60
Views
2
CrossRef citations to date
0
Altmetric
Review

Nucleic acid aptamers for targeting of shRNA-based cancer therapeutics

&
Pages 367-376 | Published online: 07 Dec 2022

Abstract

Aptamers are nucleic acid ligands which have been validated to bind to epitopes with a specificity similar to that of monoclonal antibodies. Aptamers have been primarily investigated for their direct function in terms of inhibition of protein targets; however, recent evidence gives reason to actively explore aptamers as targeting moieties for delivery of anticancer therapeutics. Many aptamers have been developed to bind to extracellular membrane domains of proteins overexpressed on cancer cells and have the potential to be modified for use in targeting cancer therapeutics. The use of DNA vector-based short hairpin RNA (shRNA) for RNA interference (RNAi) is a precise means for the disruption of target gene expression but its clinical usage in cancer is limited by obstacles related to delivery into cancer cells. Nucleic acid aptamers are attractive candidates for targeting of shRNA therapies. Their small size, ease of production and modification, and high specificity are valued attributes in comparison to other targeting moieties currently being tested. Here we review the development of aptamers directed to PSMA, Nucleolin, HER-3, RET, TN-C, and MUC1 and focus on their potential for use in targeting of shRNA-based cancer therapeutics.

Introduction

Delivery vehicles optimal for shRNA delivery ideally are targeted in order to maximize biodistribution to the cancer cell population, thereby increasing their effectiveness in the clinical setting and minimizing potentially adverse off target (ie, nonmalignant) cell effects. Tumor cellular uptake at the target site(s) can be enhanced by conferring to the delivery vehicle the ability to recognize surface features of the target cells. This can be accomplished most readily by recognizing surface antigens or promoting receptor mediated endocytosis. Delivery vehicles can be complexed with monoclonal antibodies, peptides, small molecule ligands, and aptamers to recognize cell surface markers (CitationLu and Low 2002; CitationDyba et al 2004; CitationStefanidakis and Koivunen 2004; CitationHughes and Rao 2005).

Aptamers are noncoding nucleic acid oligomers made from either RNA or DNA. Much like tRNA, they fold through intramolecular bonds into varied tertiary structures. The folded structure of aptamers creates binding pockets that allow the molecules to interact with the three dimensional structure of specific domains on a protein’s surface. Aptamers bind to epitopes with affinity comparable to that of monoclonal antibodies. Their dissociation constants are often in the picomolar to low nanomolar range. Because of their high surface area, the area in contact with the target is large, minimizing the chances of off target effects by increasing binding specificity. In some cases aptamers can inhibit the function of a target protein by binding to its active site, or inducing a conformational change, or inhibiting dimerization or DNA binding (CitationLupold et al 2002; CitationUmehara et al 2004; CitationShi et al 2007).

The idea of developing nucleic acid ligands for therapeutic purposes was born out of the study of activation of viral gene expression during HIV infection. There exists a nucleic acid ligand that binds specifically to a transactivator of human immunodeficiency virus type 1 (HIV-1) gene called Tat. The Tat protein normally binds to an RNA stem-loop structure called a trans-acting response element (TAR) which occurs within the first 60 nucleotides of all HIV-1 transcripts, thus activating expression of viral genes. A series of studies showed that overexpression of TAR sequence decoys conferred HIV-1 resistance to cells, presumably because the TAR oligonucleotides bound specifically to the Tat protein, thus inhibiting it from activating viral gene expression (CitationSullenger et al 1990; CitationSullenger et al 1991; CitationHeld et al 2006).

Since this initial discovery, a wide range of potential therapeutic applications for aptamers have been proposed and pursued (CitationBunka and Stockley 2006). Aptamers have been successfully developed against extracellular protein ligands, such as transforming growth factor receptors, platelet-derived growth factor, basic fibroblast growth factor, and vascular endothelial growth factor (VEGF) as well as intracellular proteins such as NFkb (CitationJellinek et al 1995; CitationGolden et al 2000; CitationCassiday and Maher 2001; CitationPietras et al 2001; CitationOhuchi et al 2005, Citation2006; CitationMcCauley et al 2006). Pegaptanib, an aptamer that binds to and inhibits VEGF has been approved by the FDA for treatment of age related macular generation (CitationNg et al 2006). Aptamers that bind to blood coagulation factors VIIa and IXa are being investigated as anticoagulants (CitationRusconi et al 2002). A subset of aptamers that have been developed for therapeutic purposes have the potential to be used as targeting moieties for shRNA.

Aptamers represent an attractive alternative to traditional methods of targeting cancer gene therapy by circumventing many of the stumbling blocks that inhibit the effectiveness of these methods. Aptamers are smaller than monoclonal antibodies. With a molecular weight of 10–15 kDa they are one order of magnitude lighter than antibodies used for targeting, which have an average molecular weight of 150 kDa (CitationWhite et al 2000). Aptamers are also less immunogenic than antibodies (CitationDrolet et al 2000; CitationPestourie et al 2005; CitationApte et al 2007), making them more suited to systemic administration and long term therapy.

Aptamers can facilitate cell entry of an shRNA delivery vehicle in similar ways as traditional targeting moieties. Aptamers to the extracellular domains of transmembrane receptor proteins can penetrate the cell membrane by receptor mediated endocytosis (CitationHughes and Rao 2005). When aptamers bind to cell surface proteins they increase the proximity of the delivery vehicle and the cell membrane. This facilitates charge interactions between a positively charged delivery vehicle and the negatively charged membrane that result in internalization of the payload. Here we will briefly review the process of aptamer development and then give examples of the current development of aptamers directed to good candidate proteins for tumor targeting.

Here we review the development of aptamers directed to PSMA, Nucleolin, HER-3, RET, TN-C, and MUC1 and focus on their potential for use in targeting of shRNA-based cancer therapeutics. Though most of these aptamers have not yet been used as shRNA targeting moieties in vivo this selection of aptamers represents a group that is well suited to the targeting shRNA-based cancer therapies thereby enhancing their efficiency and safety.

