of the 3D printed moulds and PDMS channels
varying TFRs of 1, 3 and 6 ml/min to demonstrate devices’ usability for
producing liposomes at high-throughput. Three samples were collected, The morphology and roughness of the flat base of the 3D printed
at each TFR and for each device tested. master are important characteristics affecting spatial uniformity and
durability of the sealing. From the morphological examination, diag-
onal grooves can be observed due to the oblique motion of the nozzle in
Characterisation of nanoparticles the x-y plane during printing (see Fig. 1a). The average roughness (Ra)
value of the master mould is 6.56 μm (Fig. 1a), and a maximum peak-to-
The UV–vis characterization of silver nanospheres (SNSs) was car- valley (Rz) value of 85.13 μm is detected in proximity to the diagonal
ried out using a Varian Cary300Bio UV–vis Spectrophotometer. All features over the base surface (representative cross sectional profiles
measurements were performed in the 200–800 nm range, with an in- are shown in Figure SI-1a). Notably, we observed that grooves could
crement step of 0.5 nm. SNS analytical samples (1 ml) were collected promote fluid leakage at flow rates greater than 5 ml/min.
from the flow reactors, and diluted to 3 ml with Milli-Q water into a In order to overcome this limitation, the PDMS mixture (monomer/
quartz cuvette for the spectrophotometric characterization. The base- curing agent) was prepared using 8% curing agent (instead of the
line was subtracted from each experimental condition (i.e., considering commonly used 10% by mass), to reduce PDMS stiffness. Notably, a
the specific Milli-Q/IPA volume ratio). softer PDMS could be deformed more easily by applying external
Transmission electron microscopy (TEM) characterization of SNSs pressure during sealing. Manual compression using a plastic spatula
was also performed. Images were acquired using the TEM Hitachi was initially performed (at an estimated pressure of 1 bar), followed by
HT7700. Silver nanoparticles were prepared by drop-casting of the compression at ∼0.5 bar for 1 h. In this way, no leakages were observed
colloidal synthesis solution (5 μl), on carbon and Formvar coated Cu/Pd even at a total flow rate of 20 ml/min (see Supporting information
200 mesh grids, and left to dry under atmospheric conditions at room video, at https://youtu.be/EpmnLZDXtBo), confirming the effective-
temperature. ness of the sealing procedure for continuous-flow synthesis at high-
A dynamic light scattering (DLS) technique was used instead to throughput.
Scheme 2. Schematic of the experimental set-up and list of chemicals injected through syringes A and B for the production of SNSs and liposomes, respectively.
3
D.A. Cristaldi et al. New BIOTECHNOLOGY 47 (2018) 1–7
Fig. 1. Morphology and cross-sectional shape of the 3D printed moulds. a) Morphological characterisation of the 3D printed base, including average roughness (Ra) and maximum peak-
to-valley value (Rz). b) Cross section of a representative channel design of 0.60 × 0.80 mm (width × height), obtained from optical profilometry.
Accurate reproduction of the designed (i.e., nominal) channel size Fig. 2a). The radius of curvature of the inlet channels was also very
and shape should ideally be achieved by the 3D printing process. In this comparable between different moulds (mean value = 1.770 ±
respect, the high resolution (HR) resin 3D printer (Object Connex 350) 0.012 mm), and only slightly differed from the nominal value of
was tested against the Ultimaker 2+ by printing channels of different 1.75 mm (see Fig. 2b).
cross-sectional dimension, as illustrated in Figure SI-1b. The HR 3D
printer was able to create smaller and smoother channels; however, Synthesis of silver nanospheres and liposomes via 3DPM-C/Tape & μMi-
reproduction of the cross sectional shape was less accurate, even for the REM*Glass reactors
relatively large channel dimension of 1.00 × 1.00 mm (see cross section
in Figure SI-1c obtained from profilometry). Thus, the Ultimaker 2+ Nanoparticle synthesis via 3DPM-C/Tape reactor was performed in
was selected to generate the master moulds in the remaining experi- parallel with the already established μMi-REM*Glass technique. In
ments. order to compare the performance of the two fabrication methods, the
Two additional test channels were printed having different size but physical properties of the produced nanomaterials were measured and
the same aspect ratio of 1.33, using the Ultimaker 2+. The mean ex- evaluated at varying fluid dynamic boundary conditions.
