You are on page 1of 25

www.acsami.

org Review

Surface-Mediated Intracellular Delivery by Physical Membrane


Disruption
Yangcui Qu,§ Yanxia Zhang,§ Qian Yu,* and Hong Chen

Cite This: ACS Appl. Mater. Interfaces 2020, 12, 31054−31078 Read Online

ACCESS Metrics & More Article Recommendations


Downloaded via HONG KONG UNIV SCIENCE TECHLGY on December 2, 2022 at 10:48:23 (UTC).
See https://pubs.acs.org/sharingguidelines for options on how to legitimately share published articles.

ABSTRACT: Effective and nondestructive intracellular delivery of exogenous molecules and


other functional materials into living cells is of importance for diverse biological fundamental
research and therapeutic applications, such as gene editing and cell-based therapies. However,
for most exogenous molecules, the cell plasma membrane is effectively impermeable and thus
remains the greatest barrier to intracellular delivery. In recent years, methods based on surface-
mediated physical membrane disruption have attracted considerable attention. These methods
exploit the physical properties of the surface to transiently increase the membrane permeability
of cells come in contact thereto, thereby facilitating the efficient intracellular delivery of
molecules regardless of molecule or target cell type. In this Review, we focus on recent
progress, particularly over the past decade, on these surface-mediated membrane disruption-
based delivery systems. According to the membrane disruption mechanism, three categories
can be recognized: (i) mechanical penetration, (ii) electroporation, and (iii) photothermal
poration. Each of these is discussed in turn and a brief perspective on future developments in
this promising area is presented.
KEYWORDS: intracellular delivery, surface-mediated gene transfection, membrane disruption, mechanical penetration, electroporation,
photothermal poration

1. INTRODUCTION for recalcitrant cells. Moreover, an inherent shortcoming of all


Intracellular delivery of exogenous molecules and other carrier-based methods is lack of “universality”, that is, a given
functional materials into living cells without compromising carrier may not be suitable for a given molecule because of the
cell viability plays an important role in various biological variability in molecular properties, such as size, charge, and
fundamental research and therapeutic applications, ranging from hydrophobicity.12−15
gene editing, such as clustered regularly interspaced short In contrast to carrier-based methods, membrane disruption-
palindromic repeats/associated protein 9 (CRISPR/Cas9) based methods, and in particular, physical membrane disruption
technology for regulation of cell fate and behavior to ex vivo (PMD)-based methods, deliver the molecules and materials into
cells by membrane penetration or permeabilization via optical,
cell-based therapies, such as the use of modified immune cells for
electrical, mechanical, magnetic, thermal, and acoustic
cancer treatment, and induced pluripotent stem cells for tissue
means.16−18 PMD-based methods are also less dependent on
repair and regeneration.1−4 However, the 5−6 nm thick plasma
the physicochemical properties of the delivered molecule and
membrane that encloses the cell remains an impenetrable barrier
thus are able to deliver almost any exogenous molecule into the
for most exogenous molecules;5 effective and nondestructive
cell. A series of PMD-based intracellular delivery systems
delivery methods are, therefore, urgently needed.
Existing intracellular delivery methods can be divided into two including direct penetration (e.g., microinjection),19 electro-
poration,20 magnetic perforation,21 acoustic perforation,22 and
categories according to the delivery mechanism, that is, carrier-
based methods and membrane disruption-based methods.6−9 In optoporation23 have been developed; two categories can be
distinguished, that is, solution-mediated and surface-mediated.
the former, the molecules to be delivered are packaged with a
Solution-mediated methods rely on physical “agents”, such as
carrier before entering the cell by endocytosis or by fusion with
the cell membrane.10,11 In recent years, various viral and
nonviral carriers have been developed. All of them, however, Received: April 16, 2020
have their own significant limitations. For example, viral carriers Accepted: June 19, 2020
raise concerns regarding cytotoxicity and immunogenicity, while Published: June 19, 2020
nonviral carriers, such as liposomes, cationic polymers, and
inorganic nanoparticles, usually require complex synthetic
procedures and suffer from low delivery efficiency, especially

© 2020 American Chemical Society https://dx.doi.org/10.1021/acsami.0c06978


31054 ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Scheme 1. Schematic Illustration of Three Categories of Surface-Mediated Membrane Disruption-Based Intracellular Delivery
Systems

electric field and laser irradiation applied in solution to generate cell membrane permeability intrinsic mechanisms, as well as
transient holes in the cell membrane for high-efficiency delivery properties and functions of delivered molecules for the details.9
of dispersed molecules.24−27 However, these methods suffer
generally from a lack of control of the process leading to 2. SURFACE-MEDIATED INTRACELLULAR DELIVERY
randomness, poor uniformity, and poor repeatability. In BY MECHANICAL PENETRATION
addition, the required high voltage field or high energy laser
radiation may result in excessive membrane damage causing cell Mechanical penetration is one of the most direct approaches for
death. intracellular delivery by generating localized holes in the cell
In contrast, surface-mediated PMD-based methods depend membrane, through which the exogenous molecules enter the
on the physical properties of the surface to which the target cells cells. Surface-mediated mechanical penetration generally relies
come in contact to increase membrane permeability tran- on the nanoX arrays to penetrate the cell membrane.32−36 In the
siently.9,10 Efficient delivery of the molecule (loaded on the past decade, two possible mechanisms of nanoX arrays for
surface or in solution) in an electric field or under laser intracellular delivery have been proposed. One mechanism is
irradiation to virtually any cell type is thereby facilitated. that nanoX arrays with optimal density and aspect ratio can
Compared with solution-mediated methods, surface-mediated penetrate cell membrane spontaneously, but the penetration is
methods possess the additional merit that the surface can serve limited for some specific cell types.37−42 In 2013, Melosh et al.
as a reservoir for the molecules to be delivered, as well as an found that stiff cells could be penetrated by nanowires more
agent for the localized delivery of molecules to surface-attached easily compared to regular and soft cells.32 Later, they
cells.28 Also, because of the enhancing effect of surface-cell systematically studied the interaction between cells and hollow
interactions, the required voltage or laser energy is relatively low nanowires and explored the influence of the geometry of
thereby decreasing the loss of cell viability. Additionally, besides nanowires, the rigidity of the cell, and the adhesion of the cells
surface-mediated PMD-based methods, there are also some on the interaction between cell and nanowires. Their results
reports about surface-mediated chemical membrane disruption- implied that spontaneous cell penetration by nanoX arrays is
based methods that rely on some chemical promotors, such as possible, yet limited in scope, time scale, and cell types.42
detergents to promote molecular delivery to cells by either Another mechanism is that the nanoX arrays themselves cannot
perturbing cell membrane or facilitating the internalization of penetrate the cell membrane, but they are justly engulfed by cell
nanoneedles.29,30 These methods are covered in other membrane.43−46 This claim was supported by the following
publications elsewhere.18,31 finding that only 7.1% hollow nanowires could spontaneously
In this Review, we mainly focus on recent developments of penetrate cell membrane.47 However, the penetration efficiency
surface-mediated PMD-based methods for intracellular delivery can be increased with the assistance of external force.48,49
of exogenous molecules. These methods are discussed under Although the definite mechanism is still not clear, the nanoX
three headings: (i) mechanical penetration using nanoX arrays arrays have been widely used for intracellular delivery because of
(here X can be wire/needle/pillar/straw of uniform length that the wide range of delivered molecules and target cell types. In
is oriented perpendicular to the substrate) (see section 2); (ii) this section, we will introduce the development of surface-
electroporation using microfluidics, microelectrodes, micro/ mediated mechanical penetration approaches for intracellular
nanopore membranes, or nanostraw arrays (see section 3); and delivery that are divided into two categories based on the
(iii) photothermal poration using surface-immobilized photo- structure of the nanoX arrays: (i) solid nanoX arrays and (ii)
thermal agents such as gold nanomaterials and polydopamine hollow nanostraw arrays (as illustrated in Scheme 2 and
(see section 4) as illustrated in Scheme 1. A brief perspective is summarized in Table 1).
then presented on approaches that show promise for the future. 2.1. Solid NanoX Arrays. 2.1.1. Silicon Nanowire Arrays.
It is noted that our Review does not want to cover all the aspects Silicon nanowire arrays (SiNWAs) refer to arrays of dense
of intracellular delivery by membrane disruption because of the silicon nanowires that are oriented perpendicular to the
scope limit, so interested readers are suggested to read an substrate.83 Including the intrinsic biocompatibility of silicon,
excellent review about the basic concepts, such as cell structures, the high surface-to-volume ratio and unique nanoporous
31055 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Scheme 2. Schematic Illustration of Different Surface- with various chemicals (e.g., cationic polymers,50−53 poly-
Mediated Mechanical Penetration Approaches Based on dopomine,54 or supramolecular nanoparticles55,56) to increase
NanoX Arraysa the loading amount of delivered molecules as well as the contact
between target cells and molecules. For example, we modified
SiNWAs with various cationic polymers, such as branched
polyethylenimine (PEI)50 and poly(N,N-dimethylamino)ethyl
methacrylate (PDMAEMA)51 and aminated poly(glycidyl
methacrylate) (PGMA)52 for loading pDNA and achieved
enhanced transfection of pDNA to HeLa cells. However, there
are still some limitations in these systems. Because these cationic
polymers were firmly immobilized on the surface of SiNWAs,
they cannot provide enough protection and quick release of the
pDNA into cytoplasm, restricting the transfection efficiency. In
addition, the high positive charge carried by these cationic
polymers caused the loss of viability of target cells. To further
a
(a) Intracellular delivery by mechanical penetration using solid improve the transfection efficiency and decrease the cytotox-
nanoX arrays without external force. (b, c) Intracellular delivery by icity, we synthesized a branched PEI cross-linked by disulfide
mechanical penetration using solid nanoX arrays with external force. bonds for modification of SiNWAs to load pDNA.53 When the
(b) The nanoX arrays were placed on the bottom and the target cells modified SiNWAs penetrated the membrane of target cells, the
adhered on the surface of nanoX arrays. (c) The nanoX arrays were disulfide bonds were broken by the high concentration of
placed on the top to penetrate into cell monolayer. (d) Intracellular
glutathione in the cytoplasm, leading to the fast release of PEI
delivery by mechanical penetration using hollow nanostraw arrays,
where the exogenous molecules are stored in bottom fluid storage and preloaded pDNA. The “engineered” cells could be
device. efficiently harvested by trypsin digestion and the SiNWAs
surface could be regenerated by dithiothreitol treatment due to
the replaceable properties of disulfide bonds (Figure 2a).53 It is
structure and of SiNWAs provide them great potential for noted that the process of modification of SiNWAs with polymers
biotechnology and biomedical applications.84 In general, there was relatively complicated. Alternatively, Ito et al. developed a
are two main strategies for fabrication of SiNWAs including the polydopamine (PDA) coating on SiNWAs by a simple
top-down strategy (e.g., metal-assisted chemical etching deposition method (Figure 2b).54 The biocompatible PDA
(MACE) and reactive ion etching (RIE)) and the bottom-up coating could enhance the adhesion, spreading, and growth of
strategy (e.g., chemical vapor deposition (CVD) and laser cells. Moreover, they provided a uniform porous layer on the
ablation).85−87 The SiNWAs discussed in this section are mainly surface of nanowires to increase the area of surface with
fabricated using CVD, MACE, and RIE methods. functional groups for adsorption of siRNA. Compared with flat
In 2007, for the first time, Yang and Conklin et al. silicon surface and unmodified SiNWAs, PDA-coated SiNWAs
demonstrated that SiNWAs containing nanowires with small exhibited more efficient delivery of siRNA to knock down the
diameter and high aspect ratio could deliver plasmid DNA expression of a specific gene via RNA interference.54
(pDNA) to cells.37 The SiNWAs could directly penetrate mouse Codelivery or sequential delivery of diverse genes into one cell
embryonic stem cells (mESCs) without using external forces plays a crucial role in cell reprogramming.1,88 One typical
(Figure 1a and b), and the penetrated cells survived and example is to deliver four genes (Oct4, Sox2, KLF4, and c-MYC
proliferated for up to 3 days. Moreover, their preliminary results (OSKM)) to fibroblasts for preparation of induced pluripotent
indicated that the penetration of SiNWAs was beneficial for gene stem cells (iPSCs).89,90 However, it is still challenging to deliver
transfection using HEK293T cells as model cell lines. However, diverse genes together effectively because of the large volume of
the transfection efficiency was very low (<1%).37 Later, Park et complex of these genes. To solve this problem, Tseng et al.
al. applied SiNWAs for gene-specific manipulation of various developed a new gene delivery system based on adamantine
human and murine immune cells without compromising cell (Ad) modified SiNWAs (Ad-SiNWAs) and supramolecular
viability.39 In particular, they delivered siRNA for gene silencing nanoparticle (SNP) vectors.56 The SNP vectors were synthe-
to study the potential basis of clinical heterogeneity in chronic sized through a modular self-assembled approach from
lymphocytic leukemia.40 In addition, to deeply understand the polyamidoamine dendrimer anchored with Ad groups (Ad-
influence of physical parameters of SiNWAs on intracellular PAMAM), poly(ethylene glycol) anchored with an Ad group
delivery, a series of SiNWAs with different geometrical (Ad-PEG), branched polyethylenimine grafted with β-cyclo-
structures were fabricated and the effects of physical parameters dextrin (CD) grafted (CD-PEI), and delivered pDNA. Because
of SiNWAs on intracellular delivery were examined. As shown in of the host−guest interactions between Ad and CD, as well as
Figure 1c−h, efficient delivery of molecules in smaller electrostatic interactions between PEI and pDNA, the resulted
suspension immune cells (e.g., native mouse B and T cells) SNP vectors (DNA⊂SNPs) could encapsulate and compress
needed the use of SiNWAs in which nanowires were denser four kinds of pDNA to sharply reduce the volume of the complex
(0.3−1 μm−2) and longer (2−3 μm). In contrast, larger and (Figure 3a).55 The DNA⊂SNPs were enriched on the surface of
adherent immune cells (e.g., DCs and MΦs) required the use of Ad-SiNWAs and could be introduced through the cell
SiNWAs in which nanowires were less dense (0.15−0.2 μm−2) membranes to release pDNA inside cytoplasm after dynamic
and slightly shorter (1−2 μm). The SiNWAs with fairly long dissociation. Using this system, direct reprogramming/trans-
nanowires (>3 μm) might cause the nucleus rupture of adherent differentiation of fibroblasts into a stem cell-like state or a
immune cells, leading to severe cytotoxicity even cell death.40 neuronal cell-like state were realized successfully (Figure 3b).56
The delivery efficiency of pristine SiNWAs is generally quite They further extended the application range of this system,
low. To improve the efficiency, several groups modified SiNWAs achieving successful transfection for not only various adherent
31056 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Table 1. Summary of Surface-Mediated Intracellular Delivery by Mechanical Penetrationa


materials delivered molecule cell line delivery efficiency cell viability ref
SiNWAs pDNA HEK293T <1% 37
SiNWAs pDNA/siRNA/protein/peptide HeLa/hFs/NPCs/HNs/DCs 95% 39
B/NK/MΦs/T cells 69−90% without 40
affecting
viability
SiNWAs pDNA HeLa 50
pDNA HeLa 51
siRNA/pDNA HeLa 80%(siRNA to HeLa) 90% 52
pDNA HeLa/SMMCs >60% >90% 53
SiNWAs siRNA A549-luc cells 70−80% 54
SiNWAs pDNA U87/HeLa/NIH3T3/MCF7/BC-1/Ramos/ 70−98% 55
Jurkat/BJ/hDFs/J1/mEFs/hNSCs
pDNA (OSKM/pNTFs) fibroblast cells 30−50% 56
SiNWAs pDNA hDPSCs 77% 57
SiNWAs pDNA HEK293/HeLa/hDPSCs/hFFs >85% (hDPSCs and >90% 58
HEK 293)
silicon pillar arrays pDNA hFFs/DPSCs 75% 59
silicon nanoneedle dextran DU145/HeLa 50% (calcein to 90% 60
arrays HeLa)
silicon nanoneedle pDNA NIH3T3 34% (pDNA) 42% 61
arrays (Cre recombinase)
silicon nanoneedle siRNA/pDNA/VEGF-165 gene HeLa 90% 41
arrays
silicon nanoneedle FITC-dextran/pYFP NIH3T3 45% (FITC-dextran, 62
arrays 70 kDa)
silicon nanoneedle quantum dots HeLa/skin/muscle 63
arrays
silicon microneedle PI HeLa 229 97.9% 97.7% 64
arrays
diamond luminescent iridium polypyridine A549 92% 65
nanoneedle arrays EthD1/Dextran/antibody/pDNA/ NIH3T3/neurons 45% (pGFP to 90% 48
quantum dots/polystyrene nanoparticles neurons)
carbon nanofiber pDNA CHO 66,
arrays 67
carbon nanofiber shRNA/pDNA CHO 53.1% (shRNA) 68
arrays
carbon nanofiber pDNA V97 69
arrays
carbon nanofiber pDNA V97 38
arrays
carbon nanosyringe quantum dots/pGFP NIH3T3/MSCs 34% (pGFP to 85% 70
arrays NIH3T3)
30% (pGFP to
MSCs)
carbon nanosyringe gene: VEGF-mRNA/pGFP HeLa/PC3/MDA-MB-231/MCF7/ 46% (pGFP to 71
arrays lymphocyte)
proteins: GFP/RFP/caspase-3 T47D/HepG2/primary lymphocyte 70.3% (GFP to
T47D)
79% (RFP to HepG2)
ZnO nanowire Hyg3/peptide/pDNA MCF7/Microalgal 6.52 × 10−3 49,
arrays 72
ZnO nanowire pGFP HEK293 60% 73
arrays 8% (pGFP)
CuO nanowires pDNA HeLa <1% without 74
affecting
viability
hollow nanoneedle dextran/pRFP HEK293 70−90% 29
arrays
hollow carbon PI/dextran/quantum dots/pYFP HEK293/L6 99% for dye delivery 97% 75
nanostraw arrays 84% for pDNA
transfection
hollow tapered quantum dots microalgae 9.8% 76
microtube arrays
Al2O3 nanostraw fluorescence dye/Co2+/EDTA/pGFP CHO 70% (Co2+) 77
arrays 75% (EDTA)
10% (pGFP)
Al2O3 nanostraw Ca2+ indicator CHO 90% 78
arrays