Aptamer production

RNA and DNA aptamers with specific binding properties can now be amplified in vitro from libraries through a streamlined, automated iterative selection process. Because of their small, relatively simple structure, the chemical modification of aptamers to increase serum stability or bioavailability can be accomplished readily. The production and modification of aptamers is very uncomplicated when compared to the production of monoclional antibodies, which require prokaryotic or eukaryotic expression systems. The chemical methods involved are well defined and easily conformable to good manufacturing process requirements, which has eased the transition of aptamers into the clinical setting.

Aptamers that bind to a specific target are isolated from a group of candidate oligonucleotides, either RNA or DNA, through ‘systematic evolution of ligands by exponential enrichment’ (SELEX) (CitationTuerk and Gold 1990). SELEX is an in vitro process that can be automated to allow the rapid production of aptamers (CitationCox et al 2002; CitationEllington et al 2005).

The SELEX process begins with a degenerate sequence library of ≥1015 molecules. With a library of this size it is likely that there is a molecule that will bind to almost any target (CitationEllington and Szostak 1990). The library is then treated with the target molecule at physiological pH and temperature. After some aptamers bind the aptamer–target complexes are partitioned from the nonbinding aptamers and bound species are regenerated and amplified by PCR in the case of DNA aptamers or RT-PCR in the case of RNA aptamers. The amplified molecules are then subjected to another round of the selection process. Over this repetitive process of binding and amplification the pool is enriched for aptamers that bind most efficiently to the target (CitationGopinath 2007). SELEX can also be tailored by ligation and cleavage of small 10nt fixed primer sites before and after amplification. These smaller primer sequences allow the isolation of shorter aptamer sequences which can later be more readily synthesized chemically (CitationVater et al 2003).

Whole-cell SELEX is a procedure that has been developed for the selection of aptamers to extracellular matrix proteins and transmembrane proteins. It involves the introduction of cells overexpressing the protein target of interest to oligomer libraries. This procedure offers the advantage of selecting molecules more suited to recognizing their target in its natural glycosylation state under physiological conditions. Because of the great variety of proteins of the cell surface, a second set of traditional SELEX with the purified protein is sometimes necessary to obtain a purified specific aptamer product (CitationHicke et al 2001; CitationCerchia et al 2005; CitationOhuchi et al 2005). This method has been used to isolate aptamers that bind to human transforming growth factor-beta type III receptor displayed on cell surface (CitationOhuchi et al 2005, Citation2006).

Naked nucleotide oligomers are in general very unstable in the blood and are unsuitable for therapeutic or targeting applications. One study showed that an unmodified aptamer against thrombin had an in vivo serum half-life of only 108 seconds (CitationGriffin et al 1993). A variety of chemical modifications can be applied to nucleic acid aptamers during and after the SELEX process in order to increase their suitability for systemic tumor targeting (). Natural nucleotides within aptamers can be substituted with 2′ fluoro, O-methyl, or amino modified or 4′ thio modified nucleotides that are poor substrates for nuclease degradation (CitationBeigelman et al 1995; CitationGold et al 1995; CitationBurmeister et al 2005; CitationKato et al 2005) 3′ end caps can also be modified to confer nuclease resistance (CitationBurmeister et al 2005). Aptamers can be bound to bulky lipids or polymers to reduce their renal clearance and increase their serum half-life (CitationBoomer et al 2005). Aptamer intramolecular bonds can be altered to increase structural stability (CitationSchmidt et al 2004). Beyond chemical modifications to increase stability, aptamers can also be modified with small functional groups to facilitate their attachment as targeting moieties to polymeric or lipid shRNA delivery vehicles. Amino groups have been added to aptamers in order to facilitate their conjugation to a nanoparticle composed of poly(lactic acid)-block-polyethylene glycol copolymer with a terminal carboxylic acid functional group (CitationFarokhzad et al 2004).

Table 1 Common chemical modifications of aptamers for tumor targeting

Likely candidates for aptamer mediated cancer targeting

The following cell surface proteins are good candidates for tumor targeting. Aptamers are actively being developed for all of them, whether for therapeutic or for targeting purposes ().

Table 2 Nucleotide aptamers in development with potential for use in targeting of shRNA therapy

Prostate-specific membrane antigen (PSMA)

The most extensively studied aptamer for cancer targeting binds to the prostate-specific membrane antigen (PSMA). PSMA is a well described protein that is expressed in prostate cancer epithelial cells (CitationIsraeli et al 1994; CitationMurphy et al 1998). Its use in targeting of many different types of cancer therapeutics is a constantly developing field. In normal prostate epithelial cells, it is primarily expressed as the intracellular protein referred to as PSM’ but in prostate carcinoma the splice variant termed PSMA is overexpressed. PSMA, which is more pertinent for targeting purposes, is a type 2 integral membrane glycoprotein folate carboxypeptidase expressed exclusively in prostate acinar epithelium (CitationSu et al 1995). Increased PSMA expression has been documented in prostatic intraepithelial neoplasia, prostatic adenocarcinoma, and in tumor associated neovasculature. Its expression generally increases with disease progression and metastasis (CitationBostwick et al 1998).

PSMA was originally described as the target for mAb 7E11in the androgen-dependent LNCaP human prostatic adenocarcinoma cell line (CitationHoroszewicz et al 1987). Antibodies that bind to the extracellular domain of the PSMA are used clinically for imaging and staging of prostate cancer (CitationFreeman et al 2002) and are in clinical trials to determine therapeutic potential (CitationBander et al 2005). Monoclonal antibodies to PSMA have also been used to target delivery of gene therapy in vivo (CitationMoffatt et al 2006).

The practicality of PSMA as a target for various kinds of anticancer therapies has placed it in center stage of aptamer-based cancer targeting research. The first reported RNA aptamers selected to bind a tumor-associated membrane antigen were two 2′-fluoro-pyrimidine RNA oligomers that bind to the extracellular domain of the PSMA. These two aptamers, called xPSM-A9 and xPSM-A10, which share no consensus sequences, bind to the PSMA at different extracellular epitopes. Aptamer xPSM-A9 was shown to inhibit the PSMA noncompetitively with an average K(i) of 2.1 nM, while aptamer xPSM-A10 inhibits the PSMA competitively with an average K(i) of 11.9 nM (CitationLupold et al 2002).

