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

REVIEW

www.advnanobiomedres.com

Cellular Ingestible DNA Nanostructures for Biomedical


Applications
Rui Xu, Yujie Li, Chenyou Zhu, Dongsheng Liu,* and Yuhe R. Yang*

(LNPs) have been approved to be an effec-


Delivering functional substances across the cell membrane barriers has been a tive carrier for mRNA vaccines.[3] However,
great challenge in biomedicine. Among various strategies to improve cellular different carrier materials have correspond-
uptake, DNA nanostructures, with different shapes and sizes, supply as suitable ing drawbacks. For example, the potential
toxicity of synthetic nanoparticles cannot
platforms for delivering a wide range of functional substances to cells. With great
be ignored due to their nondegradability
programmability, biocompatibility, and biostability, DNA nanostructure-based in vivo,[4] and the limited DNA packaging
delivery systems have shown and have succeeded in anticancer therapy, anti- capacity and carcinogenesis restricted
bacterial treatment, gene editing, vaccine development, and stem cell bioengi- further biomedical applications of viral
neering. Herein, the development and construction of various cellular ingestible vectors.[5] Thus, it is urgent to expand the
pool of carrier materials with excellent cell
DNA nanostructures are described, the loading strategies of functional sub-
permeability, high loading capacity, and
stances are summarized, current biomedical applications are overviewed, and biocompatibility.
finally the remaining challenges and opportunities are discussed. DNA nanotechnology, an emerging
technique that utilizes the DNA as building
blocks to program well-defined nanostruc-
tures, is considered as a suitable option for
1. Introduction drug delivery due to the intrinsic biocompatibility, programma-
bility, and biostability of DNA nanostructures. Since the concept
While the cell membrane functions as a barrier that separates of DNA nanotechnology was proposed by Seeman in 1982,[6] this
harmful foreign substances from the cytoplasm, it also prevents technology has been rapidly developing. Previous studies have
the therapeutic substances from cellular entry. Functional sub- demonstrated that some DNA nanostructures can be actively
stances (i.e., small molecule drugs, siRNA, Cas9/gRNA, etc.) absorbed by cells, which have attracted extensive attention for
need to penetrate the cell membrane barrier to realize efficient their wide range of applications such as biosensing, tissue cul-
intracellular functions.[1] Popular delivery vehicles for the import ture, drug delivery, etc.[7] In this review, we will focus on cellular
of various functional substances into cells include inorganic ingestible DNA nanostructure-based functional materials and
nanomaterials (i.e., Au nanoparticles, carbon nanomaterials), their biomedical applications. Within this framework, we compre-
polymer materials (i.e., liposome, chitosan), and biomimetic hensively review the development, construction strategy, and
materials (i.e., viral vector).[2] For instance, lipid nanoparticles biomedical applications of DNA nanostructures with particular
emphasis on DNA-functionalized nanoparticles, DNA microgel,
and programmable DNA self-assembly nanostructures. We
R. Xu, Y. Li, C. Zhu, D. Liu mainly discuss their application prospects and current challenges
Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology
and further propose several improvement strategies for broader
(Ministry of Education)
Department of Chemistry therapeutic applications. Owing to space constraints, DNA nano-
Tsinghua University structures working extracellularly or in vitro will not be discussed
Beijing 100084, China in this review but are touched on in companion reviews.[8]
E-mail: liudongsheng@tsinghua.edu.cn
Y. R. Yang
CAS Key Laboratory of Nanosystem and Hierarchical Fabrication 2. Development and Construction
CAS Center for Excellence in Nanoscience
National Center for Nanoscience and Technology
Beijing 100190, China In past decades, numerous efforts have been devoted to the devel-
E-mail: yangyh@nanoctr.cn opment and improvement of manufacture strategies of different
DNA nanostructures. Here we analyze pros and cons for each strat-
The ORCID identification number(s) for the author(s) of this article
can be found under https://doi.org/10.1002/anbr.202200119. egy and describe the reaction time, yield, scale, cost, etc. as follows.

© 2022 The Authors. Advanced NanoBiomed Research published by


Wiley-VCH GmbH. This is an open access article under the terms of 2.1. DNA-Functionalized Nanoparticles
the Creative Commons Attribution License, which permits use,
distribution and reproduction in any medium, provided the original Au/Ag─S bond is widely employed to mediate ssDNA–metal
work is properly cited. nanoparticle conjugates. In 1996, Mirkin and Alivisatos indepen-
DOI: 10.1002/anbr.202200119 dently reported the synthetic approach for DNA-functionalized

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (1 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

gold nanoparticles (AuNPs) for the first time.[9] Subsequently, mixture at 20 °C for 2 h and then thawed the mixture at room
various nanoparticles have been functionalized by DNA and temperature.[12] This method can increase the surface density of
obtained different applications. Herein, we focused on the syn- DNA up to 110 DNA per 13 nm AuNP. Moreover, based on
thesis of DNA-functionalized AuNPs for their broad applications DNA-functionalized AuNPs, the DNA with specific functions (i.e.,
in biomedicine field. i-motif,[13] a structure responding to pH,[14] electric signal,[15]
Various efforts have been used to improve the synthesis of light,[16] etc.) has been employed to modify AuNPs and
DNA-functionalized AuNPs in efficiency of process and density achieved pH-responsive assembly.[17] Number-controlled DNA-
of DNA modification (Figure 1A). The high-density DNA- functionalized AuNPs have also been developed for programma-
functionalized AuNP was achieved by Mirkin et al.[10] using a ble assembly of AuNPs into well-defined superstructures.[18]
method referred as salt aging. They successfully promoted higher Furthermore, similar covalent conjugation approaches have been
surface density of DNA (up to 90 DNA per 13 nm AuNP) by established, which utilize dibenzocyclooctyne-modified DNA to
increasing the concentration of sodium chloride (NaCl) in reac- functionalize azide-modified liposomal or protein and apply
tion solution up to 2 mol L1 to prevent the electrostatic exclu- tocopherol modified to functionalize liposome via hydrophobic
sion between the AuNPs and DNA strands. However, this salt interaction.[19]
aging method requires over 24 h for reaction; it has been opti-
mized by Liu et al. with a low-pH method to enable rapid synthesis 2.2. DNA Microgels
(only 5 min) of high-density DNA-functionalized AuNPs.[11] To
further enhance the surface density and modification efficiency, DNA microgels, a nanoscale version of DNA hydrogels, have
Liu et al. additionally introduced a method that froze the reaction been developed to improve cellular applicability due to its

Figure 1. DNA-functionalized AuNP and DNA microgels. A) Three main methods for constructing DNA-functionalized AuNP. B) DNA hydrogels con-
structed by Y scaffold and linker. Reproduced with permission.[24] Copyright 2011, Wiley-VCH. C) DNA nanoflower constructed by long DNA chains.
Reproduced with permission.[33] Copyright 2013, American Chemical Society.

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (2 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