31057 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Table 1. continued
materials delivered molecule cell line delivery efficiency cell viability ref
Al2O3 nanostraw Co2+/EDTA CHO 95% 47
arrays
Al2O3 nanostraw functional probes of glycolysylation CHO near 100% 79
arrays
Al2O3 nanostraw pRFP CHO 1.5% near 100% 80
arrays
hierarchical spiky staurosporine MCF7 84.6% 94.5% 81
microstraw arrays
Au nanostraw arrays calcein NIH3T3 85% 90% 82
a
Abbreviation: Silicon nanowire arrays (SiNWAs); Oct4, Sox2, KLF4, and c-MYC (OSKM, a pDNA encoding reprogramming factors, 13 kbp);
Ascl1, Brn2, Myt1l, and NeuroD1 (pNTFs, a pDNA encoding neuron specific transcription factors); pDNA encoding red/yellow/green
fluorescence protein (pRFP/pYFP/pGFP); green/red fluorescence protein (GFP, 26 kDa/RFP); ZnO-binding peptide (ZBP) conjugated with
FITC (ZBP-FITC); propidium iodide (PI, 0.66 kDa); vascular endothelial growth factor (VEGF); ethylenediamine-tetra-acetic acid (EDTA);
dextran (3 kDa, 70 kDa, 500 kDa); pDNA (5.2 kbp, 6 kbp); quantum dots (15−20 nm in diameter); polystyrene nanoparticles (200 nm in
diameter); human embryonic kidney cells (HEK293); human fibroblasts (hFs); rat neural progenitor cells (NPCs); hippocampal neurons (HNs);
dendritic cells (DCs); nature killer (NK) cells; microphages (MΦs); smooth muscle cells (SMMCs); breast cancer-1 cell (BC-1); human dermal
fibroblast (hDFs); mouse embryo fibroblast (mEFs); human neural stem cells (hNSCs); human dental pulp stem cells (hDPSCs); human foreskin
fibroblasts (hFFs); Chinese Hamster Ovary (CHO); Chinese hamster lung cells (V79); mesenchymal stem cells (MSCs).

Figure 1. Intracellular delivery by membrane penetration using SiNWAs fabricated via chemical vapor deposition. (a) Typical confocal microscopy
image and (b) a typical scanning electronic microscopy (SEM) image of mESCs penetrated with silicon nanowires. Adapted with permission from ref
37. Copyright 2007 American Chemical Society. (c−h) Representative SEM images of (c) mouse bone-marrow DCs, (d) B cells, (e) DCs, (f) MΦs,
(g) NK cells, and (h) T cells on top of SiNWAs with different geometric structures. Adapted with permission from ref 40. Copyright 2012 American
Chemical Society.

cells (HeLa, U87, MCF3, NIH3T3, and HEK293) but also 2.1.2. Other Solid NanoX Arrays. In addition to chemical
recalcitrant suspension cells (BC-1, Ramos, and Jurkat cells) and modification to increase the loading amount of delivered
primary cells (BJ, hDFs, J1, mEFs, and hNSCs). In particular, molecules, introduction of external force (e.g., centrifugal force
sequential delivery of three pDNA encoding three different and squeeze force) to nanoX arrays can also enhance the delivery
proteins (pDNA encoding green fluorescence protein (pGFP), efficiency by increasing the extent of membrane penetration. In
mCherry (pmCherry), and E2-Crimson (pE2-Crimson)) to general, there are two classic models for applying external force
U87 cells was achieved by adding three respective SNP vectors on nanoX arrays. The first one is to culture cells on the top of the
with encapsulated pDNA 12, 24, and 36 h after the cell nanoX arrays and then to apply a top-down centrifugal force or
settlement on Ad-SiNWAs. In addition, Ad-SiNWAs were impact force on the adhered cells to promote deep penetration
of nanoX (as illustrated in Scheme 2b).49,65,71,72 For example,
subcutaneously transplanted into one side of the mouse body,
with the assistance of centrifugal force, successful transfection of
followed by local injection of SNPs encapsulated with plasmid
pDNA was achieved not only for easy-to-transfect HepG2 cells
encoding luciferase reporter (pGL3⊂SNPs). The expression of (more than 70% transfection efficiency) but also for hard-to-
luciferase in site with Ad-SiNWAs was significantly increased transfect lymphocytes (about 46% transfection efficiency) using
compared to that detected in site without Ad-SiNWAs or in site carbon nanosyringe arrays (CNSAs), expanding the application
with smooth silicon surface modified with Ad, showing the range of cell types and improving the transfection efficiency
application potential of this system for in vivo gene delivery (Figure 4).71 The second one is to place nanoX arrays on the top
(Figure 3c).55 However, it should be noted that this system has a of cells to squeeze the cells monolayer, promoting the
drawback regard to the hard harvest of “engineered” cell from penetration of nanoX for the entry of exogenous molecules
the surface of SiNWAs, which might limit its ex vivo into cells (as illustrated in Scheme 2c).48,61,62 For example, by
applications. applying external squeeze force on diamond nanoneedle arrays,
31058 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Figure 2. (a) Scheme of intracellular delivery of DNA and regeneration of SiNWAs modified with a branched PEI cross-linked by disulfide bonds.
PEICBA represents a branched PEI cross-linked by disulfide bonds that are sensitive to glutathione. Adapted with permission from ref 53. Copyright
2019 American Chemical Society. (b) Schematic illustration of preparation of PDA-coated SiNWAs for delivery of siRNA into cells. siRNA could be
easily bound to the surface of PDA-coated SiNWAs through the electrostatic interaction. Adapted with permission from ref 54. Copyright 2016
American Chemical Society.

various exogenous molecules including fluorescent dyes, layers: a glass manifold layer associated with a pump (top layer),
dextran, antibodies, quantum dots, nanoparticles, and pGFP a monolithic polydimethylsiloxane (PDMS) membrane (inter-
were delivered to NIH3T3 cells and hard-to-transfect primary layer), and a microchannel layer with ZnO nanowire arrays
neuron cells effectively. The transfection efficiency of pDNA to (bottom layer). Among them, the top layer and interlayer
neurons was improved significantly from ∼1−5% by using together served as a pneumatic microvalve (Figure 5a). Once the
lipofection approach to ∼45% by using this system. Moreover, target cells were loaded in microchannel, the PDMS membrane
after delivery process, the diamond nanoneedle arrays could be was properly bent down by supplying air through the pump, and
removed from the cells to return back the normal cell culture cells were squeezed onto ZnO nanowire arrays preloaded with
conditions.48 delivered molecules. After delivery, the PDMS membrane was
Carbon nanofiber arrays (CNFAs) are also a kind of nanoX bent upward by supplying vacuum through the pump, thus the
arrays for intracellular delivery. In 2003, McKnight et al. “engineered” cells were harvested due to reverse-directional
demonstrated that penetration of CNFAs into living cells could pressure (Figure 5b).49
provide efficient delivery and expression of exogenous genes. 2.2. Hollow Nanostraw Arrays. Although solid nanoX
They prepared a tapered spiked CNFAs with a small tip arrays are promising for intracellular delivery, they are still
diameter of 20−50 nm for insertion directly into cells and a wide restricted by some limitations. For example, as the delivery
base diameter of 1 μm for providing mechanical strength. The process goes on, the molecules loaded on solid nanoX arrays
pDNA loaded on the CNFAs by physical adsorption or covalent gradually taper off, causing a lack of continuous supply of
interaction was delivered in CHO cells by penetration of CNFAs delivered molecules. In addition, it remains challenging to
alone or in combination with centrifugal force or pressing control the dosage of delivered molecules precisely, and it is
force.66,67 In the following work, the CNFAs were coated with a cumbersome to codeliver or sequential deliver diverse molecules
thin gold layer as an anchor for immobilization of pDNA into the same cell repeatedly. To this end, nanostraw arrays
containing thiol or amino groups via covalent bonds. Compared consisting of a dense array of hollow nanotubes have been
with unmodified CNFAs, the gold-coated CNFAs exhibited developed.92−95 The bottom of nanostraw arrays is connected to
increased loading capacity of pDNA, thus enhancing trans- a fluid storage device or a microfluidic channel, thus the cells
fection efficiency of pDNA encoding yellow fluorescent protein adhered on the top of the nanostraw arrays can be continuously
(pYFP) to V97 cells.69 To better release the pDNA on the and repeatedly delivered with the same or different molecules
CNFAs, they further optimized this platform by binding DNA through the bottom device/microchannel (as illustrated in
via disulfide bonds. When the nanofibers penetrated into the Scheme 2d).77,96,97 In addition, nanostraw arrays can achieve
cells, the pDNA was released effectively because of the breakage precisely control of delivery dosage by modulating the
of disulfide bonds by glutathione in cytoplasm, further composition of external fluids. More importantly, codelivery
improving the transfection efficiency.38 or sequential delivery of diverse molecules to the same cell can
Although the above-mentioned systems realize effective be realized simply by adding or changing molecules in the fluid
intracellular delivery, one main drawback of most nanoX- storage/microchannel without repeated penetration of cell
array-based systems is the insufficient cell harvest capability, membrane, those together solving the above-mentioned
limiting their broad applications in ex vivo cell-based therapy.3,91 problems related to the solid nanoX arrays.
In some reports, the “engineered” cells were harvested by trypsin In 2012, Sivan et al. developed silicon nanostraw arrays that
digestion but the viability might be compromised. Aiming at this were established continuous fluidic access with a fluidic storage
limitation, Seo and Park et al. developed a microdevice device. Multiple delivery of fluorescently labeled dextran to the
integrated with “intracellular delivery” and “cell harvest” same HEK293 cells was achieved. Especially, only the cells
functions together. The microdevice was composed of three connecting with nanostraws expressed red fluorescence when
31059 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Figure 3. (a) Schematic illustration of preparation of DNA⊂SNPs and the interaction between Ad-SiNWAs and DNA⊂SNPs. The host−guest
interaction between the Ad motifs on Ad-SiNWAs and the β-cyclodextrin (CD) motifs of DNA⊂SNPs leads to dynamic assembly and local
enrichment of DNA⊂SNPs onto Ad-SiNWAs. Reproduced with permission from ref 55. Copyright 2014 American Chemical Society. (b) Schematic
illustration of intracellular delivery of OSKM encoding reprogramming factors and pNTFs encoding neuron specific transcription factors to fibroblasts
for cell reprogramming and cell transdifferentiation using Ad-SiNWAs, respectively. Reproduced with permission from ref 56. Copyright 2015 John
Wiley and Sons. (c) Schematic illustration of gene delivery in vivo using Ad-SiNWAs. Reproduced with permission from ref 55. Copyright 2014
American Chemical Society.

31060 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Figure 4. Schematic illustration of intracellular delivery using CNSAs


combined with centrifugal force, where the CNSAs was placed on the
bottom and the target cells adhered on the surface of CNSAs. Adapted
with permission from ref 71. Copyright 2016 John Wiley and Sons.

Figure 6. (a) Schematic illustration of a cross-section view of a


representative nanostraw arrays-incorporated device for intracellular
delivery. (b, c) Representative SEM images of cells cultured on
nanostraw arrays with diameter of 100 nm, height of 1−2 μm and
density of 108 nanostraws/cm2. (d−f) Typical fluorescence images of
sequential delivery of two different dyes: (d) Alexa-Fluor 488-hydrazide
dye (green color), (e) Alexa-Fluor 568-hydrazide dye (red color), and
Figure 5. Schematic illustration of (a) fabrication of the ZnO nanowire (f) a merged image indicating colocalization of dyes to CHO cells using
array-incorporated microdevice. The microdevice was composed of nanostraw arrays. Adapted with permission from ref 77. Copyright 2012
three layers: a glass manifold layer associated with a pump (top layer), a American Chemical Society.
monolithic polydimethylsiloxane (PDMS) membrane (interlayer), and
a microchannel layer with ZnO nanowire arrays (bottom layer). (b)
Schematic illustration of the functions of “intracellular molecular cells on nanostraw arrays surface were delivered with Co2+ and
beacon delivery” and “cell harvest” of this microdevice, where the ethylenediamine-tetra-acetic acid (EDTA) for quenching and
“engineered” cells were harvested due to reverse-directional pressure. dequenching GFP fluorescence, respectively. Introduction of
Adapted with permission from ref 49. Copyright 2015 John Wiley and Co2+ in the microfluidic channel for one minute led to significant
Sons. reduction in fluorescence of the cells adhered on nanostraws,
while no changes in fluorescence of the cells existed in the
transfected plasmid encoding red fluorescence protein (pRFP), culture medium were observed. Subsequent delivery of EDTA
confirming the potential for spatial control of gene trans- enabled 75% of quenched cells to recover fluorescence after 4
fection.29 In another work, Melosh et al. developed Al2O3 min. This fluorescent “blinking” demonstrated that it is possible
nanostraw arrays that established continuous fluidic access to directly manipulate intracellular contents using nanostraw
into cytoplasm for the delivery of genetic materials, proteins, and arrays with precise spatiotemporal control.77
small molecules (Figure 6a). The nanostraws could sponta- The delivery of exogenous molecules into various cell types
neously penetrate the cell membrane (Figure 6b and c), may require nanostraw arrays with different geometry, hence a
providing permanent pipelines between exogenous molecules simple method for preparation of nanostraw arrays with different
stored in fluidic chamber/microfluidic channel underneath the diameters, lengths, and densities is needed. Conventionally,
nanostraw arrays and interior of cells adhered on surface of nanostraw arrays are prepared using nanoporous polycarbonate
nanostraw arrays.77 Furthermore, they delivered second (PC) membranes as starting templates.47,78,79,98 However,
membrane-impermeable dyes (red color) 12 h after delivering nanoporous PC membranes with custom-designed diameters
the first membrane-impermeable dyes (green color) to the cells. and densities are often expensive or commercially unavailable.
As shown in Figure 6d−f, the two dyes colocalized in most of the Regarding this issue, Xie et al. introduced a novel method
cells, confirming that the nanostraw−cytosol connection applying O2 plasma to fabricate a series of membrane template
remained open over extended periods after initial penetration. to prepare nanostraw arrays, in which the structural parameters
In addition, green fluorescence protein (GFP)-expressing CHO of nanostraws could be adjusted simply by controlling the
31061 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Figure 7. Schematic illustration of (a) a microfluidic device integrated with anti-EpCAM coated HS-MSAs for capture of cancer cells and intracellular
delivery, the illustration of conjugating anti-EpCAM biomolecules onto surface of HS-MSAs by biotin−streptavidin interaction was shown in right
image. (b) Fabrication process of HS-MSAs combining sequential atomic layer deposition, mechanical polishing, O2 etching, hydrothermal growth,
and Al2O3 deposition processes. (c) Integration of HS-MSAs with a microfluidic device. Reproduced with permission from ref 81. Copyright 2019
John Wiley and Sons.

