AU2020234406A1 - Microfluidic system for intracellular delivery of materials and method therefor - Google Patents

Microfluidic system for intracellular delivery of materials and method therefor Download PDF

Info

Publication number
AU2020234406A1
AU2020234406A1 AU2020234406A AU2020234406A AU2020234406A1 AU 2020234406 A1 AU2020234406 A1 AU 2020234406A1 AU 2020234406 A AU2020234406 A AU 2020234406A AU 2020234406 A AU2020234406 A AU 2020234406A AU 2020234406 A1 AU2020234406 A1 AU 2020234406A1
Authority
AU
Australia
Prior art keywords
cell
vortex
microfluidic system
junction
present disclosure
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
AU2020234406A
Inventor
Aram Chung
Jeong Soo Hur
Gum-Young KANG
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Mxt Biotech
Original Assignee
Mxt Biotech
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Mxt Biotech filed Critical Mxt Biotech
Priority claimed from PCT/KR2020/003411 external-priority patent/WO2020184992A1/en
Publication of AU2020234406A1 publication Critical patent/AU2020234406A1/en
Assigned to MxT Biotech reassignment MxT Biotech Request for Assignment Assignors: KOREA UNIVERSITY RESEARCH AND BUSINESS FOUNDATION
Priority to AU2023229574A priority Critical patent/AU2023229574A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L3/00Containers or dishes for laboratory use, e.g. laboratory glassware; Droppers
    • B01L3/50Containers for the purpose of retaining a material to be analysed, e.g. test tubes
    • B01L3/502Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures
    • B01L3/5027Containers for the purpose of retaining a material to be analysed, e.g. test tubes with fluid transport, e.g. in multi-compartment structures by integrated microfluidic structures, i.e. dimensions of channels and chambers are such that surface tension forces are important, e.g. lab-on-a-chip
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2200/00Solutions for specific problems relating to chemical or physical laboratory apparatus
    • B01L2200/06Fluid handling related problems
    • B01L2200/0647Handling flowable solids, e.g. microscopic beads, cells, particles
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B01PHYSICAL OR CHEMICAL PROCESSES OR APPARATUS IN GENERAL
    • B01LCHEMICAL OR PHYSICAL LABORATORY APPARATUS FOR GENERAL USE
    • B01L2300/00Additional constructional details
    • B01L2300/08Geometry, shape and general structure

Abstract

Provided is a microfluidic system for delivering external substances into cells by cell perforation performed using inertia, the system comprising a fluid channel structure through which a solution containing cells and external substances continuously flows, wherein the fluid channel structure includes a junction between one or more channels, a local vortex occurs near the interface of the junction, the cells are deformed by the vortex, the vortex causes a temporary discontinuity in the cell membrane, and the external substances are introduced into the cells by solution exchange between the cells and the fluid around the cells.

