Minireviews Open Access
Copyright ©The Author(s) 2019. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastroenterol. Jul 14, 2019; 25(26): 3359-3369
Published online Jul 14, 2019. doi: 10.3748/wjg.v25.i26.3359
Screening of aptamers and their potential application in targeted diagnosis and therapy of liver cancer
Guo-Qing Zhang, Li-Ping Zhong, Nuo Yang, Yong-Xiang Zhao, National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
ORCID number: Guo-Qing Zhang (0000-0003-3865-0378); Li-Ping Zhong (0000-0003-1532-4107); Nuo Yang (0000-0003-0606-7313); Yong-Xiang Zhao (0000-0003-1219-0908).
Author contributions: Zhang GQ, Zhong LP, Yang N, and Zhao YX wrote the paper.
Conflict-of-interest statement: All the authors have no conflict of interest related to the manuscript.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Corresponding author: Yong-Xiang Zhao, MD, PhD, Full Professor, National Center for International Research of Biological Targeting Diagnosis and Therapy, Guangxi Key Laboratory of Biological Targeting Diagnosis and Therapy Research, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, 22 Shuangyong Road, Nanning 530021, Guangxi Zhuang Autonomous Region, China. yongxiang_zhao@126.com
Telephone: +86-15177120015 Fax: +86-771-5317061
Received: May 18, 2019
Peer-review started: May 18, 2019
First decision: May 30, 2019
Revised: June 2, 2019
Accepted: June 8, 2019
Article in press: June 8, 2019
Published online: July 14, 2019

Abstract

Aptamers are a class of single oligonucleotide molecules (DNA or RNA) that are screened from random DNA or RNA oligonucleotide chain libraries by the systemic evolution of ligands by exponential enrichment technology. The selected aptamers are capable of specifically binding to different targeting molecules, which is achieved by the three-dimensional structure of aptamers. Aptamers are similar in function to monoclonal antibodies, and therefore, they are also referred to as "chemical antibodies". Due to their high affinity and specificity and low immunogenicity, aptamers are topics of intense interest in today's biological targeting research especially in tumor research. They not only have high potential for clinical advances in tumor targeting detection but also are highly promising as targeted tumor drug carriers for use in tumor therapy. Various experimental studies have shown that aptamer-based diagnostic and therapeutic methods for liver cancer have great potential for application. This paper summarizes the structure, characteristics, and screening methods of aptamers and reviews the recent research progress on nucleic acid aptamers in the targeted diagnosis and treatment of liver cancer.

Key Words: Aptamer, Systemic evolution of ligands by exponential enrichment, Liver cancer, Outlook

Core tip: For liver cancer, conventional diagnostic methods have low sensitivity, therapeutic drugs have great side effects, and tumor monitoring is difficult, which limit the diagnosis and treatment of liver cancer. However, the diagnosis and treatment of liver cancer based on aptamers have great application prospects. This review describes the screening of aptamers and reviews the recent advances on aptamers in the targeted diagnosis and treatment of liver cancer.



INTRODUCTION

Tumors are one of the leading causes of human death worldwide. With the development of society, the population has increased, and the risk of tumors has also greatly increased. Tumor diagnosis and treatment have become an issue of intense concern. Liver cancer is a common malignant tumor, and its diagnosis and treatment are the research focus of many scholars.

Aptamer refers to a type of single oligonucleotide molecule (DNA or RNA) that is screened and enriched from a library of random single-stranded oligonucleotide sequences constructed in vitro, and aptamers can specifically bind to target molecules, such as small molecules, proteins, and even whole cells[1-3]. In 1990, Ellington et al[4] and Tuerk et al[5] screened for a random oligonucleotide that binds to T4 DNA polymerase with high affinity and specificity and confirmed the ability of this oligonucleotide sequence to bind to the target. The identified oligonucleotide was referred to as an aptamer, and the screening method was named systematic evolution of ligands by exponential enrichment (SELEX). The first aptamer drug, pegaptanib sodium (trade name: Macugen), was approved by the United States Food and Drug Administration in 2004 and is used to treat age-related macular degeneration[6]. Now, scientists have acquired a variety of aptamers screened using the SELEX technology, which have a special three-dimensional structure that can bind different target molecules[7,8]. The more detailed explanation for aptamers is given below.

APTAMERS
Structure and function of aptamers

An aptamer is a specific three-dimensional structure formed by the folding of a single-stranded nucleic acid molecule. The binding of an aptamer to the target molecule is achieved by its own special three-dimensional structure, with features such as hairpins, internal loops, bulges, pseudoknots, and G-tetramers[9]. The factors by which an aptamer binds to the target molecule are the complementary effects of the geometric shape, the interactions of aromatic ring overlap, the electrostatic interactions of the charged groups, the bases of the nucleic acid aptamer, the van der Waals forces, and the hydrogen bonds. Targets that can be bound include small molecules, proteins, cells, and even entire tissues. Proteins are the most numerous targets[10,11].

DNA and RNA aptamers

Both DNA and RNA aptamers can produce similar specificities and affinities, but each has its own distinct advantages. The primary difference between DNA and RNA lies in the sugar. The GH bond of deoxyribose in DNA has low reactivity and a stable structure, making it better able to resist the action of enzymes. The ribose in RNA is more reactive and less stable than deoxyribose, and double-stranded regions of RNA can stimulate the immune system via the Toll-like receptor. An RNA aptamer obtained from an RNA oligonucleotide chain library using the SELEX method needs to be modified for improved stability[12].

In the early stages, many researchers were more likely to screen for RNA aptamers because they have more diverse stereoscopic three-dimensional structures than DNA and higher affinity for targets[13]. However, in-depth studies of aptamers have presented increasing evidence that DNA aptamers are superior to RNA aptamers in application. For example, DNA aptamers have better structural stability and are easy to screen. Therefore, an increasing number of researchers are now more inclined to explore DNA aptamers, and in 2013, it was reported that more than 85% of aptamers were obtained from DNA oligonucleotide libraries[14].