discrete size, high stimuli–response rate, and cellular uptake effi- including stimuli responsiveness, self-healing, drug sustained
ciency.[20] The current design of DNA microgels is built on the release, which provides DNA hydrogels with high potential
formation strategies of DNA hydrogels. Herein, we reviewed the application value, especially in the field of biomedicine.[32]
development of DNA hydrogels first and discussed the construc- Based on the generation methods of DNA hydrogels, DNA
tion of DNA hydrogels and DNA microgels. microgels similarly can also be composed by the tangle of
DNA hydrogels can be constructed through chemical or phys- long DNA chains and self-assembly of DNA building blocks.
ical interaction among DNA-based building blocks. In 2006, Luo Tan et al. used long DNA chains prepared via RCA to construct
et al. initially prepared chemically crosslinked DNA hydrogel by a DNA nanoflower with annealing process (Figure 1C).[33]
DNA assembly and ligation.[21] The reformation of duplexes in Furthermore, Gu et al. reported kinds of DNA microgels called
rehydration process endowed the hydrogels with amazing shape DNA nanoclews (NCs) which also adopted RCA process.[34] The
memory properties, but also limited their structural dynamic.[22] sizes of the DNA nanoflower and NC were generally more than
Luo et al. elongated DNA chains through rolling circle amplifi- 100 nm. More recently, Tan et al. also used Y unit and linker to
cation (RCA) and multiprimed chain amplification (MCA) and synthesize self-assembled DNA microgels with a well-defined net-
then weaved them noncovalently into hydrogels.[23] This strategy work. They employed an aptamer to block the Y-unit and inhibit
provides high convenience for the synthesis of a large quantity of the extension of nanostructures, which can regulate the size of
DNA hydrogels without the requirement to design specific DNA nanohydrogels from 100, 144, to 268 nm through controlling
sequence. Besides, Liu et al. developed a series of self- the proportion of the aptamer-blocked Y-unit and normal Y-unit.[35]
assembled-based DNA hydrogels with a programmable network.
In 2011, they designed self-assembled DNA hydrogels with 2.3. Programmable DNA Self-Assembly Nanostructures for
designable thermal and enzymatic responsiveness via the hybrid- Biomedical Applications
ization of two building blocks: a Y-scaffold and a linker
(Figure 1B).[24] Next, they dividedly heated Y-scaffold and linker 2.3.1. DNA Polyhedron
DNA strands to 95 °C and cooled down to room temperature after
2 h and then proportionally mixed two solutions, which allowed The synthesis of DNA polyhedron has experienced various
to obtain the DNA hydrogels through one-pot reaction. Based on evolutions. In 1991, Seeman et al. reported the first DNA poly-
this strategy, they established a series of stimuli–responsive DNA hedron, in which they constructed DNA cube in a DNA ring
hydrogels: photoresponsiveness, thermal responsiveness, and nested manner.[36] In 2004, two individual studies using
metal–ion responsiveness.[25,26] Recently, they constructed rein- ssDNA to assemble polyhedron structures in a one-pot annealing
forced DNA hydrogels through adding polymeric multiple-unit process were reported. Turberfield et al. utilized four ssDNA to
linkers to this system, which enabled strong improvement of the build DNA tetrahedron successfully (Figure 2A),[37] and
mechanical strength.[27] A similar strategy with Y-scaffold and Shih et al. created DNA octahedrons with a long ssDNA and five
i-motif was used to construct a pH-responsive hydrogel.[28] This short ssDNAs.[38] In 2008, Mao et al. described a new method of
method is also suitable for the construction of hybrid DNA hydro- constructing various kinds of DNA polyhedrons by linking
gels with peptides,[29] proteins,[30] cucurbit[8]uril,[31] etc. The DNA three (or four, five)-point-star motifs though sticky-ended
self-assembly-based strategy possess more specific properties, (Figure 2A).[39] They constructed DNA tetrahedrons, DNA

Figure 2. Programable DNA self-assembly nanostructures. A) Two strategies to produce DNA polyhedron. Reproduced with permission.[37,39] Copyright
2004, Royal Society of Chemistry, Copyright 2008, Nature Publishing Group. B) The design and construction of DNA origami. Reproduced with
permission.[41,44,70] Copyright 2006, Nature Publishing Group, Copyright 2009, The American Association for the Advancement of Science,
Copyright 2014, American Chemical Society. C) The method of SST to construct different shapes. Reproduced with permission.[51,52] Copyright 2012,
The American Association for the Advancement of Science, Copyright 2012, Nature Publishing Group.

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (3 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

dodecahedrons, and DNA buckyballs through regulating the DNA origami). Besides, the assembly procedure, scale, yield,
length of the central single-stranded loop from five-base to and other practical factors need to be considered comprehen-
three-base long. In the subsequent research, they employed star sively when selecting DNA nanostructures as targets in biomed-
motifs to create other polyhedrons, such as DNA cubes, DNA ical applications in vivo.
octahedrons, and DNA triangular prisms. In addition, in
2015, a new method for combination with the motif-based 2.3.3. Single-Stranded DNA Tiles
polyhedron assembly and circular long-stranded DNA origami
strategy was developed by Yan et al., namely, wireframe DNA The concept of single-stranded DNA tiles (SSTs) was proposed by
origami.[40] Similarly, the length of the central single-stranded Yin et al. in 2008.[50] An SST consists of a 42-base strand of DNA
loop also played a vital role in adjusting the size and structure entirely composed of concatenated sticky ends, binding to four
of the DNA polyhedrons. local neighbors during self-assembly (Figure 2C). Researchers
At present, DNA tetrahedrons were mainly constructed by have used SSTs to construct various complex shapes (e.g., DNA
four single-stranded DNA for its simple process and relatively triangle, DNA rectangle, and DNA tube). In 2012, Yin et al. also
high yield. With only one annealing step spending about 4 h, put forward the concept of DNA bricks which extended the
the DNA tetrahedrons could be obtained with a yield of 95%. SST assembly method from 2D to 3D.[51] A canonical DNA brick
DNA polyhedrons with more complex structures were usually consists of a 32-nt ssDNA with four 8-nt binding domains.
generated using star motifs. Depending on the size and complex- Each 32-nt brick is a modular component and binds to four local
ity, the annealing time varied from 4 h to more than 48 h whereas neighbors which can be removed or added independently.
the yield dropped from 90% (i.e., DNA tetrahedrons) to less than A unique feature of SST-based assembly is the unlimited size
69% (i.e., DNA buckyballs). The process of 12 h annealing was of the desired shape which was previously defined by the limited
necessitated for the generation of wireframe DNA origami. length of the scaffold DNA in DNA origami strategy. Purification
Usually involving a M13 scaffold strand, and the size of DNA and exact stoichiometry of DNA components are not essential in
polyhedrons varies from 50 nm to hundreds of nanometers with SST-based assembly. The annealing rate varies from 17 to 72 h
yield from 70% to 80%. Based on all the designs of polyhedron, depending on the structure of the final product as well as the
tradeoffs among the yield, assembly time, and cost, DNA tetra- yield differing greatly from 80% to 4%. The large range of size
hedrons are becoming the most widely used nanostructures in adjustment enables generation of nanostructures to micrometer
biomedical field nowadays for its broad applications, relatively scale with this SST method (around 10–200 nm, even up to
large-scale production, and facile synthesis process. 1 μm),[52] which compensates for the deficiency of the program-
mable nanomaterials described previously. However, the rela-
2.3.2. DNA Origami tively low yield and high cost of SST-based methods are the
major obstacles. Further development and improvement in these
DNA origami, a revolutionary method for constructing various aspects are required, which will expand their biomedical
unprecedented DNA structures with fantastic stability and applications.
extensive applications, was proposed by Rothemund in 2006
(Figure 2B).[41] With rigorous design, DNA origami with arbi-
trary shapes can theoretically be formed by a one-step annealing 3. Methods of Loading Various Functional
process which involves a long, ssDNA scaffold (e.g., M13mp18 Groups
genomic DNA) and hundreds of short oligonucleotide staple
strands. With the development of user-friendly software (e.g., In addition to the diverse compositions, sizes, and structures of
CADnano,[42] Tiamat[43]), a large number of nanostructures from DNA nanostructures, the unique modifiability of DNA enables
simple 2D rectangular shapes to complex 3D shapes (e.g., twisted them to carry a variety of target cargos of interest, including small
nanoscale shapes,[44] DNA boxes,[45] flower vase,[46] DNA molecules, nucleic acids, proteins, etc. Depending on the differ-
origamiþ,[47] square nut[48]) have been constructed using DNA ent types and properties of the functional groups, various loading
origami strategy. methods are categorized into covalent conjugation, intercalation,
The one-step annealing process and purification-free synthe- base pairing, and packaging, which will be described as follows.
sis of staple strands are relatively convenient. Due to the excess
addition of staples to the reaction mixture, stoichiometry reduces 3.1. Complementary Base Pairing
restrictions compared with other methods such as tile-based
assembly.[39] The annealing parameters for different DNA Nucleic acid-based drugs (i.e., mRNA, miRNA, siRNA, antisense
origami structures vary from 1.5 h for simple structures to more nucleic acid, aptamer) play an increasingly important role in
than 24 h for the complex structures. The yield also varies modern medicine aiming for cancer treatment, hyperglycemia,
dramatically from >70% to <4% with increasing complexity. antivirus, and other diseases.[53] All kinds of DNA nanostructures
DNA origami method has enabled the creation of fancy DNA can potentially be applied as the natural carriers for the
structures through simple delicate design that could replicate nucleic acid-based drugs through complementary base pairing
art pieces in nanometer scale (e.g., smiley,[41] Mona Lisa,[49]). (Figure 3A). A large number of studies have reported the appli-
However, until now, the DNA origami structures used to carry cations of various DNA nanostructures to load nucleic acid-based
materials into cells were limited to relatively simple structures drugs including DNA origami assemblies, DNA-functionalized
(i.e., triangle DNA origami, square DNA origami, and tube NPs, DNA microgels, and polyhedrons.[7] These nucleic acid