Description

WO 2020/184992 PCT/KR2020/003411
DESCRIPTION MICROFLUIDIC SYSTEM FOR INTRACELLULAR DELIVERY OF SUBSTANCES AND METHOD THEREFOR TECHNICAL FIELD
[0001] The present disclosure relates to a microfluidic system for intracellular delivery of materials and a method therefor.
BACKGROUND ART
[0002] Intracellular delivery of materials is one of the most key steps in cell engineering, in which materials are traditionally delivered by using a carrier or by physically
making nanopores in a cell/nuclear membrane. For virus or lipid carrier techniques, it is possible to deliver materials with high efficiency when optimized, but there are drawbacks such as low safety, slow delivery speed, labor/cost-intensive
carrier preparation process, and low reproducibility.
[0003] On the other hand, for methods of physically making a nanopore by applying energy to a cell membrane (e.g., electroporation or microneedle), there is an advantage that
relatively various materials are able to be delivered to various cell lines. However, low cell viability, denaturation of delivery material, and low throughput, which are caused by the invasiveness of the methods, are pointed
out as major limitations.
[0004] To address the above-mentioned drawbacks, microfluidic devices capable of processing a large number of
cells are prominently used. Typically, there is a platform that creates nanopores in cell membranes through physical
WO 2020/184992 PCT/KR2020/003411
deformation of cells when the cells pass through the
bottleneck section. However, the approach has major
drawbacks such as clogging of the bottleneck section itself
during the experiment and inconsistent material delivery
efficiency.
[0005] For example, US Patent No. 2014-0287509 discloses a
technology for inducing cell deformation by applying pressure
to cells through a channel having a bottleneck structure.
However, in this case, there is a drawback that the cells
block the bottleneck structure, and there is a drawback that
it is possible to deliver materials only to cells smaller
than the size of a constriction. Furthermore, there is also
a drawback that the cost rises because a channel having a
fine diameter has to be used.
[0006] Therefore, it is urgent to develop an innovative
next-generation intracellular material delivery platform
capable of delivering various materials into cells uniformly
and with high efficiency while making use of the high
processing function of the microfluidic device. DISCLOSURE OF THE INVENTION TECHNICAL PROBLEM
[0007] Accordingly, in order to solve the above-mentioned
problems, an object of the present disclosure is to provide a
system and method capable of delivering materials to a large
number of cells with high efficiency without using a new
active delivery means. TECHNICAL SOLUTION
[0008] In order to solve the problems described above,
according to an aspect of the present disclosure, there is
provided a microfluidic system delivering external materials
into a cell by cell mechanoporation using inertia and
WO 2020/184992 PCT/KR2020/003411
inertial effects, the microfluidic system including a fluidic
channel structure through which a solution containing a cell
and external materials flows continuously, in which the
fluidic channel structure includes a junction between one or
more channels, a localized vortex is generated near an
interface of the junction, the cell is deformed by the vortex,
and transient discontinuities are generated in a cell
membrane by fluidic cell deformation and the external
materials are introduced into the cell via solution exchange
between the cell and fluid around the cell.
[0009] In an embodiment of the present disclosure, the
fluidic channel structure including the junction between one
or more channels may include a junction including a T, Y,
cross shape, or a combination thereof.
[0010] In an embodiment of the present disclosure, the
fluidic channel structure may include a cavity near a fluid
stagnation point when the fluidic channel structure is a
channel of the T or Y shape.
[0011] In an embodiment of the present disclosure, the
cavity may have a shape of a circle, an ellipse, an elongate
slit, a square, a rectangle, a trapezoid, a polygon, and a
combination thereof, and a modification thereof.
[0012] In an embodiment of the present disclosure, a
diameter of the cavity may be determined according to a
diameter of the cell.
[0013] In an embodiment of the present disclosure, the
microfluidic system may further include fluid control unit
for allowing a solution to flow in the fluidic channel
structure, and the fluid control unit may allow the solution
to flow in the fluidic channel at a velocity that is at a
WO 2020/184992 PCT/KR2020/003411
level capable of generating a localized vortex near the
interface of the junction.
[0014] In an embodiment of the present disclosure, the fluid
control unit may be a syringe pump or pneumatic system.
[0015] In an embodiment of the present disclosure, a
Reynolds number (Re) of the solution may be 1 to 1000.
[0016] In an embodiment of the present disclosure, the
vortex feature may be determined by the Reynolds number.
[0017] In an embodiment of the present disclosure, the
vortex may be in a form of a closed or open recirculating
flow.
[0018] In an embodiment of the present disclosure, the
microfluidic system may be formed by combining the
microfluidic system according to any one of claims 1 to 12 in
series, parallel, or a combination thereof.
[0019] According to another aspect of the present disclosure,
there is provided a method of delivering external materials
into a cell by cell mechanoporation using inertia and
inertial effects, the method including: allowing a solution
containing the cell and external materials to continuously
flow a fluidic channel; forming a vortex by vortex generating
means near the junction; deforming the cell by the vortex;
and allowing the external materials to be introduced into the
cell through a pore created in a cell membrane by the
deforming of the cell.
[0020] in an embodiment of the present disclosure, the
vortex generating means may be a junction structure of the
fluidic channels.
[0021] In an embodiment of the present disclosure, the
fluidic channel may include a junction including a T, Y,
WO 2020/184992 PCT/KR2020/003411
cross shape, or a combination thereof. ADVANTAGEOUS EFFECTS
[0022] According to the present disclosure, a vortex is
generated by allowing a cell and external materials to flow
into a fluidic channel structure including at least one
junction, and the resulting inertia and inertial effects
deform the cell to induce transient discontinuity in a cell
membrane, thereby perforating the cell membrane. Then, a
solution exchanges between the cell and fluid around the cell
occurs through the perforated cell membrane, and as a result,
the external materials are introduced into the cell. Thus,
the present disclosure does not require vectors or active
cell delivery means (e.g., electric fields). Therefore, the
present disclosure may directly deliver external materials
(e.g., genes, plasmids, nanoparticles, or the like) into
cells with high efficiency and low cost only by solution and
channel structure features.
BRIEF DESCRIPTION OF THE DRAWINGS
[0023] FIG. 1 is a diagram for illustrating a formation of a
spiral vortex in a + junction channel.
[0024] FIG. 2 is a schematic diagram of cell deformation
caused by a spiral vortex (1) and shows high-speed microscopy
images showing rotational cell motion (scale bar: 10 pm)(2).
[0025] FIG. 3 is a diagram for describing a mechanism of
material delivery with cell deformation according to an
embodiment of the present disclosure.
[0026] FIG. 4 is a schematic diagram of two types of channel
structures according to an embodiment of the present
disclosure and a diagram for describing cell deformation in
each channel.
WO 2020/184992 PCT/KR2020/003411
[0027] FIG. 5 is a schematic diagram of a microfluidic
system in which two types of fluidic channels of FIG. 4 are
connected in series in a fluid flow direction.
[0028] FIG. 6 shows a result of an experiment on behavior
according to a Reynolds number of fluid flowing in opposite
directions toward a junction (intersection) in a + junction
channel.
[0029] FIG. 7 shows high-speed microscopy images showing a
cell (MDA-MB-231) trapping behavior in the + junction
channel.
[0030] FIG. 8 shows a graph of normalizing cell trapping and
cell deformation time by vortex breakdown of FIG. 7 as a
function of the Reynolds number.
[0031] FIG. 9 is a diagram for illustrating an intracellular
material delivery platform according to an embodiment of the
present disclosure.
[0032] FIGS. 10 to 12 are diagrams for illustrating a method
for intracellular delivery of materials using the
intracellular material delivery platform according to an
embodiment of the present disclosure.
[0033] FIG. 13 is a schematic diagram of a T-junction
channel according to an embodiment of the present disclosure.
[0034] FIG. 14 shows high-speed microscopy images
illustrating a cell deformation and vortex deformation
mechanism of a T-junction channel of a cavity structure.
Here, each arrow indicates a fluid flow direction.
[0035] FIG. 15 shows a vortex deformability index (VDI)
analysis result in a fluid having different Reynolds numbers.
[0036] FIG. 16 is a diagram for illustrating an
intracellular material delivery platform according to an
WO 2020/184992 PCT/KR2020/003411
embodiment of the present disclosure.
[0037] FIGS. 17 to 19 are diagrams for illustrating a method
for intracellular delivery of materials using the
intracellular material delivery platform according to an
embodiment of the present disclosure.
[0038] FIGS. 20 and 21 show analysis results of the amount
of materials delivered into cells.
[0039] FIG. 22 shows a result of measuring cell viability by
varying the Reynolds number under the same conditions as in
FIG. 19.
[0040] FIG. 23 shows an analysis result of delivery
efficiency for various dextran sizes.
[0041] FIG. 24 shows a result of measuring dextran delivery
efficiency in the complex microfluidic system of FIG. 5 (a
combination of the + junction channel and the T-junction
channel).
[0042] FIG. 25 shows a comparison result of the delivery
efficiency of electroporation in the related art (a neon
transfection system (Thermo Fisher Scientific, Waltham, MA,
USA)) and the method for delivery of materials based on
inertia (hydroporation) according to the present disclosure.
[0043] FIGS. 26 and 27 are photographs for analyzing the
intracellular delivery effect of gold nanoparticles (GNPs),
and a result of counting scattering spots, respectively.
[0044] FIG. 28 shows a result of measuring cell viability
depending on GNP delivery.
[0045] FIG. 29 shows a result of calculating a yield
normalized by multiplying the number of scattering spots by
the cell viability.
[0046] FIG. 30 shows an analysis result of intracellular
WO 2020/184992 PCT/KR2020/003411
delivery efficiency of mesooporous silica nanoparticles (MSN)
used as a drug, protein, and nucleic acid nanocarrier instead
of gold nanoparticles.
[0047] FIG. 31 shows an analysis result of a time-dependent
cell viability for the analyte of FIG. 30.
[0048] FIG. 32 is a histogram of fluorescence intensity
obtained by measuring a delivery effect to K562 cells using
mRNA (996-nucleotide mRNA fragment, green fluorescent
protein) as an external material.
[0049] FIG. 33 shows fluorescence images after delivery of
mRNA into K562 by (C) endocytosis and (D) the microfluidic
system according to the present disclosure.