The nuclease resistant xPSMA-A10 aptamer has been exploited to target the delivery of various nanoparticulate drug delivery systems. It has peen particularly well studied as a targeting moiety for biodegradable nanoparticles conjugated with chemotherapeutics. In one study, the xPSMA-A10 aptamer was bound to a poly(lactic acid)-block-polyethylene glycol (PEG) copolymer with a terminal carboxylic acid functional group (PLA-PEG-COOH) and used to deliver rhodamine-labeled dextran as a model drug in vitro. This study showed that the targeted nanoparticles preferentially bind to and are taken up in vitro by LNCaP prostate epithelial cells which express PSMA, but not by PC3 prostate epithelial cells, which do not express detectable levels of the PSMA (CitationFarokhzad et al 2004).

In a more recent study, 5′-NH2modified xPSMA-A10 aptamers were conjugated to poly(D,L-lactic-co-glycolic acid)-block-poly(ethylene glycol) nanoparticles with terminal carboxylic acid groups (PLGA-PEG-COOH). These nanoparticles were then encapsulated with the chemotherapeutic docetaxel and tested in vitro and in vivo. The docetaxel-encapsulated nanoparticle-aptamer (Dtxl-NP-Apt) bioconjugates enhanced in vitro cellular toxicity with LNCaP prostate epithelial cells as compared with nanoparticles that lack the PSMA aptamer. After a single intratumoral injection of Dtxl-NP-Apt bioconjugates using a murine LNCaP xenograft model of prostate cancer, complete tumor reduction was noted in five of seven nude mice. 100% of the mice that received the Dtxl-NP-Apt bioconjugate survived the 109-day study. In contrast, only two of seven mice that received the non-targeted Dtxl-NP had complete tumor reduction and only 57% of the mice in this group survived the 109-day study. Only 14% of mice treated with Dtxl alone survived the 109-day study (CitationFarokhzad et al 2006). A similar study tested PLGA-PEG-COOH-xPSMA-A10 conjugates encapsulating docetaxel and 14C labeled paclitaxel. It was found that the surface association of nanoparticles with targeting aptamer significantly enhanced delivery to tumors when compared to equivalent but nontargeted nanoparticles in a murine LNCaP xenograft model (CitationCheng et al 2007).

PSMA aptamers are particularly attractive as targeting moities for RNAi-based cancer therapeutics because the PSMA itself participates in membrane recycling, becoming internalized through ligand induced endocytosis, thus facilitating entry of the therapeutic payload into the cytoplasm (CitationGhosh and Heston 2004). This property of the PSMA has been exploited in the targeting of therapeutic siRNAs to prostate cancers using aptamer-siRNA chimeras and conjugates. In one study, a 27mer anti-Lamin siRNA was noncovalently bound to xPSMA-A9 aptamers using a modular streptavidin bridge and tested in vitro to transfect PC3 cells and LNCaP cells that naturally expressed the PSMA. The conjugates were successfully transfected with this method and inhibition of lamin expression was achieved. The conjugates displayed similar transfection efficiency to oligofectamine controls but better specificity, as they were only taken up by PSMA expressing LNCaP cells and not PSMA-negative PC3 cells (CitationChu et al 2006).

In another study, aptamer-siRNA chimeras were used to target prostate cancer cells and knock out expression of survival genes. Chimeras of the xPSMA-A10 aptamer and polo-like kinase 1 (PLK1) and BCL2 siRNAs were generated via in vitro transcription from DNA templates. The chimeras were shown be internalized and promote apoptosis preferentially in PSMA expressing LNCaP cells and not PC3 control cells. This specificity was retained in murine LNCaP xenograft tumor models where chimeras bearing PLK1 siRNAs mediated pronounced tumor regression while having no statistically significant effect on non-PSMA expressing PC3 xenografts (CitationMcNamara et al 2006).

Nucleolin

An aptamer (AS1411) that binds to the external domain of the membrane protein nucleolin is has been well characterized because of its therapeutic potential. Due to the properties of the aptamer and the properties of nucleolin itself AS1411 is an excellent candidate for aptamer mediated targeting of gene therapy.

Human nucleolin is an abundant multifunctional 76 kDa membrane associated phosphoprotein that has 707 amino acids in its sequence (CitationDerenzini et al 1995; CitationSarcevic et al 1997). It is composed of three main domains, an acidic histone-like NH2 terminus, a central domain containing four RNA-binding sub-domains, and a carboxy terminal domain that contains its four sites of phosphorylation (CitationLapeyre et al 1987; CitationGinisty et al 1999). Though it was originally characterized as a nucleolar protein, nucleolin is also present on the external cell membrane where it functions as a signaling receptor of poorly defined function (CitationHovanessian et al 2000; CitationStorck et al 2007).

Though its function on the surface of cancer cells has not been clearly defined, Nucleolin has been extensively investigated in the context of cancer. It has been shown to be associated with survival, growth and proliferation of cells, nuclear transport, transcription, packing and transport of rRNA, and replication and recombination of DNA (CitationSrivastava and Pollard 1999). In addition, overexpression of nucleolin is related to poor clinical prognosis for some cancer types (CitationDerenzini et al 1995; CitationDerenzini 2000).

One pathophysiologic function of cell surface nucleolin that makes it ideal for targeting anticancer therapies is its involvement in internalization of specific ligands. Nucleolin has been shown to bind to and internalize the heparin-binding growth factor midkine (CitationSaid et al 2002), the iron-binding lycoprotein lactoferrin (CitationLegrand et al 2004), acharan sulfate (CitationJoo et al 2005), as well as the tumor-homing 34-amino acid peptide (F3) (CitationChristian et al 2003).

