Open Access

Aptamer‑based therapy for targeting key mediators of cancer metastasis (Review)

  • Authors:
    • Yahya Alhamhoom
    • Homood M. As Sobeai
    • Sary Alsanea
    • Ali Alhoshani
  • View Affiliations

  • Published online on: April 14, 2022     https://doi.org/10.3892/ijo.2022.5355
  • Article Number: 65
  • Copyright: © Alhamhoom et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Cancer‑related deaths remain a challenging and devastating obstacle to defeat despite the tremendous advances in cancer treatment. Cancer metastasis is the major cause of these cancer‑related deaths. Metastasis involves sequential steps during cancer cells' journey to a new site. These steps are coordinately regulated by specific intracellular regulators and cellular interactions between the cancer cells and the supporting microenvironment of the different organs. The development of aptamer‑based therapeutics is a promising strategy to fight cancer metastasis as it holds potential advantages. Oligonucleotide and peptide aptamers are short sequences of single‑stranded nucleic acids or amino acids, respectively, that target proteins, genetic materials, and cells. Antimetastatic aptamer‑based therapeutics exert their pharmacological effect by direct interaction with the signaling pathways inside the cancer cells or the communications between cancer cells and the tumor microenvironment. In addition, aptamers have been utilized as a guiding ligand to deliver a therapeutic moiety to cancer cells or the supporting microenvironment. The selected aptamer possesses high specificity since it is designed to recognize and interact with its target. This review summarizes recent advances in the development of aptamer‑based therapeutics targeting mediators of cancer metastasis. In addition, potential opportunities are discussed to inspire researchers in the field to develop novel aptamer‑based antimetastatic treatments.

1. Introduction

Cancer consists of more than 200 types with incomplete knowledge of the origin, tumorigenesis, and progression (1-3). According to the most recent statistics, approximately 19.3 million cases of new patients have been diagnosed with cancer, and a fatality of approximately 10 million (4). The majority of these massive numbers of deaths are caused by cancer metastasis (5). Cancer metastasis involves the process of tumor cells spreading from a primary tumor mass to different sites through blood and lymphatic vessels. It involves a series of events known as the cancer metastasis cascade (Fig. 1). First, the epithelial cancer cells in primary tumors invade the extracellular matrix (ECM) and stromal cell layers within the site and enter into the lumina of the blood vessels. After that, cancer cells deal with several unfavorable conditions during their transport in the circulation until they arrive at specific organs. Next, cancer cells extravasate into the parenchyma of these organs, survive in these new microenvironments and initiate micrometastases. Finally, cancer cells restart their proliferative ability at the metastatic sites, thereby generating neoplastic growths known as 'metastatic colonization' (6).

During the last few decades, cumulative scientific discoveries that have been made in the field of molecular and cellular oncology have transformed clinical practice. For instance, the precise detection of cancer at an earlier stage and tailoring a therapeutic approach toward tumor type-specific intervention have been improved significantly (7). Molecular phenotypes associated with cancer metastasis have been extensively studied which has led to identifying potential metastasis-associated targets. The design of molecular-targeted therapies that specifically interact with those targets, whether they are components of a specific intracellular pathway or cell-cell communication signaling, have demonstrated potential benefits by overcoming systemic toxicities associated with traditional treatments such as chemotherapy and radiation therapy as well as improving the pharmacokinetics and pharmacodynamics of these traditional treatments (8,9).

One of the most recently developed molecular-targeted therapy is short sequences of single-stranded nucleic acids (ssDNA or RNA) or amino acids which are known as oligonucleotide or peptide aptamers, respectively (Fig. 2). Aptamers mimic antibodies in which they possess high selectivity when they bind with the selected targets. The first observation of the binding capability of aptamers was reported when researchers found that a subpopulation of isolated RNA molecules was able to bind specific ligands. An in vitro technique called SELEX (Systemic Evolution of Ligands by Exponential Enrichment) was introduced as a procedure to generate these oligonucleotide aptamers (10,11). The three-dimensional structure of a short sequence (20-100 bases long) RNA or ssDNA gives aptamers the capability to interact specifically with particular ligands with selectivity and affinity similar to those of antibodies. However, aptamers are more favorable compared to antibodies due to their cheap and rapid synthesis (12,13). A few years after oligonucleotide aptamers had been introduced, a research study demonstrated that designing a short peptide (5 to 20 amino acids long) then embedding it in a protein scaffold generated a protein with high specificity to a selected target. Therefore, a new concept launched for this type of aptamer is called peptide aptamer (14-16). To date, peptide and oligonucleotide aptamers have been developed and exploited for different diagnostic and therapeutic purposes. Aptamers have been employed for diagnostic applications in cancer detection and imaging (17) and infectious disease (18). For therapeutic purposes, aptamers have been utilized to treat different diseases such as cancer (17,19), infectious diseases (18,20), coagulation disorders (21), diabetic nephropathy (22) and ocular vascular diseases (23). This review aims to summarize recent advances in utilizing aptamers as a treatment strategy against cancer metastasis specifically and highlight key mediators and molecular factors involved during different metastasis stages. Moreover, potential opportunities to improve aptamer-based antimetastatic therapeutics are discussed. We used Web of Science and PubMed databases to retrieve the most recent information in this review.

2. Cancer metastasis cascade

Local invasion

Local invasion is the access of the cancer cells that were contained within the primary tumor into the surrounding stroma, and then into the adjoining normal parenchymal tissue. To enter the stroma, the carcinoma cells modify the ECM, which has a significant function in arranging epithelial tissues. The integrins (transmembrane proteins) bind to the ECM and form the integrin-mediated cell-matrix adhesions (24). This binding initiates several pathways leading to signal transduction events within the carcinoma cells that leads to disturbances in cell polarity, proliferation, invasiveness, and survival (25).

A majority of the carcinomas have the ability to invade as cohesive multicellular units known as a collective invasion. However, a single cancer cell may attack through two different mechanisms: integrin-dependent (mesenchymal invasion) and integrin-independent, Rho/ROCK-dependent (amoeboid invasion) pathways (26). It may be noted that the cancer cells may interconvert between these mechanisms due to the change in the microenvironment (27). While the patterns of cancer cell invasion are classified as collective and individual cell migration (28), the individual cell invasion mechanism is not compatible with an important element of epithelial tissue organization, specifically the E-cadherin-mediated intercellular junctions that lead to the development of the epithelial cell sheets and remain associated with the surrounding epithelial cells. To attenuate tight junctions and cellular polarity, carcinoma cells may undergo epithelial-mesenchymal transition (EMT). EMT is vital for different facets of normal embryonic morphogenesis which ultimately help to liberate cancer cells from epithelial cell sheets (29). Several transcription factors, namely Slug, Snail, Twist, zinc finger E-box-binding homeobox 1 (ZEB1), and ZEB2 play significant roles in EMT. They trigger mesenchymal entry by downregulating the expression of E-cadherin and other epithelial markers such as cytokeratin, zonula occludens-1 (ZO-1), laminin-1, and α1(IV) collagen (29). In addition, other regulatory non-coding genes, such as microRNAs (miRNAs/miRs), govern EMT. The miR-200 family regulates EMT programs by post-transcriptionally suppressing the expression of ZEB1 and ZEB2, while on the other hand, ZEB1 and ZEB2 inhibit the transcription of the miR-200 family. Such a relationship establishes a double-negative-feedback loop that operates as a bistable switch controlling the fate of cancer cells to go in either the mesenchymal or epithelial state (29). The debilitation of the ECM is further aggravated by active proteolysis activated by the matrix metalloproteinases (MMPs), which promotes the invasion of carcinoma cells to the stromal compartment. As the stroma becomes more chronically inflamed upon tumor progression, cancer cells are challenged by fibroblasts, endothelial cells, adipocytes, mesenchymal stem cells from the bone marrow, macrophages, and other immune cells (30). These stromal cells further influence the aggressiveness of carcinoma cells via different types of heterotypic signaling. For example, the invasiveness of breast cancer is stimulated by IL-6 secreted by the adipocytes (31). Similarly, it has also been demonstrated that the cathepsin protease activity in tumor-associated macrophages is activated by the secretion of IL-4, which fuels the invasiveness of carcinoma cells (32).

Intravasation

Intravasation occurs when the carcinoma cells enter into the lumina of lymphatic or blood vessels. This process is normally observed in human tumors and signifies a vital prognostic marker for its progression; dissemination through blood vessels is the key mechanism for the spread of metastatic carcinoma cells (33). Intravasation is enhanced by molecular variations that increase the capacity of carcinoma cells to penetrate the microvessels that are composed of pericyte and endothelial cells. Intravasation is influenced by the vascular endothelial growth factors (VEGFs) secreted by the tumor cells which enhance the generation of new blood vessels within their local microenvironment through neoangiogenesis. In comparison to the normal blood vessels, the neovasculature developed by carcinoma cells is prone to leakiness and is subjected to continuous reconfiguration (34). Furthermore, it has been reported earlier that cyclooxygenase-2 (COX-2), epiregulin (EREG), MMP-1, and MMP-2 synergistically promote breast carcinoma intravasation due to their capacity to stimulate neoangiogenesis (35).

Survival in the circulation

After the intravasation has been achieved, the carcinoma cells are widely transported through the systemic circulation, known as circulating tumor cells (CTCs). Before reaching other organs, the CTCs deal with different types of stresses for its survival, such as the absence of the integrin-dependent adhesion to ECM components that is required for cell survival. Consequently, the epithelial cells undergo anoikis, which is a form of apoptosis that is activated by the loss of attachment to the substratum (36). However, the tyrosine kinase TrkB has been observed to suppress anoikis (37). Additionally, the tumor cells also face potential damage due to the hemodynamic shear forces and the innate immune system, specifically natural killer cells. These challenges have been observed to be evaded simultaneously through the formation of large emboli through the interactions with blood platelets (regulated by the tissue factor and/or L- and P-selectins by the carcinoma cells) (30). In this way, the platelet-coated tumor cells escape immune detection until they are arrested at distant organ sites.

Arrest and extravasation at a distant organ site

Despite the capacity of the CTCs to spread to a wide range of distant organ sites, it has been previously observed that specific carcinoma types metastasize to specific organs which leads to the proposal of the 'seed and soil hypothesis' (6). Seed and soil hypothesis suggests that cancer cells (seed) disseminated from the primary tumor spread to all organs, but only specific microenvironments (fertile soil) that support metastatic tumor formation in specific organs. Once the cancer cells reach the suitable organs, the organ microenvironment supports the attachment and seeding of cancer cells in the specific organ. This is because some carcinoma cells depend on specific adhesive interactions in particular tissues that assist their trapping. For example, the generation of metadherin in breast cancer cells initiates the spread of carcinoma cells to the lungs through increased binding to the pulmonary vasculature (38). In addition, it has been observed that a proinflammatory environment in the liver causes Kupffer cells to secrete chemokines which upregulate vascular adhesion receptors and ultimately enable adhesion of the circulating colorectal and lung carcinoma cells to the liver microvasculature (39). Then, the trapped cancer cells grow inside the vasculature and develop a microcolony that ultimately penetrates through the surrounding vessels, resulting in the straight contact of the tumor cells with the tissue parenchyma (40). In another way, the carcinoma cells might rupture from vessel lumina into the tissue parenchyma by going through the endothelial cell and pericyte layers that separate the vessel lumina from the stromal microenvironment. This process is known as extravasation. Further, the physical barriers to extravasation may be breached due to the ability of the primary tumors to secrete factors that cause an imbalance in the microenvironments and induce vascular hyperpermeability. For example, the factors protein angiopoietin-like-4 (Angptl4), epiregulin (EREG), COX-2, MMP-1, and MMP-2, disrupt pulmonary vascular endothelial cell-cell junctions in order to facilitate the extravasation of breast carcinoma cells in the lungs (35,41).