[0050] FIG. 34 shows a result of measuring delivery
efficiency (E) and viability (F) when mRNA is delivered
according to the present disclosure as shown in FIGS. 32 and
33.
[0051] FIG. 35 shows fluorescence images after mRNA (996
nucleotide mRNA fragment, green fluorescent protein) is
delivered into K562 cells by endocytosis and a method of
Embodiment 2.
[0052] FIG. 36 shows an analysis result of (b) mRNA
transfection efficiency and (c) mean fluorescence intensity
depending on mRNA concentration.
[0053] FIG. 37 shows fluorescence images after delivery of
plasmid DNA into HEK293t by the endocytosis (control) and the
method of Embodiment 2.
[0054] FIG. 38 shows a graph of (e) analyzing plasmid DNA
(pDNA) transfection efficiency of HEK293t cells and (f) mean
intensity of HEK293t cells depending on plasmid DNA
concentration.
WO 2020/184992 PCT/KR2020/003411
[0055] FIGS. 39 and 40 are a result of western blot analysis
of the ITGal gene knockdown effect of HeLa cells (al subunit
of an integrin transmembrane receptor) into which siRNA is
delivered, and a result of comparing relative expression
levels, respectively.
[0056] FIG. 41 shows an analysis result of transfection
yields by Lipofectamine 3000, electroporation, and the
present disclosure.
[0057] FIGS. 42 to 44 are analysis results of transfection
efficiencies, cell viability, and transfection yields for
human MSC, human ADSC, and mouse BMDC.
[0058] FIG. 45 shows confocal microscopy images showing
delivery of quantum dots (qdot625) into MDA-MB-231 cells by
the present disclosure (p-Hydroporator), electroporator, and
Lipofectamine 3000.
[0059] FIG. 46 shows an analysis result of spot number
counts of quantum dots (Qdot 625) per cell.
[0060] FIG. 47 is a fluorescence intensity histogram of
Embodiment (p-Hydroporator, Ncelu = 5,000 per sample) in which
nanospheres (green fluorescent silica nanospheres, Micromod,
Germany) are delivered into K562 cells, by a negative control
(nontreated K562 cells), a positive control (K562 cells co
incubated with nanospheres, the same time and concentration
as those in Embodiment), and the present disclosure (p
Hydroporator).
[0061] FIG. 48 shows a result of measuring relative mean
fluorescence intensity.
[0062] FIGS. 49 and 50 show confocal images of two of the
groups of FIGS. 47 and 48.
BEST MODE FOR CARRYING OUT THE INVENTION
WO 2020/184992 PCT/KR2020/003411
[0063] Hereinafter, preferred embodiments of a system for intracellular delivery of materials based on inertia according to the present disclosure will be described in detail with reference to the accompanying drawings. For reference, it should be understood that the terms used in the
specification and the appended claims should not be construed as limited to general and dictionary meanings, but should be interpreted based on the meanings and concepts corresponding to technical aspects of the present disclosure on the basis
of the principle that the inventor is allowed to define terms appropriately for the best explanation. In addition, the embodiment disclosed in the present disclosure and configurations shown in the accompanying drawings are just
one preferred embodiment of the present disclosure and do not represent all technical ideas of the present disclosure. Therefore, it should be understood that the present disclosure covers various modifications and variations
provided they come within the scope of the appended claims and their equivalents at the time of filing of this application.
[0064] In order to solve the problems described above, the present disclosure provides a microfluidic system delivering external materials into a cell by cell mechanoporation using inertia and inertial effects, the microfluidic system including a fluidic channel structure through which a
solution containing a cell and external materials flows continuously, in which the fluidic channel structure includes a junction between one or more channels, a localized vortex
is generated near an interface of the junction, the cell is deformed by the vortex, and a transient discontinuous shape
WO 2020/184992 PCT/KR2020/003411
of a cell membrane is generated by the vortex and the external materials are introduced into the cell by solution exchange between the cell and fluid around the cell.
[0065] In addition, the present disclosure provides a method of delivering external materials into a cell by cell
mechanoporation using inertia and inertial effects, the method including: allowing a solution containing the cell and external materials to continuously flow a fluidic channel; forming a vortex by a vortex generating means near the
junction; deforming the cell by the vortex; and allowing the external materials to be introduced into the cell through a pore created in a cell membrane by the deforming of the cell. MODE FOR CARRYING OUT THE INVENTION
[0066] A method and system for intracellular delivery of materials according to an embodiment of the present disclosure are based on a technical feature of generating a vortex in a fluidic channel and deforming cells by the vortex
to create a pore in cell membranes. In particular, the present disclosure improves the intracellular material delivery efficiency by cell deformation caused by the vortex and cell deformation by subsequent vortex breakdown, when
cells flow into the vortex. In an embodiment of the present disclosure, the vortex is generated through a physical structure of a fluidic channel, such as a junction, but the scope of the present disclosure is not limited thereto.
[0067] According to an embodiment in which a vortex generating means is the junction, the present disclosure provides a microfluidic system having a fluidic channel
structure including one or more junctions at which at least two channels are connected, as a system for delivering an
WO 2020/184992 PCT/KR2020/003411
external material outside a cell into the cell. In the
present disclosure, "junction" means a point at which each
channel meets another channel when two or more channels are
connected in the form of T, Y, + or a combination thereof,
and in the present disclosure, at least one junction may be
provided in a single channel defined by an inlet and an
outlet of a fluid.
[0068] In the microfluidic system according to an embodiment
of the present disclosure, a vortex is generated at an
interface of the junction as the solution containing cells
and external materials flows into the junction, and at this
time, cells that continuously experience the vortex and
vortex breakdown are continuously trapped and deformed by
inertia and inertial effects, and then pores in cell
membranes, which are pathways for intracellular delivery of
materials, are formed one after another as the deformation
progresses. That is, the microfluidic system according to
the present disclosure may greatly improve the intracellular
material delivery efficiency by utilizing a fluid property
such as a Reynolds number (1 to 1000) and the channel
structure in the form of T, Y, + or a combination thereof.
[0069] In the microfluidic system of the following
embodiments of the present disclosure, a mold with channels
according to the present disclosure formed is first made by
etching a SU-8 mold or a silicon wafer through a normal
photolithography process. Then, a polydimethylsiloxane
(PDMS)-based chip is made through PDMS, and at this time, an
inlet and an outlet are made in the chip made as mentioned
above, and general slide glass is combined using plasma
treatment (Cute, Femto Science, South Korea), thereby making
WO 2020/184992 PCT/KR2020/003411
a platform device. Then, cells in the suspended state and
materials to be delivered are mixed, and then the mixture is
injected into the made chip by using a syringe pump. In this
case, the intracellular delivery of materials may be
controlled by adjusting the flow rate of the syringe pump,
and after the delivery, only the cells are separated by using
a centrifuge, and then cultured or analyzed or used according
to the purpose. However, the scope of the present disclosure
is not limited to the embodiments themselves.
[0070]
[0071] Embodiment 1: + Junction Structure Microfluidic
System
[0072] FIG. 1 is a diagram for illustrating a formation of a
spiral vortex in a + junction channel.
[0073] Referring to FIG. 1, as fluid flows in opposite
directions at both ends of the + channel, the fluid exits
into two other channels intersecting the channel, and at this
time, fluid instability near a stagnation point induces a
strong spiral localized vortex near a proximal point. The
local vortex explains the reasons for the phenomena of the
cell deformation and intracellular delivery of materials
described below. In the present disclosure, "near" refers to
a region in which a vortex of a fluid flowing into a junction
may be generated due to a junction structure.
[0074] FIG. 2 is (1) a schematic diagram of cell deformation
caused by a spiral vortex and shows high-speed microscopy
images (scale bar: 10 pm) showing rotational cell motion.
[0075] Referring to FIG. 2, in the present disclosure, when
allowing fluid to flow to a junction channel with a medium
Reynolds number (1 to 1000), cells move spirally instead of
WO 2020/184992 PCT/KR2020/003411
symmetrical cell elongation, and at this time, the cells show
a large deformation as the cells approach a stagnation point
of the vortex flow.
[0076] FIG. 3 is a diagram for describing a mechanism of
material delivery with cell deformation according to an
embodiment of the present disclosure.
[0077] Referring to FIG. 3, a nanopore is created in a cell
membrane by the cell deformation on the left, and the
external materials are introduced into the cell by solution
exchange between the cell and fluid around the cell. Then,
within 1 to 10 minutes, the cell membrane self-recovers and
closes, and the recovery time may be controlled by adjusting
the concentration of an electrolyte such as calcium in the
solution.
[0078] FIG. 4 is a schematic diagram of two types of channel
structures according to an embodiment of the present
disclosure and a diagram for describing cell deformation in
each channel.
[0079] The two types of channel structures in FIG. 4 are a
+ structure in which oppositely flowing fluid collides near the junction to form a vortex near the junction, and a T
shaped structure in which the introduced cells collide with a
channel wall.
[0080] FIG. 5 is a schematic diagram of a microfluidic
system in which two types of fluidic channels of FIG. 4 are
connected in series in a fluid flow direction.
[0081] Referring to FIG. 5, the microfluidic system of the
present disclosure has (1) a + junction channel (cross
junction) and (2) a T-junction channel (T-junction) in which
a fluid guided while passing through the vortex from the +
WO 2020/184992 PCT/KR2020/003411
junction channel collides with the channel wall.
[0082] For the above-mentioned system, cells out of the
stagnation point in the + junction channel (cross-junction)
are again guided to a channel center by inertia and collide
with the channel wall again. That is, the T, Y, and + shaped microfluidic structures according to the present
disclosure may be designed in series, parallel, or a
combination thereof with respect to fluid flow, all of which
fall within the scope of the present disclosure. In this
case, a plurality of junctions may be formed that connects a
plurality of unit channels on a single channel defined by an
inlet and an outlet in terms of cells.
[0083] FIG. 6 shows a result of an experiment on behavior
according to the Reynolds number of fluid flowing in opposite