SELEX METHOD FOR SCREENING APTAMERS
Basic principles of SELEX technology

The SELEX technology combines multiple fields, such as molecular biology, bioinformatics, and materials science, to develop aptamers for a variety of targets[15]. A large number (approximately 1014-15) of random DNA or RNA oligonucleotide chains are first synthesized, and then multiple screening and enrichment cycles are performed to select oligonucleotide chains from this library that can bind target molecules with high specificity and high affinity. New base sequences will appear during the screening process, and as the cycle progresses, the specificity and affinity with which the selected oligonucleotide strand binds the target molecule will gradually increase. The final oligonucleotide chain has the highest specificity and affinity for the target molecule and is referred to as the aptamer of the specific target molecule[16,17].

Establishment of a random oligonucleotide chain library

As a raw material, the oligonucleotide chain for aptamer screening is fixed at both ends and random in the middle. The random sequences can specifically bind to the target molecule, and the fixed sequences can be used for amplification and screening. Screening of DNA or RNA aptamers starts with synthesis of random DNA or RNA oligonucleotide chain libraries.

The first step in building a library is to design fixed sequences at both ends of the oligonucleotide chain, which are usually 20-30 nucleotides in length. The fixed sequences generally contain primer binding sites and restriction endonuclease binding sites required for PCR amplification, while binding sites containing RNA polymerase are needed when preparing RNA aptamers. The transcribed RNA sequences must also be able to bind to reverse transcriptase[18]. In designing the intermediate sequences, attention should be paid to length. Short chains are easier to design and fabricate and less expensive, but if the sequence is too short, it cannot form secondary structures that specifically and stably bind to target molecules, or there are not enough secondary structures in the library for screening. As the length of the intermediate sequence increases, the capacity of the library increases, but increasing the length of the intermediate sequence beyond a certain threshold will not significantly increase the number of unique stereo structures available for screening. Generally, the length of the intermediate sequence is 30-50 nucleotides, which can theoretically form approximately 1014-1024 random oligonucleotide chains. The stereo structure formed by this number of nucleotides is sufficient to bind a target molecule[19]. After the sequence design is completed, a large number of DNA oligonucleotide chains with random sequences in the middle can be synthesized with a DNA synthesizer, and a DNA oligonucleotide chain library can be established.

Selection and enrichment of aptamers

After establishment of a DNA oligonucleotide chain library, it is necessary to screen for and enrich oligonucleotide chains that can bind to target molecules with high specificity and affinity. Mature screening techniques include magnetic bead separation[20], high-speed centrifugation, affinity chromatography[21], capillary electrophoresis[22], and nitrocellulose membrane filtration[23]. For example, studies have shown that a target aptamer can be obtained in only four cycles of capillary electrophoresis SELEX[24].

On the basis of traditional screening methods, many screening methods have been developed, such as counter SELEX[25], subtractive SELEX[26], photo SELEX[27], and negative SELEX[28], which can improve the selectivity of aptamers. Blended SELEX[29], complex targeting SELEX[30], expression cassette SELEX[31], and toggle SELEX[32] can improve the universality of aptamers. In addition, automated SELEX[33], non-SELEX[34], and FluMag SELEX[35] can shorten the screening cycle. Methods for combining different pools, such as tailored SELEX[36] and genomic SELEX[37], can increase the likelihood of screening for aptamers of interest. At present, cell-based screening (cell-SELEX) is widely used[38] (Figure 1).

Figure 1
Figure 1 Schematic diagram of cell systemic evolution of ligands by exponential enrichment strategy. The target cells are used to screen and enrich the selection pools, and clonal sequencing is performed to obtain the required aptamers. SELEX: Systemic evolution of ligands by exponential enrichment.

To shorten the screening process, the following screening methods have been developed. Multiple GO-SELEX employs graphene oxide (GO) for screening. This method is simple, fast, and high throughput, and thus reduces the screening cost and shortens the screening time[39]. Single-walled carbon nanotube-assisted cell-SELEX (SWCNT-assisted cell-SELEX) requires only six rounds of screening compared with the 15 rounds necessary in standard cell-SELEX and has a high screening efficiency[40]. On-chip cell-SELEX, which incorporates microfluidic chips into the cell-SELEX process, requires only five rounds of screening, greatly shortening the screening process[41]. Sequence-constructive SELEX modifies the sequence of aptamers to improve the affinity and specificity for the targets[42]. Furthermore, Hi-Fi SELEX is a protocol for rapid and efficient DNA aptamer selection[43].

The general procedures for screening and enrichment of DNA aptamers are as follows: in the first round of screening, target molecules are used to screen DNA oligonucleotide chain libraries and the oligonucleotide chain DNA is amplified via PCR; in the second round of screening, the amplified products are rescreened and the DNA is amplified via PCR; after 8-16 repeated rounds of screening, flow cytometry and clonal sequencing are performed to obtain the required high specificity and high affinity aptamers[15]. However, depending on the SELEX method used, the number of screening and enrichment rounds will vary. In most cases, too many rounds will waste consumable reagents without significantly improving the screening rate. However, too few rounds may not identify the best aptamers or impurities. The appropriate number of screening rounds is generally the number at which the affinity does not continue to increase. Because of the low fidelity of the polymerase used in SELEX, each screening enrichment cycle may produce variants, and thus, the screening enrichment cycles actually increase the capacity of oligonucleotide chain libraries. In addition to the effect of screening enrichment, the number of cycles needed for screening enrichment is one of the criteria for evaluating the advantages and disadvantages of screening enrichment methods. Many of the methods mentioned above are optimized in terms of the number of cycles.

Base sequencing, characterization, and molecular structure modification of aptamers

Screening of enriched aptamers requires base sequencing to obtain well-defined base sequences. Then, structural analysis and characterization of these screened aptamers need to be performed, to assess factors such as specificity, affinity, and stability.

The chemical synthesis of aptamers is more stable than antibodies. However, despite their high stability and tolerance to temperature, pH range, and some organic solvents, aptamers can be readily degraded or removed by the kidneys in vivo. Because of the limited interaction between natural base pairs and drugs, in vivo use requires prolonging the survival time of aptamers under physiological conditions, that is, optimizing the structure of aptamers. Conventional optimization methods include the following: (1) Substituting a natural nucleotide for a nucleotide modified with a group to avoid degradation of the nucleic acid[44]; (2) Chemically synthesizing a mirror image sequence of the selected aptamers with high plasma and serum stability to enhance the stability of aptamers to make them have significant therapeutic potential[45]; (3) Modifying aptamers with locked nucleic acids to enhance their ribozyme resistance. Methylene-linked 2'-oxygen and 4'-carbon in carbohydrate residues have strong ability to hybridize with nucleic acid molecules and are not easily degraded by enzymes[46]; and (4) Covalently attaching PEG or cholesterol to aptamers, prolonging their residence time in the human body and reducing renal filtration rate[47,48]. The half-life of survival of aptamers in human physiological environment can be extended from a few minutes to several hours by one or more of the above methods, and aptamers with modifiable properties are superior to antibodies or polypeptides in the diagnosis and treatment of tumor diseases.

