You are on page 1of 10

DOI: 10.1002/asia.

201900574 Minireview

Multifunctional DNA Origami Nanoplatforms for Drug Delivery


Xuehe Lu,[a, b] Jianbing Liu,*[b] Xiaohui Wu,[b, c] and Baoquan Ding*[a, b, c]

Chem. Asian J. 2019, 14, 2193 – 2202 2193 T 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Minireview

Abstract: DNA nanotechnology has been employed in the therapeutic drugs, cytosine–phosphate–guanine, functional
construction of self-assembled nano-biomaterials with uni- proteins, gene drugs, and their combinations for optoacous-
form size and shape for various biological applications, such tic imaging, photothermal therapy, chemotherapy, immuno-
as bioimaging, diagnosis, or therapeutics. Herein, recent suc- logical therapy, gene therapy, and coagulation-based thera-
cessful efforts to utilize multifunctional DNA origami nano- py are summarized. The challenges and opportunities for
platforms as drug-delivery vehicles are reviewed. Diagnostic DNA-based nanocarriers for biological applications are also
and therapeutic strategies based on gold nanorods, chemo- discussed.

1. Introduction up fabrication of DNA origami, RNA–DNA hybrid origami, 3D


DNA origami nanostructures, and some related computational
DNA is not only genetic material, but also a representative tools are described.
building block that employs classic Watson–Crick base pairing
for molecular self-assembly. The programmability of DNA nano-
structures and precise molecular recognition provide a tailored 2.1. Self-Assembled DNA Origami Nanostructures
strategy for the construction of functional DNA-based nano- DNA origami represents a remarkable advance in the field of
platforms. DNA nanotechnology, invented in the 1980s by the DNA nanotechnology and molecular self-assembly. In 2006,
group of Seeman, opened up a new research area.[1] In recent Rothemund first reported this fascinating technique and suc-
years, much progress in biological applications of multifunc- cessfully constructed a series of DNA origami nanostructures,
tional DNA nanoplatforms has been reported. DNA hydrogels such as squares, triangles, disks, and five-pointed stars, in a
are employed in a variety of biological applications, such as simple one-pot reaction (Figure 1).[8] This attractive technique
drug delivery,[2] sensing,[3] and 3D cell culture.[4] DNA tetrahedra
have been developed for drug delivery,[5] molecular diagnosis,[6]
and bioimaging.[7]
In particular, functional nanoplatforms based on the DNA or-
igami technique have attracted increasing interest in the fields
of bioimaging and cancer therapy. Rationally designed multi-
functional DNA origami nanoplatforms with structural pro-
grammability, spatial addressability, and excellent biocompati-
bility display several unique advantages for various biological
applications. The development of a DNA origami based drug-
delivery system is a gradual evolutionary process. There are
challenges and great opportunities to utilize multifunctional
DNA origami nanoplatforms for biomedical applications. Figure 1. Examples of DNA origami nanostructures and the corresponding
AFM images. Reprinted from Ref. [8] with permission from Nature Publishing
Group, Copyright 2006.

2. Evolution of DNA Origami Nanotechnology


employed a single-stranded DNA of 7249 bases as the “scaffold
In this section, we introduce the history of DNA origami nano- strand” (obtained from the M13mp18 bacteriophage) and
technology and its rapid developments in recent years. Scale- about 200 chemically synthesized short oligonucleotides as
“staple strands” (complementary to different regions of the
[a] X. Lu, Prof. B. Ding
scaffold) to construct desired DNA nanostructures with high
School of Materials Science and Engineering
Zhengzhou University, Zhengzhou 450001 (P.R. China)http://www.bdinglab.- yield and reliable reproducibility. Since then, a wide variety of
com/ 2D DNA nanostructures have been reported by many groups
E-mail: dingbq@nanoctr.cn on the basis of the DNA origami technique. Subsequently,
[b] X. Lu, Dr. J. Liu, X. Wu, Prof. B. Ding nanoscale shapes of dolphins[9] and a Chinese map[10] were de-
CAS Key Laboratory of Nanosystem and Hierarchical Fabrication
signed and assembled by the groups of Kjems and He, respec-
CAS Center for Excellence in Nanoscience
National Center for Nanoscience and Technology tively.
11 BeiYiTiao, ZhongGuanCun, Beijing 100190 (P.R. China) DNA origami nanotechnology has compelling advantages
E-mail: liujb@nanoctr.cn for the construction of different nanostructures with controlled
[c] X. Wu, Prof. B. Ding sizes and shapes. One important feature of DNA origami is ex-
University of Chinese Academy of Sciences
cellent addressability on the surface of DNA origami. Function-
Beijing 100049 (P.R. China)
al ligands, biomolecules, and nanoscale objects can be precise-
The ORCID identification number(s) for the author(s) of this article can be
found under: ly organized on the desired position of DNA origami nano-
https://doi.org/10.1002/asia.201900574. structures.

