Uptake and stability of DNA nanostructures in cells: a cross-sectional overview of the current state of the art

Divita Mathur *a, Angelica Rose Galvan bc, Christopher M. Green c, Kevin Liu a and Igor L. Medintz c
aDepartment of Chemistry, Case Western Reserve University, Cleveland, OH 44106, USA. E-mail: divita.mathur@case.edu
bFischell Department of Bioengineering, University of Maryland, College Park, MD 20742, USA
cCenter for Bio/Molecular Science and Engineering, Code 6900, US Naval Research Laboratory, Washington, DC 20375, USA

Received 22nd October 2022 , Accepted 16th January 2023

First published on 25th January 2023


Abstract

The physical and chemical properties of synthetic DNA have transformed this prototypical biopolymer into a versatile nanoscale building block material in the form of DNA nanotechnology. DNA nanotechnology is, in turn, providing unprecedented precision bioengineering for numerous biomedical applications at the nanoscale including next generation immune-modulatory materials, vectors for targeted delivery of nucleic acids, drugs, and contrast agents, intelligent sensors for diagnostics, and theranostics, which combines several of these functionalities into a single construct. Assembling a DNA nanostructure to be programmed with a specific number of targeting moieties on its surface to imbue it with concomitant cellular uptake and retention capabilities along with carrying a specific therapeutic dose is now eminently feasible due to the extraordinary self-assembling properties and high formation efficiency of these materials. However, what remains still only partially addressed is how exactly this class of materials is taken up into cells in both the native state and as targeted or chemically facilitated, along with how stable they are inside the cellular cytosol and other cellular organelles. In this minireview, we summarize what is currently reported in the literature about how (i) DNA nanostructures are taken up into cells along with (ii) what is understood about their subsequent stability in the complex multi-organelle environment of the cellular milieu along with biological fluids in general. This allows us to highlight the many challenges that still remain to overcome in understanding DNA nanostructure–cellular interactions in order to fully translate these exciting new materials.


Introduction

The importance of formulating a predictable relationship between the properties of DNA nanostructures (DNA NS or NS) and their structural integrity, along with their intracellular behavior, is emphasized in their now frequently demonstrated potential as designer nanoscale platforms for targeted medical interventions.1–4 DNA NS have shown potential superiority in delivering pharmaco-chemical or nucleic acid therapeutics against several diseases in comparison to current drugs. The targeted delivery of chemotherapeutic drugs via DNA NS instead of direct intravenous administration can dramatically reduce off-target toxicity.5–8 Their versatility is highlighted by examples where thrombin-DNA aptamer chemistry has been leveraged in DNA platforms to, on one hand, induce targeted damage in the vasculature of tumor sites in Chinese hamster animal models9 and, on the other hand, prevent thrombosis during hemodialysis by sequestering protein activity from plasma.10,11 In another example of this material's versatility, RNA-targeting protein RNAse A bound to DNA platforms via sulfo-SMCC protein–DNA chemistry was able to induce cell death in cancer MCF-7 cells.12 Optimizing the display of ligands,13–16 aptamers,17–19 and antigens20,21 on the NS surface to match the spatial pattern of membrane receptors improves the therapeutic efficacy, particularly the B-cell recognition, of engineered DNA NS as vaccines,19,20 cancer treatment, or immunogenic treatments. In addition to interfacing with cells, DNA NS can potentially be programmed to remain inactive until the right biological signal stimulates a mechano-chemical actuation; these cues can include that originating from light-induced antigen binding,22 aptamer-protein binding,9,23 antigen-ligand binding,24 and many others that are yet to be incorporated into prototypical biomedical systems.25,26 Combining imaging with therapeutic application is also possible due to the multiplexed functionality of DNA NS.27 Alongside performing the aforementioned delivery functionalities, the off-target immune response elicited by DNA NS is often non-toxic up to considerably high doses.28 Most excitingly, delivering genes for potential Cas9-mediated genome editing is possible using the superior structural properties of DNA NS.29

As synthetic DNA-based therapeutic NS mature and start being prototyped for the aforementioned applications, confident prediction of the stability and structural integrity of a DNA NS inside the body at any point in time persists as a significant challenge. There are general observations that suggest that – depending on the structural and/or functional “complexity” of a DNA NS – it could remain “stable” up to a certain number of hours within the extracellular environment, serum, blood, and the cytosol (vide infra). Whole animal delivery studies show that the shape of a DNA NS imparts certain renal targeting advantages over unfolded plasmid DNA which can be leveraged for renal intervention.30 Collectively processing such observations to formulate a framework that could guide future design criteria has yet to be done and this is something that is crucial prior to clinical trial scrutiny.

The complexity of the journey that a DNA NS will undergo for delivery into a cell should be appreciated. Using the context of targeting a solid tumor in an animal model for illustration, the NS will most likely be first administered as a bolus or by slower infusion in the blood stream, intramuscularly, to the lymphatic system, or even in a transdermal manner. Assuming a targeting moiety is attached to the NS, such as an arginylglycylaspartic acid (RGD) peptide motif or folic acid targeting tumors displaying integrin or folic acid receptors, respectively, the NS will then selectively bind to the tumor cell surfaces.31 The DNA NS then are usually taken up into the cells by some form of endocytosis that in this case is most likely receptor mediated, but which for untargeted NS may be a more non-specific mechanism, these are reviewed in ref. 2 and 32. Now once inside the endolysosomal system and entrapped in endosomal vesicles, the DNA NS need to escape this harsh environment to access the cytosol and subsequently enter the nucleus or mitochondria if those organelles are the required targets of the intervention. Fig. 1 provides an excellent overview of the interactions occurring during the endocytic uptake of a DNA nanorod into H1299 (transformed lung carcinoma epithelial) cells as followed by transmission electron microscopy (TEM), which, in this case, also shows the hydrolysis of the structure after being present in acidic lysosomes over 24 h.33 Suffice to say that DNA NS have already been shown to discreetly access these different components of a mammalian cell – they can robustly match the spatial pattern and maximize their interaction with membrane receptors,20,21 undergo endocytosis to access intracellular space,34,35 and make deliveries into the cytosol and even the nucleus.12,35 How one DNA NS accomplishes all this while another different assembly architecture does not, how they can be engineered specifically enough, and the fundamental rules that determine a given intracellular pathway remain poorly understood.


image file: d2nr05868e-f1.tif
Fig. 1 DNA nanoparticle endocytosis into cells. Visualization of the uptake and endocytic pathway in H1299 cells of a rod-shaped DNA NS labeled with spherical gold nanoparticles which allow for tracking via TEM imaging. (a) Schematic and representative electron microscopy image of the DNA nanorod with 6 gold nanospheres displayed along the length of the DNA NS. (b) Schematic representation of the uptake mechanism hypothesized for DNA nanorods through the four stages of endocytosis. (c–f) Step-wise representation of the transport of gold-labeled DNA nanorods from extracellular membrane to lysosomes where it can be seen that the nanorod has disintegrated (clustered instead of linearly arranged gold spheres). Reprinted (adapted) with permission from ref. 33 Copyright 2018 American Chemical Society.

Herein, a progress report consolidating the knowledge gained so far in elucidating the uptake mechanisms and stability of DNA NS in cellular and biological environments is presented. The DNA NS discussed here constitute 10–200 nanometer (nm) structures, or sometimes larger, as prepared by self-assembly of a pool of single stranded DNA (ssDNA) molecules via thermal annealing. DNA materials of this kind are typically designed based on two overarching strategies, namely, DNA origami36 and complete ssDNA assembly.37,38 For more information on the DNA materials themselves and their properties, along with potential applications in the current context, the interested reader is referred to ref. 1, 4, 39, and 40. Of particular interest are DNA NS that are examined for their innate cellular uptake abilities rather than when coupled with transfection agents4 such as chitosan,41 gold nanoparticles,42 condensing agents,7,43 or viral capsids.44 These examples along with delivery of materials that are electroporated into cells, while valuable, are outside the current scope. A more extensive discussion on the in vivo implications of DNA NS is also reviewed recently.1,45 The context of interest being examined is that of understanding which properties of DNA NS facilitate interaction with the cell membrane or otherwise affect the rest of the internalization path at this stage of their development.

