Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2012 Sep 1.
Published in final edited form as: Curr Med Chem. 2011 Sep 1;18(27):4185–4194. doi: 10.2174/092986711797189547

Tumor-Targeted Drug Delivery with Aptamers

Yin Zhang 1, Hao Hong 2, Weibo Cai 1,2,3,*
PMCID: PMC3205327  NIHMSID: NIHMS305435  PMID: 21838687

Abstract

Cancer is one of the leading causes of death around the world. Tumor-targeted drug delivery is one of the major areas in cancer research. Aptamers exhibit many desirable properties for tumor-targeted drug delivery, such as ease of selection and synthesis, high binding affinity and specificity, low immunogenicity, and versatile synthetic accessibility. Over the last several years, aptamers have quickly become a new class of targeting ligands for drug delivery applications. In this review, we will discuss in detail about aptamer-based delivery of chemotherapy drugs (e.g. doxorubicin, docetaxel, daunorubicin, and cisplatin), toxins (e.g. gelonin and various photodynamic therapy agents), and a variety of small interfering RNAs. Although the results are promising which warrants enthusiasm for aptamer-based drug delivery, tumor homing of aptamer-based conjugates after systemic injection has only been achieved in one report. Much remains to be done before aptamer-based drug delivery can reach clinical trials and eventually the day-to-day management of cancer patients. Therefore, future directions and challenges in aptamer-based drug delivery are also discussed.

Keywords: Aptamers, drug delivery, cancer, nanomedicine, theranostics, RNA interference, siRNA

INTRODUCTION

Cancer is one of the leading causes of death around the world. It is estimated that there were about 12.7 million cancer cases and 7.6 million cancer deaths in 2008 [1]. Among these, about 56% of the cases and 64% of the deaths occurred in the economically developing world. In the United States, cancer is the second leading cause of death (http://www.cdc.gov). In 2010, a total of 1,529,560 new cancer cases and 569,490 deaths from cancer were estimated to occur [2]. Over the last four decades, tremendous investment and effort has been devoted towards the “War on Cancer”, which has become one of the top priorities in pharmaceutical industry and the National Institutes of Health. With the development of new anti-cancer drugs with better efficacy and fewer side effects, accurate and efficient delivery of these agents to the tumor sites in cancer patients is of utmost importance [3,4]. A recent search of “drug delivery AND cancer” in PubMed returned > 19,000 publications. For successful delivery of anti-cancer drugs, many aspects have to be optimized simultaneously which include encapsulation, targeting, delivery, controlled release, among many others. Ideally, a combination of specific targeting and controlled drug release should be able to deliver sufficient doses of cytotoxic drugs to cancer cells (as well as cancer stem cells [5,6]) over an extended period of time without significantly affecting the surrounding normal tissue. Although a large number of anti-cancer drugs have been approved by the Food and Drug Administration (FDA) [7], most of them are not molecularly targeted which can give rise to significant toxicity and side effects.

Aptamers, typically generated through Systematic Evolution of Ligands by EXponential enrichment (SELEX; Fig. (1)) [8,9], have quickly emerged as a novel and powerful class of ligands with excellent potential for diagnostic and therapeutic applications [10]. These single-stranded DNA/RNA oligonucleotides (with a molecular weight of 5–40 kDa) can fold into well-defined 3D structures and bind to their target molecules with high affinity and specificity. To date, aptamers have been selected against a wide range of targets such as proteins, phospholipids, sugars, nucleic acids, whole cells, among others. Since wild-type RNA and DNA molecules can be easily degraded by nucleases, various strategies have been adopted to synthesize aptamers with enhanced in vitro/in vivo stability, such as the use of chemically modified oligonucleotides [1113], unnatural internucleotide linkages [14], polyethylene glycol (PEG) conjugation [15], Spiegelmers (where the sugars are enantiomers of wild-type nucleic acid sugars) [16,17], among many others [10].

Fig. 1.

Fig. 1

A schematic depiction of SELEX (systematic evolution of ligands by exponential enrichment). The target can be either proteins or cancer cells. For cell-based SELEX, typically the nucleic acid library is first incubated with non-target cells. Only unbound nucleic acids are used for selection against the target cells. Typically, aptamer selection can be completed after 10–20 rounds of selection process. Adapted from [42].

Aptamers possess several advantages over other ligands typically used in drug delivery such as antibodies. First, production of aptamers does not rely on biological systems hence is much easier to scale up with low batch-to-batch variability; Second, aptamers are quite thermally stable and can be denatured and renatured multiple times without significant loss of activity [18]; Third, the smaller size of aptamers than intact antibodies (~150 kDa) can lead to better tissue penetration in solid tumors; Fourth, lack of immunogenicity is another favorable advantage of aptamers over antibodies; Lastly, conjugation chemistry for the attachment of various imaging labels or functional groups to aptamers are orthogonal to nucleic acid chemistry, hence they can be readily introduced during aptamer synthesis. On the other hand, the disadvantages of aptamers include faster excretion than antibodies due to smaller size, potentially weaker binding to targets than antibodies, unpredictable toxicity and other systemic properties, susceptibility to serum degradation when unmodified aptamers are used, and intellectual property-related issues [10]. During the last two decades since aptamers were first selected through SELEX [8,9], Pegaptanib (Pfizer/Eyetech), an aptamer that binds to human vascular endothelial growth factor (VEGF), has been approved by the FDA for clinical use in treating age-related macular degeneration (AMD). A variety of aptamers against other molecular targets are currently in clinical investigation [10].

In this review, we will focus on the use of aptamers for tumor-targeted drug delivery. Although aptamers themselves can be employed for therapeutic applications in various diseases such as cancer, this aspect is out of the scope of this review. Generally speaking, the therapeutic agents that have been delivered using aptamers as the targeting ligands can be categorized into three major classes: chemotherapy drugs, toxins, and small interfering RNAs (siRNAs).

DELIVERY OF CHEMOTHERAPY DRUGS

Metastases are the cause of 90% of human cancer deaths [19,20]. Chemotherapy of cancer metastasis, although effective in some patients, has significant toxicity because of non-specific distribution of the cytotoxic drugs which severely limits the maximum allowable dose. However, rapid elimination and widespread distribution into non-targeted organs/tissues requires the administration of large doses. This vicious cycle of large doses and the concurrent toxicity is a major limitation of current cancer therapy. Sometimes patients succumb to the adverse effects of the drug far earlier than the tumor burden [21,22]. Therefore, the development of an effective drug delivery strategy can significantly improve metastatic cancer patient management.

Doxorubicin (Dox), an anthracycline antibiotic which can intercalate within the double-stranded CG sequences of DNA and RNA, has been used in treating many cancer types such as hematological malignancies, carcinomas, and soft tissue sarcomas [2325]. In many cases, the use of Dox can lead to dose-dependent cardiotoxicity such as dilated congestive heart failure and cardionyopathy [26], which mandates the development of efficient tumor-targeted delivery strategies for Dox. In one report, an aptamer-Dox (Apt-Dox) physical conjugate (via intercalation) was prepared and tested in prostate cancer cells [27]. A10, a 2′-fluoropyrimidine RNA aptamer that binds to the extracellular domain of the prostate specific membrane antigen (PSMA) with a Kd of 2.1 nM [28], was used in this study. Composed of 57 base pairs, the molecular weight of A10 is 18.5 kDa. The Apt-Dox conjugate exhibited good stability in cell culture medium and was shown to target PSMA-expressing cells with high efficiency [27]. It was suggested that this strategy may be employed to develop novel targeted therapeutic agents for more effective cancer chemotherapy in the future.

