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 + 3140 word(s) 3140 2021-12-28 08:23:59

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.
Camorani, S.; Fedele, M.; Cerchia, L. Profiling Cancer Cells by Cell-SELEX. Encyclopedia. Available online: https://encyclopedia.pub/entry/18549 (accessed on 27 July 2024).
Camorani S, Fedele M, Cerchia L. Profiling Cancer Cells by Cell-SELEX. Encyclopedia. Available at: https://encyclopedia.pub/entry/18549. Accessed July 27, 2024.
Camorani, Simona, Monica Fedele, Laura Cerchia. "Profiling Cancer Cells by Cell-SELEX" Encyclopedia, https://encyclopedia.pub/entry/18549 (accessed July 27, 2024).
Camorani, S., Fedele, M., & Cerchia, L. (2022, January 20). Profiling Cancer Cells by Cell-SELEX. In Encyclopedia. https://encyclopedia.pub/entry/18549
Camorani, Simona, et al. "Profiling Cancer Cells by Cell-SELEX." Encyclopedia. Web. 20 January, 2022.
Profiling Cancer Cells by Cell-SELEX
Edit

The identification of tumor cell-specific surface markers is a key step towards personalized cancer medicine, allowing early assessment and accurate diagnosis, and development of efficacious targeted therapies. What mainly limits the number of ideal clinical biomarkers is the high complexity and heterogeneity of several human cancers and still-limited methods for molecular profiling of specific cancer types. The cell-SELEX (Systematic Evolution of Ligands by Exponential Enrichment) technology for the differential selection of oligonucleotide aptamers against a specific cancer-cell type has become the selection technique for the discovery of cell-surface markers. Indeed, it allows selection, at the same time, of a set of aptamers acting as highly efficacious recognition elements for functional surface signatures of target cells. Importantly, these aptamers may be used to identify cell-surface molecules whose role is still unexplored. This fulfills the great challenge of simultaneously targeting multiple proteins whose alterations, in concert, define the pathological state of the cell and are thus more informative for biomarker discovery than the alteration of a single protein.

cell-SELEX aptamer cancer cell phenotype biomarker discovery cell-profiling targeted therapy