Micrometastasis formation

The formation of micrometastases is initiated following the survival of the extravasated cancer cells in the parenchyma of distant tissues. It is worth mentioning that several factors contribute to this stage of cancer metastasis including the type of stromal cells, ECM constituents, growth factors, and cytokines. In the beginning, cancer cells establish a 'premetastatic niche' to ascertain the compatibility in the foreign microenvironment (42). For this to happen, the primary tumors liberate systemic signals consisting of lysyl oxidase (LOX) (43), that stimulates organ-specific upregulation of fibronectin from fibroblasts, which in turn, activates VEGF receptor 1-positive (VEGFR1+) hematopoietic progenitor cells from the bone marrow to these prospective sites of metastasis through homing interactions between the deposited fibronectin and its cognate receptor, integrin a4b1that is expressed by the hematopoietic progenitor cells. The hematopoietic progenitor cells secrete MMP-9 that changes the immediate microenvironments at these loci. The MMP-9 activation triggers the release of various integrins and discharge of molecules from the ECM, such as the carcinoma cell chemoattractant stromal cell-derived factor 1 (SDF-1) (42). All of these changes convert the distant microenvironments into growth sites for the disseminated tumor cells.

Metastatic colonization

Even after the successful survival of the tumor cells in the new microenvironment, it is still not ensured that they can grow and form metastases, the process known as metastatic colonization. Instead, it has been observed that a large number of tumor cells either slowly perish over a period of time or are sustained as microcolonies during long-term dormancy, retaining the overall cell number (44). These dormant microcolonies may continue to remain dormant because of incompatibilities with the foreign microenvironments that surround them (44), such as in mammary carcinoma cells, where the focal adhesion kinase (FAK), integrin b1, and Src pathways are unable to engage within distant tissues (45-47). However, they may escape dormancy to initiate active proliferation cell-nonautonomous mechanisms that are stimulated by osteopontin (OPN) or SDF-1 (48,49). Secondly, the dormant microcolonies may proliferate continuously but the overall number may remain the same due to the high apoptotic rate. For example, the prostate tumor cell secretes prosaposin (Psap) that inhibits metastatic colonization by upregulating the anti-angiogenic factor thrombospondin-1 in stromal cells (50). Metastatic colonization is also dependent on another attribute known as 'tumor-initiating cells' (TICs), which possess such an extensive self-renewal capacity to achieve malignant growth. The entry into the TIC state is promoted by miRNAs and the EMT-promoting transcription factors, such as Snail, Twist, and ZEB1, as already discussed (51).

3. Aptamers as targeted therapeutics

Approaches of targeted therapy

Exploiting aptamers as a new generation of therapeutics has attracted the attention of the scientific community due to various advantages that are offered by aptamers. An individual aptamer possesses high specificity since it depends on its three-dimensional conformation to bind to its specific target. The aptamer molecule binds to the target through a hydrogen bond, electrostatic interaction, van der Waals, hydrophobic interactions, or stacking interactions (14,52). Moreover, aptamers can be chemically synthesized with flexible customization providing an opportunity to improve pharmacokinetics and meet a wide range of applications needed (53,54). They can bind diverse targets, ranging from small molecules, proteins to viruses and cells (55-58). From a therapeutic perspective, aptamers can be used to exert pharmacological action by themselves as agonists (target activation), antagonists (target inhibition), or to act as ligands for targeted delivery of therapeutics (12) (Fig. 2). Most of the developed aptamers fall into the former category. Pegaptanib (Macugen®) is the first FDA-approved aptamer and works by antagonizing the action of vascular endothelial growth factor (VEGF) (23). For targeted delivery purposes, aptamers can be constructed to deliver a wide range of diagnostic or therapeutic moieties such as fluorescent materials, radioisotopes, cytotoxic drugs, RNA oligonucleotides, and nanoparticles (59-63). In addition, a multifunctional, aptamer-based theranostic conjugate can be designed by coupling a diagnostic marker and a therapeutic moiety with the aptamer for simultaneous diagnosis and treatment (64-66).

Pharmacokinetic considerations when designing aptamers

Once an aptamer enters the body, it is susceptible to various factors that minimize or prevent its therapeutic action. It is vital to implement strategies to overcome obstacles and produce aptamers suitable for clinical settings. Stability in the circulation and tumor microenvironment, rapid excretion by the kidney, and delivery to intracellular targets affect the therapeutic actions of the aptamers. First, oligonucleotide aptamers can undergo enzymatic degradation by nucleases present in the circulation and tumor microenvironment. Strategies to produce nuclease-resistant aptamers include chemical modification of the structure by attaching a functional group to one of the following positions: 2′position of monosaccharide and 3′ or 5′termini of the aptamer (67,68). Pegaptanib aptamer, an approved therapeutic aptamer against age-related macular degeneration, is an example of such a modification. Another strategy is synthesizing an RNA or DNA backbone composed of L-ribose or L-deoxyribose for RNA and ssDNA aptamers, respectively. Spiegelmer is the name used to refer to these types of aptamers. This strategy relies on the fact that nucleases degrade D-oligonucleotide while the L-oligonucleotide is resistant to degradation (68,69). A second challenge facing an aptamer inside the body is the rapid excretion of the aptamer by the kidneys. The low molecular weight of the aptamer is responsible for the short-time presence of an aptamer in the circulation. An effective approach to overcome this obstacle is by conjugating the aptamer with cholesterol or high molecular weight moieties, such as proteins and polyethylene glycol (70-72). Aptamer-PEG conjugate possesses an enhanced half-life in the circulation compared to the unconjugated form of the aptamer (72). Crossing the cell membrane is another obstacle that can prevent an aptamer from exerting its pharmacological action. While the majority of targeted aptamers can easily interact with targets that are present in the circulation or on the surface of the cancer cells, some aptamers face difficulties crossing the cell membrane to reach their intercellular target. To overcome this obstacle, several strategies have been explored. The first strategy is the use of cell-internalization SELEX to develop a therapeutic cell-internalizing aptamer which has the capability to bind its target on the cell membrane and subsequently internalize to exert its action (73-75). The second strategy relies on coupling the therapeutic aptamer to protein transduction domains (PTDs), also known as cell-penetrating peptide. PTDs can cause the internalization of the therapeutic aptamer (76-78). The third strategy depends on the cellular uptake of a vector containing the DNA sequence of the therapeutic aptamer which is subsequently expressed intracellularly. The expressed aptamer is called an intramer. After cellular uptake of the vector by the targeted cell, intramer expression takes place inside the cells which makes the intramer available to exert its action (79,80).

4. Targeting key mediators of metastasis by aptamer-based therapeutics

Targeting cancer stem cells (CSCs)

A small population of cancer cells inside the tumor mass, known as cancer stem cells (CSCs), have been found to possess the capability of self-renewal and generation of cancer progeny cells (81). These cells defeat the process of anoikis, a type of programmed cell death that is triggered when the cells are removed from the surrounding ECM. In addition, research has found that these CSCs are resistant to therapeutic drugs since there is an upregulation in the expression of the ATP binding cassette transporter which results in drug efflux. In vitro and in vivo studies show that CSCs play a role in the formation of metastatic nodules (81-83). For example, it has been reported that the pluripotent genes octamer-binding transcription factor 4 (OCT4) and NANOG force-expressed in lung adenocarcinoma (LAC) increase the tumorigenic and metastatic capability of the cells since they induce EMT through the Slug protein. In addition to the increase in the number of CD133+ cells, the new cells have increased sphere-forming ability, drug resistance, and migration (84). In addition, CD24−/low breast cancer stem cells were found to have an increased capability for the formation of the tumor as compared to CD24+ breast cancer cells. Several important pathways are engaged in the CSC self-renewal process, such as the Wnt/β-catenin and NOTCH pathways. Further, the stemness of CSCs is maintained through the hepatocyte growth factor (HGF), and the transcription factors, OCT4, NANOG, SOX2, and BMI-1 (85-89). OCT4 expression has been reported to induce dysplasia and expansion of progenitor cells in the intestines (90). Furthermore, SRY-box transcription factor 2 (SOX2) maintains the vital signaling cascades for tumorigenesis. BMI1 proto-oncogene, polycomb ring finger (BMI-1) has a vital role in the self-renewal of normal stem cells. It has been observed that the knockdown of BMI-1 expression in CD133+ laryngeal cancer cells led to the restriction of cell growth, colonization, cell invasion in vitro, and tumorigenesis in vivo (91). It has been earlier demonstrated that the Y-box binding protein 1 could augment the stemness characteristics of human hepatoma cell lines (92,93). Moreover, the KRAS signal c-MYC axis supports the advancement of stem cell characteristics in pancreatic cancer (94).

The selective targeting capability of aptamers has been exploited to deliver cytotoxic drugs or nanoparticles carrying different types of gene therapy or small-molecule drugs to CSCs (Fig. 3). These therapeutic aptamers selectively target proteins overexpressed on the surface of the cell membrane of CSCs such as, but not limited to, human epidermal growth factor receptor 2 (HER2), CD133, CD44, CD20, and EpCAM receptors. In addition, CSC-targeted aptamers were developed against different types of cancer (95-100). A multifunctional nanoparticle decorated with RNA aptamer that can specifically target the HER2 receptor overexpressed on the surface of human breast cancer cells was examined in vitro and in vivo. The HER2 aptamer was able to deliver the nanoparticle carrying two different siRNAs against mediator complex subunit 1 (MED1) to HER2-overexpressing breast cancer cells and inhibit metastatic behavior of the cells in vitro. An in vivo study demonstrated that the targeted nanoparticle eliminated breast cancer metastatic nodules in the lung and significantly downregulated the expression levels of proteins involved in cancer metastasis such as c-Myc, MMP-9, Trefoil factor 1 (TFF-1), and cyclin D1. Most importantly, the examination of the stem cell population showed a significant decline and complete depletion of CD44+/CD24−/low stem cells after monotherapy and combination therapy with tamoxifen, respectively (98). Moreover, RNA aptamers targeting CD133 receptors that present on the surface of breast cancer cell stem cells were used to deliver nanoparticles carrying anti-microRNA to inhibit microRNA-21. An in vitro study demonstrated selective nanoparticle uptake by breast cancer stem cells compared to a control cell line with a loss of metastatic capability of the stem cells (96). In addition, functional nanoparticles decorated with two different aptamers against CD44 and transmembrane glycoprotein mucin 1 antigen (MUC1) were able to simultaneously deliver doxorubicin to CSCs and cancer cells, respectively. Dual-aptamer targeting nanoparticles inhibited breast cancer metastasis to the lung in an animal model of breast cancer metastasis (100).

Targeting circulating tumor cells (CTCs)

Following the progression of the disease in the primary organ, the cancer cells leave the organ, survive in the blood circulation, and metastasize in an organ-specific manner. This process was first reported by Stephen Paget (101). Research evidence has demonstrated the vital role of CTCs in the development of metastasis. Poor prognosis is associated with the increased numbers of CTCs in the circulation. This makes targeting CTCs a novel strategy to disrupt cancer metastasis cascade (102). Comprehensive knowledge established concerning molecular changes such as overexpression of adhesion molecules that support the attachment of CTCs to the tissues of the distant organs led scientists to develop therapeutics that block or prevent CTCs from attaching to the new organ. A clinical study examining the impact of using anti-EpCAM monoclonal antibodies, which target EpCAM, which is overexpressed in CTCs, on patients with metastatic colorectal carcinoma was conducted. The study demonstrated a significant prolongation of patient survival (103). Another treatment strategy that showed prolonged survival of an animal model of metastatic ovarian cancer involved utilizing nanotechnology and a magnetic field to isolate CTCs (104).

