Europe PMC

This website requires cookies, and the limited processing of your personal data in order to function. By using the site you are agreeing to this as outlined in our privacy notice and cookie policy.

Abstract 


Nucleic acid aptamers, also regarded as chemical antibodies, show potential as targeted therapeutic and delivery agents since they possess unique advantages over antibodies. Generated by an iterative selection and amplification process from oligonucleotide libraries using cultured cells, the aptamers bind to their target molecules expressed on the cell surface. Excitingly, most aptamers also demonstrate a cell-internalizing property in native living cells, allowing them to directly enter the cells via endocytosis depending on the target. In this review, we discuss selection methods in generating cell-internalizing aptamers via a cell-based selection process, along with their challenges and optimization strategies. We highlight the cellular uptake routes adopted by the aptamers and also their intracellular fate after the uptake, to give an overview of their mechanism of action for applications as promising pharmacological agents.

Free full text 


Logo of rscmedchemLink to Publisher's site
RSC Med Chem. 2021 Oct 20; 12(10): 1640–1649.
Published online 2021 Jul 24. https://doi.org/10.1039/d1md00199j
PMCID: PMC8528270
PMID: 34778766

Uptake mechanisms of cell-internalizing nucleic acid aptamers for applications as pharmacological agents

Abstract

Nucleic acid aptamers, also regarded as chemical antibodies, show potential as targeted therapeutic and delivery agents since they possess unique advantages over antibodies. Generated by an iterative selection and amplification process from oligonucleotide libraries using cultured cells, the aptamers bind to their target molecules expressed on the cell surface. Excitingly, most aptamers also demonstrate a cell-internalizing property in native living cells, allowing them to directly enter the cells via endocytosis depending on the target. In this review, we discuss selection methods in generating cell-internalizing aptamers via a cell-based selection process, along with their challenges and optimization strategies. We highlight the cellular uptake routes adopted by the aptamers and also their intracellular fate after the uptake, to give an overview of their mechanism of action for applications as promising pharmacological agents.

I. Introduction

Nucleic acid aptamers are synthetic single-stranded oligonucleotides that bind specifically to their target molecules when the aptamers are folded into unique tertiary conformations depending on their sequences. The binding affinities of aptamers to target molecules are the results of various binding forces such as hydrophobic interactions, hydrogen bonding, van der Waals interactions, aromatic ring stacking, and shape complementary fitting.1

Emerging as an alternative to antibodies in diagnostics and therapeutics,2 the aptamers are selected from a pool of 1014–1016 randomized nucleic acid library, through an in vitro molecular evolution method called Systematic Evolution of Ligands by EXponential enrichment (SELEX).3,4

Dubbed as nucleic acid antibodies, the best-elected aptamers possess notable advantages over protein-based antibodies, such as better stability, high affinity and specificity, more structural flexibility for various types of chemical modifications, and being small in size (approximately 10 to 30 kDa).5,6 These properties allow aptamers to bind stably to more specific epitopes, especially at low antigen density.6 Remarkably, aptamers also demonstrate a cell-internalizing property in native living cells, which means that they could directly enter the cells without the need for permeabilization, via a receptor-dependent system.7

Despite their distinct advantages, however, applications of aptamers are still less explored in comparison with antibodies (antibodies have been studied for more than a century). Antibodies have been largely exploited as labelling and imaging agents, in addition to being popularly used as a carriage in targeted drug delivery via antibody–drug conjugates (ADCs).8 While antibodies are proven to be powerful tools in diagnostic and therapeutic applications, they are not without limitations. For instance, most monoclonal antibodies can only be produced biologically in animals with high manufacturing cost, in addition to complicated quality control. Antibodies also suffer from poor cell/tissue penetration due to their large size (~150 kDa), which then makes intracellular target labelling under native conditions not feasible. Consequently, in recent years, researchers have been attracted to the utilization of cell-internalizing aptamers as pharmacological agents in diagnostics and therapeutics.