Chem. Asian J. 2019, 14, 2193 – 2202 www.chemasianj.org 2194 T 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Minireview

2.2. Development of DNA Origami Nanotechnology structures that could be reversibly reconfigured in three differ-
ent conformational states, depending on the indicated concen-
The DNA origami technique can potentially be used to con- tration of cations in solution (Figure 2 f).[25] A closed DNA tetra-
struct various functional nanostructures with customized sizes hedral origami nanostructure was designed and fabricated by
and shapes. However, there is still a limitation to using this Ke et al. (Figure 2 g).[26] Yan and co-workers constructed DNA
strategy for nanoscale construction that originates from the origami with complex 3D curvatures (Figure 2 h).[27] Hçgberg
finite length of the standard DNA scaffold. This single-stranded
DNA scaffold limits the size and surface area of DNA origami.
Thus, large functional components cannot be accommodated
on regular DNA origami nanostructures. To overcome this Xuehe Lu received his BS degree (2015) from
shortcoming, many strategies have been developed to fabri- Huaiyin Normal University and MS degree
(2018) from Shanghai University. He is cur-
cate larger DNA nanostructures. For instance, Winfree et al. de-
rently a PhD candidate, working under the su-
veloped an algorithmic self-assembly from origami seeds for pervision of Professor Baoquan Ding, at the
programmable bottom-up fabrication (Figure 2 a).[11] Liu and National Center for Nanoscience and Technol-
co-workers obtained larger and more complex nanostructures ogy and Zhengzhou University. His research
interests focus on the chemical modification
through a linker-strand connection strategy (Figure 2 b).[12]
of DNA and biomedical applications.
Qian and co-workers used multiple origami tiles to draw micro-
meter-scale DNA origami images, such as the Mona Lisa (Fig-
ure 2 c).[13] Sugiyama and co-workers used multiple origami
structures named 2D DNA jigsaw pieces to scale up origami
structures.[14] The group of Seeman developed DNA origami
arrays to scale up 2D DNA origami.[15] Liu and co-workers used Jianbing Liu received his PhD (2016) in chemi-
cal biology from Nankai University. Currently,
eight-helix bundles as staples to extend the sizes of DNA origa- he is an assistant research professor at the
mi.[16] Based on long, single-stranded polymerase chain reac- National Center for Nanoscience and Technol-
tion (PCR) amplification products, the group of Woolley suc- ogy. His research interests focus on the devel-
cessfully obtained scaffold strands with different lengths.[17] In opment of delivery systems for gene therapy
based on DNA nanocarriers.
research by Shin and co-workers, double-stranded (ds) DNA
could be employed as a scaffold to construct two discrete
nanoscale objects.[18] Li and co-workers prepared four long
single-stranded DNA with sequences of 10 563, 10 782, 21 261,
and 31 274 nucleotides to be used as scaffolds to construct
larger DNA origami nanostructures.[19]
In addition to the classical DNA-templated scaffold, RNA
Xiaohui Wu received her BS degree from Tian-
transcripts can also be employed as RNA scaffolds to build jin University in 2018. She is currently an MS
RNA–DNA hybrid origami. The group of Sugiyama successfully candidate, working under the supervision of
fabricated seven-helix bundled rectangular structures and six- Professor Baoquan Ding, at the National
Center for Nanoscience and Technology. Her
helix bundled tubular structures by using RNA transcripts as
research interests focus on the self-assembly
scaffolds and DNA sequences as staple strands.[20] The physical of RNA–DNA hybrid origami for biological ap-
properties of the assembled RNA–DNA hybrid origami can be plications.
modified by the functionalized RNA scaffold. Moreover, various
functions based on the structure of RNA, such as RNA catalysis
and RNA interference (RNAi), can be introduced into the RNA–
DNA hybrid origami. Meanwhile, Mao and co-workers also con-
structed ribbon, rectangle, and triangle RNA–DNA hybrid origa-
mi structures, depending on the length of the RNA scaffolds.[21] Baoquan Ding received his bachelor degree in
In addition to building 2D DNA nanostructures, various 3D chemistry from Jilin University in 2000. He ob-
nanostructures can also be constructed through the DNA ori- tained his PhD in 2006 from the Department
of Chemistry, New York University, under the
gami technique. For instance, Shih and co-workers presented supervision of Professor Nadrian Seeman. He
the first attempt to construct various 3D nanostructures, such then worked as a postdoctoral research fellow
as monoliths, square nuts, a railed bridge, a genie’s bottle, at the Molecular Foundry, Lawrence Berkeley
stacked crosses, and slotted crosses (Figure 2 d).[22] Gothelf and National Laboratory. He joined the Biodesign
Institute, Arizona State University, as a re-
co-workers successfully created an addressable 3D DNA box, search assistant professor in October 2009. He
with a size of 42 V 36 V 36 nm3, which could be opened by pre- became a professor at the National Center for
designed DNA “keys” (Figure 2 e).[23] At the same time, Komiya- Nanoscience and Technology, P.R. China, in
na and Kuzuya constructed another box-shaped 3D DNA origa- November 2010. His research interests include
DNA nanotechnology, coassembled biomole-
mi by selective closing of a preformed open motif.[24] Dietz and cules, and drug delivery.
co-workers constructed origami-based dynamic DNA nano-

Chem. Asian J. 2019, 14, 2193 – 2202 www.chemasianj.org 2195 T 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Minireview