etching power and etching time.80 Furthermore, they applied surrounding environment are effectively delivered into cells
Al2O3 nanostraw arrays with differing structures alone or in through electrophoresis or promotion of diffusion effect.20,99 In
combination with external force for delivery of pRFP to CHO general, electroporation can be classically grouped into three
cells. When using nanostraw arrays alone, only the nanostraws categories: bulk electroporation (BEP), microelectroporation
with small diameter and length could transfect pRFP to CHO (MEP), and nanoelectroporation (NEP).20 BEP is generally a
cells but with an extremely low efficiency (1.5%). In contrast, the kind of solution-mediated electroporation, and MEP/NEP
transfection efficiency was significantly enhanced with the includes both solution-mediated electroporation and surface-
assistance of squeezing force to promote the membrane mediated electroporation. For the purposes of this Review, we
penetration.80 In the following work, they fabricated a only discuss those techniques that involve using a surface to
hierarchical spiky microstraw arrays (HS-MSAs) by hydro- focus the electric field locally on the cell membrane. Although
thermal growth method to increase the surface-to-volume ratio. BEP has been widely applied as a commercial technique for gene
After conjugated with specific antibodies, antiepithelial cell transfection, it still has some concerns and limitations. The
adhesion molecules (anti-EpCAM), the HS-MSAs could traditional BEP requires a high voltage (>1000 V) for effective
capture cancer cells efficiently and selectively because of the transfection, which however, always lead to severe cell damage.
synergistic effect of hierarchical structures of HS-MSAs and the Moreover, the typical BEP usually treats plenty of cells at the
specific recognition of anti-EpCAM (Figure 7a-b). Delivery of same time, and it is difficult for electric field to be evenly
staurosporine to the captured cancer cells was realized with distributed at the single cell level, resulting in randomness of cell
precise dose and spatiotemporal control using the HS-MSAs in transfection.24,100,101 Alternatively, surface-mediated electro-
combination with microfluidic device (Figure 7c), following poration can enhance the local electric field strength around cells
with detection of the cancer cells in situ and promotion of cell because of contact between cells and surfaces, reducing the total
apoptosis. Because the capture, delivery and detection of cells applied voltage to alleviate cell damage during delivery
were realized in a single device, side effects related to releasing process.102−105 In this section, we will introduce the develop-
captured cells were avoided, which were inevitable for traditional
ment of surface-mediated electroporation approaches for
cancer cells detection methods.81 In addition to the above-
intracellular delivery that are divided into four categories
mentioned Al2O3 nanostraw arrays, carbon nanostraw arrays,75
based on the devices used: (i) microfluidics (Scheme 3a), (ii)
and Au nanostraw arrays82 were also developed as intracellular
microelectrodes (Scheme 3b), (iii) micro/nanopore mem-
molecular delivery platforms.
branes (Scheme 3c), and (iv) nanostraw arrays (Scheme 3d) as
summarized in Table 2.
3. SURFACE-MEDIATED INTRACELLULAR DELIVERY 3.1. Microfluidics. Surface-mediated microfluidic electro-
BY ELECTROPORATION poration is a method for delivery of molecules to the cells by
In addition to mechanical penetration, other external forces, applying a certain voltage at both ends or both sides of the
such as electric field, can also be used for intracellular delivery.26 microfluidic channel after cell attachment.133,134 Lu et al.
Under a certain electric field, a potential difference across the cell developed a microfluidic device integrated with multifunctions
membrane forms. When the potential difference reaches a including intracellular protein delivery, cell culture, and
critical magnitude of voltage, the formation of transient pores in fluorescence imaging (Figure 8a). Effective delivery of a
cell membrane occurs, thus exogenous cargoes in the fluorescence resonance energy transfer (FRET) biosensor in
31062 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Scheme 3. Schematic Illustration of Different Surface- with precisely controlled dosage (Figure 8d). Moreover, this
Mediated Electroporation Approachesa system enabled on-chip optimization of electroporation
parameters for cells with changing properties.111 It should be
noted that most of the microfluidic electroporation systems
mentioned above usually handle a small amount of target cells
(less than 100) at one time, which is more suitable for single-cell
research but not for the clinical applications where a large
amount of cells are required.
3.2. Microelectrodes. In addition to microfluidics, electro-
poration based on microelectrodes has also been developed for
surface-mediated intracellular delivery, where the targeted cells
come in contact to surface of microelectrodes that produce
localized electric field. For example, Lin et al. developed an
electroporation-based system including interdigitated electrodes
at bottom and a flat plate electrode on top. Because of the
a
(a) Microfluidics electroporation, (b) microelectrodes electro-
electrostatic force generated between the two electrodes, the
poration, (c) micro/nanopores membrane electroporation, and (d) negatively charged pDNA in the solution accumulated on the
nanostraw arrays electroporation. surface of interdigitated electrodes, facilitating their delivery to
the cells come in contact on the interdigitated electrodes under
electric field.114 This interdigitated electrodes was fabricated by
its protein form to mEFs was achieved by applying a voltage traditional microfabrication technology that was time-consum-
across the microchannel. The FRET signal was then recorded in ing and required special equipment. To solve this problem, Xing
situ to reflect the intracellular molecular activity with high et al. fabricated a 3D interdigitated electrode microchip using a
spatiotemporal resolution.106 Moreover, they also used this simpler and more economical standard printed circuit board
microfluidic electroporation system to deliver quantum dots to technology, using which to deliver pDNA into suspension Jurkat
cells for live-cell imaging and cell-molecule tracking.107 cells.115 Liang et al. developed a novel electroporation microchip
However, this electroporation system is unable to control the that was compatible with the standard multiwell plates used in
dose of delivered molecules precisely. Lee et al. developed a biological research (Figure 9a). Tens to hundreds of cells were
microfluidic device containing a unique microchannel-nano- loaded on surface of the novel annular interdigitated electrode
channel-microchannel structure by a DNA combing and underneath the microchip and were transfected with pDNA with
imprinting (DCI) method,135 achieving delivery of precise 90% of transfection efficiency and 80% of cell viability. The
amounts of molecules into cells without compromising cell results of numerical simulations proved that the application of a
viability.108 The target cells and delivered molecules were voltage of 150 V was enough to generate a uniform electric field,
located in two microchannels separately. When an electrical which was significantly lower than the voltage used in traditional
pulse was delivered between the two microchannels, a large BEP.112 However, random transfection is inevitable in the
electric field was generated in the nanochannel to drive the process of microelectrode electroporation. To overcome this
molecules through the cell membrane adjacent to the nano- limitation, instead of a single microelectrode, microelectrode
channel by electrophoresis. The dose of delivered molecules arrays consisting of conductive nanospikes with uniform
could be precisely controlled by modulating the duration and dimensions were developed for electroporation (Figure 9b).
number of pulses. However, the yield of channels prepared using Because of the high aspect ratio, these conductive nanospikes
manual DCI method was relatively low. In addition, the brittle could generate a highly concentrated electric field under a low
resin printed on the channel surface often formed small voltage (<5 V) to deliver molecules to single cell or multiple cells
microcracks, which might lead to leakage within the device to with high delivery efficiency and good cell viability.117
reduce the service life. To this end, they improved the DCI 3.3. Micro/Nanopore Membranes. Precise intracellular
process by replacing manual peeling method with dip-combing delivery of molecules with well-defined amounts is crucial for a
and using a softer resin, increasing the preparation speed and variety of biological researches and practical applications, such
reducing the product defects. Moreover, they shortened the as cell reprogramming and gene therapy.3,88,91 To this end,
length of microchannel from 3000 to 50 μm, reducing the micro/nanopore membranes with multiple pores were devel-
obstacles for cells to move from the microchannel to the oped for electroporation, in which only the small areas of cell
nanochannel to improve cell loading efficiency.109 membrane adjacent to the pores were exposed to a high localized
Intracellular delivery to rare cells, such as MSCs, is crucial for electric field intensity. Therefore, delivered molecules are
both fundamental research and patient-derived-patient-used electrophoretically driven into the cytoplasm directly during
transplantations.136 Valero et al. developed a microfluidic device electroporation and precise dosage control can be achieved
containing two channels, which are connected by microholes for without compromising cell viability.
trapping living cells (Figure 8b and c). Because of the pressure Lee et al. developed a series of sandwich-structured
difference between the two channels, the target cells were electroporation systems based on micropore membranes.118,119
trapped to the microholes. The enhanced local electric field led The system was composed of two poly(ethylene terephthalate)
to delivery of pDNA to MSCs with transfection efficiency of 75% (PET) nuclear pore membranes with an average pore diameter
and cell viability of about 100% at a low voltage (<4 V).110 In of 0.4 and 3 μm as the bottom layer and top layer, respectively,
another work, Hur et al. developed a robust, efficient, and between which were the target cells. The transfected pDNA was
versatile on-chip vortex-assisted microfluidic electroporation added from the top membrane, enriched on or around the
system, enabling simultaneous enrichment of target cells and exposed cell membrane by electrophoresis under low voltage,
sequential delivery of diverse molecules into different cell types and then delivered to the cytoplasm under a relatively large
31063 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Table 2. Summary of Surface-Mediated Intracellular Delivery by Electroporationa


electric field
style parameters delivered molecules cell type delivery efficiency cell viability ref
microfluidics 800 V/cm; 15 ms FRET mEFs 55% 106
microfluidics 400 V/cm; 15 ms quantum dots CHO 60 quantum dots/cell 71% 107
microchannel-nano- 220 V/2 mm ODN-Cy3/siRNA/GAPDH- Jurkat/K562 12% (ODN-Cy3 to Jurkat) 108
channel-microchan- MB
nel
microchannel-nano- pGFP/OSKM mEFs 90% 109
channel-microchan-
nel
microfluidics <4 V PI/pDNA C2C12/MSCs 75% (pDNA to C2C12) 100% 110
65% (pDNAto MSCs)
microfluidics <20 V proteins/siRNA/miRNA HEK293/MCF7 26%-71% (single protein to HEK293) 111
microelectrodes 40−150 V; siRNA/pDNA HEK293/HUVECs/ 90% 80% 112,
0.1−1 ms MDCK/HL-60 113
microelectrodes 150−424 V/cm; pDNA HEK293/HepG2/HU- 114
20 ms VECs/MC3T3 × 10−1
microelectrodes 60−70 V pEGFP-N1 HeLa/MCF7/COS7/Ju- 75% (HeLa)/65% (MCF7)/75% >60% 115
rkat/3T3-L1 (COS7)/55% (Jurkat)/40%
(3T3-L1)
microelectrode arrays 66−82 V calcein/FITC-BSA/pGFP hPCs 50% (calcein) >80% 116
microelectrode arrays 2V PI/EB HeLa >93% >93% 117
micropore membranes 35 V/cm; 500 ms pEGFP/pSEAP NIH3T3 90% 118
micropore membranes 150 V/cm pmaxGFP/gWiz SEAP mESCs 75% 119
micropore membranes 4−10 V; 10 ms GATA2 molecular beacon leukemia cells 83.4% 89−96% 120
micropore membranes 150 V/cm pSEAP mESCs 85% 121
micropore membranes 3−15 V lucifer yellow/pDNA HeLa >80% 122,
123
micropore membranes 20 V PI/pGFP HeLa/HT1080/neurons 95% (PI to HeLa) >90% 124
50% (pGFP to HT1080)
micropore membranes 25 V; 20 ms oligonucleotides/dacarba- myofibroblasts/Human >90% 95.6% 125
zine/CRISPR-Cas9 plas- A375 melanoma cells
mids
nanopore membranes 10−15 V; 5 ms PI/BSA/pmCherry HT1080/MDA-MB 231 95% (PI to HT 1080) 95% (PI to 126
cells 70% (pmCherry) HT 1080)
nanopore membranes 100−140 V FAM-ODN/PI/pGFP/ H9C2 cells/mEFs 10% (OSKM to mEFs) >90% 127
OSKM
nanopore membranes 20 V/30 V mCherry RNA/pGFP/Cas9 HeLa/HEK293/3T3/Ju- >80% (HeLa) 95% 128
RNP rkat 75% (RNA to Jurkat)
50% (pGFP to Jurkat)
nanostraw arrays 30 V; 500 μs pRFP CHO 57.6% 90% 80
nanostraw arrays 5−20 V; 20−200 PI/pRFP/pGFP CHO/HEK293T 67−95% >98% 129
μs
nanostraw arrays 20 V; 20 s GFP/mCherry-mRNA HEK293T/hiPSC-CMs/ 60−90% >90% 130
ESCs/hFs/mGs/mNs
nanostraw arrays 15 V; 200 μs PI/pDNA MCF-7 64.2% (pDNA) >95% 131
83.8% (PI)
nanostraw arrays 5 V; 200 μs PI HeLa 80% >95% 132
a
Abbreviation: fluorescence resonance energy transfer (FRET); 18-mer oligodeoxynucleotide conjugated with Cy3 (ODN-Cy3); GAPDH
molecular beacon (GAPDH-MB); Ethidium bromide (EB); plasmid gWiz GFP (5757 bp) (pEGFP); plasmid secreted alkaline phosphatase
(pSEAP, 6569 bp); FAM labeled oligonucleotides (FAM-ODN); Cas9 ribonucleoproteins (Cas9 RNP); fluorescein isothiocyanate-labeled bovine
serum albumin (FITC-BSA); bovine serum albumin (BSA); quantum dots (15−20 nm in diameter); oligonucleotides (molecular beacons, 22 bp),
pDNA (CRISPR-Cas9 expression vectors, > 9 kbp); OSKM (13 kbp); GFP/mCherry-mRNA (∼922 nt); human umbilical vein endothelial cells
(HUVECs); human prostate cancer cells (hPC); mouse embryonic stem cells (mESCs); human iPSC-derived cardiomyocytes (hiPSC-CMs);
mouse primary glia cells (mGs); mouse primary neuron cells (mNs).

voltage. In this process, cells come in contact to the underlying when they are removed from culture plates for BEP.137 To
membrane by applying a vacuum force, which might cause overcome these challenges, Lee et al. presented a nanofiber-
certain damages to the cells. Alternatively, Sooryakumar and Lee based sandwich electroporation device, which could deliver
et al. combined cells and magnetic microspheres and applied low nucleic acid to ESCs in culture without removing them from the
magnetic field to bind the cells on the micropore membrane, culture substrates. As shown in Figure 10a, a nanofibrous
alleviating the cell damage. Using this system, they delivered electrospun nanofiber of poly(ε-caprolactone)/gelatin was used
signal molecules (GATA2 molecular beacon) to leukemia cells to provide optimum conditions for cell culture, and a
with a high throughput of 40 000 cells/cm2.120 Genetically nanoporous membrane was placed on the top of target cells.
modified primary ESCs have been widely used for biological Under a highly focused electric field, plasmids were delivered
research, which are commonly prepared via BEP in suspension. through the nanopores to ESCs with high efficiency. Moreover, a
However, most ESCs suffer the problems to reform colonies spacer could be integrated to this system for modulation the
31064 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Figure 8. (a) Setup of microfluidic electroporation for delivery of FRET biosensor to cells integrating electroporation delivery, cell culture, and
fluorescence imaging, where the microfluidic channel was coated with fibronectin to facilitate cell adhesion and culture. Reproduced with permission
from ref 106. Copyright 2014 The Royal Society of Chemistry. (b, c) Setup for electroporation-based molecular delivery to 9 cells using a microfluidic
device including 9 traps: (b) 3D scheme of trapped cells and (c) microfluidic chip layout. Reproduced with permission from ref 110. Copyright 2008
The Royal Society of Chemistry. (d) Schematic illustration of the structure, operational principle of a vortex-assisted electroporation system and its
applications on sequential delivery, gene knockdown and phenotype alteration. Reproduced with permission from ref 111. Copyright 2017 Springer
Nature.

shape and size of the cell colonies to enhance the focused electric micro/nanopore membrane electroporation system capable of
field on cells.121 delivering molecules to adherent cells as well as to suspension
In most sandwich-structured micropore membranes electro- cells. Recently, Yang et al. presented a novel nanopore
poration systems, the target cells come in contact to the bottom membrane device consisting of two flat titanium electrodes
membrane, leaving a gap between cells and the top nanoporous and a PDMS holder with a track-etched PC nanoporous
membrane, through which the delivered molecules were added. membrane. To improve cell adhesion property, the membrane
Therefore, it is difficult to deliver the same or different molecules was precoated with fibronectin or poly-L-lysine. Diverse
to one cell repeatedly. To solve this limitation, a series of molecules including nucleic acids, functional proteins, and
electroporation systems were developed by integration of Cas9 ribonucleoproteins were delivered to various cell types
nanoporous aluminum membranes or nanoporous polycarbon- including adherent cells (HeLa cells) and suspension cells
ate membranes with an underlying PDMS chamber as a (Jurkat cells) using this device (Figure 11). The permeability of
molecule storage device. In these systems, the molecules could a small area of the cell membrane could be enhanced under low
be delivered continuously into the same cells through the electric field due to the intimate contact between cells and
nanopores.122−124,126 For example, Lee et al. fabricated a 3D nanopores, making it easier for entry of exogenous molecules to
nanopore membrane electroporation chip with uniform and cells through electrophoresis. Especially, the integrity of the cells
parallel nanopores (650 nm for pore diameter) by combining was retained well. In addition, this system did not require
projection photolithography and deep reactive ion-etching complicated fabrication processes, expensive materials, speci-
(Figure 10b). When delivering cargo with large molecular alized buffers, or cell manipulation, providing a flexible and
weight (13 kbp transcriptional plasmid), this system showed effective method for intracellular delivery.128
significant higher efficiency as compared to the BEP system 3.4. Nanostraw Arrays. Nanostraw arrays electroporation
(∼10% vs <1%).127 is a novel electroporation method, in which a flat electrode is
Many cell types, such as lymphocytes and circulating tumor located at the bottom of nanostraw arrays and the other
cells, exist in suspension. Successful delivery of functional electrode is inserted into the upper cell medium. The unique
molecules to such suspension cells is of considerable importance structure of nanostraws provide localized electric field to
in immune therapy and cancer diagnostics,138,139 however, is less enhance the membrane permeability transiently only over a
investigated but more challenging compared with the delivery to small area. More importantly, unlike other electroporation
adherent cells. One main limitation of the above-mentioned systems in which the delivered molecules are randomly
systems is that they are not suitable for delivery to suspension suspended in the fresh opened pores, in nanostraw arrays the
cells because of the low adhesion and insufficient duration of molecules are directly drew into the cells, thereby improving the
cell-surface contact.140 Therefore, it is crucial to develop a speed and accuracy of delivery process.132
31065 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Figure 9. (a) Multiple-well microelectrodes electroporation device consisting of multiple microchips. Left: Impression of microchip consisting of two
juxtaposed chrome and gold layers sputtered on a glass substrate. Scale bar: 5 μm. Reproduced with permission from ref 112. Copyright 2011 The
Royal Society of Chemistry. (b) Schematic illustration of an electroporation chip containing nanospike arrays. The electric field at nanospike arrays was
enhanced due to its high aspect ratio. Reproduced with permission from ref 117. Copyright 2017 Elsevier.