Internalization of aptamers is particularly crucial for in vivo applications in targeted delivery or therapeutics. Several comprehensive reviews on modification strategies and applications of modified intracellular aptamers have recently been published. Hirao's group reported developments in the creation of artificial extra base pairs for enhanced properties of DNA aptamers.9 In early 2021, Hollenstein's group discussed non-native modifications along with the challenges faced in the design and synthesis of novel modified oligonucleotides.10 Rossi's and Obika's groups reported nice reviews on versatile intracellular applications of aptamers and biological applications of nucleic acid analogues, respectively.11,12 In this review, hence, we will focus our discussion on the various methods for generating cell-internalizing aptamers particularly via cell-based SELEX. Importantly, we will highlight the internalization property of the aptamers, along with their uptake mechanisms and subcellular localization in cells.

II. Aptamer generation by cell-SELEX

Aptamers are largely generated in vitro through the SELEX technique, which provides a tunable process in a controlled evolutionary manner, by increasing the stringent conditions to select high-affinity and high-specificity aptamers to targets of interest. A randomized oligonucleotide library with 1014–1016 diversity is used to isolate oligonucleotides that bind to the selected targets, such as small molecules, sugars, peptides or proteins.3,4,13,14 Once aptamer sequences are obtained by SELEX, the winning aptamers are synthesized chemically in a large scale production.

In aptamer isolation against proteins, the SELEX process requires high-quality purified recombinant proteins to eliminate off-target binders. However, some pharmacologically relevant proteins, such as G protein-coupled receptors (GCPRs) and serine/threonine kinase receptors, cannot be fully purified in their intact forms, due to their complexity in the purification process.15 Additionally, when a protein-binding aptamer is obtained via SELEX using recombinant proteins, more often than not, the aptamer would bind to the region of the proteins involving their membrane-embedded and/or intracellular domains, not exclusively to the preferable extracellular cell-surface domain. Consequently, this method is not preferable for cell-surface protein targets.

To this end, the SELEX method against whole living cells (termed as cell-SELEX) has emerged. Cell-SELEX allows the generation of aptamers which bind to the cell-surface areas of target molecules in their native state without prior knowledge of the target identity.16,17 Since the aptamer recognizes its targets in their native conformations on the target cells, it also leads to the discovery of novel biomarkers and unexpected cellular events.18 In addition, target recognition for biological applications in in vivo settings is more likely. Toward practical use in the required settings, high affinities to the targets are also one of the key challenges to be attained.

Similar to SELEX targeting simple molecules, the process for cell-SELEX involves repeated cycles of selection and amplification (Fig. 1). The main key process can be divided into three steps for each cycle:

An external file that holds a picture, illustration, etc.
Object name is d1md00199j-f1.jpg
Simplified schematic representation of the cell-SELEX process. Each cycle starts with incubation of the oligonucleotide library with the target cells, followed by removal of the unbound oligonucleotides by washing to collect the cell-bound oligonucleotides, and finally ends with amplification of the cell-bound oligonucleotides by PCR.

i) incubation of an oligonucleotide library with target cells;

ii) removal of unbound oligonucleotides and collection of the cell-bound oligonucleotides;

iii) amplification of the cell-bound oligonucleotides by PCR for preparation of the next selection–amplification cycle.

The cycle is repeated for several rounds until the oligonucleotides that specifically and tightly bind to the target cells are sufficiently enriched, and they are subsequently sequenced to identify the isolated aptamer candidates. Usually, a pre- and/or post-counter selection step by using control cells with no potential targets of interest is implemented in the selection cycles, to eliminate nonspecific binders. This step will then enhance the target specificity of the enriched oligonucleotides.