Figure 2. Some examples of self-assembled DNA nanostructures. a) 2D DNA origami by seeds. Reproduced from Ref. [11b] with permission from the National
Academy of Sciences, Copyright 2009. b) 2D DNA origami by linkers. Reproduced from Ref. [12] with permission from the American Chemical Society, Copy-
right 2010. c) 2D DNA origami by titles. Reproduced from Ref. [13] with permission from Nature Publishing Group, Copyright 2017. d) Multilayer 3D origami.
Reproduced from Ref. [22] with permission from Nature Publishing Group, Copyright 2009. e) Openable 3D DNA box. Reproduced from Ref. [23] with permis-
sion from Nature Publishing Group, Copyright 2009. f) 3D nanorobot origami. Reproduced from Ref. [25] with permission from the American Association for
the Advancement of Science, Copyright 2015. g) Closed DNA tetrahedral origami. Reproduced from Ref. [26] with permission from American Chemical Society,
Copyright 2009. h) 3D curvature. Reproduced from Ref. [27] with permission from the American Association for the Advancement of Science, Copyright 2011.
i) Wireframe DNA origami. Reproduced from Ref. [28] with permission from Nature Publishing Group, Copyright 2015.

and co-workers created wireframe DNA origami by means of and scattering in the near-infrared (NIR) region can be em-
graph theory (Figure 2 i).[28] Thus, the construction of 3D DNA ployed as excellent candidates for optoacoustic imaging and
nanostructures has dramatically increased the diversification of photothermal therapy.[33]
DNA nanostructures. Recently, Ding and co-workers successfully developed a self-
Nowadays, several computational tools have been devel- assembled DNA origami–GNR complex for dual-functional
oped for the design of various DNA nanostructures, such as nanotheranostics by combining the features of GNRs and DNA
SARSE-DNA,[29] caDNAno,[30] and CanDo.[31] Recently, software origami nanostructures (Figure 3 a).[34] Compared with bare
with a fully automatic inverse design procedure, named DAE- GNRs, DNA origami–GNR complexes achieved a significant en-
DALUS, was developed by Veneziano et al. to construct various hancement of cellular uptake in human MCF7 breast cancer
DNA nanostructures with tailored sizes and shapes.[32] This soft- cells. In particular, the triangular DNA origami–GNR complex
ware grants easy access to the DNA origami technique to re- demonstrated optimal cellular accumulation. In contrast to
searchers from different backgrounds. Multifunctional DNA bare GNRs, the triangular DNA origami–GNR complex showed
nanostructures can be rationally designed with these software enhanced photothermolysis against tumor cells in vitro and in
tools for a wide variety of applications. vivo.
After observing the efficient photothermal therapy effect
based on the DNA origami–GNR complex, Tian and co-workers
3. Biological Applications
subsequently developed GNR-loaded triangular DNA origami
In this section, DNA origami nanostructures as vehicles for the to function as an optoacoustic imaging agent.[35] GNR-loaded
delivery of gold nanorods (GNRs), chemotherapeutic drugs, cy- DNA origami can act as an efficient contrast agent to achieve
tosine–phosphate–guanine (CpG), functional proteins, gene enhanced imaging quality with a decreased dose. Photother-
drugs, and their combinations for optoacoustic imaging, pho- mal treatment based on this DNA origami–GNR complex dem-
tothermal therapy, chemotherapy, immunological therapy, onstrated efficient inhibition of tumor growth in vivo (Fig-
gene therapy, and coagulation-based therapy are described. ure 3 b).

3.1. Delivery of GNRs 3.2. Delivery of Small-Molecule Drugs


Noble-metal nanoparticles with attractive size-dependent Chemotherapeutic drugs, such as doxorubicin (Dox) and dau-
properties present great promise for electronic, optical, and norubicin, can efficiently inhibit the proliferation of tumor cells
biomedical applications. Specially, GNRs with strong absorbing by intercalating into DNA base pairs to block the replication

Chem. Asian J. 2019, 14, 2193 – 2202 www.chemasianj.org 2196 T 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Minireview

Figure 3. Examples of DNA origami for GNR delivery in biological applications. a) GNR-loaded DNA origami for photothermal therapy. DO-GNR = DNA origa-
mi–GNR complex. Reproduced from Ref. [34] with permission from Wiley-VCH, Copyright 2015. b) GNR-loaded DNA origami for optoacoustic imaging and
photothermal therapy. PBS = phosphate-buffered saline, D-AuNR = gold nanorods with DNA nanostructures. Reproduced from Ref. [35] with permission from
Wiley-VCH, Copyright 2016.