CHARACTERISTICS OF APTAMERS AND COMPARISON WITH MONOCLONAL ANTIBODIES
Characteristics of aptamers

Aptamers have many advantages, such as high affinity and specificity, a wide range of action, small size, good stability, low cost, and easy synthesis and screening compared with traditional immunological and chemical recognition molecules, and aptamers have shown broad application prospects in the diagnosis and treatment of diseases[49].

Comparison of aptamers and monoclonal antibodies

There are some similarities between aptamers and monoclonal antibodies (mAbs). In terms of function, both aptamers and mAbs can bind target molecules with high specificity and affinity. In terms of application, both aptamers and mAbs can be used in medical research and in clinical diagnosis and treatment; therefore, aptamers are known as "chemical antibodies". Compared with mAbs, aptamers do not exhibit immunogenicity or antigenicity and do not cause a related immune response or immune rejection in the human body. This property makes aptamers reusable for the same patient in the diagnosis and treatment of clinical diseases[50].

APPLICATION OF APTAMERS IN THE TARGETED DIAGNOSIS OF LIVER CANCER

The early stage of tumorigenesis may present no symptoms, leading to tumors in the middle and late stages before symptoms are identified, which hinders the treatment of tumors. However, tumor markers already exist in the early stage of tumorigenesis, which is of great significance for the early diagnosis and treatment of tumors. The main difficulty in the early diagnosis of tumors is the identification of the characteristics of tumors and tumor cells. Tumor markers are important molecules in the diagnosis and treatment of tumors. They can target whole tumors or tumor cells. Fluorescence imaging of tumors and cells can diagnose tumors and monitor their treatment. Due to the high specificity and affinity of aptamers, a goal currently attracting intense interest is to screen for aptamers as tumor markers and apply them to the targeted diagnosis of tumors. Aptamers have become potential probe molecules for targeted diagnosis of liver cancer, and their application in vitro and in vivo of liver cancer can improve detection specificity and sensitivity[51] (Figure 2).

Figure 2
Figure 2 Schematic diagram of aptamer targeting to liver cancer. The aptamers bind to the surface of the liver cancer cells with high specificity and high affinity after incubation with the liver cancer cells. After the aptamers are injected into the mouse model through the tail vein, they specifically bind to the liver cancer tissue site.
Aptamers for in vitro detection of liver cancer

In recent years, with the in-depth study of aptamers, increasing numbers of liver cancer detection methods based on aptamers have been developed to provide different in vitro detection methods for the diagnosis of liver cancer (Table 1).

Table 1 Current literature summary of aptamers for in vitro detection of liver cancer.
First authorYearAptamerTargetSystem for detectionMethod for detection
Hu et al[51]2017TLS11aHepG2 cellsBio-TLS11a and SA-FSNPsFluorescence Microscopy
Wang et al[52]2016sLex-APCTCsCTC-BioT ChipFluorescence Microscopy
Joo et al[53]2017AFB1-AptAFB1FAM modified AFB1-Apt and GOFluorescence Microscopy
Kaur et al[54]2019SL2-BHepG2 cellsQDs labeled SL2-BFluorescence Microscopy
Zhou et al[55]2019Slow-off aptamer (TD10); fast-off aptamerDKK1 protein5′ amino C6 modified TD10-based ELISA; 5′ FAM labeled fast-off aptamerELISA (TD10); dot blot and flow cytometry (fast-off aptamer)

The aptamer-nanoparticle system (Apt-NP) formed by the conjugation of liver cancer-related aptamers and nanoparticles, can be used for the diagnosis of liver cancer. Recently, Hu et al[52] labeled HepG2 cells with biotin-conjugated TLS11a aptamer (Bio-TLS11a)., and then co-incubated with streptavidin-conjugated fluorescent silica nanoparticles (SA-FSNPs). HepG2 cells were detected in vitro by the interaction between biotin and streptavidin. Hu et al[52] pointed out that these SA-FSNPs can specifically and sensitively detect aptamer-labeled HepG2 cells and show no obvious toxic effects, showing the good application prospect of this method for liver cancer cells and other types of cancer cells.

With the deeper research on nano-molecules, more and more nano-molecules have been discovered and used together with aptamers for targeted detection of liver cancer cells. Wang et al[53] established a new method for detecting circulating tumor cells (CTCs) in hepatocellular carcinoma (HCC). They combined the aptamer of the carbohydrate sialic acid Lewis X with a hydroxyapatite/chitosan (HA/CTS) nanofilm to form the CTC-BioTChip platform, which effectively captures and identifies HCC CTCs. A small amount of aflatoxin B1 can cause liver cancer to occur. Joo et al[54] developed a rapid method for the determination of aflatoxin B1 (AFB1) based on GO and fluorescein imide modified AFB1-specific aptamer. When the aptamer binds to AFB1, the conformation of the aptamer changes and interacts with GO such that the fluorescence intensity is reduced or even quenched.

In the past few years, the potential role of quantum dots in fluorescence imaging has gradually attracted the attention and research of more scholars. QD-labeled aptamers are also a new strategy for detecting liver cancer cells. It has been found that the SL2-B aptamer labeled with QDs can specifically recognize HepG2 cells and be taken up by HepG2 cells, and the results can be detected by fluorescence microscopy. Moreover, as the incubation time of HepG2 cells with the conjugate increased, it also showed potential antiproliferative activity against HepG2 cells[55]. This technique of QDs labeling aptamers provides a new strategy for in vitro detection and research of liver cancer.