In another report, Dox was conjugated to a DNA aptamer (termed “sgc8c”) via a hydrazone linker for specific killing of target cells [29]. The sgc8c DNA aptamer, which was selected using human T-cell acute lymphoblastic leukemia cells, can recognize protein tyrosine kinase 7 (PTK7) with nM binding affinity [30]. The high specificity and well-characterized DNA structure of sgc8c makes it capable of distinguishing target leukemia cells from normal human bone marrow aspirate, as well as identifying cancer cells closely related to the target cell line in clinical specimens. It was demonstrated that the sgc8c-Dox conjugate specifically bound to target cancer cells with high affinity, which led to high efficiency cell killing [29]. In some cases, chemical modification may change the binding characteristics of aptamers, as well as the safety and efficacy profile of the drugs. Therefore, Daunorubicin (Dau), another chemotherapy drug of the anthracycline family commonly used in the treatment of leukemia, has been physically bound to the sgc8 aptamer through intercalation instead of chemical linkage [31]. It was found that sgc8, serving as an escort aptamer, could specifically deliver Dau into PTK7-positive cells with high potency.

DELIVERY OF CHEMOTHERAPY DRUGS WITH NANOTECHNOLOGY

Nanotechnology can have a revolutionary impact on imaging and therapy of cancer [3236]. With sizes smaller than a few hundred nanometers [37,38], several orders of magnitude smaller than human cells, nanomaterials can exhibit properties distinct from both molecules and bulk solids. Thus, they can offer unprecedented interactions with biomolecules both on the surface of and inside the cells, which may result in cancer detection and treatment in ways unimaginable before. Because of the large surface area/loading capacity and versatile chemistry, nanomaterials are excellent carriers for targeted delivery of anti-cancer drugs. Many nano-systems have been developed for systemic cancer therapy, such as liposomes, albumin-based particles, PEGylated proteins, biodegradable polymer-drug composites, polymeric micelles, dendrimers, among others [39]. The combination of nanoparticles and aptamers can have broad applications ranging from diagnostics to targeted delivery of drugs [4042]. Over the last decade, a variety of nanoparticle-aptamer (NP-Apt) conjugates have been constructed for drug delivery applications.

In one early study, the anti-cancer efficacy of a NP-Apt conjugate (where the A10 RNA aptamer was used as the targeting ligand) was shown both in vitro and in vivo [43]. Docetaxel (Dtxl; an anti-mitotic chemotherapy drug [44,45]) was encapsulated into a NP-Apt conjugate where the nanoparticle was composed of a biocompatible and biodegradable copolymer, poly(D, L-lactic-co-glycolic acid)-block-poly(ethylene glycol) (i.e. PLGA-b-PEG). It was found that the NP-Apt-Dtxl conjugate bound to PSMA on the cancer cell surface. Subsequent endocytosis into the cells led to significantly enhanced cellular toxicity than the non-targeted nanoparticles (i.e. NP-Dtxl). In addition, therapeutic efficacy and reduced toxicity were observed in vivo, where a single intratumoral injection of NP-Apt-Dtxl led to significant anti-tumor effect. The enhanced efficacy of NP-Apt-Dtxl over NP-Dtxl was attributed to the intracellular Dtxl release upon PSMA-mediated endocytosis of the former, while NP-Dtxl may release the drug in the extracellular space, thereby causing systemic distribution and absorption, increased toxicity, and decreased efficacy. Subsequently, similar strategy was also used to deliver cisplatin (another chemotherapy drug that can bind to and cause cross-linking of DNA [46]) into cancer cells [47].

In another report, a smart Bi-FRET QD-Apt-Dox conjugate (FRET and QD denote fluorescence resonance energy transfer and quantum dot, respectively) was constructed as both an imaging agent and a drug delivery vehicle (Fig. (2)) [48]. QDs are inorganic fluorescent semiconductor nanoparticles with many desirable properties for imaging applications such as high quantum yields, high molar extinction coefficients, strong resistance to photobleaching and chemical degradation, narrow emission spectra, large effective Stokes shifts, among others [49,50]. The A10 RNA aptamer was attached covalently to the QD surface, while Dox was intercalated in the double-stranded stem region of the A10 aptamer [48]. Since the fluorescence of QD was quenched by the absorbance of Dox and the fluorescence of Dox was quenched by the A10 aptamer, this QD-Apt-Dox conjugate is initially in the fluorescence “OFF” state. After taken up by PSMA-positive cancer cells, Dox was gradually released from the conjugate which led to a recovery of the fluorescence signal from QD and Dox (i.e. the “ON” state). Therefore, this multifunctional QD-Apt-Dox conjugate can be used for not only drug delivery to targeted cells, but also monitoring the delivery/release of the drug and concurrently imaging the target cells. Cytotoxicity of QD-Apt-Dox was found to be significantly higher in PSMA-positive cells as compared to PSMA-negative cells. Subsequently, similar strategies have also been adopted for cancer imaging and therapy applications by several other groups, where the A10 aptamer was attached to various nanoparticles such as thermally cross-linked superparamagnetic iron oxide nanoparticles (TCL-SPION, which could act as both a magnetic resonance contrast agent and a carrier for Dox) and gold nanoparticles (which served as both a contrast agent for computed tomography and a carrier for Dox) [51,52].

Fig. 2.

Fig. 2

A QD-Apt-Dox conjugate. A. QD-Apt-Dox is initially “off” as the fluorescence of QD is transferred to Dox and the fluorescence of Dox is quenched by the aptamer, both by fluorescence resonance energy transfer. B. Once QD-Apt-Dox is inside cancer cells, Dox is gradually released from the conjugate and the fluorescence of QD is recovered. C. Microscopy images of PSMA-positive cells after incubation with QD-Apt-Dox. QD and Dox are shown in green and red, respectively. Scale bar: 20 μm. Adapted from [42,48].

Due to the relatively large size and surface area of nanoparticles, dual-targeting can be employed for drug delivery when more than one type of aptamers are attached to the nanoparticle surface. Recently, one such dual-aptamer conjugate was constructed for targeting both PSMA-positive and PSMA-negative prostate cancer cells (Fig. (3)) [53]. Both the A10 RNA aptamer and DUP-1 peptide aptamer (selected against PSMA-negative cells [54]) were conjugated to streptavidin to form a dual-aptamer complex which can enable targeted delivery of drugs to both PSMA-positive and PSMA-negative cells. Dox was loaded on the stem region of the A10 aptamer similar as mentioned above [48]. Furthermore, Dox-loaded A10 RNA aptamer and DUP-1 peptide aptamer were immobilized on the surface of TCL-SPION for targeted delivery of Dox in cell culture. This proof-of-principle study demonstrated that tumor-specific drug delivery to multiple targets can be achieved using this dual-aptamer conjugate, which may potentially also be applied for image-guided drug delivery, as well as monitoring the therapeutic response.

Fig. 3.

Fig. 3

A dual-aptamer (A10 RNA and DUP-1 peptide) probe that can target both PSMA-positive and PSMA-negative cells. The red parts indicate the cells and blue dots represent the stained superparamagnetic iron oxide nanoparticles. Adapted from [53].