A. Challenges in aptamer generation using cell-SELEX

The cell-SELEX technique is a rather complex process and has some technical challenges. The protocol depends on the quality and viability of the cells used during the selection process. Hence, proper cell culture maintenance is crucial. This includes appropriate growth media conditions and consistent cell confluency and cell passage number used in the procedure.19 Poor cell culture conditions and prolonged cell growth would not only affect the cell morphology, but also perturb the native conformation and expression level of target proteins on the cell surface, which consequently reduce the efficiency of aptamer generation by cell-SELEX.1,16

Interference from dead cells is highly undesirable during the selection process. The presence of dead cell population would lead to nonspecific binding of the aptamers which causes false positive sequences (due to enrichment of off-target or unwanted sequences), resulting in selection failure.19,20 Mayer's group reported the implementation of the fluorescence-activated cell sorting (FACS) technique in the selection procedure to separate certain subpopulations of cells from other subpopulations, such as dead-cell/live-cell populations, within cultured cell mixtures.19 This technique has been shown to serve as a more efficient route for reducing false positives in cell-SELEX.

False positives can also arise due to nonspecific internalization of DNA aptamers into target cells when incubation is performed at physiological temperature. For this reason, the aptamer–target cell incubation process is commonly performed at 4 °C, despite the concern that aptamers selected at low temperatures may display dissimilar binding affinity and specificity to those selected at physiological temperatures.16

Some other approaches emerged to eliminate false positives in aptamer selection. For instance, Tan's group recently developed a strategy to remove nonspecific and weak binders from the living cell surface via aptamer displacement reaction using complementary DNA (cDNA), toehold-mediated cDNA (tcDNA), and single-stranded binding protein (SSB).21 In the study, they demonstrated that nonspecific cell surface-bound ssDNAs interact tightly with SSB, while the winning aptamer sequence binds to its target cells firmly (Fig. 2A). On this basis, non-specific binders with low affinity can be separated from target cells by displacement reaction, and eventually could be eliminated from the selection process by washing or centrifugation. This hence improves the efficiency of cell-SELEX. Note that fast kinetics to minimize the internalization of nonspecific aptamers is essential for the success of the displacement reaction.21

An external file that holds a picture, illustration, etc.
Object name is d1md00199j-f2.jpg
A) Schematic representation of SSB-assisted cell-SELEX and conventional cell-SELEX. B) Schematic illustration of the cell-internalization SELEX.

B. Cell-SELEX and its variety

Cell-SELEX technology has become increasingly popular. Many aptamers are generated through this technique on more than 80 different cell lines targeting various proteins.18Table 1 summarizes some examples of aptamers obtained by cell-SELEX and their identified target proteins.

Cell-specific aptamers obtained by cell-SELEX, and their target proteins. The aptamers' names along with their types and sequence/nucleotide lengths are included
Target cellTarget proteinAptamer nameAptamer typeAptamer lengthRef.
T cell acute lymphoblastic leukemia CCRF-CEM cellsProtein tyrosine kinase-7 (PTK7)sgc8DNA88 45
Acute myelogenous leukemia NB4 cellsSialic-acid-binding immunoglobulin-like lectins (siglec-5)K19DNA285 46
Burkitt's lymphoma Ramos B cell linesImmunoglobulin heavy muTD05DNA48 47
Glioblastoma U251 cellsTenascin-CGBI-10DNA69 48
Differentiated neuroblastoma SH-SY5Y cellsNeural cell adhesion molecule L1 (L1CAM)yly12DNA51 49
Lung adenocarcinoma cellsCathepsin DLC-183DNA80 50
Breast cancer MCF-7 cellsAnnexin A2ACE4RNA95 51
Pancreatic adenocarcinoma PL45 cellsCD71XQ-2dDNA56 52
Ovarian cancer TOV-21G cellsStress-induced phosphoprotein 1 (STIP1)TOV6DNA84 53
Glioblastoma U-87 MG cellsIntegrin α5β1H02RNA75 54

While the cell-SELEX strategy has successfully generated aptamers as promising tools in cell detection and therapy,22,23 there are two major issues that may limit their usefulness for greater applications as delivery and therapeutic agents: 1) susceptibility of aptamers to degradation by serum or nuclease, and 2) inefficient internalization by living cells.24