directions toward a junction (intersection) in the
+ junction channel.
[0084] Referring to Fig. 6, at the lowest Reynolds number,
the interface between the two fluid streams remains clearly
symmetrical, and the three-dimensional vortex motion clearly
starts at the Reynolds number 37.9 (critical Reynolds number).
Then, with the increase in the Reynolds number, the
intersection between the two fluid streams becomes stronger
and more complex (see (1)).
[0085] (2) and (3) in FIG. 6 are confocal microscopy images,
where, with the increase in the Reynolds number, the
counterclockwise spiral vortex develops beyond the critical
Reynolds number, and the spiral expands vertically and
horizontally. The above result means that the vortex is
generated near the junction and its shape, pattern, velocity,
and the like vary depending on the Reynolds number of the
WO 2020/184992 PCT/KR2020/003411
fluid. This also suggests that delivery efficiency may be
improved by varying the Reynolds number depending on the
desired cell type and the type of materials to be delivered.
[0086] The system according to the present disclosure has a
specific additional effect of trapping cells and inducing
deformation thereof by cell deformation due to vortex
formation and vortex breakdown after the delivery of
materials.
[0087] FIG. 7 shows high-speed microscopy images showing a
cell (MDA-MB-231) trapping behavior in the + junction
channel.
[0088] Referring to FIG. 7, a specific trapping phenomenon
of cells is observed for a certain period of time as the
cells leave a cell deformation region by the first vortex.
That is, slightly above the stagnation point, the cells
repeatedly move up and down for approximately 30 ps and then
go out again toward the outlet. This means that immediately
after leaving a vortex region, cells are deformed for a
certain period of time and then stay in a region near the
junction (a region near the stagnation point) by the inertia
caused by the vortex breakdown, and thus the intracellular
material delivery efficiency may be greatly improved.
[0089] FIG. 8 shows a graph of normalizing cell trapping and
cell deformation time by vortex breakdown of FIG. 7 as a
function of the Reynolds number.
[0090] Referring to FIG. 8, with the increase in the
Reynolds number, the cell trapping and deformation times
increase, and cell deformation for a certain period of time
after the formation of the vortex can greatly improve the
intracellular material delivery efficiency.
WO 2020/184992 PCT/KR2020/003411
[0091] More specifically, the + junction structure microfluidic system according to the present disclosure will
be described.
[0092] FIG. 9 is a diagram for illustrating an intracellular
material delivery platform according to an embodiment of the
present disclosure.
[0093] Referring to FIG. 9, the intracellular material
delivery platform according to an embodiment of the present
disclosure includes: a first channel 100 through which a
fluid including cells and delivery material flows; a second
channel 200 that vertically intersects with the first channel
100; and a first fluid control unit 300 provided on one side
of the first channel 100 to control a fluid velocity in the
first channel 100 in a first direction.
[0094] According to an embodiment of the present disclosure,
the fluid in the first channel 100 flows in opposite
directions to the point where the first channel 100
vertically intersects with the second channel 200, and the
first fluid control unit 300 applies, to cells in the first
channel 100, kinetic energy for causing cell membrane
deformation at a level at which nanopores are formed in the
cells by the vortex formed at the point where the first
channel 100 and the second channel 200 vertically intersect
each other.
[0095] In addition to that, a second fluid control unit 300'
for controlling the fluid velocity in the first channel 100
in a second direction may be further included on the other
side of the first channel 100.
[0096] In particular, in the first channel 100, the vortex
may be formed at the point where the first channel 100 and
WO 2020/184992 PCT/KR2020/003411
the second channel 200 vertically intersect each other by the
first and second fluid control units 300 and 300' performing
controls in opposite directions, and due to the inertial
force and inertial flow that are generated in this way,
physical deformation may occur in the cell and the cell
membrane may be deformed accordingly.
[0097] Meanwhile, the intracellular material delivery
platform according to the present disclosure may deliver
nucleic acids, proteins, transcription factors, vectors,
plasmids, genetic-scissors materials, nanoparticles, and the
like. However, the present disclosure is not limited thereto.
[0098] Furthermore, the intracellular material delivery
platform is not limited in application to regenerative
medicine, cancer immunotherapy, genomic editing, or other
fields.
[0099]
[00100] FIGS. 10 to 12 are diagrams for illustrating a method
for intracellular delivery of materials using the
intracellular material delivery platform according to an
embodiment of the present disclosure.
[00101] Referring to FIG. 10, the fluid containing cells and
delivery material flows in the first channel 100 by the first
fluid control unit 300. At this time, the second fluid
control unit 300' formed on the other side of the first
channel 100 also operates such that the delivery material
flows in a direction opposite to the fluid controlled by the
first fluid control unit 300.
[00102] In an embodiment of the present disclosure, the
delivery material includes all materials that may be
delivered into cells, and specifically, genetic-scissors
WO 2020/184992 PCT/KR2020/003411
materials, plasmids, nucleic acids, proteins, nanoparticles,
and the like may all the delivery materials.
[00103]
[00104] Referring to FIG. 11, the fluid accelerated by the
first fluid control unit 300 forms a vortex along the
interface with the opposing fluid near the junction, and the
cells trapped by the vortex are deformed. Then, a nanopore
is formed in the cell by the cell deformation. The delivery
material is delivered into the cell through the nanopore.
Accordingly, it is desirable that the first and second fluid
control units 300 and 300' apply kinetic energy having a
Reynolds number of a level at which the vortex of the fluid
is formed near the junction.
[00105] According to another embodiment of the present
disclosure, vortex breakdown formed after passing through the
vortex formed in the fluid makes another cell deformation.
[00106] Referring to FIG. 12, cells passing through the
vortex experience the vortex breakdown, and cell deformation
occurs again due to inertia. Through the nanopore formed in
the cell membrane by another cell deformation occurring at
this time, the delivery material is again delivered into the
cell, which greatly improves intracellular material delivery
efficiency.
[00107]
[00108] Embodiment 2: T or Y Junction Structure Microfluidic
System
[00109] Another embodiment of the present disclosure provides
an intracellular material delivery system using a T or Y
junction channel having a cavity. In the present disclosure,
a cavity refers to an empty space formed in a channel at a
WO 2020/184992 PCT/KR2020/003411
stagnation point, which is a structure in which when the
cells of the solution and the channel wall collide at the
junction, a collision area between the cells and a channel
wall is eliminated or reduced.
[00110] In this case, similar to the + junction channel
described above, a localized vortex is formed near the T
junction, and sequentially, cell deformation, formation of
the nanopore in the cell membrane, intracellular delivery of
external materials, and closure of the nanopore in the cell
membrane are performed.
[00111] FIG. 13 is a schematic diagram of a T-junction
channel according to an embodiment of the present disclosure.
[00112] Referring to FIG. 13, it can be seen that a channel
shaped cavity 1 is formed near a junction of a T-channel. In
an embodiment of the present disclosure, cells mixed with
external materials delivered into the cells are injected into
the microfluidic channel of FIG. 13 by using a syringe pump
in the related art (PHD 2000, Harvard Apparatus, USA), as a
fluid control unit, at an appropriate Reynolds number.
[00113] Upon injection, the cells are concentrated in the
channel center by inertia, and the cells collided with the
channel wall. Each cell penetrates a portion of the above
mentioned cavity ((1) of FIG. 13) and is deformed (refer to
the photographs on the right of FIG. 13). Then, after the
first cell deformation by the collision, the cell is trapped
in a localized vortex near the stagnation point (point (2) in
FIG. 13) and is hydrodynamically deformed to form nanopores
in the cell membrane, as described in FIG. 3.
[00114] Then, the cell that has passed through the vortex
region are trapped and then deformed by vortex breakdown
WO 2020/184992 PCT/KR2020/003411
again ((3) of Fig. 13). An advantage of such additional cell
trapping and deformation is the improvement of intracellular
material delivery efficiency as described above in FIG. 7.
[00115] In an embodiment of the present disclosure, the
cavity is used in the T-junction channel structure, the
advantage of which is to reduce cell damage due to collision
with a rigid solid channel wall by allowing cells to collide
with the channel wall of a fluid form, instead of the channel
wall of a rigid solid form. Another advantage is to
virtually prevent cell clogging by creating a stagnation
point upstream in the fluid flow direction to support complex
fluid behavior patterns. Accordingly, the form, size, shape,
and the like of the cavity structure may vary depending on
the cell. For example, the cavity may include not only an
elongated slit structure as shown in FIG. 13 but also a
circular, oval, elongated slit, square, rectangular,
trapezoidal, polygonal, a combination thereof, and a modified
form thereof, and at least as long as the introduced cells do
not collide with the channel wall as they are, they all
belong to the cavity of the present disclosure.
[00116] In addition, the cavity diameter is determined
depending on the cell diameter, and in particular, the cavity
diameter is preferably on the order of 10% to 5 times the
cell size. The cavity diameter according to the present
disclosure is within the scope of the present disclosure, at
least as long as the cavity diameter can reduce the collision
force caused by the collision between the cells, which are
introduced to the junction through the solution, and the
channel wall.
[00117] FIG. 14 shows high-speed microscopy images
WO 2020/184992 PCT/KR2020/003411
illustrating a cell deformation and vortex deformation
mechanism of a T-junction channel of a cavity structure.
Here, each arrow indicates a fluid flow direction.
[00118] Referring to FIG. 14, trapping by the localized
vortex near the stagnation point ((1) in FIG. 14) and
trapping of the cell passing through the stagnation point by
vortex breakdown due to fluid instability near the stagnation
point ((2) in FIG. 14) can be confirmed, which shows that the
vortex, the trapping by the vortex breakdown, and the
deformation occur continuously, similar to the channel of the
+ channel described above. In particular, in (2) of FIG. 14, it can be seen that the cell maintains a static state for
about 20 ps and then exits downstream.
[00119] FIG. 15 shows a vortex deformability index (VDI)