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (4 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

Figure 3. Methods of loading various functional groups. A) Complementary base pairing. B) Intercalation. C) Covalent Conjugation. D) FGA strategy for
multiple loading. Reproduced with permission.[64] Copyright 2017, Wiley-VCH.

nanostructures are mainly loaded with nucleic acid drugs in two six ssDNAs with six double-stranded siRNAs, in which the solu-
ways. 1) One-step method. By extending the nucleic acid-based tion was heated to 90 °C for 2 min and rapidly cooled down to
drug sequences as a part of the DNA nanostructures, they can be 4 °C.[54] 2) Two-step method. In this way, a short single-stranded
loaded into DNA nanostructures through one-pot annealing. In probe is extruded out of the DNA nanoparticle to hybridize with
spite of convenience, the process appears to be destructive for the part of the nucleic acid-based drugs. This relatively mild con-
some RNA-based drugs (i.e., mRNA, siRNA, miRNA) since dition (usually <37 °C) is not only suitable for RNA-based
the annealing procedure usually involves an initial step of high drugs,[55] but also enables the coloading of multiple drugs.[56]
temperature to denature all the DNA components. Therefore, However, the sequence of probe needs to be accurately designed
most of the reported works are used for loading more to form stable tethering while minimizing misfolding.[57]
thermostable DNA-based drugs (i.e., antisense DNA,[35] DNA
aptamer[33]). Some RNA drugs can accommodate assembly con- 3.2. Intercalation
ditions with short heating time and fast cooling rate for loading
onto easy-to-form DNA nanostructures. Anderson et al. described Intercalation is the most simple and convenient method which
the assembly of tetrahedron–siRNA complex by directly mixing only needs an incubation step of the drugs with corresponding

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (5 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

DNA nanostructures. Some small molecules, especially the which will avoid drug leakage and realize the gradual release
anthracyclines drugs containing an anthracene ring connected of drugs.
to an amino sugar, (e.g., doxorubicin, daunorubicin), have been
found to be able to intercalate between two base pairs of the DNA 3.5. Combination of Multiple Methods
(Figure 3B). Various DNA nanostructures carrying Dox have
been widely studied and applied to treat cancers benefiting from The loading methods described above can be further combined
their high efficiency to be loaded onto DNA nanostructures by to codeliver various functional materials to specific sites. Liu et al.
simply mixing.[58] Besides doxorubicin, a highly potent platinum- employed the frame-guided assembly (FGA) strategy to develop a
based DNA intercalator, 56MESS, can also be loaded into the modularly designable vesicle for sequential multiple loading
double-stranded DNA (dsDNA) with this method.[59] (Figure 3E).[64] With this vesicle, they accomplished programmat-
ically loading of four different functional substances through
layer-by-layer assembly. The FGA strategy provided a potential
3.3. Covalent Conjugation
approach for the delivery of multiple functional groups and
realized the combination of therapeutic function and tracking
Covalent conjugation is a universal method for loading
purpose.
small-molecular compounds (e.g., cisplatin drugs, paclitaxel),
nucleic acid drugs, and proteinaceous drugs on all types of
DNA nanostructures. 3’ end and 5’ end of ssDNA are the two 4. Biomedical Applications
common sites for modification. Commonly the used chemical
groups modified to the 3’end and 5’ end of DNA include amino DNA nanostructures have become a widely used material in bio-
(–NH3), sulfhydryl (–SH), azide (–N3), and alkyne (–C≡CH) medicine due to their programmability, biocompatibility, and
(Figure 3C). Mirkin et al. applied 28mer oligonucleotides con- bio-stability. 1) Programmability: As described above, the size
taining a terminal dodecyl amine to conjugate with a platinum of DNA nanostructures ranges from a few nanometers to micro-
(IV) complex.[60] Besides the modification on 3’ end and 5’ end of meters, with diverse shape, which endows DNA nanomaterials
DNA, the phosphodiester linker between each nucleotide can with the different pathways of cellular uptake as well as distinct
also be modified with phosphorothioate (PS) in which one of targeting and trafficking behavior in animal models.[7,65]
the oxygen atoms (O) is replaced by sulfur atom (S) to improve 2) Biocompatibility: DNA nanostructures inherit the low toxicity
the reactivity of DNA with other compounds. For example, the from ssDNA and dsDNA and the excellent biocompatibility ena-
carbonethyl bromide-modified drug molecules (e.g., camptothe- bles them to be applied in vivo, including cells, tissues, and
cin[61]) can be employed to directly react with PS-modified DNAs. organisms.[66] 3) Biostability: With significantly less exposed free
In addition to the methods described above, using modified ssDNA, the densely packed DNA nanostructures usually exhibit
deoxynucleotide to synthesize base-modified DNA through better resistance to degradation by nucleases.[67] Resulting from
solid-phase synthesis is also an effective method to load medici- improved stability in physiological environment, the longer
nally valuable base analogs (e.g., 5-bromouracil, mercaptopurine, retention time in animal models expands the applications in
2,6-diaminopurine, etc.). Zhu et al. replaced thymine with flox- long-term drug release, targeted drug delivery, etc. These excel-
uridine (an anticancer drug for blocking DNA synthesis and lent properties of DNA nanostructures open up new frontiers of
inhibiting RNA formation) to synthesize DNA strands and con- biomedical applications which are summarized as follows:
structed different DNA polyhedrons for anticancer therapy.[62] cancer treatment, antibacterial treatment, gene editing, vaccine
However, the modification of different functional substances carrier, and cell differentiation.
may increase the complexity, which will lead to the arduous
assembly, limited yield, and unpredictable cost of DNA 4.1. DNA Nanostructures for Anticancer Treatment
nanostructures.
Great progress has been made in the development of anticancer
therapy in recent years. However, even for the most successful
3.4. Encapsulation and effective anticancer drugs, both innate and acquired mech-
anisms of resistance are increasingly commonplace. An increas-
Encapsulation is generally applicable to DNA hydrogels for ing number of reports found that the resistance of some tumor
loading drugs (Figure 3D). Depending on the mesh size of cells originated from inhibiting the uptake of drugs in cancer
the hydrogels, it can be used to immobilize small-molecule cells.[68] Furthermore, direct injection of anticancer drugs into
drugs, nucleic acids, and proteins through encapsulation. the body may cause serious side effects due to the leakage of anti-
Liu et al. employed the DNA nanoflowers to load both TMPyP4 cancer drugs to normal tissues. DNA nanostructures exhibit
(a porphyrin-based photosensitizer) and catalase (an enzyme).[63] superior properties in addressing two serious problems
Similar to DNA microgels, DNA-functionalized liposomes are mentioned above, which improve the efficacy of drugs and avoid
also capable of loading drugs through encapsulation. Using damage to healthy tissue cells.
encapsulation to deliver various functional substances possesses Due to the enhanced permeability and retention effect (EPR
two distinctive advantages. First, it can achieve coloading of mul- effect) of nanostructures, DNA nanostructures can passively tar-
tiple drugs, including small molecules, proteins, and other poly- get cancer tissues in vivo without any auxiliary assistance.[69]
mers. Second, the drugs are embedded in the nanostructures Some reports have illustrated the relationship between the struc-
instead of merely displaying on the surface of nanostructures ture of DNA nanostructures and cellular uptake efficiency.[7]

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (6 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

In 2007, Mirkin et al. found that uptake efficiency depends on the AuNPs through amide linkages can facilitate the cellular uptake
density of DNA on the surface of AuNPs. Ding and Du et al. of platinum complex to increase cytotoxicity significantly.[60]
demonstrated that the triangle-shaped DNA origami possessed Subsequently, it has been demonstrated that various DNA nano-
enhanced passive targeting of tumor and long-lasting properties structures exhibited their ability to carry drug molecules into
at the tumor region compared with square and tube DNA cancer cells and significantly increase the cytotoxicity to drug-
origami (Figure 4A).[70] Based on the EPR effect, various drug resistant cancer cells (Figure 4B).[71]
molecules can also be directly taken up by cells after being immo- In addition, the modification of some targeting agents can pro-
bilized on DNA nanostructures, thereby destroying the drug vide DNA nanostructures with the ability of active targeting
resistance of cancer cells. Mirkin et al. first reported that connect- (Figure 4B). Aptamer, an oligonucleotide selected by systematic
ing platinum (IV) drug to the surface of DNA-functionalized evolution of ligands by exponential enrichment (SELEX)

Figure 4. DNA nanostructures for anticancer treatment. A) Passive targeting to various tissues of different structures and sizes of DNA origami.
Reproduced with permission.[70] Copyright 2014, American Chemical Society. B) Various DNA nanostructures can be uptaken by cancer cells.
Reproduced with permission.[70] Copyright 2014, American Chemical Society. C) Recognition methods between DNA nanostructures and cells.
D) Responsive release of drugs in cancer cells.