Uptake of DNA NS into mammalian cells

Tables 1 and 2 summarize published reports describing the state of the investigation into the uptake properties of DNA nanoparticles. Table 1 focuses on DNA NS applied in different therapeutic contexts while Table 2 summarizes studies geared towards fundamental understanding and typically do not contain DNA NS that were functionally modified with a drug or protein. The uptake studies listed in Table 1 fall under the context of three overarching therapeutic applications, namely, immune-modulation, nucleic acid delivery, and pharmacochemical delivery. For each study, the type of DNA NS studied, cell type, displayed targeting moieties, dosage, and the tracked end-point are identified. Abbreviations of cell types are further defined in the table's footnotes. Predominantly, the DNA tetrahedron (∼10 nm) is used in these studies but over the last several years larger DNA origami structures (∼100 nm) have become more common, likely due to the availability of more sites to couple biocompatible signaling molecules. We observed that the display of targeting antigens or ligands on the surface of the DNA NS is not a requirement for uptake and that DNA NS with21,31,46,47 and without5,34,48,49 targeting moieties have been reported to be internalized by cells. The spatial arrangement of antigens on the DNA NS does, however, perform significantly better in immunostimulatory applications since it leverages B/T-cell receptor antigen pattern recognition and macrophage functionality.20,21,50
Table 1 Overview of application-specific studies reporting uptake of DNA nanostructure by mammalian cell and animal models
Application DNA nanostructure (size nm/mass kDa) Cell type primary/transformed Targeting moiety on DNA/drug or Protein Dosage (in ×104 cells)a Tracked end-point Cytotoxicityb Ref. (year)
Abbreviations: Adpgk = neoantigen peptide derived from MC-38 colon carcinoma, CpG = unmethylated cytosine–guanine dinucleotide motifs, DOX = doxorubicin, gp100 = peptide vaccine from melanoma antigen glycoprotein, MUC1 = mucin 1 protein overexpressed in malignant cells, OVA = ovalbumin peptide epitope presented by class I major histocompatibility complex, PTO = kinase substrate, siRNA = small interfering RNA, inj = tail vein injection, BBB = blood brain barrier. Cell types: BMDC = bone marrow derived dendritic cells; COS = monkey kidney fibroblast cells; DMS53 = human small cell lung cancer cells; H1299 = human non-small cell lung cancer cells; H441 = human distal lung epithelial cells; HEK293 = human embryonic epithelial kidney cells; HeLa = human cervical cancer cells; Huh7 = human epithelial cancer cells; HUVEC = human umbilical vein endothelial cells; MCF-7 = human breast cancer cells; MDA-MB = human epithelial breast cancer cells; NSCLC = human non-small cell lung cancer cells; NIH-3T3 = mouse embryonic fibroblast cells; RAW264.7 = mouse macrophage cells; THP-1 = human leukemia monocytic cells; U87MG = human glioblastoma.a Dosage is given as ×104 cells. For example, first entry represents 3 × 104 cells.b Cytotoxicity in the presence of bare DNA NS only (not the treatment in some cases).
Immune-stimulation: Rectangle reconfigured as tube (90 nm) BMDC, mice OVA, gp100, and Adpgk peptides 2 nM in 3× Endosome 98 (2021)
Octahedron (50 nm), tube (400 nm/4 MDa), nanorod (89 nm/4 MDa) 3T3, HEK-293, H441 None 1 nM in 1× None >7 d 79 (2014)
Disk (80 nm/4 MDa) RAW264.7 CpG 10–1000 nM in 2× Endosome None >5 h 21 (2022)
Tetrahedron (7 nm/82 kDa) RAW264.7 CpG 100 nM in 50× None >24 h 99 (2011)
Rectangle (90 nm/4 MDa) RAW264.7, THP-1 IgG 1 μM DNA in 3× 50 (2021)
Tube (80 nm/4 MDa) Splenic macrophages CpG, CpG PTO, CpG chimera 2.4 nM 50 μL DNA in 40× Lysosomes None >18 h 72 (2011)
Tetrahedron (7 nm/82 kDa) RAW 264.7, primary dendritic cells CpG, streptavidin 62.5 nM in 2.5×–25× Lysosomes 53 (2012)
 
Nucleic acid delivery: Tetrahedron (7 nm/82 kDa) HeLa siRNA 10–80 nM in 15× None >24 h 31 (2012)
Tube (27 nm/140 kDa) HeLa None 10 nM Endosome None >24 h 100 (2014)
Rectangular prism (10 nm/160 kDa) HeLa DNA trigger strand 75 nM in 1× None >72 h 26 (2016)
Cube (7 nm/82 kDa) HeLa, primary B-lymphocytes DNA trigger strand 250 pmole in 10× 101 (2014)
Rectangle, tube (32 nm–64 nm) DMS53, NSCLC siRNA 16.7 nM in 1× Cytoplasm None >8 h 46 and 47 (2020)
Tetrahedron (7 nm/82 kDa) Mice SARS-CoV-2 aptamer 500 nM 100 μL inj. None >12 h 19 (2022)
Tetrahedron (7 nm/82 kDa) Mice TGF-β1 mRNA 1 μM 200 μL inj. Liver targeting None >24 h 102 (2022)
Tetrahedron (7 nm/82 kDa) Mice BACE1 aptamer 1 μM 100 μL inj. Cross BBB 18 (2022)
 
Pharmaco-chemical/drug: Triangle (120 nm), rod (400 nm) MCF-7 None/DOX 50–100 μM None >48 h 6 (2012)
Disk (62 nm/4 MDa), donut (44 nm/4 MDa), sphere (62 nm/4 MDa) MCF-7, MDA-MB-231 MUC1 aptamer/DOX 1 nM in 0.3× None >48 h 5 (2022)
Cross, rectangle, triangle (50 nm/4 MDa) MDA-MB-231 None/DOX 1.25–5 nM in 0.3× Lysosomes 8 (2018)
T-nanotube, S-nanotube (100 nm/4 MDa) MDA-MB-231, MDA-MB-468, and MCF-7 None/DOX 50–15[thin space (1/6-em)]000 nM Dox in 20× Endosomes 103 (2012)
Tetrahedron (7 nm/82 kDa) U87MG Tumor penetrating peptide/DOX 50 nM in 0.4× 104 (2016)
3D DNA NS (100 nm MCF-7, mice MUC1 aptamer 4 μM in 10× 105 (2022)
 
Protein delivery: Rectangle reconfigured as a tube (90 nm/4 MDa) HUVEC, mice AS1411 aptamer/thrombin None >72 h 9 (2018)
Rectangle (90 nm/4 MDa) MCF-7 MUC1 aptamer/RnaseA 1 nM None >48 h 12 (2019)


Table 2 Studies focused on DNA nanostructure uptake in mammalian cells
DNA nanostructure (size nm/mass kDa) Cell type primary/transformed Targeting moiety displayed on the DNA NS Dosage (in ×104 cells)a Tracked end-point Cytotoxicityc Ref. (year)
Cell types: BMDC = bone marrow derived dendritic cells; COS = monkey kidney fibroblast cells; DMS53 = human small cell lung cancer cells; H1299 = human non-small cell lung cancer cells; HEK293 = human embryonic epithelial kidney cells; HeLa = human cervical cancer cells; Huh7 = human epithelial cancer cells; HUVEC = human umbilical vein endothelial cells; DC2.4 = immortalized murine dendritic cells; NIH-3T3 = mouse embryonic fibroblast cells; RAW264.7 = mouse macrophage cells.a Dosage is given as ×104 cells. For example, first entry represents 10 × 104 cells (or 105 cells).b The different cells were first treated with artificial membrane receptor (AMR) that enabled uptake and targeting of the DNA NS.c Cytotoxicity in the presence of bare DNA NS only (not the treatment in some cases).
Thin rod, thick rod, small ring, large ring, barrel, octahedron, block (50 nm–400 nm/4 MDa) BDMC, HEK293, HUVEC None 1 nM in 10× None >12 h 106 (2018)
Octahedron (11 nm/160 kDa) COS fibroblasts None 2 μg mL−1 in 100× Cytoplasm, endosomes 34 (2016)
Tetrahedral nanocage (8 nm/82 kDa), octahedral nanocage (11 nm/180 kDa), chainmail (25 nm/180 kDa), square box (35 nm), rectangle (80 nm/4 MDa) COS-7 None 6 μg ml−1 in 100× Lysosomes 69 (2019)
Tetrahedron (7 nm/82 kDa) HEK None 1 μg in 30× Cytoplasm None >72 h 49 (2011)
Cube, tetrahedron nanocages (10 nm) HeLa None 150 nM Lysosome, mitochondria 52 (2019)
Tetrahedron (7 nm/82 kDa) HeLa None 150 nM None >3 h 55 (2021)
Tetrahedron (7 nm/82 kDa) HeLa, COS-7 None 100 nM in 8× Lysosome, nucleus 35 (2014)
Small tetrahedron (11 nm), large tetrahedron (47 nm), small rod (32 nm), large rod (127 nm) HeLa, H1299, DMS53 None 250 nM Lysosome None >1 h 33 (2018)
6-Helix bundle (6 nm × 7 nm) HepG2, Huh7 Oligolysine 50 nM in 10× Endosome, lysosome, cytoplasm None >24 h 48 (2021)
Concave-DNA origami structure (34 nm/4 MDa) HeLa, HepG2, MCF-7, DC2.4 pH low insertion peptideb 15 nM in 10× Endoplasmic reticulum 107 (2022)
Icosahedron (10 nm/160 kDa) Caenorhabditis elegans 10 kDa FITC-dextran 12–15 μM Endosome 108 (2011)
Tube (400 nm/4 MDa) NIH-3T3 None 10 nM Lysosome 109 (2016)
Tetrahedron (7 nm/82 kDa) RAW264.7 None 10 nM in 2.5× Cytoplasm None >6 h 110 (2013)