The development of aptamer-based therapeutics that target receptors that are involved in CTC attachment has shown effective prevention of cancer metastasis. DNA aptamer that displayed the capability to selectively target carcinoembryonic antigen (CEA), which plays a critical role in CTC adhesion and implantation, has been developed. Researchers mixed the anti-adhesive DNA aptamer with CEA-overexpressing colonic carcinoma cells before they injected these pretreated cells intraperitoneally in a mouse model. The in vivo study showed a significant decrease in the numbers and volumes of metastatic nodules compared to the control group (105,106). Another study developed a DNA aptamer against sialyl Lewis X (sLex), which helps cancer cells to bind endothelial-selectin (E-selectin) and metastasize to distant organs. In vitro study demonstrated that treating metastatic hepatocellular carcinoma cells (HepG2) with the DNA aptamer against sLex significantly inhibited adhesion and migration of HepG2 cells (107,108).

Targeting tumor-secreted extracellular vesicles

Extracellular vesicles (EVs) are cell-derived, cytosol-containing vesicles that are involved in cell communications with the surrounding environment. They have been categorized based on the size into three types: exosomes (30-100 nm diameter), microvesicles (MVs) (100-1000 nm diameter), and larger vesicles known as oncosomes (1-10 µm diameter) (109-112). It has been found that EVs play a critical role in supporting metastasis. Tumor-secreted EVs contain thousands of active constituents such as lipids, proteins, and genetic materials for intercellular communication (113-116), which act as messengers in local and distant microenvironments (117-119). RNAs function as the chief bioactive factor of tumor cell-derived EVs, together with other non-coding RNAs namely microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (120-122). These non-coding RNAs delivered by exosomes to recipient cells can control the expression of many genes to support oncogenic reprogramming of malignant cells, tumor growth, local invasion, and premetastatic or metastatic niches formation (123-126). For example, it has also been reported that exosomes can destroy the blood-brain barrier by the action of miR-181c that increases central nervous system (CNS) metastasis (127). Furthermore, the breast cancer cell-derived exosomes were found to utilize miR-122 to create a metastatic niche in the brain and enhance disease progression by downregulating glucose uptake in non-tumor cells and rerouting the available nutrients for cancer cells themselves (128). Moreover, the exosomes also adversely affect the immune function leading to an immunosuppressed phenotype and facilitating tumor progression (129). It has been observed that the c-Myc mRNA in the recipient microglia and macrophages was decreased due to the uptake of glioblastoma-derived exosomes by microglia/macrophages that led to an immunosuppressive phenotype mediated by the transport of miR-21 and miR-451 (130). It is worth mentioning that EVs can be released from stroma cells and support tumor metastasis. For example, exosomes released by astrocytes were found to downregulate phosphatase and tensin homolog (PTEN), a tumor-suppressor gene, in brain tumor cells leading to elevated oncogenicity (131).

The growing body of evidence regarding the crucial role of EVs in the development of cancer metastasis has attracted some researchers to identify aptamers that target EVs and seek to develop aptamer-based therapeutics to treat cancer metastasis. Recently, a novel modification of SELEX technology, which has been called Exo-SELEX, has been established to identify aptamers that specifically bind cell-derived exosomes. Using the most aggressive subtypes of breast cancer, triple-negative and HER2+, Exo-SELEX helped researchers to identify novel Ex-50.T aptamer. In addition, in vitro assessment of the therapeutic activity of EX-50.T was carried out using MDA-MB-231 and MCF-7 cell lines. The study relied on the fact that treating MCF-7 cells with exosomes derived from the highly metastatic MDA-MB-231 cells stimulates MCF-7 migration. The results demonstrated that MCF-7 cell migration was significantly inhibited when treating MCF-7 cells with exosomes that were pre-incubated with Ex-50.T (132). Another approach for targeting cancer cell-derived EVs is with the help of nanotechnology. Researchers have exploited the well-known fact regarding the liver uptake of mesoporous silica nanoparticles and the subsequent elimination of the nanoparticles into the small intestine to develop nanoparticles that specifically bind and eliminate circulating EVs from the blood. Mesoporous silica nanoparticles decorated with an aptamer that specifically target epidermal growth factor receptor (EGFR-targeting aptamer) that presents on the surface of EVs were developed. In vivo study to monitor the biodistribution of injected, labeled exosomes derived from metastatic lung cancer showed that the aptamer-targeted nanoparticles were able to significantly increase the accumulation of labeled exosomes to the liver and consecutively to the small intestine. In addition, aptamer-targeted nanoparticles inhibited the pulmonary metastasis formation in a subcutaneous murine tumor model (133).

Targeting tumor-associated immune cells

Immune cells can play dual opposite roles in cancer. Immune cells can recognize and kill immunogenic cancer cells and inhibit tumor growth. Conversely, they can promote tumor growth by inducing the formation of new blood vessels, known as angiogenesis, and facilitating cancer metastasis (134-136). Intercellular communication between cancer cells, immune cells, and other stroma cells in the tumor microenvironment is responsible for this observed plasticity of immune cell function. Myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs) recruited to the primary and secondary tumor sites contribute to the immunosuppressive environment. They are recruited in response to numerous growth factors, chemokines, and cytokines that are secreted by the cancer cells and other stromal cells (137-141). Through different mechanisms, MDSCs and Tregs negatively affect the antitumor activity of NK cells and prevent tumor infiltration of antitumor cytotoxic CD8+ T cells (139,142). In addition, the types and levels of bioactive molecules and growth factors secreted in the tumor microenvironment and involved in signaling pathways can modulate the function of immune cells. For example, pro-inflammatory cytokines stimulate the antitumor activity of the immune cells while anti-inflammatory cytokines create an immunosuppressive environment that shifts the function of immune cells toward tissue repair and regeneration. Interleukin-4 (IL-4) and interleukin-13 (IL-13) can shift tumor-associated macrophages toward the alternatively activated M2 phenotype which promotes tumor growth and stimulates the metastatic behavior of cancer cells. Similarly, transforming growth factor-β (TGF-β) shifts tumor-associated neutrophils toward a protumor phenotype (141,143,144). The communication between cancer cells and the immune cells continues during different stages of the cancer metastasis cascade. During cancer cell invasion and intravasation, immune cells secrete enzymes, such as MMPs, that contribute to ECM remodeling. In addition, the newly developed blood vessels facilitate cancer cell spread to different organs. Receptor activator of nuclear factor-B ligand (RANKL) which is secreted by T regulator cells improves CTC survival during the circulation and contributes to the development of the pre-metastatic niche in the secondary site (145,146). More details regarding the role of immune cells in cancer metastasis are reviewed elsewhere (147,148).

The use of aptamer-based therapeutics to inhibit the recruitment and function of immunosuppressive cells and modulate the immune cell phenotype from a protumor to an antitumor phenotype are promising strategies by which to inhibit cancer metastasis. The development of aptamer-based therapeutics that prevent immunosuppressive cells from exerting their action has been exploited. The interleukin 4 receptor-α (IL4R-α) antagonist aptamer that targets IL4R-α-expressing M2 tumor-associated macrophages and myeloid-derived suppressor cells led to the elimination of immunosuppressive cells in vivo. Subsequently, the numbers of antitumor CD8+ T cells were increased which suppressed the formation of pulmonary metastasis of 4T1 metastatic breast cancer cells (149). The utilization of aptamers to deliver therapeutics to tumor metastases also has been developed. Doxorubicin-loaded liposomes decorated with a T1 aptamer that target MDSCs showed significant depletion of MDSCs and increased intratumoral accumulation of cytotoxic T cells in animals with bone metastasis of MDA-MB-231 breast cancer cells (150). In addition, the 4-1BB aptamer was used to deliver a small non-coding antisense RNA (sasRNA) to Treg cells for transcriptional gene silencing of a key regulator of the immunosuppressive phenotype of Treg cells. Aptamer-sasRNA conjugate inhibited the Treg immunosuppressive phenotype in vitro and significantly improved coadministered antitumor vaccine against B16F10 metastatic melanoma cells that were grown subcutaneously in vivo (142,151). Similar findings have been reported after using an aptamer that targets cytotoxic T lymphocyte-associated antigen 4 (CTLA4) to deliver STAT3 siRNA to Treg cells. The CTLA4-STAT3 siRNA conjugate depleted Treg cells in the primary and metastatic sites and prevented pulmonary metastasis of B16 melanoma cells in vivo (19). In addition, aptamers that target and prevent the functions of anti-inflammatory cytokines or their receptors can inhibit immune cell polarization toward a protumor phenotype. Aptamers that target IL-6, IL-6R, IL-10R have been developed (152-155), yet their therapeutic activities against metastasis need to be examined.

5. Limitations and future perspectives

The development of aptamer-based anticancer therapeutics has been increasingly growing in the last few years. Encouraging findings introduce aptamer-based therapeutics as a potential class of agents in the treatment of primary tumors. However, cancer metastasis is the main cause for cancer-related deaths, thus preventing or treating cancer metastasis is pivotal to improving survival rates. Recently, extensive efforts have been directed toward developing and evaluating aptamer-based therapeutics against cancer metastasis. Table I documents examples of aptamer-based therapeutics that have been examined against cancer metastasis in preclinical and clinical settings and reported between 2019 and 2022 (133,156-161). Unfortunately, no aptamer-based therapeutic has been approved by the FDA to treat cancer metastasis thus far limiting the use of aptamer-based therapeutics in cancer metastasis treatment. It has been mentioned earlier in this review that some strategies had been introduced to overcome the challenges concerning aptamer pharmacokinetics such as enzymatic degradation, rapid excretion by the kidney, and poor delivery to intracellular targets. Nevertheless, other challenges still exist, and overcoming these challenges is crucial before aptamer-based technology can be approved for use against cancer metastasis. For example, immunological reactions resulting from aptamer administration have been observed. A randomized clinical trial evaluating the effect of REG1 anticoagulation system, which is an RNA aptamer-based inhibitor of the coagulation factor IXa, was terminated due to a severe allergic reaction (162). Research has demonstrated that allergic reactions observed with aptamer-based therapeutics result from factors such as the CpG-containing sequence and PEGylation (163-165). Understanding the underlying mechanisms by which the immunological reactions are triggered as well as finding the design elements contributing to immunogenicity is important for the successful development of safe aptamers. Furthermore, it is extremely important to identify unwanted immunological reactions when using aptamers that target and modulate immune cells involved in cancer metastasis. In addition, information concerning the long-term consequences of targeting tumor-associated immune cells, and whether the impact can be extended to affect other cells in the immune system network are lacking. In addition, insufficient toxicity data and lack of appropriate regulatory guidelines for preclinical toxicity assessment studies are challenges that could restrain aptamers from being approved for treatment against cancer metastasis. Toxicity observed with other oligonucleotide-based therapeutics such as antisense oligonucleotides have raised a red flag about possible toxicity that could not be detected with current study designs when assessing the toxicity of oligonucleotide aptamers (166,167). Pegaptanib is the only FDA-approved aptamer-based therapeutic for the treatment of neovascular age-related macular degeneration. However, postmarketing surveillance of the adverse drug reactions of pegaptanib suggests a critical need for a long-term assessment for possible severe adverse drug reactions and toxicity (168). Moreover, a survey conducted by the Oligonucleotide Working Group of the European Federation of Pharmaceutical Industries and Associations (EFPIA) demonstrated a discrepancy in preclinical safety assessment studies performed for oligonucleotide-based therapeutics (169). Thus, establishing optimal toxicity assessment guidelines is critical for fully characterizing aptamer toxicity. Another limitation is the lengthy and complex production process which is an obstacle for large-scale production for clinical use. The SELEX process is carried out in multiple rounds in which each round consists of multiple steps: incubation of the target with a random library, removing the unbound sequences from the bound sequence, elution of the desirable bound sequence, and amplification of the desirable bound sequence (11). Moreover, a successful selection of an aptamer necessitates optimizing the experimental conditions such as temperature, pH, ionic strength, and ratio of the target to the random library which creates a complex process. Therefore, identification of strategies to improve the aptamer selection process will help facilitate the large-scale production of aptamer-based therapeutics.