Xie et al. developed an Al2O3 nanostraw arrays electro- arrays were biocompatible and could serve as a plat electrode at
poration system to deliver exogenous molecules (propidium the bottom without setting up another plat electrode. As a
iodide (PI) and pRFP) to HEK293T cells with high delivery demonstration, Pt nanostraw arrays were integrated with a low-
efficiency (95% for PI delivery, 81% for pRFP transfection) and voltage nanoelectroporation system for delivery of drug and
98% cell viability (Figure 12a).129 The nanostraws with small sensing of intracellular enzymes (Figure 13).132 Effective
diameter (250 nm) were unable to penetrate the cell membrane, delivery of molecules in living cells with spatial control was
thereby avoiding the leakage of intracellular proteins and ions realized at ∼5 V, while nonconductive nanostraw arrays usually
from the persistently opened pores of cell membrane. They also require approximately 20 V for cell poration.129,130
investigated the mechanism of electroporation in this system.
Exogenous PI was added into the microfluidic channel at 4. SURFACE-MEDIATED INTRACELLULAR DELIVERY
intervals of 10 s and 10 min after electroporation. A few cells BY PHOTOTHERMAL PORATION
showed red fluorescence after 10 s, and almost no cells showed In addition to mechanical penetration and electroporation,
red fluorescence after 10 min (Figure 12b), indicating that the photothermal poration based on the photothermal agents
cell membrane resealed within 10 min after electroporation. (PTAs) is another effective physical approach for intracellular
Moreover, simultaneous delivery or sequential delivery of two delivery. Under irradiation of laser with suitable wavelength and
different pDNA (pGFP and pRFP) could be realized (Figure intensity, the PTAs convert the absorbed light energy into
12c). This system was not only suitable for a small amount of thermal energy to increase the membrane permeabilization or
rare cells (about 5 cells) but also for a large amount of cells produce transient pores in cell membrane, facilitating entry of
(greater than 100 000 cells) at one time.129 The dosage of exogenous molecules into cell interior. The possible photo-
delivered molecules could also be precisely control, which is very thermal poration mechanisms are that the increase in local
important for the efficiency of cell reprogramming that are temperature might (i) lead to the local dissociation of
highly affected by the relative ratio of different functional constituent lipid molecules that make up the bilayer or
molecules. denaturation of integral glycoproteins141−143 and (ii) cause
The Al2O3 nanostraw arrays were fabricated by using atomic the evaporation of water molecules around the PTAs to form
layer deposition and Cl2/BCl3 reactive ion etching approaches, vapor nanobubbles (VNBs), the collapse of which would create
which involved sophisticated equipment and complex proce- transient pores in the cell membrane.144−147 Compared with the
dures, and failed to produce conductive nanostraw arrays. To above-mentioned mechanical penetration and electroporation,
solve these limitations, Xie et al. developed an economical and photothermal poration has better spatiotemporal control of
commonly accessible electrodeposition method for fabrication delivery. For example, efficient delivery of macromolecules
of a series of conductive nanostraw arrays based on metals (e.g., suspended in the surrounding cell medium was achieved by
Pt and Au) or conductive polymers (e.g., poly(3,4-ethyl- allowing gold nanoparticles (GNPs) in solution to bind to the
enedioxythiophene), PEDOT). These conductive nanostraw cell membrane to create transient pores upon near-infrared
31066 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

with the delivered molecules during irradiation, and limited


delivery efficiency because the amount of PTAs attached to the
cell membrane is limited. Alternatively, surface-mediated
photothermal poration approaches based on the surfaces on
which the PTAs are firmly immobilized show their own
advantages including low cytotoxicity by avoiding the
endocytosis of suspended PTAs, and improved delivery
efficiency due to the enhanced interaction between the
surface-immobilized PTAs and the surface-attached cells, and/
or increased chance of cell-to-molecule contact.149−151 In this
section, we will introduce the recent development of surface-
mediated photothermal poration approaches that are divided
into three categories based on the PTAs: gold nanomaterials,
PDA, and other photothermal materials (as summarized in
Table 3).
4.1. Gold Nanomaterials. Gold nanomaterials (such as
GNPs and gold nanorods (GNRs)) are typical PTAs with
excellent photothermal effects based on the localized surface
plasmon resonance (LSPR) and good biocompatibility,165
making them suitable candidates for fabrication of surfaces
with photothermal poration capability. In 2010, Chiou et al.
immobilized GNPs on the cell-culture Petri dishes. Under 532
nm visible light irradiation, the GNPs converted the absorbed
Figure 10. (a) Schematic illustration of an electroporation system light energy into heat to generate explosive VNBs, promoting
based on a sandwich-structured micropore membrane. The top layer is the delivery of fluorescent dye (calcein) to attached HEK293T
PET membrane and the bottom layer is Al2O3 membrane coated with cells.149 After this pioneering work, a series of photothermal
electrospun nanofiber. Reproduced with permission from ref 121.
surfaces were developed to deliver diverse molecules into
Copyright 2013 American Chemical Society. (b) Schematic illustration
of a 3D nano electroporation (NEP) system consisting of a 3D NEP various cell types. For example, Mazur et al. developed a
chip, support platform, PDMS holders and electrodes. The exogenous plasmonic nanostructured substrate with gold-coated tipless
molecules stored in bottom chamber were delivered into cells came in pyramids arrays. Under NIR irradiation, the surface produced
contact surface of chip under electric pulse. Reproduced with heat to create nanobubbles and pressure wave, increasing the
permission from ref 127. Copyright 2016 The Royal Society of permeability of cell membrane transiently and thus facilitating
Chemistry. the delivery of fluorescence molecules (calcein) in HeLa cells
with a delivery efficiency of 80%.159 In the following work, they
further developed a gold-coated thermoplasmonic substrate and
achieved delivery of molecules with different molecular weights
(0.6−2000 kDa) into HeLa CCL-2 cells with high delivery
efficiency (up to 95%) and high cell viability (up to 98%). The
throughput was as high as 50 000 cells/min, and could be further
enlarged by scaling up the substrate.160 However, it should be
noted that the thermoplasmonic-based substrates are relatively
expensive with complicated fabrication process, limiting their
broad applications.
Gold nanoparticle layer (GNPL) refer to a layer of dense
immobilized GNPs on a substrate, which has been recognized as
a versatile nanostructured platform for biomedical applications
due to the excellent properties of GNPs and unique two-
dimensional surface topography.166,167 In general, the fabrica-
tion of GNPL is simple, facile and does not need sophisticated
equipment. Compared with dispersed GNPs in solution, the
aggregated GNPs on GNPL exhibit a more efficient light-to-heat
conversion. In addition, GNPL exhibits unique micronanoto-
Figure 11. Schematic illustration of a nanopore membrane-based pography to provide multiple sites for cell contacting and
electroporation system for intracellular delivery to both adherent cells enhance local temperature induced by irradiation.168,169 On the
and suspension cells, where suspension cells came in contact bottom basis of these properties of GNPL, we developed a high-
surface under effect of centrifuge force and adherent cells were cultured
overnight before delivery procedure. The cargoes were delivered into
throughput platform for intracellular delivery with broad
cell interior through electrophoresis. Adapted with permission from ref applicability for both delivered molecules and target cell
128. Copyright 2019 National Academy of Sciences. types.150 As illustrated in Figure 14a, under NIR irradiation,
the increase in surface temperature resulted in the enhanced
membrane transient permeability for the diffusion of molecules
(NIR) irradiation.25,148 However, there are some drawbacks of into cells from the surrounding medium. The molecules were
such solution-mediated photothermal poration such as possible demonstrated to be delivered to the interior of cells rather than
cytotoxicity caused by the PTAs that enter the cells together merely attached to the cell membrane (Figure 14b). Using
31067 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Figure 12. (a) Schematic illustration of Al2O3 nanostraw arrays electroporation for intracellular delivery. The electric field was localized inducing
transient membrane permeability over a small area and decreasing cell damage. (b) Study of cell membrane recovery conditions associated with Al2O3
nanostraw arrays electroporation. Cells cultured on nanostraw arrays under electric field were recovered for 0 s, 10 s, and 10 min, respectively. Then PI
were added in the microfluidic channel at each time point. Scale bar: 50 μm. (c) Simultaneous transfection and sequential transfection with a 24-h
interval of pRFP and pGFP to cells under electroporation. Scale bar: 50 μm. Adapted with permission from ref 129. Copyright 2013 American
Chemical Society.

Figure 13. Schematic illustration of a device based on conductive nanostraw arrays achieving intracellular delivery, extraction of cytosol contents from
live cells, and detection of cell number. The conductive nanostraw arrays could be served as a planar electrode without setting up another planar
electrode at the bottom. Reproduced with permission from ref 132. Copyright 2019 American Chemical Society.

GNPL, high transfection efficiency (∼100%) of pGFP to easy- ± 6.3% for HUVECs). Moreover, besides model pDNA (e.g.,
to-transfect cells (e.g., HeLa cells) was obtained with negligible pGFP), we also delivered a functional pDNA encoding ZNF580
cytotoxicity, however, the transfection efficiencies to hard-to- gene (pZNF580) to HUVECs using this system; the transfected
transfect cells (e.g., 53% for mEFs and 9% for HUVECs) were cells exhibited improvement in both initial attachment and
still limited, which might be due to the possible degradation of proliferation for long-term culture.
naked pDNA by endonucleases. To improve the transfection GNRs are also gold nanomaterials with high photothermal
efficiency of such cells, we incorporated a chemical carrier conversion capability resulting from their distinctive LSPR
(polyethylenimine of low molecular weight (LPEI, 2 kDa)) into effect. In addition, because of their adjustable aspect ratios,
the GNPL-mediated photothermal poration system to combine GNRs can absorb light energy in a wider range (from visible to
their respective advantages together.151 In this upgraded NIR region).170 Utilizing the photothermal effect of GNRs, we
approach, the pDNA was first protected by LPEI to avoid the developed a NIR activated core−sheath structure electrospun
degradation during the transfection process and the resulted nanofiber for in situ controllable gene transfection.152 The
LPEI/pDNA complex was delivered into cytosol by GNPL nanofiber was composed of a core layer containing complex of
under NIR irradiation with dramatically improved delivery fibroblast growth factor-encoding pDNA (pFGF) and LPEI and
efficiencies of recalcitrant cells (88.5 ± 9.2% for mEFs and 94.0 a sheath layer of biocompatible and biodegradable gelatin/L-
31068 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Table 3. Summary of Surface-Mediated Intracellular Delivery by Photothermal Porationa


PTAs delivered molecules cell types delivery efficiency cell viability ref
GNPs fluorescent dye HEK293T 149
GNPL TRITC-dextran/pGFP HeLa/mEFs/HUVECs/ 100% (HeLa) near 100% 150
mESCs 53% (pGFP to mEFs)
44% (pGFP to HUVECs)
pGFP mEFs/HUVECs 88.5% (pGFP to mEFs) 94% (pGFP 80% 151
to HUVECs)
GNRs plasmid bFGF NIH3T3 >85% >90% 152
GNRs DOX A498 cells 153
GNTs PI NIH3T3 ≥95% 154
PDA pDNA HUVECs 85% 155
pDNA HUVECs/HEK293T 40% 156
dextran/BSA/pGFP/pZNF580 HeLa/mEFs/HUVECs/ >99% >90% (HeLa/mEFs/ 157
hDFs/DCs HUVECs) 60%(DCs)
P-MNPs dextran/pDNA HeLa 67% >92.6% 158
TPS-Au FITC-dextran/Calcein HeLa CCL-2 up to 95% up to 98% 159,
160
TPS-TiN calcein dye HeLa CCL-2 >80% >90% 161
Ti thin FITC-dextran/polystyrene beads/ HeLa/hDFs/PB- 58% (bacteria to HeLa) >90% 162
films enzyme/bacteria MDMs/RPTECs 90% (FITC-dextran to hDFs and
RPTECs)
60%(FITC-dextran to PB-MDMs)
Ti thin calcein/FITC-dextran/bacterial enzyme β- Ramos suspension B cells 84% (calcein)/45% (FITC- >89% (pGFP) 163
films lactamase/pGFP dextran)/ 58% (pGFP)
SiNWAs RBITC-BSA/pGFP HeLa/Ramos/T cells 100% (HeLa)/83% (Ramos)/80% >95% 164
(T cells)
a
Abbreviations: Gold nanoparticles (GNPs); gold nanoparticle layer (GNPL); gold nanorods (GNRs); gold nanotubes (GNTs); porous magnetic
iron oxide nanoparticles (P-MNPs); gold-coated thermoplasmonic substrates (TPS-Au); TiN-coated thermoplasmonic substrates (TPS-TiN);
Calcein (0.6 kDa); Tetramethylrhodamine isothiocyanate-labeled dextran (TRITC-dextran, 4.4 kDa); fluorescein isothiocyanate-labeled dextran
(FITC-dextran, 0.6, 4, 40, 2000 kDa); pDNA encoding fibroblast growth factor/ZNF580 gene (pbFGF, 6825 bp/pZNF580); polystyrene beads
(20 nm to 1 μm); bacterial enzyme β-lactamase (29 kDa); doxorubicin (DOX); rhodamine B isothiocyanate-labeled bovine serum albumin
(RBITC-BSA, 66 kDa); peripheral blood monocyte-derived macrophages (PB-MDMs); renal proximal tubule epithelial cells (RPTECs).