Several research groups have attempted to address these problems by improving the selection techniques. Giangrande's group modified the cell-based selection process, termed cell-internalization SELEX, to optimize the cellular uptake by incubating the oligonucleotide library at 37 °C for a period of time and subsequently removing the unbound and surface-bound aptamers by stringent 0.5 M NaCl salt washing (Fig. 2B).25,26 This approach enables the recovery of rapidly-internalized aptamers while eliminating non- and slowly-internalized binders. Thiel et al. generated aptamers that are efficiently internalized into target cells upon binding with their target protein, human epidermal growth factor receptor 2 (HER2). The aptamers were further applied to deliver chemo-sensitizing siRNAs targeting anti-apoptotic gene Bcl-2 in breast cancer cells.25 In another study, insertion of G-rich sequences at the 3′-terminus was largely found to increase cellular uptake, especially for standard phosphodiester oligonucleotides.27

Adding functionality to natural nucleotides could enhance their binding affinity and/or specificity, which enlarges their structural properties and expands their physicochemical properties targeting the cells. The addition can be by introducing either modified nucleotides or unnatural bases as the fifth- and sixth-letters.9,10,28–39 Common strategies to modify nucleotides include modifications on the sugar ring and/or phosphodiester backbone for resisting nuclease degradation, and on the nucleobase for improving binding affinity and specificity.40–44

Our group developed a new cell-SELEX method by introducing an additional highly hydrophobic unnatural base, 7-(2-thienyl)imidazo[4,5-]pyridine (Ds), on top of the four natural nucleotides (A, C, G, T), in a random sequence library.35,55–58 This method using genetic alphabet Expansion SELEX is called cell-ExSELEX34,58 and the bound oligonucleotides are subjected to PCR amplification involving the third base pair, Ds, and its pairing partner, Px (2-nitro-4-propynylpyrrole).57 Contrasting the hydrophilic nature of nucleic acids, the unnatural hydrophobic bases strengthen the interactions between the aptamers and hydrophobic parts of target proteins, resulting in efficient aptamer generation with superior affinity.32,35,59–61 By using the cell-ExSELEX system, our group successfully identified several high-affinity DNA aptamers with excellent stability targeting specifically breast cancer cells, such as MCF-7, MDA-MB-231 and T-47D.58 All of the aptamers are internalized within the bound cells. It was found that one of the aptamers, named 05-MB231, binds to all of the cancer cells that were tested but not to noncancerous cells. Interestingly, the 05-MB231 sequence contains one Ds base that is crucial for its binding affinity. This aptamer also showed outstandingly fast uptake into T-47D cells, followed by efficient internalization and distinct accumulation in the perinuclear region (unpublished data). This shows a promising property for cancer therapy.

Tan's and Benner's group reported an artificially expanded genetic information system (AEGIS) in which they introduced two nonstandard Z and P nucleotides to make a six-letter GACTZP DNA library.36,62 The nucleotide Z contains a nitro functionality not found in the four natural nucleotides. By using the AEGIS cell-SELEX technique, they reported aptamer ZAP-2012 that binds to breast cancer cell line MDA-MB-231 with a dissociation constant value of 30 nM. Importantly, the binding affinity is reduced significantly when Z and/or P is replaced by natural nucleotides, suggesting substantial contributions of the nonstandard nucleotides to the aptamer–target binding interactions.36

Obika et al. utilized cell-internalization SELEX with an artificial DNA library to generate aptamers with high cell-internalizing ability.63 The library contains a hydrophobic uracil derivative (5-((3-indolyl)propionamide-N-allyl)-2′-deoxyuridine, Utrp). The study showed that all Utrp-containing aptamers are internalized into cells more easily compared to the unmodified DNA random library. The internalization ability of the aptamers is reduced when Utrp is substituted with deoxythymidine, signifying the important role of Utrp in the cell-penetrating function of aptamers. The authors then demonstrated a successful application of the base-modified aptamers to deliver antisense oligonucleotides (ASOs) into A549 cells.63

The chemical structures of the modified hydrophobic bases are summarized in Fig. 3.