Table I

Examples of aptamer-based therapeutics that have been examined against cancer metastasis in preclinical and clinical settings and reported between 2019 and 2022.

Table I

Examples of aptamer-based therapeutics that have been examined against cancer metastasis in preclinical and clinical settings and reported between 2019 and 2022.

A, Aptamer-based therapeutics with confirmed efficacy in animal models of cancer metastasis
Aptamer nameAptamer typeTargetFunctionCancer typeAnimal modelCell line used(Refs.)
TR4
P19
RNA RNAHuman transferrin receptor 1 (hTfR1) PDAC cell specificTargeted delivery and internalization of C/EBPα-saRNA (C/EBPα-saRNA-TR4 or C/EBPα-saRNA-P19 conjugates)Pancreatic ductal adenocarcinoma (PDAC)Direct intrahepatic implantation of pancreatic cancer (liver-metastatic model of pancreatic cancer)PANC-1(156)
Gint4.TRNAPlatelet-derived growth factor receptor β (PDGFRβ)Inhibition of ligand-dependent receptor activation and its downstream signalingTriple-negative breast cancerSpontaneous lung metastasis in orthotopic mouse model of TNBC4T1(157)
GL21.TRNAAXL, an oncogenic tyrosine kinase receptorInhibition of AXL downstream signaling and as targeted delivery of miR-148b (axl-miR-148b conjugate)Breast cancer MelanomaSpontaneous lung and liver metastases in orthotopic breast cancer or melanoma subcutaneous mouse models4175-TGL/MA-2(158)
AIA1RNAMetastasis phenotypeInvasion inhibitionProstate cancer OsteosarcomaWidespread metastases of prostate cancer using intracardiac injection/Spontaneous lung metastasis in orthotopic primary model of osteosarcomaPC-3/82L(159)
EpCAM aptamerRNAEpithelial cell adhesion molecule (EpCAM)Targeted delivery of miR-203b-3p (EpCAM-Apt/miR-203b-3p)Ovarian cancerOvarian peritoneal metastasis modelSK-OV-3 ES2(160)
Anti-EGFRDNAEpidermal growth factor receptor (EGFR)Helping nanoparticles for specific recognition of oncogenic exosomes and decreasing exosomes circulationLung cancerSpontaneous lung metastases in subcutaneous mouse model of human lung cancerA549(133)
P42PeptideSRY (sex determining region Y)-box 2 (SOX2)Inhibition of SOX2 function and its downstream targetsEsophageal squamous cell carcinoma (ESCC)Zebrafish model of human esophageal squamous cell carcinoma metastasisKYSE-450(161)

B, Aptamer-based therapeutics evaluated against cancer metastasis in clinical trials
Aptamer nameAptamer typeTargetPhaseStatusCancer typeClinicalTrials.gov Identifier
Olaptesed (NOX-A12)RNAC-X-C motif chemokine 12 (CXCL12)Phase I/IICompleted March 2020Metastatic colorectal cancer
Metastatic pancreatic cancer
NCT03168139

Targeting key mediators of cancer metastasis using aptamer-based therapeutics is a promising strategy against metastasis; yet, researchers should solve concerns with the current research. First, exploring new targets and assessing the efficacy of aptamer-based therapy in relevant in vivo models are still limited. Enormous molecular targets and factors that regulate cancer metastasis have not been explored as potential targets for aptamer-based antimetastatic therapy. For example, miRNAs are RNAs with short, highly conserved, non-coding sequences. By binding to the 3′untranslated region of target mRNAs, miRNAs control gene expression at the posttranscriptional level. Recent findings demonstrate that miRNAs control metastasis by regulating metastasis-related genes in cancer stem cells and during the processes of EMT and metastatic colonization (170,171). Using aptamers to bind oncogenic precursor microRNAs to inhibit functional miRNA formation or deliver tumor suppressor miRNAs in the form of aptamer-miRNA conjugate may show antitumor activity (172-175). Furthermore, circular RNAs (circRNAs), which range in length from a few hundred to thousands of nucleotides, is a type of RNA that has been recently re-recognized (176). Contrary to linear RNAs that have 5′caps and 3′tails, circRNAs are single-stranded with covalently closed circular transcripts (177). They were earlier known to be the products of mis-splicing or by-products of pre-mRNA processing with poor abundance, but now they have been recognized as a class of non-coding RNAs following the use of high-throughput RNA sequencing (RNA-seq) technologies. circRNAs are observed to be involved in the pathogenesis of cancer tumorigenesis, metastasis, and therapy resistance (178,179). These potential strategies have been examined in vitro or clinically irrelevant in vivo models (142,173,175). Thus, assessment of the efficacy of developed aptamer technology against newly identified targets in appropriate animal models of cancer metastasis is crucial to develop effective aptamer-based therapeutics.

6. Conclusion

In conclusion, this review summarizes recent advances in the development of aptamer-based antimetastatic therapeutics. Studies discussed here demonstrate that targeted aptamers possess a promising future in fighting cancer metastasis. Despite the potential findings in the field of molecular and cellular oncology, there are still opportunities to explore aptamer technology against potential targets. This could result in the development of aptamer-based antimetastatic therapy to target cancer metastasis. In addition, more research is needed to examine the existing antimetastatic aptamers in situations that mimic cancer metastasis inside the body.

Availability of data and materials

All information provided in this review is documented by relevant references.

Authors' contributions

YA and HA contributed in the idea and writing the manuscript. SA and AA participated in writing and editing the manuscript. All authors revised and approved the final version.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Authors' information

Author names and ORCID nos. are as follows: Yahya Alhamhoom: 0000-0002-8368-9047; Homood M. As Sobeai: 0000-0003-3073-3072; Sary Alsanea: 0000-0002-5323-5781; Ali Alhoshani: 0000-0002-3450-283X.

Competing interests

The authors declare no competing interests.

Acknowledgments

Not applicable.

Funding

The authors reported there is no funding associated with this work.

Abbreviations:

Angptl4

angiopoietin-like-4

CEA

carcinoembryonic antigen

circRNA

circular RNA

COX-2

cyclooxygenase-2

CSCs

cancer stem cells

CTCs

circulating tumor cells

CTLA4

cytotoxic T lymphocyte-associated antigen 4

ECM

extracellular matrix

EGFR

epidermal growth factor receptor

EMT

epithelial-mesenchymal transition

EREG

epiregulin

EVs

extracellular vesicles

FAK

focal adhesion kinase

HepG2

hepatocellular carcinoma cells

HER2

human epidermal growth factor receptor 2

HGF

hepatocyte growth factor

IL

interleukin

IL4R-α

Interleukin 4 receptor-α

LAC

lung adenocarcinoma

lncRNAs

long non-coding RNAs

LOX

lysyl oxidase

MDSCs

myeloid-derived suppressor cells

MED1

mediator complex subunit1

miRNAs/miRs

microRNAs

MMPs

matrix metalloproteinases

MUC1

mucin 1 antigen

MVs

microvesicles

OPN

osteopontin

Psap

prosaposin

PTDs

protein transduction domains

RANKL

receptor activator of nuclear factor-B ligand

RNA-seq

RNA sequencing

sasRNA

small antisense RNA

SDF-1

stromal cell-derived factor 1

SELEX

Systemic Evolution of Ligands by Exponential Enrichment

sLex

sialyl Lewis X

ssDNA/RNA

sequences of single-stranded nucleic acids

TGF-β

transforming growth factor-β

TICs

tumor-initiating cells

Tregs

regulatory T cells

VEGFR1+

VEGF receptor 1-positive

VEGFs

vascular endothelial growth factors

References

1 

Chiang CJ, Lo WC, Yang YW, You SL, Chen CJ and Lai MS: Incidence and survival of adult cancer patients in Taiwan, 2002-2012. J Formos Med Assoc. 115:1076–1088. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Liu CM, Peng CY, Liao YW, Lu MY, Tsai ML, Yeh JC, Yu CH and Yu CC: Sulforaphane targets cancer stemness and tumor initiating properties in oral squamous cell carcinomas via miR-200c induction. J Formos Med Assoc. 116:41–48. 2017. View Article : Google Scholar

3 

Tsai CE, Wu KL, Chiu YC, Chuah SK, Tai WC, Hu ML and Liang CM: The incidence and clinical associated factors of interval colorectal cancers in Southern Taiwan. J Formos Med Assoc. 117:185–190. 2018. View Article : Google Scholar

4 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Dillekas H, Rogers MS and Straume O: Are 90% of deaths from cancer caused by metastases? Cancer Med. 8:5574–5576. 2019. View Article : Google Scholar : PubMed/NCBI

6 

Fidler IJ: The pathogenesis of cancer metastasis: The 'seed and soil' hypothesis revisited. Nat Rev Cancer. 3:453–458. 2003. View Article : Google Scholar : PubMed/NCBI

7 

Stewart CM and Tsui DWY: Circulating cell-free DNA for non-invasive cancer management. Cancer Genet. 228-229:169–179. 2018. View Article : Google Scholar : PubMed/NCBI

8 

Hafeez U, Gan HK and Scott AM: Monoclonal antibodies as immunomodulatory therapy against cancer and autoimmune diseases. Curr Opin Pharmacol. 41:114–121. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Dominiak A, Chelstowska B, Olejarz W and Nowicka G: Communication in the cancer microenvironment as a target for therapeutic interventions. Cancers (Basel). 12:12322020. View Article : Google Scholar

10 

Tuerk C and Gold L: Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science. 249:505–510. 1990. View Article : Google Scholar : PubMed/NCBI

11 

Ellington AD and Szostak JW: In vitro selection of RNA molecules that bind specific ligands. Nature. 346:818–822. 1990. View Article : Google Scholar : PubMed/NCBI

12 

Zhou J and Rossi J: Aptamers as targeted therapeutics: Current potential and challenges. Nat Rev Drug Discov. 16:181–202. 2017. View Article : Google Scholar

13 

Toh SY, Citartan M, Gopinath SC and Tang TH: Aptamers as a replacement for antibodies in enzyme-linked immunosorbent assay. Biosens Bioelectron. 64:392–403. 2015. View Article : Google Scholar

14 

Mascini M, Palchetti I and Tombelli S: Nucleic acid and peptide aptamers: Fundamentals and bioanalytical aspects. Angew Chem Int Ed Engl. 51:1316–1332. 2012. View Article : Google Scholar : PubMed/NCBI

15 

Colas P, Cohen B, Jessen T, Grishina I, McCoy J and Brent R: Genetic selection of peptide aptamers that recognize and inhibit cyclin-dependent kinase 2. Nature. 380:548–550. 1996. View Article : Google Scholar : PubMed/NCBI

16 

Reverdatto S, Burz DS and Shekhtman A: Peptide aptamers: Development and applications. Curr Top Med Chem. 15:1082–1101. 2015. View Article : Google Scholar : PubMed/NCBI

17 

Han J, Gao L and Wang J and Wang J: Application and development of aptamer in cancer: From clinical diagnosis to cancer therapy. J Cancer. 11:6902–6915. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Pan Q, Luo F, Liu M and Zhang XL: Oligonucleotide aptamers: Promising and powerful diagnostic and therapeutic tools for infectious diseases. J Infect. 77:83–98. 2018. View Article : Google Scholar : PubMed/NCBI

