Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2311 2022-12-01 06:55:16 |
2 format Meta information modification 2311 2022-12-02 03:08:59 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Gholikhani, T.;  Kumar, S.;  Valizadeh, H.;  Mahdinloo, S.;  Adibkia, K.;  Zakeri-Milani, P.;  Barzegar-Jalali, M.;  Jimenez, B. Advances in Aptamers-Based Applications in Breast Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/37548 (accessed on 30 June 2024).
Gholikhani T,  Kumar S,  Valizadeh H,  Mahdinloo S,  Adibkia K,  Zakeri-Milani P, et al. Advances in Aptamers-Based Applications in Breast Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/37548. Accessed June 30, 2024.
Gholikhani, Tooba, Shalen Kumar, Hadi Valizadeh, Somayeh Mahdinloo, Khosro Adibkia, Parvin Zakeri-Milani, Mohammad Barzegar-Jalali, Balam Jimenez. "Advances in Aptamers-Based Applications in Breast Cancer" Encyclopedia, https://encyclopedia.pub/entry/37548 (accessed June 30, 2024).
Gholikhani, T.,  Kumar, S.,  Valizadeh, H.,  Mahdinloo, S.,  Adibkia, K.,  Zakeri-Milani, P.,  Barzegar-Jalali, M., & Jimenez, B. (2022, December 01). Advances in Aptamers-Based Applications in Breast Cancer. In Encyclopedia. https://encyclopedia.pub/entry/37548
Gholikhani, Tooba, et al. "Advances in Aptamers-Based Applications in Breast Cancer." Encyclopedia. Web. 01 December, 2022.
Advances in Aptamers-Based Applications in Breast Cancer
Edit

Aptamers are synthetic single-stranded oligonucleotides (such as RNA and DNA) evolved in vitro using Systematic Evolution of Ligands through Exponential enrichment (SELEX) techniques. Aptamers are evolved to have high affinity and specificity to targets; hence, they have a great potential for use in therapeutics as delivery agents and/or in treatment strategies. Aptamers can be chemically synthesized and modified in a cost-effective manner and are easy to hybridize to a variety of nano-particles and other agents which has paved a way for targeted therapy and diagnostics applications such as in breast tumors.

aptamer drug delivery cancer diagnostics

1. Introduction

Breast cancer is considered as one of the top five causes of mortality among women worldwide since 1980 [1]. Primary diagnosis and therapy are of pivotal aspects in the prognosis of breast cancer. Recently, constant education on breast cancer symptoms and how to spot breast differences require a medical opinion, and complementarily to the current diagnosis techniques such as mammograms, ultrasounds, MRI scans and biopsies, the survival rate of breast cancer is improved.
The most common ways of diagnosis are based on mammogram (imaging). However, molecular diagnosis techniques such as hormone receptor and protein expression methods (ER, PR, HER2, HERmark™), serum methods (CellSearch®, Biomarker translation test), and gene expression profiling methods (Mammaprint™, OncoVue® Test, GeneSearch™ breast lymph node, nipple fluid aspiration, DiaGenic BCtect®, Oncotype DX®, Theros H/ISM and MGISM) are also used [2]. These techniques has resulted in a 1.8% to 3.4% reduction per year in the breast cancer mortality rate in the last 30 years among U.S woman [3]. However, the current mortality rate is still attributable to a late diagnosis because of the patients’ fear of cancer diagnosis and treatment, competing life priorities, financial problems, embarrassment about having a breast examination, use of traditional methods, lack of information and mammography misinterpretation [4]. Therefore, there is still a need to innovate new strategies to reduce the current mortality rates that are less invasive, fast, do not require woman to expose their breasts and do not require expensive equipment or specialized personnel to interpret the results [5]. Every year 2.26 million new patients are diagnosed with breast cancer and 684,966 deaths are recorded globally due to breast cancer [6]. Woman from rural and remote areas are among the most affected; for example, in rural areas in China, where woman have less access to health care facilities and have a lower income, the mortality of breast cancer has increased in the last 25 years [7]. In 2019, in US approximately 232,000 women were diagnosed with breast cancer and of those, 40,000 died [8].
The proliferation index Ki-67 is an essential indicator of uncontrolled cellular proliferation in malignancy and critical to make a distinction between ‘Luminal A’ and ‘Luminal B (HER2 negative)’ subtypes [9][10][11][12]. Understanding the distinction between different tumor subtypes, has allowed researchers to implement specific treatments for each subtype specially for luminal A and B subtypes [13].

2. Affinity SELEX

Affinity column SELEX is suitable for target molecules such as metal ions, small organic and inorganic molecules, peptides, proteins and various cell types [14][15]. The target molecule is conjugated to a solid phase such as sepharose beads, magnetic particles, nitrocellulose membranes or graphene oxide polymers using functional groups such as amines, carboxyl and thiol groups [16]. The type of chemistry used for the target conjugation to the substrate depends on the substrate and the functional group. Following co-incubation of oligo pool with the affixed target, the oligo demonstrating affinity to the functionalized target is separated and enriched using PCR [17].
Blok et al. [18] used a thrombin affinity column prepared with concanavalin A, to find aptamers specific for thrombin.
Xie et al. [19] adopted carboxylated magnetic beads for HBsAg immobilization by some deoxynucleotide aptamers which can bind with high specificity to the surface antigen of hepatitis B virus (HBV) [20]. In immobilized metal affinity chromatography (IMAC), metal ions such as Zn(II), Cu(II), Ni (II), and Co(II) are immobilized on a support to fractionate and purify proteins in solutions; the IMAC method is used to enrich most of the phosphopeptides from whole cell lysates, and it is reported that titanium dioxide (TiO2) beads have an equivalent ability to enrich phosphopeptides [21][22].