It is generally agreed that the primary mechanism of NS uptake into cells without active delivery modalities such as electroporation is almost universally that of endocytosis. Indeed, almost all the studies listed in Tables 1 and 2 directly or indirectly align with this consensus. The most common mechanism of uptake was found to be receptor-mediated endocytosis wherein the DNA NS interacted with membrane receptors and are internalized via endocytic vesicles (Fig. 1). A clear uptake mechanism explaining the efficiency of internalization or the interaction of DNA NS with membrane receptors is still not fully elucidated. Colocalization of the fluorescence of dyes (typically cyanine 5 – Cy5) coupled to DNA NS prototypes and cell lysosomal staining suggests that the structures reach the lysosomes in a NS-dependent timeframe.51 The DNA NS in lysosomes may or may not be fully intact; some clarity is needed as to what extent the Cy5 fluorescence colocalization within lysosomes is attributed to stable DNA NS.32,52 Macrophage cells such as RAW264.7 (established from a tumor in a male mouse induced with the Abelson murine leukemia virus) are known to perform phagocytosis on what the human body would normally consider foreign material, but in the case of DNA NS, the distinction between their endo- and phagocytosis and other uptake mechanisms by these cells is yet to be studied further.50 Preliminary studies have tracked certain DNA NS in endosomes and even lysosomes of RAW264.7 cells through fluorescence colocalization studies.21,53

The shape or structural complexity of DNA NS is a factor that modulates the timeframes in which the structures enter the cell and collect in the lysosomes before complete degradation.33 While it is not immediately evident from Tables 1 and 2 how the physicochemical properties of DNA NS determine which endocytic pathway they might trigger (since 7–400 nm large rods, tubes, polyhedra, disks, and other shapes are amenable to uptake), preliminary experimental and computational analysis suggest that the vertices of polygonal DNA NS offer the path of least resistance in terms of interfacing with the negatively charged cell membrane for endocytosis.54,55 More recent work by Bhatia and co-workers systematically corroborates that tetrahedron endocytosis occurs more readily than for other larger polyhedral structures, and the size of the tetrahedral shape (14.3 nm “medium” size versus “large” 32 nm or “small” 11 nm) can affect its endocytic efficacy.56 Cancerous cell lines have higher uptake propensity for DNA NS than non-cancerous. The implication of understanding the role of NS shape in uptake can be considered crucial to formalizing the physical factors of DNA NS that determine their cellular interaction.

Stability of DNA NS in biological environments

Over the last decade, considerable effort by the scientific community has helped decipher the preliminary structural stability of DNA nanoparticles in some extracellular in vitro-related environments; knowledge of this kind helps overcome one of the primary barriers to the development of any translational biomedical products. These environments include cell culture media, blood sera (most commonly fetal bovine serum – FBS), and various buffers with salt and pH conditions mimicking what could be experienced in vivo for mammalian cells and the body, as summarized in Tables 3 and 4. In biological environments, the integrity of DNA NS can be rapidly compromised through attacks by various DNA nucleases (DNases), low divalent cation concentrations (common to nearly all physiological environments), and large variations in ionic strength and pH, particularly upon uptake into cells or ingestion into the stomach with its inherently acidic environment which is naturally hydrolytic.39,40 For eventual therapeutic applications, a DNA NS must be sufficiently robust to maintain structural integrity long enough to enter the body, travel to a target of interest, and undergo any desired interactions, such as uptake, macrophage capture, or viral capture.57,58 The timescales for such interactions range from minutes to tens of hours, though most applications require lifetimes on the order of several hours in the body.
Table 3 Stability of DNA nanostructures in serum
Environment DNA nanostructure Dimensions/mass Modifications for stability Reported stability Reporting Notes Ref. (year)
Abbreviations: fetal bovine serum (FBS), Dulbecco's modified Eagle's medium (DMEM), Roswell Park Memorial Institute (RPMI) medium, Tris acetate EDTA (TAE), agarose gel electrophoresis (AGE), polyacrylamide gel electrophoresis (PAGE), transmission electron microscopy (TEM), hexaethylene glcol (HEG), single stranded DNA (ssDNA), phosphorothioate (PS), half-life (τ1/2), hours (h), internal (int).
Media/buffers with serum
DMEM-Mg with 10% FBS, 37 °C ssDNA 63, 96 nt/24, 36 kDa ± HEG termination ssDNA τ1/2: 0.8–4 h PAGE denat. Cy dye position: 3′ > int > 5′ 52 (2019)
Tetrahedron (Tet.) 7 nm/82 kDa 3′, int., 5′ Cy dyes Tet. τ1/2: 7–8 h PAGE HEG termination > native
Cube 7 nm/124 kDa Cube τ1/2: 3.5–5.5 h Tet. ≫ cube > ssDNA
DMEM with 10% FBS, 37 °C Disc origami 62 nm dia./4 MDa Disc <12 h AGE No degradation observed. 5 (2022)
Donut origami 44 nm dia./4 MDa Donut <24 h
Sphere origami 42 nm dia./4 MDa Sphere <12 h
RPMI with 10% FBS, 37 °C Octahedron origami 45 nm dia./4 MDa <24 h AGE, TEM No degradation observed. 79 (2014)
DMEM with 10% FBS, 37 °C Triangular prism 10 nm/160 kDa PS τ 1/2: 2–7 h (decreased w/PS) AGE, PAGE, denat. PAGE While PS decreased structure stability, single strand stability increased 111 (2014)
10% FBS, 37 °C Tetrahedron (Tet.) 7 nm/82 kDa Enzymatic ligation FBS: Tet. >24 h, DS ∼4 h AGE Tet. >3× more stable than DS 78 (2009)
Linear dsDNA (DS) 63 bp/41 kDa DNase: Tet. <10 min, DS ∼3 min
50% FBS, 37 °C Tetrahedron (Tet.) 7 nm/82 kDa Tet. <24 h AGE Tet. >4× more stable than DS 99 (2011)
Duplex 63 bp/41 kDa Duplex <4 h
DMEM with 10% FBS, 37 °C Triangular prism 10 nm/160 kDa HEG ended DNA oligos Prism: ∼2 h, HEG prism: >24 h PAGE HEG >10× increased stability 67 (2013)
ssDNA 63 bp/41 kDa ssDNA: <1 h, HEG ssDNA: 28 h
RPMI with 10% FBS, 37 °C Barrel origami 60 nm dia./4 MDa Oligolysine (K4–K20) For native/K10/K10:P5k in FBS: τ1/2 = 5 min/55 min/36 h AGE, TEM Best nuclease stability with K10:P5k 65 (2017)
PEG oligolysine (K10:P1k–10k)
10% FBS, 37 °C Tetrahedron (Tet.) 7 nm/82 kDa Enzymatic ligation (Tet., Oct.) Native structures: τ1/2 ∼12 h AGE Tet. most stable structure 69 (2019)
Octahedron (Oct.) 11 nm/180 kDa Click-chemistry ligation (CM) Ligated structures: τ1/2 ∼36 h
Chainmail rod (CM) 25 nm/180 kDa Native origami: τ1/2 ∼24 h
Rectangular origami 90 × 60 nm/4 MDa
10% FBS in TAE + Mg, 37 °C Pyramid 10 nm/94 kDa 2′-Fluoro-RNA (2F) Native structures: <1 h AGE >24× improved stability with modified backbones 71 (2019)
Triangular prism 12 nm/105 kDa 2′-O-Methyl-RNA (2OMe) 2F structures: >24 h
Cube 12 nm/140 kDa Enantiomeric L-DNA 2OMe structures: >24 h
Rugby-ball 18 nm/140 kDa L-DNA structures: >24 h