The aptamer known as AS1411 was first developed as AGRO100 by Aptamera (Louisville, KY). It is an unmodified guanosine rich 26-mer, which binds to nucleolin. AS1411 was originally investigated because of observations that guanosine-rich oligonucleotides (GRO) had nonantisense-based in vitro antiproliferative properties against cancer cells (CitationBates et al 1999). These oligonucleotides were further studied and it was found that the unmodified DNA phosphodiester GRO analog GRO29A-OH, which retains the G quartet structure essential for nucleolin binding (CitationDapic et al 2003), displayed resistance against serum nuclease degradation (CitationDapic et al 2002). AS1411 is simply GRO29A-OH with its three 5′ thymidines cleaved.

A nontraditional aptamer because of its G quartet-based binding to its target, AS1411 has been shown to bind to cell surface nucleolin and inhibit growth of many cancer cell lines in vitro such as the prostate cancer cell line DU145, the breast cancer cell lines MDA-MB-231 and MCF-7, the lung cancer cell line A549, as well as the cervical cancer cell line HeLa (CitationBates et al 1999; CitationDapic et al 2002).

In anticipation of its use as a therapeutic, biodistribution studies were realized to characterize the in vivo pharmacodymanics, distribution, and metabolism of the aptamer following IV administration. AS1411 metabolism was studied in mice bearing lung and renal human tumor xenografts derived from A549 and A498 cells using IV administration of AS1411 radiolabeled with tritiated thymidine. Following 1, 10, and 25 mg/kg IV boluses of [3H]AS1411, 63% of the radiolabeled aptamer was excreted in urine within 5 hours while less than 1% of the injected dose was detected upon analysis of bile, thus suggesting that the main mode of metabolism is renal. This same study also showed that AS1411 displayed dose related pharmacokinetics with an elimination half life of 2 days in plasma and whole blood. Regardless of the dose administered, tumor/blood ratios were 4:6 for the renal carcinoma xenograft and 2:4 and for the lung carcinoma xenograft, showing that the aptamer effectively accumulated in tumor tissue (CitationIreson and Kelland 2006).

In vivo xenograft efficacy studies have collectively shown that AS1411 results in cytostasis rather than cytotoxicity. After treatment with AS1411 cells are blocked at S-phase. Because of this observation, there has been interest in combining AS1411 with nucleoside analogs such as gemcitabine, which act on cells during S-phase. Early in vitro and in vivo studies of AS1411 in combination with gemcitabine against PANC-1 pancreatic cancer cells have shown enhanced anti-tumor activity in comparison to either agent alone (CitationIreson and Kelland 2006).

Preclinical toxicology of AS1411 has been evaluated in rats and dogs with IV bolus dosages of up to 100 mg/kg in rats and 96-hour continuous IV infusions at doses up to 10 mg/kg/d. No significant toxicity was observed in either species, as determined by clinical observations, clinical pathology evaluation, and gross examination of tissues at necroscopy (CitationIreson and Kelland 2006).

AS1411 was the first nucleic acid-based aptamer approved for Phase I clinical testing for the treatment of cancer in humans. Results have been published on the progress of 17 patients with advanced solid tumors receiving AS1411 at the Brown Cancer Center in Louisville, KY (CitationLaber et al 2005). This report describes a continuous IV infusion dose escalation study starting from 1 mg/kg/d and thus far reported up to 10 mg/kg/d. Fifteen patients received a 4 day infusion while 2 patients received a 7 day infusion. No serious toxicity related to AS1411 administration was observed. A patient with renal cancer achieved a sustained partial response at 16 months post-treatment and 41% of patients were reported to have stable disease at 2 months post-treatment. From these initial results the Phase I study has continued with a focus on renal and non–small cell lung cancers (CitationIreson and Kelland 2006; CitationLaber et al 2006).

The extensive preclinical and burgeoning clinical data amassed about AS1411 makes it an ideal candidate for targeting of cancer gene therapy in humans.

Human epidermal growth factor receptor 3 (HER-3)

Human epidermal growth factor receptor 3 (HER-3) is a receptor tyrosine kinase that is overexpressed on the surface of many different cancer cells (CitationYokota et al 1988; CitationSchneider et al 1989; CitationSlamon et al 1989; CitationBerchuck et al 1990; CitationDougall et al 1993). The extra-cellular domains of receptor tyrosine kinases are rational and accessible targets for anticancer therapies. The most prominent example of a therapy targeted to the extra-cellular domain of a receptor tyrosine kinase is Herceptin, a humanized monoclonal antibody that targets the extra-cellular domain of HER-2. This molecule has proved effective in the treatment of HER2-overexpressing breast cancers (CitationPegram et al 1999).

An aptamer to the extracellualar domain of HER-3, called A30, has been isolated using SELEX. A30 was demonstrated to have high binding specificity to HER-3, despite the sequence similarities shared by the extracellular domains of HER-2 and HER-3. In vitro, A30 inhibits the growth stimulatory effects of HER-3’s natural ligand, heregulin, though it does not compete for its binding site. A30 also inhibits HER-3 from binding to HER-2 which is a necessary step in HER-mediated growth induction (CitationChen et al 2003).

Though A30 is still in the early stages of development and would likely need to undergo modifications to enhance its serum stability prior to its introduction in the clinical setting, it represents a possible future targeting moiety for shRNA delivery in the therapy of cancer.

RET

Another receptor tyrosine kinase for which an aptamer has been developed is the RET oncoprotein. Mutated forms of the RET Receptor tyrosine kinase are implicated as the cause of the dominant inherited cancer syndrome MEN2 (CitationHansford and Mulligan 2000). In MEN2, germline to ‘missense mutations of the RET gene result in the conversion of cysteine 634 to tyrosine (RETC634Y), which causes cancers of the neuroendocrine organs. These mutations affect the cysteine rich extracellular domain of RET causing the loss of an intramolecular disulfide bond. The loss of this bond allows the formation dimmers of mutated RET monomers via an intermolecular disulfide bond. Dimerization results in a constitutively activated receptor (CitationIwashita et al 1999).