1. Profiling Cancer Cells by Cell-SELEX

One of the greatest advantages of cell-SELEX is the possibility to perform the selection against a specific cell type without the prior knowledge of protein targets present on its surface. In such a way, a panel of aptamers can be generated that specifically recognizes the surface signature of the target cells and, through it, can distinguish those cells from the cells chosen for the counter-selection. These aptamers are applied as useful bioreagents for active cancer targeting and, notably, can be used as bait for the identification of new protein targets, playing an important role in biomarker discovery (Figure 1).
Figure 1. Aptamer discovery and development process. The image shows the sequential steps from the generation of aptamers by SELEX to their preclinical development as cancer-targeted therapeutics, imaging agents for diagnostics and theranostic molecules. Briefly, cancer cell-specific aptamers are generated by cell-SELEX against a specific cell type, without the prior knowledge of the molecular target. High affinity ligands are identified by classical cloning and/or high-throughput sequencing and bioinformatic analysis. The best aptamer candidates are tested in vitro for their affinity and specificity, optimized to increase their binding efficacy and tested in vitro/in vivo as cancer-targeted agents. The best binding aptamers are used as bait to identify their cell-surface targets, thus leading to discovery of novel biomarkers.
Cell-SELEX has been applied to different tumor types, including highly aggressive and heterogeneous cancers that lack well-defined biomarkers for a targeted therapy, with the intent to discover new aptamers able to bind to surface proteins, which differ in expression level between healthy and unhealthy cells or among different cancer cell phenotypes.
The first group who realized the opportunity to dissect biological complex targets using SELEX technology was Morris and Jensen’s group in 1998. They used human red blood cell membrane preparations as a complex mixture of potential targets to select a set of cell-specific ssDNA aptamers with high affinity for different cell membrane proteins[1].t was not long before there was an understanding that this approach could be also applied to decode the specific surface signature of different kinds of cancer cells.
Tan’s group applied the SELEX technology to living cells to explore membrane protein biomarkers. In their pioneering work, they screened a DNA library on cultured precursor T cell acute lymphoblastic leukemia (ALL) to generate a panel of aptamers specifically discriminating target cells from human Burkitt’s lymphoma cells, used in the counter-selection steps. In particular, five aptamers (sgc8, sgc3, sgd3, sgc4, and sgd2) were able to bind to target cells at high affinity, with KD values ranging from 0.80 ± 0.09 nM (sgc8) to 26.6 ± 2.1 nM (sgc4), and other ALL cell lines, but not to cultured B cells and acute myeloid leukemia (AML) cells [2]. In addition, these aptamers could discriminate different leukemia cells (T-ALL, B-ALL, AML) in clinical samples, thus detecting subtle molecular differences among individual samples from leukemia patients in the same category [3]. Post-SELEX target identification, based on aptamer-mediated affinity purification and mass spectrometry, allowed the match of sgc8 aptamer with its target, the transmembrane receptor protein tyrosine kinase 7 (PTK7) [4]. Subsequently, as discussed below, this aptamer was applied as a new therapeutic tool for haemato-oncological malignancies [5]. By using a similar strategy, Tan’s group also identified the immunoglobin heavy mu chain as the target for TD05 aptamer, which was selected by using the Burkitt’s lymphoma cell line Ramos as the target [6]. Both of these studies demonstrate that this two-step strategy, the development of high-quality aptamer probes and the identification of their target proteins, is a powerful approach for biomarker discovery.
Through the use of several protocols, essentially based on altering positive selection steps on the chosen target cells and counter-selection steps on off-target cells, several aptamers have been to date developed with binding to unknown but unique characteristic surface proteins of target cells. For instance, aptamers have been generated that are able to discriminate high-metastatic from low-metastatic cancer cells of different tumor types, including colorectal carcinoma [7], breast cancer [8][9][10], osteosarcoma [11], prostate cancer [12][13] hepatocellular carcinoma [14][15][16] and colon cancer [17][18]. The generation of the above aptamer probes specifically targeting metastatic cancer cells provides a significant tool for diagnosis and treatment of the metastatic disease. For one of them, the investigation has already progressed to the target identification. Indeed, the RNA Apt63 aptamer, generated by a cell-SELEX approach for differential binding to prostate cancer cell lines with high vs. low metastatic potential, when used for aptamer-based affinity purification combined with mass spectrometry, matched to the plasma membrane ATP synthase beta subunit (ecto-ATP5B). Testing of the aptamer in vitro, as well as in xenograft models and human samples, proved this protein is a new marker for predicting and treating metastatic breast and prostate cancers [13]. Moreover, applied to cancer cells belonging to different tumor types (pancreatic cancer PANC-1 cells vs. hepatocarcinoma Huh7 cells) a blind cell-SELEX approach raised an aptamer able to regulate epithelial–mesenchymal transition (EMT) and inhibit metastasis in pancreatic cancer by binding to cell-surface vimentin, as revealed by post-SELEX liquid chromatography tandem mass spectrometry analyses [19].
It is known that cancer stem cells (CSCs) represent a small fraction of cells within a tumor mass exhibiting self-renewal and tumor-initiating capabilities, which contribute to recurrence, metastasis and therapeutic resistance. Unfortunately, specific biomarkers for CSCs are lacking, thus it remains extremely hard to eradicate them by effective therapeutic strategies [20]. Notably, differential cell-SELEX has been applied to identify new markers of CSCs. Some groups succeeded in generating aptamers able to differentiate glioma stem cells from differentiated cells [21][22], highlighting the potential of aptamers to target a molecular signature of CSCs for therapeutic applications. Similarly, aptamers have been generated to bind stemness-enriched cells in colorectal [23], pancreatic [24] and prostate [25] cancers. The identification of the molecular targets of these aptamers may reveal new CSC biomarkers.
Further, to address the issue of resistance to therapy, cell-SELEX has been applied to discriminate drug-resistant cancer cells from sensitive counterparts. Recently, by using vemurafenib-resistant melanoma cells as a target of the selection and sensitive cells in counter-selection steps, Li et al. identified an aptamer specifically binding to CD63 on the surface of cancer cells [26], thus opening the possibility to interfere with the TIMP-1/CD63 interaction at the cell surface, which recently emerged as a driver of malignant progression in melanoma and other human cancers [27]. Furthermore, by a SELEX approach using taxol-resistant colon cancer cells in the positive selection and parental cells in counter-selection steps, Zhang et al. identified a DNA aptamer binding to human TfR at affinity comparable to that of human Tf. Importantly, they proved the ability of the aptamer to cross the intestinal epithelium barrier through TfR-mediated transcytosis, indicating its potential as a carrier for active drug delivery [28].
In addition, we applied the differential cell-SELEX approach to different tumors, including NSCLC [29] and glioblastoma (GBM) [30], to generate aptamers able to target, within the same tumor type, cells characterized by a phenotype more aggressive than that of the cells used in the counter-selection, in terms of resistance to chemotherapy and tumorigenicity. Combining different post-SELEX biochemical approaches, we were able to identify EGFR and platelet-derived growth factor receptor beta (PDGFRβ) as the molecular targets of CL4 and Gint4.T aptamers, respectively, with the first coming from the selection on NSCLC cells [29] and the second from that on GBM cells [30]. Both 2′-fluoro-pyrimidine (2′-F-Py) containing RNA aptamers were subsequently validated as ligands and inhibitors of their proper receptor targets, not only when applied to cancer cells used for their selection but also in different tumor types, thus contributing to insight on the oncogenic role of these two RTKs, depending on the specific tumor expressing them. For instance, the anti-EGFR CL4 aptamer exerts a strong apoptotic effect on human NSCLC [29], blocks the invasiveness of GBM cells expressing either the EGFRwt or EGFRvIII mutant [30][31] and prevents the EGFR/integrin αvβ3 interaction on the surface of TNBC cells with a mesenchymal stem-like phenotype, which we first found to be required for vasculogenic mimicry and tumor growth of aggressive and poorly differentiated TNBC subtype [32][33].
Similarly, the Gint4.T aptamer acts as a neutralizing ligand for PDGFRβ in cell lines, primary cultures and xenografts models of GBM [30]. Further, it has been applied as a highly effective tool for imaging and suppression of TNBC lung metastases, thus indicating PDGFRβ as a reliable biomarker of a subgroup of TNBCs with invasive and stemlike phenotype [33][34]. More recently, Gint4.T has been proven to potentiate the efficacy of immunotherapy with anti-programmed death-ligand 1 (PD-L1) mAb in the inhibition of tumor growth and metastasis in a syngeneic TNBC mouse model [35].
Moreover, several cell-SELEX protocols have been applied to cancer cell lines by using normal cells for the counter-selection with the aim to identify novel cancer biomarkers for improving early diagnosis and therapy. For instance, 2′-F-Py RNA aptamers were selected on two different pancreatic cancer cell lines able to bind target cells and discriminate them from normal pancreatic ductal cells. Interestingly, aptamer-based target pull-down experiments on cell membrane lysates, combined with a genome-wide microarray analysis in cells targeted or not by the aptamer, identified the oncofetal protein alkaline phosphatase placental-like 2 (ALPPL-2) as the target of one of the selected aptamers and attributed a novel function to this protein as promoter of pancreatic cancer cell growth and invasion [36]. By a similar approach, four aptamers were identified that differentiate nasopharyngeal cancer cells from nonmalignant nasopharyngeal cells, the cell-surface receptor CD109 identified as the target of one of them [37]. The overexpression of CD109 in many human cancers and its association with metastasis and chemoresistance makes it an attractive target for diagnosis and therapy [38].
Moreover, in order to generate aptamers against tumor cells in more physiologically conditions, 3D cell-SELEX protocols have been applied to spheroids of prostate [39] and breast [40] cancer cells by using nontumor cells for the negative selection.