Table 4 Stability of DNA NS in buffers and media, buffers with nucleases, and intra/extracellular
Environment DNA nanostructure Dimensions/mass Modifications for stability Reported stability Reporting Notes Ref. (year)
Buffers and media
1× PBS 6-HB origami 7 nm/36 kDa T m = 51 °C in PBS T m (melting temperature) No added peptide stability 48 (2021)
DMEM-Mg, 37 °C Disk origami 60 nm dia./4.5 MDa DMEM: >5 h AGE No serum or nucleases 21 (2022)
MES buffer pH 5.5, 37 °C MES: >5 h
RPMI media low Mg, 37 °C Barrel origami (×4) 30–90 nm dia./4 MDa Oligolysine (K4–K20) All but PEI stabilized AGE, TEM Best salt stability with K6 65 (2017)
5 mM Tris with 1 mM EDTA, 37 °C Block origami 50 nm/4 MDa PEG oligolysine Structures in RPMI All oligolysines stabilized all structures
24/6-HB origami 100–400 nm/4 MDa Spermidine, spermine PEI compromised structures
Octahedron origami 60 nm dia./4 MDa Polyethylenimine (PEI) R6 compromised shape
Oligoarginine (R6)
6 M urea, 23–42 °C Triangle origami 120 nm/4 MDa Strand redesign Up to 30 min AFM Ligation increased Tm by 8 °C 112 (2019)
Enzymatic ligation
 
Buffers with nucleases
1 U DNase I 18-HB origami 140 nm/4 MDa Origami:<1 h in DNase I AGE, TEM Origami stable in all but DNase I and T7 exo 80 (2011)
10 U endonucl. (T7, MseI) 24-HB origami 100 nm/4 MDa–70 nm/4 MDa Partial degradation in 1 h by T7 exo Origami ≫ plasmid
10 U exonucl. (T7, E. coli, lambda) 32-HB origami 5309 bp/3.3 MDa Plasmid: <5 min in DNase I
All in NE buffer 4, 37 °C pET24b ds plasmid
1 U μL−1 DNase I, 37 °C Wide barrel origami 90 nm dia./4 MDa PEG oligolysine (K10P) Native: τ1/2 < 1 min AGE, TEM K10P: ∼400-fold improved 81 (2020)
Barrel origami 60 nm dia./4 MDa Crosslinked K10P (×K10P) With K10P: τ1/2 ∼16 min ×K10P: ∼105-fold improved
With ×K10P: τ1/2 ∼66 h Over native structures
0.2 U DNase I, 37 °C Tetrahedron (Tet.) 7 nm/82 kDa Enzymatic ligation Tet. <10 min, DS ∼3 min AGE Tet. >3× more stable than DS 78 (2009)
Linear dsDNA (DS) 63 bp/41 kDa
RPMI with varying DNase I, 37 °C Barrel origami 60 nm dia./4 MDa Oligolysine (K4–K20) DNase for degradation in 1 h: 0.5 U mL−1 native, >500 U mL−1 K10:P5k AGE, TEM Best nuclease stability with K10:P5k 65 (2017)
PEG oligolysine (K10:P1k–10k)
 
Intra/extracellular
Cell cytosol (COS-1, fibroblasts, astrocytes, A549, and HeLa cells) Tetrahedron (Tet.) 7 nm/82 kDa Tet. >1 h in all cell types Three-dye FRET Tetrahedron ≫ crosshair 86 (2022)
4-Arm crosshair (4ac) 23 nm/104 kDa 4ac <15 min in HeLa cells
Cell lysate at 4 °C, 25 °C Rectangle origami 90 × 60 nm/4 MDa All structures stable >12 h AGE, TEM, AFM No degradation observed. 113 (2011)
Triangle origami 120 nm/4 MDa
Block origami 30 nm/4 MDa
70% human serum, 37 °C Nanotweezer (Ntw) 10 nm/53 kDa Ntw: >24 h in serum/blood AGE, FRET DS more stable than Ntw 62 (2015)
97.5% whole human blood, 37 °C dsDNA probe (DS) 22 bp/17 kDa DS: >24 h in serum/blood
ssDNA 32 nt/10 kDa ssDNA: <2 h in serum/blood
Flowing whole blood Tetrahedron (Tet.) 7 nm/82 kDa Tet. >1 h in all cell types Three-dye FRET Tetrahedron ≫ crosshair 9 (2018)
4-Arm crosshair (4ac) 23 nm/104 kDa 4ac <15 min in HeLa cells


In vitro studies have played a particularly outsized role in deciphering the relationship between DNA NS design and resilience in physiological environments, owing to the fine-grained control of environmental conditions that cannot be achieved in vivo, as well as compatibility with large, multiplexed assays that can probe many design criteria and conditions simultaneously. Studies of the resilience of DNA NS against nucleases have shown that stability varies widely depending on the physical properties of the NS, as highlighted in several recent reviews,32,39,40,59–61 and structures with greater topological complexity, helical density, and crossover density have been shown to possess greater resistance to degradation by nucleases.62–64 Unfortunately, DNA NS with these characteristics tend to possess reduced thermal stability and require higher divalent cation concentrations to remain stable in solution. This is particularly notable in closely packed structures with high charge densities and to a lesser extent in structures with high crossover densities, often achieved with crossovers at nonideal positions and/or by shortening the length of complementary domains in the structure.63,64 Despite the potential drawbacks, these modifications can improve the stability of native DNA NS in the presence of nucleases by an order of magnitude or more and are thus useful tools in the design of nuclease-resistant nanostructures.

Beyond the use of native DNA NS, many strategies to modify and/or protect DNA NS in biological environments have been successfully demonstrated, such as polymer coatings adhered to structures electrostatically or chemically,41,65–67 staple strand ligation and cross-linking (Fig. 2) with chemical,68,69 enzymatic,69 and photoactivated strategies,70 incorporation of nucleic acids with nuclease-resistant backbones such as enantiomeric L-DNA,71 phosphorothioate DNA,72 peptide nucleic acids (PNA),77 and various modified RNAs,73,74 as well as protection with hard coatings such as silica.75,76 Such modifications can potentially extend the lifetimes of DNA NS in biological environments beyond that achievable with native structures, often without compromising and sometimes improving thermal and low-salt stability. A recent review article by Chandrasekaran highlights several approaches to creating nuclease resistance DNA NS.40Fig. 2 is an example of small DNA NS that possess better serum stability when the strands are covalently linked within them versus when they are not.69 From the various reports in the literature gathered here, some general trends for structures in similar environments can be identified. The lifetime of single-stranded and duplexed DNA prior to assembly into DNA NS is typically on the order of an hour or less in serum/media and 10 min or less in buffer with nucleases.52,67,78 The lifetimes of native DNA NS are improved to several hours or longer in serum/media,5,69,79 though native structures are often fully degraded within an hour in buffer with nucleases.78,80,81 Modified DNA NS tend to have lifetimes in serum/media on the order of days and several hours in buffer with nucleases,65,69,71 though some oligolysine-based coatings have been shown to extend lifetimes to days even in exposure to high nuclease concentrations.81


image file: d2nr05868e-f2.tif
Fig. 2 Stability of selected DNA NS in FBS containing serum without and with non-covalent (nc) linkage. The NS studied, namely, (a and b) tetrahedral (TD) and (c and d) chainmail (CM), were covalently interlocked via alkyne–azide linkage between ends of adjacent DNA molecules. Stability was studied in the presence of 10% FBS serum and measured via agarose gel electrophoresis with the results of the corresponding densitometric analysis shown in the accompanying plots. Reproduced from ref. 69 with permission from the Royal Society of Chemistry. Copyright 2019.