Using 16 cycles of whole-cell SELEX several RNA aptamers that bind to the extracellular domain of RETC634Y have been isolated. PC12 cells which expressed the mutant receptor were used for the selection procedure. The aptamer with the best signaling inhibition and the most nuclease resistance, termed D4, was selected for further development (CitationCerchia et al 2005).

An aptamer that binds specifically to RET could be useful for targeting of specific anticancer gene therapies and it also is another example of the use of whole-cell SELEX to effectively develop aptamers that bind to transmembrane proteins in their natural glycosylation states. This method could be employed to develop aptamers to other potential targets.

Tenascin-C (TN-C)

Tenascin-C is a hexameric extracellular matrix protein that is highly expressed during active tissue development processes including wound healing, angiogenesis, embryonic development, and tumor growth (CitationMackie et al 1988; CitationErickson and Bourdon 1989; CitationKoukoulis et al 1991; CitationZagzag et al 1995). TN-C is an ideal candidate for targeting of anticancer shRNA therapy because it is highly expressed in tumor and in general its expression is expression is limited in normal tissues (CitationBorsi et al 1992). High TN-C expression has been noted in osteosarcomas, lymphomas, glioblastomas, melanomas as well as carcinomas of breast, colon lung, and prostate (CitationChiquet-Ehrismann 2004; CitationOrend 2005; CitationOrend and Chiquet-Ehrismann 2006).

Using SELEX with TN-C expressing U251 glioblastoma cells RNA aptamers that bind to the extracellular fibrinogen-like domain of TN-C with high affinity were isolated. Several modifications were made to the aptamer in order to increase its usefulness as a targeting moiety. Its size was reduced in order to increase tissue penetration and efficiency of synthesis. It was 2′-OCH3 substituted, and 3′ capped in order to increase nuclease stability. A 5′ amine was also added to serve as a conjugation site. After these modifications the resultant aptamer, termed TTA1, had an increased equilibrium Kd but the final value of 5 × 10−9M still represents a very high affinity (CitationHicke et al 2001).

In a later experiment the potential of TTA1 to be used for in vivo drug delivery was evaluated. Uptake and tumor distribution of rhodamine red-X-labeled TTA1 was studied using fluorescence microscopy in a U251 human glioblastoma cell line xenograft nude mouse model. Within 10 minutes after IV injection of the fluorescently labeled aptamer, bright perivascular fluorescence was noted in the xenografts. The fluorescence then diffused throughout the tumor stroma over the following 3 hours (CitationHicke et al 2006).

Biodistribution and radioimaging studies were performed using (99m)Tc labeled TTA1 and glioblastoma (U251) and breast cancer (MDA-MB-435) tumor xenografts in nude mice. IV administered (99m)Tc labeled TTA1 showed rapid blood clearance with a serum half-life of less than 2 min, and rapid tumor penetration. TTA1 was cleared much more rapidly from the blood than from the tumor. Both renal and hepatobiliary clearance pathways were observed, but it is estimated that due to rapid nuclease degredation of the aptamer the clearance patterns observed were those of the aptamer metabolites. In these studies it was found that TTA1 affectively targeted a wide variety of TN-C expressing tumor types including colon (SW620), breast (MDA-MB-468, MDA-MB-435), glioblastoma (U251), rhabdomyosarcoma (A673). As a control TTA1 was also tested with a squamous cell carcinoma of the head and neck (KB) xenograft that did not express TN-C. As expected, KB xenografts did not display appreciable aptamer uptake (CitationHicke et al 2006).

The results of these studies are promising in terms of TTA1s potential use in tumor targeting. Before its use in targeting of shRNA-based cancer therapies it would probably need further modifications to increase its serum half life. While short half lives and rapid elimination are beneficial for minimizing potential toxicity, such a short serum half life may inhibit aptamer penetration in areas of the target tumor that are poorly perfused (CitationHicke et al 2006).

MUC1

MUC1 is a large, highly glycosylated transmembrane epithelial cell surface protein. Mucin proteins are expressed on the apical membrane of various epithelial cell types. They have many roles which facilitate the function of mucosal cells such as lubrication of epithelial cell surfaces, prevention of tissue dehydration, protection of cells from proteolytic degradation and provision of a barrier against infection (Citationvon Mensdorff-Pouilly et al 2000). Mucins also function as signal transduction receptors involved in triggering cellular responses such as proliferation, differentiation, and apoptosis (CitationTaylor-Papadimitriou et al 1999; CitationHollingsworth and Swanson 2004). MUC1 overexpression has been well documented in many human adenocarcinomas including breast, pancreatic, ovarian, colorectal, lung, prostate, esophageal (CitationMaeshima et al 1997; CitationAoki et al 1998; CitationZhang et al 1998; CitationHough et al 2000; CitationGinestier et al 2002; CitationKohlgraf et al 2003; CitationBurjonrappa et al 2007). Its expression has also been documented in other types of tumors including astrocytoma, melanoma, neuroblastoma, as well as hematological malignancies (CitationOosterkamp et al 1997; CitationBrossart et al 2001).

The cell surface expression pattern of MUC1 across human malignancies makes it an excellent and versatile target for cancer targeting with an aptamer. Using 10 rounds of in vitro SELEX, a group of DNA aptamers has been isolated that binds selectively to synthetic MUC1 peptides. This pool of aptamers, when fluorescently labeled has been verified to bind specifically to MUC1 expressing cancer cell lines in vitro (CitationFerreira et al 2006). In a later study this group of aptamers was demonstrated by radio-imaging to have good tumor penetration in a murine xenograft (CitationPerkins and Missailidis 2007). These aptamers are in the early stages of development and before their transition into the clinic they would need further modification to minimize size and maximize nuclease resistance.

Conclusions

Aptamer development could ameliorate many of the traditional problems with the introduction of exogenous genetic material for therapeutic purposes. There is a wide range of experimentation occurring, from the laboratory to the preclinical setting, which has the potential to revolutionize the targeting of gene-based therapy. The aptamers reviewed here represent the most promising areas of current development pertinent to aptamer-based targeting of shRNA cancer therapeutics and will likely be among the first to make transition to targeting in the clinic.