19 

Herrmann A, Priceman SJ, Swiderski P, Kujawski M, Xin H, Cherryholmes GA, Zhang W, Zhang C, Lahtz C, Kowolik C, et al: CTLA4 aptamer delivers STAT3 siRNA to tumor-associated and malignant T cells. J Clin Invest. 124:2977–2987. 2014. View Article : Google Scholar : PubMed/NCBI

20 

Ospina-Villa JD, Zamorano-Carrillo A, Castanon-Sanchez CA, Ramirez-Moreno E and Marchat LA: Aptamers as a promising approach for the control of parasitic diseases. Braz J Infect Dis. 20:610–618. 2016. View Article : Google Scholar : PubMed/NCBI

21 

Liu M, Zaman K and Fortenberry YM: Overview of the therapeutic potential of aptamers targeting coagulation factors. Int J Mol Sci. 22:38972021. View Article : Google Scholar : PubMed/NCBI

22 

Ninichuk V, Clauss S, Kulkarni O, Schmid H, Segerer S, Radomska E, Eulberg D, Buchner K, Selve N, Klussmann S and Anders HJ: Late onset of Ccl2 blockade with the Spiegelmer mNOX-E36-3′PEG prevents glomerulosclerosis and improves glomerular filtration rate in db/db mice. Am J Pathol. 172:628–637. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Ng EW, Shima DT, Calias P, Cunningham ET Jr, Guyer DR and Adamis AP: Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular disease. Nat Rev Drug Discov. 5:123–132. 2006. View Article : Google Scholar : PubMed/NCBI

24 

Gauthier NC and Roca-Cusachs P: Mechanosensing at integrin-mediated cell-matrix adhesions: From molecular to integrated mechanisms. Curr Opin Cell Biol. 50:20–26. 2018. View Article : Google Scholar : PubMed/NCBI

25 

Bissell MJ and Hines WC: Why don't we get more cancer? A proposed role of the microenvironment in restraining cancer progression. Nat Med. 17:320–329. 2011. View Article : Google Scholar : PubMed/NCBI

26 

Friedl P and Wolf K: Tumour-cell invasion and migration: Diversity and escape mechanisms. Nat Rev Cancer. 3:362–374. 2003. View Article : Google Scholar

27 

Zheng X, Yu C and Xu M: Linking tumor microenvironment to plasticity of cancer stem cells: Mechanisms and application in cancer therapy. Front Oncol. 11:6783332021. View Article : Google Scholar : PubMed/NCBI

28 

Krakhmal NV, Zavyalova MV, Denisov EV, Vtorushin SV and Perelmuter VM: Cancer invasion: Patterns and mechanisms. Acta Naturae. 7:17–28. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Thiery JP, Acloque H, Huang RY and Nieto MA: Epithelial-mesenchymal transitions in development and disease. Cell. 139:871–890. 2009. View Article : Google Scholar : PubMed/NCBI

30 

Joyce JA and Pollard JW: Microenvironmental regulation of metastasis. Nat Rev Cancer. 9:239–252. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Dirat B, Bochet L, Dabek M, Daviaud D, Dauvillier S, Majed B, Wang YY, Meulle A, Salles B, Le Gonidec S, et al: Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 71:2455–2465. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Gocheva V, Wang HW, Gadea BB, Shree T, Hunter KE, Garfall AL, Berman T and Joyce JA: IL-4 induces cathepsin protease activity in tumor-associated macrophages to promote cancer growth and invasion. Genes Dev. 24:241–255. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Gupta GP and Massague J: Cancer metastasis: Building a framework. Cell. 127:679–695. 2006. View Article : Google Scholar

34 

Carmeliet P and Jain RK: Principles and mechanisms of vessel normalization for cancer and other angiogenic diseases. Nat Rev Drug Discov. 10:417–427. 2011. View Article : Google Scholar : PubMed/NCBI

35 

Gupta GP, Nguyen DX, Chiang AC, Bos PD, Kim JY, Nadal C, Gomis RR, Manova-Todorova K and Massagué J: Mediators of vascular remodelling co-opted for sequential steps in lung metastasis. Nature. 446:765–770. 2007. View Article : Google Scholar : PubMed/NCBI

36 

Guo W and Giancotti FG: Integrin signalling during tumour progression. Nat Rev Mol Cell Biol. 5:816–826. 2004. View Article : Google Scholar : PubMed/NCBI

37 

Douma S, Van Laar T, Zevenhoven J, Meuwissen R, Van Garderen E and Peeper DS: Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature. 430:1034–1039. 2004. View Article : Google Scholar : PubMed/NCBI

38 

Brown DM and Ruoslahti E: Metadherin, a cell surface protein in breast tumors that mediates lung metastasis. Cancer Cell. 5:365–374. 2004. View Article : Google Scholar : PubMed/NCBI

39 

Auguste P, Fallavollita L, Wang N, Burnier J, Bikfalvi A and Brodt P: The host inflammatory response promotes liver metastasis by increasing tumor cell arrest and extravasation. Am J Pathol. 170:1781–1792. 2007. View Article : Google Scholar : PubMed/NCBI

40 

Al-Mehdi AB, Tozawa K, Fisher AB, Shientag L, Lee A and Muschel RJ: Intravascular origin of metastasis from the proliferation of endothelium-attached tumor cells: A new model for metastasis. Nat Med. 6:100–102. 2000. View Article : Google Scholar

41 

Padua D, Zhang XH, Wang Q, Nadal C, Gerald WL, Gomis RR and Massagué J: TGFbeta primes breast tumors for lung metastasis seeding through angiopoietin-like 4. Cell. 133:66–77. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Psaila B and Lyden D: The metastatic niche: Adapting the foreign soil. Nat Rev Cancer. 9:285–293. 2009. View Article : Google Scholar

43 

Erler JT, Bennewith KL, Cox TR, Lang G, Bird D, Koong A, Le QT and Giaccia AJ: Hypoxia-induced lysyl oxidase is a critical mediator of bone marrow cell recruitment to form the premetastatic niche. Cancer Cell. 15:35–44. 2009. View Article : Google Scholar

44 

Chambers AF, Groom AC and MacDonald IC: Dissemination and growth of cancer cells in metastatic sites. Nat Rev Cancer. 2:563–572. 2002. View Article : Google Scholar : PubMed/NCBI

45 

Barkan D, El Touny LH, Michalowski AM, Smith JA, Chu I, Davis AS, Webster JD, Hoover S, Simpson RM, Gauldie J and Green JE: Metastatic growth from dormant cells induced by a col-I-enriched fibrotic environment. Cancer Res. 70:5706–5716. 2010. View Article : Google Scholar : PubMed/NCBI

46 

Barkan D, Kleinman H, Simmons JL, Asmussen H, Kamaraju AK, Hoenorhoff MJ, Liu ZY, Costes SV, Cho EH, Lockett S, et al: Inhibition of metastatic outgrowth from single dormant tumor cells by targeting the cytoskeleton. Cancer Res. 68:6241–6250. 2008. View Article : Google Scholar : PubMed/NCBI

47 

Shibue T and Weinberg RA: Integrin beta1-focal adhesion kinase signaling directs the proliferation of metastatic cancer cells disseminated in the lungs. Proc Natl Acad Sci USA. 106:10290–10295. 2009. View Article : Google Scholar : PubMed/NCBI

48 

Hiratsuka S, Duda DG, Huang Y, Goel S, Sugiyama T, Nagasawa T, Fukumura D and Jain RK: C-X-C receptor type 4 promotes metastasis by activating p38 mitogen-activated protein kinase in myeloid differentiation antigen (Gr-1)-positive cells. Proc Natl Acad Sci USA. 108:302–307. 2011. View Article : Google Scholar :

49 

McAllister SS, Gifford AM, Greiner AL, Kelleher SP, Saelzler MP, Ince TA, Reinhardt F, Harris LN, Hylander BL, Repasky EA and Weinberg RA: Systemic endocrine instigation of indolent tumor growth requires osteopontin. Cell. 133:994–1005. 2008. View Article : Google Scholar : PubMed/NCBI

50 

Kang SY, Halvorsen OJ, Gravdal K, Bhattacharya N, Lee JM, Liu NW, Johnston BT, Johnston AB, Haukaas SA, Aamodt K, et al: Prosaposin inhibits tumor metastasis via paracrine and endocrine stimulation of stromal p53 and Tsp-1. Proc Natl Acad Sci USA. 106:12115–12120. 2009. View Article : Google Scholar : PubMed/NCBI

51 

Shimono Y, Zabala M, Cho RW, Lobo N, Dalerba P, Qian D, Diehn M, Liu H, Panula SP, Chiao E, et al: Downregulation of miRNA-200c links breast cancer stem cells with normal stem cells. Cell. 138:592–603. 2009. View Article : Google Scholar : PubMed/NCBI

52 

Cai S, Yan J, Xiong H, Liu Y, Peng D and Liu Z: Investigations on the interface of nucleic acid aptamers and binding targets. Analyst. 143:5317–5338. 2018. View Article : Google Scholar : PubMed/NCBI

53 

Ni S, Zhuo Z, Pan Y, Yu Y, Li F, Liu J, Wang L, Wu X, Li D, Wan Y, et al: Recent progress in aptamer discoveries and modifications for therapeutic applications. ACS Appl Mater Interfaces. 13:9500–9519. 2021. View Article : Google Scholar

54 

Zhang Y, Lai BS and Juhas M: Recent advances in aptamer discovery and applications. Molecules. 24:9412019. View Article : Google Scholar :

55 

Dong Y, Zhang T, Lin X, Feng J, Luo F, Gao H, Wu Y, Deng R and He Q: Graphene/aptamer probes for small molecule detection: from in vitro test to in situ imaging. Mikrochim Acta. 187:1792020. View Article : Google Scholar : PubMed/NCBI

56 

Cho SJ, Woo HM, Kim KS, Oh JW and Jeong YJ: Novel system for detecting SARS coronavirus nucleocapsid protein using an ssDNA aptamer. J Biosci Bioeng. 112:535–540. 2011. View Article : Google Scholar : PubMed/NCBI

57 

Li HY, Jia WN, Li XY, Zhang L, Liu C and Wu J: Advances in detection of infectious agents by aptamer-based technologies. Emerg Microbes Infect. 9:1671–1681. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Hassan EM, Willmore WG and DeRosa MC: Aptamers: Promising tools for the detection of circulating tumor cells. Nucleic Acid Ther. 26:335–347. 2016. View Article : Google Scholar : PubMed/NCBI

59 

Orava EW, Cicmil N and Gariepy J: Delivering cargoes into cancer cells using DNA aptamers targeting internalized surface portals. Biochim Biophys Acta. 1798:2190–2200. 2010. View Article : Google Scholar : PubMed/NCBI

60 

Zhang GX, Liu YL, Yang M, Huang WS and Xu JH: An aptamer-based, fluorescent and radionuclide dual-modality probe. Biochimie. 171-172:55–62. 2020. View Article : Google Scholar : PubMed/NCBI

61 

Hashemitabar S, Yazdian-Robati R, Hashemi M, Ramezani M, Abnous K and Kalalinia F: ABCG2 aptamer selectively delivers doxorubicin to drug-resistant breast cancer cells. J Biosci. 44:392019. View Article : Google Scholar : PubMed/NCBI

62 

Kruspe S and Giangrande PH: Aptamer-siRNA chimeras: Discovery, progress and future prospects. Biomedicines. 5:452017. View Article : Google Scholar

63 

Jiang L, Wang H and Chen S: Aptamer (AS1411)-conjugated liposome for enhanced therapeutic efficacy of miRNA-29b in ovarian cancer. J Nanosci Nanotechnol. 20:2025–2031. 2020. View Article : Google Scholar