2. Unravel Cancer Heterogenicity by Cell-SELEX

One of the major factors limiting the number of effective targeted therapies is represented by cancer heterogeneity. Indeed, cancer is a dynamic disease and various cell subpopulations develop during its course, which are characterized by unique genotypes and phenotypes correlating to different biological behaviors and sensitivity to treatments. Distinct molecular signatures can be found among different tumors or inside the same one (inter- and intratumor, respectively). At the basis of the intratumor heterogeneity, there is a continuous process of evolution that involves all tumor cells, not only in time but also in space. For instance, Zhao et al. observed that the location of genetically distinct subclones in a tumor reveals their evolution, with the most aggressive and prone to metastasis located in the center of the tumor [41].
Phenotypic heterogeneity in different cell populations contributes to cancer drug resistance and hampers treatment outcome [42], but sufficient characteristics have not yet been determined to allow discrimination of the subpopulations present in a tumor. Cell-SELEX is an important approach for unraveling differently expressed cell-surface proteins of heterogeneous cancers such as GBM, breast and pancreatic cancers, which still lack personalized treatment protocols.

2.1. GBM

GBMs, classified as grade 4 astrocytomas, are the most frequent and aggressive malignant tumors occurring in the human brain and, according to gene expression, they were classified by the TCGA consortium into four molecular subtypes (proneural, mesenchymal, neuronal and classical) associated with different prognosis [43]. One of the main reasons for GBM aggressiveness is its intrinsic intratumor heterogeneity; indeed, it is composed of tumor stem cells, differentiated tumor cells, cells from the blood vessels, and inflammatory cells. The existence of these different subpopulations and their interactions with the TME components contributes to resistance to conventional treatments [44].
In order to create new opportunities for better treatment options, Lin et al. [45] applied a cell-SELEX protocol to human glioma SHG44 cells, a grade 3 human anaplastic astrocytoma, by using human astrocyte SVGp12 cells, for counter-selection. Aptamer S6-1b, obtained by truncating the long version coming from the selection, showed excellent specificity, discriminating target cells from SVGp12 cells and other cancer cell lines, including GBM U87, T98G and U251 cells. Cy5-labeled aptamers, intravenously injected in mice bearing subcutaneous SHG44-derived tumors, rapidly accumulated in the tumor and persisted for over 4 h, resulting in useful tools for noninvasive imaging of the tumor. Aptamer-mediated affinity purification of cell membrane proteins identified fibronectin, an extracellular matrix protein overexpressed in glioma, as the potential target of S6-1b.
Further, using SELEX technology on T98G target cells and SVGp12 off-target cells, Wu et al. selected two aptamers, WYZ-41a and WYZ-50a, able to bind to T98G but not to either U87 and U251 GBM cell lines [46] consistently, with major differences at the cell surface among them [47]. The aptamers were stable for 2 h and retained their binding capability in cerebral spinal fluid, thus indicating a great potential in using these aptamers for biomedical applications.
In addition, among a group of DNA aptamers selected on a cell line (K308) derived from gliosarcoma, a variant of GBM, WQY-9 aptamer was capable of recognizing gliosarcoma cells and gliosarcoma tissues, differentiating gliosarcoma from GBM [48].