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (7 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

targeting to different aptamer targets, can be designed into the


sequence of DNA chain directly. Liu et al. encoded the sequence
of sgc8c aptamer to the DNA templates of RCA and achieved the
targeted delivery of DNA nanosponges in mouse models. Within
step-wise strategy, aptamer can be connected to DNA nanostruc-
tures through covalent conjugation or base pairing.[72] Liu and
Ding et al. used triangle DNA origami to link with MUC-1
aptamer and achieve codelivery of Dox and Au nanorods into
cancer cells.[73] Other targeted agents such as folic acid[54,74]
and antibodies[59] have also been employed on DNA nanostruc-
tures for targeting cancer cells(Figure 4C).
Besides the passive and active targeting strategy, another
method is to design switchable devices to achieve controlled
release of drugs in cancer cells. The first example is the
telomerase-response release (Figure 4D). Gu and Qian et al. com- Figure 5. DNA 6HB nanotubes for codelivering antisense oligonucleotide
(ASO) and silver ions into bacterium. Reproduced with permission.[82]
bined FITC-labeled hairpin DNA sequences hybridized with
Copyright 2021, American Chemical Society.
telomerase primers to construct DNA-functionalized AuNPs.[75]
It would precisely release doxorubicin in response to the telome-
rase existing only in the tumor cells and prevent toxicity to challenge for codelivery of all the components to realize complete
normal organs. In addition, disulfide linkages endow DNA nano- function of this large complex with high homology-directed
structures the ability to be degraded by glutathione (GSH), which repair (HDR) efficiency and low off-target effect.[84] DNA nano-
has realized the controlled release of ASO,[35] shRNA,[76] etc. structures have been proved to be a scaffold with controllable
The responsiveness of light,[77] pH,[56] and other factors[78] was sizes to load multiple components, which match the requirement
also used for the selective release of anticancer drugs in cancer of delivering CRISPR-Cas systems. Gu et al. designed a DNA NC
cells, which reduced the lethality of normal tissue cells. constructed by RCA-generated ssDNA and loaded the NC with
Cas9/sgRNA ribonucleoprotein (RNP) complexes by hybridizing
4.2. DNA Nanostructures for Antibacterial partial sequence of the NC and sgRNA (Figure 6A).[85] They suc-
cessfully delivered the complexes to the nucleus of human cells
One of the most important issues of antibacterial is that some with the assistance of nuclear-localization-signal (NLS) peptides
antibacterial drugs such as ASO and antibiotics are unable to fused to Cas9 protein. They also employed polyethylenimine
penetrate bacterial cell wall and membrane directly into drug- (PEI) to coat the complex for inducing endosomal escape.
resistant bacteria.[79] Several studies have reported that certain After cells were treated by the Cas9/sgRNA/NC-12/PEI, the assay
DNA nanostructures such as tetrahedrons had the ability to accel- revealed mutation frequencies of 28%. The similar strategies
erate the ingestion process of bacterial cells. In 2014, Leong et al. have been applied to the successful delivery of CRISPR/
first discovered that DNA tetrahedrons were able to delivery red- Cas12a complex.[86] Ding et al. constructed the sgRNA/Cas9/
emissive glutathione-protected gold nanoclusters (GSH-Au NCs) Antisense complex to codeliver CRISPR Cas9 system and
and actinomycin D (AMD) into bacterial cells and achieved ASO through coassembly of branched DNA, linker (antisense
bacterial detection and treatment at the same time.[80] In addition with two disulfide linkage), and 3’ terminal extended sgRNA
to the DNA tetrahedrons, other bacteria ingestible DNA nano- (Figure 6B).[87] With such smart design, these DNA nanostruc-
structures have been discovered. Kaminski et al. adopted DNA tures achieved the dual function of gene editing in nucleus and
origami as the delivering vehicle for the antibacterial enzyme gene silencing in cytoplasm with 80% loss of EGFP fluorescent
lysozyme.[81] Li et al. reported DNA six-helix bundle (6HB) nano- signal. They also combined sgRNA/Cas9 system with gold nano-
tubes to codeliver ASO and Agþ into bacterium with high rods to achieve the combination of gene editing and photother-
antibacterial efficacy (Figure 5).[82] They employed Agþ to destroy mal therapy for efficient tumor therapy.[88] Recently, Ding et al.
the cell wall of the bacterium and the ASO to inhibit the expres- construct a double-stranded DNA–ribonucleoprotein (RNP)
sion of the ftsZ gene. However, only a small amount of DNA hybrid nanostructure to serve as the platform of CRIPSR/dead
nanostructures has been proved to be able to enter the bacterial Cas (dCas) system. They fold double-stranded DNA with a
cells, and the mechanism is not revealed, which needs further covalently bivalent between dCas9 RNPs and dCas12a RNPs
investigation. to codeliver the dCas9 and dCas12a systems.[89]

4.3. DNA Nanostructures for Gene Editing 4.4. DNA Nanostructures for Vaccine Carrier

Gene editing is a powerful strategy to modify genes and treat There is a critical need for delivery vehicles to enhance the
genetic diseases. The Clustered regularly interspaced short immune response due to the deficient immunogenicity of some
palindromic repeats (CRISPR) gene editing system is the most antigens in vivo. DNA nanostructures provide plenty of alterna-
efficient and widely used next-generation technology involving tive platforms for loading antigens. Chang and Yan et al. first
CRISPR-associated nuclease (Cas) proteins, guide-RNA (gRNA), used DNA tetrahedron as a scaffold to load a model antigen,
donor DNA, and other factors.[83] It has been a long-existing streptavidin (STV), and CpG adjuvants together, which resulted

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (8 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

Figure 6. DNA nanostructures for gene editing. A) DNA NC loading Cas9/sgRNA complexes by through partially complementary to the sgRNA.
Reproduced with permission.[85] Copyright 2015, Wiley-VCH. B) Branched DNA structure loading a sgRNA/Cas9/antisense complex to achieve the
combination of gene editing and gene silencing. Reproduced with permission.[87] Copyright 2019, American Chemical Society.

in strong B cell response (Figure 7A).[90] Bathe et al. applied scaf- promote the proliferation and differentiation of stem cells
folded DNA origami to carry HIV immunogens with controllable (e.g., embryonic stem cells, endothelial cells, neural stem cells
organization and revealed the impact of antigen spacing.[91] [NSCs], dental pulp stem cells, etc.).[94]
Mirkin et al. encapsulated the receptor-binding domain (RBD) DNA hydrogel is a kind of unique biomaterial containing a
subunit into liposomes and decorated the core with a dense shell series of characteristics comparable to natural tissue, (i.e., high
of CpG toll-like receptor 9 (TLR9) agonist oligonucleotides. Such water content (90%), suitable mechanical properties, and shape
a DNA-functionalized liposome vaccine exhibited high resistance plasticity). DNA hydrogels have been applied to serve as excellent
to the SARS-Cov-2 virus.[92] Tubular DNA origami can serve as a biomaterials in biological sensing,[95] tissue regeneration,[96] and
carrier to encapsulate functional substances and achieve the stem cell culture [25c,97] for their high permeability, good mechan-
stimuli–response.[93] Ding et al. incorporated a lock and key ical strength, and thixotropic properties. Especially, DNA hydro-
design into a rectangular DNA nanoplatform coloaded with gels exhibited excellent properties in regulating the proliferation
peptide antigen and TLR) agonists (i.e., double-stranded RNA and differentiation of stem cells. Liu et al. employed DNA supra-
(dsRNA) and CpG DNA) and achieved the protection of the anti- molecular hydrogel to carry bone marrow mesenchymal stem
gen and TLR agonists from degradation (Figure 7B).[56] After cells (BMSCs) and demonstrated that it would protect the
entering CD8þ cytotoxic T lymphocyte (CTL), the nanodevice BMSCs from the strong shear force and enhance the therapeutic
would open in low pH (pH 5.5) and expose the antigen and effect of BMSCs (Figure 8A).[97a] Liu et al. designed a DNA hydro-
TLR agonists to activate a strong immune response. gel encapsulating NSCs to repair a 2 mm spinal cord gap in rats.
The sufficient and tunable mechanical strength, swift thixotropy,
and excellent biocompatibility contributed to the proliferation
4.5. DNA Nanostructures for Stem Cell Proliferation and and differentiation of the NSCs (Figure 8B).[96a] They demon-
Differentiation strated that a renascent neural network with basic hindlimb
function was formed within 8 weeks through the test of various
After being ingested by cells, the DNA nanostructures have tools, and DNA hydrogels with high mechanical strength could
shown potential effects on cell behaviors, such as inducing provide cells with excellent antifriction protection and 3D sup-
immunological rejection, stimulating cell signaling pathways, port. Therefore, they can serve as a suitable filler between osteo-
etc. For example, DNA tetrahedrons have been discovered to articular. More recently, Liu et al. prepared L-DNA hydrogels with