poly(lactic acid) with uniformly distributed GNRs (Figure 15a). cells.156 The potential delivery mechanism was examined by
Under NIR irradiation, the heat generated by the encapsulated Sytox (a membrane-impermeable dye) staining assay and lactate
GNRs could (i) increase the permeability of the nanofibers to dehydrogenase (LDH, a cytosolic enzyme present inside the
achieve the controlled release of pFGF from the core layer and cell) assay, both of which were commonly used methods to test
(ii) generate transient holes in the membranes of NIH3T3 the membrane integrity. As shown in Figure 16b, without NIR
fibroblasts on the nanofibers to enhance the delivery of pFGF irradiation, the attached cells did not show any fluorescence,
(Figure 15b). The proliferation and migration of pFGF indicating the intact healthy cell membranes. In contrast, the
transfected fibroblasts were effectively improved, indicating cells emitted bright red fluorescence under NIR irradiation and
great potential of such nanofibers with localized surface- the number of cells representing fluorescence increased with
mediated gene transfection capability. increasing irradiation time. Moreover, quantification of the
4.2. Polydopamine. Polydopamine (PDA) is well-known as released LDH showed that NIR irradiation resulted in an
a mussel-inspired material, which has been extensively studied in obvious increase in LDH release. Taken together, it is suggested
surface chemistry. Besides strong adhesive property, good that the intracellular delivery of this system was due to the
biocompatibility, easy functionalization, PDA also has excellent disturbance and destabilization of cell membrane resulted from
photothermal property to effectively absorb and transfer NIR the localized hyperthermia generated by PDA.156
light energy to heat.171−173 Taking advantages of these Besides intracellular delivery of functional molecules with
properties, Ji and Ren et al. reported a photothermal poration high efficiency, efficient and nontraumatic cell harvest is also
surface based on poly(D,L-lactide-co-glycolide) (PLGA) poly- important and plays a critical role in many applications, in
meric spongy film modified with PDA coating (Figure 16a).155 particular ex vivo cell-based therapy, where the engineered cells
Because of the microporous structure, the PEI/pDNA with delivered molecules are to be injected to a patient.1,3
complexes could be loaded by a simple wicking effect and the However, the cell harvest capability is rarely mentioned in above
amount of loaded complexes could be varied by changing the surface-mediated photothermal poration systems. Recently, we
deposition time or solution concentration. Under NIR developed a “two-in-one” platform integrated with intracellular
irradiation, the primary endothelial cells came in contact to delivery and cell harvest functions (Figure 17).157 This surface
the spongy film were delivered with loaded pDNA with high was fabricated by deposition of a vinyl group-containing PDA
efficiency (85%) and low cytotoxicity. In addition, the spatial layer, followed by polymerization to graft a thermoresponsive
control delivery could be achieved via NIR irradiation in the polymer poly (N-isopropylacrylamide) (PNIPAAm). The
defined area. In the following work, they simplified the underlying PDA layer provided good photothermal effect for
fabrication process by codeposition of PDA and PEI on the effective delivery of a variety of exogenous molecules (e.g.,
substrate and successfully transfected pGFP to endothelial polysaccharide, protein, and pDNA) to diverse cell types
31069 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Figure 14. (a) Schematic illustration of intracellular delivery using the GNPL-mediated photothermal poration system. Left: Complete experimental
procedure of GNPL-mediated intracellular delivery. Right: Permeability of cell membrane was enhanced to facilitate entry of exogenous
macromolecules into cell interior. (b) Representative confocal microscopy images of HeLa cells delivered with TRITC-dextran using GNPL under
NIR irradiation measured using the Z-stack scan mode of confocal microscopy. Adapted with permission from ref 150. Copyright 2016 John Wiley and
Sons.

including three typical hard-to-transfect cells (mEFs, HUVECs, HUVECs exhibited significantly increased proliferation and
and DCs). The grafted PNIPAAm did not influence the migration capabilities compared to unmodified cells.
photothermal properties of the PDA layer, but allowed the 4.3. Other Photothermal Materials. In addition to gold
surface to release the “engineered” cells by decreasing the nanomaterials and PDA, other photothermal materials have also
temperature of cell medium to induce the transition of surface been exploited for photothermal poration intracellular delivery.
property from cell-adhesive to cell-repellent.174 The harvested For example, Chiou et al. developed a massively parallel
cells maintained their viability for subculture and the functions photothermal biophotonic laser-assisted surgery tool (BLAST)
of delivered molecules were conserved. Using HUVECs and enabling the entry of large micrometer-sized cargoes into
pZNF580 as a pair of model cell line and functional molecule, cells.162 The BLAST platform consisted of a silicon chip with a
the feasibility and effectiveness of this PDA−PNIPAAm thin porous SiO2 membrane on top, providing an array of
platform was demonstrated as the harvested “engineered” micrometer-wide, trans-film holes whose side walls were
31070 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Figure 15. (a) Schematic illustration of fabrication of the core−shell structured electrospun fiber composed of pbFGF/PEI complex as core layer and
GNRs/gelatin/L-poly(lactic acid) as shell layer. (b) Intracellular delivery of pbFGF to NIH3T3 fibroblasts using the core−shell structured electrospun
fiber under NIR irradiation. Left: Experimental procedure of electrospun fiber-mediated intracellular delivery. Right: Amplified images of cell
membrane with/without NIR irradiation. Reproduced with permission from ref 152. Copyright 2020 American Chemical Society.

Figure 16. (a) Schematic illustration of a PDA coated microporous spongy film platform for intracellular delivery via photothermal poration, where the
pDNA was first protected by bPEI to form a pDNA/bPEI complex, then the complex was loaded onto surface of PDA-coated microporous spongy film
by simple wicking effect. Reproduced with permission from ref 155. Copyright 2019 American Chemical Society. (b) Representative fluorescence
images of cells on PDA−PEI films without irradiation, with irradiation for 70 or 140 s, respectively. Red fluorescence is from delivered Sytox.
Reproduced with permission from ref 156. Copyright 2019 The Royal Society of Chemistry.

asymmetrically coated with crescent-shaped titanium thin films. Unfortunately, the above-mentioned surface-mediated photo-
This BLAST platform harvested the light energy to induce rapid thermal poration delivery systems are not suitable for suspension
heating and vaporization of adjacent water molecules to trigger cells due to the poor cell-surface contact.140 To solve this
limitation, Chiou et al. developed a photothermal 3D microwells
cavitation bubbles. These cavitation bubbles grew, coalesced,
with sharp nanoscale metal-coated tips, permitting self-position
and collapsed within 110 ns. The rapid bubble “explosion” led to
of suspension cells by synergetic effect of gravity with each
strong fluid flows to disrupt the nearby cell membrane, microwell and size effect of microwells in direct contact with
facilitating the entry of the exogenous large cargoes including eight sharp tips (Figure 18).163 Under NIR irradiation, a range of
proteins, bacteria, and nanoparticles to various cell types. cargoes were delivered into suspension Ramos B cells stayed in
31071 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

enhance the interactions between the surface of SiNWAs and


cell membranes, leading to a high capture capacity for both
adherent cells (e.g., HeLa cells) and suspension cells (e.g.,
Ramos cells and T cells) that overexpressed sialic acid on the
membrane. Under NIR irradiation, the local heat generated by
SiNWAs efficiently enhanced membrane permeability of
captured cells transiently for delivery of diverse macromolecules
(protein and pDNA). The transfection efficiency of pDNA to
the hard-to-transfect suspension immune T cells was more than
80%, significantly higher than that using the commercial
transfection reagents and other reported methods.178,179
Moreover, because of the sugar-responsiveness of phenyl-
boronate ester bonds,180,181 the “engineered” cells could be
released from the surface by sugar treatment. As sugars were
natural biomolecules and are generally nontoxic, the harvested
“engineered” cells maintained high viability for normal growth
and proliferation, suggesting the potentials for further
fundamental research and clinical application.164

Figure 17. Schematic illustration of the universal “two-in-one” platform 5. SUMMARY AND PERSPECTIVES
integrated with intracellular delivery and cell harvest functions based on
The decade just passed has witnessed rapid developments in
a PDA layer grafted with PNIPAAm. The cells were first delivered with
exogenous molecules under NIR irradiation, and then, the “engineered” surface-mediated PMD-based methods for the intracellular
cells could be harvested from the surface by decreasing cell medium delivery of exogenous molecules and functional materials. In this
temperature, finally, the harvested cells could be recultured for next use Review, we systematically summarize achievements in intra-
or research. Reproduced with permission from ref 157. Copyright 2019 cellular delivery systems based on mechanical penetration,
American Chemical Society. electroporation, and photothermal poration and discuss their
respective characteristics. PMD-based methods allow “crossing”
microwells with a high throughput of >100 000 cells delivered the outer cell membrane “barrier”, thus facilitating direct
per minute. delivery of diverse types of exogenous molecule into target
cells. Compared with traditional carrier-mediated methods and
solution-mediated PMD-based methods, surface-mediated
PMD-based methods exhibit high delivery efficiency, low
cytotoxicity, and broad applicability to diverse molecules and
cell types.
Although considerable progress has been made, many
challenges remain and these should be the focus of future
research. From fundamental point of view, a greater under-
standing of the mechanisms of physical membrane disruption,
such as whether the transient pores are produced in cell
Figure 18. Schematic illustration of intracellular photothermal delivery membrane, the size of pores, the resealing of pores, and the
for suspension cells using sharp nanoscale tips in microwells. Adapted possible leakage of cell contents is increasingly important for this
with permission from ref 163. Copyright 2019 American Chemical field, but little detailed research has been done in this area
Society. related to intracellular delivery, hence these factors require in-
depth investigation. Assessing the safety and effectiveness of
Although the photothermal microwell-based system achieved engineered cells is of importance for ex vivo cell therapy,
macromolecular delivery to suspension cells, the fabrication however, most of current researches merely focused on
process was complicated and no cell harvest function involved. investigating cell viability, but ignoring the further detection
Ideally, a surface-mediated photothermal poration system such as the DNA damage, changes of cell fate and cell functions,
should have high delivery efficiency, high cell viability, and which requires engineering technology researchers to develop
high cell harvest capability for both adherent cells and more accurate detection tools and techniques to detect changes
suspension cells. In addition, it is pointed out that low- of cell interior. From practical point of view, most of the
temperature treatment for cell harvest used in our PDA− researches to date have investigated the delivery of model
PNIPAAm system may decrease cell viability.175 Therefore, a molecules (such as pGFP) into model cell types, while the
gentle way to trigger cell release is more promising. To this end, delivery of functional molecules to edit cells (e.g., CRISPR/
we developed a smart photothermal poration platform that Cas9-based genome editing for disease modeling and therapy)
combined three sequential functions including cell capture, to prepare therapeutic cells for in vitro or ex vivo cell-therapy
intracellular delivery, and cell harvest together to engineer living (e.g., chimeric antigen receptor (CAR)-T cells immunotherapy
cells (Figure 19).164 This platform was fabricated by the for human cancer) or molecular probes and nanomaterial
modification of SiNWAs possessing photothermal effect with a sensors to analyze cell behaviors (e.g., live-cell imaging) remains
polymer containing phenylboronic acid (PBA) groups with largely unexplored. Moreover, many platforms now only
sugar-responsive property. The unique 3D nanostructure of involved intracellular delivery, while the upstream cell capture
SiNWAs84,176 could increase the local density of PBA groups or downstream cell detection and cell harvest are ignored.
that can form phenylboronate ester bonds with sialic acid177 and Therefore, it is urgent to develop multifunctional platforms that
31072 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Figure 19. Schematic illustration of a universal platform based on SiNWAs modified with sugar-responsive polymers for engineering cells. Left: Cells
including adherent and suspension cells were captured due to the synergistic effects of “nano-enhancement” of SiNWAs and chemical recognition of
PHB. Middle: Cells came in contact surface of SN-PHB were delivered with exogenous molecules under NIR irradiation. Right: “Engineered” cells
were harvested from surface by fructose treatment. Reproduced with permission from ref 164. Copyright 2020 John Wiley and Sons.

can conduct multiple cell manipulation processes, which can be Hospital, Soochow University, Suzhou 215007, P. R. China;
better applied to the in situ visualization of single cell analysis, in orcid.org/0000-0002-1920-6709
situ chemical manipulation, and analysis of captured circulating Hong Chen − State and Local Joint Engineering Laboratory for
tumor cells and ex vivo cell-based therapies. Novel Functional Polymeric Materials, College of Chemistry,
Finally, it is hoped that this Review will be of benefit to the Chemical Engineering and Materials Science, Soochow University,
broad community engaged in engineering manufacture, Suzhou 215123, P. R. China; orcid.org/0000-0001-7799-
automatic research, intracellular molecular delivery, immuno- 4961
therapy, regenerative medicine, tissue engineering, surface Complete contact information is available at:
chemistry, and other fields. For engineers, we expect to provide https://pubs.acs.org/10.1021/acsami.0c06978
them with inspirations to develop more compact and portable
devices integrating all cell manipulation processes and a robot Author Contributions
matching array-type and flow-through systems to achieve high
level of automation. With the development of surface-mediated Contributions to this Review were made by all authors, and all
intracellular delivery methods by physical membrane disruption, have seen and approved the submitted manuscript.
researchers need to develop precise toolboxes to study the Author Contributions
§
mechanisms of physical membrane disruption from the micro/ Y.Q. and Y.Z. contributed equally.
nano scale. It is hoped that future research will be able to regulate Funding
cell function and detect the intracellular environment through
The National Natural Science Foundation of China (21774086
intracellular delivery strategies, thereby providing effective
and 21935008), the Natural Science Foundation of Jiangsu
strategies for drug delivery, cell therapy, gene editing, stem cell
Province (BK20180093), the Suzhou Municipal Science and
transfection, reprogramming of induced pluripotent stem cells,
and neuronal analysis to contribute to various cancer treatments Technology Foundation (SYS2018026), and the Priority
and gene-related diseases. Academic Program Development of Jiangsu Higher Education


Institutions (PAPD).
AUTHOR INFORMATION Notes
The authors declare no competing financial interest.


Corresponding Author
Qian Yu − State and Local Joint Engineering Laboratory for Novel ACKNOWLEDGMENTS
Functional Polymeric Materials, College of Chemistry, Chemical
Engineering and Materials Science, Soochow University, Suzhou This work was supported by the National Natural Science
215123, P. R. China; orcid.org/0000-0003-3612-6951; Foundation of China (21774086 and 21935008), the Natural
Email: yuqian@suda.edu.cn Science Foundation of Jiangsu Province (BK20180093), the
Suzhou Municipal Science and Technology Foundation
Authors (SYS2018026), and the Priority Academic Program Develop-
Yangcui Qu − State and Local Joint Engineering Laboratory for ment of Jiangsu Higher Education Institutions (PAPD). The
Novel Functional Polymeric Materials, College of Chemistry, authors thank Prof. John L. Brash for his helpful discussions and
Chemical Engineering and Materials Science, Soochow University, Mr. Yang Zhou for his assistance on graphic works. The authors
Suzhou 215123, P. R. China also sincerely appreciated the anonymous reviewer for his/her
Yanxia Zhang − Institute for Cardiovascular Science and constructive comments, which are very helpful to improve the
Department of Cardiovascular Surgery of the First Affiliated thoroughness and presentation of this paper.
31073 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces


www.acsami.org Review

REFERENCES (23) Antkowiak, M.; Torres-Mapa, M. L.; Stevenson, D. J.; Dholakia,


K.; Gunn-Moore, F. J. Femtosecond Optical Transfection of Individual
(1) Shi, Y.; Inoue, H.; Wu, J. C.; Yamanaka, S. Induced Pluripotent Mammalian Cells. Nat. Protoc. 2013, 8, 1216−1233.
Stem Cell Technology: A Decade of Progress. Nat. Rev. Drug Discovery (24) Geng, T.; Zhan, Y.; Wang, H. Y.; Witting, S. R.; Cornetta, K. G.;
2017, 16, 115−130. Lu, C. Flow-through Electroporation Based on Constant Voltage for
(2) Wang, H.-X.; Li, M.; Lee, C. M.; Chakraborty, S.; Kim, H.-W.; Bao, Large-Volume Transfection of Cells. J. Controlled Release 2010, 144,
G.; Leong, K. W. Crispr/Cas9-Based Genome Editing for Disease 91−100.
Modeling and Therapy: Challenges and Opportunities for Nonviral (25) Xiong, R.; Raemdonck, K.; Peynshaert, K.; Lentacker, I.; De
Delivery. Chem. Rev. 2017, 117, 9874−9906. Cock, I.; Demeester, J.; De Smedt, S. C.; Skirtach, A. G.; Braeckmans, K.
(3) June, C. H.; O’Connor, R. S.; Kawalekar, O. U.; Ghassemi, S.; Comparison of Gold Nanoparticle Mediated Photoporation: Vapor
Milone, M. C. Car T Cell Immunotherapy for Human Cancer. Science Nanobubbles Outperform Direct Heating for Delivering Macro-
2018, 359, 1361−1365. molecules in Live Cells. ACS Nano 2014, 8, 6288−6296.
(4) Zhang, Y.; Roise, J. J.; Lee, K.; Li, J.; Murthy, N. Recent (26) Lamichhane, T. N.; Raiker, R. S.; Jay, S. M. Exogenous DNA
Developments in Intracellular Protein Delivery. Curr. Opin. Biotechnol. Loading into Extracellular Vesicles Via Electroporation is Size-
2018, 52, 25−31. Dependent and Enables Limited Gene Delivery. Mol. Pharmaceutics
(5) Yang, N. J.; Hinner, M. J. Getting across the Cell Membrane: An 2015, 12, 3650−3657.
Overview for Small Molecules, Peptides, and Proteins. Methods Mol. (27) Raes, L.; Van Hecke, C.; Michiels, J.; Stremersch, S.; Fraire, J. C.;
Biol. 2015, 1266, 29−53. Brans, T.; Xiong, R.; De Smedt, S.; Vandekerckhove, L.; Raemdonck,
(6) Zhang, S.; Zhao, Y.; Zhao, B.; Wang, B. Hybrids of Nonviral K.; Braeckmans, K. Gold Nanoparticle-Mediated Photoporation
Vectors for Gene Delivery. Bioconjugate Chem. 2010, 21, 1003−1009. Enables Delivery of Macromolecules over a Wide Range of Molecular
(7) Yin, H.; Kanasty, R. L.; Eltoukhy, A. A.; Vegas, A. J.; Dorkin, J. R.; Weights in Human CD4+ T Cells. Crystals 2019, 9, 411.
Anderson, D. G. Non-Viral Vectors for Gene-Based Therapy. Nat. Rev. (28) Wang, C.-H. K.; Pun, S. H. Substrate-Mediated Nucleic Acid
Genet. 2014, 15, 541−555. Delivery from Self-Assembled Monolayers. Trends Biotechnol. 2011, 29,
(8) Stewart, M. P.; Sharei, A.; Ding, X.; Sahay, G.; Langer, R.; Jensen, 119−126.
K. F. In Vitro and Ex Vivo Strategies for Intracellular Delivery. Nature (29) Peer, E.; et al. Hollow Nanoneedle Array and Its Utilization for
2016, 538, 183−192. Repeated Administration of Biomolecules to the Same Cells. ACS Nano
(9) Stewart, M. P.; Langer, R.; Jensen, K. F. Intracellular Delivery by 2012, 6, 4940−4946.
Membrane Disruption: Mechanisms, Strategies, and Concepts. Chem. (30) Aalipour, A.; Xu, A. M.; Leal-Ortiz, S.; Garner, C. C.; Melosh, N.
Rev. 2018, 118, 7409−7531. A. Plasma Membrane and Actin Cytoskeleton as Synergistic Barriers to
(10) Guo, X.; Huang, L. Recent Advances in Nonviral Vectors for Nanowire Cell Penetration. Langmuir 2014, 30, 12362−12367.
Gene Delivery. Acc. Chem. Res. 2012, 45, 971−979. (31) Liu, J.; Fraire, J. C.; De Smedt, S. C.; Xiong, R.; Braeckmans, K.
(11) Bordet, T.; Behar-Cohen, F. Ocular Gene Therapies in Clinical Intracellular Labeling with Extrinsic Probes: Delivery Strategies and
Practice: Viral Vectors and Nonviral Alternatives. Drug Discovery Today Applications. Small 2020, 16, 2000146.
2019, 24, 1685−1693. (32) Xie, X.; Xu, A. M.; Angle, M. R.; Tayebi, N.; Verma, P.; Melosh,
(12) Bråve, A.; Ljungberg, K.; Wahren, B.; Liu, M. A. Vaccine Delivery N. A. Mechanical Model of Vertical Nanowire Cell Penetration. Nano
Methods Using Viral Vectors. Mol. Pharmaceutics 2007, 4, 18−32. Lett. 2013, 13, 6002−6008.
(13) Stewart, M. P.; Lorenz, A.; Dahlman, J.; Sahay, G. Challenges in (33) Elnathan, R.; Kwiat, M.; Patolsky, F.; Voelcker, N. H.
Carrier-Mediated Intracellular Delivery: Moving Beyond Endosomal Engineering Vertically Aligned Semiconductor Nanowire Arrays for
Barriers. Wiley Interdiscip. Rev.: Nanomed. Nanobiotechnol. 2016, 8, Applications in the Life Sciences. Nano Today 2014, 9, 172−196.
465−478. (34) Kwak, M.; Han, L.; Chen, J. J.; Fan, R. Interfacing Inorganic
(14) Tong, S.; Moyo, B.; Lee, C. M.; Leong, K.; Bao, G. Engineered Nanowire Arrays and Living Cells for Cellular Function Analysis. Small
Materials for in Vivo Delivery of Genome-Editing Machinery. Nat. Rev. 2015, 11, 5600−5610.
Mater. 2019, 4, 726−737. (35) Lee, K.; Lingampalli, N.; Pisano, A. P.; Murthy, N.; So, H.
(15) Zhao, L.; Skwarczynski, M.; Toth, I. Polyelectrolyte-Based Physical Delivery of Macromolecules Using High-Aspect Ratio
Platforms for the Delivery of Peptides and Proteins. ACS Biomater. Sci. Nanostructured Materials. ACS Appl. Mater. Interfaces 2015, 7,
Eng. 2019, 5, 4937−4950. 23387−23397.
(16) Meacham, J. M.; Durvasula, K.; Degertekin, F. L.; Fedorov, A. G. (36) Higgins, S. G.; Becce, M.; Belessiotis-Richards, A.; Seong, H.;
Physical Methods for Intracellular Delivery: Practical Aspects from Sero, J. E.; Stevens, M. M. High-Aspect-Ratio Nanostructured Surfaces
Laboratory Use to Industrial-Scale Processing. JALA 2014, 19, 1−18. as Biological Metamaterials. Adv. Mater. 2020, 32, 1903862.
(17) Du, X.; Wang, J.; Zhou, Q.; Zhang, L.; Wang, S.; Zhang, Z.; Yao, (37) Kim, W.; Ng, J. K.; Kunitake, M. E.; Conklin, B. R.; Yang, P.
C. Advanced Physical Techniques for Gene Delivery Based on Interfacing Silicon Nanowires with Mammalian Cells. J. Am. Chem. Soc.
Membrane Perforation. Drug Delivery 2018, 25, 1516−1525. 2007, 129, 7228−7229.
(18) He, G.; Hu, N.; Xu, A. M.; Li, X.; Zhao, Y.; Xie, X. Nanoneedle (38) Peckys, D. B.; Melechko, A. V.; Simpson, M. L.; McKnight, T. E.
Platforms: The Many Ways to Pierce the Cell Membrane. Adv. Funct. Immobilization and Release Strategies for DNA Delivery Using Carbon
Mater. 2020, 30, 1909890. Nanofiber Arrays and Self-Assembled Monolayers. Nanotechnology
(19) Fumoto, S.; Nakajima, S.; Mine, T.; Yoshikawa, N.; Kitahara, T.; 2009, 20, 145304.
Sasaki, H.; Miyamoto, H.; Nishida, K. Efficient in Vivo Gene Transfer (39) Shalek, A. K.; Robinson, J. T.; Karp, E. S.; Lee, J. S.; Ahn, D.-R.;
by Intraperitoneal Injection of Plasmid DNA and Calcium Carbonate Yoon, M.-H.; Sutton, A.; Jorgolli, M.; Gertner, R. S.; Gujral, T. S.;
Microflowers in Mice. Mol. Pharmaceutics 2012, 9, 1962−1970. MacBeath, G.; Yang, E. G.; Park, H. Vertical Silicon Nanowires as a
(20) Shi, J. F.; Ma, Y. F.; Zhu, J.; Chen, Y. X.; Sun, Y. T.; Yao, Y. C.; Universal Platform for Delivering Biomolecules into Living Cells. Proc.
Yang, Z. G.; Xie, J. A Review on Electroporation-Based Intracellular Natl. Acad. Sci. U. S. A. 2010, 107, 1870−1875.
Delivery. Molecules 2018, 23, 3044. (40) Shalek, A. K.; Gaublomme, J. T.; Wang, L.; Yosef, N.; Chevrier,
(21) Jenkins, S. I.; Pickard, M. R.; Granger, N.; Chari, D. M. Magnetic N.; Andersen, M. S.; Robinson, J. T.; Pochet, N.; Neuberg, D.; Gertner,
Nanoparticle-Mediated Gene Transfer to Oligodendrocyte Precursor R. S.; Amit, I.; Brown, J. R.; Hacohen, N.; Regev, A.; Wu, C. J.; Park, H.
Cell Transplant Populations Is Enhanced by Magnetofection Strategies. Nanowire-Mediated Delivery Enables Functional Interrogation of
ACS Nano 2011, 5, 6527−6538. Primary Immune Cells: Application to the Analysis of Chronic
(22) Maciulevicius, M.; Tamosiunas, M.; Jurkonis, R.; Venslauskas, M. Lymphocytic Leukemia. Nano Lett. 2012, 12, 6498−6504.
S.; Satkauskas, S. Analysis of Metrics for Molecular Sonotransfer in (41) Chiappini, C.; De Rosa, E.; Martinez, J. O.; Liu, X.; Steele, J.;
Vitro. Mol. Pharmaceutics 2015, 12, 3620−3627. Stevens, M. M.; Tasciotti, E. Biodegradable Silicon Nanoneedles

31074 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Delivering Nucleic Acids Intracellularly Induce Localized In vivo (59) Harding, F. J.; Surdo, S.; Delalat, B.; Cozzi, C.; Elnathan, R.;
neovascularization. Nat. Mater. 2015, 14, 532−539. Gronthos, S.; Voelcker, N. H.; Barillaro, G. Ordered Silicon Pillar
(42) Xie, X.; Aalipour, A.; Gupta, S. V.; Melosh, N. A. Determining the Arrays Prepared by Electrochemical Micromachining: Substrates for
Time Window for Dynamic Nanowire Cell Penetration Processes. ACS High-Efficiency Cell Transfection. ACS Appl. Mater. Interfaces 2016, 8,
Nano 2015, 9, 11667−11677. 29197−29202.
(43) Gopal, S.; Chiappini, C.; Penders, J.; Leonardo, V.; Seong, H.; (60) Park, S.; Choi, S. O.; Paik, S. J.; Choi, S.; Allen, M.; Prausnitz, M.
Rothery, S.; Korchev, Y.; Shevchuk, A.; Stevens, M. M. Porous Silicon Intracellular Delivery of Molecules Using Microfabricated Nanoneedle
Nanoneedles Modulate Endocytosis to Deliver Biological Payloads. Arrays. Biomed. Microdevices 2016, 18, 10.
Adv. Mater. 2019, 31, 1806788. (61) Matsumoto, D.; Rao Sathuluri, R.; Kato, Y.; Silberberg, Y. R.;
(44) Chen, Y.; Aslanoglou, S.; Gervinskas, G.; Abdelmaksoud, H.; Kawamura, R.; Iwata, F.; Kobayashi, T.; Nakamura, C. Oscillating
Voelcker, N. H.; Elnathan, R. Cellular Deformations Induced by High-Aspect-Ratio Monolithic Silicon Nanoneedle Array Enables
Conical Silicon Nanowire Arrays Facilitate Gene Delivery. Small 2019, Efficient Delivery of Functional Bio-Macromolecules into Living
15, 1904819. Cells. Sci. Rep. 2015, 5, 15325.
(45) Hanson, L.; Lin, Z. C.; Xie, C.; Cui, Y.; Cui, B. Characterization (62) Matsumoto, D.; Yamagishi, A.; Saito, M.; Sathuluri, R. R.;
of the Cell-Nanopillar Interface by Transmission Electron Microscopy. Silberberg, Y. R.; Iwata, F.; Kobayashi, T.; Nakamura, C. Mechanopo-
Nano Lett. 2012, 12, 5815−5820. ration of Living Cells for Delivery of Macromolecules Using
(46) Berthing, T.; Bonde, S.; Rostgaard, K. R.; Madsen, M. H.; Nanoneedle Array. J. Biosci. Bioeng. 2016, 122, 748−752.
Sorensen, C. B.; Nygard, J.; Martinez, K. L. Cell Membrane (63) Chiappini, C.; Martinez, J. O.; De Rosa, E.; Almeida, C. S.;
Conformation at Vertical Nanowire Array Interface Revealed by Tasciotti, E.; Stevens, M. M. Biodegradable Nanoneedles for Localized
Fluorescence Imaging. Nanotechnology 2012, 23, 415102. Delivery of Nanoparticles in Vivo: Exploring the Biointerface. ACS
(47) Xu, A. M.; Aalipour, A.; Leal-Ortiz, S.; Mekhdjian, A. H.; Xie, X.; Nano 2015, 9, 5500−5509.
Dunn, A. R.; Garner, C. C.; Melosh, N. A. Quantification of Nanowire (64) Teichert, G. H.; Burnett, S.; Jensen, B. D. A Microneedle Array
Penetration into Living Cells. Nat. Commun. 2014, 5, 3613. Able to Inject Tens of Thousands of Cells Simultaneously. J. Micromech.
(48) Wang, Y.; Yang, Y.; Yan, L.; Kwok, S. Y.; Li, W.; Wang, Z.; Zhu, Microeng. 2013, 23, 095003.
X.; Zhu, G.; Zhang, W.; Chen, X.; Shi, P. Poking Cells for Efficient (65) Chen, X.; Zhu, G.; Yang, Y.; Wang, B.; Yan, L.; Zhang, K. Y.; Lo,
Vector-Free Intracellular Delivery. Nat. Commun. 2014, 5, 4466. K. K.; Zhang, W. A Diamond Nanoneedle Array for Potential High-
(49) Kim, K. H.; Kim, J.; Choi, J. S.; Bae, S.; Kwon, D.; Park, I.; Kim, Throughput Intracellular Delivery. Adv. Healthcare Mater. 2013, 2,
D. H.; Seo, T. S. Rapid, High-Throughput, and Direct Molecular 1103−1107.
Beacon Delivery to Human Cancer Cells Using a Nanowire- (66) McKnight, T. E.; Melechko, A. V.; Griffin, G. D.; Guillorn, M. A.;
Incorporated and Pneumatic Pressure-Driven Microdevice. Small
Merkulov, V. I.; Serna, F.; Hensley, D. K.; Doktycz, M. J.; Lowndes, D.
2015, 11, 6215−6224.
H.; Simpson, M. L. Intracellular Integration of Synthetic Nanostruc-
(50) Pan, J.; Lyu, Z.; Jiang, W.; Wang, H.; Liu, Q.; Tan, M.; Yuan, L.;
tures with Viable Cells for Controlled Biochemical Manipulation.
Chen, H. Stimulation of Gene Transfection by Silicon Nanowire Arrays
Nanotechnology 2003, 14, 551−556.
Modified with Polyethylenimine. ACS Appl. Mater. Interfaces 2014, 6,
(67) McKnight, T. E.; Melechko, A. V.; Hensley, D. K.; Mann, D. G. J.;
14391−14398.
Griffin, G. D.; Simpson, M. L. Tracking Gene Expression after DNA
(51) Pan, J.; Yuan, Y.; Wang, H.; Liu, F.; Xiong, X.; Chen, H.; Yuan, L.
Efficient Transfection by Using Pdmaema-Modified Sinwas as a Delivery Using Spatially Indexed Nanofiber Arrays. Nano Lett. 2004, 4,
Platform for Ca(2+)-Dependent Gene Delivery. ACS Appl. Mater. 1213−1219.
Interfaces 2016, 8, 15138−15144. (68) Mann, D. G. J.; McKnight, T. E.; McPherson, J. T.; Hoyt, P. R.;
(52) Huang, X.; Li, Z.; Wu, J.; Hang, Y.; Wang, H.; Yuan, L.; Chen, H. Simpson, M. L.; Sayler, G. S.; et al. Inducible Rna Interference-
Small Addition of Zn2+ in Ca2+@DNA Results in Elevated Gene Mediated Gene Silencing Using Nanostructured Gene Delivery Arrays.
Transfection by Aminated Pgma-Modified Silicon Nanowire Arrays. J. ACS Nano 2008, 2, 69−76.
Mater. Chem. B 2019, 7, 566−575. (69) Peckys, D. B.; de Jonge, N.; Simpson, M. L.; McKnight, T. E.
(53) Liu, Y. P.; Wang, J. H.; Huang, X. J.; Zhao, Y. X.; Sun, Y.; Zhang, End-Specific Strategies of Attachment of Long Double Stranded DNA
S. X.; Wang, H. W.; Yuan, L.; Chen, H. Glutathione-Sensitive Silicon onto Gold-Coated Nanofiber Arrays. Nanotechnology 2008, 19, 435301.
Nanowire Arrays for Gene Transfection. ACS Appl. Mater. Interfaces (70) Park, S.; Kim, Y.-S.; Kim, W. B.; Jon, S. Carbon Nanosyringe
2019, 11, 46515−46524. Array as a Platform for Intracellular Delivery. Nano Lett. 2009, 9, 1325−
(54) Nair, B. G.; Hagiwara, K.; Ueda, M.; Yu, H. H.; Tseng, H. R.; Ito, 1329.
Y. High Density of Aligned Nanowire Treated with Polydopamine for (71) Choi, M.; Lee, S. H.; Kim, W. B.; Gujrati, V.; Kim, D.; Lee, J.;
Efficient Gene Silencing by Sirna According to Cell Membrane Kim, J. I.; Kim, H.; Saw, P. E.; Jon, S. Intracellular Delivery of Bioactive
Perturbation. ACS Appl. Mater. Interfaces 2016, 8, 18693−18700. Cargos to Hard-to-Transfect Cells Using Carbon Nanosyringe Arrays
(55) Peng, J.; Garcia, M. A.; Choi, J.-s.; Zhao, L.; Chen, K.-J.; under an Applied Centrifugal G-Force. Adv. Healthcare Mater. 2016, 5,
Bernstein, J. R.; Peyda, P.; Hsiao, Y.-S.; Liu, K. W.; Lin, W.-Y.; Pyle, A. 101−107.
D.; Wang, H.; Hou, S.; Tseng, H.-R. Molecular Recognition Enables (72) Bae, S.; Park, S.; Kim, J.; Choi, J. S.; Kim, K. H.; Kwon, D.; Jin, E.;
Nanosubstrate-Mediated Delivery of Gene-Encapsulated Nanopar- Park, I.; Kim, D. H.; Seo, T. S. Exogenous Gene Integration for
ticles with High Efficiency. ACS Nano 2014, 8, 4621−4629. Microalgal Cell Transformation Using a Nanowire-Incorporated
(56) Hou, S.; Choi, J. S.; Chen, K. J.; Zhang, Y.; Peng, J.; Garcia, M. A.; Microdevice. ACS Appl. Mater. Interfaces 2015, 7, 27554−27561.
Yu, J. H.; Thakore-Shah, K.; Ro, T.; Chen, J. F.; Peyda, P.; Fan, G.; Pyle, (73) Sharma, P.; Cho, H. A.; Lee, J. W.; Ham, W. S.; Park, B. C.; Cho,
A. D.; Wang, H.; Tseng, H. R. Supramolecular Nanosubstrate- N. H.; Kim, Y. K. Efficient Intracellular Delivery of Biomacromolecules
Mediated Delivery for Reprogramming and Transdifferentiation of Employing Clusters of Zinc Oxide Nanowires. Nanoscale 2017, 9,
Mammalian Cells. Small 2015, 11, 2499−2504. 15371−15378.
(57) Elnathan, R.; Isa, L.; Brodoceanu, D.; Nelson, A.; Harding, F. J.; (74) Mumm, F.; Beckwith, K. M.; Bonde, S.; Martinez, K. L.; Sikorski,
Delalat, B.; Kraus, T.; Voelcker, N. H. Versatile Particle-Based Route to P. A Transparent Nanowire-Based Cell Impalement Device Suitable for
Engineer Vertically Aligned Silicon Nanowire Arrays and Nanoscale Detailed Cell-Nanowire Interaction Studies. Small 2013, 9, 263−272.
Pores. ACS Appl. Mater. Interfaces 2015, 7, 23717−23724. (75) Golshadi, M.; Wright, L. K.; Dickerson, I. M.; Schrlau, M. G.
(58) Elnathan, R.; Delalat, B.; Brodoceanu, D.; Alhmoud, H.; Harding, High-Efficiency Gene Transfection of Cells through Carbon Nanotube
F. J.; Buehler, K.; Nelson, A.; Isa, L.; Kraus, T.; Voelcker, N. H. Arrays. Small 2016, 12, 3014−3020.
Maximizing Transfection Efficiency of Vertically Aligned Silicon (76) Durney, A. R.; Frenette, L. C.; Hodvedt, E. C.; Krauss, T. D.;
Nanowire Arrays. Adv. Funct. Mater. 2015, 25, 7215−7225. Mukaibo, H. Fabrication of Tapered Microtube Arrays and Their