2.2. Breast Cancer

Luminal A, Luminal B, HER-2 enriched and TNBC represent the four main subtypes of breast cancer [49]. They are characterized by different behavior and therapeutic sensitivity. Hence, dissecting the heterogeneity of its subpopulations is also necessary for the management of breast cancer. A differential cell-SELEX was applied to HER2-overexpressing breast cancer SK-BR-3 cells by using a pool of three counter-selections against Luminal A MCF-7 cells, breast normal MCF-10A cells and TNBC MDA-MB-231 cells. The length-optimized sk6Ea aptamer, resulting in the best binding candidate, was able to bind specifically to the target cells without recognizing the three counter-selection cell lines both in vitro and in vivo, upon intravenous injection in subcutaneous tumor-bearing mice. Moreover, sk6Ea when cemented on breast cancer tissue sections, efficiently discriminated the three different breast cancer subtypes [50].
TNBC, defined by the lack of ER, progesterone receptor (PR), HER-2 expression, is significantly more aggressive than other breast cancers, diagnosed at a later stage and more likely to develop recurrence. It is highly heterogeneous and the presence of different subtypes, related to different cancer features, makes it difficult to establish an appropriate therapy [49]. Lehman et al. classified TNBC into four subtypes: Basal-like 1, Basal-like 2, mesenchymal and luminal androgen receptor [51][52]. Recently, we applied a cell-SELEX method for the specific recognition of MDA-MB-231 cells, a highly metastatic human cell line representing an established model for the aggressive and undifferentiated mesenchymal TNBC subtype [53]. At each round, the positive selection step on TNBC cells was preceded by counter-selection against non-TNBC breast cancer BT-474 cells, which express high level of ER, PR, and HER-2, and A431 epidermoid cancer cells. By using high-throughput NGS and bioinformatics, we identified a panel of six 2′-F-Py RNA aptamers able to bind to human TNBC cell lines (MDA-MB-231, BT-549, MDA-MB-436, DU4475, MDA-MB-468) covering different TNBC subtypes, and to distinguish them from both normal cells and non-TNBC breast cancer cell lines representative of luminal A (MCF-7 and T47D) and HER2-positive (SK-BR-3) molecular categories. Further, these aptamers can be used for staining of histological tissues to differentiate TNBC human samples, showing a distinct pattern of binding on different tumors [53]. In addition to their binding capability, the aptamers actively internalize into target cells and inhibit the mammosphere-forming ability of TNBC cell lines. Therefore, we anticipate that the identification of their targets will help to expand the limited repertoire of actionable TNBC biomarkers.

2.3. Pancreatic Cancer

Pancreatic cancer is classified as basal, with the worst prognosis and classical clinical subtypes. More than 70% of patients are unresponsive to the current therapies, and despite many efforts made in past years there are no improvements, principally due to its great heterogeneity [54]. To determine if there were molecular markers differently expressed on pancreatic cancer, Yoon et al. investigated the expression of seven different markers (five EMT markers, one proliferation marker, and one leukocyte antigen marker) on human cancer tissues by using a multiplexed tissue imaging mass cytometer platform. They observed a substantial heterogeneity both intratumor and in tumors of the same grade [55]. Nevertheless, a cell-SELEX protocol using pancreatic cancer PANC-1 cells for the positive selection and hepatocellular carcinoma cells for counter-selection led to the identification of mitochondrial heat shock protein 70 or mortalin, as a potential biomarker of pancreatic cancer [56]. This protein is expressed on the surface of cells belonging to some human cancers, such as colorectal, neuronal and pancreatic, but not to normal tissues.

2.4. TME

The role of stroma microenvironment in both solid tumors as well as malignant haematological disease is well-accepted. Based on the high level of heterogeneity in the nature of mutations present between patients and even within patients during the different stages of their disease as well as the clear role of tumor/stroma interactions in chemoresistance, targeting interactions between tumor cells and their stroma provide new therapeutic approaches [57]. Because of the role of tumor-associated macrophages (TAMs) in supporting tumor progression and dissemination and their linking with worse clinical outcome and resistance to conventional therapies, many efforts are focused on the development of TAMs-targeted strategies for cancer treatment. To this aim, Sylvestre et al. aimed to identify by cell-SELEX an aptamer able to target this cell population, discriminating it from resident (M0-like) or tumoricidal (M1-like) macrophages [58]. Despite the use of negative selection screens on monocytes and M0-like macrophages, and positive selection on M2-like macrophages, resembling TAMs, they selected an aptamer, named A2, that binds to both human M0- and M2-like macrophages and monocytes, probably due to overlapping receptor expression between M0- and M2-like macrophages. Conversely, A2 does not recognize M1-like macrophages or other leukocyte populations. Because of its binding and internalization into CD14+ but not CD16+ monocytes, which is also observed in vivo, A2 holds great potential for drug delivery approaches targeting monocytes. Based on the binding behavior of A2 to monocytes, they hypothesized and then confirmed that the aptamer binds to the CD14 receptor, which is expressed by both monocytes and macrophages.