DELIVERY OF TOXINS

Besides targeted delivery of chemotherapy drugs, aptamers have also been used for directing toxins to cancer cells. Gelonin, a ribosome-inactivating toxin with a molecular weight of 28 kDa, can cause cell death by cleaving a specific glycosidic bond in rRNA thereby interrupting protein synthesis [55]. Gelonin has low intrinsic cytotoxicity since it cannot be efficiently internalized into cells by itself. Over the years, gelonin has been conjugated to various antibodies or other proteins for targeted delivery to tumor cells [5659]. In one study, gelonin was conjugated to an RNA aptamer A9, which also binds to PSMA with nM affinity [60]. The A9-gelonin conjugate exhibited dramatically enhanced potency to cells that express PSMA. Interestingly, it was also found that the A9-gelonin conjugate had 3-fold lower toxicity in PSMA-negative cells than unconjugated gelonin, suggesting that non-specific uptake of gelonin or presentation of the toxin may be inhibited by the A9 aptamer. This study suggested that aptamers may also be used to reduce the side effects of certain toxins or chemotherapy drugs.

Phototoxic-agent conjugated apatmers that can selectively enter and destroy cancer cells were reported [61]. DNA aptamers were selected for specific binding of short O-glycan peptides expressed only on the surface of certain cancer cells, and were further modified at their 5′-ends with chlorin e6 (a photodynamic therapy agent that can be activated by laser illumination to produce cytotoxic singlet oxygen species [62,63]) via amide bond formation. Upon light activation, the modified aptamers had significantly enhanced toxicity than the drug or aptamer alone in targeted cells, while no toxicity was observed in cells lacking the O-glycan peptides [61].

A similar strategy was adopted to link another photodynamic agent, 5,10,15,20-tetrakis(1-methylpyridinium-4-yl)porphyrin (TMPyP4), to an AS1411 aptamer which could recognize and bind to nucleolin with high affinity and specificity [64]. Upon crossing the plasma membrane into cancer cells via nucleolin-mediated internalization of AS1411-TMPyP4, significantly higher phototoxicity was observed in nucleolin-expressing cancer cells than the control cells after light exposure (i.e. photodynamic therapy). The fact that AS1411 is currently in Phase II clinical trials as a cancer therapeutic [65,66] may speed up the translation of future AS1411-based conjugates for clinical use.

DELIVERY OF siRNA

Since the initial discovery in 1998, RNA interference (RNAi) has emerged as a powerful tool for therapeutic gene silencing due to its unique specificity, broad applicability, and high efficiency [67]. SiRNAs composed of 20–30 nucleotides can lead to the degradation of a specific, targeted mRNA through the RNA-induced silencing complex (RISC) [68,69]. Although the mechanisms and the advantages of RNAi have been elucidated and many clinical trials are ongoing [7072], clinical use of RNAi still faces a number of challenges. There is an urgent need for more efficient and specific delivery methods to deliver siRNAs to target cells or organs [70,73]. The ultimate goal of achieving RNAi-based therapies cannot be attained without improving the safety, effectiveness, and reliability of the delivery systems for siRNA or small hairpin RNA (shRNA). The attractive characteristics of aptamers, such as small size, high binding affinity/specificity, similar chemical composition as siRNA/shRNA, and desirable stability, make them excellent candidates for targeted delivery of siRNA.

In general, there are two major strategies for attaching siRNA to aptamers: 1) chemical conjugation and 2) covalently linking a siRNA and a RNA aptamer to form an aptamer-siRNA chimera. In a proof-of-principle study, biotinylated A9 aptamer was linked to a biotinylated 27mer anti-lamin A/C siRNA via a modular streptavidin bridge [74]. The disulfide linker between the sense strand of siRNA and biotin in the construct could be cleaved, after entering the cells, to enhance siRNA release. Effective PSMA-mediated internalization and siRNA-mediated inhibition of target gene expression was achieved in tumor cells with this multivalent construct. Since streptavidin is a tetramer with four binding sites for biotin, it is possible to have more than one kind of aptamers/siRNAs within the construct which can be mixed and matched. However, the immunogenicity of streptavidin may limit its use as a delivery vehicle for siRNA. Less immunogenic streptavidin variants or linkers other than streptavidin would be desirable for future studies.

A truncated form of the A10 RNA aptamer was covalently linked to a siRNA specific for eukaryotic elongation factor 2 (EEF2) mRNA, forming monovalent or bivalent A10-anti-EEF2-siRNA constructs [75]. These conjugates were processed by the enzyme Dicer for siRNA release, after they were internalized by the targeted cells. Both the monovalent and bivalent A10-anti-EEF2-siRNA conjugates induced a sequence-specific knock-down of EEF2 mRNA and protein expression in vitro. The bivalent conjugate, with higher affinity than and same specificity as the monovalent conjugate, was found to be internalized much more efficiently.

A 2′-fluoro-modified A10 RNA aptamer was appended covalently to a 21mer siRNA which could knock down the expression of two survival genes commonly overexpressed in human tumors, polo-like kinase 1 (PLK1) and B-cell lymphoma 2 (BCL2) [76]. The aptamer-siRNA chimeras were shown to selectively enter PSMA-positive cells, effectively silence target gene expression, and lead to depletion of target proteins and cell death both in vitro and in vivo (after intratumoral injection). Specificity of the aptamer-siRNA chimera was confirmed with a number of controls including the use of PSMA-negative cells, a non-silencing siRNA, and a mutated A10 aptamer. It is worth pointing out that although the aptamers contained significant duplex structures, the chimeras did not induce any interferon response, suggesting that this strategy may be able to avoid certain adverse symptoms commonly observed in siRNA-based therapies [77].

To date, most of the aptamer-based anti-cancer agents were only investigated in vitro, with very few conjugates showing any efficacy in vivo. Direct injection of aptamer-based conjugates intratumorally has been reported to be effective in a few studies [43,76]. However, for potential clinical applications, systemic administration will be needed in most scenarios. Generally speaking, much higher therapeutic doses are required for systemic administration than local injection, which can lead to greater risk for harmful side effects due to unwanted exposure of the conjugates to normal non-targeted tissues. Further improvement of aptamer-siRNA chimeras that can minimize the dose and reduce the cost for efficacious anti-cancer therapy is of great value for future clinical investigation.

The abovementioned A10-anti-PLK1 siRNA chimera has been further optimized to achieve enhanced inhibition of prostate cancer xenograft growth after systemic administration (Fig. (4)) [78]. The aptamer portion of the chimera was truncated from 71 to 39 nucleotides, which still exhibited nM binding affinity. Such modification also made the chimera amenable to chemical synthesis because of significantly shorter nucleotide sequence. Furthermore, to increase the gene silencing activity and specificity of the chimera, several additional structural modifications were made to the siRNA to facilitate Dicer recognition, increase silencing specificity, as well as improve loading of the guide strand into the RISC. The optimized aptamer-siRNA chimera exhibited potent efficacy in vivo upon systemic administration, where ~70% of tumors in the treated mice completely regressed by the end of the treatment with no observed toxicity to normal organs. Subsequently, appending a 20 kDa PEG increased both the in vivo circulation half-life (from < 35 min to > 30 h) and serum stability of the chimera, without affecting its targeting and gene silencing capability. As a result, the therapeutic dose of the second-generation aptamer-siRNA chimera was reduced from 10 nmol to 1.25 nmol in 10 days, which not only reduced the cost of treatment but also minimized the risk of harmful side effects. Although the results observed in this study were very promising, the mechanism regarding how the siRNA escaped from endosomes still remains unclear.

Fig. 4.