perimental values obtained with the optical profilometer (re-
presentative test Channel 1 in Fig. 1b) are compared with the nominal
Synthesis of silver nanospheres
dimensions of both channels in Table 1. The channel height is accu-
rately reproduced, whereas the width is affected by a 0.09 mm differ-
Silver nanospheres (SNSs) were synthetized using the chemical re-
ence for both test channels 1 and 2 (a 3D cross section of test Channel 2
duction method [15], which was adapted for usage in a continuous-flow
is shown in Figure SI-1c). This is due to the orientation angle (90°) of
reactor. Specifically, NaBH4 solution was prepared initially by dissol-
the side walls of the channels with respect to the light source of the
ving the solid in 1.5 ml of Milli-Q water, followed by dilution to 15 ml
profilometer. This effect is clearly visible in Fig. 1b in which the side
with IPA. The use of IPA was to minimise the generation of H2, pro-
walls of the channel are represented as the projection of the edge of the
duced from the degradation of NaBH4 in water [21]. This drastically
channel roof towards the base, causing overestimation of the measured
reduces the formation of gas bubbles within the channels, which may
width.
significantly alter the flow field or cause clogging. This effect may
Nonetheless, in order to assess repeatability of the reactor’s fabri-
however be less problematic in millimetre-scale channels. An alter-
cation method, three replicas of the 3D printed moulds were manu-
native approach may involve the generation of a strong basic condition
factured using the Ultimaker 2+ (Fig. 2). The width of the mixing
(NaOH, 14 M) in water, as described by Barber et al. [16]. However,
channel (measured at five separate and equidistant locations along the
with the protocol described in this study, we were able to produce SNSs
channel) and the radius of curvature of the inlet channels were mea-
at all FRRs investigated, obtaining the maximum absorbance (Amax) of
sured, and results are shown in Fig. 2a and b respectively. Insets il-
2.9 ± 0.2 nm after 1:3 dilution with Milli-Q water. Nevertheless, the
lustrate the positions at which measurements were taken.
stoichiometry played a critical role in SNSs synthesis, and although the
The width of the mixing channel is comparable between the three
numerical results (see Figure SI-2b) show a marginal increase in mixing
different moulds, as illustrated in Fig. 2a. The maximum difference is at
efficiency at FRR = 11, the SNSs production performance was more
6 mm from the junction, between mould n.1 and n.3, and is equal to
effective at FRR = 7.
0.0011 mm only. All moulds had a slightly larger channel width in
Fig. 3a reports the UV–vis spectra of SNSs prepared using the 3DPM-
proximity to the junction, which gradually reduced along the channel
C/Tape and the μMi-REM*Glass reactors, applying the same operational
and reached a plateau value at about 8–10 mm from the junction (see
conditions in both devices (TFR = 1 ml/min; FRR = 7). In order to
assess the reproducibility of the reaction, samples were produced in
Table 1 triplicate under the same conditions for both devices.
Size comparison between the CAD design and the 3D printed channels. The typical absorption spectra, due to SNSs surface plasmon re-
sonance (SPR) [22], are observed. Notably, the SNSs spectra obtained
Design-Morphology Width × Height (mm) Width × Height (mm)
using the two different fabrication methods are almost overlapping
Comparisons Test Channel 1 Test Channel 2
across the whole wavelength spectrum. The mean value of maximum
CAD Design 0.60 × 0.80 0.80 × 1.33 absorbance (Amax) is equal to 3.252 ± 0.050 and 3.236 ± 0.068 for
the Tape and Glass reactors, respectively (see Fig. 3a and inset). More
3D Printed mould 0.69 × 0.80 0.89 × 1.33
importantly, in both types of reactor the maximum absorption value of
profile (mean
values) 398.0 nm is obtained, further indicating a comparable performance
between them. The absorbance value is also related to the particle size
4
D.A. Cristaldi et al. New BIOTECHNOLOGY 47 (2018) 1–7
Fig. 2. a) Width of the mixing channel at 2, 4, 6, 8, and 10 mm from the junction between inlets, for three different 3D printed moulds. An image of a representative channel (at
2.5 × magnification) showing the measurement lines (red lines) is reported in the inset. b) Radius of curvature of both right (R) and left (L) inlet channels, for three different 3D printed
moulds. A graphical representation of the measurement method is reported in the inset, for the right inlet channel.