References

  1. Kevin N. Morris; Kirk Jensen; Carol M. Julin; Michael Weil; Larry Gold; High affinity ligands from in vitro selection: Complex targets. Proceedings of the National Academy of Sciences 1998, 95, 2902-2907, 10.1073/pnas.95.6.2902.
  2. Dihua Shangguan; Y. Li; Z. Tang; Z. C. Cao; H. W. Chen; Prabodhika Mallikaratchy; K. Sefah; C. J. Yang; W. Tan; Aptamers evolved from live cells as effective molecular probes for cancer study. Proceedings of the National Academy of Sciences 2006, 103, 11838-11843, 10.1073/pnas.0602615103.
  3. Dihua Shangguan; Zehui Charles Cao; Ying Li; Weihong Tan; Aptamers Evolved from Cultured Cancer Cells Reveal Molecular Differences of Cancer Cells in Patient Samples. Clinical Chemistry 2007, 53, 1153-1155, 10.1373/clinchem.2006.083246.
  4. Dihua Shangguan; Zehui Cao; Ling Meng; Prabodhika Mallikaratchy; Kwame Sefah; Hui Wang; Ying Li; Weihong Tan; Cell-Specific Aptamer Probes for Membrane Protein Elucidation in Cancer Cells. Journal of Proteome Research 2008, 7, 2133-2139, 10.1021/pr700894d.
  5. Estefanía Sicco; Jessica Baez; Manuel Ibarra; Marcelo Fernández; Pablo Cabral; María Moreno; Hugo Cerecetto; Victoria Calzada; Sgc8-c Aptamer as a Potential Theranostic Agent for Hemato-Oncological Malignancies. Cancer Biotherapy and Radiopharmaceuticals 2020, 35, 262-270, 10.1089/cbr.2019.3402.
  6. Prabodhika Mallikaratchy; Zhiwen Tang; Sefah Kwame; Ling Meng; Dihua Shangguan; Weihong Tan; Aptamer Directly Evolved from Live Cells Recognizes Membrane Bound Immunoglobin Heavy Mu Chain in Burkitt's Lymphoma Cells. Molecular & Cellular Proteomics 2007, 6, 2230-2238, 10.1074/mcp.m700026-mcp200.
  7. Baoyin Yuan; Xiaochun Jiang; Yuanyuan Chen; Qiuping Guo; Kemin Wang; Xiangxian Meng; Zhixiang Huang; Xiaohong Wen; Metastatic cancer cell and tissue-specific fluorescence imaging using a new DNA aptamer developed by Cell-SELEX. Talanta 2017, 170, 56-62, 10.1016/j.talanta.2017.03.094.
  8. Wan-Ming Li; Lin-Lin Zhou; Min Zheng; Jin Fang; Selection of Metastatic Breast Cancer Cell-Specific Aptamers for the Capture of CTCs with a Metastatic Phenotype by Cell-SELEX. Molecular Therapy - Nucleic Acids 2018, 12, 707-717, 10.1016/j.omtn.2018.07.008.
  9. Xilan Li; Weiyun Zhang; Lu Liu; Zhi Zhu; Gaoliang Ouyang; Yuan An; Chunyi Zhao; Chaoyong James Yang; In Vitro Selection of DNA Aptamers for Metastatic Breast Cancer Cell Recognition and Tissue Imaging. Analytical Chemistry 2014, 86, 6596-6603, 10.1021/ac501205q.
  10. Elina Zueva; Laila Illán Rubio; Frédéric Ducongé; Bertrand Tavitian; Metastasis-focused cell-based SELEX generates aptamers inhibiting cell migration and invasion. International Journal of Cancer 2010, 128, 797-804, 10.1002/ijc.25401.
  11. Liping Wang; Peipei Li; Xue Xiao; Jingying Li; Juan Li; Huang-Hao Yang; Weihong Tan; Generating lung-metastatic osteosarcoma targeting aptamers for in vivo and clinical tissue imaging. Talanta 2018, 188, 66-73, 10.1016/j.talanta.2018.05.011.
  12. Minlan Duan; Yuqian Long; Cai Yang; Xiaoqiu Wu; Yang Sun; Jianglin Li; Xiaoxiao Hu; Wei Lin; Dongmei Han; Yifan Zhao; et al.Jing LiuMao YeWeihong Tan Selection and characterization of DNA aptamer for metastatic prostate cancer recognition and tissue imaging. Oncotarget 2016, 7, 36436-36446, 10.18632/oncotarget.9262.
  13. S. Speransky; P. Serafini; Jimmy Caroli; S. Bicciato; M. E. Lippman; N. H. Bishopric; A novel RNA aptamer identifies plasma membrane ATP synthase beta subunit as an early marker and therapeutic target in aggressive cancer. Breast Cancer Research and Treatment 2019, 176, 271-289, 10.1007/s10549-019-05174-3.
  14. Fu-Bing Wang; Yuan Rong; Min Fang; Jing-Ping Yuan; Chun-Wei Peng; Shao-Ping Liu; Yan Li; Recognition and capture of metastatic hepatocellular carcinoma cells using aptamer-conjugated quantum dots and magnetic particles. Biomaterials 2013, 34, 3816-3827, 10.1016/j.biomaterials.2013.02.018.
  15. Hao Chen; Chun-Hui Yuan; Yi-Fei Yang; Chang-Qing Yin; Qing Guan; Fu-Bing Wang; Jian-Cheng Tu; Subtractive Cell-SELEX Selection of DNA Aptamers Binding Specifically and Selectively to Hepatocellular Carcinoma Cells with High Metastatic Potential. BioMed Research International 2016, 2016, 1-9, 10.1155/2016/5735869.
  16. Yuan Rong; Hao Chen; Xue-Feng Zhou; Chang-Qing Yin; Bi-Cheng Wang; Chun-Wei Peng; Shao-Ping Liu; Fu-Bing Wang; Identification of an aptamer through whole cell-SELEX for targeting high metastatic liver cancers. Oncotarget 2016, 7, 8282-8294, 10.18632/oncotarget.6988.