3. Aptamer Utilization for Breast Cancer

Many reports have listed aptamers for diagnosis and therapeutic applications of various cancer targets. In particular, various groups have reported the identification of aptamers against biomarkers for breast cancer. Among the certified biomarkers against breast cancer, HER2 is considered as one of the most suitable and pivotal biomarkers and applied either for molecular division or for the targeted therapy of breast tumors in medical treatment. Namazi et al. [23] utilized Cell-SELEX to select an anti-HER2 single-strand DNA aptamer (known as H2) that demonstrated affinity to HER2 with a Kd of 270 nM. Qaureshi et al. [24] developed a label-free capacitive apta-sensor using H2 and continued micro-electrodes of the capacitor for HER2 identification in solutions. The highly sensitive apta-sensor was used to identify HER2 in specimen within a reasonable level of 0.2–2 ng/mL. In addition to HER2, other biomarkers of breast tumors were targeted to develop aptamers for detection of breast tumor cells [25][26][27][28][29]. Ahirware et al. [30] applied the HT-SELEX method to develop an ERα-specific DNA aptamer. The aptamer was internalized by breast tumor cells positively expressing Erα. Following internalization, the aptamer localized in the nucleus. The aptamer was utilized to represent mRNA level of ERα in breast tumor cells, and the findings were related to IHC identification of ERα in breast tumor tissues that were either positive or negative for ERα. Lie et al. [31] developed a label-free biosensor for a regulator protein, i.e., nucleolin, which has a modulating role in the stability of Bcl-2 mRNA in tumor cells. The AS1411 aptamer was applied on alarm cantilevers in the microcantilever array. AS1411 interactions with nucleolin in clinical samples stimulated surface stress alterations, causing various flaws between the reference and sensor cantilevers [32][33]. The complex showed increased sensitivity with a LOD of 1.0 nM.
Identification of breast tumors particularly by targeting circulating tumor cells (CTCs) in the serum of patients is pivotal for initial prognosis, diagnosis and monitoring of treatment effects of cancer [34]. Aptamers developed from either SELEX or Cell-SELEX is a potential option for identifying breast tumors [35][36][37][38][39]. Caei et al. [40] improved aptamer-based fluorescence detection by targeting mucin 1 (MUC1) using ssDNA aptamers combined with luminescent terbium (TbIII) for sensitive identification of breast tumor cells. In the presence of breast tumors, the aptamer attaches to MUC1 on the cell surface and the stimulation of Tb3+ induced fluorescence dictates the positive signal [41]. The apta-sensor displayed great sensitivity towards breast tumors with a confinement level of detection as low as 65 cells/mL. Moreover, Joe et al. [42] developed an apta-sensor that detected HER2 and MUC1 in of MCF-7 cells reaching 10 cells/mL level of detection sensitivity. Li et al. [43][44] combined aptamers with silver nanoparticles using MUC1 aptamers for the imaging of MCF-7 cells. The system could effectively differentiate breast tumor MCF-7 cells from more metastatic A549 human lung cancer and MDAMB-231 breast tumors.

4. Therapeutic Use of Aptamers

The applicability of aptamers is not only limited to analytical systems but also paves a way for therapeutic uses [45][46]. Aptamers with small molecular weights of ~20,000 Da are easy to penetrate tumors tissues so they have been considered as drugs [47][48]. Currently, there are a number of aptamers that have been examined for the treatment of clinical diseases, while some of them have already received FDA approval for the therapeutic utilization, such as Macugen (Pegaptanib Sodium Injection) for treatment of macular degeneration [33][49][50]. AGRO100 is an aptamer that binds to nucleolin, a protein found intranuclear in all cells, but is uniquely expressed on the surface of tumor cells. Pre-clinical testing demonstrated the inhibitory effect of AGRO100 on nucleolin function and proved its anti-cancer effects against lung, prostate, breast, cervical, and colon cancer, as well as malignant melanoma and leukemia.
In vivo therapeutic application of aptamers is limited due to nuclease decomposition and metabolism of aptamers in physiological conditions [51][52]. To reduce the decomposition and enhance effects of aptamer utility in in vivo therapeutics application, the original aptamers generally require chemical alteration. Modifications such as internucleotide linkage with 3–3′ and 5–5′capping in the terminus with an inverted nucleotide [53][54], 2′-substitutions and phosphodiester linkage replacement with 2-F, 2-NH2, 2-OMe, and sugar rings [55][56], incorporating unnatural nucleotides into the oligonucleotide chain [57], cyclization of nucleic acids by linking 5- and 3-termini [58][59], and dialkyl lipid/PEG/cholesterol modifications at the 5′-End [60][61][62] on the oligo backbone inhibit the nucleases from digesting the oligo aptamers.