Dickkopf-1 (DKK1) has potential application value for the early diagnosis of HCC. Zhou et al[56] obtained two kinds of aptamers that recognize DKK1, one is slow-off aptamer, and the experimental results show that it can be used for aptamer-based ELISA assay, while another fast-off aptamer is more suitable for flow cytometry and spot-blot. In addition, the combination of molecular beacons[57], which have stem-ring structures and contain fluorescent and quenching agents, and fluorescent dyes[58] with aptamers targeting tumors has also been proven to be useful for detecting or imaging tumors. This provides more potential methods for in vitro detection of liver cancer.

Aptamers for in vivo imaging of liver cancer

In vivo imaging is still a common method for the clinical diagnosis of tumors. Techniques include magnetic resonance imaging (MRI), ultrasound molecular imaging, computed tomography (CT), and fluorescence molecular tomography, which have been verified in small rodents[59]. Nowadays, due to the extremely high incidence of liver cancer and the increasingly intensive research on aptamer, the application of various aptamer-based contrast agents, probes, and fluorescent groups to in vivo oncology imaging has become the research topic of numerous scholars (Table 2).

Table 2 Current literature summary of aptamers for in vivo imaging of liver cancer.
First authorYearAptamerType of tumorProbe for detectionMethod for detection
Yan et al[59]2018CD105-AptTiny HCCConjugated CD105-Apt to paramagnetic and fluorophore on the G5 dendrimerMRI
Zhong et al[60]2019mENDHCCmEND-Fe O @CMCSMRI
Zhao et al[61]2018AP613-1HCC expressing GPC3Apt-USPIOMRI
Babaei et al[69]2017EpCAM-AptHCCEpCAM-PEG-Au@Si-5-FU NPsCT

MRI is a non-invasive tumor diagnosis technique. To improve the diagnostic efficacy of MRI, Yan et al[60] obtained an aptamer capable of binding to endoglin (CD105) with high specificity by SELEX. This aptamer is conjugated to paramagnetic and fluorophore on the G5 dendrimer to form a targeted nanoprobe that can be used to detect tiny HCCs. Zhong et al[61] developed a nanoprobe targeting CD105 for MRI by the use of aptamer-modified magnetic carboxymethyl chitosan, which can improve the early diagnosis of HCC. In addition, Zhao et al[62] prepared a targeted MRI molecular probe based on ultrasmall superparamagnetic iron oxide (USPIO) mediated by phosphatidylinositol-3 (GPC3) aptamer. The probe was verified in xenograft mice, and the experimental results confirmed the targeted imaging effect of the probe on HCC expressing GPC3.

Various studies have shown that aptamers have great potential advantages in the targeted diagnosis of liver cancer, but this application is still in its infancy. For some malignant tumors with a low expression rate of tumor markers, further exploration is still needed[63].

APPLICATION OF APTAMERS IN TARGETED THERAPY OF LIVER CANCER

In addition to acting as therapeutic agents, aptamers can also be used as targeting agents to conjugate with antitumor drugs, oligonucleotides, or nanocarriers for targeted tumor therapy. A drug delivery system based on aptamers has many advantages. It can not only significantly improve the pharmacokinetic properties of drugs in vivo, but also can reduce toxic and side effects to some extent certain due targeting properties. Because aptamers are small (nanosized) molecules, the whole drug delivery system can effectively penetrate all levels of blood vessels into the tumor site[64].

The traditional methods of liver cancer therapy have poor targeting, inefficient drug delivery, poor efficacy, and severe side effects, which together present great challenges in the therapy of liver cancer. The targeted therapy of liver cancer is now the focus of scientists' research. Currently, aptamers have been widely studied and applied in various fields because of their characteristics. Modified aptamers have been applied to the treatment of liver cancer, improving the efficiency of treatment and greatly reducing the side effects of drug treatment[65]. Aptamers can be used as therapeutic agents to intervene in the treatment of liver cancer. They can also be conjugated with antineoplastic drugs, oligonucleotides, or nanocarriers for the treatment of tumors, such as chemotherapy, gene therapy, immune therapy, radiotherapy, and phototherapy[66] (Table 3).

Table 3 Current literature summary on the application of aptamers in targeted therapy for liver cancer.
First authorYearAptamerTargetSystem for therapyFunction
Hu et al[66]2018TLS11aH22 cellsTLS11a/CD3Immunity therapy
Xiao et al[67]2017EpDT3EpCAM-positive cells HepG2EpDT3-PEG-Ad5-PTEN (EPAP)Gene therapy
Babaei et al[69]2017EpCAM-AptEpCAM-positive cells HepG2EpCAM-PEG-Au@Si-5-FU NPsChemotherapy
Lan et al[70]2019TLS11aHepG2 cellsApt-BMSF@PtPhototherapy
Liu et al[72]2016ST21SK-Hep-1 cellsFasudilST21-H3R5-PEG miR195Chemotherapy and gene therapy
Cheng et al[73]2019TLS1cMEAR cellsBioTL Cab/lipoChemotherapy

Hu et al[67] constructed a bispecific system using the hepatoma cells-specific aptamer TLS11a and anti-CD3, which mediates T cell activation, targets cytotoxic T cells to the liver cancer site, and thus enhances the anti-tumor effect. The experimental results show that TLS11a/CD3 can connect hepatoma cells and T cells, avoiding the occurrence of immune escape and mediating the lysis of hepatoma cells and the release of cytokines, and the released cytokines can continue to enhance anti-tumor effects. This aptamer-based bispecific system provides a new technology platform for targeted immunotherapy of liver cancer.

The recombinant adenovirus carrying the PTEN gene (Ad5-PTEN) is an effective anti-tumor agent, but its application is greatly limited due to its poor stability and toxic effects to normal tissues. To overcome the application limitations of Ad5-PTEN, Xiao et al[68] formed a novel gene delivery system, EPAP, by linking EpDT3, an aptamer that specifically binds to epithelial cell adhesion molecule (EpCAM), to Ad-PTEN. The results show that EPAP is non-toxic to normal cells and tissues, can target and inhibit EpCAM-positive hepatoma cells, enhance anti-tumor activity, and improve the gene therapy efficiency of liver cancer.

Today, mesoporous silica nanoparticles (MSNs) are a potential therapeutic platform for cancer therapy. MSNs are a good nano-drug delivery platform because of its good stability, high load, high biocompatibility, adjustable pore size, and protection against inactivation or degradation of the incorporated drug[69]. Babaei et al[70] developed a PEGylated MSN system capable of targeting EpCAM and controlling the release of 5-fluorouracil. First, the authors loaded 5-fluorouracil into mesoporous silica, hybridized with gold nanoparticles to its outer surface, PEGylated, and then conjugated with EpCAM aptamer, and the resulting complex can be used for HCC treatment. This system has potential application prospects in the targeted therapy of early liver cancer. In addition, it can also be used for CT imaging.