and transcription of functional genes. However, similar to other efflux pumps in leukemia cells at clinically relevant drug con-
traditional anticancer drugs, these chemotherapeutic drugs centrations.
also have adverse side effects and poor selectivity during
tumor therapy.[36] Targeted delivery of these chemotherapeutic
3.3. Delivery of CpG
drugs would achieve efficient inhibition of tumor growth and
avoid systemic toxicity. CpG oligodeoxynucleotides (ODNs) have strong immunostimu-
Ding and co-workers demonstrated that DNA origami nano- latory effects. The natural immune system can recognize exter-
structures had tremendous potential to act as a biocompatible nal CpG sequences and initiate immune responses.[41] Because
carrier of Dox for cancer therapy (Figure 4 a).[37] Dox can be effi- CpG-containing ODNs have exhibited great potential as immu-
ciently loaded into the DNA duplex through intercalation with nostimulatory agents, various delivery systems for CpG-con-
base pairs. The Dox-loaded DNA origami nanoplatform demon- taining ODNs have been developed. Liedl and co-workers used
strated prominent cytotoxicity to drug-resistant human breast a tubular DNA origami to load unmethylated CpG sequences
cancer cells and successfully circumvented drug resistance in through DNA hybridization (Figure 5).[42] This DNA origami,
vitro. Ding and co-workers subsequently employed Dox-loaded containing 62 CpG-ODNs, triggered higher immunostimulation
triangular DNA origami to inhibit tumor growth in vivo (Fig- than that of equal amounts of CpG motifs delivered by Lipo-
ure 4 b).[38] The origami-based drug-delivery system demon- fectamine. The CpG-modified DNA origami can be recognized
strated efficient antitumor efficacy in vivo without inducing by TLR9 and induces remarkable cytokine production.
observable systemic toxicity.
Hçgberg and co-workers employed two DNA origami nano-
3.4. Delivery of Functional Proteins
structures to load Dox for the treatment of three different
breast cancer cell lines (MDA-MB-231, MDA-MB-468, and MCF- The delivery of therapeutic proteins to disease sites is an effec-
7; Figure 4 c).[39] Depending on different degrees of twisting of tive strategy to treat cancer or other diseases.[43] A suitable de-
the DNA origami, the drug release kinetics can be rationally livery system is essential to achieve therapeutic effects. A func-
controlled. This drug-loaded DNA origami also showed high tional DNA origami nanoplatform can also be designed to real-
degrees of internalization and efficient inhibition of prolifera- ize efficient protein delivery.
tion of tumor cells in vitro. Castro and co-workers employed a Transferrin (Tf) plays a key role in the transportation and me-
rodlike DNA origami nanostructure to deliver the widely used tabolism of iron through receptor-mediated endocytosis.[44]
chemotherapeutic drug daunorubicin in a leukemia model Moreover, Tf has been successfully employed to facilitate the
(Figure 4 d).[40] This drug-loaded DNA nanocarrier also demon- delivery of macromolecular payloads[45] and various DNA con-
strated efficient circumvention of drug resistance mediated by structs[46] in target cells. Kjems and co-workers first equipped a

Chem. Asian J. 2019, 14, 2193 – 2202 www.chemasianj.org 2197 T 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Minireview

Figure 4. Examples of DNA origami for small-molecule drug delivery in biomedical applications. a) DNA origami for Dox delivery in vitro. Reproduced from
Ref. [37] with permission from the American Chemical Society, Copyright 2012. b) DNA origami for Dox delivery in vivo. Reproduced from Ref. [38] with per-
mission from the American Chemical Society, Copyright 2014. c) DNA origami for Dox delivery in vitro. Reproduced from Ref. [39] with permission from the
American Chemical Society, Copyright 2012. d) DNA origami for daunorubicin delivery. Reproduced from Ref. [40] with permission from Wiley-VCH, Copyright
2016.

planar DNA origami with Tf ligand to increase cellular uptake gen recognition, the loaded antibodies can bind to cell-surface
in a cancer cell line (Figure 6 a).[47] The Tf-modified DNA origami receptors to inhibit the proliferation of target tumor cells.
nanostructure demonstrated efficient cellular uptake through Thrombin is an important blood coagulation protease that
receptor-mediated endocytosis. stops bleeding. Zhao and co-workers constructed a tubular
Douglas and co-workers constructed a hexagonal DNA ori- DNA nanorobot to load thrombin in its inner cavity (Fig-
gami barrel (about 40 nm) to transport molecular payloads, ure 6 c).[49] This multifunctional DNA nanorobot can induce co-
such as antibodies, to target cells (Figure 6 b).[48] Antibody Fab’’ agulation in tumor-associated vessels, leading to efficient
fragments can be loaded into the molecular barrel through tumor tissue thrombosis and tumor necrosis in tumor-bearing
DNA hybridization. Once the barrels are opened through anti- mouse models.

Chem. Asian J. 2019, 14, 2193 – 2202 www.chemasianj.org 2198 T 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Minireview

Figure 5. DNA origami for CpG delivery. Reproduced from Ref. [42] with permission from the American Chemical Society, Copyright 2011.