analysis result in a fluid having different Reynolds numbers.
Here, VDI is defined as the following formula, which can be
interpreted as the degree of cell deformability index and the
duration of cell membrane permeation, which in turn indicates
the intracellular material delivery efficiency.
[00120] VDI=t(1-c)U/D
[00121] where t is the cell trapping time in the vortex, c is
the circularity (c , where A and P are the area and
radius in the state with maximum deformation, respectively),
U is the average velocity of the fluid, and D is the cell
diameter.
[00122] Referring to FIG. 15, with the increase in the
Reynolds number, the VDI increases during (1) the vortex and
(2) the vortex breakdown, indicating that a high Reynolds
WO 2020/184992 PCT/KR2020/003411
number has a higher intracellular material delivery
efficiency.
[00123] Hereinafter, the T-junction structure microfluidic
system according to the present disclosure will be described
in more detail.
[00124] FIG. 16 is a diagram for illustrating an
intracellular material delivery platform according to an
embodiment of the present disclosure.
[00125] The intracellular material delivery platform
according to the present disclosure includes: a third channel
101 forming a pathway through which a fluid including cells
and delivery material moves; a fourth channel 201 vertically
extending to both sides of the third channel 101 at an end of
the third channel 101; and a fluid control unit 301 provided
at the third channel 101 to control a fluid velocity in the
third channel 101.
[00126] FIGS. 17 to 19 are diagrams for illustrating a method
for intracellular delivery of materials using the
intracellular material delivery platform according to an
embodiment of the present disclosure.
[00127] Referring to FIG. 17, the fluid containing cells and
delivery material flows in the third channel 101 by the fluid
control unit 301. In an embodiment of the present disclosure,
the delivery material includes all materials that may be
delivered into cells, and genetic-scissors materials,
plasmids, nucleic acids, proteins, nanoparticles, and the
like are all the delivery materials.
[00128] Referring to FIG. 18, the cells in the third channel
101 accelerated by the fluid control unit 301 are trapped by
the vortex formed near the junction and then deformed. This
WO 2020/184992 PCT/KR2020/003411
is the same as that described with reference to FIGS. 13 and
14. Then, the cells collide with a partition wall of the
fourth channel 201 connected to the end of the third channel
101.
[00129] At this time, the fourth channel 201 is provided with
a slit-shaped cavity 401 formed in the same direction as the
fluid flow direction in the third channel 101, and the cavity
produces effects of 1) preventing cell damage by physical
collision, 2) preventing clogging, and 3) forming the
stagnation point upstream.
[00130] In the present disclosure, as described above, a
plurality of unit microfluidic systems may be connected in
series or parallel or a combination thereof to construct an
entire system. In this case, the vortex may occur in the
recirculated stream in a circulation mode, where the vortex
may be a closed or open stream.
[00131]
[00132] Experimental examples
[00133] Experimental Example 1
[00134] In the experimental example, delivery characteristics
of the microfluidic system based on Embodiment 1 were
analyzed.
[00135] FIG. 19 shows a result of comparing the intracellular
material delivery efficiencies after 18 hours by the
endocytosis (control group) and the intracellular mass
transfer method according to the present disclosure (spiral
hydroporation). Here, 3-5 kDa fluorescein isothiocyanate
(FITC)-conjugated dextran was delivered into MDA-MB-231 cells,
and the result was shown as a fluorescence image (scale bar:
pm). In particular, it should be noted here that the
WO 2020/184992 PCT/KR2020/003411
delivery of dextran into MDA-MB-231 cells is known to be very
difficult. In the experiments described below, unless
otherwise noted, cells are MDA-MB-231.
[00136] Referring to FIG. 19, the intracellular material
delivery effect according to the present disclosure can be
confirmed through multiple FITC signals on the right.
[00137] FIGS. 20 and 21 show analysis results of the amount
of materials delivered into cells. In the analysis, the
delivery efficiency was defined as a fraction of fluorescence
signals equal to or greater than 5%, which corresponds to the
red dotted line.
[00138] Referring to FIGS. 20 and 21, it can be seen that at
Reynolds number 366, a delivery efficiency of approximately
96.5% was achieved, and the fluorescence intensity increases
with the increase in Re and the histogram profile shifted to
the right. This means that the amount of materials delivered
into cells may be adjusted by adjusting the Reynolds number.
[00139] FIG. 22 shows a result of measuring cell viability by
varying the Reynolds number under the same conditions as in
FIG. 19.
[00140] Referring to FIG. 22, with regard to cell viability,
a decrease in viability was observed at a higher Reynolds
number, presumably because the higher the Reynolds number,
the greater the cell perturbation.
[00141] Furthermore, in order to further investigate cell
viability, a standard MTT assay was performed via metabolic
function and the result is shown in FIG. 22. The overall
trends of the trypan blue exclusion method and the MTT assay
were in good agreement with each other, whereas the MTT assay
showed slightly lower viability values at high Reynolds
WO 2020/184992 PCT/KR2020/003411
numbers.
[00142] FIG. 23 shows an analysis result of delivery
efficiency for various dextran sizes. In the analysis,
dextrans of molecular weights ranging from 3 to 2000 kDa,
corresponding to a hydraulic diameter of 2 to 55 nm, were
used, where FITC-dextrans (3 to 5, 70, 150, 500, and 2000
kDa) of 5 different sizes and the same concentration were
delivered into MDA-MB-231 cells in the same manner as in
Embodiment 1 under the same flow conditions (Re = 366) and
the efficiency was calculated.
[00143] Referring to FIG. 23, it was shown that relatively
small dextran (<70 kDa) had higher delivery efficiency than
relatively large dextran. This is because, for small dextran,
convective and diffuse transport of dextran across the cell
membrane occurs, whereas for large dextran, convective
transport is the dominant transport.
[00144] FIG. 24 shows a result of measuring dextran delivery
efficiency in the complex microfluidic system of FIG. 5 (a
combination of the + junction channel and the T-junction
channel).
[00145] Referring to FIG. 24, it can be seen that the complex
microfluidic system of FIG. 5, in which continuous cell
deformation was performed, exhibited relatively higher
delivery efficiency than the stand-alone one.
[00146] FIG. 25 shows a comparison result of the delivery
efficiency of electroporation in the related art (a neon
transfection system (Thermo Fisher Scientific, Waltham, MA,
USA)) and the method for delivery of materials based on
inertia (hydroporation) according to the present disclosure.
In the experimental example, 500 and 2000 kDa FITC-dextrans
WO 2020/184992 PCT/KR2020/003411
were used as external materials.
[00147] Referring to FIG. 25, for the 2000 kDa FITC-dextran,
the efficiency of the present disclosure was approximately 4
times higher than that of electroporation. In particular,
the method according to the present disclosure greatly
improves the delivery efficiency of macromolecules that are
difficult to be delivered into cells by electroporation,
which suggests the possibility of intracellular delivery of
molecular weight materials that is not possible to be
achieved by electroporation techniques in the related art.
[00148] FIGS. 26 and 27 are photographs for analyzing the
intracellular delivery effect of gold nanoparticles (GNPs),
and a result of counting scattering spots, respectively.
Here, (A) shows a result of hydroporation (SH) according to
the present disclosure, (B) shows a result of electroporation
(EP), and (C) shows a result of endocytosis (EC).
[00149] Referring to FIGS. 26 and 27, it can be seen that
very large GNPs (200 nm in diameter) were successfully
delivered into MDAMB-231 cells through the fluidic system
according to the present disclosure (A). On the other hand,
it can be seen that only a few scattering points were
detected although electroporation was able to deliver GNPs
(B). Furthermore, the endocytosis mechanism exhibited lower
particle delivery efficiency than that of electroporation (C).
[00150] FIG. 28 shows a result of measuring cell viability
depending on GNP delivery.
[00151] Referring to FIG. 28, it can be seen that the method
and system for intracellular delivery of materials according
to the present disclosure exhibited higher viability than
that of electroporation.
WO 2020/184992 PCT/KR2020/003411
[00152] FIG. 29 shows a result of calculating a yield
normalized by multiplying the number of scattering spots by
the cell viability (FIG. 5F).
[00153] Referring to FIG. 29, it can be seen that the
material delivery efficiency according to the present
disclosure was three times higher than that of
electroporation or endocytosis.
[00154] FIG. 30 shows an analysis result of intracellular
delivery efficiency of mesoporous silica nanoparticles (MSN)
used as a drug, protein, and nucleic acid nanocarrier instead
of gold nanoparticles. In the experiment, doxorubicin (DOX),
an anticancer drug, was loaded into MSNs and the MSNs were
delivered into MDA-MB-231 cells.
[00155] Referring to FIG. 30, it can be seen that the method
according to the present disclosure (spiral control)
exhibited a number of bright fluorescence points compared to
endocytosis. This suggests that a carrier carrying a
functional material can be effectively delivered into the
cell by the method according to the present disclosure.
[00156] FIG. 31 shows an analysis result of a time-dependent
cell viability for the analyte of FIG. 30.
[00157] Referring to FIG. 31, DOX cytotoxicity to MDA-MB-231
cells was measured by the trypan blue exclusion method, and
it can be seen that approximately 85% of cancer cells were
killed after 6 hours. The result indicates that the
microfluidic system according to the present disclosure may
investigate DOX-induced cell death without using a surface
ligand-based DOX delivery method in the related art.
[00158] FIG. 32 is a histogram of fluorescence intensity
obtained by measuring a delivery effect to K562 cells using
WO 2020/184992 PCT/KR2020/003411
mRNA (996-nucleotide mRNA fragment, green fluorescent
protein) as an external material. In FIG. 32, (A) shows a
result of endocytosis and (B) shows a result of the
microfluidic system according to the present disclosure,
where EGFP protein expression was assessed based on mRNA
delivery (2 pg/mL) using a flow cytometer.
[00159] Referring to FIG. 32, it can be seen that the method
according to the present disclosure exhibited a very high
protein expression level. This means that the method and
system for intracellular delivery of materials according to
the present disclosure may be used for chimeric antigen
receptor-expressing T-cells (CAR-T) without the use of
vectors or vaccines.
[00160] FIG. 33 shows fluorescence images after delivery of
mRNA into K562 by (C) endocytosis and (D) the microfluidic
system according to the present disclosure.
[00161] Referring to FIG. 33, in the method according to the
present disclosure, strong EGFP signals were detected
compared to endocytosis, which results in higher mRNA
delivery efficiency.