Black Phosphorus Quantum Dots (BPQDs) have a unique photocatalytic activity and have potential applications in photodynamic therapy, but the application of BPQD is limited due to the instability of the tumor microenvironment. To solve this problem, Lan et al[71] constructed a nanostructure based on the TLS11a aptamer. Lan et al[71] synthesized BPQDs/Pt hybrid mesoporous silica (BMSF@Pt), and then conjugated the TLS11a aptamer to BMSF@Pt to form Apt-MBMSF@Pt nanostructure. This structure can specifically target to HCC cells, killing HCC cells by self-compensation of oxygen, enhancing photodynamic efficacy, and providing a new method for targeted therapy of HCC.

MiRNA-195 (miR195) is a potent gene drug that inhibits vascular endothelial growth factor (VEGF), and some studies have demonstrated the inhibitory effect of miR195 in HCC[72]. Liu et al[73] conjugated histidine-modified disulfide cross-linked stearyl polyarginine peptide (H3R5) to cell penetrating peptide-modified aptamer, ST21, which is capable of specifically binding to HCC cells. Then, under the action of PEG, miR195 and Fasudil were loaded to the conjugation. Finally, a nano-system, FasudilST21-H3R5-PEGmiR195, based on ST21-H3R5-PEG was constructed. The results showed that FasudilST21-H3R5-PEGmiR195 has a strong ROCK2 blocking effect and VEGF silencing effect, showing a higher anti-tumor effect, and has great application prospects for targeted therapy of HCC.

Liposomes are complex structures composed of cholesterol and phospholipids that can contain hydrophilic and lipophilic drugs and reduce the cytotoxicity of drugs. Liposomes are very effective drug nanocarriers. Cabazitaxel is a broad-spectrum antineoplastic drug, but its clinical adverse reactions hinder its use. Cheng et al[74] modified capsaicin liposomes with a TLS1c aptamer targeting MEAR cells and demonstrated the highly specific targeting ability and strong antitumor effect of this complex on MEAR cells. This drug delivery system is also an effective potential therapeutic strategy. Nanoparticles, polymer micelles, and biomimetic nanocarriers can also be used as drug carriers to construct drug delivery systems, providing potential for the further development of targeted therapy for liver cancer[75].

CONCLUSION

In conclusion, the diagnosis and treatment of liver cancer developed on the basis of aptamer solve the problems of low diagnostic sensitivity, large side effects of anticancer drugs, and difficulty in tumor monitoring. Various experimental results have confirmed the great potential of these strategies in the clinical application to liver cancer, and provide good scientific evidence for the clinical application of aptamers in liver cancer.

As “chemical antibodies”, aptamers have been considered an important research direction in the targeted diagnosis and treatment of tumors. With the continuous development of aptamers with different targeting sites, the application of aptamers with different targeting sites has become increasingly extensive. Aptamers have been modified and applied to various aspects of targeting research for liver cancer. With the development of different detection methods and treatment methods based on aptamers, aptamers will continue to develop in the clinical application of liver cancer, and finally realize early diagnosis and treatment of liver cancer.

Footnotes

Manuscript source: Unsolicited manuscript

Specialty type: Gastroenterology and hepatology

Country of origin: China

Peer-review report classification

Grade A (Excellent): A

Grade B (Very good): 0

Grade C (Good): C

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Mohamed SY, Tanabe S S-Editor: Yan JP L-Editor: Wang TQ E-Editor: Zhang YL