64 

Meng HM, Liu H, Kuai H, Peng R, Mo L and Zhang XB: Aptamer-integrated DNA nanostructures for biosensing, bioimaging and cancer therapy. Chem Soc Rev. 45:2583–2602. 2016. View Article : Google Scholar : PubMed/NCBI

65 

Xiang D, Shigdar S, Qiao G, Wang T, Kouzani AZ, Zhou SF, Kong L, Li Y, Pu C and Duan W: Nucleic acid aptamer-guided cancer therapeutics and diagnostics: The next generation of cancer medicine. Theranostics. 5:23–42. 2015. View Article : Google Scholar : PubMed/NCBI

66 

Wu C, Han D, Chen T, Peng L, Zhu G, You M, Qiu L, Sefah K, Zhang X and Tan W: Building a multifunctional aptamer-based DNA nanoassembly for targeted cancer therapy. J Am Chem Soc. 135:18644–18650. 2013. View Article : Google Scholar : PubMed/NCBI

67 

Kuwahara M and Sugimoto N: Molecular evolution of functional nucleic acids with chemical modifications. Molecules. 15:5423–5444. 2010. View Article : Google Scholar : PubMed/NCBI

68 

Mayer G: The chemical biology of aptamers. Angew Chem Int Ed Engl. 48:2672–2689. 2009. View Article : Google Scholar : PubMed/NCBI

69 

Turner JJ, Hoos JS, Vonhoff S and Klussmann S: Methods for L-ribooligonucleotide sequence determination using LCMS. Nucleic Acids Res. 39:e1472011. View Article : Google Scholar : PubMed/NCBI

70 

Lee CH, Lee SH, Kim JH, Noh YH, Noh GJ and Lee SW: Pharmacokinetics of a Cholesterol-conjugated aptamer against the Hepatitis C Virus (HCV) NS5B protein. Mol Ther Nucleic Acids. 4:e2542015. View Article : Google Scholar : PubMed/NCBI

71 

Dougan H, Lyster DM, Vo CV, Stafford A, Weitz JI and Hobbs JB: Extending the lifetime of anticoagulant oligodeoxynucleotide aptamers in blood. Nucl Med Biol. 27:289–297. 2000. View Article : Google Scholar

72 

Healy JM, Lewis SD, Kurz M, Boomer RM, Thompson KM, Wilson C and McCauley TG: Pharmacokinetics and biodistribution of novel aptamer compositions. Pharm Res. 21:2234–2246. 2004. View Article : Google Scholar

73 

Marshall ML and Wagstaff KM: Internalized functional DNA aptamers as alternative cancer therapies. Front Pharmacol. 11:11152020. View Article : Google Scholar : PubMed/NCBI

74 

Thiel WH, Thiel KW, Flenker KS, Bair T, Dupuy AJ, McNamara JO II, Miller FJ and Giangrande PH: Cell-internalization SELEX: Method for identifying cell-internalizing RNA aptamers for delivering siRNAs to target cells. Methods Mol Biol. 1218:187–199. 2015. View Article : Google Scholar :

75 

Alamudi SH, Kimoto M and Hirao I: Uptake mechanisms of cell-internalizing nucleic acid aptamers for applications as pharmacological agents. RSC Med Chem. 12:1640–1649. 2021. View Article : Google Scholar : PubMed/NCBI

76 

Nagel-Wolfrum K, Buerger C, Wittig I, Butz K, Hoppe-Seyler F and Groner B: The interaction of specific peptide aptamers with the DNA binding domain and the dimerization domain of the transcription factor Stat3 inhibits transactivation and induces apoptosis in tumor cells. Mol Cancer Res. 2:170–182. 2004.PubMed/NCBI

77 

Taylor RE and Zahid M: Cell penetrating peptides, novel vectors for gene therapy. Pharmaceutics. 12:2252020. View Article : Google Scholar :

78 

Moutal A, Francois-Moutal L, Brittain JM, Khanna M and Khanna R: Differential neuroprotective potential of CRMP2 peptide aptamers conjugated to cationic, hydrophobic, and amphipathic cell penetrating peptides. Front Cell Neurosci. 8:4712014.

79 

Famulok M, Blind M and Mayer G: Intramers as promising new tools in functional proteomics. Chem Biol. 8:931–939. 2001. View Article : Google Scholar : PubMed/NCBI

80 

Kunz C, Borghouts C, Buerger C and Groner B: Peptide aptamers with binding specificity for the intracellular domain of the ErbB2 receptor interfere with AKT signaling and sensitize breast cancer cells to Taxol. Mol Cancer Res. 4:983–998. 2006. View Article : Google Scholar : PubMed/NCBI

81 

Shiozawa Y, Nie B, Pienta KJ, Morgan TM and Taichman RS: Cancer stem cells and their role in metastasis. Pharmacol Ther. 138:285–293. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Velasco-Velazquez MA, Popov VM, Lisanti MP and Pestell RG: The role of breast cancer stem cells in metastasis and therapeutic implications. Am J Pathol. 179:2–11. 2011. View Article : Google Scholar : PubMed/NCBI

83 

Eyler CE and Rich JN: Survival of the fittest: Cancer stem cells in therapeutic resistance and angiogenesis. J Clin Oncol. 26:2839–2845. 2008. View Article : Google Scholar

84 

Chiou SH, Wang ML, Chou YT, Chen CJ, Hong CF, Hsieh WJ, Chang HT, Chen YS, Lin TW, Hsu HS and Wu CW: Coexpression of Oct4 and Nanog enhances malignancy in lung adenocarcinoma by inducing cancer stem cell-like properties and epithelial-mesenchymal transdifferentiation. Cancer Res. 70:10433–10444. 2010. View Article : Google Scholar : PubMed/NCBI

85 

Chiou SH, Yu CC, Huang CY, Lin SC, Liu CJ, Tsai TH, Chou SH, Chien CS, Ku HH and Lo JF: Positive correlations of Oct-4 and Nanog in oral cancer stem-like cells and high-grade oral squamous cell carcinoma. Clin Cancer Res. 14:4085–4095. 2008. View Article : Google Scholar : PubMed/NCBI

86 

Leis O, Eguiara A, Lopez-Arribillaga E, Alberdi MJ, Hernandez-Garcia S, Elorriaga K, Pandiella A, Rezola R and Martin AG: Sox2 expression in breast tumours and activation in breast cancer stem cells. Oncogene. 31:1354–1365. 2012. View Article : Google Scholar

87 

Lonardo E, Hermann PC, Mueller MT, Huber S, Balic A, Miranda-Lorenzo I, Zagorac S, Alcala S, Rodriguez-Arabaolaza I, Ramirez JC, et al: Nodal/Activin signaling drives self-renewal and tumorigenicity of pancreatic cancer stem cells and provides a target for combined drug therapy. Cell Stem Cell. 9:433–446. 2011. View Article : Google Scholar : PubMed/NCBI

88 

Paranjape AN, Balaji SA, Mandal T, Krushik EV, Nagaraj P, Mukherjee G and Rangarajan A: Bmi1 regulates self-renewal and epithelial to mesenchymal transition in breast cancer cells through Nanog. BMC Cancer. 14:7852014. View Article : Google Scholar : PubMed/NCBI

89 

Villodre ES, Kipper FC, Pereira MB and Lenz G: Roles of OCT4 in tumorigenesis, cancer therapy resistance and prognosis. Cancer Treat Rev. 51:1–9. 2016. View Article : Google Scholar

90 

Hochedlinger K, Yamada Y, Beard C and Jaenisch R: Ectopic expression of Oct-4 blocks progenitor-cell differentiation and causes dysplasia in epithelial tissues. Cell. 121:465–477. 2005. View Article : Google Scholar

91 

Wei X, He J, Wang J, Yang X and Ma B: Bmi-1 is essential for the oncogenic potential in CD133(+) human laryngeal cancer cells. Tumour Biol. 36:8931–8942. 2015. View Article : Google Scholar : PubMed/NCBI

92 

Chao HM, Huang HX, Chang PH, Tseng KC, Miyajima A and Chern E: Y-box binding protein-1 promotes hepatocellular carcinoma-initiating cell progression and tumorigenesis via Wnt/beta-catenin pathway. Oncotarget. 8:2604–2616. 2017. View Article : Google Scholar

93 

Chen YR, Sekine K, Nakamura K, Yanai H, Tanaka M and Miyajima A: Y-box binding protein-1 down-regulates expression of carbamoyl phosphate synthetase-I by suppressing CCAAT enhancer-binding protein-alpha function in mice. Gastroenterology. 137:330–340. 2009. View Article : Google Scholar : PubMed/NCBI

94 

Guerra C, Schuhmacher AJ, Cañamero M, Grippo PJ, Verdaguer L, Pérez-Gallego L, Dubus P, Sandgren EP and Barbacid M: Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. Cancer Cell. 11:291–302. 2007. View Article : Google Scholar : PubMed/NCBI

95 

Macdonald J, Henri J, Goodman L, Xiang D, Duan W and Shigdar S: Development of a bifunctional aptamer targeting the transferrin receptor and epithelial cell adhesion molecule (EpCAM) for the treatment of brain cancer metastases. Chronic pancreatitis is essential for induction of pancreatic ductal adenocarcinoma by K-Ras oncogenes in adult mice. ACS Chem Neurosci. 8:777–784. 2017. View Article : Google Scholar

96 

Yin H, Xiong G, Guo S, Xu C, Xu R, Guo P and Shu D: Delivery of Anti-miRNA for Triple-negative breast cancer therapy Using RNA nanoparticles targeting stem cell marker CD133. Mol Ther. 27:1252–1261. 2019. View Article : Google Scholar : PubMed/NCBI

97 

Zeng YB, Yu ZC, He YN, Zhang T, Du LB, Dong YM, Chen HW, Zhang YY and Wang WQ: Salinomycin-loaded lipid-polymer nanoparticles with anti-CD20 aptamers selectively suppress human CD20+ melanoma stem cells. Acta Pharmacol Sin. 39:261–274. 2018. View Article : Google Scholar :

98 

Zhang Y, Leonard M, Shu Y, Yang Y, Shu D, Guo P and Zhang X: Overcoming tamoxifen resistance of human breast cancer by targeted gene silencing using multifunctional pRNA nanoparticles. ACS Nano. 11:335–346. 2017. View Article : Google Scholar

99 

Zhou G, Da Won Bae S, Nguyen R, Huo X, Han S, Zhang Z, Hebbard L, Duan W, Eslam M, Liddle C, et al: An aptamer-based drug delivery agent (CD133-apt-Dox) selectively and effectively kills liver cancer stem-like cells. Cancer Lett. 501:124–132. 2021. View Article : Google Scholar

100 

Kim DM, Kim M, Park HB, Kim KS and Kim DE: Anti-MUC1/CD44 dual-aptamer-conjugated liposomes for cotargeting breast cancer cells and cancer stem cells. ACS Applied Bio Materials. 2:4622–4633. 2019. View Article : Google Scholar

101 

Talmadge JE and Fidler IJ: AACR centennial series: The biology of cancer metastasis: Historical perspective. Cancer Res. 70:5649–5669. 2010. View Article : Google Scholar : PubMed/NCBI

102 

Sleeman JP, Nazarenko I and Thiele W: Do all roads lead to Rome? Routes to metastasis development. Int J Cancer. 128:2511–2526. 2011. View Article : Google Scholar : PubMed/NCBI

103 

Liljefors M, Nilsson B, Fagerberg J, Ragnhammar P, Mellstedt H and Frodin JE: Clinical effects of a chimeric anti-EpCAM monoclonal antibody in combination with granulocyte-macrophage colony-stimulating factor in patients with metastatic colorectal carcinoma. Int J Oncol. 26:1581–1589. 2005.PubMed/NCBI