[23], which ranged from 10 to 20 nm as shown in the TEM images min and FRR = 25, have a diameter of 188.61 ± 1.62 nm and
(Fig. 3b). 191.37 ± 3.19 nm, respectively. Moreover, by increasing the TFR from
1 to 6 ml/min caused only a slight increase in liposome diameter in
Synthesis of liposomes both devices, which is consistent with the numerical simulations
showing a relatively small difference in the mixing index at the dif-
Having demonstrated that flow reactors fabricated with different ferent flow regimes investigated (see Figure SI-2b). Fig. 4b shows that
methods have comparable performance when employed to produce liposomes have a relatively small dispersity, and that PDI slightly in-
silver nanoparticles, they were also tested for continuous-flow synthesis creased with increasing the TFR from 1 to 6 ml/min.
of liposomes. Devices were operated at total flow rates which were Liposomes produced by the 3DPM-C/Tape reactor had a larger
significantly higher than those typically used in microfluidic reactors mean dispersity at the higher flow rates investigated, which could be
[20], to demonstrate their potential utility for scaled-up synthesis. potentially attributed to the surface roughness of the PDMS channels.
Fig. 4 shows the size (z-average) and polydispersity index (PDI) of Further investigations will be performed to better understand the effect
liposomes produced at varying TFR values (1, 3, 6 ml/min) and a fixed of surface properties on the transport of fluids and chemical species in
FRR of 25. A representative intensity-based liposome size distribution these devices.
(at TFR = 1 ml/min and FRR = 25) is also shown in Figure SI-3a, for In addition, the diameter of liposomes produced by 3DPM-C/Tape
the μMi-REM*Glass reactor. Notably, liposome size and dispersity are reactor (at FRR of 25 and TFR of 1 ml/min) was measured up to 30 days
not significantly different across different devices, for all the hydro- from production, at two different storage temperatures (4 °C and 25 °C),
dynamic conditions investigated. For instance, liposomes produced as shown in Fig. 4c. The size of liposomes stored at 4 °C increased over
with the 3DPM-C/Tape and μMi-REM*Glass reactors at TFR = 1 ml/ time, and reached a maximum% increase of approximately 15% after
Fig. 3. a) UV–vis characterization of SNSs prepared using the 3DPM-C/Tape (black) and the μMi-REM*Glass (red) reactors. Spectra are shown as the mean of triplicate samples prepared
using both types of reactor, at the same operating conditions (TFR = 1 ml/min; FRR = 7) (individual spectra are shown in the inset). b) Representative TEM image of the SNSs prepared
with the 3DPM-C/Tape (TFR = 1 ml/min; FRR = 7); with a magnified view shown in the inset.
5
D.A. Cristaldi et al. New BIOTECHNOLOGY 47 (2018) 1–7
Fig. 4. Comparison of liposome size and dispersity. Size (z-average) (a) and dispersity (PDI) (b) of liposomal formulations produced by μMi-REM*Glass and 3DPM-C/Tape reactors. Each
experiment was performed at TFR of 1 ml/min, 3 ml/min and 6 ml/min, at a fixed FRR of 25. Data are reported as the mean of three independent samples, with the corresponding
standard deviation. c) Size stability of liposomal formulations produced by 3DPM-C/Tape reactor, at FRR of 25 and TFR of 1 ml/min. Liposome size was measured every 5 days and up to
30 days, at storage temperatures of both 4 °C and 25 °C.
30 days. These results are comparable with the ones previously reported (liposomes). They exhibited comparable performance to reactors fab-
in the literature [24], although they refer to different Phospholi- ricated using more laborious and expensive fabrication methods. The
pon90G-based liposomal formulations. The increase in vesicle diameter manufacturing technique presented in this study is therefore potentially
could be attributed to sterical hindrance of bilayer stability [25] or suitable for scaling-up, as devices can be fabricated at low-cost, and
aggregation, which may result in liposome coalescence [26]. Con- without resorting to sophisticated instrumentation and time-consuming
versely, the size of liposomes stored at 25 °C was almost unchanged multistep procedures. Moreover, devices produced with this technique
over time. Moreover, the zeta potential of liposomes produced using the can be operated at relatively high total flow rates ( > 20 ml/min),
3DPM-C/Tape rector (at FRR of 25 and TFR of 1 ml/min) was which is a desirable characteristic for application in continuous-flow
−15.06 mV, which is coherent with the literature [27]. chemical synthesis.