Therapeutic Application of Aptamers in Breast Cancer

A therapeutic agent is something that acts directly on the target. Aptamers are able to modulate the performance of the targeted protein or mRNA, and could influence their physiological roles such as the initiation of apoptosis [63][64][65]. Balae et al. [66][67] adopted glutathione-attaching RNA aptamers to induce the apoptosis of tumor cells in breast cancer. The aptamers gathered reactive oxygen species (ROS), responsible for the regulation of caspases function in breast tumors. The AS1411 aptamer, capable of binding the Bcl-2 mRNA-attaching protein nucleolin, was assessed for the capability to stimulate Bcl-2 gene cytotoxicity and instability in MDA-MB-231 and MCF-7 breast tumors [68][69]. The AS1411 aptamer could suppress the homeostasis of MDA-MB-231 and MCF-7 cells, reducing the life-span of Bcl-2 genes in tumor cells, and hinder nucleolin from attaching to the full element of the AU region in the Bcl-2 gene, eventually triggering apoptotic pathway. Varshney et al. [70] adopted RNA aptamers (hTERTapt8.1, hTERTapt7.7, hTERTapt 9.5, and CR4/CR5) that were specified for the sequence of RNA interactive domain 2 (RID2) of Human telomerase reverse transcriptase (hTERT) [71]. It can specifically and tightly attach to the peptide of hTERT and modulate the performance of the enzyme of telomerase in MCF-7 tumor cells, indicating the RNA aptamers’ potential in the treatment of patients with breast cancer [71].

5. Drug Delivery Pathways of Aptamer

Chemotherapy is the key therapeutic strategy utilized for the treatment of the tumor cells of cancers [72]. Some chemotherapeutic medicines can act on normal healthy cells in addition to tumoral cells thus leading to multiple adversary effects. Targeted delivery of chemotherapeutic substances could reduce adverse effects improving the specificity and efficacy of the therapeutic agent [73]. Aptamers’ ability to specifically bind to its target allows for aptamers’ utility as a targeted drug delivery system in breast cancer treatment. Liu et al. [74] developed an aptamer to target HER2 and made a complex with doxorubicin (a drug commonly used in breast cancer treatment). The doxorubicin–aptamer complex was used as a targeted drug delivery system to HER2-positive breast tumor cells. The aptamer–doxorubicin complex successfully delivered the complex to HER2-positive breast tumors, while minimal cytotoxicity was reported for normal cells. Dai et al. [75] improved a MUC1-targeting system of drug delivery using a MUC1–aptamer complex. The DNA tetrahedron (Td) contained the drug inside its DNA structure. The complex of aptamer–Td can adoptively attach and present doxorubicin (Dox) to MUC1 positive breast tumors, resulting in increased cytotoxicity towards MUC1-positive MCF-7 breast tumors versus normal cells, which are negative for the MUC1 marker in vitro (p < 0.01). Tao and Wei, et al. reported that they can produce upgraded polydopamine (pD)-reformed nano-substance-aptamer bio-conjugates (Apt-pDDTX/NPs) for the therapy of breast tumor cells with in vivo applications [76]. Both in vivo animal and in vitro cell experiments showed that the Apt-pD-DTX/NPs can improve targeted medicinal delivery, decrease the deleterious aspects of the drugs and enhance the wellbeing of the surviving patients in the treatment of breast tumors [77].
In last decades, aptamer nanomaterial conjugates have been used as medicine delivery agents in breast cancer. Beqa et al. [78] generated a novel mixture of nano-material which consisted of gold nano-substances for photothermal treatment of breast tumors [79]. By using gold nanoparticle-decorated SWCNTs with SKBR-3 breast cancer cell specific S6 aptamers, SK-BR-3 breast tumors were eliminated by applying 10 min of laser radiation at 785 nm with a power of 1.5 w/cm2. S6 aptamers interact with cancer cells specifically, then conjugated AuPOP-decorated SWCNT aggregates on the surface of cancer cells so that the application of a laser will kill that particular cell [80]. Similarly, other scientists considered utilizing the benefits of gold nanomaterials due to the ability to absorb extended wavelength of plasmon (700–1000 nanometer) [81]. Cell-SELEX was utilized to develop a new KW16-13 DNA aptamer, which had affinity for metastatic tumors in the breast tissue [35]. Conjugation of AuNPs–aptamer (KW16-13–AuNPs) demonstrated a 71-fold affinity for metastatic tumors of breast tissue compared to KMF2–1a-AuNPs [82][83]. Maik et al. [84] conjugated aptamer AS1411 to gold nanospheres (AuNSs) for targeted therapy of breast cancer. AS1411–AuNSs demonstrated high durability both in serum and solutions, and was simply internalized by target cells in higher doses and lead to an elevated anti-cytotoxic/proliferative effects when compared to either the pure AS1411 aptamer or AuNSs [85]. Moreover, compared to unconjugated AS1411 or GNS linked to control oligonucleotides, the injection of AS1411–AuNSs in vivo remarkably suppressed the growth of xenografted tumors in mice without any toxic side effects, indicating AS1411–AuNSs is a suitable therapeutic candidate for clinical application in breast cancer treatment.
In addition to nanomaterials and chemical drugs, aptamers can deliver small interfering RNA for treatment of gene disorders [86][87][88]. Theil et al. [87] attached an RNA aptamer specific for HER2 to siRNA targeting the anti-apoptotic Bcl-2 gene for utility in HER2 positive tumors. The aptamer–siRNA conjugate was internalized by HER2-positive tumor cells and silenced expressed levels of Bcl-2, improving the sensitivity of HER2-positive breast tumors to chemotherapy. Wang et al. [89] developed an aptamer–siRNA conjugate; survivin exerts a negative feedback of the cell death pathway in cancerous stem cells of DOX-resistant breast cancer cells. Epithelial cell adhesion molecule (EpCAM), which is present at low levels in normal epithelial cells is highly overexpressed (up to 800-fold) in many solid cancers. Therefore, utilizing an active targeting system containing the RNA aptamer specific for EpCAM presented an elevated dose of siRNA to cancerous stem cells that silenced survivin and elevated tumor cell chemosensitivity, thus finally inhibiting the cancerous cell growth, and extending the mice survival carrying xenograft mammalian tumors.