Fig. 4

An improved second generation aptamer-siRNA chimera exhibited excellent anti-cancer efficacy in a xenograft model of prostate cancer. A. The first and second generation chimeras. B. Bioluminescence imaging of mice bearing luciferase-expressing PSMA-positive tumors showed significantly lower signal after being treated with the second generation aptamer-siRNA chimera as compared to saline control. C. H&E staining of tumor tissue after treatment showed readily detectable areas of necrosis (asterisks) in the second generation aptamer-siRNA chimera-treated tumors, but not frequently in saline-treated tumors. TUNEL staining was detected in scattered cells throughout the tumor section of each group. Adapted from [78].

The large surface areas of nanoparticles can allow for the loading of a combination of anti-cancer drugs, which may lead to improved therapeutic efficacy if the drug combination is chosen rationally. In one recent report, co-delivery of Dox and Bcl-xL-specific shRNA was accomplished with the A10 RNA aptamer [79]. Branched polyethyleneimine (PEI) was grafted to PEG and aptamers were conjugated to the surface of the PEI-PEG construct. Dox was intercalated onto the aptamers and Bcl-xL-specific shRNA was complexed with the Dox-intercalated construct (i.e. PEI-PEG-Apt-Dox) through electrostatic interaction to generate the final conjugate. This conjugate efficiently and selectively delivered both shRNA and Dox to PSMA-positive cells through aptamer-mediated binding, which led to synergistic tumoricidal effect.

CONCLUSIONS AND FUTURE PERSPECTIVES

One of the major areas in cancer research is targeted delivery of drugs to cancerous cells, which can not only increase the therapeutic efficacy but also reduce the adverse side effects of the drugs [4,33]. Significant progress has been made over the last two decades since the initial appearance of aptamers. As therapeutics, one aptamer (i.e. Pegaptanib) has been approved by the FDA and many others are in late phase clinical trials. As delivery agents, aptamers have only been investigated for a few years. To date, a variety of anti-cancer agents (e.g. chemotherapy drugs, toxins, and siRNAs) have been successfully delivered to cancer cells in vitro. However, to the best of our knowledge, successful drug delivery to the tumor tissue after systemic injection of aptamer-based conjugates has only been achieved in one report [78]. Much remains to be done before aptamer-based drug delivery can reach clinical trials and eventually the day-to-day management of cancer patients.

The development of more efficient selection methods and easier strategies for aptamer conjugation are needed for potential clinical translation. With more and more academic groups and companies getting involved in this area of research after the earliest aptamer intellectual property expires [80], the development of aptamer-based therapeutics will speed up and aptamers will find their own niches in cancer treatment and bring revolution into targeted drug delivery as well as cancer diagnosis. Besides the three classes of agents mentioned in this review, aptamers can also be used to deliver other anti-cancer agents such as therapeutic radioisotopes (e.g. α-particle emitters such as 211At and 225Ac, β-particle emitters such as 90Y, 67Cu, 131I, 177Lu, 186Re, and 188Re, and Auger electron emitters such as 111In and 125I). Another interesting avenue of future research is to use an aptamer to deliver other aptamers that were selected against intracellular targets (e.g. nuclear factor-κB [81], high mobility group A1 [82], among others).

To date, researchers have identified high-affinity aptamers that target a broad array of protein families including cytokines, proteases, kinases, cell-surface receptors, cell-adhesion molecules, among others [10,8385]. Since aptamers can be selected against most protein targets, the possible therapeutic applications of aptamer-based drug delivery range far and wide. One disadvantage of aptamers is the hard-to-predict and poorly understood pharmacokinetics, toxicity, and other properties upon systemic delivery. Although aptamers are unlikely to illicit an immune response, at least at the antibody level, the safety profile of each individual aptamer (conjugate) must be examined carefully. Off-target effects due to inhibition/activation of other proteins with similar structural conformations as the target protein need to be investigated empirically since they will likely differ for each aptamer.

The most significant barrier to the widespread use of RNA interference in the clinic is efficient delivery. Solving this problem will require the development of clinically suitable, safe, and effective gene delivery systems. Aptamer-siRNA chimeras represent a platform technology that can be applied to many other aptamers/siRNAs in the future [76,78]. Nanotechnology will also play a key role in the delivery of siRNAs (as well as other anti-cancer agents such as chemotheapry drugs and toxins) in the future [39,86].

There are many challenges facing nanoparticle-based drug delivery, and the most important of all is the (tumor) targeting efficacy. Although passive targeting based on the enhanced permeability and retention effect can be therapeutically efficacious in some cases and many liposome-based drugs have already been approved by the FDA for clinical use [39,87], this is by no means optimal. Long circulation half-life of nanoparticles is a double-edged sword. Although it can lead to higher level of passive targeting to the tumor, it also causes prolonged exposure of normal organs to the drug which can give rise to undesired toxicity. Based on the literature data to date, tumor vasculature targeting is the best bet for nanoparticles since many of these nanoparticles are too large to extravasate [8891]. A circulation half-life of several hours, rather than a few days or weeks for liposomes, may be sufficient to allow for efficient tumor (vasculature) targeting. Nanoparticles are usually large enough to enable multiple targeting aptamers on their surfaces to simultaneously bind to multiple targets, an aspect that has been virtually unexplored to date. Targeting multiple different, but closely-related, receptors by incorporating different targeting aptamers on the same nanoparticle, with spacers of suitable length, require robust chemistry to minimize batch-to-batch difference and improve the reproducibility. For nanoparticle-based drug delivery, aptamers should be selected against those targets that are expressed on the tumor vasculature, such as integrin αvβ3 [90,92,93], VEGF receptors [90,94], CD105/endoglin [95,96], Tie-2 [97,98], c-Met [99,100], etc.

The future of nanomedicine lies in multifunctional nanoplatforms which combine both therapeutic components and multimodality imaging (Fig. (5)). The integration of diagnostic imaging capability with therapeutic interventions, sometimes termed “theranostics” [101,102], is critical to addressing the challenges of cancer heterogeneity and adaptation. The ultimate goal of nanomedicine is that nanoparticle-based agents can allow for efficient, specific in vivo delivery of therapeutic agents (drugs, genes, radioisotopes, etc.) without systemic toxicity, and the dose delivered as well as the therapeutic efficacy can be accurately measured non-invasively over time. Much remains to be done before this can be a clinical reality. Continuous multidisciplinary efforts on the use/optimization of such nanoplatforms will shed new light on molecular diagnostics and molecular therapy. With the capacity to provide enormous sensitivity and flexibility, aptamer-conjugated nanoparticles have the potential to profoundly impact anti-cancer drug delivery and patient management in the near future.

Fig. 5.

Fig. 5

A multifunctional nanoplatform incorporating multiple receptor targeting with different aptamers, multimodality imaging, and multiple therapeutic entities. Not all functional moieties will be necessary and only suitably selected components are needed for each individual application. The various functional moieties may be either on the surface of or encapsulated inside the nanoparticle (NP).

Table 1.

A brief summary of aptamer-based drug delivery in vitro and in vivo.

Aptamer Target Anti-cancer drug Drug linkage Delivery vehicle In vivo/In vitro Ref.