RNase A can efficiently degrade RNA molecules inside cells and exhibited high inhibition of multidrug-resistant tumor
to induce high cytotoxicity. Ding and co-workers employed an growth in vivo without apparent systemic toxicity.
aptamer-modified rectangular DNA origami nanosheet to load RNAi is a tailored technology to knock down the expression
and deliver RNase A into target tumor cells in vitro (Fig- of MDR-related genes. Ding and co-workers employed the
ure 6 d).[50] The RNase A loaded DNA origami nanoplatform can DNA origami nanoplatform to load and deliver small hairpin
efficiently penetrate the cell membrane and induce high inhibi- RNA (shRNA) transcription templates that targeted MDR-associ-
tion of proliferation of target tumor cells. ated genes P-gp and Survivin (Figure 7 c).[54] The Dox and
shRNA transcription templates for coloading DNA origami with
aptamer modifications can efficiently enter target cells and
induce a remarkable antitumor effect. This multifunctional
3.5. Codelivery of Multiple Therapeutic Components
DNA origami nanoplatform can achieve the synergistic inhibi-
Multidrug resistance (MDR) is a major obstacle in the clinical tion of tumor growth against multidrug-resistant tumors in
treatment of cancer. Much higher doses and different combi- vivo.
nations of chemotherapeutic drugs have been administered to
achieve a considerable therapeutic effect. This widely em-
ployed treatment strategy may function at an earlier stage, but
4. Summary and Outlook
results in much stronger MDR for patients in the long run.[51]
Permeability glycoprotein (P-gp) is a member of the trans- In recent decades, various DNA nanostructures have been con-
membrane adenosine triphosphate (ATP)-binding cassette structed and utilized for biomedical applications. Herein, we
transporter superfamily. P-gp is overexpressed in many drug- summarized recent progress in employing multifunctional DNA
resistant tumor cells and functions as an efflux pump for many origami nanoplatforms as drug-delivery vehicles. DNA origami
chemotherapeutic drugs. Ding and co-workers constructed a with structural programmability, spatial addressability, and ex-
multifunctional DNA origami nanoplatform through the combi- cellent biocompatibility is tailored to load a variety of function-
nation of Dox and GNRs to downregulate the expression of P- al components at desired sites. Diagnostic and therapeutic
gp and circumvent drug resistance of multidrug-resistant MCF- strategies based on GNRs, chemotherapeutic drugs, CpG, func-
7 cells in vitro (Figure 7 a).[52] Aptamer-modified multifunctional tional proteins, gene drugs, and their combinations for bio-
DNA origami nanoplatforms can achieve efficient cellular imaging and cancer therapy were highlighted.
uptake, as verified by means of fluorescent and two-photon lu- Although various multifunctional DNA origami nanoplat-
minescence imaging. After NIR laser irradiation, P-gp expres- forms have been successfully developed for drug delivery, chal-
sion was efficiently downregulated to achieve combined che- lenges still remain for their further application in vivo. First, a
motherapeutic and photothermal therapy. better understanding of the systemic pharmacokinetics, includ-
P53, as an important tumor suppressor gene, plays a key ing internal circulation, biodistribution, and metabolism of
role in the inhibition of the proliferation of tumor cells. Ding DNA origami nanocarriers with different sizes and shapes, is
and co-workers fabricated a triangular DNA origami containing still needed.[55] Second, the endocytosis mechanism and intra-
a linear tumor therapeutic gene (p53) and a chemotherapeutic cellular fate of different DNA origami nanocarriers should be
drug (Dox) for combined therapy of multidrug-resistant tumors further investigated. Finally, the possible immune response in-
(Figure 7 b).[53] The aptamer-modified kitelike DNA origami duced by exogenous DNA should also be taken into account
nanoplatform demonstrated efficient gene-delivery efficacy carefully.

Chem. Asian J. 2019, 14, 2193 – 2202 www.chemasianj.org 2199 T 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Minireview

Figure 6. Examples of DNA origami for protein delivery. a) Tf-loaded DNA origami for enhanced cellular uptake. Reproduced from Ref. [47] with permission
from Wiley-VCH, Copyright 2016. b) DNA nanorobot for antibody fragment delivery. Reproduced from Ref. [48] with permission from the American Association
for the Advancement of Science. Copyright 2012. c) DNA origami nanorobot for thrombin delivery. Reproduced from Ref. [49] with permission from Nature
Publishing Group, Copyright 2018. d) DNA origami for ribonuclease A (RNase A) delivery. Reproduced from Ref. [50] with permission from American Chemical
Society, Copyright 2019.

Despite the abovementioned challenges, DNA origami with Acknowledgements


customized functions can serve as a promising candidate for
efficient drug delivery. We believe that further developments This work was supported by the National Natural Science
in DNA origami nanotechnology will provide a more intelligent Foundation of China (21573051, 21708004, 51761145044); the
nanoplatform for diagnosis and disease therapy. Science Fund for Creative Research Groups of the National Nat-
ural Science Foundation of China (21721002); the National
Basic Research Program of China (2016YFA0201601,

Chem. Asian J. 2019, 14, 2193 – 2202 www.chemasianj.org 2200 T 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Minireview

Figure 7. Examples of DNA origami for the codelivery of multiple therapeutic components. a) DNA origami for Dox and GNR codelivery. Reproduced from
Ref. [52] with permission from the Royal Society of Chemistry, Copyright 2017. b) DNA origami for Dox and p53 gene codelivery. Reproduced from Ref. [53]
with permission from the American Chemical Society, Copyright 2018. c) DNA origami for Dox and RNAi codelivery. Reproduced from Ref. [54] with permission
from Wiley-VCH, Copyright 2018.