References
1.  Gold L, Polisky B, Uhlenbeck O, Yarus M. Diversity of oligonucleotide functions. Annu Rev Biochem. 1995;64:763-797.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 633]  [Cited by in F6Publishing: 686]  [Article Influence: 23.7]  [Reference Citation Analysis (0)]
2.  Pestourie C, Tavitian B, Duconge F. Aptamers against extracellular targets for in vivo applications. Biochimie. 2005;87:921-930.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 131]  [Cited by in F6Publishing: 114]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
3.  Cibiel A, Pestourie C, Ducongé F. In vivo uses of aptamers selected against cell surface biomarkers for therapy and molecular imaging. Biochimie. 2012;94:1595-1606.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 33]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
4.  Ellington AD, Szostak JW. In vitro selection of RNA molecules that bind specific ligands. Nature. 1990;346:818-822.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7090]  [Cited by in F6Publishing: 6659]  [Article Influence: 195.9]  [Reference Citation Analysis (0)]
5.  Tuerk C, Gold L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science. 1990;249:505-510.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7422]  [Cited by in F6Publishing: 7021]  [Article Influence: 206.5]  [Reference Citation Analysis (0)]
6.  Ng EW, Shima DT, Calias P, Cunningham ET, Guyer DR, Adamis AP. Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease. Nat Rev Drug Discov. 2006;5:123-132.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 994]  [Cited by in F6Publishing: 996]  [Article Influence: 55.3]  [Reference Citation Analysis (0)]
7.  Ahmadyousefi Y, Malih S, Mirzaee Y, Saidijam M. Nucleic acid aptamers in diagnosis of colorectal cancer. Biochimie. 2019;156:1-11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 12]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
8.  Zhou G, Latchoumanin O, Hebbard L, Duan W, Liddle C, George J, Qiao L. Aptamers as targeting ligands and therapeutic molecules for overcoming drug resistance in cancers. Adv Drug Deliv Rev. 2018;134:107-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 44]  [Article Influence: 7.3]  [Reference Citation Analysis (0)]
9.  Perez-Gonzalez C, Lafontaine DA, Penedo JC. Fluorescence-Based Strategies to Investigate the Structure and Dynamics of Aptamer-Ligand Complexes. Front Chem. 2016;4:33.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 44]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
10.  Cai S, Yan J, Xiong H, Liu Y, Peng D, Liu Z. Investigations on the interface of nucleic acid aptamers and binding targets. Analyst. 2018;143:5317-5338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 141]  [Article Influence: 28.2]  [Reference Citation Analysis (0)]
11.  Takahashi M. Aptamers targeting cell surface proteins. Biochimie. 2018;145:63-72.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 35]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
12.  Zhu B, Hernandez A, Tan M, Wollenhaupt J, Tabor S, Richardson CC. Synthesis of 2'-Fluoro RNA by Syn5 RNA polymerase. Nucleic Acids Res. 2015;43:e94.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 25]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
13.  Zhang Y, Lai BS, Juhas M. Recent Advances in Aptamer Discovery and Applications. Molecules. 2019;24:pii: E941.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 261]  [Cited by in F6Publishing: 317]  [Article Influence: 63.4]  [Reference Citation Analysis (0)]
14.  Wang T, Chen C, Larcher LM, Barrero RA, Veedu RN. Three decades of nucleic acid aptamer technologies: Lessons learned, progress and opportunities on aptamer development. Biotechnol Adv. 2019;37:28-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 263]  [Article Influence: 43.8]  [Reference Citation Analysis (0)]
15.  Blind M, Blank M. Aptamer Selection Technology and Recent Advances. Mol Ther Nucleic Acids. 2015;4:e223.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 214]  [Article Influence: 26.8]  [Reference Citation Analysis (0)]
16.  Kinghorn AB, Fraser LA, Lang S, Shiu SCC, Tanner JA. Aptamer Bioinformatics. Int J Mol Sci. 2017;18:pii: E2516.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 108]  [Cited by in F6Publishing: 79]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
17.  Bayat P, Nosrati R, Alibolandi M, Rafatpanah H, Abnous K, Khedri M, Ramezani M. SELEX methods on the road to protein targeting with nucleic acid aptamers. Biochimie. 2018;154:132-155.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 113]  [Cited by in F6Publishing: 94]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
18.  Fitzwater T, Polisky B. A SELEX primer. Methods Enzymol. 1996;267:275-301.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 123]  [Cited by in F6Publishing: 131]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
19.  Cowperthwaite MC, Ellington AD. Bioinformatic analysis of the contribution of primer sequences to aptamer structures. J Mol Evol. 2008;67:95-102.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 67]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
20.  Wang L, Wang C, Li H. Selection of DNA aptamers and establishment of an effective aptasensor for highly sensitive detection of cefquinome residues in milk. Analyst. 2018;143:3202-3208.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 14]  [Cited by in F6Publishing: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
21.  Yang L, Ding P, Luo Y, Wang J, Lv H, Li W, Cao Y, Pei R. Exploration of Catalytic Nucleic Acids on Porphyrin Metalation and Peroxidase Activity by in Vitro Selection of Aptamers for N-Methyl Mesoporphyrin IX. ACS Comb Sci. 2019;21:83-89.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 17]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
22.  Luo Z, Zhou H, Jiang H, Ou H, Li X, Zhang L. Development of a fraction collection approach in capillary electrophoresis SELEX for aptamer selection. Analyst. 2015;140:2664-2670.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 37]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
23.  Song KM, Lee S, Ban C. Aptamers and their biological applications. Sensors (Basel). 2012;12:612-631.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 503]  [Cited by in F6Publishing: 450]  [Article Influence: 37.5]  [Reference Citation Analysis (0)]
24.  Zhuo Z, Yu Y, Wang M, Li J, Zhang Z, Liu J, Wu X, Lu A, Zhang G, Zhang B. Recent Advances in SELEX Technology and Aptamer Applications in Biomedicine. Int J Mol Sci. 2017;18:pii: E2142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 195]  [Cited by in F6Publishing: 229]  [Article Influence: 32.7]  [Reference Citation Analysis (0)]
25.  Jenison RD, Gill SC, Pardi A, Polisky B. High-resolution molecular discrimination by RNA. Science. 1994;263:1425-1429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 860]  [Cited by in F6Publishing: 796]  [Article Influence: 26.5]  [Reference Citation Analysis (0)]
26.  Li K, Xiu CL, Gao LM, Shi M, Zhai Y. [Subtractive SELEX using agar beads for screening DNA aptamers with specific affinity to HIV gp41 antigen]. Nan Fang Yi Ke Da Xue Xue Bao. 2016;36:1592-1598.  [PubMed]  [DOI]  [Cited in This Article: ]
27.  Golden MC, Collins BD, Willis MC, Koch TH. Diagnostic potential of PhotoSELEX-evolved ssDNA aptamers. J Biotechnol. 2000;81:167-178.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 110]  [Cited by in F6Publishing: 113]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