Acknowledgment

Thank you to the Newsome Fund for their financial support.

References

  • AokiRTanakaS1998MUC-1 expression as a predictor of the curative endoscopic treatment of submucosally invasive colorectal carcinomaDis Colon Rectum41101262729788390
  • ApteRModiMMasonsonHfor the Macugen AMD Study Group2007Pegaptanib 1-year systemic safety results from a safety-pharmacokinetic trial in patients with neovascular age-related macular degenerationOphthalmology114917021217509689
  • BanderNHMilowskyMINanusDM2005Phase I trial of 177lutetium-labeled J591, a monoclonal antibody to prostate-specific membrane antigen, in patients with androgen-independent prostate cancerJ Clin Oncol2321459160115837970
  • BatesPJKahlonJBThomasSD1999Antiproliferative activity of G-rich oligonucleotides correlates with protein bindingJ Biol Chem27437263697710473594
  • BeigelmanLMcSwiggenJADraperKG1995Chemical modification of hammerhead ribozymes. Catalytic activity and nuclease resistanceJ Biol Chem270432570287592749
  • BerchuckAKamelAWhitakerR1990Overexpression of HER-2/neu is associated with poor survival in advanced epithelial ovarian cancerCancer Res50134087911972347
  • BoomerRMLewisSDHealyJM2005Conjugation to polyethylene glycol polymer promotes aptamer biodistribution to healthy and inflamed tissuesOligonucleotides1531839516201906
  • BorsiLCarnemollaBNicoloG1992Expression of different tenascin isoforms in normal, hyperplastic and neoplastic human breast tissuesInt J Cancer525688921385335
  • BostwickDGPacelliABluteM1998Prostate specific membrane antigen expression in prostatic intraepithelial neoplasia and adenocarcinoma: a study of 184 casesCancer82112256619610707
  • BrossartPSchneiderADillP2001The epithelial tumor antigen MUC1 is expressed in hematological malignancies and is recognized by MUC1-specific cytotoxic T-lymphocytesCancer Res611868465011559560
  • BunkaDHStockleyPG2006Aptamers come of age – at lastNat Rev Microbiol485889616845429
  • BurjonrappaSCReddimasuSNawazZ2007Mucin expression profile in Barrett’s, dysplasia, adenocarcinoma sequence in the esophagusIndian J Cancer4411517401217
  • BurmeisterPELewisSDSilvaRF2005Direct in vitro selection of a 2′-O-methyl aptamer to VEGFChem Biol121253315664512
  • CassidayLAMaherLJ3rd2001In vivo recognition of an RNA aptamer by its transcription factor targetBiochemistry4082433811327864
  • CerchiaLDucongéFPestourieC2005Neutralizing aptamers from whole-cell SELEX inhibit the RET receptor tyrosine kinasePLoS Biol34e12315769183
  • ChenCHChernisGAHoangVQ2003Inhibition of heregulin signaling by an aptamer that preferentially binds to the oligomeric form of human epidermal growth factor receptor-3Proc Natl Acad Sci USA1001692263112874383
  • ChengJTeplyBASherifiI2007Formulation of functionalized PLGA-PEG nanoparticles for in vivo targeted drug deliveryBiomaterials2858697617055572
  • Chiquet-EhrismannR2004TenascinsInt J Biochem Cell Biol3669869015094113
  • ChristianSPilchJAkermanME2003Nucleolin expressed at the cell surface is a marker of endothelial cells in angiogenic blood vesselsJ Cell Biol1634871814638862
  • ChuTCTwuKYEllingtonAD2006Aptamer mediated siRNA deliveryNucleic Acids Res3410e7316740739
  • CoxJCHayhurstAHesselberthJ2002Automated selection of aptamers against protein targets translated in vitro: from gene to aptamerNucleic Acids Res3020e10812384610
  • DapićVAbdomerovićVMarringtonR2003Biophysical and biological properties of quadruplex oligodeoxyribonucleotidesNucleic Acids Res318209710712682360
  • DapićVBatesPJTrentJO2002Antiproliferative activity of G-quartet-forming oligonucleotides with backbone and sugar modificationsBiochemistry411136768511888284
  • DerenziniM2000The AgNORsMicron3121172010588056
  • DerenziniMSirriVTrereD1995The quantity of nucleolar proteins nucleolin and protein B23 is related to cell doubling time in human cancer cellsLab Invest7344975027474921
  • de SmidtPCLe DoanTde FalcoS1991Association of antisense oligonucleotides with lipoproteins prolongs the plasma half-life and modifies the tissue distributionNucleic Acids Res191746957001891360
  • DougallWCQianXGreeneMI1993Interaction of the neu/p185 and EGF receptor tyrosine kinases: implications for cellular transformation and tumor therapyJ Cell Biochem53161737901229
  • DouganHLysterDMVoCV2000Extending the lifetime of anticoagulant oligodeoxynucleotide aptamers in bloodNucl Med Biol2732899710832086
  • DroletDWNelsonJTuckerCE2000Pharmacokinetics and safety of an anti-vascular endothelial growth factor aptamer (NX1838) following injection into the vitreous humor of rhesus monkeysPharm Res171215031011303960
  • DybaMTarasovaNIMichejdaCJ2004Small molecule toxins targeting tumor receptorsCurr Pharm Des101923113415279611
  • EllingtonADCoxJCHayhurstA2005Automated in vitro selection and microarray applications for functional RNA sequencesThe RNA World3rd EdCold Spring Harbor, New YorkCold Spring Harbor Laboratory Press683719
  • EllingtonADSzostakJW1990In vitro selection of RNA molecules that bind specific ligandsNature3466287818221697402
  • EricksonHPBourdonMA1989Tenascin: an extracellular matrix protein prominent in specialized embryonic tissues and tumorsAnnu Rev Cell Biol571922480799
  • FarokhzadOCChengJTeplyBA2006Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivoProc Natl Acad Sci USA1031663152016606824