2018YFA0208900); the Key Research Program of Frontier Scien- [3] a) A. Baeissa, N. Dave, B. D. Smith, J. Liu, ACS Appl. Mater. Interfaces
ces, CAS (grant QYZDB-SSW-SLH029); the CAS Interdisciplinary 2010, 2, 3594 – 3600; b) H. Y. Lee, H. Jeong, I. Y. Jung, B. Jang, Y. C. Seo,
H. Lee, Adv. Mater. 2015, 27, 3513 – 3517; c) X. Wei, T. Tian, S. Jia, Z. Zhu,
Innovation Team; and the K. C. Wong Education Foundation.
Y. Ma, J. Sun, Z. Lin, C. J. Yang, Anal. Chem. 2015, 87, 4275 – 4282; d) Y.
Song, B. Lin, T. Tian, X. Xu, W. Wang, Q. Ruan, J. Guo, Z. Zhu, C. Yang,
Anal. Chem. 2019, 91, 388 – 404.
Conflict of interest [4] a) C. Li, A. Faulkner-Jones, A. R. Dun, J. Jin, P. Chen, Y. Z. Xing, Z. Q.
Yang, Z. B. Li, W. M. Shu, D. S. Liu, R. R. Duncan, Angew. Chem. Int. Ed.
The authors declare no conflict of interest. 2015, 54, 3957 – 3961; Angew. Chem. 2015, 127, 4029 – 4033; b) J. Jin,
Y. Z. Xing, Y. L. Xi, X. L. Liu, T. Zhou, X. X. Ma, Z. Q. Yang, S. T. Wang, D. S.
Liu, Adv. Mater. 2013, 25, 4714 – 4717; c) J. Gacănin, J. Hedrich, S. Sieste,
Keywords: biological activity · DNA · DNA origami · drug G. Glaßer, I. Lieberwirth, C. Schilling, S. Fischer, H. Barth, B. Knçll, C. V.
Synatschke, T. Weil, Adv. Mater. 2018, 31, 185044.
delivery · nanotechnology
[5] a) J. Li, H. Pei, B. Zhu, L. Liang, M. Wei, Y. He, N. Chen, D. Li, Q. Huang,
C. H. Fan, ACS Nano 2011, 5, 8783 – 8789; b) H. Lee, A. K. Lytton-Jean, Y.
[1] N. R. Kallenbach, R.-I. Ma, N. C. Seeman, Nature 1983, 305, 829 – 831. Chen, K. T. Love, A. I. Park, E. D. Karagiannis, A. Sehgal, W. Querbes, C. S.
[2] a) J. Song, K. Im, S. Hwang, J. Hur, J. Nam, G. O. Ahn, S. Hwang, S. Kim, Zurenko, M. Jayaraman, C. G. Peng, K. Charisse, A. Borodovsky, M. Mano-
N. Park, Nanoscale 2015, 7, 9433 – 9437; b) R. Hu, H. Yuan, B. Wang, L. haran, J. S. Donahoe, J. Truelove, M. Nahrendorf, R. Langer, D. G. Ander-
Liu, F. Lv, S. Wang, ACS Appl. Mater. Interfaces 2014, 6, 11823 – 11828; son, Nat. Nanotechnol. 2012, 7, 389 – 393; c) P. Charoenphol, H. Bermu-
c) L. Sun, N. Hu, J. Peng, L. Chen, J. Weng, Adv. Funct. Mater. 2014, 24, dez, Mol. Pharm. 2014, 11, 1721 – 1725; d) K. R. Kim, D. R. Kim, T. Lee,
6905 – 6913; d) M. Shahbazi, T. Bauleth-Ramos, H. A. Santos, Adv. Ther. J. Y. Yhee, B. S. Kim, I. C. Kwon, D. R. Ahn, Chem. Commun. 2013, 49,
2018, 1, 1800042.