31075 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

Application as a Microalgal Injection Platform. ACS Appl. Mater. (97) Xu, A.; Shieh, P.; Sanman, L.; Melosh, N. Nanostraws: A
Interfaces 2016, 8, 34198−34208. Nanofabricated Platform for Delivery of Cell-Impermeable, Synthetic
(77) VanDersarl, J. J.; Xu, A. M.; Melosh, N. A. Nanostraws for Direct Biomolecules. Biophys. J. 2015, 108, 149a.
Fluidic Intracellular Access. Nano Lett. 2012, 12, 3881−3886. (98) Cao, Y.; Hjort, M.; Chen, H.; Birey, F.; Leal-Ortiz, S. A.; Han, C.
(78) Xu, A. M.; Kim, S. A.; Wang, D. S.; Aalipour, A.; Melosh, N. A. M.; Santiago, J. G.; Paşca, S. P.; Wu, J. C.; Melosh, N. A. Nondestructive
Temporally Resolved Direct Delivery of Second Messengers into Cells Nanostraw Intracellular Sampling for Longitudinal Cell Monitoring.
Using Nanostraws. Lab Chip 2016, 16, 2434−2439. Proc. Natl. Acad. Sci. U. S. A. 2017, 114, E1866−E1874.
(79) Xu, A. M.; Wang, D. S.; Shieh, P.; Cao, Y.; Melosh, N. A. Direct (99) Sun, M.; Duan, X. Recent Advances in Micro/Nanoscale
Intracellular Delivery of Cell-Impermeable Probes of Protein Intracellular Delivery. Nanotechnol. Precis. Eng. 2020, 3, 18−31.
Glycosylation by Using Nanostraws. ChemBioChem 2017, 18, 623− (100) Huang, Y.; Rubinsky, B. Flow-through Micro-Electroporation
628. Chip for High Efficiency Single-Cell Genetic Manipulation. Sens.
(80) He, G.; Chen, H.-J.; Liu, D.; Feng, Y.; Yang, C.; Hang, T.; Wu, J.; Actuators, A 2003, 104, 205−212.
Cao, Y.; Xie, X. Fabrication of Various Structures of Nanostraw Arrays (101) Potter, H.; Heller, R. Transfection by Electroporation. Curr.
and Their Applications in Gene Delivery. Adv. Mater. Interfaces 2018, 5, Protoc. Mol. Biol. 2018, 121, 9.3.1−9.3.13.
1701535. (102) Wang, S.; Lee, L. J. Micro-/Nanofluidics Based Cell
(81) He, G.; Yang, C.; Feng, J.; Wu, J.; Zhou, L.; Wen, R.; Huang, S.; Electroporation. Biomicrofluidics 2013, 7, 011301.
Wu, Q.; Liu, F.; Chen, H. J.; Hang, T.; Xie, X. Hierarchical Spiky (103) Chang, L. Q.; Li, L.; Shi, J. F.; Sheng, Y.; Lu, W.; Gallego-Perez,
Microstraws-Integrated Microfluidic Device for Efficient Capture and D.; Lee, L. J. Micro-/Nanoscale Electroporation. Lab Chip 2016, 16,
in Situ Manipulation of Cancer Cells. Adv. Funct. Mater. 2019, 29, 4047−4062.
1806484. (104) Lee, W. G.; Demirci, U.; Khademhosseini, A. Microscale
(82) Zhang, B.; Shi, Y.; Miyamoto, D.; Nakazawa, K.; Miyake, T. Electroporation: Challenges and Perspectives for Clinical Applications.
Nanostraw Membrane Stamping for Direct Delivery of Molecules into Integr. Biol. 2009, 1, 242−251.
Adhesive Cells. Sci. Rep. 2019, 9, 6806. (105) Geng, T.; Lu, C. Microfluidic Electroporation for Cellular
(83) Yu, Q.; Liu, H.; Chen, H. Vertical Sinwas for Biomedical and Analysis and Delivery. Lab Chip 2013, 13, 3803−3821.
Biotechnology Applications. J. Mater. Chem. B 2014, 2, 7849−7860. (106) Sun, C.; Ouyang, M.; Cao, Z.; Ma, S.; Alqublan, H.;
(84) Liu, X.; Wang, S. Three-Dimensional Nano-Biointerface as a Sriranganathan, N.; Wang, Y.; Lu, C. Electroporation-Delivered
New Platform for Guiding Cell Fate. Chem. Soc. Rev. 2014, 43, 2385− Fluorescent Protein Biosensors for Probing Molecular Activities in
2401. Cells without Genetic Encoding. Chem. Commun. 2014, 50, 11536−
(85) Schmidt, V.; Wittemann, J. V.; Senz, S.; Gösele, U. Silicon 11539.
Nanowires: A Review on Aspects of Their Growth and Their Electrical (107) Sun, C.; Cao, Z.; Wu, M.; Lu, C. Intracellular Tracking of Single
Properties. Adv. Mater. 2009, 21, 2681−2702. Native Molecules with Electroporation-Delivered Quantum Dots. Anal.
(86) Schmidt, V.; Wittemann, J. V.; Gösele, U. Growth, Thermody- Chem. 2014, 86, 11403−11409.
namics, and Electrical Properties of Silicon Nanowires. Chem. Rev. (108) Boukany, P. E.; Morss, A.; Liao, W.-c.; Henslee, B.; Jung, H.;
2010, 110, 361−388. Zhang, X.; Yu, B.; Wang, X.; Wu, Y.; Li, L.; Gao, K.; Hu, X.; Zhao, X.;
(87) Dasgupta, N. P.; Sun, J.; Liu, C.; Brittman, S.; Andrews, S. C.;
Hemminger, O.; Lu, W.; Lafyatis, G. P.; Lee, L. J. Nanochannel
Lim, J.; Gao, H.; Yan, R.; Yang, P. 25th Anniversary Article:
Electroporation Delivers Precise Amounts of Biomolecules into Living
Semiconductor Nanowires–Synthesis, Characterization, and Applica-
Cells. Nat. Nanotechnol. 2011, 6, 747−754.
tions. Adv. Mater. 2014, 26, 2137−2184.
(109) Gao, K.; Li, L.; He, L.; Hinkle, K.; Wu, Y.; Ma, J.; Chang, L.;
(88) Ronaghi, M.; Erceg, S.; Moreno-Manzano, V.; Stojkovic, M.
Zhao, X.; Perez, D. G.; Eckardt, S.; McLaughlin, J.; Liu, B.; Farson, D.
Challenges of Stem Cell Therapy for Spinal Cord Injury: Human
F.; Lee, L. J. Design of a Microchannel-Nanochannel-Microchannel
Embryonic Stem Cells, Endogenous Neural Stem Cells, or Induced
Pluripotent Stem Cells? Stem Cells 2010, 28, 93−99. Array Based Nanoelectroporation System for Precise Gene Trans-
(89) Maherali, N.; Hochedlinger, K. Guidelines and Techniques for fection. Small 2014, 10, 1015−1023.
the Generation of Induced Pluripotent Stem Cells. Cell Stem Cell 2008, (110) Valero, A.; Post, J. N.; van Nieuwkasteele, J. W.; Ter Braak, P.
3, 595−605. M.; Kruijer, W.; van den Berg, A. Gene Transfer and Protein Dynamics
(90) Gonzalez, F.; Boue, S.; Izpisua Belmonte, J. C. Methods for in Stem Cells Using Single Cell Electroporation in a Microfluidic
Making Induced Pluripotent Stem Cells: Reprogramming a La Carte. Device. Lab Chip 2008, 8, 62−67.
Nat. Rev. Genet. 2011, 12, 231−242. (111) Ouyang, M.; Hill, W.; Lee, J. H.; Hur, S. C. Microscale
(91) Rosenberg, S. A.; Restifo, N. P. Adoptive Cell Transfer as Symmetrical Electroporator Array as a Versatile Molecular Delivery
Personalized Immunotherapy for Human Cancer. Science 2015, 348, System. Sci. Rep. 2017, 7, 44757.
62−68. (112) Huang, H.; Wei, Z.; Huang, Y.; Zhao, D.; Zheng, L.; Cai, T.;
(92) Persson, H.; Beech, J. P.; Samuelson, L.; Oredsson, S.; Prinz, C. Wu, M.; Wang, W.; Ding, X.; Zhou, Z.; Du, Q.; Li, Z.; Liang, Z. An
N.; Tegenfeldt, J. O. Vertical Oxide Nanotubes Connected by Efficient and High-Throughput Electroporation Microchip Applicable
Subsurface Microchannels. Nano Res. 2012, 5, 190−198. for Sirna Delivery. Lab Chip 2011, 11, 163−172.
(93) Xu, A. M.; Aalipour, A.; Melosh, N. A. Nanostraw-Mediated (113) Li, Y.; Wu, M.; Zhao, D.; Wei, Z.; Zhong, W.; Wang, X.; Liang,
Intracellular Delivery: Direct Observation of Cell/Nanotube Interfaces. Z.; Li, Z. Electroporation on Microchips: The Harmful Effects of Ph
Biophys. J. 2013, 104, 194a. Changes and Scaling Down. Sci. Rep. 2015, 5, 17817.
(94) Bonde, S.; Buch-Manson, N.; Rostgaard, K. R.; Andersen, T. K.; (114) Lin, Y. C.; Li, M.; Wu, C. C. Simulation and Experimental
Berthing, T.; Martinez, K. L. Exploring Arrays of Vertical One- Demonstration of the Electric Field Assisted Electroporation Micro-
Dimensional Nanostructures for Cellular Investigations. Nanotechnol- chip for in Vitro Gene Delivery Enhancement. Lab Chip 2004, 4, 104−
ogy 2014, DOI: 10.1088/0957-4484/25/36/362001. 108.
(95) Fox, C. B.; Cao, Y.; Nemeth, C. L.; Chirra, H. D.; Chevalier, R. (115) Xu, Y.; Su, S.; Zhou, C.; Lu, Y.; Xing, W. Cell Electroporation
W.; Xu, A. M.; Melosh, N. A.; Desai, T. A. Fabrication of Sealed with a Three-Dimensional Microelectrode Array on a Printed Circuit
Nanostraw Microdevices for Oral Drug Delivery. ACS Nano 2016, 10, Board. Bioelectrochemistry 2015, 102, 35−41.
5873−5881. (116) Choi, S. O.; Kim, Y. C.; Lee, J. W.; Park, J. H.; Prausnitz, M. R.;
(96) Xu, A.; Kim, S. A.; Aalipour, A.; Melosh, N. A. Rapid Calcium Allen, M. G. Intracellular Protein Delivery and Gene Transfection by
Modulation in Cells: Direct Intracellular Access Using Nanostraws. Electroporation Using a Microneedle Electrode Array. Small 2012, 8,
Biophys. J. 2014, 106, 432a. 1081−1091.