  • FarokhzadOCJonSKhademhosseiniA2004Nanoparticle-aptamer bioconjugates: a new approach for targeting prostate cancer cellsCancer Res642176687215520166
  • FerreiraCSMatthewsCSMissailidisS2006DNA aptamers that bind to MUC1 tumour marker: design and characterization of MUC1-binding single-stranded DNA aptamersTumour Biol27628930117033199
  • FreemanLMKrynyckyiBRLiY2002The role of (111)In Capromab Pendetide (Prosta-ScintR) immunoscintigraphy in the management of prostate cancerQ J Nucl Med462131712114876
  • GhoshAHestonWD2004Tumor target prostate specific membrane antigen (PSMA) and its regulation in prostate cancerJ Cell Biochem9135283914755683
  • GinestierCCharafe-JauffretEBertucciF2002Distinct and complementary information provided by use of tissue and DNA microarrays in the study of breast tumor markersAm J Pathol161412233312368196
  • GinistyHSicardHRogerB1999Structure and functions of nucleolinJ Cell Sci112Pt 67617210036227
  • GoldLPoliskyBUhlenbeckO1995Diversity of oligonucleotide functionsAnnu Rev Biochem64763977574500
  • GoldenMCCollinsBDWillisMC2000Diagnostic potential of PhotoSELEX-evolved ssDNA aptamersJ Biotechnol812316778
  • GopinathSC2007Methods developed for SELEXAnal Bioanal Chem38711718217072603
  • GriffinLCTidmarshGFBockLC1993In vivo anticoagulant properties of a novel nucleotide-based thrombin inhibitor and demonstration of regional anticoagulation in extracorporeal circuitsBlood8112327168507864
  • HansfordJRMulliganLM2000Multiple endocrine neoplasia type 2 and RET: from neoplasia to neurogenesisJ Med Genet37118172711073534
  • HeldDMKisselJDPattersonJT2006HIV-1 inactivation by nucleic acid aptamersFront Biosci118911216146716
  • HickeBJMarionCChangYF2001Tenascin-C aptamers are generated using tumor cells and purified proteinJ Biol Chem27652486445411590140
  • HickeBJStephensAWGouldT2006Tumor targeting by an aptamerJ Nucl Med4746687816595502
  • HollingsworthMASwansonBJ2004Mucins in cancer: protection and control of the cell surfaceNat Rev Cancer41456014681689
  • HoroszewiczJSKawinskiEMurphyGP1987Monoclonal antibodies to a new antigenic marker in epithelial prostatic cells and serum of prostatic cancer patientsAnticancer Res75B927352449118
  • HoughCDSherman-BaustCAPizerES2000Large-scale serial analysis of gene expression reveals genes differentially expressed in ovarian cancerCancer Res60226281711103784
  • HovanessianAGPuvion-DutilleulFNisoleS2000The cell-surface-expressed nucleolin is associated with the actin cytoskeletonExp Cell Res26123122811112338
  • HughesJARaoGA2005Targeted polymers for gene deliveryExpert Opin Drug Deliv211455716296741
  • IresonCRKellandLR2006Discovery and development of anticancer aptamersMol Cancer Ther51229576217172400
  • IsraeliRSPowellCTCorrJG1994Expression of the prostate-specific membrane antigenCancer Res5471807117511053
  • IwashitaTKatoMMurakamiH1999Biological and biochemical properties of Ret with kinase domain mutations identified in multiple endocrine neoplasia type 2B and familial medullary thyroid carcinomaOncogene182639192210445857
  • JellinekDGreenLSBellC1995Potent 2′-amino-2′-deoxypyrimidine RNA inhibitors of basic fibroblast growth factorBiochemistry343611363727547864
  • JooEJten DamGBvan KuppeveltTH2005Nucleolin: acharan sulfate-binding protein on the surface of cancer cellsGlycobiology1511915329357
  • KatoYMinakawaNKomatsuY2005New NTP analogs: the synthesis of 4′-thioUTP and 4′-thioCTP and their utility for SELEXNucleic Acids Res33929425115914669
  • KohlgrafKGGawronAJHigashiM2003Contribution of the MUC1 tandem repeat and cytoplasmic tail to invasive and metastatic properties of a pancreatic cancer cell lineCancer Res631650112012941828
  • KoukoulisGKGouldVEBhattacharyyaA1991Tenascin in normal, reactive, hyperplastic, and neoplastic tissues: biologic and pathologic implicationsHum Pathol227636431712747
  • LaberDASharmaVRBhupalamL2005Update on the first phase I study of AGRO100 in advanced cancerJ Clin Oncol (Meet Abstr)233064
  • LaberDATaftBSKloeckerGH2006Extended phase I study of AS1411 in renal and non-small cell lung cancersJ Clin Oncol24[18SJune 20 Supplement13098
  • LapeyreBBourbonHAmalricF1987Nucleolin, the major nucleolar protein of growing eukaryotic cells: an unusual protein structure revealed by the nucleotide sequenceProc Natl Acad Sci USA846147263470736
  • LegrandDVigiéKSaidEA2004Surface nucleolin participates in both the binding and endocytosis of lactoferrin in target cellsEur J Biochem27123031714717698
  • LuYLowPS2002Folate-mediated delivery of macromolecular anticancer therapeutic agentsAdv Drug Deliv Rev5456759312204598
  • LupoldSEHickeBJLinY2002Identification and characterization of nuclease-stabilized RNA molecules that bind human prostate cancer cells via the prostate-specific membrane antigenCancer Res621440293312124337
  • MackieEJHalfterWLiveraniD1988Induction of tenascin in healing woundsJ Cell Biol1076 Pt 22757672462568
  • MaeshimaAMiyagiAHiraiT1997Mucin-producing adenocarcinoma of the lung, with special reference to goblet cell type adenocarcinoma: immunohistochemical observation and Ki-ras gene mutationPathol Int477454609234384
  • McCauleyTGKurzJCMerlinoPG2006Pharmacologic and pharmacokinetic assessment of anti-TGFbeta2 aptamers in rabbit plasma and aqueous humorPharm Res2323031116411148
  • McNamaraJO2ndAndrechekERWangY2006Cell type-specific delivery of siRNAs with aptamer-siRNA chimerasNat Biotechnol24810051516823371