An external file that holds a picture, illustration, etc.
Object name is d1md00199j-f3.jpg
Chemical structures of the natural (A, T, G, C) and modified bases (Ds, Px, Z, P, Utrp) discussed in the text. R: functional groups.

III. Mechanisms of aptamer internalization and subcellular localization

Internalization of aptamers into a specific cell is one of the important properties for the development of aptamer-based therapeutics, especially in cancer research. To this end, exploration and investigation of their cellular uptake mechanism are of important interest.

Aptamers are widely generated against cell surface targets. Ideally, once they bind to the receptor targets, they would enter the cells via endocytic pathways. Endocytosis is an energy-requiring process, in which internal membranes are formed from the invagination of the plasma membrane. By this means, extracellular materials (macromolecules, liquid, and so on) hence can be internalized into the cells. One protein that largely participates in this fission of membrane process is dynamin.64

A high rate of endocytosis is particularly critical; however, the specific mechanisms underlying aptamer movement into and within target cells have not been widely investigated. There are well-defined endocytic pathways that are involved in the internalization mechanism of aptamers upon binding with their target receptors on the surface of the cell membrane. In general, they include receptor-mediated endocytosis and macropinocytosis.65,66 Receptor-mediated endocytosis can be divided into two types: clathrin-mediated endocytosis (CME) and caveolae-mediated endocytosis (CvME). CME and CvME are regulated by the activities of cargo molecules and other proteins. As aptamer internalization may involve multiple and interchangeable endocytic pathways,67 elucidating the aptamer uptake and cellular fate is essential for a deeper understanding of its potential therapeutic and delivery activities.

Once molecules like aptamers are internalized into mammalian cells, they go through complex intracellular pathways leading to various destinations. Most, if not all, of them are transported into endocytic vessels in the cytoplasmic space and sorted at two major sorting sites, namely the early and late endosomes.68 At the early endosomes, the molecules are either initially sorted to the route back to the cell surface through recycling endosomes or trafficked to lysosomes for degradation.69 This intracellular journey is regulated by intricate protein machinery,69 such as the Rab GTPase family,70 the ESCRT complex71 and the retromer complex.72 Generally, most aptamers are subsequently destined to localize in the endosomes, lysosomes, Golgi apparatus, endoplasmic reticulum, and rarely in the mitochondria.11

A. Clathrin-mediated endocytosis (CME)

CME is the uptake of materials bound to the cellular surface into the cells through the aid of clathrin-coated vesicles. This endocytic portal is fundamental to various plasma membrane activities and crucial to a wide range of eukaryotic cells. The soluble major coat protein, clathrin, forms a triskelion structure composed of three clathrin heavy chains, with tightly associated clathrin light chains. It then polymerizes into a lattice-like coat, which is then also described as “vesicles in a basket”.66

Upon aptamer binding onto the receptor, CME proceeds through several stages: 1) initiation of clathrin-coated pits on the aptamer–receptor binding zone, 2) curvature and invagination of the membrane, 3) fission of the membrane that is catalyzed by dynamin, and 4) the release of clathrin-coated vesicles that are rapidly uncoated and fused with endosomes (Fig. 4A).73

An external file that holds a picture, illustration, etc.
Object name is d1md00199j-f4.jpg
Mechanisms of aptamer internalization. Aptamers can be internalized into cells via several endocytic pathways: A) clathrin-mediated endocytosis (CME), B) caveolae-mediated endocytosis (CvME), and C) macropinocytosis. Most internalized aptamers accumulate in the endosomes, while some are sorted back to the cell surface or other cellular compartments such as lysosomes for degradation.