A10 RNA PSMA Dox Intercalation A10 In vitro [27]
Sgc8c DNA PTK 7 Dox Hydrazone linker Sgc8c In vitro [29]
Sgc8 DNA PTK 7 Dau Intercalation Sgc8 In vitro [31]
A10 RNA PSMA Dtxl Encapsulation PLGA-b-PEG NP In vivo (intratumoral) [43]
A10 RNA PSMA Cisplatin Encapsulation PLGA-b-PEG NP In vitro [47]
A10 RNA PSMA Dox Intercalation QD In vitro [48]
A10 RNA PSMA Dox Intercalation TCL-SPION In vitro [51]
A9 RNA PSMA Dox Intercalation Gold NP In vitro [52]
A10 RNA & DUP1 Peptide Prostate cancer Dox Intercalation TCL-SPION In vitro [53]
A9 RNA PSMA Gelonin Covalent A9 In vitro [60]
DNA aptamer MUC1 glycoform Chlorin e6 Covalent DNA aptamer In vitro [61]
AS1411 Nucleolin TMPyP4 Intercalation & outside binding AS1411 In vitro [64]
A9 RNA PSMA Anti-lamin A/C siRNA Steptavidin-biotin A9 In vitro [74]
Truncated A10 RNA PSMA Anti-EEF2 siRNA Covalent Truncated A10 In vitro [75]
A10 RNA PSMA Anti-PLK1/BCL2 siRNA Covalent A10 RNA In vivo (Intratumoral) [76]
Optimized A10 RNA PSMA Anti-PLK1 siRNA Covalent Optimized A10 In vivo (Systemic) [78]
A10 RNA PSMA Dox and anti-Bcl- xL shRNA Intercalation & electrostatic PEI-PEG NP In vitro [79]

Acknowledgments

The authors acknowledge financial support from the University of Wisconsin Carbone Cancer Center, NCRR 1UL1RR025011, and a DOD PCRP IDEA Award.