  • MoffattSPapasakelariouCWiehleS2006Successful in vivo tumor targeting of prostate-specific membrane antigen with a highly efficient J591/PEI/DNA molecular conjugateGene Ther1397617216453011
  • MurphyGPElgamalAASuSL1998Current evaluation of the tissue localization and diagnostic utility of prostate specific membrane antigenCancer83112259699840525
  • NgEWShimaDTCaliasP2006Pegaptanib, a targeted anti-VEGF aptamer for ocular vascular diseaseNat Rev Drug Discov521233216518379
  • OhuchiSPOhtsuTNakamuraY2005A novel method to generate aptamers against recombinant targets displayed on the cell surfaceNucleic Acids Symp Ser (Oxf)493512
  • OhuchiSPOhtsuTNakamuraY2006Selection of RNA aptamers against recombinant transforming growth factor-beta type III receptor displayed on cell surfaceBiochimie88789790416540230
  • OosterkampHMScheinerLStefanovaMC1997Comparison of MUC-1 mucin expression in epithelial and non-epithelial cancer cell lines and demonstration of a new short variant form (MUC-1/Z)Int J Cancer72187949212228
  • OrendG2005Potential oncogenic action of tenascin-C in tumorigenesisInt J Biochem Cell Biol37510668315743679
  • OrendGChiquet-EhrismannR2006Tenascin-C induced signaling in cancerCancer Lett24421436316632194
  • PegramMHsuSLewisG1999Inhibitory effects of combinations of HER-2/neu antibody and chemotherapeutic agents used for treatment of human breast cancersOncogene181322415110327070
  • PerkinsACMissailidisS2007Radiolabelled aptamers for tumour imaging and therapyQ J Nucl Med Mol Imaging
  • PestourieCTavitianBDucongeF2005Aptamers against extracellular targets for in vivo applicationsBiochimie879109213015733730
  • PietrasKOstmanASjoquistM2001Inhibition of platelet-derived growth factor receptors reduces interstitial hypertension and increases transcapillary transport in tumorsCancer Res61729293411306470
  • RusconiCPScardinoELayzerJ2002RNA aptamers as reversible antagonists of coagulation factor IXaNature419690290412214238
  • RusconiCPRobertsJDPitocGA2004Antidote-mediated control of an anticoagulant aptamer in vivoNat Biotechnol22111423815502817
  • SaidEAKrustBNisoleS2002The anti-HIV cytokine midkine binds the cell surface-expressed nucleolin as a low affinity receptorJ Biol Chem277403749250212147681
  • SarcevicBLilischkisRSutherlandRL1997Differential phosphorylation of T-47D human breast cancer cell substrates by D1-, D3-, E-, and Atype cyclin-CDK complexesJ Biol Chem2725233327379407125
  • SchmidtKSBorkowskiSKurreckJ2004Application of locked nucleic acids to improve aptamer in vivo stability and targeting functionNucleic Acids Res321957576515509871
  • SchneiderPMHungMCChioccaSM1989Differential expression of the c-erbB-2 gene in human small cell and non-small cell lung cancerCancer Res49184968712569928
  • ShiHFanXSevilimeduA2007RNA aptamers directed to discrete functional sites on a single protein structural domainProc Natl Acad Sci USA104103742617360423
  • SlamonDJGodolphinWJonesLA1989Studies of the HER-2/neu proto-oncogene in human breast and ovarian cancerScience2444905707122470152
  • SrivastavaMPollardHB1999Molecular dissection of nucleolin’s role in growth and cell proliferation: new insightsFaseb J131419112210544174
  • StefanidakisMKoivunenE2004Peptide-mediated delivery of therapeutic and imaging agents into mammalian cellsCurr Pharm Des102430334415379666
  • StorckSShuklaMDimitrovS2007Functions of the histone chaperone nucleolin in diseasesSubcell Biochem411254417484127
  • SuSLHuangIPFairWR1995Alternatively spliced variants of prostate-specific membrane antigen RNA: ratio of expression as a potential measurement of progressionCancer Res557144137882349
  • SullengerBAGallardoHFUngersGE1990Overexpression of TAR sequences renders cells resistant to human immunodeficiency virus replicationCell63360182225067
  • SullengerBAGallardoHFUngersGE1991Analysis of trans-acting response decoy RNA-mediated inhibition of human immunodeficiency virus type 1 transactivationJ Virol6512681161942253
  • Taylor-PapadimitriouJBurchellJMilesDW1999MUC1 and cancerBiochim Biophys Acta14552330113
  • TuerkCGoldL1990Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymeraseScience2494968505102200121
  • UmeharaTFukudaKNishikawaF2004Designing and analysis of a potent bi-functional aptamers that inhibit protease and helicase activities of HCV NS3Nucleic Acids Symp Ser (Oxf)481956
  • VaterAJaroschFBuchnerK2003Short bioactive Spiegelmers to migraine-associated calcitonin gene-related peptide rapidly identified by a novel approach: tailored-SELEXNucleic Acids Res3121e13014576330
  • von Mensdorff-PouillySSnijdewintFGVerstraetenAA2000Human MUC1 mucin: a multifaceted glycoproteinInt J Biol Markers1543435611192832
  • WhiteRRSullengerBARusconiCP2000Developing aptamers into therapeuticsJ Clin Invest10689293411032851
  • WillisMCCollinsBDZhangT1998Liposome-anchored vascular endothelial growth factor aptamersBioconjug Chem95573829736491
  • YokotaJYamamotoTMiyajimaN1988Genetic alterations of the c-erbB-2 oncogene occur frequently in tubular adenocarcinoma of the stomach and are often accompanied by amplification of the v-erbA homologueOncogene2328373281095
  • ZagzagDFriedlanderDRMillerDC1995Tenascin expression in astrocytomas correlates with angiogenesisCancer Res554907147531617
  • ZhangSZhangHSReuterVE1998Expression of potential target antigens for immunotherapy on primary and metastatic prostate cancersClin Cancer Res422953029516914