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (9 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

Figure 7. DNA nanostructures for vaccine carrier. A) DNA tetrahedron as a vaccine carrier to load antigen. Reproduced with permission.[88] Copyright
2012, American Chemical Society. B) Locked DNA origami tube to codeliver peptide antigen, CpG loop, and double-strand RNA. Reproduced with
permission.[56] Copyright 2020, The Author(s), under exclusive licence to Springer Nature Limited.

univariate mechanical strength, improved biostability, and low applications in biomedical field, including anticancer treatment,
inflammation response which greatly promoted the application antibacterial treatment, gene editing and cellular regulation, etc.
of DNA hydrogels in the field of stem cell culture.[98] Although DNA nanostructures are becoming the key materials to
realize safe delivery of functional substances in vivo, there are
still many limitations demanding prompt solution.
5. Conclusion and Future Prospects First, the imperfect monodispersity, high cost, strict storage
conditions, small preparation scale of DNA nanostructures limit
This review has focused on the construction methods and bio- their further biomedical and clinical applications. When drug
medical applications of multiple cellular ingestible DNA nano- carriers are applied in organisms, especially human bodies,
structures. With numerous efforts in the past decades devoted the homogeneity is strictly required to avoid side effects caused
in designing artificial DNA structures,[8b] DNA nanostructures by the uncertainty of drug dosage. However, construction of the
with various shapes, structures, and sizes have been constructed. DNA nanostructures via the methods mentioned above
Additional modularity has also been achieved through modifica- cannot guarantee their sizes and structures 100% identical.
tion of DNA strands via covalent connection, complementary Furthermore, the number of functional molecules loaded onto
base pairing, or other interactions, which enabled the convenient DNA nanostructures through various interactions is still uncer-
loading of functional substances (e.g., drugs, fluorophores). The tain. Therefore, to achieve applications in human bodies, the syn-
development of DNA nanotechnology, combined with excellent thesis and loading process of DNA nanostructures need to be
biocompatibility, and optimal stability of DNA molecules, makes further improved. Liu et al. developed a FGA to prepare hetero-
DNA nanostructures be excellent materials for a wide range of vesicles with programmed geometry and dimensions.[99]

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (10 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

has not been reported. In addition, the studies on explaining


the effects of DNA nanostructures on cells (i.e., induction of
immune response, regulation of cytokine expression, and inspi-
ration of cell proliferation, migration, and differentiation) stim-
ulated by the structural properties (e.g., size, shape, loop) or
specific sequence are not sufficient currently, which are signifi-
cantly demanded.
Third, the gene safety and long-term effects of DNA nano-
structures have not been investigated systematically. DNA, as
the main element of DNA nanostructures, remains the probabil-
ity to be integrated into the cellular genome through genetic
recombination. The harmful genetic recombination will lead
to an irreversible change of cellular function and even cell carci-
nogenesis. Furthermore, at present, most of the experiments
were applied in the bodies of mice and the cycle is 3–6 months,
which cannot satisfy the requirement of long-term biosecurity
validation for clinical applications.
To summarize, cellular ingestible DNA nanostructures are
still worth to be explored for potential applications in many fields.
The optimizations and improvements of the synthesis and load-
ing strategies of these DNA nanostructures would make up the
huge gap between the labs and clinical use for perfect monodis-
persity, low cost, simple storage conditions, and mass produc-
tion. While current studies focus on anticancer treatment due
to the excellent permeability of DNA nanostructures to cancer
cells, it has been ignored to apply DNA nanostructures to other
diseases. With the developing research on the interaction and
function mechanism of DNA nanostructures with cells, treating
Figure 8. DNA Nanostructures for stem cell proliferation and differentia- other cell types is also expected to take the advantage of DNA
tion. A) DNA hydrogel loading BMSCs to improve the therapy for nanostructures, but the balance between specific target efficiency
severe osteoarthritis. Reproduced with permission.[97a] Copyright 2021, and high permeability should be thoroughly considered. Besides,
Wiley-VCH. B) DNA hydrogel loading NSCs to repair a 2 mm-long spinal combining all the DNA nanotechnology-based delivery system
cord cavity. Reproduced with permission.[96a] Copyright 2021,
mentioned above, DNA-functionalized nanostructures can be
Wiley-VCH.
an alternative carrier for mRNA vaccine.[108] However, more
studies on strategy to maintain the DNA nanostructures’ integ-
Versatile core material such as AuNPs and DNA origami[100] rity in extracellular fluid and to ensure the release of full-length
can be used as the scaffolds to construct well-programmed het- sequence mRNA after entering the cell are urgently needed to
erovesicles with diverse responsiveness to temperature,[101] achieve highly efficient protein expression. Additionally, intact
pH,[102] and redox substances.[103] The FGA strategy provided validation in both cell level and animal models needs to be veri-
a generalized platform with improved monodispersity of DNA fied for further clinical applications.
nanostructures.[104] Besides monodispersity, the large-scale
synthesis and low cost are the two key factors to realize the indus-
trialization of DNA nanostructures for clinical applications. Acknowledgements
Compared with silica nanostructures (cost about $0.1 g1), poly-
This work was supported by National Natural Science Foundation of China
mer nanostructures (cost less than $1g1), and carbon nanotubes (nos. 21890731, 21821001, 21971248, 21890730), National Basic Research
(cost less than $50 g1), the expense of DNA molecules is rela- Plan of China (2018YFA0208900), and National Center for Nanoscience
tively higher for about $50–300 g1, which is too expensive to and Technology, Chinese Academy of Sciences (E1l0511ZX and
achieve the broad clinical application of DNA nanostructures.[105] E1XT2611FT).
Second, the interaction mode and function mechanism of
DNA nanostructures with cells have not been clearly explained,
as well as the mechanism and pathway by which the DNA nano- Conflict of Interest
structures are internalized by cells. Several researches have
The authors declare no conflict of interest.
reported the factors influencing the entrance of nanostructures
into cells (e.g., the DNA density of DNA-functionalized
nanoparticles,[106] the shape and size of DNA origami[65a]).
Fan et al. explained the entrance pathways of DNA tetrahedron
and its transportation pathways after entering cell.[65b,107]
Keywords
However, the detailed pathway of other DNA nanostructures biomedical applications, cellular uptake, DNA nanostructures

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (11 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