  17. Xilan Li; Yuan An; Jiang Jin; Zhi Zhu; Linlin Hao; Lu Liu; Yongquan Shi; Daiming Fan; Tianhai Ji; Chaoyong James Yang; et al. Evolution of DNA Aptamers through in Vitro Metastatic-Cell-Based Systematic Evolution of Ligands by Exponential Enrichment for Metastatic Cancer Recognition and Imaging. Analytical Chemistry 2015, 87, 4941-4948, 10.1021/acs.analchem.5b00637.
  18. Wan-Ming Li; Tao Bing; Jia-Yi Wei; Zhe-Zhou Chen; Di-Hua Shangguan; Jin Fang; Cell-SELEX-based selection of aptamers that recognize distinct targets on metastatic colorectal cancer cells. Biomaterials 2014, 35, 6998-7007, 10.1016/j.biomaterials.2014.04.112.
  19. Sorah Yoon; Brian Armstrong; Nagy Habib; John J. Rossi; Blind SELEX Approach Identifies RNA Aptamers That Regulate EMT and Inhibit Metastasis.. Molecular Cancer Research 2017, 15, 811-820, 10.1158/1541-7786.MCR-16-0462.
  20. Tannishtha Reya; Sean Morrison; Michael F. Clarke; Irving L. Weissman; Stem cells, cancer, and cancer stem cells. Nature 2001, 414, 105-111, 10.1038/35102167 35102167 [pii].
  21. Youngmi Kim; Qiulian Wu; Petra Hamerlik; Masahiro Hitomi; Andrew E. Sloan; Gene H. Barnett; Robert J. Weil; Patrick Leahy; Anita B. Hjelmeland; Jeremy N. Rich; et al. Aptamer Identification of Brain Tumor–Initiating Cells. Cancer Research 2013, 73, 4923-4936, 10.1158/0008-5472.can-12-4556.
  22. Qiaoyi Wu; Ningqin Lin; Tian Tian; Zhi Zhu; Liang Wu; Hongyao Wang; Dengliang Wang; Dezhi Kang; Ruijun Tian; Chaoyong Yang; et al. Evolution of Nucleic Acid Aptamers Capable of Specifically Targeting Glioma Stem Cells via Cell-SELEX. Analytical Chemistry 2019, 91, 8070-8077, 10.1021/acs.analchem.8b05941.
  23. Wang Chih-Hung; Chih-Hung Wang; Yu-Jui Che; Chien-Yu Fu; Hwan-You Chang; Kuan Wang; Gwo-Bin Lee; Screening of aptamers specific to colorectal cancer cells and stem cells by utilizing On-chip Cell-SELEX. Scientific Reports 2015, 5, 10326-10326, 10.1038/srep10326.
  24. Yoon-Jin Kim; Hee Seung Lee; Dawoon E. Jung; Jeong Mi Kim; Si Young Song; The DNA aptamer binds stemness-enriched cancer cells in pancreatic cancer. Journal of Molecular Recognition 2017, 30, e2591, 10.1002/jmr.2591.
  25. Kwame Sefah; Kyung-Mi Bae; Joseph A. Phillips; Dietmar W. Siemann; Zhen Su; Steve McClellan; Johannes Vieweg; Weihong Tan; Cell-based selection provides novel molecular probes for cancer stem cells. International Journal of Cancer 2013, 132, 2578-2588, 10.1002/ijc.27936.
  26. Hui Li; Juan Liu; Xiaojuan Xiao; Shuming Sun; Hui Zhang; Yibin Zhang; Weihua Zhou; Mridul Roy; Hong Liu; Mao Ye; et al.Zi WangFeng Liu-SmithJing Liu A Novel Aptamer LL4A Specifically Targets Vemurafenib-Resistant Melanoma through Binding to the CD63 Protein. Molecular Therapy - Nucleic Acids 2019, 18, 727-738, 10.1016/j.omtn.2019.10.005.
  27. Richard B. Warner; Abdo J. Najy; Young Suk Jung; Rafael Fridman; Seongho Kim; Hyeong-Reh Choi Kim; Establishment of Structure-Function Relationship of Tissue Inhibitor of Metalloproteinase-1 for Its Interaction with CD63: Implication for Cancer Therapy. Scientific Reports 2020, 10, 1-8, 10.1038/s41598-020-58964-x.
  28. Nan Zhang; Tao Bing; Luyao Shen; Le Feng; Xiangjun Liu; Dihua Shangguan; A DNA Aptameric Ligand of Human Transferrin Receptor Generated by Cell-SELEX. International Journal of Molecular Sciences 2021, 22, 8923, 10.3390/ijms22168923.
  29. Carla Esposito; Diana Passaro; Immacolata Longobardo; Gerolama Condorelli; Pina Marotta; Andrea Affuso; Vittorio De Franciscis; Laura Cerchia; A Neutralizing RNA Aptamer against EGFR Causes Selective Apoptotic Cell Death. PLOS ONE 2011, 6, e24071, 10.1371/journal.pone.0024071.
  30. Simona Camorani; Carla Esposito; Anna Rienzo; Silvia Catuogno; Margherita Iaboni; Gerolama Condorelli; Vittorio de Franciscis; Laura Cerchia; Inhibition of Receptor Signaling and of Glioblastoma-derived Tumor Growth by a Novel PDGFRβ Aptamer. Molecular Therapy 2014, 22, 828-841, 10.1038/mt.2013.300.
  31. Simona Camorani; Elvira Crescenzi; David Colecchia; Andrea Carpentieri; Angela Amoresano; Monica Fedele; Mario Chiariello; Laura Cerchia; Aptamer targeting EGFRvIII mutant hampers its constitutive autophosphorylation and affects migration, invasion and proliferation of glioblastoma cells. Oncotarget 2015, 6, 37570-37587, 10.18632/oncotarget.6066.