The common method to investigate the aptamer uptake by CME is by using internalization inhibitors, such as dynasore (a dynamin inhibitor) and chlorpromazine.11,74 DNA aptamer R13, which targets ovarian cancer cells, was reported to possess good targeting ability in tissue sections. The time-dependent internalization of R13 indicated an active uptake process and its uptake occurs through the CME pathway. In the study, it was found that the internalization of R13 decreased significantly in target cells pre-treated with dynasore (inhibitor of CME), but no change upon treatment with amiloride (inhibitor of macropinocytosis).75

The application of aptamers as targeted delivery tools has now become more apparent.76 Aptamers with defined structures would allow assembly with drug molecules, fluorescent dyes or nanoparticles, without loss of function.77 RNA aptamer A10, which has 2′-fluoro modification in pyrimidines, has extensively been studied as a delivery tool. It recognizes prostate-specific membrane-antigen (PSMA) on prostate cancer cells.7 Aptamer A10 was shown to deliver drug-encapsulated nanoparticles to treat tumors in mice and also to carry siRNA molecules into tumor sites, thus possessing mRNA knockdown activity.78,79 This cargo delivery occurs upon the binding of A10 onto cell surface PSMA through a CME mechanism.

The CME mechanism of aptamers is also examined by colocalization studies with fluorescently labeled transferrin. A G-rich DNA aptamer C10.36 (36-nucleotide length) was reported to recognize Burkitt's lymphoma cells selectively. Aptamer C10.36 was folded into a G-quadruplex structure and taken up by the cells with a relatively fast kinetics (a maximum uptake was reached after 15 minutes).80 The colocalization study revealed high staining correlation of C10.36 with transferrin, in which its cellular uptake is mainly characterized via clathrin-mediated endocytosis.81 Moreover, when the cells were pre-incubated with dynasore, the C10.36 uptake was greatly reduced.80

B. Caveolae-mediated endocytosis (CvME)

Similar to CME, the aptamer internalization by CvME is a receptor-mediated process. It involves the formation of caveolae vesicles that is catalyzed by dynamin upon aptamer–receptor interactions. Caveolae are membrane invaginations that constitute membrane proteins called caveolins and cavins, and are augmented with cholesterol and sphingolipid (lipid raft).82 After binding to their targeted surface receptor, aptamers may move along the membrane to caveolae invagination.83 Afterwards, dynamin mediates the fission of caveolae from the plasma membrane, which then turn into cytosolic caveolar vesicles. The vesicles would then reach the caveosome or endosome before being recycled to the plasma membrane (Fig. 4B).84 Lactosylceramide (LacCer), a glycosphingolipid that localizes in lipid rafts, has previously been used as a CvME marker. A tyrosine-kinase inhibitor, genistein, interferes with CvME by disrupting the actin network at the site of endocytosis and inhibiting dynamin II recruitment to the site of internalization, thus disrupting the budding of caveolae.74,85

One of the most studied DNA aptamers is sgc8 that binds to acute lymphoblastic leukemia (ALL) T-cells with high affinity (Kd = 0.8 nM).86 Upon binding with its target protein, human protein tyrosine kinase-7 (PTK7) on the cell membrane, invagination on the plasma membrane binding area is initiated. It subsequently forms an intracellular vesicle that eventually reaches the endosome.45,87

In 2019, Tan group's investigated the endocytic pathway of sgc8 and its aptamer–drug conjugate (ApDC) with anticancer drug 5-fluorouracil (sgc8-5FU), through the single-particle tracking (SPT) technique.88 After treating MCF-7 cells with genistein, the uptake of sgc8-5FU was significantly reduced, indicating that the cellular uptake of the aptamer involves the CvME pathway. In addition, fluorescence imaging with the caveolin protein marker showed that fluorescently labeled-sgc8 exhibited similar fluorescent trajectory to a GFP-tagged caveolin when entering MCF-7 cells, with a rapid and direct motion. The labeled aptamer also displayed a high colocalization degree with the caveolin (~80%) and cholera toxin subunit B (CtxB), a CvME marker (~71%). When observed using the real-time SPT technique, sgc8-5FU exhibited cellular cytoskeleton-dependent transport on the microfilament in the peripheral region and on microtubules from the cell peripheral to the perinuclear area. Confocal microscopy revealed the final location of sgc8-5FU in the lysosome as shown by the high colocalization degree of the Cy3-sgc8-5FU signal with the lysosomal marker LysoTracker near the cell perinuclear region in the cytoplasm.88