Received: August 3, 2022 [26] X. Dai, Z. Zhu, Y. Li, B. Yang, J. Xu, Y. Dong, X. Zhou, L. Yan, D. Liu,
Revised: October 7, 2022 J. Am. Chem. Soc., 2022, 144, 19017.
Published online: December 4, 2022 [27] Y. Li, Y. Ding, B. Yang, T. Cao, J. Xu, Y. Dong, Q. Chen, L. Xu, D. Liu,
CCS Chem. 2022, 1, https://doi.org/10.31635/ccschem.022.202101523.
[28] E. Cheng, Y. Xing, P. Chen, Y. Yang, Y. Sun, D. Zhou, L. Xu, Q. Fan,
[1] H. Derakhshankhah, S. Jafari, Biomed. Pharmacother. 2018, 108, 1090. D. Liu, Angew. Chem., Int. Ed. Engl. 2009, 48, 7660.
[2] Y. Zhang, H. F. Chan, K. W. Leong, Adv. Drug Delivery Rev. 2013, 65, [29] C. Li, A. Faulkner-Jones, A. R. Dun, J. Jin, P. Chen, Y. Xing, Z. Yang,
104. Z. Li, W. Shu, D. Liu, R. R. Duncan, Angew. Chem., Int. Ed. Engl. 2015,
[3] J. Y. Chung, M. N. Thone, Y. J. Kwon, Adv. Drug Delivery Rev. 2021, 54, 3957.
170, 1. [30] J. Gačanin, A. Kovtun, S. Fischer, V. Schwager, J. Quambusch,
[4] a) N. Chen, Y. He, Y. Su, X. Li, Q. Huang, H. Wang, X. Zhang, R. Tai, S. L. Kuan, W. Liu, F. Boldt, C. Li, Z. Yang, D. Liu, Y. Wu, T. Weil,
C. Fan, Biomaterials 2012, 33, 1238; b) H. Lv, S. Zhang, B. Wang, H. Barth, A. Ignatius, Adv. Healthcare Mater. 2017, 6, 1700392.
S. Cui, J. Yan, J. Controlled Release 2006, 114, 100. [31] C. Li, M. J. Rowland, Y. Shao, T. Cao, C. Chen, H. Jia, X. Zhou, Z. Yang,
[5] X. Guo, L. Huang, Acc. Chem. Res. 2012, 45, 971. O. A. Scherman, D. Liu, Adv. Mater. 2015, 27, 3298.
[6] N. C. Seeman, J. Theor. Biol. 1982, 99, 237. [32] B. Zhou, C. Li, D. Liu, W. Liu, Fundam. Res., 2022, https://doi.org/10.
[7] Q. Hu, H. Li, L. Wang, H. Gu, C. Fan, Chem. Rev. 2019, 119, 6459. 1016/j.fmre.2022.09.003.
[8] a) F. Hong, F. Zhang, Y. Liu, H. Yan, Chem. Rev. 2017, 117, 12584; [33] G. Zhu, R. Hu, Z. Zhao, Z. Chen, X. Zhang, W. Tan, J. Am. Chem. Soc.
b) N. C. Seeman, H. F. Sleiman, Nat. Rev. Mater. 2017, 3, 17068. 2013, 135, 16438.
[9] a) A. P. Alivisatos, K. P. Johnsson, X. Peng, T. E. Wilson, C. J. Loweth, [34] W. Sun, T. Jiang, Y. Lu, M. Reiff, R. Mo, Z. Gu, J. Am. Chem. Soc. 2014,
M. P. Bruchez, P. G. Schultz, Nature 1996, 382, 609; b) C. A. Mirkin, 136, 14722.
R. L. Letsinger, R. C. Mucic, J. J. Storhoff, Nature 1996, 382, 607. [35] J. Li, C. Zheng, S. Cansiz, C. Wu, J. Xu, C. Cui, Y. Liu, W. Hou,
[10] R. Elghanian, J. J. Storhoff, R. C. Mucic, R. L. Letsinger, C. A. Mirkin, Y. Wang, L. Zhang, I. T. Teng, H. H. Yang, W. Tan, J. Am. Chem.
Science 1997, 277, 1078. Soc. 2015, 137, 1412.
[11] X. Zhang, M. R. Servos, J. Liu, J. Am. Chem. Soc. 2012, 134, 7266. [36] J. Chen, N. Seeman, Nature 1991, 350, 631.
[12] B. Liu, J. Liu, J. Am. Chem. Soc. 2017, 139, 9471. [37] R. P. Goodman, R. M. Berry, A. J. Turberfield, Chem. Commun. 2004,
[13] a) L. Xu, D. Liu, Chem. Soc. Rev. 2010, 39, 150; b) D. Liu, E. Cheng, 1372.
Z. Yang, NPG Asia Mater. 2011, 3, 109; c) H. Liu, D. Liu, Chem. [38] A. Liu, R. Jones, L. Liao, D. Samara-Rubio, D. Rubin, O. Cohen,
Commun. 2009, 19, 2625. R. Nicolaescu, M. Paniccia, Nature 2004, 427, 615.
[14] a) D. Liu, S. Balasubramanian, Angew. Chem. 2003, 115, 5912; [39] Y. He, T. Ye, M. Su, C. Zhang, A. E. Ribbe, W. Jiang, C. Mao, Nature
b) S. Wang, H. Liu, D. Liu, X. Ma, X. Fang, L. Jiang, Angew. Chem. 2008, 452, 198.
2007, 119, 3989; c) W. Shu, D. Liu, M. Watari, C. K. Riener, [40] F. Zhang, S. Jiang, S. Wu, Y. Li, C. Mao, Y. Liu, H. Yan, Nat.
T. Strunz, M. E. Welland, S. Balasubramanian, R. A. McKendry, Nanotechnol. 2015, 10, 779.
J. Am. Chem. Soc. 2005, 127, 17054; d) D. Liu, A. Bruckbauer, [41] P. W. Rothemund, Nature 2006, 440, 297.
C. Abell, S. Balasubramanian, D.-J. Kang, D. Klenerman, D. Zhou, [42] S. M. Douglas, A. H. Marblestone, S. Teerapittayanon, A. Vazquez,
J. Am. Chem. Soc. 2006, 128, 2067; e) C. Chen, M. Li, Y. Xing, G. M. Church, W. M. Shih, Nucleic Acids Res. 2009, 37, 5001.
Y. Li, C. C. Joedecke, J. Jin, Z. Yang, D. Liu, Langmuir 2012, 28, 17743. [43] S. Williams, K. Lund, C. Lin, P. Wonka, S. Lindsay, H. Yan, DNA
[15] Y. Yang, G. Liu, H. Liu, D. Li, C. Fan, D. Liu, Nano Lett. 2010, 10, 1393. Computing, Springer Berlin Heidelberg, Berlin, Heidelberg 2009,
[16] H. Liu, Y. Xu, F. Li, Y. Yang, W. Wang, Y. Song, D. Liu, Angew. Chem. pp. 90–101.
2007, 119, 2567. [44] H. Dietz, S. M. Douglas, W. M. Shih, Science 2009, 325, 725.
[17] W. Wang, H. Liu, D. Liu, Y. Xu, Yang, D. Zhou, Langmuir 2007, 23, [45] E. S. Andersen, M. Dong, M. M. Nielsen, K. Jahn, R. Subramani,
11956. W. Mamdouh, M. M. Golas, B. Sander, H. Stark, C. L. Oliveira,
[18] a) P. Chen, T. Zhang, T. Zhou, D. Liu, RSC Adv. 2016, 6, 70553; J. S. Pedersen, V. Birkedal, F. Besenbacher, K. V. Gothelf, J. Kjems,
b) Z. Li, E. Cheng, W. Huang, T. Zhang, Z. Yang, D. Liu, Z. Tang, Nature 2009, 459, 73.
J. Am. Chem. Soc. 2011, 133, 15284; c) T. Zhang, P. Chen, Y. Sun, [46] D. Han, S. Pal, J. Nangreave, Z. Deng, Y. Liu, H. Yan, Science 2011,
Y. Xing, Y. Yang, Y. Dong, L. Xu, Z. Yang, D. Liu, Chem. Commun. 332, 342.
2011, 47, 5774; d) T. Zhang, Y. Dong, Y. Sun, P. Chen, Y. Yang, [47] a) Y. Zhang, C. Wang, Y. Dong, D. Wang, T. Cao, S. Wang, D. Liu,
C. Zhou, L. Xu, Z. Yang, D. Liu, Langmuir 2012, 28, 1966; Adv. Funct. Mater. 2019, 29, 1809097; b) C. Zhou, Y. Zhang,
e) T. Zhang, Z. Yang, D. Liu, Nanoscale 2011, 3, 4015. Y. Dong, F. Wu, D. Wang, L. Xin, D. Liu, Adv. Mater. 2016, 28,
[19] H. Yu, Z. Liang, L. Zhu, C. Zhao, X. He, K. Chen, W. Xu, Adv. Mater. 9819.
Interfaces 2022, 9, 2101246. [48] G. Tikhomirov, P. Petersen, L. Qian, Nature 2017, 552, 67.
[20] F. Li, D. Lyu, S. Liu, W. Guo, Adv. Mater. 2020, 32, 1806538. [49] S. M. Douglas, H. Dietz, T. Liedl, B. Högberg, F. Graf, W. M. Shih,
[21] S. H. Um, J. B. Lee, N. Park, S. Y. Kwon, C. C. Umbach, D. Luo, Nature 2009, 459, 414.
Nat. Mater. 2006, 5, 797. [50] P. Yin, R. F. Hariadi, S. Sahu, H. M. T. Choi, S. H. Park, T. H. LaBean,
[22] Y. Shao, H. Jia, T. Cao, D. Liu, Acc. Chem. Res. 2017, 50, 659. J. H. Reif, Science 2008, 321, 824.
[23] J. B. Lee, S. Peng, D. Yang, Y. H. Roh, H. Funabashi, N. Park, [51] Y. Ke, L. L. Ong, W. M. Shih, P. Yin, Science 2012, 338, 1177.
E. J. Rice, L. Chen, R. Long, M. Wu, D. Luo, Nat. Nanotechnol. [52] B. Wei, M. Dai, P. Yin, Nature 2012, 485, 623.
2012, 7, 816. [53] J. A. Kulkarni, D. Witzigmann, S. B. Thomson, S. Chen, B. R. Leavitt,
[24] Y. Xing, E. Cheng, Y. Yang, P. Chen, T. Zhang, Y. Sun, Z. Yang, D. Liu, P. R. Cullis, R. van der Meel, Nat. Nanotechnol. 2021, 16, 630.
Adv. Mater. 2011, 23, 1117. [54] H. Lee, A. K. Lytton-Jean, Y. Chen, K. T. Love, A. I. Park,
[25] a) J. Shi, Z. Shi, Y. Dong, F. Wu, D. Liu, ACS Appl. Bio Mater. 2020, 3, E. D. Karagiannis, A. Sehgal, W. Querbes, C. S. Zurenko,
2827; b) B. Yang, L. Sun, Y. Pan, Y. Dong, Y. Sun, D. Liu, Acta Polym. M. Jayaraman, C. G. Peng, K. Charisse, A. Borodovsky,
Sin. 2021, 52, 996; c) B. Yang, Z. Zhao, Y. Pan, J. Xie, B. Zhou, Y. Li, M. Manoharan, J. S. Donahoe, J. Truelove, M. Nahrendorf,
Y. Dong, D. Liu, ACS Appl. Mater. Interfaces 2021, 13, 48414. R. Langer, D. G. Anderson, Nat. Nanotechnol. 2012, 7, 389.