It remains a challenge to identify more than basic trends from the consolidated studies of DNA NS stability due to a general lack of standardization and best practices between studies. Studies often vary in several ways between nanostructure design, buffer conditions, nuclease activity, types of nucleases, methods of characterizing structural integrity, and reported measures of stability.1,32,40,82 This makes it extremely hard to correlate results amongst these studies. Additionally, it is not yet common practice to calibrate the enzymatic activity of nucleases in sera and buffers, which can vary with aging and handling, prior to their use,32,61,79 thus it is possible for studies to report different values for stability of the same DNA NS under identical conditions. Until some form of standards are adopted, we must rely on the consistency within individual studies to elucidate the preliminary rules dictating DNA NS stability in biological environments.

Conclusions and future outlook

From reviewing the consolidated summary of DNA NS uptake behavior (Table 1), it can be surmised that the overall size of the NS tested thus far ranges from a few nm (with 7 nm tetrahedron being the dominant test-bed) to 100 nm per side, which translates to an approximate range of 30 kilodaltons (kDa) up to 4 megadaltons (MDa) in terms of globular mass. The arbitrarily different DNA NS that have been administered in uptake related studies, however, do not bear a direct correlation to NS mass, making it challenging to compare their performance in a systematic manner. This review also does not focus on the intracellular mechanisms of DNA NS endocytosis since a definitive consensus or enough data is still being sought by the field. A similar issue emerges for the stability studies highlighted in Tables 3 and 4, and if these issues persist, every DNA NS employed for therapeutic applications will require a case-by-case stability evaluation. It would be extremely beneficial if the field could collaborate and actively formulate a protocol for standardized quality control expected from each study that will be undertaken in the future.83 Such assay cascades have been put into place for other nanomaterials being developed for potential clinical use.84

The progress in recent decades on biomedically applied DNA NS is exemplative of a promising role of the field in targeted and combinatorial therapeutic interventions. Future directions that the field has already seen some movement towards are in vaccine development through strategic antigen display or through delivery of antigenic cargo to immune cells. Combinatorial therapies, wherein adjuvants such as CpG, nanoparticles, and intercalating drugs are simultaneously delivered via a targeted DNA NS, are also of high interest. Lastly, delivery of functional nucleic acids through DNA NS such as silencing RNA (siRNA), or even epitope-expressing genes, warrant further investigation.

Towards achieving a status that currently favors lipid and viral-based nanoparticles as delivery systems, a few concerted efforts are needed and already underway. Consolidation into a repository of DNA NS that have been designed and engineered by the rapidly growing expert community will help repurpose successful NS in application-specific translational studies, much like the tetrahedron (originally designed by Goodman et al.37), has become prototypical in fundamental studies. Nanobase.org by Poppleton et al. is one such endeavor that is creating a database of DNA and RNA NS.85 From such collaborations, one can anticipate certain DNA NS to emerge (or evolve) as superior cell- and intervention-specific (immune-stimulation versus nucleic acid delivery versus pharmacochemical delivery) options, but will more importantly be informed with recommendations on dosage, targeting moieties, and site-directed modifications by relevant studies in the literature.

The need of the hour is more cell-directed studies elucidating the programmable release and stability of DNA NS inside the cell. Fig. 3 shows results from a recent report where dye-labeled DNA structures were directly microinjected into the cytosol of a variety of different primary and transformed cells and the stability of the structures evaluated by tracking the resulting FRET over time.86 This format was exploited to bypass the complications of endocytic uptake so that the only variable became that of cytosolic stability. This study confirmed that the high-torsion DNA tetrahedron is more stable in the cellular cytosol than linear structures. Perhaps it is worth exploring the feasibility of adopting techniques of cytosolic delivery through endocytosis that have shown promise for other non-nucleic acid nanoparticles. Like DNA NS, calcium phosphate nanoparticles (CPN) aim to be carriers that introduce medication into the cell in a noninvasive manner.87,88 Both nanoparticle types are taken into the cell via endocytosis and face stability issues as they are transported into the lysosome.87,88 Some stability issues can be alleviated if the CPNs can undergo endosomal escape into the cytoplasm.87–90 Research using CPNs has shown successful endosomal escape of the nanoparticle via destabilization,87,88 thus we can attempt to integrate destabilizing methods, like cationic polymers88,89 and pH sensitive polymers87,89 in DNA NS administration to investigate if the same effect is observed. Furthermore, expanding cytosolic studies to other DNA NS types as well as integration of stabilizing techniques are needed.86


image file: d2nr05868e-f3.tif
Fig. 3 A study on the cytosolic stability of small DNA nanoparticles. Schematic with scale shown of the (a) nanocrosshair and (b) tetrahedron structures whose stability was observed directly in the cytosol of live cells. The location of dye labels and FRET pathway on each structure are also shown. COS-1 cells microinjected with the nanocrosshair (c) and (d) tetrahedron shown at t = 0 min and t = 60 min. After 60 min the FRET changed based on the change in stability of the DNA structure can be seen. (e) Plot of the change in acceptor dye to donor dye fluorescence ratio over time in the nanocrosshair versus tetrahedron DNA structure. (f) Plot of the change in acceptor dye to donor dye fluorescence ratio over time for microinjected samples of the nanocrosshair supplemented with the indicated concentrations of aurintricarboxylic acid nuclease inhibitor providing evidence that degradation resulted from nuclease activity. Reprinted (adapted) with permission from ref. 86 Copyright 2022 American Chemical Society.

Feasibility of DNA NS based therapies at a scale where patient impact is significant are required as well. For example, to what extent are DNA NS based anti-cancer interventions advantageous from an economic standpoint is being evaluated.91 It is also notable that the dosage of DNA NS tested in Tables 1 and 2 are in the nM scale, urging careful evaluation of scalable synthesis needed for translating DNA NS based therapies into in vivo studies, let alone clinical use. Formulations testing oral or topical administration of DNA-based treatments are slowly emerging as well.92,93 Additionally aerosolization, lyophilized storage,94 scalable production,95 and long term stability96,97 of DNA NS have seen preliminary success in some cases but are topics in which ongoing research is actively engaged. In order to realize the widespread applicability of these fascinating materials within a medical context, everything comes back full circle to understanding their fundamental structural integrity and functional properties intracellularly and this still remains the first challenge.

Conflicts of interest

There are no conflicts to declare.

Acknowledgements

The authors acknowledge the US Naval Research Laboratory (NRL), the Office of Naval Research, and the NRL's Nanoscience Institute for programmatic funding. D. M. was supported by the National Institute of Biomedical Imaging and Bioengineering of the National Institutes of Health under award number R00EB030013-03. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