104 

Scarberry KE, Mezencev R and McDonald JF: Targeted removal of migratory tumor cells by functionalized magnetic nanoparticles impedes metastasis and tumor progression. Nanomedicine (Lond). 6:69–78. 2011. View Article : Google Scholar

105 

Orava EW, Abdul-Wahid A, Huang EH, Mallick AI and Gariepy J: Blocking the attachment of cancer cells in vivo with DNA aptamers displaying anti-adhesive properties against the carcinoembryonic antigen. Mol Oncol. 7:799–811. 2013. View Article : Google Scholar : PubMed/NCBI

106 

Abdul-Wahid A, Huang EH, Cydzik M, Bolewska-Pedyczak E and Gariepy J: The carcinoembryonic antigen IgV-like N domain plays a critical role in the implantation of metastatic tumor cells. Mol Oncol. 8:337–350. 2014. View Article : Google Scholar : PubMed/NCBI

107 

Wang XK, Peng Y, Tao HR, Zhou FF, Zhang C, Su F, Wang SP, Liu Q, Xu LH, Pan XK, et al: Inhibition of adhesion and metastasis of HepG2 hepatocellular carcinoma cells in vitro by DNA aptamer against sialyl Lewis X. J Huazhong Univ Sci Technolog Med Sci. 37:343–347. 2017. View Article : Google Scholar

108 

Brodt P, Fallavollita L, Bresalier RS, Meterissian S, Norton CR and Wolitzky BA: Liver endothelial E-selectin mediates carcinoma cell adhesion and promotes liver metastasis. Int J Cancer. 71:612–619. 1997. View Article : Google Scholar : PubMed/NCBI

109 

Kowal J, Arras G, Colombo M, Jouve M, Morath JP, Primdal-Bengtson B, Dingli F, Loew D, Tkach M and Théry C: Proteomic comparison defines novel markers to characterize heterogeneous populations of extracellular vesicle subtypes. Proc Natl Acad Sci USA. 113:E968–E977. 2016. View Article : Google Scholar : PubMed/NCBI

110 

Minciacchi VR, Freeman MR and Di Vizio D: Extracellular vesicles in cancer: Exosomes, microvesicles and the emerging role of large oncosomes. Semin Cell Dev Biol. 40:41–51. 2015. View Article : Google Scholar : PubMed/NCBI

111 

Raposo G and Stoorvogel W: Extracellular vesicles: Exosomes, microvesicles, and friends. J Cell Biol. 200:373–383. 2013. View Article : Google Scholar : PubMed/NCBI

112 

Yang F, Ning Z, Ma L, Liu W, Shao C, Shu Y and Shen H: Exosomal miRNAs and miRNA dysregulation in cancer-associated fibroblasts. Mol Cancer. 16:1482017. View Article : Google Scholar : PubMed/NCBI

113 

Colombo M, Raposo G and Thery C: Biogenesis, secretion, and intercellular interactions of exosomes and other extracellular vesicles. Annu Rev Cell Dev Biol. 30:255–289. 2014. View Article : Google Scholar : PubMed/NCBI

114 

Gould SJ and Raposo G: As we wait: Coping with an imperfect nomenclature for extracellular vesicles. J Extracell Vesicles. 2:203892013. View Article : Google Scholar

115 

Li W, Ma H, Zhang J, Zhu L, Wang C and Yang Y: Unraveling the roles of CD44/CD24 and ALDH1 as cancer stem cell markers in tumorigenesis and metastasis. Sci Rep. 7:138562017. View Article : Google Scholar : PubMed/NCBI

116 

van Niel G, D'Angelo G and Raposo G: Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 19:213–228. 2018. View Article : Google Scholar : PubMed/NCBI

117 

Hannafon BN and Ding WQ: Intercellular communication by exosome-derived microRNAs in cancer. Int J Mol Sci. 14:14240–14269. 2013. View Article : Google Scholar : PubMed/NCBI

118 

Milane L, Singh A, Mattheolabakis G, Suresh M and Amiji MM: Exosome mediated communication within the tumor microenvironment. J Control Release. 219:278–294. 2015. View Article : Google Scholar : PubMed/NCBI

119 

Valadi H, Ekstrom K, Bossios A, Sjostrand M, Lee JJ and Lotvall JO: Exosome-mediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells. Nat Cell Biol. 9:654–659. 2007. View Article : Google Scholar : PubMed/NCBI

120 

Gezer U, Ozgur E, Cetinkaya M, Isin M and Dalay N: Long non-coding RNAs with low expression levels in cells are enriched in secreted exosomes. Cell Biol Int. 38:1076–1079. 2014.PubMed/NCBI

121 

Li Z, Huang C, Bao C, Chen L, Lin M, Wang X, Zhong G, Yu B, Hu W, Dai L, et al: Exon-intron circular RNAs regulate transcription in the nucleus. Nat Struct Mol Biol. 22:256–264. 2015. View Article : Google Scholar : PubMed/NCBI

122 

Sun T, Kalionis B, Lv G, Xia S and Gao W: Role of exosomal noncoding RNAs in lung carcinogenesis. Biomed Res Int. 2015:1258072015. View Article : Google Scholar : PubMed/NCBI

123 

Hood JL, San RS and Wickline SA: Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 71:3792–3801. 2011. View Article : Google Scholar : PubMed/NCBI

124 

Lobb RJ, Lima LG and Moller A: Exosomes: Key mediators of metastasis and pre-metastatic niche formation. Semin Cell Dev Biol. 67:3–10. 2017. View Article : Google Scholar : PubMed/NCBI

125 

Luga V, Zhang L, Viloria-Petit AM, Ogunjimi AA, Inanlou MR, Chiu E, Buchanan M, Hosein AN, Basik M and Wrana JL: Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell. 151:1542–1556. 2012. View Article : Google Scholar : PubMed/NCBI

126 

Peinado H, Alečković M, Lavotshkin S, Matei I, Costa-Silva B, Moreno-Bueno G, Hergueta-Redondo M, Williams C, García-Santos G, Ghajar C, et al: Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat Med. 18:883–891. 2012. View Article : Google Scholar : PubMed/NCBI

127 

Tominaga N, Kosaka N, Ono M, Katsuda T, Yoshioka Y, Tamura K, Lötvall J, Nakagama H and Ochiya T: Brain metastatic cancer cells release microRNA-181c-containing extracellular vesicles capable of destructing blood-brain barrier. Nat Commun. 6:67162015. View Article : Google Scholar : PubMed/NCBI

128 

Fong MY, Zhou W, Liu L, Alontaga AY, Chandra M, Ashby J, Chow A, O'Connor ST, Li S, Chin AR, et al: Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat Cell Biol. 17:183–194. 2015. View Article : Google Scholar : PubMed/NCBI

129 

Szajnik M, Czystowska M, Szczepanski MJ, Mandapathil M and Whiteside TL: Tumor-derived microvesicles induce, expand and up-regulate biological activities of human regulatory T cells (Treg). PLoS One. 5:e114692010. View Article : Google Scholar : PubMed/NCBI

130 

van der Vos KE, Abels ER, Zhang X, Lai C, Carrizosa E, Oakley D, Prabhakar S, Mardini O, Crommentuijn MH, Skog J, et al: Directly visualized glioblastoma-derived extracellular vesicles transfer RNA to microglia/macrophages in the brain. Neuro Oncol. 18:58–69. 2016. View Article : Google Scholar

131 

Zhang L, Zhang S, Yao J, Lowery FJ, Zhang Q, Huang WC, Li P, Li M, Wang X, Zhang C, et al: Microenvironment-induced PTEN loss by exosomal microRNA primes brain metastasis outgrowth. Nature. 527:100–104. 2015. View Article : Google Scholar : PubMed/NCBI

132 

Esposito CL, Quintavalle C, Ingenito F, Rotoli D, Roscigno G, Nuzzo S, Thomas R, Catuogno S, de Franciscis V and Condorelli G: Identification of a novel RNA aptamer that selectively targets breast cancer exosomes. Mol Ther Nucleic Acids. 23:982–994. 2021. View Article : Google Scholar : PubMed/NCBI

133 

Xie X, Nie H, Zhou Y, Lian S, Mei H, Lu Y, Dong H, Li F, Li T, Li B, et al: Eliminating blood oncogenic exosomes into the small intestine with aptamer-functionalized nanoparticles. Nat Commun. 10:54762019. View Article : Google Scholar : PubMed/NCBI

134 

Dunn GP, Old LJ and Schreiber RD: The immunobiology of cancer immunosurveillance and immunoediting. Immunity. 21:137–148. 2004. View Article : Google Scholar : PubMed/NCBI

135 

Leek RD, Lewis CE, Whitehouse R, Greenall M, Clarke J and Harris AL: Association of macrophage infiltration with angiogenesis and prognosis in invasive breast carcinoma. Cancer Res. 56:4625–4629. 1996.PubMed/NCBI

136 

DeNardo DG, Barreto JB, Andreu P, Vasquez L, Tawfik D, Kolhatkar N and Coussens LM: CD4(+) T cells regulate pulmonary metastasis of mammary carcinomas by enhancing protumor properties of macrophages. Cancer Cell. 16:91–102. 2009. View Article : Google Scholar : PubMed/NCBI

137 

Moaaz M, Lotfy H, Elsherbini B, Motawea MA and Fadali G: TGF-beta Enhances the Anti-inflammatory effect of tumor-infiltrating CD33+11b+HLA-DR myeloid-derived suppressor cells in gastric cancer: A possible relation to MicroRNA-494. Asian Pac J Cancer Prev. 21:3393–3403. 2020. View Article : Google Scholar : PubMed/NCBI

138 

Shitara K and Nishikawa H: Regulatory T cells: A potential target in cancer immunotherapy. Ann N Y Acad Sci. 1417:104–115. 2018. View Article : Google Scholar : PubMed/NCBI

139 

Kumar V, Patel S, Tcyganov E and Gabrilovich DI: The Nature of Myeloid-derived suppressor cells in the tumor microenvironment. Trends Immunol. 37:208–220. 2016. View Article : Google Scholar :

140 

Sarvaiya PJ, Guo D, Ulasov I, Gabikian P and Lesniak MS: Chemokines in tumor progression and metastasis. Oncotarget. 4:2171–2185. 2013. View Article : Google Scholar : PubMed/NCBI

141 

Briukhovetska D, Dorr J, Endres S, Libby P, Dinarello CA and Kobold S: Interleukins in cancer: From biology to therapy. Nat Rev Cancer. 21:481–499. 2021. View Article : Google Scholar :

142 

Manrique-Rincón AJ, Ruas LP, Fogagnolo CT, Brenneman RJ, Berezhnoy A, Castelucci B, Consonni SR, Gilboa E and Bajgelman MC: Aptamer-mediated transcriptional gene silencing of Fox 3 inhibits regulatory T cells and potentiates antitumor response. Mol Ther Nucleic Acids. 25:143–151. 2021. View Article : Google Scholar

143 

Borsig L, Wolf MJ, Roblek M, Lorentzen A and Heikenwalder M: Inflammatory chemokines and metastasis-tracing the accessory. Oncogene. 33:3217–3224. 2014. View Article : Google Scholar

144 

Fridlender ZG, Sun J, Kim S, Kapoor V, Cheng G, Ling L, Worthen GS and Albelda SM: Polarization of tumor-associated neutrophil phenotype by TGF-beta: 'N1' versus 'N2' TAN. Cancer Cell. 16:183–194. 2009. View Article : Google Scholar : PubMed/NCBI

145 

Tan W, Zhang W, Strasner A, Grivennikov S, Cheng JQ, Hoffman RM and Karin M: Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL-RANK signalling. Nature. 470:548–553. 2011. View Article : Google Scholar : PubMed/NCBI