28.  Ellington AD, Szostak JW. Selection in vitro of single-stranded DNA molecules that fold into specific ligand-binding structures. Nature. 1992;355:850-852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 608]  [Cited by in F6Publishing: 559]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
29.  Charlton J, Kirschenheuter GP, Smith D. Highly potent irreversible inhibitors of neutrophil elastase generated by selection from a randomized DNA-valine phosphonate library. Biochemistry. 1997;36:3018-3026.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 44]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
30.  Morris KN, Jensen KB, Julin CM, Weil M, Gold L. High affinity ligands from in vitro selection: Complex targets. Proc Natl Acad Sci USA. 1998;95:2902-2907.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 268]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
31.  Martell RE, Nevins JR, Sullenger BA. Optimizing aptamer activity for gene therapy applications using expression cassette SELEX. Mol Ther. 2002;6:30-34.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 63]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
32.  White R, Rusconi C, Scardino E, Wolberg A, Lawson J, Hoffman M, Sullenger B. Generation of species cross-reactive aptamers using "toggle" SELEX. Mol Ther. 2001;4:567-573.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 206]  [Cited by in F6Publishing: 206]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
33.  Cox JC, Hayhurst A, Hesselberth J, Bayer TS, Georgiou G, Ellington AD. Automated selection of aptamers against protein targets translated in vitro: From gene to aptamer. Nucleic Acids Res. 2002;30:e108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 139]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
34.  Lisi S, Fiore E, Scarano S, Pascale E, Boehman Y, Ducongé F, Chierici S, Minunni M, Peyrin E, Ravelet C. Non-SELEX isolation of DNA aptamers for the homogeneous-phase fluorescence anisotropy sensing of tau Proteins. Anal Chim Acta. 2018;1038:173-181.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 37]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
35.  Stoltenburg R, Reinemann C, Strehlitz B. FluMag-SELEX as an advantageous method for DNA aptamer selection. Anal Bioanal Chem. 2005;383:83-91.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 256]  [Cited by in F6Publishing: 229]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
36.  Vater A, Jarosch F, Buchner K, Klussmann S. Short bioactive Spiegelmers to migraine-associated calcitonin gene-related peptide rapidly identified by a novel approach: Tailored-SELEX. Nucleic Acids Res. 2003;31:e130.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 107]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
37.  Shimada T, Ogasawara H, Ishihama A. Genomic SELEX Screening of Regulatory Targets of Escherichia coli Transcription Factors. Methods Mol Biol. 2018;1837:49-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 21]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
38.  Chen M, Yu Y, Jiang F, Zhou J, Li Y, Liang C, Dang L, Lu A, Zhang G. Development of Cell-SELEX Technology and Its Application in Cancer Diagnosis and Therapy. Int J Mol Sci. 2016;17:pii: E2079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 64]  [Article Influence: 8.0]  [Reference Citation Analysis (0)]
39.  Lee BH, Kim SH, Ko Y, Park JC, Ji S, Gu MB. The sensitive detection of ODAM by using sandwich-type biosensors with a cognate pair of aptamers for the early diagnosis of periodontal disease. Biosens Bioelectron. 2019;126:122-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 28]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
40.  Tan Y, Guo Q, Xie Q, Wang K, Yuan B, Zhou Y, Liu J, Huang J, He X, Yang X, He C, Zhao X. Single-walled carbon nanotubes (SWCNTs)-assisted cell-systematic evolution of ligands by exponential enrichment (cell-SELEX) for improving screening efficiency. Anal Chem. 2014;86:9466-9472.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 24]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
41.  Hung LY, Wang CH, Hsu KF, Chou CY, Lee GB. An on-chip Cell-SELEX process for automatic selection of high-affinity aptamers specific to different histologically classified ovarian cancer cells. Lab Chip. 2014;14:4017-4028.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 64]  [Cited by in F6Publishing: 56]  [Article Influence: 5.6]  [Reference Citation Analysis (0)]
42.  Wang CY, Wu CY, Hung TC, Wong CH, Chen CH. Sequence-constructive SELEX: A new strategy for screening DNA aptamer binding to Globo H. Biochem Biophys Res Commun. 2014;452:484-489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
43.  Ang A, Ouellet E, Cheung KC, Haynes C. Highly Efficient and Reliable DNA Aptamer Selection Using the Partitioning Capabilities of ddPCR: The Hi-Fi SELEX Method. Methods Mol Biol. 2018;1768:531-554.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 1]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
44.  Maio G, Enweronye O, Zumrut HE, Batool S, Van N, Mallikaratchy P. Systematic optimization and modification of a DNA aptamer with 2'-O-methyl RNA analogues. ChemistrySelect. 2017;2:2335-2340.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 16]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
45.  Wang Z, Xu W, Liu L, Zhu TF. A synthetic molecular system capable of mirror-image genetic replication and transcription. Nat Chem. 2016;8:698-704.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 136]  [Cited by in F6Publishing: 137]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
46.  Gissberg O, Zaghloul EM, Lundin KE, Nguyen CH, Landras-Guetta C, Wengel J, Zain R, Smith CI. Delivery, Effect on Cell Viability, and Plasticity of Modified Aptamer Constructs. Nucleic Acid Ther. 2016;26:183-189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
47.  Haruta K, Otaki N, Nagamine M, Kayo T, Sasaki A, Hiramoto S, Takahashi M, Hota K, Sato H, Yamazaki H. A Novel PEGylation Method for Improving the Pharmacokinetic Properties of Anti-Interleukin-17A RNA Aptamers. Nucleic Acid Ther. 2017;27:36-44.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 34]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
48.  Lee CH, Lee SH, Kim JH, Noh YH, Noh GJ, Lee SW. Pharmacokinetics of a Cholesterol-conjugated Aptamer Against the Hepatitis C Virus (HCV) NS5B Protein. Mol Ther Nucleic Acids. 2015;4:e254.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 49]  [Cited by in F6Publishing: 56]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
49.  Ali MH, Elsherbiny ME, Emara M. Updates on Aptamer Research. Int J Mol Sci. 2019;20:pii: E2511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 107]  [Cited by in F6Publishing: 91]  [Article Influence: 18.2]  [Reference Citation Analysis (0)]
50.  Xu J, Liao S, Tian C, Zhang L, Zhai M, Chen C, Tang J, Zeng J. [Screening of nucleic acid aptamer of lung cancer cells based on cell exponential enrichment ligand system evolution and its application in tumor diagnosis and treatment]. Sheng Wu Yi Xue Gong Cheng Xue Za Zhi. 2018;35:964-969.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
51.  Li G, Zeng J, Liu H, Ding P, Liang J, Nie X, Zhou Z. A fluorometric aptamer nanoprobe for alpha-fetoprotein by exploiting the FRET between 5-carboxyfluorescein and palladium nanoparticles. Mikrochim Acta. 2019;186:314.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 22]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