References

  1. Parkin, D.; Läärä, E.; Muir, C. Estimates of the worldwide frequency of sixteen major cancers in 1980. Int. J. Cancer 1988, 41, 184–197.
  2. Zoon, C.K.; Starker, E.Q.; Wilson, A.M.; Emmert-Buck, M.R.; Libutti, S.K.; Tangrea, M.A. Current molecular diagnostics of breast cancer and the potential incorporation of microRNA. Expert Rev. Mol. Diagn. 2009, 9, 455–466.
  3. Hendrick, R.E.; Baker, J.A.; Helvie, M.A. Breast cancer deaths averted over 3 decades. Cancer 2019, 125, 1482–1488.
  4. Maghous, A.; Rais, F.; Ahid, S.; Benhmidou, N.; Bellahamou, K.; Loughlimi, H.; Marnouche, E.; Elmajjaoui, S.; Elkacemi, H.; Kebdani, T. Factors influencing diagnosis delay of advanced breast cancer in Moroccan women. BMC Cancer 2016, 16, 1–8.
  5. Liu, M.; Li, Z.; Yang, J.; Jiang, Y.; Chen, Z.; Ali, Z.; He, N.; Wang, Z. Cell-specific biomarkers and targeted biopharmaceuticals for breast cancer treatment. Cell Prolif. 2016, 49, 409–420.
  6. Cai, Z.; Liu, Q. Understanding the Global Cancer Statistics 2018: Implications for cancer control. Sci. China Life Sci. 2019, 64, 1017–1020.
  7. Zidan, H.E.; Karam, R.A.; El-Seifi, O.S.; Abd Elrahman, T.M. Circulating long non-coding RNA MALAT1 expression as molecular biomarker in Egyptian patients with breast cancer. Cancer Genet. 2018, 220, 32–37.
  8. DeSantis, C.E.; Fedewa, S.A.; Goding Sauer, A.; Kramer, J.L.; Smith, R.A.; Jemal, A. Breast cancer statistics, 2015: Convergence of incidence rates between black and white women. CA Cancer J. Clin. 2016, 66, 31–42.
  9. Lukong, K.E. Understanding breast cancer–The long and winding road. BBA Clin. 2017, 7, 64–77.
  10. Maleki, S.; Dorokhova, O.; Sunkara, J.; Schlesinger, K.; Suhrland, M.; Oktay, M.H. Estrogen, progesterone, and HER-2 receptor immunostaining in cytology: The effect of varied fixation on human breast cancer cells. Diagn. Cytopathol. 2013, 41, 864–870.
  11. Toss, A.; Cristofanilli, M. Molecular characterization and targeted therapeutic approaches in breast cancer. Breast Cancer Res. 2015, 17, 60.
  12. Wu, M.; Ma, J. Association between imaging characteristics and different molecular subtypes of breast cancer. Acad. Radiol. 2017, 24, 426–434.
  13. Goldhirsch, A.; Winer, E.P.; Coates, A.; Gelber, R.; Piccart-Gebhart, M.; Thürlimann, B.; Senn, H.-J.; Albain, K.S.; André, F.; Bergh, J. Personalizing the treatment of women with early breast cancer: Highlights of the St Gallen International Expert Consensus on the Primary Therapy of Early Breast Cancer 2013. Ann. Oncol. 2013, 24, 2206–2223.
  14. Paniel, N.; Istamboulié, G.; Triki, A.; Lozano, C.; Barthelmebs, L.; Noguer, T. Selection of DNA aptamers against penicillin G using Capture-SELEX for the development of an impedimetric sensor. Talanta 2017, 162, 232–240.
  15. Spiga, F.M.; Maietta, P.; Guiducci, C. More DNA–aptamers for small drugs: A capture–SELEX coupled with surface plasmon resonance and high-throughput sequencing. ACS Comb. Sci. 2015, 17, 326–333.
  16. Chatterjee, B.; Kalyani, N.; Anand, A.; Khan, E.; Das, S.; Bansal, V.; Kumar, A.; Sharma, T.K. GOLD SELEX: A novel SELEX approach for the development of high-affinity aptamers against small molecules without residual activity. Microchim. Acta 2020, 187, 1–13.
  17. Yang, L.; Gao, T.; Li, W.; Luo, Y.; Ullah, S.; Fang, X.; Cao, Y.; Pei, R. Ni-Nitrilotriacetic acid affinity SELEX method for selection of DNA aptamers specific to the N-cadherin protein. ACS Comb. Sci. 2020, 22, 867–872.
  18. Bock, L.C.; Griffin, L.C.; Latham, J.A.; Vermaas, E.H.; Toole, J.J. Selection of single-stranded DNA molecules that bind and inhibit human thrombin. Nature 1992, 355, 564–566.
  19. Xi, Z.; Huang, R.; Li, Z.; He, N.; Wang, T.; Su, E.; Deng, Y. Selection of HBsAg-specific DNA aptamers based on carboxylated magnetic nanoparticles and their application in the rapid and simple detection of hepatitis B virus infection. ACS Appl. Mater. Interfaces 2015, 7, 11215–11223.