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (12 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

[55] L. Zhou, A. R. Chandrasekaran, M. Yan, V. A. Valsangkar, [83] A. Pickar-Oliver, C. A. Gersbach, Nat. Rev. Mol. Cell Biol. 2019, 20,
J. I. Feldblyum, J. Sheng, K. Halvorsen, Nanoscale Adv. 2021, 3, 4048. 490.
[56] S. Liu, Q. Jiang, X. Zhao, R. Zhao, Y. Wang, Y. Wang, J. Liu, Y. Shang, [84] H. X. Wang, M. Li, C. M. Lee, S. Chakraborty, H. W. Kim, G. Bao,
S. Zhao, T. Wu, Y. Zhang, G. Nie, B. Ding, Nat. Mater. 2021, 20, 421. K. W. Leong, Chem. Rev. 2017, 117, 9874.
[57] J. Fu, Y. R. Yang, A. Johnson-Buck, M. Liu, Y. Liu, N. G. Walter, [85] W. Sun, W. Ji, J. M. Hall, Q. Hu, C. Wang, C. L. Beisel, Z. Gu, Angew.
N. W. Woodbury, H. Yan, Nat. Nanotechnol. 2014, 9, 531. Chem., Int. Ed. Engl. 2015, 54, 12029.
[58] Q. Jiang, C. Song, J. Nangreave, X. Liu, L. Lin, D. Qiu, Z. G. Wang, [86] W. Sun, J. Wang, Q. Hu, X. Zhou, A. Khademhosseini, Z. Gu, Sci.
G. Zou, X. Liang, H. Yan, B. Ding, J. Am. Chem. Soc. 2012, 134, 13396. Adv. 2020, 6, eaba2983.
[59] T. Wu, J. Liu, M. Liu, S. Liu, S. Zhao, R. Tian, D. Wei, Y. Liu, Y. Zhao, [87] J. Liu, T. Wu, X. Lu, X. Wu, S. Liu, S. Zhao, X. Xu, B. Ding, J. Am.
H. Xiao, B. Ding, Angew. Chem., Int. Ed. Engl. 2019, 58, 14224. Chem. Soc. 2019, 141, 19032.
[60] S. Dhar, W. L. Daniel, D. A. Giljohann, C. A. Mirkin, S. J. Lippard, [88] W. Tang, L. Han, X. Lu, Z. Wang, F. Liu, Y. Li, S. Liu, S. Liu, R. Tian,
J. Am. Chem. Soc. 2009, 131, 14652. J. Liu, B. Ding, ACS Appl. Mater. Interfaces 2021, 13, 20974.
[61] J. Zhang, Y. Guo, F. Ding, G. Pan, X. Zhu, C. Zhang, Angew. Chem., [89] T. Wu, Y. Cao, Q. Liu, X. Wu, Y. Shang, J. Piao, Y. Li, Y. Dong, D. Liu,
Int. Ed. Engl. 2019, 58, 13794. H. Wang, J. Liu, B. Ding, J. Am. Chem. Soc. 2022, 144, 6575.
[62] Q. Mou, Y. Ma, G. Pan, B. Xue, D. Yan, C. Zhang, X. Zhu, Angew. [90] X. Liu, Y. Xu, T. Yu, C. Clifford, Y. Liu, H. Yan, Y. Chang, Nano Lett.
Chem., Int. Ed. 2017, 56, 12528. 2012, 12, 4254.
[63] M. Pan, Q. Jiang, J. Sun, Z. Xu, Y. Zhou, L. Zhang, X. Liu, Angew. [91] R. Veneziano, T. J. Moyer, M. B. Stone, E. C. Wamhoff, B. J. Read,
Chem., Int. Ed. Engl. 2020, 59, 1897. S. Mukherjee, T. R. Shepherd, J. Das, W. R. Schief, D. J. Irvine,
[64] Y. Zhang, D. Bao, S. Wang, Y. Dong, F. Wu, H. Li, D. Liu, Small 2018, M. Bathe, Nat. Nanotechnol. 2020, 15, 716.
14, 1703259. [92] M. H. Teplensky, M. E. Distler, C. D. Kusmierz, M. Evangelopoulos,
[65] a) M. M. C. Bastings, F. M. Anastassacos, N. Ponnuswamy, H. Gula, D. Elli, A. Tomatsidou, V. Nicolaescu, I. Gelarden,
F. G. Leifer, G. Cuneo, C. Lin, D. E. Ingber, J. H. Ryu, W. M. Shih, A. Yeldandi, D. Batlle, D. Missiakas, C. A. Mirkin, Proc. Natl.
Nano Lett. 2018, 18, 3557; b) L. Liang, J. Li, Q. Li, Q. Huang, Acad. Sci. U.S.A. 2022, 119, e2119093119.
J. Shi, H. Yan, C. Fan, Angew. Chem., Int. Ed. Engl. 2014, 53, 7745. [93] a) D. Wang, Y. Zhang, M. Wang, Y. Dong, C. Zhou, M. A. Isbell,
[66] a) D. Bila, Y. Radwan, M. A. Dobrovolskaia, M. Panigaj, K. A. Afonin, Z. Yang, H. Liu, D. Liu, Nanoscale 2016, 8, 3944; b) D. Wang,
Molecules 2021, 26, 4231; b) S. Messaoudi, A. A. Greschner, Z. Da, B. Zhang, M. A. Isbell, Y. Dong, X. Zhou, H. Liu,
M. A. Gauthier, Adv. Ther. 2019, 2, 1900144. J. Y. Y. Heng, Z. Yang, RSC Adv. 2015, 5, 58734.
[67] a) D. A. G. Matthew, D. Massich, D. S. Seferos, L. E. Ludlow, [94] a) M. Wei, S. Li, Z. Yang, C. Cheng, T. Li, W. Le, Nanomedicine 2021,
C. M. Horvath, C. A. Mirkin, Mol. Pharmaceutics 2009, 6, 1934; 34, 102375; b) S. Li, T. Tian, T. Zhang, X. Cai, Y. Lin, Mater. Today
b) J. Hahn, S. F. J. Wickham, W. M. Shih, S. D. Perrault, ACS 2019, 24, 57.
Nano 2014, 8, 8765. [95] T. Cao, H. Jia, Y. Dong, S. Gui, D. Liu, ACS Appl. Mater. Interfaces
[68] R. A. Ward, S. Fawell, N. Floc’h, V. Flemington, D. McKerrecher, 2020, 12, 4185.
P. D. Smith, Chem. Rev. 2021, 121, 3297. [96] a) T. Yuan, Y. Shao, X. Zhou, Q. Liu, Z. Zhu, B. Zhou, Y. Dong,
[69] a) H. Mok, S. H. Lee, J. W. Park, T. G. Park, Nat. Mater. 2010, 9, 272; N. Stephanopoulos, S. Gui, H. Yan, D. Liu, Adv. Mater. 2021, 33,
b) G. Jeon, S. Y. Yang, J. K. Kim, J. Mater. Chem. 2012, 22, 14814. 2102428; b) Y. Jin, Y. Li, S. Song, Y. Ding, Y. Dong, Y. Lu,
[70] Q. Zhang, Q. Jiang, N. Li, L. Dai, Q. Liu, L. Song, J. Wang, Y. Li, J. Tian, D. Liu, C. Zhang, Adv. Mater. Interfaces 2022, 9, 2101321.
B. Ding, Y. Du, ACS Nano 2014, 8, 6633. [97] a) X. Yan, B. Yang, Y. Chen, Y. Song, J. Ye, Y. Pan, B. Zhou,
[71] a) G. Zhu, J. Zheng, E. Song, M. Donovan, K. Zhang, C. Liu, W. Tan, Y. Wang, F. Mao, Y. Dong, D. Liu, J. Yu, Adv. Mater. 2021, 33,
Proc. Natl. Acad. Sci. U.S.A. 2013, 110, 7998; b) Y.-X. Zhao, A. Shaw, 2104758;
X. Zeng, E. Benson, A. M. Nystrom, B. Hogberg, ACS Nano 2012, 6, b) Y. Wang, Y. Shao, X. Ma, B. Zhou, A. Faulkner-Jones, W. Shu,
8684. D. Liu, ACS Appl. Mater. Interfaces 2017, 9, 12311.
[72] H. Pei, N. Lu, Y. Wen, S. Song, Y. Liu, H. Yan, C. Fan, Adv. Mater. [98] B. Yang, B. Zhou, C. Li, X. Li, Z. Shi, Y. Li, C. Zhu, X. Li, Y. Hua,
2010, 22, 4754. Y. Pan, J. He, T. Cao, Y. Sun, W. Liu, M. Ge, Y. R. Yang,
[73] L. Song, Q. Jiang, J. Liu, N. Li, Q. Liu, L. Dai, Y. Gao, W. Liu, D. Liu, Y. Dong, D. Liu, Angew. Chem., Int. Ed. 2022, 61, e202202520.
B. Ding, Nanoscale 2017, 9, 7750. [99] Y. Dong, Y. Sun, L. Wang, D. Wang, T. Zhou, Z. Yang,
[74] H. L. Seung, H. Ko, Y. Chen, C. Mao, Biomacromolecules 2008, 9, 3039. Z. Chen, Q. Wang, Q. Fan, D. Liu, Angew. Chem., Int. Ed. Engl.
[75] Y. Ma, Z. Wang, M. Zhang, Z. Han, D. Chen, Q. Zhu, W. Gao, Z. Qian, 2014, 53, 2607.
Y. Gu, Angew. Chem., Int. Ed. Engl. 2016, 55, 3304. [100] Y. Dong, Y. R. Yang, Y. Zhang, D. Wang, X. Wei, S. Banerjee,
[76] J. Liu, L. Song, S. Liu, S. Zhao, Q. Jiang, B. Ding, Angew. Chem., Int. Y. Liu, Z. Yang, H. Yan, D. Liu, Angew. Chem., Int. Ed. 2017, 56,
Ed. Engl. 2018, 57, 15486. 1586.
[77] X. Tan, B. B. Li, X. Lu, F. Jia, C. Santori, P. Menon, H. Li, B. Zhang, [101] Z. Zhao, C. Chen, Y. Dong, Z. Yang, Q. H. Fan, D. Liu, Angew.
J. J. Zhao, K. Zhang, J. Am. Chem. Soc. 2015, 137, 6112. Chem., Int. Ed. Engl. 2014, 53, 13468.
[78] a) F. Li, Y. Liu, Y. Dong, Y. Chu, N. Song, D. Yang, J. Am. Chem. Soc. [102] C. Wang, Y. Zhang, Y. Shao, X. Tian, J. Piao, Y. Dong, D. Liu, Giant
2022, 144, 4667; b) F. Li, Z. Lv, X. Zhang, Y. Dong, X. Ding, Z. Li, S. Li, 2020, 1, 100006.
C. Yao, D. Yang, Angew. Chem. Int. Ed. Engl. 2021, 60, 25557. [103] S. Wang, Y. Zhang, Y. Dong, D. Liu, Polymer 2019, 175, 146.
[79] M. C. McManus, Am. J. Health-Syst. Pharm. 1997, 54, 1420. [104] Y. Dong, Y. Yang, C. Lin, D. Liu, Acc. Chem. Res. 2022, 55, 1938.
[80] M. I. Setyawati, R. V. Kutty, C. Y. Tay, X. Yuan, J. Xie, D. T. Leong, [105] B. Kelley, mAbs 2009, 1, 443.
ACS Appl. Mater. Interfaces 2014, 6, 21822. [106] D. S. S. David, A. Giljohann, P. C. Patel, J. E. Millstone, N. L. Rosi,
[81] I. Mela, P. P. Vallejo-Ramirez, S. Makarchuk, G. Christie, D. Bailey, C. A. Mirkin, Nano Lett. 2007, 7, 3818.
R. M. Henderson, H. Sugiyama, M. Endo, C. F. Kaminski, [107] H. Ding, J. Li, N. Chen, X. Hu, X. Yang, L. Guo, Q. Li, X. Zuo,
Angew. Chem., Int. Ed. Engl. 2020, 59, 12698. L. Wang, Y. Ma, C. Fan, ACS Cent. Sci. 2018, 4, 1344.
[82] Q. Long, B. Jia, Y. Shi, Q. Wang, H. Yu, Z. Li, ACS Appl. Mater. [108] A. Mokhtarzadeh, H. Vahidnezhad, L. Youssefian, J. Mosafer,
Interfaces 2021, 13, 47987. B. Baradaran, J. Uitto, Trends Mol. Med. 2019, 25, 1066.