52.  Hu Z, Tan J, Lai Z, Zheng R, Zhong J, Wang Y, Li X, Yang N, Li J, Yang W, Huang Y, Zhao Y, Lu X. Aptamer Combined with Fluorescent Silica Nanoparticles for Detection of Hepatoma Cells. Nanoscale Res Lett. 2017;12:96.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 29]  [Cited by in F6Publishing: 24]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
53.  Wang S, Zhang C, Wang G, Cheng B, Wang Y, Chen F, Chen Y, Feng M, Xiong B. Aptamer-Mediated Transparent-Biocompatible Nanostructured Surfaces for Hepotocellular Circulating Tumor Cells Enrichment. Theranostics. 2016;6:1877-1886.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 42]  [Article Influence: 5.3]  [Reference Citation Analysis (0)]
54.  Joo M, Baek SH, Cheon SA, Chun HS, Choi SW, Park TJ. Development of aflatoxin B1 aptasensor based on wide-range fluorescence detection using graphene oxide quencher. Colloids Surf B Biointerfaces. 2017;154:27-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 36]  [Article Influence: 5.1]  [Reference Citation Analysis (0)]
55.  Kaur H. Aptamer Conjugated Quantum Dots for Imaging Cellular Uptake in Cancer Cells. J Nanosci Nanotechnol. 2019;19:3798-3803.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
56.  Zhou Y, Li W, Tseng Y, Zhang J, Liu J. Developing slow-off dickkopf-1 aptamers for early-diagnosis of hepatocellular carcinoma. Talanta. 2019;194:422-429.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 10]  [Article Influence: 1.7]  [Reference Citation Analysis (0)]
57.  Yu Q, Zhai F, Zhou H, Wang Z. Aptamer Conformation Switching-Induced Two-Stage Amplification for Fluorescent Detection of Proteins. Sensors (Basel). 2018;19:pii: E77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 6]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
58.  Bayat P, Taghdisi SM, Rafatpanah H, Abnous K, Ramezani M. In vitro selection of CD70 binding aptamer and its application in a biosensor design for sensitive detection of SKOV-3 ovarian cells. Talanta. 2019;194:399-405.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
59.  Bouvier-Müller A, Ducongé F. Application of aptamers for in vivo molecular imaging and theranostics. Adv Drug Deliv Rev. 2018;134:94-106.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 54]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
60.  Yan H, Gao X, Zhang Y, Chang W, Li J, Li X, Du Q, Li C. Imaging Tiny Hepatic Tumor Xenografts via Endoglin-Targeted Paramagnetic/Optical Nanoprobe. ACS Appl Mater Interfaces. 2018;10:17047-17057.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 26]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
61.  Zhong L, Zou H, Huang Y, Gong W, He J, Tan J, Lai Z, Li Y, Zhou C, Zhang G, Li G, Yang N, Zhao Y. Magnetic Endoglin Aptamer Nanoprobe for Targeted Diagnosis of Solid Tumor. J Biomed Nanotechnol. 2019;15:352-362.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 2.6]  [Reference Citation Analysis (0)]
62.  Zhao M, Liu Z, Dong L, Zhou H, Yang S, Wu W, Lin J. A GPC3-specific aptamer-mediated magnetic resonance probe for hepatocellular carcinoma. Int J Nanomedicine. 2018;13:4433-4443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 29]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
63.  Cao H, Fang X, Li H, Li H, Kong J. Ultrasensitive detection of mucin 1 biomarker by immuno-loop-mediated isothermal amplification. Talanta. 2017;164:588-592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 16]  [Cited by in F6Publishing: 16]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
64.  Bates PJ, Laber DA, Miller DM, Thomas SD, Trent JO. Discovery and development of the G-rich oligonucleotide AS1411 as a novel treatment for cancer. Exp Mol Pathol. 2009;86:151-164.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 635]  [Cited by in F6Publishing: 575]  [Article Influence: 38.3]  [Reference Citation Analysis (0)]
65.  Soldevilla MM, Meraviglia-Crivelli de Caso D, Menon AP, Pastor F. Aptamer-iRNAs as Therapeutics for Cancer Treatment. Pharmaceuticals (Basel). 2018;11:pii: E108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 30]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
66.  Zhu G, Chen X. Aptamer-based targeted therapy. Adv Drug Deliv Rev. 2018;134:65-78.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 314]  [Cited by in F6Publishing: 253]  [Article Influence: 42.2]  [Reference Citation Analysis (0)]
67.  Hu Z, He J, Gong W, Zhou N, Zhou S, Lai Z, Zheng R, Wang Y, Yang X, Yang W, Zhong L, Lu X, Zhao Y. TLS11a Aptamer/CD3 Antibody Anti-Tumor System for Liver Cancer. J Biomed Nanotechnol. 2018;14:1645-1653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
68.  Xiao S, Liu Z, Deng R, Li C, Fu S, Chen G, Zhang X, Ke F, Ke S, Yu X, Wang S, Zhong Z. Aptamer-mediated gene therapy enhanced antitumor activity against human hepatocellular carcinoma in vitro and in vivo. J Control Release. 2017;258:130-145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 24]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
69.  Sábio RM, Meneguin AB, Ribeiro TC, Silva RR, Chorilli M. New insights towards mesoporous silica nanoparticles as a technological platform for chemotherapeutic drugs delivery. Int J Pharm. 2019;564:379-409.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 60]  [Article Influence: 12.0]  [Reference Citation Analysis (0)]
70.  Babaei M, Abnous K, Taghdisi SM, Amel Farzad S, Peivandi MT, Ramezani M, Alibolandi M. Synthesis of theranostic epithelial cell adhesion molecule targeted mesoporous silica nanoparticle with gold gatekeeper for hepatocellular carcinoma. Nanomedicine (Lond). 2017;12:1261-1279.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 52]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
71.  Lan S, Lin Z, Zhang D, Zeng Y, Liu X. Photocatalysis Enhancement for Programmable Killing of Hepatocellular Carcinoma through Self-Compensation Mechanisms Based on Black Phosphorus Quantum-Dot-Hybridized Nanocatalysts. ACS Appl Mater Interfaces. 2019;11:9804-9813.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 38]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
72.  Wang R, Zhao N, Li S, Fang JH, Chen MX, Yang J, Jia WH, Yuan Y, Zhuang SM. MicroRNA-195 suppresses angiogenesis and metastasis of hepatocellular carcinoma by inhibiting the expression of VEGF, VAV2, and CDC42. Hepatology. 2013;58:642-653.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 160]  [Cited by in F6Publishing: 176]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
73.  Liu Y, Wu X, Gao Y, Zhang J, Zhang D, Gu S, Zhu G, Liu G, Li X. Aptamer-functionalized peptide H3CR5C as a novel nanovehicle for codelivery of fasudil and miRNA-195 targeting hepatocellular carcinoma. Int J Nanomedicine. 2016;11:3891-3905.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 27]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
74.  Cheng Y, Ou Z, Li Q, Yang J, Hu M, Zhou Y, Zhuang X, Zhang ZJ, Guan S. Cabazitaxel liposomes with aptamer modification enhance tumor targeting efficacy in nude mice. Mol Med Rep. 2019;19:490-498.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 7]  [Article Influence: 1.2]  [Reference Citation Analysis (0)]
75.  Qu L, Xu J, Tan X, Liu Z, Xu L, Peng R. Dual-aptamer modification generates a unique interface for highly sensitive and specific electrochemical detection of tumor cells. ACS Appl Mater Interfaces. 2014;6:7309-7315.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 47]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]