References

  1. D. Mathur and I. L. Medintz, Adv. Healthcare Mater., 2019, 8, e1801546 CrossRef PubMed .
  2. S. Jiang, Z. Ge, S. Mou, H. Yan and C. Fan, Chem, 2021, 7, 1156–1179 CAS .
  3. A. H. Okholm and J. Kjems, Expert Opin. Drug Delivery, 2017, 14, 137–139 CrossRef PubMed .
  4. A. Keller and V. Linko, Angew. Chem., Int. Ed., 2020, 59, 15818–15833 CrossRef CAS PubMed .
  5. A. Udomprasert, C. Wootthichairangsan, R. Duangrat, S. Chaithongyot, Y. Zhang, R. Nixon, W. Liu, R. Wang, M. Ponglikitmongkol and T. Kangsamaksin, ACS Appl. Bio Mater., 2022, 5, 2262–2272 CrossRef CAS PubMed .
  6. Q. Jiang, C. Song, J. Nangreave, X. Liu, L. Lin, D. Qiu, Z. G. Wang, G. Zou, X. Liang, H. Yan and B. Ding, J. Am. Chem. Soc., 2012, 134, 13396–13403 CrossRef CAS PubMed .
  7. S. Palazzolo, M. Hadla, C. Russo Spena, I. Caligiuri, R. Rotondo, M. Adeel, V. Kumar, G. Corona, V. Canzonieri, G. Toffoli and F. Rizzolio, Cancers, 2019, 11, 1997 CrossRef CAS PubMed .
  8. Y. Zeng, J. Liu, S. Yang, W. Liu, L. Xu and R. Wang, J. Mater. Chem. B, 2018, 6, 1605–1612 RSC .
  9. S. Li, Q. Jiang, S. Liu, Y. Zhang, Y. Tian, C. Song, J. Wang, Y. Zou, G. J. Anderson, J. Y. Han, Y. Chang, Y. Liu, C. Zhang, L. Chen, G. Zhou, G. Nie, H. Yan, B. Ding and Y. Zhao, Nat. Biotechnol., 2018, 36, 258–264 CrossRef CAS .
  10. S. Zhao, R. Tian, J. Wu, S. Liu, Y. Wang, M. Wen, Y. Shang, Q. Liu, Y. Li, Y. Guo, Z. Wang, T. Wang, Y. Zhao, H. Zhao, H. Cao, Y. Su, J. Sun, Q. Jiang and B. Ding, Nat. Commun., 2021, 12, 358 CrossRef CAS PubMed .
  11. A. Rangnekar, A. M. Zhang, S. S. Li, K. M. Bompiani, M. N. Hansen, K. V. Gothelf, B. A. Sullenger and T. H. LaBean, Nanomedicine, 2012, 8, 673–681 CrossRef CAS PubMed .
  12. S. Zhao, F. Duan, S. Liu, T. Wu, Y. Shang, R. Tian, J. Liu, Z. G. Wang, Q. Jiang and B. Ding, ACS Appl. Mater. Interfaces, 2019, 11, 11112–11118 CrossRef CAS PubMed .
  13. J. Hellmeier, R. Platzer, A. S. Eklund, T. Schlichthaerle, A. Karner, V. Motsch, M. C. Schneider, E. Kurz, V. Bamieh, M. Brameshuber, J. Preiner, R. Jungmann, H. Stockinger, G. J. Schutz, J. B. Huppa and E. Sevcsik, Proc. Natl. Acad. Sci. U. S. A., 2021, 118, e2016857118 CrossRef CAS PubMed .
  14. J. Hellmeier, R. Platzer, V. Muhlgrabner, M. C. Schneider, E. Kurz, G. J. Schutz, J. B. Huppa and E. Sevcsik, ACS Nano, 2021, 15, 15057–15068 CrossRef CAS PubMed .
  15. R. Dong, T. Aksel, W. Chan, R. N. Germain, R. D. Vale and S. M. Douglas, Proc. Natl. Acad. Sci. U. S. A., 2021, 118, e2109057118 CrossRef PubMed .
  16. T. Fang, J. Alvelid, J. Spratt, E. Ambrosetti, I. Testa and A. I. Teixeira, ACS Nano, 2021, 15, 3441–3452 CrossRef CAS PubMed .
  17. Y. Sakai, M. S. Islam, M. Adamiak, S. C. Shiu, J. A. Tanner and J. G. Heddle, Genes, 2018, 9, 571 CrossRef PubMed .
  18. Y. Wang, J. Zhu, W. Jia, H. Xiong, W. Qiu, R. Xu and Y. Lin, ACS Appl. Mater. Interfaces, 2022, 14, 44228–44238 CrossRef CAS PubMed .
  19. S. Wan, S. Liu, M. Sun, J. Zhang, X. Wei, T. Song, Y. Li, X. Liu, H. Chen, C. J. Yang and Y. Song, ACS Nano, 2022, 16, 15310–15317 CrossRef CAS PubMed .
  20. R. Veneziano, T. J. Moyer, M. B. Stone, E. C. Wamhoff, B. J. Read, S. Mukherjee, T. R. Shepherd, J. Das, W. R. Schief, D. J. Irvine and M. Bathe, Nat. Nanotechnol., 2020, 15, 716–723 CrossRef CAS PubMed .
  21. A. Comberlato, M. M. Koga, S. Nussing, I. A. Parish and M. M. C. Bastings, Nano Lett., 2022, 22, 2506–2513 CrossRef CAS PubMed .
  22. I. Seitz, H. Ijas, V. Linko and M. A. Kostiainen, ACS Appl. Mater. Interfaces, 2022, 14, 38515–38524 CrossRef CAS PubMed .
  23. S. M. Douglas, I. Bachelet and G. M. Church, Science, 2012, 335, 831–834 CrossRef CAS PubMed .
  24. W. Engelen, C. Sigl, K. Kadletz, E. M. Willner and H. Dietz, J. Am. Chem. Soc., 2021, 143, 21630–21636 CrossRef CAS .
  25. P. Pitikultham, Z. Wang, Y. Wang, Y. Shang, Q. Jiang and B. Ding, ChemMedChem, 2022, 17, e202100635 CrossRef CAS PubMed .
  26. K. E. Bujold, J. C. C. Hsu and H. F. Sleiman, J. Am. Chem. Soc., 2016, 138, 14030–14038 CrossRef CAS PubMed .
  27. D. Jiang, Y. Sun, J. Li, Q. Li, M. Lv, B. Zhu, T. Tian, D. Cheng, J. Xia, L. Zhang, L. Wang, Q. Huang, J. Shi and C. Fan, ACS Appl. Mater. Interfaces, 2016, 8, 4378–4384 CrossRef CAS PubMed .
  28. C. R. Lucas, P. D. Halley, A. A. Chowdury, B. K. Harrington, L. Beaver, R. Lapalombella, A. J. Johnson, E. K. Hertlein, M. A. Phelps, J. C. Byrd and C. E. Castro, Small, 2022, 18, e2108063 CrossRef PubMed .
  29. E. Lin-Shiao, W. G. Pfeifer, B. R. Shy, M. Saffari Doost, E. Chen, V. S. Vykunta, J. R. Hamilton, E. C. Stahl, D. M. Lopez, C. R. Sandoval Espinoza, A. E. Deyanov, R. J. Lew, M. G. Poirer, A. Marson, C. E. Castro and J. A. Doudna, Nucleic Acids Res., 2022, 50, 1256–1268 Search PubMed .
  30. D. Jiang, Z. Ge, H. J. Im, C. G. England, D. Ni, J. Hou, L. Zhang, C. J. Kutyreff, Y. Yan, Y. Liu, S. Y. Cho, J. W. Engle, J. Shi, P. Huang, C. Fan, H. Yan and W. Cai, Nat. Biomed. Eng., 2018, 2, 865–877 CrossRef CAS PubMed .
  31. H. Lee, A. K. Lytton-Jean, Y. Chen, K. T. Love, A. I. Park, E. D. Karagiannis, A. Sehgal, W. Querbes, C. S. Zurenko, M. Jayaraman, C. G. Peng, K. Charisse, A. Borodovsky, M. Manoharan, J. S. Donahoe, J. Truelove, M. Nahrendorf, R. Langer and D. G. Anderson, Nat. Nanotechnol., 2012, 7, 389–393 CrossRef CAS PubMed .
  32. C. M. Green, D. Mathur and I. L. Medintz, J. Mater. Chem. B, 2020, 8, 6170–6178 RSC .
  33. P. Wang, M. A. Rahman, Z. Zhao, K. Weiss, C. Zhang, Z. Chen, S. J. Hurwitz, Z. G. Chen, D. M. Shin and Y. Ke, J. Am. Chem. Soc., 2018, 140, 2478–2484 CrossRef CAS PubMed .