References

  • 1.Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics. CA Cancer J Clin. 2011;61:69–90. doi: 10.3322/caac.20107. [DOI] [PubMed] [Google Scholar]
  • 2.Jemal A, Siegel R, Xu J, Ward E. Cancer statistics, 2010. CA Cancer J Clin. 2010;6:277–300. doi: 10.3322/caac.20073. [DOI] [PubMed] [Google Scholar]
  • 3.Rajendran L, Knolker HJ, Simons K. Subcellular targeting strategies for drug design and delivery. Nat Rev Drug Discov. 2010;9:29–42. doi: 10.1038/nrd2897. [DOI] [PubMed] [Google Scholar]
  • 4.Rosen H, Abribat T. The rise and rise of drug delivery. Nat Rev Drug Discov. 2005;4:381–385. doi: 10.1038/nrd1721. [DOI] [PubMed] [Google Scholar]
  • 5.Winquist RJ, Furey BF, Boucher DM. Cancer stem cells as the relevant biomass for drug discovery. Curr Opin Pharmacol. 2010;10:385–390. doi: 10.1016/j.coph.2010.06.008. [DOI] [PubMed] [Google Scholar]
  • 6.Singh A, Settleman J. EMT, cancer stem cells and drug resistance: an emerging axis of evil in the war on cancer. Oncogene. 2010;29:4741–4751. doi: 10.1038/onc.2010.215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Blagosklonny MV. Analysis of FDA approved anticancer drugs reveals the future of cancer therapy. Cell Cycle. 2004;3:1035–1042. [PubMed] [Google Scholar]
  • 8.Ellington AD, Szostak JW. In vitro selection of RNA molecules that bind specific ligands. Nature. 1990;346:818–822. doi: 10.1038/346818a0. [DOI] [PubMed] [Google Scholar]
  • 9.Tuerk C, Gold L. Systematic evolution of ligands by exponential enrichment: RNA ligands to bacteriophage T4 DNA polymerase. Science. 1990;249:505–510. doi: 10.1126/science.2200121. [DOI] [PubMed] [Google Scholar]
  • 10.Keefe AD, Pai S, Ellington A. Aptamers as therapeutics. Nat Rev Drug Discov. 2010;9:537–550. doi: 10.1038/nrd3141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Lin Y, Nieuwlandt D, Magallanez A, Feistner B, Jayasena SD. High-affinity and specific recognition of human thyroid stimulating hormone (hTSH) by in vitro-selected 2′-amino-modified RNA. Nucleic Acids Res. 1996;24:3407–3414. doi: 10.1093/nar/24.17.3407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Rusconi CP, Scardino E, Layzer J, Pitoc GA, Ortel TL, Monroe D, Sullenger BA. RNA aptamers as reversible antagonists of coagulation factor IXa. Nature. 2002;419:90–94. doi: 10.1038/nature00963. [DOI] [PubMed] [Google Scholar]
  • 13.Pagratis NC, Bell C, Chang YF, Jennings S, Fitzwater T, Jellinek D, Dang C. Potent 2′-amino-, and 2′-fluoro-2′-deoxyribonucleotide RNA inhibitors of keratinocyte growth factor. Nat Biotechnol. 1997;15:68–73. doi: 10.1038/nbt0197-68. [DOI] [PubMed] [Google Scholar]
  • 14.King DJ, Safar JG, Legname G, Prusiner SB. Thioaptamer interactions with prion proteins: sequence-specific and non-specific binding sites. J Mol Biol. 2007;369:1001–1014. doi: 10.1016/j.jmb.2007.02.004. [DOI] [PubMed] [Google Scholar]
  • 15.Takafuji Y, Jo JI, Tabata Y. Simple PEG Modification of DNA Aptamer Based on Copper Ion Coordination for Tumor Targeting. J Biomater Sci Polym Ed. 2010 doi: 10.1163/092050610X501713. [DOI] [PubMed] [Google Scholar]
  • 16.Eulberg D, Klussmann S. Spiegelmers: biostable aptamers. Chembiochem. 2003;4:979–983. doi: 10.1002/cbic.200300663. [DOI] [PubMed] [Google Scholar]
  • 17.Vater A, Klussmann S. Toward third-generation aptamers: Spiegelmers and their therapeutic prospects. Curr Opin Drug Discov Devel. 2003;6:253–261. [PubMed] [Google Scholar]
  • 18.Liss M, Petersen B, Wolf H, Prohaska E. An aptamer-based quartz crystal protein biosensor. Anal Chem. 2002;74:4488–4495. doi: 10.1021/ac011294p. [DOI] [PubMed] [Google Scholar]
  • 19.Mehlen P, Puisieux A. Metastasis: a question of life or death. Nat Rev Cancer. 2006;6:449–458. doi: 10.1038/nrc1886. [DOI] [PubMed] [Google Scholar]
  • 20.Weigelt B, Peterse JL, van ‘t Veer LJ. Breast cancer metastasis: markers and models. Nat Rev Cancer. 2005;5:591–602. doi: 10.1038/nrc1670. [DOI] [PubMed] [Google Scholar]
  • 21.Cullen JM, Miller RT. The role of pathology in the identification of drug-induced hepatic toxicity. Expert Opin Drug Metab Toxicol. 2006;2:241–247. doi: 10.1517/17425255.2.2.241. [DOI] [PubMed] [Google Scholar]
  • 22.Alberts DS, Delforge A. Maximizing the delivery of intraperitoneal therapy while minimizing drug toxicity and maintaining quality of life. Semin Oncol. 2006;33:8–17. doi: 10.1053/j.seminoncol.2006.11.003. [DOI] [PubMed] [Google Scholar]
  • 23.Toma S, Palumbo R, Sogno G, Venturino A, Santi L. Doxorubicin (or epidoxorubicin) combined with ifosfamide in the treatment of adult advanced soft tissue sarcomas. Ann Oncol. 1992;3(Suppl 2):S119–123. doi: 10.1093/annonc/3.suppl_2.s119. [DOI] [PubMed] [Google Scholar]
  • 24.Gehl J, Boesgaard M, Paaske T, Jensen BV, Dombernowsky P. Paclitaxel and doxorubicin in metastatic breast cancer. Semin Oncol. 1996;23:35–38. [PubMed] [Google Scholar]
  • 25.Visani G, Isidori A. Doxorubicin variants for hematological malignancies. Nanomedicine (Lond) 2011;6:303–306. doi: 10.2217/nnm.10.152. [DOI] [PubMed] [Google Scholar]
  • 26.Jain D. Cardiotoxicity of doxorubicin and other anthracycline derivatives. J Nucl Cardiol. 2000;7:53–62. doi: 10.1067/mnc.2000.103324. [DOI] [PubMed] [Google Scholar]
  • 27.Bagalkot V, Farokhzad OC, Langer R, Jon S. An aptamer-doxorubicin physical conjugate as a novel targeted drug-delivery platform. Angew Chem Int Ed Engl. 2006;45:8149–8152. doi: 10.1002/anie.200602251. [DOI] [PubMed] [Google Scholar]
  • 28.Lupold SE, Hicke BJ, Lin Y, Coffey DS. Identification and characterization of nuclease-stabilized RNA molecules that bind human prostate cancer cells via the prostate-specific membrane antigen. Cancer Res. 2002;62:4029–4033. [PubMed] [Google Scholar]
  • 29.Huang YF, Shangguan D, Liu H, Phillips JA, Zhang X, Chen Y, Tan W. Molecular assembly of an aptamer-drug conjugate for targeted drug delivery to tumor cells. Chembiochem. 2009;10:862–868. doi: 10.1002/cbic.200800805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Shangguan D, Li Y, Tang Z, Cao ZC, Chen HW, Mallikaratchy P, Sefah K, Yang CJ, Tan W. Aptamers evolved from live cells as effective molecular probes for cancer study. Proc Natl Acad Sci USA. 2006;103:11838–11843. doi: 10.1073/pnas.0602615103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Taghdisi SM, Abnous K, Mosaffa F, Behravan J. Targeted delivery of daunorubicin to T-cell acute lymphoblastic leukemia by aptamer. J Drug Target. 2010;18:277–281. doi: 10.3109/10611860903434050. [DOI] [PubMed] [Google Scholar]
  • 32.Cai W, Chen X. Nanoplatforms for targeted molecular imaging in living subjects. Small. 2007;3:1840–1854. doi: 10.1002/smll.200700351. [DOI] [PubMed] [Google Scholar]
  • 33.Sinha R, Kim GJ, Nie S, Shin DM. Nanotechnology in cancer therapeutics: bioconjugated nanoparticles for drug delivery. Mol Cancer Ther. 2006;5:1909–1917. doi: 10.1158/1535-7163.MCT-06-0141. [DOI] [PubMed] [Google Scholar]
  • 34.Ferrari M. Cancer nanotechnology: opportunities and challenges. Nat Rev Cancer. 2005;5:161–171. doi: 10.1038/nrc1566. [DOI] [PubMed] [Google Scholar]
  • 35.Service RF. Materials and biology. Nanotechnology takes aim at cancer. Science. 2005;310:1132–1134. doi: 10.1126/science.310.5751.1132. [DOI] [PubMed] [Google Scholar]
  • 36.Hong H, Zhang Y, Sun J, Cai W. Molecular imaging and therapy of cancer with radiolabeled nanoparticles. Nano Today. 2009;4:399–413. doi: 10.1016/j.nantod.2009.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Whitesides GM. Nanoscience, nanotechnology, and chemistry. Small. 2005;1:172–179. doi: 10.1002/smll.200400130. [DOI] [PubMed] [Google Scholar]