The CvME pathway was also adopted by DNA aptamer R13. Its cellular internalization occurs not only through CME, but also through CvME. Poor uptake of R13 was observed upon cellular treatment with genistein, suggesting the involvement of multiple endocytic pathways for its internalization.75

C. Macropinocytosis

Macropinocytosis is a receptor-independent endocytic pathway that nonselectively internalizes extracellular fluid and soluble macromolecules into intracellular vesicles called macropinosomes.89 The macropinocytic uptake is driven by actin dynamics that induces the protrusions of the plasma membrane into cup-shaped membrane ruffles. The ruffles then fold back and fuse with the plasma membrane, while enclosing portions of extracellular fluids and solutes. Unlike CME and CvME, macropinocytosis is not controlled by its endocytic cargo.90 Macropinocytosis allows cells to ‘drink’ any extracellular soluble macromolecules due to its nonselective property and can accommodate macromolecules that are excluded from other endocytic vesicles, thanks to the large size of macropinosomes.91 In general, the intracellular fate of macropinosomes can be routed into two: i) to be recycled to the cell surface to release back its cargo, ii) or trafficked to the lysosome to break down its cargo (Fig. 4C).91–93

One of the most well characterized examples for the macropinocytic pathway is the uptake of aptamer AS1411. AS1411 is a G-quadruplex DNA aptamer which binds to target protein nucleolin that is expressed on the cell surface, with high affinity and specificity.95,96 Nucleolin is known to be abundantly expressed in the nucleoli of proliferating cells, as well as in the cytoplasm, inner plasma membrane and cell surface under certain conditions.97 Cell surface nucleolin has been implicated in cancer progression and expressed at high levels in most cancer cells.98 Once bound to cell surface nucleolin, AS1411 is internalized into cancer cells.95 When DU145 prostate cancer cells were pre-incubated with CME inhibitors dynasore and cytochalasin D, only slight changes in AS1411 cellular uptake were observed.67 On the other hand, its cellular uptake was greatly reduced upon pre-treatment with amiloride (a macropinocytosis inhibitor).67 The data then suggested that AS1411 was mainly internalized through the macropinocytosis pathway, as further confirmed by the colocalization study of AS1411 with dextran (a macropinocytic marker).

In another study, the uptake of AS1411 in PC3 prostate cancer cells was significantly reduced in cells that were pre-incubated with either amiloride (a macropinocytosis inhibitor) or chlorpromazine (a CME inhibitor).94 In contrast, cells pre-treated with a CvME inhibitor, genistein, did not affect the uptake of the AS1411 aptamer.94 This interesting observation suggested that macropinocytosis is not a sole pathway used in AS411 internalization as CME also plays a role in its cellular uptake. Thus, the uptake mechanism of AS1411 may vary based on the type of cancer cell used.11

The anticancer properties of aptamers may depend on the mechanism of their cellular uptake. The route that was adopted by the aptamers in their internalization process could affect their biological activity in cells. In cells, the aptamer–target complex incorporated into the CME and/or CvME endocytic cargo may go through endosomal trapping and/or lysosomal degradation, while the cargo in macropinocytic vesicles is believed to bypass degradative processing due to the ability of AS1411 to escape from macropinosomes.99,100 Hyperstimulation of macropinocytosis may also induce cancer cell death upon efficient uptake of AS1411.67,101,102

The cell-internalizing aptamers and their known uptake mechanisms discussed here are listed in Table 2.