  34. G. Vindigni, S. Raniolo, A. Ottaviani, M. Falconi, O. Franch, B. R. Knudsen, A. Desideri and S. Biocca, ACS Nano, 2016, 10, 5971–5979 CrossRef CAS PubMed .
  35. L. Liang, J. Li, Q. Li, Q. Huang, J. Shi, H. Yan and C. Fan, Angew. Chem., Int. Ed., 2014, 53, 7745–7750 CrossRef CAS PubMed .
  36. P. W. Rothemund, Nature, 2006, 440, 297–302 CrossRef CAS PubMed .
  37. R. P. Goodman, R. M. Berry and A. J. Turberfield, Chem. Commun., 2004, 12, 1372–1373 RSC .
  38. Y. Ke, L. L. Ong, W. M. Shih and P. Yin, Science, 2012, 338, 1177–1183 CrossRef CAS PubMed .
  39. S. Manuguri, M. K. Nguyen, J. Loo, A. K. Natarajan and A. Kuzyk, Bioconjugate Chem., 2023, 34, 6–17 CrossRef CAS PubMed .
  40. A. R. Chandrasekaran, Nat. Rev. Chem., 2021, 5, 225–239 CrossRef CAS PubMed .
  41. Y. Ahmadi, E. De Llano and I. Barisic, Nanoscale, 2018, 10, 7494–7504 RSC .
  42. S. Park and K. Hamad-Schifferli, ACS Nano, 2010, 4, 2555–2560 CrossRef CAS PubMed .
  43. A. Chopra, S. Krishnan and F. C. Simmel, Nano Lett., 2016, 16, 6683–6690 CrossRef CAS PubMed .
  44. J. Mikkila, A. P. Eskelinen, E. H. Niemela, V. Linko, M. J. Frilander, P. Torma and M. A. Kostiainen, Nano Lett., 2014, 14, 2196–2200 CrossRef CAS PubMed .
  45. K. Kansara, A. Kumar and D. Bhatia, Appl. Nanomed., 2022, 22, 337 Search PubMed .
  46. M. A. Rahman, P. Wang, Z. Zhao, D. Wang, S. Nannapaneni, C. Zhang, Z. Chen, C. C. Griffith, S. J. Hurwitz, Z. G. Chen, Y. Ke and D. M. Shin, Angew. Chem., Int. Ed., 2020, 59, 12575 CrossRef CAS PubMed .
  47. M. A. Rahman, P. Wang, Z. Zhao, D. Wang, S. Nannapaneni, C. Zhang, Z. Chen, C. C. Griffith, S. J. Hurwitz, Z. G. Chen, Y. Ke and D. M. Shin, Angew. Chem., Int. Ed., 2017, 56, 16023–16027 CrossRef CAS PubMed .
  48. B. Smolkova, T. MacCulloch, T. F. Rockwood, M. Liu, S. J. W. Henry, A. Frtus, M. Uzhytchak, M. Lunova, M. Hof, P. Jurkiewicz, A. Dejneka, N. Stephanopoulos and O. Lunov, ACS Appl. Mater. Interfaces, 2021, 13, 46375–46390 CrossRef CAS PubMed .
  49. A. S. Walsh, H. Yin, C. M. Erben, M. J. Wood and A. J. Turberfield, ACS Nano, 2011, 5, 5427–5432 CrossRef CAS PubMed .
  50. N. Kern, R. Dong, S. M. Douglas, R. D. Vale and M. A. Morrissey, eLife, 2021, 10, 68311 CrossRef .
  51. V. Prakash, K. Tsekouras, M. Venkatachalapathy, L. Heinicke, S. Presse, N. G. Walter and Y. Krishnan, Angew. Chem., Int. Ed., 2019, 58, 3073–3076 CrossRef CAS PubMed .
  52. A. Lacroix, E. Vengut-Climent, D. de Rochambeau and H. F. Sleiman, ACS Cent. Sci., 2019, 5, 882–891 CrossRef CAS PubMed .
  53. X. Liu, Y. Xu, T. Yu, C. Clifford, Y. Liu, H. Yan and Y. Chang, Nano Lett., 2012, 12, 4254–4259 CrossRef CAS PubMed .
  54. H. Ding, J. Li, N. Chen, X. Hu, X. Yang, L. Guo, Q. Li, X. Zuo, L. Wang, Y. Ma and C. Fan, ACS Cent. Sci., 2018, 4, 1344–1351 CrossRef CAS PubMed .
  55. X. Peng, S. Fang, B. Ji, M. Li, J. Song, L. Qiu and W. Tan, Nano Lett., 2021, 21, 6946–6951 CrossRef CAS PubMed .
  56. A. Rajwar, S. R. Shetty, P. Vaswani, V. Morya, A. Barai, S. Sen, M. Sonawane and D. Bhatia, ACS Nano, 2022, 16, 10496–10508 CrossRef CAS PubMed .
  57. C. Sigl, E. M. Willner, W. Engelen, J. A. Kretzmann, K. Sachenbacher, A. Liedl, F. Kolbe, F. Wilsch, S. A. Aghvami, U. Protzer, M. F. Hagan, S. Fraden and H. Dietz, Nat. Mater., 2021, 20, 1281–1289 CrossRef CAS PubMed .
  58. S. Ren, K. Fraser, L. Kuo, N. Chauhan, A. T. Adrian, F. Zhang, R. J. Linhardt, P. S. Kwon and X. Wang, Nat. Protoc., 2022, 17, 282–326 CrossRef CAS PubMed .
  59. N. Stephanopoulos, ChemBioChem, 2019, 20, 2191–2197 CrossRef CAS PubMed .
  60. A. Lacroix and H. F. Sleiman, ACS Nano, 2021, 15, 3631–3645 CrossRef CAS PubMed .
  61. H. Bila, E. E. Kurisinkal and M. M. C. Bastings, Biomater. Sci., 2019, 7, 532–541 RSC .
  62. S. Goltry, N. Hallstrom, T. Clark, W. Kuang, J. Lee, C. Jorcyk, W. B. Knowlton, B. Yurke, W. L. Hughes and E. Graugnard, Nanoscale, 2015, 7, 10382–10390 RSC .
  63. A. R. Chandrasekaran, J. Vilcapoma, P. Dey, S. W. Wong-Deyrup, B. K. Dey and K. Halvorsen, J. Am. Chem. Soc., 2020, 142, 6814–6821 CrossRef CAS PubMed .
  64. Y. Xin, P. Piskunen, A. Suma, C. Li, H. Ijas, S. Ojasalo, I. Seitz, M. A. Kostiainen, G. Grundmeier, V. Linko and A. Keller, Small, 2022, 18, e2107393 CrossRef PubMed .
  65. N. Ponnuswamy, M. M. C. Bastings, B. Nathwani, J. H. Ryu, L. Y. T. Chou, M. Vinther, W. A. Li, F. M. Anastassacos, D. J. Mooney and W. M. Shih, Nat. Commun., 2017, 8, 15654 CrossRef CAS PubMed .
  66. N. P. Agarwal, M. Matthies, F. N. Gur, K. Osada and T. L. Schmidt, Angew. Chem., Int. Ed., 2017, 56, 5460–5464 CrossRef CAS PubMed .
  67. J. W. Conway, C. K. McLaughlin, K. J. Castor and H. Sleiman, Chem. Commun., 2013, 49, 1172–1174 RSC .
  68. V. Cassinelli, B. Oberleitner, J. Sobotta, P. Nickels, G. Grossi, S. Kempter, T. Frischmuth, T. Liedl and A. Manetto, Angew. Chem., Int. Ed., 2015, 54, 7795–7798 CrossRef CAS PubMed .
  69. S. Raniolo, S. Croce, R. P. Thomsen, A. H. Okholm, V. Unida, F. Iacovelli, A. Manetto, J. Kjems, A. Desideri and S. Biocca, Nanoscale, 2019, 11, 10808–10818 RSC .
  70. T. Gerling, M. Kube, B. Kick and H. Dietz, Sci. Adv., 2018, 4, eaau1157 CrossRef CAS PubMed .
  71. K. R. Kim, S. J. Kang, A. Y. Lee, D. Hwang, M. Park, H. Park, S. Kim, K. Hur, H. S. Chung, C. Mao and D. R. Ahn, Biomaterials, 2019, 195, 1–12 CrossRef CAS PubMed .
  72. V. J. Schuller, S. Heidegger, N. Sandholzer, P. C. Nickels, N. A. Suhartha, S. Endres, C. Bourquin and T. Liedl, ACS Nano, 2011, 5, 9696–9702 CrossRef PubMed .
  73. C. Lin, Y. Ke, Z. Li, J. H. Wang, Y. Liu and H. Yan, Nano Lett., 2009, 9, 433–436 CrossRef CAS PubMed .
  74. K. R. Kim, H. Y. Kim, Y. D. Lee, J. S. Ha, J. H. Kang, H. Jeong, D. Bang, Y. T. Ko, S. Kim, H. Lee and D. R. Ahn, J. Controlled Release, 2016, 243, 121–131 CrossRef CAS PubMed .