  • 38.Service RF. Molecular electronics. Nanodevices make fresh strides toward reality. Science. 2003;302:1310. doi: 10.1126/science.302.5649.1310. [DOI] [PubMed] [Google Scholar]
  • 39.Davis ME, Chen ZG, Shin DM. Nanoparticle therapeutics: an emerging treatment modality for cancer. Nat Rev Drug Discov. 2008;7:771–782. doi: 10.1038/nrd2614. [DOI] [PubMed] [Google Scholar]
  • 40.Farokhzad OC, Karp JM, Langer R. Nanoparticle-aptamer bioconjugates for cancer targeting. Expert Opin Drug Deliv. 2006;3:311–324. doi: 10.1517/17425247.3.3.311. [DOI] [PubMed] [Google Scholar]
  • 41.Levy-Nissenbaum E, Radovic-Moreno AF, Wang AZ, Langer R, Farokhzad OC. Nanotechnology and aptamers: applications in drug delivery. Trends Biotechnol. 2008;26:442–449. doi: 10.1016/j.tibtech.2008.04.006. [DOI] [PubMed] [Google Scholar]
  • 42.Lee JH, Yigit MV, Mazumdar D, Lu Y. Molecular diagnostic and drug delivery agents based on aptamer-nanomaterial conjugates. Adv Drug Deliv Rev. 2010;62:592–605. doi: 10.1016/j.addr.2010.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Farokhzad OC, Cheng J, Teply BA, Sherifi I, Jon S, Kantoff PW, Richie JP, Langer R. Targeted nanoparticle-aptamer bioconjugates for cancer chemotherapy in vivo. Proc Natl Acad Sci USA. 2006;103:6315–6320. doi: 10.1073/pnas.0601755103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Bleickardt E, Argiris A, Rich R, Blum K, McKeon A, Tara H, Zelterman D, Burtness B, Davies MJ, Murren JR. Phase I dose escalation trial of weekly docetaxel plus irinotecan in patients with advanced cancer. Cancer Biol Ther. 2002;1:646–651. doi: 10.4161/cbt.314. [DOI] [PubMed] [Google Scholar]
  • 45.Miller VA, Kris MG. Docetaxel (Taxotere) as a single agent and in combination chemotherapy for the treatment of patients with advanced non-small cell lung cancer. Semin Oncol. 2000;27:3–10. [PubMed] [Google Scholar]
  • 46.Sedletska Y, Giraud-Panis MJ, Malinge JM. Cisplatin is a DNA-damaging antitumour compound triggering multifactorial biochemical responses in cancer cells: importance of apoptotic pathways. Curr Med Chem Anticancer Agents. 2005;5:251–265. doi: 10.2174/1568011053765967. [DOI] [PubMed] [Google Scholar]
  • 47.Dhar S, Gu FX, Langer R, Farokhzad OC, Lippard SJ. Targeted delivery of cisplatin to prostate cancer cells by aptamer functionalized Pt(IV) prodrug-PLGA-PEG nanoparticles. Proc Natl Acad Sci USA. 2008;105:17356–17361. doi: 10.1073/pnas.0809154105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Bagalkot V, Zhang L, Levy-Nissenbaum E, Jon S, Kantoff PW, Langer R, Farokhzad OC. Quantum dot-aptamer conjugates for synchronous cancer imaging, therapy, and sensing of drug delivery based on bi-fluorescence resonance energy transfer. Nano Lett. 2007;7:3065–3070. doi: 10.1021/nl071546n. [DOI] [PubMed] [Google Scholar]
  • 49.Cai W, Hsu AR, Li ZB, Chen X. Are quantum dots ready for in vivo imaging in human subjects? Nanoscale Res Lett. 2007;2:265–281. doi: 10.1007/s11671-007-9061-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Li ZB, Cai W, Chen X. Semiconductor quantum dots for in vivo imaging. J Nanosci Nanotechnol. 2007;7:2567–2581. doi: 10.1166/jnn.2007.628. [DOI] [PubMed] [Google Scholar]
  • 51.Wang AZ, Bagalkot V, Vasilliou CC, Gu F, Alexis F, Zhang L, Shaikh M, Yuet K, Cima MJ, Langer R, Kantoff PW, Bander NH, Jon S, Farokhzad OC. Superparamagnetic iron oxide nanoparticle-aptamer bioconjugates for combined prostate cancer imaging and therapy. ChemMedChem. 2008;3:1311–1315. doi: 10.1002/cmdc.200800091. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Kim D, Jeong YY, Jon S. A drug-loaded aptamer-gold nanoparticle bioconjugate for combined CT imaging and therapy of prostate cancer. ACS Nano. 2010;4:3689–3696. doi: 10.1021/nn901877h. [DOI] [PubMed] [Google Scholar]
  • 53.Min K, Jo H, Song K, Cho M, Chun YS, Jon S, Kim WJ, Ban C. Dual-aptamer-based delivery vehicle of doxorubicin to both PSMA (+) and PSMA (−) prostate cancers. Biomaterials. 2011;32:2124–2132. doi: 10.1016/j.biomaterials.2010.11.035. [DOI] [PubMed] [Google Scholar]
  • 54.Zitzmann S, Mier W, Schad A, Kinscherf R, Askoxylakis V, Kramer S, Altmann A, Eisenhut M, Haberkorn U. A new prostate carcinoma binding peptide (DUP-1) for tumor imaging and therapy. Clin Cancer Res. 2005;11:139–146. [PubMed] [Google Scholar]
  • 55.Stirpe F, Olsnes S, Pihl A. Gelonin, a new inhibitor of protein synthesis, nontoxic to intact cells. Isolation, characterization, and preparation of cytotoxic complexes with concanavalin A. J Biol Chem. 1980;255:6947–6953. [PubMed] [Google Scholar]
  • 56.Rosenblum MG, Cheung L, Kim SK, Mujoo K, Donato NJ, Murray JL. Cellular resistance to the antimelanoma immunotoxin ZME-gelonin and strategies to target resistant cells. Cancer Immunol Immunother. 1996;42:115–121. doi: 10.1007/s002620050260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Selbo PK, Sivam G, Fodstad O, Sandvig K, Berg K. Photochemical internalisation increases the cytotoxic effect of the immunotoxin MOC31-gelonin. Int J Cancer. 2000;87:853–859. doi: 10.1002/1097-0215(20000915)87:6<853::aid-ijc15>3.0.co;2-0. [DOI] [PubMed] [Google Scholar]
  • 58.Hsu AR, Cai W, Veeravagu A, Mohamedali KA, Chen K, Kim S, Vogel H, Hou LC, Tse V, Rosenblum MG, Chen X. Multimodality molecular imaging of glioblastoma growth inhibition with vasculature-targeting fusion toxin VEGF121/rGel. J Nucl Med. 2007;48:445–454. [PubMed] [Google Scholar]
  • 59.Veenendaal LM, Jin H, Ran S, Cheung L, Navone N, Marks JW, Waltenberger J, Thorpe P, Rosenblum MG. In vitro and in vivo studies of a VEGF121/rGelonin chimeric fusion toxin targeting the neovasculature of solid tumors. Proc Natl Acad Sci USA. 2002;99:7866–7871. doi: 10.1073/pnas.122157899. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Chu TC, Marks JW, 3rd, Lavery LA, Faulkner S, Rosenblum MG, Ellington AD, Levy M. Aptamer:toxin conjugates that specifically target prostate tumor cells. Cancer Res. 2006;66:5989–5992. doi: 10.1158/0008-5472.CAN-05-4583. [DOI] [PubMed] [Google Scholar]
  • 61.Ferreira CS, Cheung MC, Missailidis S, Bisland S, Gariepy J. Phototoxic aptamers selectively enter and kill epithelial cancer cells. Nucleic Acids Res. 2009;37:866–876. doi: 10.1093/nar/gkn967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.McMahon KS, Wieman TJ, Moore PH, Fingar VH. Effects of photodynamic therapy using mono-L-aspartyl chlorin e6 on vessel constriction, vessel leakage, and tumor response. Cancer Res. 1994;54:5374–5379. [PubMed] [Google Scholar]
  • 63.Taber SW, Fingar VH, Coots CT, Wieman TJ. Photodynamic therapy using mono-L-aspartyl chlorin e6 (Npe6) for the treatment of cutaneous disease: a Phase I clinical study. Clin Cancer Res. 1998;4:2741–2746. [PubMed] [Google Scholar]
  • 64.Shieh YA, Yang SJ, Wei MF, Shieh MJ. Aptamer-based tumor-targeted drug delivery for photodynamic therapy. ACS Nano. 2010;4:1433–1442. doi: 10.1021/nn901374b. [DOI] [PubMed] [Google Scholar]
  • 65.Bates PJ, Laber DA, Miller DM, Thomas SD, Trent JO. Discovery and development of the G-rich oligonucleotide AS1411 as a novel treatment for cancer. Exp Mol Pathol. 2009;86:151–164. doi: 10.1016/j.yexmp.2009.01.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mongelard F, Bouvet P. AS-1411, a guanosine-rich oligonucleotide aptamer targeting nucleolin for the potential treatment of cancer, including acute myeloid leukemia. Curr Opin Mol Ther. 2010;12:107–114. [PubMed] [Google Scholar]
  • 67.Fire A, Xu S, Montgomery MK, Kostas SA, Driver SE, Mello CC. Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 1998;391:806–811. doi: 10.1038/35888. [DOI] [PubMed] [Google Scholar]