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (13 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH
26999307, 2023, 1, Downloaded from https://onlinelibrary.wiley.com/doi/10.1002/anbr.202200119 by Readcube (Labtiva Inc.), Wiley Online Library on [23/10/2023]. See the Terms and Conditions (https://onlinelibrary.wiley.com/terms-and-conditions) on Wiley Online Library for rules of use; OA articles are governed by the applicable Creative Commons License
www.advancedsciencenews.com www.advnanobiomedres.com

Rui Xu graduated at Wuhan University with a B.S. in chemistry in 2021. He is now a Ph.D. student at
Tsinghua University under the supervision of Professor Dongsheng Liu, Yuanchen Dong, and Yuhe
Renee Yang. He focuses on functional biomolecules design and their biomedical applications.

Dongsheng Liu graduated from University of Science and Technology of China in 1993 and did his Ph.D.
study in the Hong Kong Polytechnic University from 1999 to 2002. He then joined the Chemistry
Department of Cambridge University as a postdoc. Dongsheng is now a full professor at Department of
Chemistry, Tsinghua University. He was awarded the 1st “CCS-RSC Young Chemist Award” in 2008 and
the 7th CCS-BASF Youth Innovation Prize in 2014. Dongsheng was invited as FRSC in 2011, became
“Changjiang” Chair Professor in 2015, and fellow of CCS in 2020. His research mainly focuses on the
assembly of DNA and its hybrids.

Yuhe Renee Yang graduated from Tsinghua University in 2007 and started her Ph.D. journey in Arizona
State University. While working with Dr. Hao Yan, she created artificial multienzyme DNA complexes and
networks that addressed important fundamental enzymology issues related to multienzyme catalysis.
During postdoc training in TSRI with Dr. Andrew B. Ward, Renee worked on solving high-resolution
cryoEM structures of HIV/flu surface protein and neutralizing antibody complexes. She started her own
lab in NCNST in 2021. She worked in DNA nanotechnology and structural biology for more than a
decade and continues to find the joy in this interdisciplinary niche.

Adv. NanoBiomed Res. 2023, 3, 2200119 2200119 (14 of 14) © 2022 The Authors. Advanced NanoBiomed Research published by Wiley-VCH GmbH

You might also like