Conclusions Acknowledgment
We have developed an easy-to-perform and cost-effective method Authors would like to thank the University of Southampton for
for the fabrication of PDMS based continuous-flow reactors, through 3D supporting the research through a PhD studentship.
printed mould casting (3DPM-C). In this method, the positive mould
was “printed” using a desktop 3D printer, followed by PDMS casting to Appendix A. Supplementary data
produce replica channels of millimetre or sub-millimetre width and
height. Channels on the PDMS replica were then sealed using a com- Supplementary data associated with this article can be found, in the
mercially available pressure-sensitive adhesive tape. It was also de- online version, at https://doi.org/10.1016/j.nbt.2018.02.002.
monstrated that the whole fabrication process can be completed within
24 h from the CAD design of the channel architecture, at an average References
cost of £5 per reactor. Simple modifications to the conventional PDMS
curing process were also implemented, in order to overcome limitations [1] Dong J, Liu J, Kang G, Xie J, Wang Y. Pushing the resolution of photolithography
down to 15 nm by surface plasmon interference. Sci Rep 2014;4:5618.
associated with the use of a relatively low-cost 3D printer. The fabri- [2] Hood RR, Wyderko T, DeVoe DL. Programmable digital droplet microfluidics using
cated reactors were further applied to the production of both inorganic a multibarrel capillary bundle. Sens Actuators B Chem 2015;220:992–9.
nanoparticles (silver nanospheres) and organic vesicular systems [3] D. Carugo, J.Y. Lee, A. Pora, R.J. Browning, L. Capretto, et al., Facile and cost-
6
D.A. Cristaldi et al. New BIOTECHNOLOGY 47 (2018) 1–7
7
Supporting information
Figure SI-1a: Cross sectional profile of the 3D printed mould base, showing the presence of diagonal grooves.
Figure SI-1b: Comparison between the Ultimaker 2+ 3D printer (white PLA material) and the High Resolution
3D printer Object Connex 350 (Veroclean® resin material). Although in this test the 0.4 mm channel was not
printed by the Ultimaker 2+, channels of 0.5 mm could be generated (these correspond to the inlet channels of
the reactor). The HR can reach smaller dimensions but the designed (i.e. nominal) cross sectional shape could
not be accurately reproduced (see SI-1c).
Figure SI-1c: a) Ultimaker 2+ mould test using the Alicona microscope. b) 3D morphological reproduction of
the designed 0.80 mm × 1.33 mm (width × height) channel. c) Object Connex 350 mould test under the Taylor-
Hobson profilometer. d) Profile graph of the actual channel (solid black line) compared with the designed
(nominal) 1.00 mm × 1.00 mm (width × height) channel (dotted line). The reproduction error for the HR 3D
printed channels is greater for smaller channel sizes.
The transport of fluids and chemical species within the reactors was characterised
numerically via computational fluid dynamic (CFD) simulations. The process comprised the
following steps: (i) design of the flow reactor geometry using Inventor Pro 2016 (Autodesk
Inc., San Rafael, CA, USA); (ii) meshing of the fluidic domain in ICEM CFD 17.0 (Ansys
Inc., Concord, MA, USA) using finite volumes of tetrahedral shape. The mesh element edge
length was equal to 0.05 mm, and was identified from a mesh dependence study as the
optimal compromise between solution accuracy and computational cost (Figure SI-2a).
Figure SI-2a: mesh dependence study performed at mesh volume sizes of 0.5, 0.1, 0.07, 0.05 and 0.03 mm. a)
velocity magnitude (in m/s) along the channel midline and b) inset of velocity (in m/s) for the mesh sizes of 0.1,
0.07, 0.05 and 0.03 mm. 48-64 processors were used for running the simulations at the TFR of 1 ml/min and
FRR of 11. Simulations at the mesh size of 0.05 mm took 3 hours and about 3500 iteration to converge, whereas
simulations at the mesh size of 0.03 mm took about 12 hours and 10,000 iteration to converge. Therefore, a
mesh size of 0.05 mm was selected as the best compromise between solution accuracy and computational cost.