Chem. Asian J. 2019, 14, 2193 – 2202 www.chemasianj.org 2201 T 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim
Minireview
2010 – 2012; e) Y. Zhang, J. Tu, D. Wang, H. Zhu, S. Kumar Maity, X. Qu, [33] a) L. Vigderman, B. P. Khanal, E. R. Zubarev, Adv. Mater. 2012, 24, 4811 –
B. Bogaert, H. Pei, H. Zhang, Adv. Mater. 2018, 30, 1703658. 4841; b) Z. Xiao, C. Ji, J. Shi, E. M. Pridgen, J. Frieder, J. Wu, O. C. Farokh-
[6] a) B. H. Pei, N. Lu, Y. L. Wen, S. P. Song, Y. Liu, H. Yan, C. H. Fan, Adv. zad, Angew. Chem. Int. Ed. 2012, 51, 11853 – 11857; Angew. Chem. 2012,
Mater. 2010, 22, 4754 – 4758; b) Z. G. Wang, Q. W. Xue, W. Z. Tian, L. 124, 12023 – 12027; c) Z. Zhang, L. Wang, J. Wang, X. Jiang, X. Li, Z. Hu,
Wang, W. Jiang, Chem. Commun. 2012, 48, 9661 – 9663; c) H. Liu, J. Luo, Y. Ji, X. Wu, C. Chen, Adv. Mater. 2012, 24, 1418 – 1423.
L. Fang, H. Huang, J. Deng, J. Huang, S. Zhang, Y. Li, J. Zheng, Biosens. [34] Q. Jiang, Y. Shi, Q. Zhang, N. Li, P. Zhan, L. Song, L. Dai, J. Tian, Y. Du, Z.
Bioelectron. 2018, 121, 47 – 53. Cheng, B. Ding, Small 2015, 11, 5134 – 5141.
[7] a) A. S. Walsh, H. F. Yin, C. M. Erben, M. J. A. Wood, A. J. Turberfield, ACS [35] Y. Du, Q. Jiang, N. Beziere, L. Song, Q. Zhang, D. Peng, C. Chi, X. Yang,
Nano 2011, 5, 5427 – 5432; b) K. R. Kim, Y. D. Lee, T. Lee, B. S. Kim, S. H. Guo, G. Diot, V. Ntziachristos, B. Ding, J. Tian, Adv. Mater. 2016, 28,
Kim, D. R. Ahn, Biomaterials 2013, 34, 5226 – 5235. 10000 – 10007.
[8] P. W. Rothemund, Nature 2006, 440, 297 – 302. [36] a) M. M. Gottesman, Annu. Rev. Med. 2002, 53, 615 – 627; b) B. A. Chab-
[9] E. S. Andersen, M. D. Dong, M. M. Nielsen, K. Jahn, A. Lind-Thomsen, W. ner, T. G. Roberts, Nat. Rev. Cancer 2005, 5, 65 – 72.
Mamdouh, K. V. Gothelf, F. Besenbacher, J. Kjems, ACS Nano 2008, 2, [37] Q. Jiang, C. Song, J. Nangreave, X. Liu, L. Lin, D. Qiu, Z.-G. Wang, G. Zou,
1213 – 1218. X. Liang, H. Yan, B. Ding, J. Am. Chem. Soc. 2012, 134, 13396 – 13403.
[10] Z. Zhang, Y. Wang, C. Fan, C. Li, Y. Li, L. Qian, Y. Fu, Y. Shi, J. Hu, L. He, [38] Q. Zhang, Q. Jiang, N. Li, L. Dai, Q. Liu, L. Song, J. Wang, Y. Li, J. Tian, B.
Adv. Mater. 2010, 22, 2672 – 2675. Ding, Y. Du, ACS Nano 2014, 8, 6633 – 6643.
[11] a) K. Fujibayashi, R. Hariadi, S. H. Park, E. Winfree, S. Murata, Nano Lett. [39] Y. Zhao, A. Shaw, X. Zeng, E. Benson, A. M. Nystrçm, B. Hçgberg, ACS
2008, 8, 1791 – 1797; b) R. D. Barish, R. Schulman, P. W. Rothemund, E. Nano 2012, 6, 8684 – 8691.
Winfree, Proc. Natl. Acad. Sci. USA 2009, 106, 6054 – 6059. [40] P. D. Halley, C. R. Lucas, E. M. McWilliams, M. J. Webber, R. A. Patton, C.
[12] Z. Zi, M. Liu, J. Nangreave, H. Yan, Y. Liu, J. Am. Chem. Soc. 2010, 132, Kural, D. M. Lucas, J. C. Byrd, C. E. Castro, Small 2016, 12, 308 – 320.
13545 – 13552. [41] a) Ref. [5a]; b) J. Vollmer, A. M. Krieg, Adv. Drug Delivery Rev. 2009, 61,
[13] G. Tikhomirov, P. Petersen, L. Qian, Nature 2017, 552, 67 – 71. 195 – 204; c) Y. Qu, J. Yang, P. Zhan, S. Liu, K. Zhang, Q. Jiang, C. Li, B.
[14] A. Rajendran, M. Endo, Y. Katsuda, K. Hidaka, H. Sugiyama, ACS Nano Ding, ACS Appl. Mater. Interfaces 2017, 9, 20324 – 20329.
2011, 5, 665 – 671. [42] V. J. Scheller, S. Heidegger, N. Sandholzer, P. C. Nickels, N. A. Suhartha, S.
[15] W. Liu, H. Zhong, R. Wang, N. C. Seeman, Angew. Chem. Int. Ed. 2011, 50, Endres, C. Bourquin, T. Liedl, ACS Nano 2011, 5, 9696 – 9702.
264 – 267; Angew. Chem. 2011, 123, 278 – 281. [43] M. Yan, J. Du, Z. Gu, M. Liang, Y. F. Hu, W. Zhang, S. Priceman, L. Wu, Z.
[16] Z. Zhao, H. Yan, Y. Liu, Angew. Chem. Int. Ed. 2010, 49, 1414 – 1417; Zhou, Z. Liu, T. Segura, Y. Tang, Y. Lu, Nat. Nanotechnol. 2010, 5, 48 – 53.
Angew. Chem. 2010, 122, 1456 – 1459. [44] R. D. Klausner, G. Ashwell, J. van Renswoude, J. B. Harford, K. R. Bridges,
[17] E. Pound, J. R. Ashton, H. A. Becerril, A. T. Woolley, Nano Lett. 2009, 9, Proc. Natl. Acad. Sci. USA 1983, 80, 2263 – 2266.