31076 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

(117) Riaz, K.; Leung, S. F.; Fan, Z. Y.; Lee, Y. K. Electric Field Arrays from DNA Combing and Imprinting. Adv. Mater. 2010, 22,
Enhanced 3d Scalable Low-Voltage Nano-Spike Electroporation 3997−4001.
System. Sens. Actuators, A 2017, 255, 10−20. (136) Dwyer, R. M.; Khan, S.; Barry, F. P.; O’Brien, T.; Kerin, M. J.
(118) Fei, Z.; Wang, S.; Xie, Y.; Henslee, B. E.; Koh, C. G.; Lee, L. J. Advances in Mesenchymal Stem Cell-Mediated Gene Therapy for
Gene Transfection of Mammalian Cells Using Membrane Sandwich Cancer. Stem Cell Res. Ther. 2010, 1, 25.
Electroporation. Anal. Chem. 2007, 79, 5719−5722. (137) Amit, M.; Carpenter, M. K.; Inokuma, M. S.; Chiu, C.-P.; Harris,
(119) Fei, Z.; et al. Micronozzle Array Enhanced Sandwich C. P.; Waknitz, M. A.; Itskovitz-Eldor, J.; Thomson, J. A. Clonally
Electroporation of Embryonic Stem Cells. Anal. Chem. 2010, 82, Derived Human Embryonic Stem Cell Lines Maintain Pluripotency
353−358. and Proliferative Potential for Prolonged Periods of Culture. Dev. Biol.
(120) Chang, L.; Howdyshell, M.; Liao, W. C.; Chiang, C. L.; Gallego- 2000, 227, 271−278.
Perez, D.; Yang, Z.; Lu, W.; Byrd, J. C.; Muthusamy, N.; Lee, L. J.; (138) Goffinet, C.; Keppler, O. T. Efficient Nonviral Gene Delivery
Sooryakumar, R. Magnetic Tweezers-Based 3d Microchannel Electro- into Primary Lymphocytes from Rats and Mice. FASEB J. 2006, 20,
poration for High-Throughput Gene Transfection in Living Cells. 500−502.
Small 2015, 11, 1818−1828. (139) Johnson, L. A.; June, C. H. Driving Gene-Engineered T Cell
(121) Fei, Z.; Wu, Y.; Sharma, S.; Gallego-Perez, D.; Higuita-Castro, Immunotherapy of Cancer. Cell Res. 2017, 27, 38−58.
N.; Hansford, D.; Lannutti, J. J.; Lee, L. J. Gene Delivery to Cultured (140) Harrison, G. S.; Wang, Y.; Tomczak, J.; Hogan, C.; Shpall, E. J.;
Embryonic Stem Cells Using Nanofiber-Based Sandwich Electro- Curiel, T. J.; Felgner, P. L. Optimization of Gene Transfer Using
poration. Anal. Chem. 2013, 85, 1401−1407. Cationic Lipids in Cell Lines and Primary Human CD4+ and CD34+
(122) Ishibashi, T.; Takoh, K.; Kaji, H.; Abe, T.; Nishizawa, M. A Hematopoietic Cells. Biotechniques 1995, 19, 816−823.
Porous Membrane-Based Culture Substrate for Localized in Situ (141) Ivanov, I. T.; Brähler, M.; Georgieva, R.; Bäumler, H. Role of
Electroporation of Adherent Mammalian Cells. Sens. Actuators, B 2007, Membrane Proteins in Thermal Damage and Necrosis of Red Blood
128, 5−11. Cells. Thermochim. Acta 2007, 456, 7−12.
(123) Takoh, K.; Takahashi, A.; Matsue, T.; Nishizawa, M. A Porous (142) Delcea, M.; Sternberg, N.; Yashchenok, A. M; Georgieva, R.;
Membrane-Based Microelectroanalytical Technique for Evaluating Bäumler, H.; Möhwald, H.; Skirtach, A. G. Nanoplasmonics for Dual-
Locally Stimulated Culture Cells. Anal. Chim. Acta 2004, 522, 45−49. Molecule Release through Nanopores in the Membrane of Red Blood
(124) Kang, W.; Giraldo-Vela, J. P.; Nathamgari, S. S.; McGuire, T.; Cells. ACS Nano 2012, 6, 4169−4180.
McNaughton, R. L.; Kessler, J. A.; Espinosa, H. D. Microfluidic Device (143) Xiong, R.; Samal, S. K.; Demeester, J.; Skirtach, A. G.; De
for Stem Cell Differentiation and Localized Electroporation of Smedt, S. C.; Braeckmans, K. Laser-Assisted Photoporation:
Postmitotic Neurons. Lab Chip 2014, 14, 4486−4495. Fundamentals, Technological Advances and Applications. Adv. Phys.:
(125) Dong, Z.; Jiao, Y.; Xie, B.; Hao, Y.; Wang, P.; Liu, Y.; Shi, J.; X 2016, 1, 596−620.
(144) Lapotko, D. Optical Excitation and Detection of Vapor Bubbles
Chitrakar, C.; Black, S.; Wang, Y.-C.; Lee, L. J.; Li, M.; Fan, Y.; Chang,
around Plasmonic Nanoparticles. Opt. Express 2009, 17, 2538.
L. On-Chip Multiplexed Single-Cell Patterning and Controllable
(145) Siems, A.; Weber, S. A. L.; Boneberg, J.; Plech, A.
Intracellular Delivery. Microsyst. Nanoeng. 2020, 6, 2.
Thermodynamics of Nanosecond Nanobubble Formation at Laser-
(126) Mukherjee, P.; Nathamgari, S. S. P.; Kessler, J. A.; Espinosa, H.
Excited Metal Nanoparticles. New J. Phys. 2011, 13, 043018.
D. Combined Numerical and Experimental Investigation of Localized
(146) Lukianova-Hleb, E. Y.; Wagner, D. S.; Brenner, M. K.; Lapotko,
Electroporation-Based Cell Transfection and Sampling. ACS Nano D. O. Cell-Specific Transmembrane Injection of Molecular Cargo with
2018, 12, 12118−12128. Gold Nanoparticle-Generated Transient Plasmonic Nanobubbles.
(127) Chang, L.; Bertani, P.; Gallego-Perez, D.; Yang, Z.; Chen, F.; Biomaterials 2012, 33, 5441−5450.
Chiang, C.; Malkoc, V.; Kuang, T.; Gao, K.; Lee, L. J.; Lu, W. 3d (147) Fraire, J. C.; Houthaeve, G.; Liu, J.; Raes, L.; Vermeulen, L.;
Nanochannel Electroporation for High-Throughput Cell Transfection Stremersch, S.; Brans, T.; Garcia-Diaz Barriga, G.; De Keulenaer, S.;
with High Uniformity and Dosage Control. Nanoscale 2016, 8, 243− Van Nieuwerburgh, F.; De Rycke, R.; Vandesompele, J.; Mestdagh, P.;
252. Raemdonck, K.; De Vos, W. H.; De Smedt, S.; Braeckmans, K. Vapor
(128) Cao, Y.; Ma, E.; Cestellos-Blanco, S.; Zhang, B.; Qiu, R.; Su, Y.; Nanobubble Is the More Reliable Photothermal Mechanism for
Doudna, J. A.; Yang, P. Nontoxic Nanopore Electroporation for Inducing Endosomal Escape of Sirna without Disturbing Cell
Effective Intracellular Delivery of Biological Macromolecules. Proc. Homeostasis. J. Controlled Release 2020, 319, 262−275.
Natl. Acad. Sci. U. S. A. 2019, 116, 7899−7904. (148) Wayteck, L.; Xiong, R.; Braeckmans, K.; De Smedt, S. C.;
(129) Xie, X.; Xu, A. M.; Leal-Ortiz, S.; Cao, Y.; Garner, C. C.; Melosh, Raemdonck, K. Comparing Photoporation and Nucleofection for
N. A. Nanostraw-Electroporation System for Highly Efficient Intra- Delivery of Small Interfering Rna to Cytotoxic T Cells. J. Controlled
cellular Delivery and Transfection. ACS Nano 2013, 7, 4351−4358. Release 2017, 267, 154−162.
(130) Cao, Y.; Chen, H.; Qiu, R.; Hanna, M.; Ma, E.; Hjort, M.; (149) Wu, T.-H.; et al. Image Patterned Molecular Delivery into Live
Zhang, A.; Lewis, R. S.; Wu, J. C.; Melosh, N. A. Universal Intracellular Cells Using Gold Particle Coated Substrates. Opt. Express 2010, 18,
Biomolecule Delivery with Precise Dosage Control. Sci. Adv. 2018, 4, 938−946.
No. eaat8131. (150) Lyu, Z.; Zhou, F.; Liu, Q.; Xue, H.; Yu, Q.; Chen, H. A Universal
(131) He, G.; Feng, J. M.; Zhang, A. H.; Zhou, L. F.; Wen, R.; Wu, J. Platform for Macromolecular Delivery into Cells Using Gold
M.; Yang, C. D.; Yang, J.; Li, C. W.; Chen, D. M.; Wang, J.; Hu, N.; Xie, Nanoparticle Layers Via the Photoporation Effect. Adv. Funct. Mater.
X. Multifunctional Branched Nanostraw-Electroporation Platform for 2016, 26, 5787−5795.
Intracellular Regulation and Monitoring of Circulating Tumor Cells. (151) Wu, J.; Xue, H.; Lyu, Z.; Li, Z.; Qu, Y.; Xu, Y.; Wang, L.; Yu, Q.;
Nano Lett. 2019, 19, 7201−7209. Chen, H. Intracellular Delivery Platform for “Recalcitrant” Cells: When
(132) Wen, R.; Zhang, A.-h.; Liu, D.; Feng, J.; Yang, J.; Xia, D.; Wang, Polymeric Carrier Marries Photoporation. ACS Appl. Mater. Interfaces
J.; Li, C.; Zhang, T.; Hu, N.; Hang, T.; He, G.; Xie, X. Intracellular 2017, 9, 21593−21598.
Delivery and Sensing System Based on Electroplated Conductive (152) Zheng, Y.; Wu, Y.; Zhou, Y.; Wu, J.; Wang, X.; Qu, Y.; Wang, Y.;
Nanostraw Arrays. ACS Appl. Mater. Interfaces 2019, 11, 43936−43948. Zhang, Y.; Yu, Q. Photothermally Activated Electrospun Nanofiber
(133) Movahed, S.; Li, D. Microfluidics Cell Electroporation. Mats for High-Efficiency Surface-Mediated Gene Transfection. ACS
Microfluid. Nanofluid. 2011, 10, 703−734. Appl. Mater. Interfaces 2020, 12, 7905−7914.
(134) Santra, T. S.; Tseng, F. G. Recent Trends on Micro/Nanofluidic (153) Kang, T. H.; Lee, S.; Kwon, J. A.; Song, J.; Choi, I.
Single Cell Electroporation. Micromachines 2013, 4, 333−356. Photothermally Enhanced Molecular Delivery and Cellular Positioning
(135) Guan, J.; Boukany, P. E.; Hemminger, O.; Chiou, N. R.; Zha, on Patterned Plasmonic Interfaces. ACS Appl. Mater. Interfaces 2019,
W.; Cavanaugh, M.; Lee, L. J. Large Laterally Ordered Nanochannel 11, 36420−36427.

31077 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078
ACS Applied Materials & Interfaces www.acsami.org Review

(154) Messina, G. C.; Dipalo, M.; La Rocca, R.; Zilio, P.; Caprettini, (172) Kwon, I. S.; Bettinger, C. J. Polydopamine Nanostructures as
V.; Proietti Zaccaria, R.; Toma, A.; Tantussi, F.; Berdondini, L.; De Biomaterials for Medical Applications. J. Mater. Chem. B 2018, 6,
Angelis, F. Spatially, Temporally, and Quantitatively Controlled 6895−6903.
Delivery of Broad Range of Molecules into Selected Cells through (173) Ryu, J. H.; Messersmith, P. B.; Lee, H. Polydopamine Surface
Plasmonic Nanotubes. Adv. Mater. 2015, 27, 7145−7149. Chemistry: A Decade of Discovery. ACS Appl. Mater. Interfaces 2018,
(155) Wang, J.; Ren, K.-F.; Gao, Y.-F.; Zhang, H.; Huang, W.-P.; Qian, 10, 7523−7540.
H.-L.; Xu, Z.-K.; Ji, J. Photothermal Spongy Film for Enhanced Surface- (174) Kobayashi, J.; Yamato, M.; Okano, T. On-Off Affinity Binding
Mediated Transfection to Primary Cells. ACS Appl. Bio Mater. 2019, 2, Modulation on Thermoresponsive Polymer-Grafted Surfaces for
2676−2684. Capture and Release of Proteins and Cells. J. Biomater. Sci., Polym.
(156) Zhang, H.; Wang, J.; Hu, M.; Li, B.-c.; Li, H.; Chen, T.-t.; Ren, Ed. 2017, 28, 939−957.
K.-F.; Ji, J.; Jing, Q.-m.; Fu, G.-s. Photothermal-Assisted Surface- (175) Wang, J.; Wei, Y.; Zhao, S.; Zhou, Y.; He, W.; Zhang, Y.; Deng,
Mediated Gene Delivery for Enhancing Transfection Efficiency. W. The Analysis of Viability for Mammalian Cells Treated at Different
Biomater. Sci. 2019, 7, 5177−5186. Temperatures and Its Application in Cell Shipment. PLoS One 2017,
(157) Wu, J.; Zheng, Y.; Jiang, S.; Qu, Y.; Wei, T.; Zhan, W.; Wang, L.; 12, No. e0176120.
Yu, Q.; Chen, H. Two-in-One Platform for High-Efficiency Intra- (176) Wei, T.; Yu, Q.; Zhan, W.; Chen, H. A Smart Antibacterial
cellular Delivery and Cell Harvest: When a Photothermal Agent Meets Surface for the on-Demand Killing and Releasing of Bacteria. Adv.
a Thermoresponsive Polymer. ACS Appl. Mater. Interfaces 2019, 11, Healthcare Mater. 2016, 5, 449−456.
12357−12366. (177) Matsumoto, A.; Sato, N.; Kataoka, K.; Miyahara, Y. Noninvasive
(158) Wang, L.; Wu, J.; Hu, Y.; Hu, C.; Pan, Y.; Yu, Q.; Chen, H. Sialic Acid Detection at Cell Membrane by Using Phenylboronic Acid
Using Porous Magnetic Iron Oxide Nanomaterials as a Facile Modified Self-Assembled Monolayer Gold Electrode. J. Am. Chem. Soc.
Photoporation Nanoplatform for Macromolecular Delivery. J. Mater. 2009, 131, 12022−12023.
Chem. B 2018, 6, 4427−4436. (178) Van Tendeloo, V. F.; Willems, R.; Ponsaerts, P.; Lenjou, M.;
(159) Courvoisier, S.; Saklayen, N.; Huber, M.; Chen, J.; Diebold, E. Nijs, G.; Vanhove, M.; Muylaert, P.; Cauwelaert, P. V.; Broeckhoven, C.
D.; Bonacina, L.; Wolf, J. P.; Mazur, E. Plasmonic Tipless Pyramid V.; Bockstaele, D. V.; Berneman, Z. High-Level Transgene Expression
Arrays for Cell Poration. Nano Lett. 2015, 15, 4461−4466. in Primary Human T Lymphocytes and Adult Bone Marrow Cd34+
(160) Saklayen, N.; Huber, M.; Madrid, M.; Nuzzo, V.; Vulis, D. I.; Cells Via Electroporation-Mediated Gene Delivery. Gene Ther. 2000, 7,
Shen, W.; Nelson, J.; McClelland, A. A.; Heisterkamp, A.; Mazur, E. 1431−1437.
Intracellular Delivery Using Nanosecond-Laser Excitation of Large- (179) Mizrahy, S.; Hazan-Halevy, I.; Dammes, N.; Landesman-Milo,
Area Plasmonic Substrates. ACS Nano 2017, 11, 3671−3680. D.; Peer, D. Current Progress in Non-Viral Rnai-Based Delivery
(161) Raun, A.; Saklayen, N.; Zgrabik, C.; Shen, W.; Madrid, M.; Strategies to Lymphocytes. Mol. Ther. 2017, 25, 1491−1500.
Huber, M.; Hu, E.; Mazur, E. A Comparison of Inverted and Upright (180) Zhan, W.; Qu, Y.; Wei, T.; Hu, C.; Pan, Y.; Yu, Q.; Chen, H.
Laser-Activated Titanium Nitride Micropyramids for Intracellular Sweet Switch: Sugar-Responsive Bioactive Surfaces Based on Dynamic
Delivery. Sci. Rep. 2018, 8, 15595. Covalent Bonding. ACS Appl. Mater. Interfaces 2018, 10, 10647−10655.
(162) Wu, Y. C.; Wu, T. H.; Clemens, D. L.; Lee, B. Y.; Wen, X.; (181) Zhou, Y.; Zheng, Y.; Wei, T.; Qu, Y.; Wang, Y.; Zhan, W.;
Horwitz, M. A.; Teitell, M. A.; Chiou, P. Y. Massively Parallel Delivery Zhang, Y.; Pan, G.; Li, D.; Yu, Q.; Chen, H. Multistimulus Responsive
of Large Cargo into Mammalian Cells with Light Pulses. Nat. Methods Biointerfaces with Switchable Bioadhesion and Surface Functions. ACS
2015, 12, 439−444. Appl. Mater. Interfaces 2020, 12, 5447−5455.
(163) Man, T.; Zhu, X.; Chow, Y. T.; Dawson, E. R.; Wen, X.;
Patananan, A. N.; Liu, T. L.; Zhao, C.; Wu, C.; Hong, J. S.; Chung, P. S.;
Clemens, D. L.; Lee, B. Y.; Weiss, P. S.; Teitell, M. A.; Chiou, P. Y.
Intracellular Photothermal Delivery for Suspension Cells Using Sharp
Nanoscale Tips in Microwells. ACS Nano 2019, 13, 10835−10844.
(164) Qu, Y.; Zheng, Y.; Yu, L.; Zhou, Y.; Wang, Y.; Zhang, Y.; Yu, Q.;
Chen, H. A Universal Platform for High-Efficiency “Engineering”
Living Cells: Integration of Cell Capture, Intracellular Delivery of
Biomolecules, and Cell Harvesting Functions. Adv. Funct. Mater. 2020,
30, 1906362.
(165) Qin, Z.; Bischof, J. C. Thermophysical and Biological Responses
of Gold Nanoparticle Laser Heating. Chem. Soc. Rev. 2012, 41, 1191−
1217.
(166) Zhou, F.; Yuan, L.; Wang, H.; Li, D.; Chen, H. Gold
Nanoparticle Layer: A Promising Platform for Ultra-Sensitive Cancer
Detection. Langmuir 2011, 27, 2155−2158.
(167) Zhou, F.; Wang, M.; Yuan, L.; Cheng, Z.; Wu, Z.; Chen, H.
Sensitive Sandwich Elisa Based on a Gold Nanoparticle Layer for
Cancer Detection. Analyst 2012, 137, 1779−1784.
(168) Qu, Y.; Wei, T.; Zhao, J.; Jiang, S.; Yang, P.; Yu, Q.; Chen, H.
Regenerable Smart Antibacterial Surfaces: Full Removal of Killed
Bacteria Via a Sequential Degradable Layer. J. Mater. Chem. B 2018, 6,
3946−3955.
(169) Wu, J.; Qu, Y.; Yu, Q.; Chen, H. Gold Nanoparticle Layer: A
Versatile Nanostructured Platform for Biomedical Applications. Mater.
Chem. Front. 2018, 2, 2175−2190.
(170) Haine, A. T.; Niidome, T. Gold Nanorods as Nanodevices for
Bioimaging, Photothermal Therapeutics, and Drug Delivery. Chem.
Pharm. Bull. 2017, 65, 625−628.
(171) Batul, R.; Tamanna, T.; Khaliq, A.; Yu, A. Recent Progress in the
Biomedical Applications of Polydopamine Nanostructures. Biomater.
Sci. 2017, 5, 1204−1229.

31078 https://dx.doi.org/10.1021/acsami.0c06978
ACS Appl. Mater. Interfaces 2020, 12, 31054−31078

You might also like