[00162] FIG. 34 shows a result of measuring delivery
efficiency (E) and viability (F) when mRNA is delivered
according to the present disclosure as shown in FIGS. 32 and
33.
[00163] Referring to FIG. 34, the delivery efficiency of up
to approximately 92% was achieved without sacrificing cell
viability, which represents the high potential of the
platform for use of the platform for immunotherapy research
although further investigations are needed to test human
immune cells in the future.
WO 2020/184992 PCT/KR2020/003411
[00164]
[00165] Experimental Example 2
[00166] In the experimental example, delivery characteristics
of the microfluidic system (p-Hydroporator) based on
Embodiment 2 were analyzed.
[00167] FIG. 35 shows fluorescence images after mRNA (996
nucleotide mRNA fragment, green fluorescent protein) is
delivered into K562 cells by endocytosis and the method of
Embodiment 2.
[00168] Referring to FIG. 35, it can be seen that, unlike
endocytosis (control), green fluorescence was detected when
mRNA was delivered into cells by the method of Embodiment 2.
This proves that the T-junction channel system according to
the present disclosure (Embodiment 2) enables intracellular
delivery of mRNA with high efficiency as in Embodiment 1
described above.
[00169] FIG. 36 shows an analysis result of (b) mRNA
transfection efficiency and (c) mean fluorescence intensity
depending on mRNA concentration.
[00170] Referring to FIG. 36, different concentrations of
mRNAs were accessed based on flow cytometry analysis, and in
2 pg/ml, transfection efficiency was achieved up to
approximately 93%. This is an efficiency that is not
possible in compression-based microfluidic systems such as
bottleneck structures in the related art.
[00171] Unlike mRNA, which binds to the cell substrate first,
DNA has to first pass through the cell membrane and enter a
nuclear membrane through a nuclear pore. Furthermore, in
terms of material delivery, naked plasmid DNA has a drawback
in that it is easily degraded by nucleases and has a high
WO 2020/184992 PCT/KR2020/003411
viscosity. In addition, high-density cytoplasm does not
provide favorable conditions for long, twisted DNA to reach
the nucleus purely by diffusion. In order to overcome the
above-mentioned drawback, the present inventors provide a
fluid-based microfluidic system according to the present
disclosure as a method for delivering plasmid DNA to a
nucleus. To this end, in the experimental example, an
experiment was performed to encode copepod GFP and deliver
7.9 kbp plasmid DNA to HEK293t cells.
[00172] FIG. 37 shows fluorescence images after delivery of
plasmid DNA into HEK293t by the endocytosis (control) and the
method of Embodiment 2.
[00173] Referring to FIG. 37, it can be seen that the method
according to the present disclosure exhibited a very strong
fluorescence image.
[00174] FIG. 38 shows a graph of analyzing plasmid DNA (pDNA)
transfection efficiency (E) of HEK293t cells and mean
intensity (F) of HEK293t cells depending on plasmid DNA
concentration.
[00175] Referring to FIG. 38, it can be seen that with an
increase in the pDNA concentration, the transfection
efficiency increases, and the mean fluorescence intensity
also increases.
[00176] FIGS. 39 and 40 are a result of western blot analysis
of the ITGal gene knockdown effect of HeLa cells (al subunit
of an integrin transmembrane receptor) into which siRNA is
delivered, and a result of comparing relative expression
levels, respectively. Here, the knockdown effect of the
ITGal gene was compared using cationic Lipofectamine 3000 in
the related art, which is known as the siRNA delivery method,
WO 2020/184992 PCT/KR2020/003411
as a comparative example.
[00177] Referring to FIGS. 39 and 40, when the present
disclosure was used, ITGA1 expression (197 kDa) was almost
eliminated; whereas, in Lipofectamine 3000, the comparative
example, only partial knockdown was observed. According to
the result, the present disclosure has at least three times
greater knockdown efficiency (97% vs. 30%), suggesting that
the present disclosure has a high potential for use in gene
editing.
[00178] In an experiment below, non-functional materials or
extremely small molecules (e.g., calcein, propidium iodide,
and 3 kDa dextran) were delivered to cell lines. In the
experiment below, mRNA was chosen as a delivery target, since
protein expression after mRNA delivery occurs in the
cytoplasm and is guided to fat, is well controllable, and is
easily comparable by dose-dependent transfection.
[00179] In the experiment, EGFP mRNA was delivered into
Harton's jelly human umbilical cord mesenchymal stem cells
(MSCs), human adipose derived stem cells (ADSCs), and mouse
bone marrow derived dendritic cells (BMDC) by using the
method of Embodiment 2 (electroporator), Lipofectamine 3000,
and the electroporation neon transfection system
(electroporator; Thermo Fisher Scientific).
[00180] FIG. 41 shows an analysis result of transfection
yields by Lipofectamin 3000, electroporation, and the present
disclosure.
[00181] Referring to FIG. 41, it can be seen that the present
disclosure exhibited a very high transfection yield compared
to Lipofectamine 3000 and electroporation. In the analysis
result, the transfection yield was defined as the product of
WO 2020/184992 PCT/KR2020/003411
transfection efficiency and cell viability, which can be
understood as the ratio of viable cells to cells transfected
by material delivery.
[00182] FIGS. 42 to 44 are analysis results of transfection
efficiencies, cell viability, and transfection yields for
human MSC, human ADSC, and mouse BMDC.
[00183] Referring to FIGS. 42 to 44, it can be seen that
lipofection exhibited improved cell viability in all cell
types compared to the present disclosure (p-Hydroporator) and
electroporation, but exhibited substantially low transfection
efficiency in all cell types.
[00184] In addition, for MSC and ADSC, the transfection
efficiency was slightly higher in the electroporator than in
the present disclosure (p-Hydroporation). However, for all
cell types, the present disclosure (p-Hydroporator) exhibited
higher cell viability without the use of special stabilized
buffer required for the electroporator. Furthermore, the
cell viability of the present disclosure may further increase
cell viability simply by adding trehalose or polymer to the
cell media.
[00185] For BMDCs, the present disclosure exhibited higher
transfection efficiency and cell viability than
electroporation, indicating that the present disclosure has a
high potential to be used for cancer immunotherapy.
[00186] The present disclosure has several advantages over
electroporation in the related art with respect to immune
cell therapy. First, electroporation is known to have a side
effect of altering important properties of primary T cells
(e.g., non-specific cytokine burst and blunted IFN-y
response), lowering the therapeutic performance. However,
WO 2020/184992 PCT/KR2020/003411
the low scalability of electroporation (treating 104 to 105
cells per run) is a drawback regarding its potential clinical
usage in cancer immunotherapy, which generally requires the
treatment of 108 cells.
[00187] However, in the present disclosure, 1 x 106 cells/min
may be processed while the same level of delivery efficiency
is maintained and this throughput is based on a single
microchannel, and thus the present disclosure may achieve the
cell throughput required for cancer immunotherapy through
multiplexing and parallelization of microchannels.
[00188] In an experiment below, quantum dots (Dibenzo
cyclooctyne (DOBI)) and silica nanospheres, which are widely
used as target molecules, were determined as intracellular
delivery materials, and delivery properties to cells (MDA-MB
231) were analyzed.
[00189] FIG. 45 shows confocal microscopy images showing
delivery of quantum dots (qdot625) into MDA-MB-231 cells by
the present disclosure (p-Hydroporator), electroporator, and
Lipofectamine 3000.
[00190] Referring to FIG. 45, all the methods showed
excellent delivery efficiency, but the result in which
quantum dots were well dispersed in the cytoplasm was shown
when intracellular delivery was performed by the method
according to the present disclosure. In cells treated with
electroporation and Lipofectamine 3000, multiple red spots
were observed, indicating the possibility of aggregation or
endosome entrapment of the quantum dots. Since such quantum
dot aggregation eventually causes a decrease in the
efficiency of intracellular delivery of materials, the result
means that the method for intracellular delivery of materials
WO 2020/184992 PCT/KR2020/003411
according to the present disclosure is also useful for
intracellular delivery of micro-materials.
[00191] FIG. 46 shows an analysis result of spot number
counts of quantum dots (Qdot 625) per cell.
[00192] Referring to FIG. 46, the analysis result represents
the degree of aggregation of quantum dots, and it can be seen
that in the present disclosure, the number of quantum dots
per cell was the lowest. That is, electroporation or
lipofection exhibited three- and four-fold levels of quantum
dot count compared to the present disclosure. This is also
consistent with the analysis result of FIG. 45, indicating
that when intracellular delivery of particles such as quantum
dots is performed according to the present disclosure,
quantum dots are well dispersed in the cytoplasm by rapid
solution exchange through the cell membrane after cell
deformation.
[00193] FIG. 47 is a fluorescence intensity histogram of
Embodiment (p-Hydroporator, Nceui = 5,000 per sample) in which
nanospheres (green fluorescent silica nanospheres, Micromod,
Germany) are delivered into K562 cells, by a negative control
(nontreated K562 cells), a positive control (K562 cells co
incubated with nanospheres, the same time and concentration
as those in Embodiment), and the present disclosure (p
Hydroporator).
[00194] Further, FIG. 48 shows a result of measuring relative
mean fluorescence intensity.
[00195] Referring to FIGS. 47 and 48, on average, higher
fluorescence intensity was observed in cells treated
according to the present disclosure, but high fluorescence
signals were detected in the co-incubation group in flow
WO 2020/184992 PCT/KR2020/003411
cytometry analysis. Considering the short incubation time
and the excessively large nanosphere size for endocytosis,
the fluorescence in the positive control presumably seems to
be obtained from nanospheres adhering to the cell surfaces.
[00196] FIGS. 49 and 50 show confocal images of two of the
groups of FIGS. 47 and 48. Here, the cell membrane was
visualized by using DiD lipophilic carbocyanine dye.
[00197] As shown in FIGS. 49 and 50, the green fluorescence
signals from the positive control (co-incubation) were
present only in the cell membrane, whereas bright green
fluorescence from the cytoplasm was only observed in cells
treated according to the present disclosure. This indicates
that only the system for intracellular delivery of materials
according to the present disclosure is possible as a
technology capable of delivering nanospheres into cells,
considering the positive control in which only the action of
nanospheres adhering to the cell surfaces by electrostatics
is confirmed.
[00198]
[00199] INDUSTRIAL APPLICABILITY
[00200] The microfluidic system for delivering external
materials into cells by cell mechanoporation using inertia
according to the present disclosure has industrial
applicability in the bio and medicine fields requiring
delivery of materials into cells.
[00201]
[00202]