  75. L. Nguyen, M. Doblinger, T. Liedl and A. Heuer-Jungemann, Angew. Chem., Int. Ed., 2019, 58, 912–916 CrossRef CAS PubMed .
  76. M.-K. Nguyen, V. H. Nguyen, A. K. Natarajan, Y. Huang, J. Ryssy, B. Shen and A. Kuzyk, Chem. Mater., 2020, 32, 6657–6665 CrossRef CAS .
  77. S. Fairweather, M. Rogers, P. Stoulig, F. O'Murphy, E. Bose, S. Tumusiime, A. Desany, M. Troisi, A. Alvarado, E. Wade, K. Cruz, K. Wlodychak and A. J. Bell Jr., Biophys. Chem., 2022, 289, 106863 CrossRef CAS PubMed .
  78. J. W. Keum and H. Bermudez, Chem. Commun., 2009, 45, 7036–7038 RSC .
  79. J. Hahn, S. F. Wickham, W. M. Shih and S. D. Perrault, ACS Nano, 2014, 8, 8765–8775 CrossRef CAS PubMed .
  80. C. E. Castro, F. Kilchherr, D. N. Kim, E. L. Shiao, T. Wauer, P. Wortmann, M. Bathe and H. Dietz, Nat. Methods, 2011, 8, 221–229 CrossRef CAS PubMed .
  81. F. M. Anastassacos, Z. Zhao, Y. Zeng and W. M. Shih, J. Am. Chem. Soc., 2020, 142, 3311–3315 CrossRef CAS PubMed .
  82. K. A. Afonin, M. A. Dobrovolskaia, G. Church and M. Bathe, ACS Nano, 2020, 14, 9221–9227 CrossRef CAS PubMed .
  83. J. J. Rennick, A. P. R. Johnston and R. G. Parton, Nat. Nanotechnol., 2021, 16, 266–276 CrossRef CAS PubMed .
  84. NCI, Assay Cascade Characterization Program, https://www.cancer.gov/nano/research/ncl/assay-cascade, (accessed September 2, 2022).
  85. E. Poppleton, A. Mallya, S. Dey, J. Joseph and P. Sulc, Nucleic Acids Res., 2022, 50, D246–D252 CrossRef CAS PubMed .
  86. D. Mathur, K. E. Rogers, S. A. Diaz, M. E. Muroski, W. P. Klein, O. K. Nag, K. Lee, L. D. Field, J. B. Delehanty and I. L. Medintz, Nano Lett., 2022, 22, 5037–5045 CrossRef CAS PubMed .
  87. A. Maitra, Expert Rev. Mol. Diagn., 2005, 5, 893–905 CrossRef CAS PubMed .
  88. F. Pittella, M. Zhang, Y. Lee, H. J. Kim, T. Tockary, K. Osada, T. Ishii, K. Miyata, N. Nishiyama and K. Kataoka, Biomaterials, 2011, 32, 3106–3114 CrossRef CAS PubMed .
  89. T. F. Martens, K. Remaut, J. Demeester, S. C. De Smedt and K. Braeckmans, Nano Today, 2014, 9, 344–364 CrossRef CAS .
  90. D. Ma, Nanoscale, 2014, 6, 6415–6425 RSC .
  91. E. L. Coleridge and K. E. Dunn, Biomed. Phys. Eng. Express, 2020, 6, 065030 CrossRef PubMed .
  92. G. Yang, H. E. Lee, S. W. Shin, S. H. Um, J. D. Lee, K.-B. Kim, H. C. Kang, Y.-Y. Cho, H. S. Lee and J. Y. Lee, Adv. Funct. Mater., 2018, 28, 1801918 CrossRef .
  93. M. Baig, S. Khan, M. A. Naeem, G. J. Khan and M. T. Ansari, Biomed. Pharmacother., 2018, 97, 1250–1258 CrossRef CAS PubMed .
  94. B. Zhu, Y. Zhao, J. Dai, J. Wang, S. Xing, L. Guo, N. Chen, X. Qu, L. Li, J. Shen, J. Shi, J. Li and L. Wang, ACS Appl. Mater. Interfaces, 2017, 9, 18434–18439 CrossRef CAS PubMed .
  95. F. Praetorius, B. Kick, K. L. Behler, M. N. Honemann, D. Weuster-Botz and H. Dietz, Nature, 2017, 552, 84–87 CrossRef CAS PubMed .
  96. Y. Xin, C. Kielar, S. Zhu, C. Sikeler, X. Xu, C. Moser, G. Grundmeier, T. Liedl, A. Heuer-Jungemann, D. M. Smith and A. Keller, Small, 2020, 16, e1905959 CrossRef PubMed .
  97. C. Kielar, Y. Xin, X. Xu, S. Zhu, N. Gorin, G. Grundmeier, C. Moser, D. M. Smith and A. Keller, Molecules, 2019, 24, 2577 CrossRef CAS PubMed .
  98. S. Liu, Q. Jiang, X. Zhao, R. Zhao, Y. Wang, Y. Wang, J. Liu, Y. Shang, S. Zhao, T. Wu, Y. Zhang, G. Nie and B. Ding, Nat. Mater., 2021, 20, 421–430 CrossRef CAS PubMed .
  99. J. Li, H. Pei, B. Zhu, L. Liang, M. Wei, Y. He, N. Chen, D. Li, Q. Huang and C. Fan, ACS Nano, 2011, 5, 8783–8789 CrossRef CAS PubMed .
  100. S. Kocabey, H. Meinl, I. S. MacPherson, V. Cassinelli, A. Manetto, S. Rothenfusser, T. Liedl and F. S. Lichtenegger, Nanomaterials, 2014, 5, 47–60 CrossRef PubMed .
  101. K. E. Bujold, J. Fakhoury, T. G. W. Edwardson, K. M. M. Carneiro, J. N. Briard, A. G. Godin, L. Amrein, G. D. Hamblin, L. C. Panasci, P. W. Wiseman and H. F. Sleiman, Chem. Sci., 2014, 5, 2449–2455 RSC .
  102. K. R. Kim, J. Kim, J. H. Back, J. E. Lee and D. R. Ahn, ACS Nano, 2022, 16, 7331–7343 CrossRef CAS PubMed .
  103. Y. X. Zhao, A. Shaw, X. Zeng, E. Benson, A. M. Nystrom and B. Hogberg, ACS Nano, 2012, 6, 8684–8691 CrossRef CAS PubMed .
  104. Z. Xia, P. Wang, X. Liu, T. Liu, Y. Yan, J. Yan, J. Zhong, G. Sun and D. He, Biochemistry, 2016, 55, 1326–1331 CrossRef CAS PubMed .
  105. Y. Jiang, H. Zhou, W. Zhao and S. Zhang, Anal. Chem., 2022, 94, 6771–6780 CrossRef CAS PubMed .
  106. M. M. C. Bastings, F. M. Anastassacos, N. Ponnuswamy, F. G. Leifer, G. Cuneo, C. Lin, D. E. Ingber, J. H. Ryu and W. M. Shih, Nano Lett., 2018, 18, 3557–3564 CrossRef CAS PubMed .
  107. C. X. Liu, B. Wang, W. P. Zhu, Y. F. Xu, Y. Y. Yang and X. H. Qian, Angew. Chem., Int. Ed., 2022, 61, e202205509 CAS .
  108. D. Bhatia, S. Surana, S. Chakraborty, S. P. Koushika and Y. Krishnan, Nat. Commun., 2011, 2, 339 CrossRef PubMed .
  109. M. Fu, L. Dai, Q. Jiang, Y. Tang, X. Zhang, B. Ding and J. Li, Chem. Commun., 2016, 52, 9240–9242 RSC .
  110. K. R. Kim, Y. D. Lee, T. Lee, B. S. Kim, S. Kim and D. R. Ahn, Biomaterials, 2013, 34, 5226–5235 CrossRef CAS PubMed .
  111. J. J. Fakhoury, C. K. McLaughlin, T. W. Edwardson, J. W. Conway and H. F. Sleiman, Biomacromolecules, 2014, 15, 276–282 CrossRef CAS PubMed .
  112. S. Ramakrishnan, L. Scharfen, K. Hunold, S. Fricke, G. Grundmeier, M. Schlierf, A. Keller and G. Krainer, Nanoscale, 2019, 11, 16270–16276 RSC .
  113. Q. Mei, X. Wei, F. Su, Y. Liu, C. Youngbull, R. Johnson, S. Lindsay, H. Yan and D. Meldrum, Nano Lett., 2011, 11, 1477–1482 CrossRef CAS PubMed .

This journal is © The Royal Society of Chemistry 2023