  • 68.Elbashir SM, Harborth J, Lendeckel W, Yalcin A, Weber K, Tuschl T. Duplexes of 21-nucleotide RNAs mediate RNA interference in cultured mammalian cells. Nature. 2001;411:494–498. doi: 10.1038/35078107. [DOI] [PubMed] [Google Scholar]
  • 69.Mello CC, Conte D., Jr Revealing the world of RNA interference. Nature. 2004;431:338–342. doi: 10.1038/nature02872. [DOI] [PubMed] [Google Scholar]
  • 70.Stevenson M. Therapeutic potential of RNA interference. N Engl J Med. 2004;351:1772–1777. doi: 10.1056/NEJMra045004. [DOI] [PubMed] [Google Scholar]
  • 71.Davis ME, Zuckerman JE, Choi CH, Seligson D, Tolcher A, Alabi CA, Yen Y, Heidel JD, Ribas A. Evidence of RNAi in humans from systemically administered siRNA via targeted nanoparticles. Nature. 2010;464:1067–1070. doi: 10.1038/nature08956. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Hong H, Zhang Y, Cai W. In vivo imaging of RNA interference. J Nucl Med. 2010;51:169–172. doi: 10.2967/jnumed.109.066878. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Sioud M. On the delivery of small interfering RNAs into mammalian cells. Expert Opin Drug Deliv. 2005;2:639–651. doi: 10.1517/17425247.2.4.639. [DOI] [PubMed] [Google Scholar]
  • 74.Chu TC, Twu KY, Ellington AD, Levy M. Aptamer mediated siRNA delivery. Nucleic Acids Res. 2006;34:e73. doi: 10.1093/nar/gkl388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Wullner U, Neef I, Eller A, Kleines M, Tur MK, Barth S. Cell-specific induction of apoptosis by rationally designed bivalent aptamer-siRNA transcripts silencing eukaryotic elongation factor 2. Curr Cancer Drug Targets. 2008;8:554–565. doi: 10.2174/156800908786241078. [DOI] [PubMed] [Google Scholar]
  • 76.McNamara JO, 2nd, Andrechek ER, Wang Y, Viles KD, Rempel RE, Gilboa E, Sullenger BA, Giangrande PH. Cell type-specific delivery of siRNAs with aptamer-siRNA chimeras. Nat Biotechnol. 2006;24:1005–1015. doi: 10.1038/nbt1223. [DOI] [PubMed] [Google Scholar]
  • 77.Sledz CA, Holko M, de Veer MJ, Silverman RH, Williams BR. Activation of the interferon system by short-interfering RNAs. Nat Cell Biol. 2003;5:834–839. doi: 10.1038/ncb1038. [DOI] [PubMed] [Google Scholar]
  • 78.Dassie JP, Liu XY, Thomas GS, Whitaker RM, Thiel KW, Stockdale KR, Meyerholz DK, McCaffrey AP, McNamara JO, 2nd, Giangrande PH. Systemic administration of optimized aptamer-siRNA chimeras promotes regression of PSMA-expressing tumors. Nat Biotechnol. 2009;27:839–849. doi: 10.1038/nbt.1560. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Kim E, Jung Y, Choi H, Yang J, Suh JS, Huh YM, Kim K, Haam S. Prostate cancer cell death produced by the co-delivery of Bcl-xL shRNA and doxorubicin using an aptamer-conjugated polyplex. Biomaterials. 2010;31:4592–4599. doi: 10.1016/j.biomaterials.2010.02.030. [DOI] [PubMed] [Google Scholar]
  • 80.Missailidis S, Hardy A. Aptamers as inhibitors of target proteins. Expert Opin Ther Pat. 2009;19:1073–1082. doi: 10.1517/13543770903042337. [DOI] [PubMed] [Google Scholar]
  • 81.Reiter NJ, Maher LJ, 3rd, Butcher SE. DNA mimicry by a high-affinity anti-NF-kappaB RNA aptamer. Nucleic Acids Res. 2008;36:1227–1236. doi: 10.1093/nar/gkm1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Maasch C, Vater A, Buchner K, Purschke WG, Eulberg D, Vonhoff S, Klussmann S. Polyetheylenimine-polyplexes of Spiegelmer NOX-A50 directed against intracellular high mobility group protein A1 (HMGA1) reduce tumor growth in vivo. J Biol Chem. 2010;285:40012–40018. doi: 10.1074/jbc.M110.178533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Thiel KW, Giangrande PH. Therapeutic applications of DNA and RNA aptamers. Oligonucleotides. 2009;19:209–222. doi: 10.1089/oli.2009.0199. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Fang X, Tan W. Aptamers generated from cell-SELEX for molecular medicine: a chemical biology approach. Acc Chem Res. 2010;43:48–57. doi: 10.1021/ar900101s. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Mayer G. The chemical biology of aptamers. Angew Chem Int Ed Engl. 2009;48:2672–2689. doi: 10.1002/anie.200804643. [DOI] [PubMed] [Google Scholar]
  • 86.Medarova Z, Pham W, Farrar C, Petkova V, Moore A. In vivo imaging of siRNA delivery and silencing in tumors. Nat Med. 2007;13:372–377. doi: 10.1038/nm1486. [DOI] [PubMed] [Google Scholar]
  • 87.Peer D, Karp JM, Hong S, Farokhzad OC, Margalit R, Langer R. Nanocarriers as an emerging platform for cancer therapy. Nat Nanotechnol. 2007;2:751–760. doi: 10.1038/nnano.2007.387. [DOI] [PubMed] [Google Scholar]
  • 88.Cai W, Chen K, Li ZB, Gambhir SS, Chen X. Dual-function probe for PET and near-infrared fluorescence imaging of tumor vasculature. J Nucl Med. 2007;48:1862–1870. doi: 10.2967/jnumed.107.043216. [DOI] [PubMed] [Google Scholar]
  • 89.Cai W, Chen X. Preparation of peptide conjugated quantum dots for tumour vasculature targeted imaging. Nat Protoc. 2008;3:89–96. doi: 10.1038/nprot.2007.478. [DOI] [PubMed] [Google Scholar]
  • 90.Cai W, Chen X. Multimodality molecular imaging of tumor angiogenesis. J Nucl Med. 2008;49(Suppl 2):113S–128S. doi: 10.2967/jnumed.107.045922. [DOI] [PubMed] [Google Scholar]
  • 91.Cai W, Shin DW, Chen K, Gheysens O, Cao Q, Wang SX, Gambhir SS, Chen X. Peptide-labeled near-infrared quantum dots for imaging tumor vasculature in living subjects. Nano Lett. 2006;6:669–676. doi: 10.1021/nl052405t. [DOI] [PubMed] [Google Scholar]
  • 92.Mi J, Zhang X, Giangrande PH, McNamara JO, 2nd, Nimjee SM, Sarraf-Yazdi S, Sullenger BA, Clary BM. Targeted inhibition of alphavbeta3 integrin with an RNA aptamer impairs endothelial cell growth and survival. Biochem Biophys Res Commun. 2005;338:956–963. doi: 10.1016/j.bbrc.2005.10.043. [DOI] [PubMed] [Google Scholar]
  • 93.Cai W, Niu G, Chen X. Imaging of integrins as biomarkers for tumor angiogenesis. Curr Pharm Des. 2008;14:2943–2973. doi: 10.2174/138161208786404308. [DOI] [PubMed] [Google Scholar]
  • 94.Cai W, Chen X. Multimodality imaging of vascular endothelial growth factor and vascular endothelial growth factor receptor expression. Front Biosci. 2007;12:4267–4279. doi: 10.2741/2386. [DOI] [PubMed] [Google Scholar]
  • 95.Cai W, Rao J, Gambhir SS, Chen X. How molecular imaging is speeding up anti-angiogenic drug development. Mol Cancer Ther. 2006;5:2624–2633. doi: 10.1158/1535-7163.MCT-06-0395. [DOI] [PubMed] [Google Scholar]
  • 96.Zhang Y, Yang Y, Hong H, Cai W. Multimodality molecular imaging of CD105 (Endoglin) expression. Int J Clin Exp Med. 2011;4:32–42. [PMC free article] [PubMed] [Google Scholar]
  • 97.Kobayashi H, Lin PC. Angiopoietin/Tie2 signaling, tumor angiogenesis and inflammatory diseases. Front Biosci. 2005;10:666–674. doi: 10.2741/1561. [DOI] [PubMed] [Google Scholar]
  • 98.Shim WS, Ho IA, Wong PE. Angiopoietin: a TIE(d) balance in tumor angiogenesis. Mol Cancer Res. 2007;5:655–665. doi: 10.1158/1541-7786.MCR-07-0072. [DOI] [PubMed] [Google Scholar]
  • 99.Yap TA, de Bono JS. Targeting the HGF/c-Met axis: state of play. Mol Cancer Ther. 2010;9:1077–1079. doi: 10.1158/1535-7163.MCT-10-0122. [DOI] [PubMed] [Google Scholar]
  • 100.You WK, McDonald DM. The hepatocyte growth factor/c-Met signaling pathway as a therapeutic target to inhibit angiogenesis. BMB Rep. 2008;41:833–839. doi: 10.5483/bmbrep.2008.41.12.833. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Del Vecchio S, Zannetti A, Fonti R, Pace L, Salvatore M. Nuclear imaging in cancer theranostics. Q J Nucl Med Mol Imaging. 2007;51:152–163. [PubMed] [Google Scholar]
  • 102.Sumer B, Gao J. Theranostic nanomedicine for cancer. Nanomed. 2008;3:137–140. doi: 10.2217/17435889.3.2.137. [DOI] [PubMed] [Google Scholar]

RESOURCES