4302 – 4305. [45] T. R. Daniels, E. Bernabeu, J. A. Rodr&guez, S. Patel, M. Kozman, D. A.
[18] B. Hçgberg, T. Liedl, W. M. Shih, J. Am. Chem. Soc. 2009, 131, 9154 – Chiappetta, E. Holler, J. Y. Ljubimova, G. Helguera, M. L. Penichet, Bio-
9155. chim. Biophys. Acta Gen. Subj. 2012, 1820, 291 – 317.
[19] X. Chen, Q. Wang, J. Peng, Q. Long, H. Yu, Z. Li, ACS Appl. Mater. Interfa- [46] S. Modi, C. Nizak, S. Surana, S. Halder, Y. Krishnan, Nat. Nanotechnol.
ces 2018, 10, 24344 – 24348. 2013, 8, 459 – 467.
[20] M. Endo, S. Yamamoto, K. Tatsumi, T. Emura, K. Hidaka, H. Sugiyama, [47] D. H. Schaffert, A. H. Okholm, R. S. Sorensen, J. S. Nielsen, T. Torring, C. B.
Chem. Commun. 2013, 49, 2879 – 2881. Rosen, A. L. Kodal, M. R. Mortensen, K. V. Gothelf, J. Kjems, Small 2016,
[21] P. Wang, S. Ko, C. Tian, C. Hao, C. Mao, Chem. Commun. 2013, 49, 5462 – 12, 2634 – 2640.
5464. [48] S. M. Douglas, I. Bachelet, G. M. Church, Science 2012, 335, 831 – 834.
[22] S. M. Douglas, H. Dietz, T. Liedl, B. Hçgberg, F. Graf, W. M. Shih, Nature [49] S. Li, Q. Jiang, S. Liu, Y. Zhang, Y. Tian, C. Song, J. Wang, Y. Zou, G. J. An-
2009, 459, 414 – 418. derson, Y. Han, Y. Chang, Y. Liu, C. Zhang, L. Chen, G. Zhou, G. Nie, H.
[23] E. S. Andersen, M. Dong, M. M. Nielsen, K. Jahn, R. Subramani, W. Mam- Yan, B. Ding, Y. Zhao, Nat. Biotechnol. 2018, 36, 258 – 264.
douh, M. M. Golas, B. Sander, H. Stark, C. L. Oliveira, J. S. Pedersen, V. Bir- [50] S. Zhao, F. Duan, S. Liu, T. Wu, Y. Shang, R. Tian, J. Liu, Z.-G. Wang, Q.
kedal, F. Besenbacher, K. V. Gothelf, J. Kjems, Nature 2009, 459, 73 – 76. Jiang, B. Ding, ACS Appl. Mater. Interfaces 2019, 11, 11112 – 11118.
[24] A. Kuzuya, M. Komiyama, Chem. Commun. 2009, 4182 – 4184. [51] a) M. M. Gottesman, T. Fojo, S. E. Bates, Nat. Rev. Cancer 2002, 2, 48 – 58;
[25] T. Gerling, K. Wagenbauer, A. Neuner, H. Dietz, Science 2015, 347, 1446 – b) G. Szak#cs, J. K. Paterson, J. A. Ludwig, C. BoothGenthe, M. M. Gottes-
1452. man, Nat. Rev. Drug Discovery 2006, 5, 219 – 314.
[26] Y. K. Ke, J. Sharma, M. H. Liu, K. Jahn, Y. Liu, H. Yan, Nano Lett. 2009, 9, [52] L. Song, Q. Jiang, J. Liu, N. Li, Q. Liu, L. Dai, Y. Gao, W. Liu, D. Liu, B.
2445 – 2447. Ding, Nanoscale 2017, 9, 7750 – 7754.
[27] D. R. Han, S. Pal, J. Nangreave, Z. T. Deng, Y. Liu, H. Yan, Science 2011, [53] J. Liu, L. Song, S. Liu, Q. Jiang, Q. Liu, N. Li, Z.-G. Wang, B. Ding, Nano
332, 342 – 346. Lett. 2018, 18, 3328 – 3334.
[28] E. Benson, A. Mohammed, J. Gardell, S. Masich, E. Czeizler, P. Orponen, [54] J. Liu, L. Song, S. Liu, S. Zhao, Q. Jiang, B. Ding, Angew. Chem. Int. Ed.
B. Hçgberg, Nature 2015, 523, 441 – 444. 2018, 57, 15486 – 15490; Angew. Chem. 2018, 130, 15712 – 15716.
[29] E. S. Andersen, A. Lind-Thomsen, B. Knudsen, S. E. Kristensen, J. H. Hav- [55] a) Y. Tokura, S. Harvey, C. Chen, Y. Wu, D. Y. W. Ng, T. Weil, Angew. Chem.
gaard, E. Torarinsson, N. Larsen, C. Zwieb, P. Sestoft, J. Kjems, J. Gorod- Int. Ed. 2018, 57, 1587 – 1591; Angew. Chem. 2018, 130, 1603 – 1607; b) Y.
kin, RNA 2007, 13, 1850 – 1859. Tokura, S. Harvey, X. Xu, C. Chen, S. Morsbach, K. Wunderlich, G. Fytas,
[30] S. M. Douglas, A. H. Marblestone, S. Teerapittayanon, A. Vazquez, G. M. Y. Wu, D. Y. W. Ng, T. Weil, Chem. Commun. 2018, 54, 2808 – 2811.
Church, W. M. Shih, Nucleic Acids Res. 2009, 37, 5001 – 5006.
[31] C. E. Castro, F. Kilchherr, D. N. Kim, D.-N. Shiao, T. Wauer, P. Wortmann,
Manuscript received: April 30, 2019
M. Bathe, H. Dietz, Nat. Methods 2011, 8, 221 – 229.
[32] R. Veneziano, S. Ratanalert, K. Zhang, F. Zhang, H. Yan, W. Chiu, M. Accepted manuscript online: May 24, 2019
Bathe, Science 2016, 352, 1534. Version of record online: June 17, 2019

Chem. Asian J. 2019, 14, 2193 – 2202 www.chemasianj.org 2202 T 2019 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim

You might also like