  32. Simona Camorani; Elvira Crescenzi; Matteo Gramanzini; Monica Fedele; Antonella Zannetti; Laura Cerchia; Aptamer-mediated impairment of EGFR-integrin αvβ3 complex inhibits vasculogenic mimicry and growth of triple-negative breast cancers. Scientific Reports 2017, 7, srep46659, 10.1038/srep46659.
  33. Simona Camorani; Billy Samuel Hill; Francesca Collina; Sara Gargiulo; Maria Napolitano; Monica Cantile; Maurizio Di Bonito; Gerardo Botti; Monica Fedele; Antonella Zannetti; et al.Laura Cerchia Targeted imaging and inhibition of triple-negative breast cancer metastases by a PDGFRβ aptamer. Theranostics 2018, 8, 5178-5199, 10.7150/thno.27798.
  34. Simona Camorani; Billy Samuel Hill; Raffaela Fontanella; Adelaide Greco; Matteo Gramanzini; Luigi Auletta; Sara Gargiulo; Sandra Albanese; Enrico Lucarelli; Laura Cerchia; et al.Antonella Zannetti Inhibition of Bone Marrow-Derived Mesenchymal Stem Cells Homing Towards Triple-Negative Breast Cancer Microenvironment Using an Anti-PDGFRβ Aptamer. Theranostics 2017, 7, 3595-3607, 10.7150/thno.18974.
  35. Simona Camorani; Margherita Passariello; Lisa Agnello; Silvia Esposito; Francesca Collina; Monica Cantile; Maurizio Di Bonito; Ilya V. Ulasov; Monica Fedele; Antonella Zannetti; et al.Claudia De LorenzoLaura Cerchia Aptamer targeted therapy potentiates immune checkpoint blockade in triple-negative breast cancer. Journal of Experimental & Clinical Cancer Research 2020, 39, 1-16, 10.1186/s13046-020-01694-9.
  36. Pooja Dua; Hye Suk Kang; Seung-Mo Hong; Ming Tsao; Soyoun Kim; Dong-Ki Lee; Alkaline Phosphatase ALPPL-2 Is a Novel Pancreatic Carcinoma-Associated Protein. Cancer Research 2013, 73, 1934-1945, 10.1158/0008-5472.can-12-3682.
  37. Wenting Jia; Caiping Ren; Lei Wang; Bin Zhu; Wei Jia; Menghui Gao; Fei Zeng; Liang Zeng; Xiaomeng Xia; Xiaobing Zhang; et al.Ting FuShasha LiCan DuXingjun JiangYuxiang ChenWeihong TanZilong ZhaoWeidong Liu CD109 is identified as a potential nasopharyngeal carcinoma biomarker using aptamer selected by cell-SELEX. Oncotarget 2016, 7, 55328-55342, 10.18632/oncotarget.10530.
  38. Pauliina Filppu; Jayendrakishore Tanjore Ramanathan; Kirsi J. Granberg; Erika Gucciardo; Hannu Haapasalo; Kaisa Lehti; Matti Nykter; Vadim Le Joncour; Pirjo Laakkonen; CD109-GP130 interaction drives glioblastoma stem cell plasticity and chemoresistance through STAT3 activity. JCI Insight 2021, 6, e141486, 10.1172/jci.insight.141486.
  39. Aline G. Souza; Karina Marangoni; Patrícia T. Fujimura; Patrícia Terra Alves; Márcio J. Silva; Victor Alexandre F Bastos; Luiz Goulart; Vivian Alonso Goulart; 3D Cell-SELEX: Development of RNA aptamers as molecular probes for PC-3 tumor cell line. Experimental Cell Research 2016, 341, 147-156, 10.1016/j.yexcr.2016.01.015.
  40. Frank Nelissen; Wenny Peeters; Timo Roelofs; Anika Nagelkerke; Paul Span; Hans Heus; Improving Breast Cancer Treatment Specificity Using Aptamers Obtained by 3D Cell-SELEX. Pharmaceuticals 2021, 14, 349, 10.3390/ph14040349.
  41. Yue Zhao; Xiao Fu; Jose I. Lopez; Andrew Rowan; Lewis Au; Annika Fendler; Steve Hazell; Hang Xu; Stuart Horswell; Scott T. C. Shepherd; et al.Lavinia SpainFiona ByrneGordon StampTim O’BrienDavid NicolMarcellus AugustineAshish ChandraSarah RudmanAntonia TonchevaLisa PickeringErik SahaiJames LarkinPaul A. BatesCharles SwantonSamra TurajlicBen ChallacombeSimon ChowdhuryWilliam DrakeArchana FernandoNicos FotiadisAndrew FurnessEmine HatipogluKaren Harrison-PhippsPeter HillCatherine HorsfieldTeresa MarafiotiJonathon OlsburghAlexander PolsonSergio QuezadaMary VariaHema VermaKevin LitchfieldTRACERx Renal Consortium Selection of metastasis competent subclones in the tumour interior. Nature Ecology & Evolution 2021, 5, 1033-1045, 10.1038/s41559-021-01456-6.
  42. Andriy Marusyk; Vanessa Almendro; Kornelia Polyak; Intra-tumour heterogeneity: a looking glass for cancer?. Nature Cancer 2012, 12, 323-334, 10.1038/nrc3261.
  43. David N Louis; Arie Perry; Pieter Wesseling; Daniel J Brat; Ian A Cree; Dominique Figarella-Branger; Cynthia Hawkins; H K Ng; Stefan M Pfister; Guido Reifenberger; et al.Riccardo SoffiettiAndreas von DeimlingDavid W Ellison The 2021 WHO Classification of Tumors of the Central Nervous System: a summary. Neuro-Oncology 2021, 23, 1231-1251, 10.1093/neuonc/noab106.
  44. Aline Becker; Blake Sells; S. Haque; Arnab Chakravarti; Tumor Heterogeneity in Glioblastomas: From Light Microscopy to Molecular Pathology. Cancers 2021, 13, 761, 10.3390/cancers13040761.