Claims (16)

WO 2020/184992 PCT/KR2020/003411 CLAIMS
1. A microfluidic system delivering external
materials into a cell by cell mechanoporation using inertia,
the microfluidic system comprising:
a fluidic channel structure through which a solution
containing a cell and external materials flows continuously,
wherein the fluidic channel structure includes a
junction between one or more channels,
a localized vortex is generated near an interface of
the junction,
the cell is deformed by the vortex, and
transient discontinuities are generated in a cell
membrane by the vortex and the external materials are
introduced into the cell by solution exchange between the
cell and fluid around the cell.
2. The microfluidic system of claim 1, wherein the
fluidic channel structure including the junction between one
or more channels includes a junction including a T, Y, cross
shape, or a combination thereof.
3. The microfluidic system of claim 2, wherein the
fluidic channel structure includes a cavity near a fluid
stagnation point when the fluidic channel structure is a
channel of the T or Y shape.
4. The microfluidic system of claim 3, wherein the
cavity has a shape of a circle, an ellipse, an elongate slit,
a square, a rectangle, a trapezoid, a polygon, and a
WO 2020/184992 PCT/KR2020/003411
combination thereof, and a modification thereof.
5. The microfluidic system of claim 3, wherein a
diameter of the cavity is determined according to a diameter
of the cell.
6. The microfluidic system of claim 3, wherein the
cavity has a structure for eliminating or reducing a
collision area between the cell and a channel wall when the
cell of the solution collides with the channel wall at the
junction.
7. The microfluidic system of any one of claims 1 to
6, further comprising a fluid control unit for allowing a
solution to flow in the fluidic channel structure,
wherein the fluid control unit allows the solution to
flow in the fluidic channel at a velocity that is at a level
capable of generating a localized vortex near the interface
of the junction.
8. The microfluidic system of claim 7, wherein the
fluid control unit is a syringe pump or pneumatic system.
9. The microfluidic system of any one of claims 1 to
8, wherein a Reynolds number (Re) of the solution is 1 to
1000.
10. The microfluidic system of claim 9, wherein the
vortex is determined by the Reynolds number.
WO 2020/184992 PCT/KR2020/003411
11. The microfluidic system of any one of claims 1 to
8, wherein the vortex is in a form of a closed or open
recirculating flow.
12. The microfluidic system of any one of claims 1 to
8, wherein the fluidic channel has a plurality of the
junctions at least in a channel between an inlet and an
outlet of the solution.
13. The microfluidic system of claim 10, wherein the
microfluidic system is formed by combining the microfluidic
system according to any one of claims 1 to 12 in series,
parallel, or a combination thereof.
14. A method of delivering external materials into a
cell by cell mechanoporation using inertia, the method
comprising:
allowing a solution containing the cell and external
materials to continuously flow a fluidic channel;
forming a vortex by a vortex generating means near the
junction;
deforming the cell by the vortex; and
allowing the external materials to be introduced into
the cell through a pore created in a cell membrane by the
deforming of the cell.
15. The method of claim 14, wherein the vortex
generating means is a junction structure of the fluidic
channels.
WO 2020/184992 PCT/KR2020/003411
16. The method of claim 15, wherein the fluidic
channel includes a junction including a T, Y, cross shape, or
a combination thereof.
[DRAWINGS]
[FIG. 1]
[FIG. 2]
[FIG. 3]
[FIG. 4]
[FIG. 5]
[FIG. 6]
[FIG. 7]
[FIG. 8]
[FIG. 9]
[FIG. 10]
[FIG. 11]
[FIG. 12]
[FIG. 13]
[FIG. 14]
[FIG. 15]
[FIG. 16]
[FIG. 17]
[FIG. 18]
[FIG. 19]
[FIG. 20]
[FIG. 21]
[FIG. 22]
[FIG. 23]
[FIG. 24]
[FIG. 25]
[FIG. 26]
[FIG. 27]
[FIG. 28]
[FIG. 29]
[FIG. 30]
[FIG. 31]
[FIG. 32]
[FIG. 33]
[FIG. 34]
[FIG. 35]
[FIG. 36]
[FIG. 37]
[FIG. 38]
[FIG. 39]
[FIG. 40]
[FIG. 41]
[FIG. 42]
[FIG. 43]
[FIG. 44]
[FIG. 45]
[FIG. 46]
[FIG. 47]
[FIG. 48]
[FIG. 49]
[FIG. 50]
AU2020234406A 2019-03-12 2020-03-11 Microfluidic system for intracellular delivery of materials and method therefor Abandoned AU2020234406A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2023229574A AU2023229574A1 (en) 2019-03-12 2023-09-15 Microfluidic system for intracellular delivery of materials and method therefor