The total number of mesh volumes was equal to 8’123’298; (iii) solving for steady state mass
and momentum conservation equations (i.e., Navier-Stokes equations at laminar flow regime,
see Equations 1 & 2 below), and species transport (i.e., advection-diffusion) equations (see
Equations 3 & 4 below), using Ansys® Fluent 17.0 (Ansys Inc., Concord, MA, USA).
∇ ∙ (𝒗) = 0 (Eq. 1)
∇ ∙ (𝜌𝒗𝑀𝑓,𝑖 ) = −∇ ∙ 𝐽 (Eq. 3)
Where v, ρ, µ and P represent fluid velocity, density, dynamic viscosity and pressure,
respectively. Mf,i is the mass fraction of species i. J is the diffusion flux, and Di is the mass
diffusion coefficient for species i.
To reproduce the experimental conditions, the following boundary conditions were imposed:
(i) mass flow boundary condition at the inlets, (ii) atmospheric pressure at the outlet, and (iii)
no-slip at the channel walls. The experimental values of TFR and FRR were replicated
numerically.
Fluids were assumed incompressible and Newtonian, and the IPA/ethanol-water diffusion
coefficient was set to 1 × 10−9 m2/s [1,2]. The effect of solvents’ mixing on fluid density and
viscosity was taken into consideration in the simulations. The mass fraction of IPA or ethanol
was calculated in a cross sectional plane located in the vicinity of the outlet, and the mixing
index in this specific location was determined for each TFR and FRR investigated.
CFD simulations were performed in order to gain insights into the flow field and the transport
of chemical species within the reactor (Figure SI-2b(a)), to aid the interpretation of the
experimental findings.
Figure SI-2b: Mixing index calculated over a cross sectional plane in the vicinity of the outlet surface, as shown
in a). Results refer to b) IPA-water at TFR of 1 ml/min and FRRs of 5, 7, 9 and 11; and c) ethanol-water at
TFRs of 1, 3 and 6 ml/min and FRR of 25. Contours of IPA or ethanol mass fraction are reported next to each
data point.
The extent of mixing was quantified via the mixing index [3], which is defined as a measure
of the homogeneity of chemical species (in this case water and ethanol/IPA), over a selected
surface in the reactor. Figure SI-2b shows the mixing index calculated at different fluidic
conditions, which correspond to those employed in the production of SNSs and liposomes, (b
& c respectively). Figure SI-2b(b) demonstrates that increasing the FRR from 5 to 11, while
keeping the TFR constant at 1 ml/min, resulted in increased mixing efficiency between water
and IPA. However, complete mixing was not achieved within the reactor, suggesting that the
production of SNSs continued within the collection tube at these flow regimes. Stratification
of different chemical species due to differences in density could also be observed.
Figure SI-2b(c) shows that increasing the TFR from 1 ml/min to 6 ml/min, at a fixed FRR of
25, resulted in a reduction of the mixing efficiency between ethanol and water. This could be
attributed to interfacial instabilities or vortical flow occurring at the higher flow rates, and
merits further investigations. It should however be noted that variations in the mixing index
are within a 10% range, for the different flow rates investigated.
Figure SI-3a: Representative liposome size distribution obtained by DLS, at TFR of 1 ml/min and FRR of 25. All
liposomes have mean hydrodynamic radius between 63 and 615 nm. The mean diameter is of 190 nm, with a
PDI of 0.180.
Bibliography
1. Bottaro, E., Mosayyebi, A., Carugo, D. & Nastruzzi, C. Analysis of the diffusion process by pH
indicator in microfluidic chips for liposome production. Micromachines 8, 1–16 (2017).
2. Hills, E. E., Abraham, M. H., Hersey, A. & Bevan, C. D. Diffusion coefficients in ethanol and in water
at 298K: Linear free energy relationships. Fluid Phase Equilib. 303, 45–55 (2011).
3. Hashmi, A. & Xu, J. On the quantification of mixing in microfluidics. J. Lab. Autom. 19, 488–91
(2014).