Cell-internalizing aptamers and their known uptake mechanisms described in the main text. CME: clathrin-mediated endocytosis, CvME: caveolae-mediated endocytosis
Aptamer nameUptake mechanismRef.
Apt-2 and Apt-10CME 63
R13CME and CvME 75
A10CME 78
C10.36CME 80
sgc8CvME 88
AS1411CME, macropinocytosis 67, 94

IV. Conclusions and outlook

Many studies have demonstrated the high potential of nucleic acid aptamers for their use in diagnostic and therapeutic fields. They exhibit target specificity, in addition to being relatively small in size, non-immunogenic and easily stabilized through chemical modifications. Since its first discovery about three decades ago, aptamer technology has shown great potential in producing bioactive and biostable pharmacological agents, as evidenced by the number of aptamers that successfully entered clinical trials and by the rapidly growing aptamer industry.5,103,104

Internalization of cell-binding aptamers is an important aspect for in vivo practical applications in targeted delivery or as drug candidates. In this review, we have discussed methods to generate cell-internalizing aptamers, along with their challenges and improvement strategies. Importantly, we have highlighted the cellular fate and uptake mechanism of the aptamers to better understand their mechanism of action.

Although significant achievement has been accomplished to date, there is still only one therapeutic aptamer that has been approved by the US FDA for clinical use (aptamer pegaptanib sodium/Macugen for patients with age-related macular degeneration).105 For practical use, high affinity and stability of aptamers would be required. With the vast growing improvement tactics to tackle major issues of aptamer utilization in vivo (such as susceptibility to degradation by serum or nuclease, and inefficient cellular internalization), it is anticipated that aptamer technology, especially cell-internalizing aptamers, will progressively find use as a new class of diagnostic and therapeutic practical agents.

Author contributions

S. H. A., M. K., and I. H. conceived and planned this review. S. H. A. took the lead in writing the manuscript and prepared the first draft including all the figures. All authors provided critical feedback and helped finalize the manuscript.

Conflicts of interest

There are no conflicts to declare.

Supplementary Material

Acknowledgments

This work was supported by the Institute of Bioengineering and Bioimaging (Biomedical Research Council, Agency for Science, Technology and Research, Singapore) and the National Research Foundation (NRF) Singapore, under its Competitive Research Programme (NRF-CRP17-2017-07).

Biographies

Biography

An external file that holds a picture, illustration, etc.
Object name is d1md00199j-p1.jpg
Samira Husen Alamudi

Samira Husen Alamudi received her PhD degree in Chemistry from the National University of Singapore under the supervision of Prof. Young-Tae Chang in 2015. She is currently a research fellow at IBB A*STAR Singapore with Dr Ichiro Hirao's group. Her research interests include rational design of background-free fluorescent probes for biomolecular and cellular imaging. She is also interested in the generation and application of unnatural base-containing nucleic acid aptamers for therapeutics and diagnostics.

Biography

Michiko Kimoto received her PhD degree from the Department of Biophysics and Biochemistry at the University of Tokyo, Japan, for her work on applications of unnatural base pair systems to RNA aptamers. She joined Hirao's team at RIKEN in Japan in 2002, and moved to A*STAR in Singapore, with Hirao's team, in 2015. Her research covers unnatural-base DNA aptamer generation and further applications of unnatural base pair systems toward diagnostics and therapeutics.

Biography

Ichiro Hirao received his PhD degree in 1983 from Tokyo Institute of Technology, Japan. In 1984, he worked at the University of Tokyo (Prof. Miura K.), as a research associate. In 1992, he became an associate professor at Tokyo University of Pharmacy and Life Sciences. In 1995, he moved to Andrew Ellington's laboratory at Indiana University. In 1997, he joined the JST ERATO project of Yokoyama S., as a group leader. He continued his xenobiology work as a professor at the University of Tokyo and a team leader at RIKEN. In 2015, he moved to A*STAR in Singapore, as a team leader.

References


Articles from RSC Medicinal Chemistry are provided here courtesy of Royal Society of Chemistry

Citations & impact 


Impact metrics

Jump to Citations

Citations of article over time

Alternative metrics

Altmetric item for https://www.altmetric.com/details/143368688
Altmetric
Discover the attention surrounding your research
https://www.altmetric.com/details/143368688

Article citations

Similar Articles 


To arrive at the top five similar articles we use a word-weighted algorithm to compare words from the Title and Abstract of each citation.


Funding 


Funders who supported this work.

Agency for Science, Technology and Research

    Biomedical Research Council

      National Research Foundation Singapore (1)