  45. Ningqin Lin; Liang Wu; Xing Xu; Qiaoyi Wu; Yuzhe Wang; Haicong Shen; Yanling Song; Hongyao Wang; Zhi Zhu; Dezhi Kang; et al.Chaoyong Yang Aptamer Generated by Cell-SELEX for Specific Targeting of Human Glioma Cells. ACS Applied Materials & Interfaces 2020, 13, 9306-9315, 10.1021/acsami.0c11878.
  46. Qiaoyi Wu; Yuzhe Wang; Hongyao Wang; Liang Wu; Huimin Zhang; Yanling Song; Zhi Zhu; Dezhi Kang; Chaoyong Yang; DNA aptamers from whole-cell SELEX as new diagnostic agents against glioblastoma multiforme cells. The Analyst 2018, 143, 2267-2275, 10.1039/c8an00271a.
  47. Rodolfo Bortolozo Serafim; Patrick da Silva; Cibele Cardoso; Luis Fernando Macedo Di Cristofaro; Renato Petitto Netto; Rodrigo de Almeida; Geovana Navegante; Camila Baldin Storti; Juliana Ferreira de Sousa; Felipe Canto de Souza; et al.Rodrigo PanepucciCristiano Gallina MoreiraLarissa Siqueira PennaWilson Araujo Jr SilvaValeria Valente Expression Profiling of Glioblastoma Cell Lines Reveals Novel Extracellular Matrix-Receptor Genes Correlated With the Responsiveness of Glioma Patients to Ionizing Radiation. Frontiers in Oncology 2021, 11, 668090, 10.3389/fonc.2021.668090.
  48. Qiaoyi Wu; Liang Wu; Yuzhe Wang; Zhi Zhu; Yanling Song; Yuyu Tan; Xing-Fu Wang; Jiuxing Li; Dezhi Kang; Chaoyong James Yang; et al. Evolution of DNA aptamers for malignant brain tumor gliosarcoma cell recognition and clinical tissue imaging. Biosens Bioelectron. 2016, 80, 1-8, 10.1016/j.bios.2016.01.031.
  49. Simona Camorani; Monica Fedele; Antonella Zannetti; Laura Cerchia; TNBC Challenge: Oligonucleotide Aptamers for New Imaging and Therapy Modalities. Pharmaceuticals 2018, 11, 123, 10.3390/ph11040123.
  50. Mei Liu; Zhifei Wang; Ting Tan; Zhongsi Chen; Xianbo Mou; Xiaocheng Yu; Yan Deng; Guangming Lu; Nongyue He; An Aptamer-Based Probe for Molecular Subtyping of Breast Cancer. Theranostics 2018, 8, 5772-5783, 10.7150/thno.28949.
  51. Brian D. Lehmann; Joshua A. Bauer; Xi Chen; Melinda E. Sanders; A. Bapsi Chakravarthy; Yu Shyr; Jennifer A. Pietenpol; Identification of human triple-negative breast cancer subtypes and preclinical models for selection of targeted therapies. Journal of Clinical Investigation 2011, 121, 2750-2767, 10.1172/jci45014.
  52. Brian D. Lehmann; Bojana Jovanović; Xi Chen; Monica Valeria Estrada; Kimberly N. Johnson; Yu Shyr; Harold L. Moses; Melinda E. Sanders; Jennifer A. Pietenpol; Refinement of Triple-Negative Breast Cancer Molecular Subtypes: Implications for Neoadjuvant Chemotherapy Selection. PLoS ONE 2016, 11, e0157368, 10.1371/journal.pone.0157368.
  53. Simona Camorani; Ilaria Granata; Francesca Collina; Francesco Leonetti; Monica Cantile; Gerardo Botti; Monica Fedele; Mario Rosario Guarracino; Laura Cerchia; Novel Aptamers Selected on Living Cells for Specific Recognition of Triple-Negative Breast Cancer. iScience 2020, 23, 100979, 10.1016/j.isci.2020.100979.
  54. Natalia Anahi Juiz; Juan Iovanna; Nelson Dusetti; Pancreatic Cancer Heterogeneity Can Be Explained Beyond the Genome. Frontiers in Oncology 2019, 9, 246, 10.3389/fonc.2019.00246.
  55. Sorah Yoon; Haiqing Li; Loren Quintanar; Brian Armstrong; John J Rossi; Uncovering Differently Expressed Markers and Heterogeneity on Human Pancreatic Cancer. Translational Oncology 2020, 13, 100749, 10.1016/j.tranon.2020.100749.
  56. Sorah Yoon; Kai-Wen Huang; Vikash Reebye; Paul Mintz; Yu-Wen Tien; Hong-Shiee Lai; Pål Sætrom; Isabella Reccia; Piotr Swiderski; Brian Armstrong; et al.Agnieszka JozwiakDuncan SpaldingLong JiaoNagy HabibJohn J Rossi Targeted Delivery of C/EBPα -saRNA by Pancreatic Ductal Adenocarcinoma-specific RNA Aptamers Inhibits Tumor Growth In Vivo. Molecular Therapy 2016, 24, 1106-1116, 10.1038/mt.2016.60.
  57. Daniela F Quail; Johanna A Joyce; Microenvironmental regulation of tumor progression and metastasis. Nature Medicine 2013, 19, 1423-1437, 10.1038/nm.3394.
  58. Meilyn Sylvestre; Christopher P. Saxby; Nataly Kacherovsky; Heather Gustafson; Stephen J. Salipante; Suzie H. Pun; Identification of a DNA Aptamer That Binds to Human Monocytes and Macrophages. Bioconjugate Chemistry 2020, 31, 1899-1907, 10.1021/acs.bioconjchem.0c00247.
More
Information
Subjects: Cell Biology
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: 291
Revision: 1 time (View History)
Update Date: 29 Mar 2022
1000/1000
Video Production Service