146 

Monteiro AC, Leal AC, Goncalves-Silva T, Mercadante AC, Kestelman F, Chaves SB, Azevedo RB, Monteiro JP and Bonomo A: T cells induce pre-metastatic osteolytic disease and help bone metastases establishment in a mouse model of metastatic breast cancer. PLoS One. 8:e681712013. View Article : Google Scholar

147 

Blomberg OS, Spagnuolo L and de Visser KE: Immune regulation of metastasis: Mechanistic insights and therapeutic opportunities. Dis Model Mech. 11:dmm0362362018. View Article : Google Scholar : PubMed/NCBI

148 

Kitamura T, Qian BZ and Pollard JW: Immune cell promotion of metastasis. Nat Rev Immunol. 15:73–86. 2015. View Article : Google Scholar : PubMed/NCBI

149 

Roth F, De La Fuente AC, Vella JL, Zoso A, Inverardi L and Serafini P: Aptamer-mediated blockade of IL4Rα triggers apoptosis of MDSCs and limits tumor progression. Cancer Res. 72:1373–1383. 2012. View Article : Google Scholar : PubMed/NCBI

150 

Liu H, Mai J, Shen J, Wolfram J, Li Z, Zhang G, Xu R, Li Y, Mu C, Zu Y, et al: A Novel DNA Aptamer for dual targeting of polymorphonuclear myeloid-derived suppressor cells and tumor cells. Theranostics. 8:31–44. 2018. View Article : Google Scholar : PubMed/NCBI

151 

Eberting CL, Shrayer DP, Butmarc J and Falanga V: Histologic progression of B16 F10 metastatic melanoma in C57BL/6 mice over a six week time period: Distant metastases before local growth. J Dermatol. 31:299–304. 2004. View Article : Google Scholar : PubMed/NCBI

152 

Meyer C, Eydeler K, Magbanua E, Zivkovic T, Piganeau N, Lorenzen I, Grötzinger J, Mayer G, Rose-John S and Hahn U: Interleukin-6 receptor specific RNA aptamers for cargo delivery into target cells. RNA Biol. 9:67–80. 2012. View Article : Google Scholar : PubMed/NCBI

153 

Gupta S, Hirota M, Waugh SM, Murakami I, Suzuki T, Muraguchi M, Shibamori M, Ishikawa Y, Jarvis TC, Carter JD, et al: Chemically modified DNA aptamers bind interleukin-6 with high affinity and inhibit signaling by blocking its interaction with interleukin-6 receptor. J Biol Chem. 289:8706–8719. 2014. View Article : Google Scholar : PubMed/NCBI

154 

Berezhnoy A, Stewart CA, McNamara JO II, Thiel W, Giangrande P, Trinchieri G and Gilboa E: Isolation and optimization of murine IL-10 receptor blocking oligonucleotide aptamers using high-throughput sequencing. Mol Ther. 20:1242–1250. 2012. View Article : Google Scholar :

155 

Levay A, Brenneman R, Hoinka J, Sant D, Cardone M, Trinchieri G, Przytycka TM and Berezhnoy A: Identifying high-affinity aptamer ligands with defined cross-reactivity using high-throughput guided systematic evolution of ligands by exponential enrichment. Nucleic Acids Res. 43:e822015. View Article : Google Scholar : PubMed/NCBI

156 

Yoon S, Huang KW, Andrikakou P, Vasconcelos D, Swiderski P, Reebye V, Sodergren M, Habib N and Rossi JJ: Targeted Delivery of C/EBPalpha-saRNA by RNA aptamers shows anti-tumor effects in a mouse model of advanced PDAC. Mol Ther Nucleic Acids. 18:142–154. 2019. View Article : Google Scholar :

157 

Camorani S, Passariello M, Agnello L, Pedone E, Pirone L, Chesta CA, Palacios RE, Fedele M and Cerchia L: Aptamer targeted therapy potentiates immune checkpoint blockade in triple-negative breast cancer. J Exp Clin Cancer Res. 39:1802020. View Article : Google Scholar :

158 

Quirico L, Orso F, Esposito CL, Bertone S, Coppo R, Conti L, Catuogno S, Cavallo F, de Franciscis V and Taverna D: Axl-148b chimeric aptamers inhibit breast cancer and melanoma progression. Int J Biol Sci. 16:1238–1251. 2020. View Article : Google Scholar : PubMed/NCBI

159 

Shelley G, Dai J, Keller JM and Keller ET: Pheno-SELEX: Engineering Anti-metastatic aptamers through targeting the invasive phenotype using systemic evolution of ligands by exponential enrichment. Bioengineering (Basel). 8:2122021. View Article : Google Scholar

160 

Li T, Li Y, Rehmani H, Guo J, Padia R, Calbay O, Ding Z, Jiang Y, Jin L and Huang S: Attenuated miR-203b-3p is critical for ovarian cancer progression and aptamer/miR-203b-3p chimera can be explored as a therapeutic. Adv Cancer Biol-Metastasis. 4:1000312022. View Article : Google Scholar

161 

Liu K, Xie F, Zhao T, Zhang R, Gao A, Chen Y, Li H, Zhang S, Xiao Z, Li J, et al: Targeting SOX2 protein with peptide aptamers for therapeutic gains against esophageal squamous cell carcinoma. Mol Ther. 28:901–913. 2020. View Article : Google Scholar : PubMed/NCBI

162 

Lincoff AM, Mehran R, Povsic TJ, Zelenkofske SL, Huang Z, Armstrong PW, Steg PG, Bode C, Cohen MG, Buller C, et al: Effect of the REG1 anticoagulation system versus bivalirudin on outcomes after percutaneous coronary intervention (REGULATE-PCI): A randomised clinical trial. Lancet. 387:349–356. 2016. View Article : Google Scholar

163 

Bruno JG: Potential inherent stimulation of the innate immune system by nucleic acid aptamers and possible corrective approaches. Pharmaceuticals (Basel). 11:622018. View Article : Google Scholar

164 

Verhoef JJ, Carpenter JF, Anchordoquy TJ and Schellekens H: Potential induction of anti-PEG antibodies and complement activation toward PEGylated therapeutics. Drug Discov Today. 19:1945–1952. 2014. View Article : Google Scholar : PubMed/NCBI

165 

Ganson NJ, Povsic TJ, Sullenger BA, Alexander JH, Zelenkofske SL, Sailstad JM, Rusconi CP and Hershfield MS: Pre-existing anti-polyethylene glycol antibody linked to first-exposure allergic reactions to pegnivacogin, a PEGylated RNA aptamer. J Allergy Clin Immunol. 137:1610–1613.e7. 2016. View Article : Google Scholar

166 

Shen W, De Hoyos CL, Sun H, Vickers TA, Liang XH and Crooke ST: Acute hepatotoxicity of 2′fluoro-modified 5-10-5 gapmer phosphorothioate oligonucleotides in mice correlates with intracellular protein binding and the loss of DBHS proteins. Nucleic Acids Res. 46:2204–2217. 2018. View Article : Google Scholar : PubMed/NCBI

167 

Burdick AD, Sciabola S, Mantena SR, Hollingshead BD, Stanton R, Warneke JA, Zeng M, Martsen E, Medvedev A, Makarov SS, et al: Sequence motifs associated with hepatotoxicity of locked nucleic acid-modified antisense oligonucleotides. Nucleic Acids Res. 42:4882–4891. 2014. View Article : Google Scholar : PubMed/NCBI

168 

Penedones A, Mendes D, Alves C and Batel Marques F: Safety monitoring of ophthalmic biologics: A systematic review of pre- and postmarketing safety data. J Ocul Pharmacol Ther. 30:729–751. 2014. View Article : Google Scholar : PubMed/NCBI

169 

Tessier Y, Achanzar W, Mihalcik L, Amuzie C, Andersson P, Parry JD, Moggs J and Whiteley LO: Outcomes of the European federation of pharmaceutical industries and associations oligonucleotide Working Group Survey on Nonclinical Practices and Regulatory Expectations for Therapeutic Oligonucleotide Safety Assessment. Nucleic Acid Ther. 31:7–20. 2021. View Article : Google Scholar

170 

Kim J, Yao F, Xiao Z, Sun Y and Ma L: MicroRNAs and metastasis: Small RNAs play big roles. Cancer Metastasis Rev. 37:5–15. 2018. View Article : Google Scholar :

171 

Chan SH and Wang LH: Regulation of cancer metastasis by microRNAs. J Biomed Sci. 22:92015. View Article : Google Scholar : PubMed/NCBI

172 

Sczepanski JT and Joyce GF: Specific inhibition of MicroRNA processing using L-RNA aptamers. J Am Chem Soc. 137:16032–16037. 2015. View Article : Google Scholar : PubMed/NCBI

173 

Daei P, Ramezanpour M, Khanaki K, Tabarzad M, Nikokar I, Hedayati Ch M and Elmi A: Aptamer-based targeted delivery of miRNA let-7d to gastric cancer cells as a novel anti-tumor therapeutic agent. Iran J Pharm Res. 17:1537–1549. 2018.PubMed/NCBI

174 

Esposito CL, Catuogno S and de Franciscis V: Aptamer-MiRNA conjugates for cancer cell-targeted delivery. Methods Mol Biol. 1364:197–208. 2016. View Article : Google Scholar

175 

Wang H, Zhao X, Guo C, Ren D, Zhao Y, Xiao W and Jiao W: Aptamer-dendrimer bioconjugates for targeted delivery of miR-34a expressing plasmid and antitumor effects in non-small cell lung cancer cells. PLoS One. 10:e01391362015. View Article : Google Scholar : PubMed/NCBI

176 

Guo JU, Agarwal V, Guo H and Bartel DP: Expanded identification and characterization of mammalian circular RNAs. Genome Biol. 15:4092014. View Article : Google Scholar : PubMed/NCBI

177 

Chen LL and Yang L: Regulation of circRNA biogenesis. RNA Biol. 12:381–388. 2015. View Article : Google Scholar : PubMed/NCBI

178 

Du WW, Yang W, Liu E, Yang Z, Dhaliwal P and Yang BB: Foxo3 circular RNA retards cell cycle progression via forming ternary complexes with p21 and CDK2. Nucleic Acids Res. 44:2846–2858. 2016. View Article : Google Scholar : PubMed/NCBI

179 

Guarnerio J, Bezzi M, Jeong JC, Paffenholz SV, Berry K, Naldini MM, Lo-Coco F, Tay Y, Beck AH and Pandolfi PP: Oncogenic role of Fusion-circRNAs derived from cancer-associated chromosomal translocations. Cell. 165:289–302. 2016. View Article : Google Scholar

Related Articles

Journal Cover

June-2022
Volume 60 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Alhamhoom Y, As Sobeai HM, Alsanea S and Alhoshani A: Aptamer‑based therapy for targeting key mediators of cancer metastasis (Review). Int J Oncol 60: 65, 2022
APA
Alhamhoom, Y., As Sobeai, H.M., Alsanea, S., & Alhoshani, A. (2022). Aptamer‑based therapy for targeting key mediators of cancer metastasis (Review). International Journal of Oncology, 60, 65. https://doi.org/10.3892/ijo.2022.5355
MLA
Alhamhoom, Y., As Sobeai, H. M., Alsanea, S., Alhoshani, A."Aptamer‑based therapy for targeting key mediators of cancer metastasis (Review)". International Journal of Oncology 60.6 (2022): 65.
Chicago
Alhamhoom, Y., As Sobeai, H. M., Alsanea, S., Alhoshani, A."Aptamer‑based therapy for targeting key mediators of cancer metastasis (Review)". International Journal of Oncology 60, no. 6 (2022): 65. https://doi.org/10.3892/ijo.2022.5355