  20. Qi, S.; Duan, N.; Khan, I.M.; Dong, X.; Zhang, Y.; Wu, S.; Wang, Z. Strategies to manipulate the performance of aptamers in SELEX, post-SELEX and microenvironment. Biotechnol. Adv. 2022, 55, 107902.
  21. Zhao, L.; Wang, Q.; Yin, Y.; Yang, Y.; Cui, H.; Dong, Y. Evolution of Interferon-Gamma Aptamer with Good Affinity and Analytical Utility by a Rational In Silico Base Mutagenesis Post-SELEX Strategy. Molecules 2022, 27, 5725.
  22. Krasitskaya, V.V.; Goncharova, N.S.; Biriukov, V.V.; Bashmakova, E.E.; Kabilov, M.R.; Baykov, I.K.; Sokolov, A.E.; Frank, L.A. The Ca2+-Regulated Photoprotein Obelin as a Tool for SELEX Monitoring and DNA Aptamer Affinity Evaluation. Photochem. Photobiol. 2020, 96, 1041–1046.
  23. Niazi, J.H.; Verma, S.K.; Niazi, S.; Qureshi, A. In vitro HER2 protein-induced affinity dissociation of carbon nanotube-wrapped anti-HER2 aptamers for HER2 protein detection. Analyst 2015, 140, 243–249.
  24. Qureshi, A.; Gurbuz, Y.; Niazi, J.H. Label-free capacitance based aptasensor platform for the detection of HER2/ErbB2 cancer biomarker in serum. Sens. Actuators B Chem. 2015, 220, 1145–1151.
  25. Guo, Q.; Li, X.; Shen, C.; Zhang, S.; Qi, H.; Li, T.; Yang, M. Electrochemical immunoassay for the protein biomarker mucin 1 and for MCF-7 cancer cells based on signal enhancement by silver nanoclusters. Microchim. Acta 2015, 182, 1483–1489.
  26. Lan, J.; Li, L.; Liu, Y.; Yan, L.; Li, C.; Chen, J.; Chen, X. Upconversion luminescence assay for the detection of the vascular endothelial growth factor, a biomarker for breast cancer. Microchim. Acta 2016, 183, 3201–3208.
  27. Meirinho, S.G.; Dias, L.G.; Peres, A.M.; Rodrigues, L.R. Development of an electrochemical aptasensor for the detection of human osteopontin. Procedia Eng. 2014, 87, 316–319.
  28. Sett, A.; Borthakur, B.B.; Sharma, J.D.; Kataki, A.C.; Bora, U. DNA aptamer probes for detection of estrogen receptor α positive carcinomas. Transl. Res. 2017, 183, 104–120.e102.
  29. Wu, J.; Wang, C.; Li, X.; Song, Y.; Wang, W.; Li, C.; Hu, J.; Zhu, Z.; Li, J.; Zhang, W. Identification, characterization and application of a G-quadruplex structured DNA aptamer against cancer biomarker protein anterior gradient homolog 2. PLoS ONE 2012, 7, e46393.
  30. Ahirwar, R.; Vellarikkal, S.K.; Sett, A.; Sivasubbu, S.; Scaria, V.; Bora, U.; Borthakur, B.B.; Kataki, A.C.; Sharma, J.D.; Nahar, P. Aptamer-assisted detection of the altered expression of estrogen receptor alpha in human breast cancer. PLoS ONE 2016, 11, e0153001.
  31. Li, H.; Bai, X.; Wang, N.; Chen, X.; Li, J.; Zhang, Z.; Tang, J. Aptamer-based microcantilever biosensor for ultrasensitive detection of tumor marker nucleolin. Talanta 2016, 146, 727–731.
  32. Chang, K.; Sun, P.; Dong, X.; Zhu, C.; Liu, X.; Zheng, D.; Liu, C. Aptamers as Recognition Elements for Electrochemical Detection of Exosomes. Chem. Res. Chin. Univ. 2022, 38, 879–885.
  33. Cruz-Hernández, C.D.; Rodríguez-Martínez, G.; Cortés-Ramírez, S.A.; Morales-Pacheco, M.; Cruz-Burgos, M.; Losada-García, A.; Reyes-Grajeda, J.P.; González-Ramírez, I.; González-Covarrubias, V.; Camacho-Arroyo, I. Aptamers as Theragnostic Tools in Prostate Cancer. Biomolecules 2022, 12, 1056.
  34. Duan, Y.; Zhang, C.; Wang, Y.; Chen, G. Research progress of whole-cell-SELEX selection and the application of cell-targeting aptamer. Mol. Biol. Rep. 2022, 49, 7979–7993.
  35. Jo, H.; Ban, C. Aptamer–nanoparticle complexes as powerful diagnostic and therapeutic tools. Exp. Mol. Med. 2016, 48, e230.
  36. Liu, F.; Zhang, Y.; Yu, J.; Wang, S.; Ge, S.; Song, X. Application of ZnO/graphene and S6 aptamers for sensitive photoelectrochemical detection of SK-BR-3 breast cancer cells based on a disposable indium tin oxide device. Biosens. Bioelectron. 2014, 51, 413–420.
  37. Liu, Q.; Jin, C.; Wang, Y.; Fang, X.; Zhang, X.; Chen, Z.; Tan, W. Aptamer-conjugated nanomaterials for specific cancer cell recognition and targeted cancer therapy. NPG Asia Mater. 2014, 6, e95.
  38. Meng, H.-M.; Fu, T.; Zhang, X.-B.; Tan, W. Cell-SELEX-based aptamer-conjugated nanomaterials for cancer diagnosis and therapy. Natl. Sci. Rev. 2015, 2, 71–84.
  39. Zhu, G.; Zhang, S.; Song, E.; Zheng, J.; Hu, R.; Fang, X.; Tan, W. Building fluorescent DNA nanodevices on target living cell surfaces. Angew. Chem. Int. Ed. 2013, 52, 5490–5496.
  40. Cai, S.; Li, G.; Zhang, X.; Xia, Y.; Chen, M.; Wu, D.; Chen, Q.; Zhang, J.; Chen, J. A signal-on fluorescent aptasensor based on single-stranded DNA-sensitized luminescence of terbium (III) for label-free detection of breast cancer cells. Talanta 2015, 138, 225–230.
  41. Zou, Y.; Wang, Y.; Wen, X.; Li, C.; Lei, L.; Guo, Q.; Sun, G.; Yu, L.; Nie, H. A DNA Aptamer Targeting Cellular Fibronectin Rather Than Plasma Fibronectin for Bioimaging and Targeted Chemotherapy of Tumors. Adv. Funct. Mater. 2022, 32, 2205002.
  42. Jo, H.; Her, J.; Ban, C. Dual aptamer-functionalized silica nanoparticles for the highly sensitive detection of breast cancer. Biosens. Bioelectron. 2015, 71, 129–136.
  43. Li, T.; Yang, J.; Ali, Z.; Wang, Z.; Mou, X.; He, N.; Wang, Z. Synthesis of aptamer-functionalized Ag nanoclusters for MCF-7 breast cancer cells imaging. Sci. China Chem. 2017, 60, 370–376.
  44. Malicki, S.; Pucelik, B.; Żyła, E.; Benedyk-Machaczka, M.; Gałan, W.; Golda, A.; Sochaj-Gregorczyk, A.; Kamińska, M.; Encarnação, J.C.; Chruścicka, B. Imaging of Clear Cell Renal Carcinoma with Immune Checkpoint Targeting Aptamer-Based Probe. Pharmaceuticals 2022, 15, 697.
  45. Zhou, J.; Rossi, J. Aptamers as targeted therapeutics: Current potential and challenges. Nat. Rev. Drug Discov. 2017, 16, 181–202.
  46. Keefe, A.D.; Pai, S.; Ellington, A. Aptamers as therapeutics. Nat. Rev. Drug Discov. 2010, 9, 537–550.
  47. Sun, H.; Zhu, X.; Lu, P.Y.; Rosato, R.R.; Tan, W.; Zu, Y. Oligonucleotide aptamers: New tools for targeted cancer therapy. Mol. Ther. Nucleic Acids 2014, 3, e182.
  48. Xiang, D.; Shigdar, S.; Qiao, G.; Wang, T.; Kouzani, A.Z.; Zhou, S.-F.; Kong, L.; Li, Y.; Pu, C.; Duan, W. Nucleic acid aptamer-guided cancer therapeutics and diagnostics: The next generation of cancer medicine. Theranostics 2015, 5, 23.
  49. Ng, E.W.; Shima, D.T.; Calias, P.; Cunningham, E.T.; Guyer, D.R.; Adamis, A.P. Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease. Nat. Rev. Drug Discov. 2006, 5, 123–132.
  50. Wandtke, T.; Wędrowska, E.; Szczur, M.; Przybylski, G.; Libura, M.; Kopiński, P. Aptamers—Diagnostic and Therapeutic Solution in SARS-CoV-2. Int. J. Mol. Sci. 2022, 23, 1412.
  51. Healy, J.M.; Lewis, S.D.; Kurz, M.; Boomer, R.M.; Thompson, K.M.; Wilson, C.; McCauley, T.G. Pharmacokinetics and biodistribution of novel aptamer compositions. Pharm. Res. 2004, 21, 2234–2246.
  52. Pourhajibagher, M.; Etemad-Moghadam, S.; Alaeddini, M.; Mousavi, M.; Bahador, A. DNA-aptamer-nanographene oxide as a targeted bio-theragnostic system in antimicrobial photodynamic therapy against Porphyromonas gingivalis. Sci. Rep. 2022, 12, 1–18.
  53. Gupta, S.; Hirota, M.; Waugh, S.M.; Murakami, I.; Suzuki, T.; Muraguchi, M.; Shibamori, M.; Ishikawa, Y.; Jarvis, T.C.; Carter, J.D. Chemically modified DNA aptamers bind interleukin-6 with high affinity and inhibit signaling by blocking its interaction with interleukin-6 receptor. J. Biol. Chem. 2014, 289, 8706–8719.
  54. Wei, J.; Song, R.; Sabbagh, A.; Marisetty, A.; Shukla, N.; Fang, D.; Najem, H.; Ott, M.; Long, J.; Zhai, L. Cell-directed aptamer therapeutic targeting for cancers including those within the central nervous system. OncoImmunology 2022, 11, 2062827.
  55. Schmidt, K.S.; Borkowski, S.; Kurreck, J.; Stephens, A.W.; Bald, R.; Hecht, M.; Friebe, M.; Dinkelborg, L.; Erdmann, V.A. Application of locked nucleic acids to improve aptamer in vivo stability and targeting function. Nucleic Acids Res. 2004, 32, 5757–5765.
  56. Ulrich, H. RNA aptamers: From basic science towards therapy. RNA Towards Med. 2006, 173, 305–326.
  57. Eulberg, D.; Klussmann, S. Spiegelmers: Biostable aptamers. ChemBioChem 2003, 4, 979–983.
  58. Liu, M.; Yin, Q.; Chang, Y.; Zhang, Q.; Brennan, J.D.; Li, Y. In vitro selection of circular DNA aptamers for biosensing applications. Angew. Chem. Int. Ed. 2019, 58, 8013–8017.
  59. Ni, S.; Zhuo, Z.; Pan, Y.; Yu, Y.; Li, F.; Liu, J.; Wang, L.; Wu, X.; Li, D.; Wan, Y. Recent Progress in Aptamer Discoveries and Modifications for Therapeutic Applications. ACS Appl. Mater. Interfaces 2020, 13, 9500–9519.
  60. Uludag, H.; Ubeda, A.; Ansari, A. At the intersection of biomaterials and gene therapy: Progress in non-viral delivery of nucleic acids. Front. Bioeng. Biotechnol. 2019, 7, 131.
  61. Gragoudas, E.S.; Adamis, A.P.; Cunningham Jr, E.T.; Feinsod, M.; Guyer, D.R. Pegaptanib for neovascular age-related macular degeneration. New Engl. J. Med. 2004, 351, 2805–2816.
  62. De Smidt, P.C.; Doan, T.L.; Falco, S.d.; Berkel, T.J.v. Association of antisense oligonucleotides with lipoproteins prolongs the plasma half-life and modifies the tissue distribution. Nucleic Acids Res. 1991, 19, 4695–4700.
  63. Camorani, S.; Crescenzi, E.; Gramanzini, M.; Fedele, M.; Zannetti, A.; Cerchia, L. Aptamer-mediated impairment of EGFR-integrin αvβ3 complex inhibits vasculogenic mimicry and growth of triple-negative breast cancers. Sci. Rep. 2017, 7, 46659.
  64. Chen, K.; Liu, J.; Tong, G.; Liu, B.; Wang, G.; Liu, H. Adipo8, a high-affinity DNA aptamer, can differentiate among adipocytes and inhibit intracellular lipid accumulation in vitro. Sci. China Chem. 2015, 58, 1612–1620.
  65. Chen, Y.; Lin, J.S. The application of aptamer in apoptosis. Biochimie 2017, 132, 1–8.
  66. Bala, J.; Bhaskar, A.; Varshney, A.; Singh, A.K.; Dey, S.; Yadava, P. In vitro selected RNA aptamer recognizing glutathione induces ROS mediated apoptosis in the human breast cancer cell line MCF 7. RNA Biol. 2011, 8, 101–111.
  67. Yuhan, J.; Zhu, L.; Zhu, L.; Huang, K.; He, X.; Xu, W. Cell-specific aptamers as potential drugs in therapeutic applications: A review of current progress. J. Control. Release 2022, 346, 405–420.
  68. Soundararajan, S.; Chen, W.; Spicer, E.K.; Courtenay-Luck, N.; Fernandes, D.J. The nucleolin targeting aptamer AS1411 destabilizes Bcl-2 messenger RNA in human breast cancer cells. Cancer Res. 2008, 68, 2358–2365.
  69. Ibarra, L.E.; Camorani, S.; Agnello, L.; Pedone, E.; Pirone, L.; Chesta, C.A.; Palacios, R.E.; Fedele, M.; Cerchia, L. Selective photo-assisted eradication of Triple-Negative breast cancer cells through aptamer decoration of doped conjugated polymer nanoparticles. Pharmaceutics 2022, 14, 626.
  70. Varshney, A.; Bala, J.; Santosh, B.; Bhaskar, A.; Kumar, S.; Yadava, P.K. Identification of an RNA aptamer binding hTERT-derived peptide and inhibiting telomerase activity in MCF7 cells. Mol. Cell. Biochem. 2017, 427, 157–167.
  71. Bayat, P.; Abnous, K.; Balarastaghi, S.; Taghdisi, S.M.; Saeedi, M.; Yazdian-Robati, R.; Mahmoudi, M. Aptamer AS1411-functionalized gold nanoparticle-melittin complex for targeting MCF-7 breast cancer cell line. Nanomed. J. 2022, 9, 164–169.
  72. Chen, Y.; Shi, S. Advances and prospects of dynamic DNA nanostructures in biomedical applications. RSC Adv. 2022, 12, 30310–30320.
  73. Chen, Z.; Zeng, Z.; Wan, Q.; Liu, X.; Qi, J.; Zu, Y. Targeted immunotherapy of triple-negative breast cancer by aptamer-engineered NK cells. Biomaterials 2022, 280, 121259.
  74. Liu, Z.; Duan, J.-H.; Song, Y.-M.; Ma, J.; Wang, F.-D.; Lu, X.; Yang, X.-D. Novel HER2 aptamer selectively delivers cytotoxic drug to HER2-positive breast cancer cells in vitro. J. Transl. Med. 2012, 10, 148.
  75. Dai, B.; Hu, Y.; Duan, J.; Yang, X.-D. Aptamer-guided DNA tetrahedron as a novel targeted drug delivery system for MUC1-expressing breast cancer cells in vitro. Oncotarget 2016, 7, 38257.
  76. Tao, W.; Zeng, X.; Wu, J.; Zhu, X.; Yu, X.; Zhang, X.; Zhang, J.; Liu, G.; Mei, L. Polydopamine-based surface modification of novel nanoparticle-aptamer bioconjugates for in vivo breast cancer targeting and enhanced therapeutic effects. Theranostics 2016, 6, 470.
  77. Chaudhuri, A.; Ramesh, K.; Kumar, D.N.; Dehari, D.; Singh, S.; Kumar, D.; Agrawal, A.K. Polymeric micelles: A novel drug delivery system for the treatment of breast cancer. J. Drug Deliv. Sci. Technol. 2022, 77, 103886.
  78. Beqa, L.; Fan, Z.; Singh, A.K.; Senapati, D.; Ray, P.C. Gold nano-popcorn attached SWCNT hybrid nanomaterial for targeted diagnosis and photothermal therapy of human breast cancer cells. ACS Appl. Mater. Interfaces 2011, 3, 3316–3324.
  79. Fu, Z.; Xiang, J. Aptamer-Functionalized Nanoparticles in Targeted Delivery and Cancer Therapy. Int. J. Mol. Sci. 2020, 21, 9123.
  80. Liu, M.; Wang, L.; Lo, Y.; Shiu, S.C.-C.; Kinghorn, A.B.; Tanner, J.A. Aptamer-Enabled Nanomaterials for Therapeutics, Drug Targeting and Imaging. Cells 2022, 11, 159.
  81. Stern, J.M.; Stanfield, J.; Kabbani, W.; Hsieh, J.-T.; Cadeddu, J.A. Selective prostate cancer thermal ablation with laser activated gold nanoshells. J. Urol. 2008, 179, 748–753.
  82. Alkilany, A.M.; Thompson, L.B.; Boulos, S.P.; Sisco, P.N.; Murphy, C.J. Gold nanorods: Their potential for photothermal therapeutics and drug delivery, tempered by the complexity of their biological interactions. Adv. Drug Deliv. Rev. 2012, 64, 190–199.
  83. Chandrasekaran, R.; Lee, A.S.W.; Yap, L.W.; Jans, D.A.; Wagstaff, K.M.; Cheng, W. Tumor cell-specific photothermal killing by SELEX-derived DNA aptamer-targeted gold nanorods. Nanoscale 2016, 8, 187–196.
  84. Malik, M.T.; O’Toole, M.G.; Casson, L.K.; Thomas, S.D.; Bardi, G.T.; Reyes-Reyes, E.M.; Ng, C.K.; Kang, K.A.; Bates, P.J. AS1411-conjugated gold nanospheres and their potential for breast cancer therapy. Oncotarget 2015, 6, 22270.
  85. Liu, Y.; Ding, M.; Guo, K.; Wang, Z.; Zhang, C.; Shubhra, Q.T. Systemic Co-delivery of drugs by a pH-and photosensitive smart nanocarrier to treat cancer by chemo-photothermal-starvation combination therapy. Smart Mater. Med. 2022, 3, 390–403.
  86. Herrmann, A.; Priceman, S.J.; Kujawski, M.; Xin, H.; Cherryholmes, G.A.; Zhang, W.; Zhang, C.; Lahtz, C.; Kowolik, C.; Forman, S.J. CTLA4 aptamer delivers STAT3 siRNA to tumor-associated and malignant T cells. J. Clin. Investig. 2014, 124, 2977–2987.
  87. Thiel, K.W.; Hernandez, L.I.; Dassie, J.P.; Thiel, W.H.; Liu, X.; Stockdale, K.R.; Rothman, A.M.; Hernandez, F.J.; McNamara, J.O.; Giangrande, P.H. Delivery of chemo-sensitizing siRNAs to HER2+-breast cancer cells using RNA aptamers. Nucleic Acids Res. 2012, 40, 6319–6337.
  88. Jeong, H.; Lee, S.H.; Hwang, Y.; Yoo, H.; Jung, H.; Kim, S.H.; Mok, H. Multivalent Aptamer–RNA Conjugates for Simple and Efficient Delivery of Doxorubicin/siRNA into Multidrug-Resistant Cells. Macromol. Biosci. 2017, 17, 1600343.
  89. Wang, T.; Gantier, M.P.; Xiang, D.; Bean, A.G.; Bruce, M.; Zhou, S.-F.; Khasraw, M.; Ward, A.; Wang, L.; Wei, M.Q. EpCAM aptamer-mediated survivin silencing sensitized cancer stem cells to doxorubicin in a breast cancer model. Theranostics 2015, 5, 1456.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , , ,
View Times: 313
Revisions: 2 times (View History)
Update Date: 02 Dec 2022
1000/1000
Video Production Service