Applications Claiming Priority (7)

Application Number Priority Date Filing Date Title
KR10-2019-0028069 2019-03-12
KR20190028069 2019-03-12
KR10-2019-0134094 2019-10-25
KR1020190134094A KR102354673B1 (en) 2019-03-12 2019-10-25 Inertia based microfluidic intracellular delivery platforms
KR1020190134095A KR102217407B1 (en) 2019-03-12 2019-10-25 Inertia based microfluidic intracellular delivery platforms and intracellular mass transfer method using the same
KR10-2019-0134095 2019-10-25
PCT/KR2020/003411 WO2020184992A1 (en) 2019-03-12 2020-03-11 Microfluidic system for intracellular delivery of substances and method therefor

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2023229574A Division AU2023229574A1 (en) 2019-03-12 2023-09-15 Microfluidic system for intracellular delivery of materials and method therefor

Publications (1)

Publication Number Publication Date
AU2020234406A1 true AU2020234406A1 (en) 2021-10-21

Family

ID=72707082

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2020234406A Abandoned AU2020234406A1 (en) 2019-03-12 2020-03-11 Microfluidic system for intracellular delivery of materials and method therefor

Country Status (3)

Country Link
KR (2) KR102217407B1 (en)
AU (1) AU2020234406A1 (en)
CA (1) CA3133045A1 (en)

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102499546B1 (en) * 2020-10-07 2023-02-14 고려대학교 산학협력단 Method for intracellular delivery via droplet deformation and chip for the same
WO2022220539A1 (en) * 2021-04-12 2022-10-20 ㈜엠엑스티바이오텍 Intracellular delivery platform

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
BR112014009346B1 (en) 2011-10-17 2020-09-15 Massachusetts Institute Of Technology INTRACELLULAR DELIVERY

Also Published As

Publication number Publication date
KR102217407B1 (en) 2021-02-22
KR102354673B1 (en) 2022-01-25
CA3133045A1 (en) 2020-09-17
KR20200109229A (en) 2020-09-22
KR20200109228A (en) 2020-09-22

Similar Documents

Publication Publication Date Title
Morshedi Rad et al. A comprehensive review on intracellular delivery
Liu et al. Current developments and applications of microfluidic technology toward clinical translation of nanomedicines
Balbino et al. Microfluidic devices for continuous production of pDNA/cationic liposome complexes for gene delivery and vaccine therapy
Kizer et al. Hydroporator: a hydrodynamic cell membrane perforator for high-throughput vector-free nanomaterial intracellular delivery and DNA origami biostability evaluation
US7811603B2 (en) Microfluidic device for forming monodisperse lipoplexes
US20180119212A1 (en) Assays and other reactions involving droplets
Hur et al. Microfluidic and nanofluidic intracellular delivery
Luo et al. Microfluidic devices in fabricating nano or micromaterials for biomedical applications
CN107429262A (en) A kind of method for being used for machinery and the transfection of hydrodynamic force microfluid and the equipment for it
US20150174549A1 (en) High-throughput synthesis of nanoparticles
US11745179B2 (en) Microfluidic systems and methods for lipoplex-mediated cell transfection
Li et al. Lipoplex‐mediated single‐cell transfection via droplet microfluidics
De La Vega et al. Uniform polymer microspheres: monodispersity criteria, methods of formation and applications
Debus et al. Optimized preparation of pDNA/poly (ethylene imine) polyplexes using a microfluidic system
AU2020234406A1 (en) Microfluidic system for intracellular delivery of materials and method therefor
US20230219086A1 (en) Method and device for cell or microvesicle isolation
Ilhan-Ayisigi et al. Advances in microfluidic synthesis and coupling with synchrotron SAXS for continuous production and real-time structural characterization of nano-self-assemblies
Eş et al. High-throughput conventional and stealth cationic liposome synthesis using a chaotic advection-based microfluidic device combined with a centrifugal vacuum concentrator
Hsieh et al. Nonviral gene vector formation in monodispersed picolitre incubator for consistent gene delivery
Jürgens et al. Lab-scale siRNA and mRNA LNP manufacturing by various microfluidic mixing techniques–an evaluation of particle properties and efficiency
Velmurugan et al. Role of microfluidics in drug delivery
EP3939701A1 (en) Microfluidic system for intracellular delivery of substances and method therefor
Lee et al. High-throughput nanoscale lipid vesicle synthesis in a semicircular contraction-expansion array microchannel
Ahmadi et al. Microfluidic devices for gene delivery systems
EP4314238A1 (en) Multistage device and method for intracellular delivery

Legal Events

Date Code Title Description
DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS: AMEND THE INVENTION TITLE TO READ MICROFLUIDIC SYSTEM FOR INTRACELLULAR DELIVERY OF MATERIALS AND METHOD THEREFOR

PC1 Assignment before grant (sect. 113)

Owner name: MXT BIOTECH

Free format text: FORMER APPLICANT(S): KOREA UNIVERSITY RESEARCH AND BUSINESS FOUNDATION

MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted