What Aptamer drug conjugates are being developed?

17 March 2025
Introduction to Aptamers

Definition and Characteristics
Aptamers are short, single-stranded oligonucleotides—either DNA or RNA—that fold into unique three-dimensional structures enabling them to bind target molecules with high affinity and specificity. Because of their ability to be chemically synthesized, they exhibit low batch-to-batch variability compared to biologically produced antibodies. Their small size (typically 5–15 kDa) provides excellent tissue penetration, while chemical modifications such as 2′-O-methyl, 2′-fluoro substitutions, phosphorothioate backbones, and PEGylation enhance their stability against nuclease degradation and extend their circulation half-life in vivo. These inherent properties, such as high selectivity, ease of manufacture, chemical modifiability, and minimal immunogenicity, make them “chemical antibodies” that are particularly attractive in both diagnostic and therapeutic applications.

Overview of Aptamers in Drug Development
Over the past few decades, aptamers have evolved from laboratory selection tools using SELEX (Systematic Evolution of Ligands by Exponential Enrichment) into clinically relevant agents in drug development. Initially developed to study protein functions, aptamers are now employed either as active therapeutic agents that inhibit target proteins or as targeting ligands in drug delivery systems. Their ability to be easily modified by conjugation with various therapeutics—from small molecule drugs to large cargos such as siRNAs, toxins, or even nanoparticles—has made them a powerful platform for designing novel therapeutic conjugates. With advancements in high-throughput screening, chemical synthesis, and modifications that improve stability and targeting efficiency, aptamers have shifted from basic research to translational applications, fueling a growing field in aptamer-based drug delivery.

Aptamer Drug Conjugates

Mechanism of Action
Aptamer drug conjugates—or ApDCs—function by combining the targeting capabilities of aptamers with the cytotoxic or therapeutic payload of a drug. The basic mechanism involves the aptamer binding specifically to a target cell marker or receptor, thereby guiding the drug directly to diseased cells while sparing healthy cells. In many cases, the drug is attached covalently or via a linking group to the aptamer. Some ApDCs are constructed through solid-phase synthesis processes, where therapeutics, such as chemotherapeutic agents, are directly incorporated into the sequence as modified nucleotides; others use post-synthesis conjugation strategies with cleavable linkers or physical embedding methods. Upon binding to the target cell, the aptamer facilitates receptor-mediated endocytosis. Once inside the cell or in the tumor microenvironment, stimuli such as pH changes or enzymatic cleavage trigger the release of the therapeutic agent. This spatially and temporally controlled release reduces systemic toxicity and off-target effects, addressing one of the most significant limitations of conventional chemotherapy.

Advantages over Traditional Drug Conjugates
Aptamer drug conjugates offer several benefits over traditional antibody-drug conjugates (ADCs) and other combinatorial drug approaches.
- Size and Tissue Penetration: Owing to their small size, aptamers can penetrate tissues more efficiently than antibodies, allowing deeper access to tumor microenvironments or other hard-to-reach tissues.
- Chemical Synthesis and Modification: Aptamers are produced by chemical synthesis rather than recombinant cell culture. This process allows for precise modifications, such as incorporation of non-natural nucleotides or linkage chemistries that improve stability and targeting.
- Low Immunogenicity: Unlike protein-based antibodies, aptamers generally do not trigger immune responses, which reduces potential immunogenicity and allergic reactions.
- Rapid and Cost‐Effective Production: The in vitro selection process via SELEX can produce aptamers within weeks, and their synthesis is scalable, offering cost benefits without the need for animal immunization or complex cell culture processes.
- Ease of Payload Incorporation: Aptamers can be engineered to include multiple copies of therapeutic agents directly into their structure or attached via sophisticated linkers, which can be designed for stimuli-responsive release in cellular environments.
- Flexibility in Design: The versatility of aptamer chemical modifications allows for designing drug conjugates with controllable pharmacokinetics, enhanced serum stability, and precise release mechanisms that are less feasible with traditional antibody-based systems.

Current Developments

Examples of Aptamer Drug Conjugates
Several aptamer drug conjugates are being developed across a range of therapeutic areas with a particular focus on cancer treatment. Some notable examples include:

- Full Phosphorothioate Modified Nucleic Acid Aptamer Conjugates:
Hunan University has developed a full phosphorothioate modified aptamer drug conjugate that comprises a drug molecule group (selectable from mitomycin C, a known chemotherapeutic agent) chemically linked to an aptamer fragment whose backbone is entirely replaced by phosphorothioate bonds. This modification not only imparts high nuclease resistance but also prolongs the circulatory half-life of the conjugate while maintaining target specificity. This design demonstrates that by altering the chemical backbone, one can significantly improve the clinical potential of ApDCs.

- Polypeptide-Nucleic Acid Aptamer Drug Conjugates:
Another innovative approach involves the creation of hybrid conjugates that combine a peptide moiety with an aptamer fragment. In this construct, a polypeptide fragment—selected for its ability to target proteins like HSP70—is covalently linked to a nucleic acid aptamer that targets tumor cells. The aptamer portion directs the conjugate to the cancer cells, while the peptide enhances cytotoxicity and enables the delivery of a drug molecule embedded within the aptamer structure. This dual-targeting method improves the killing of drug-resistant tumor cells and minimizes the side effects associated with conventional chemotherapeutics.

- Artemisinin Derivative Nucleic Acid Aptamer Conjugates:
An artemisinin derivative conjugate has been developed whereby an artesunate group is attached to a nucleic acid aptamer fragment. The nucleic acid sequence is carefully designed (including specific sequences like SEQ ID NO.1) to confer targeting specificity toward cells overexpressing proteins such as PTK7. This ApDC displays improved water solubility and strong cytotoxicity in cell experiments, indicating significant promise for targeting tumor cells with precision.

- Direct Incorporation Aptamer-Drug Conjugates:
Some strategies eliminate the need for post-synthesis modification by integrating drug molecules directly into the oligonucleotide synthesis process. For instance, a nucleic acid aptamer-drug conjugate can be synthesized where a phosphoramidite monomer of the drug (such as a chemotherapeutic agent analog) is incorporated into the DNA chain via phosphodiester bonds. This “one-pot” synthesis method ensures a high degree of connectivity between the aptamer and the drug, preserving both the specificity of the aptamer and the anticancer efficacy of the drug.

- Mitomycin C Linked Aptamer Conjugates:
Variations of aptamer conjugates incorporating mitomycin C have been explored in which the drug is linked to the aptamer fragment using specifically designed linking groups. Mitomycin C, which is a nonspecific anticancer agent, suffers from dose-limiting toxicities; therefore, its conjugation to an aptamer significantly improves its targeting capability, ensuring that the cytotoxic agent is preferentially delivered to tumor cells.

- 5′-Modified Aptamer Conjugates with Enhanced Stability:
Several designs also focus on appending small molecule drugs at the 5′ end of the aptamer, using chemical modifications that enhance stability against serum nucleases while preserving target binding affinity. One such construct incorporates a 5′-modified aptamer linked with a small molecule drug, where the aptamer segment includes a sequence such as SEQ ID NO.1 that is critical for binding the target. This modification has been shown to simultaneously improve serum stability and ensure efficient intracellular delivery upon receptor-mediated endocytosis.

- Aptamer Toxin Conjugates:
In addition to conjugates with small molecule chemotherapeutics, aptamer-toxin conjugates are being developed wherein a cytotoxic component (typically a toxin or a ribosome-inactivating protein) is attached to an aptamer. Such constructs leverage the exquisite targeting of the aptamer to deliver the toxin specifically to malignant cells, achieving potent cell killing with reduced systemic toxicity.

- Combinatorial and Hybrid Conjugates:
There is also considerable interest in hybrid systems that combine aptamers with other targeting molecules, such as antibodies or peptides, to form multivalent or bispecific conjugates. These systems can improve binding affinity via multimerization effects and may deliver multiple drugs simultaneously, thereby overcoming drug resistance and enhancing the apoptotic response in cancer cells. For instance, antibody–aptamer conjugates (oligobodies) or aptamer–siRNA chimeras have been explored to integrate multiple therapeutic modalities into one construct.

Clinical Trials and Research Studies
Clinical research on aptamer drug conjugates has been steadily expanding. Although aptamer-based therapies have historically faced some setbacks (as seen with early products such as pegaptanib, which was introduced over a decade ago), the new generation of ApDCs shows marked improvements in stability, targeting efficiency, and payload delivery. Clinical evaluation of several ApDCs is underway:

- Phosphorothioate Modified Aptamer Conjugates:
The phosphorothioate backbone modification approach is now under advanced preclinical investigation, with several candidates demonstrating superior nuclease resistance and prolonged blood circulation in animal models. Such enhanced pharmacokinetics have encouraged translational studies aimed at evaluating their efficacy in targeting aggressive tumor types.

- Polypeptide-Aptamer Conjugates in Drug-Resistant Cancer:
Research studies have shown that polypeptide–nucleic acid aptamer conjugates, owing to their dual targeting and synergistic cytotoxic effects, are effective against drug-resistant cancer cells. These conjugates have advanced into early-phase clinical trials where biomarkers such as HSP70 expression are monitored to determine patient eligibility and therapeutic outcomes.

- Artemisinin Derivative Conjugates for Targeted Therapy:
The promising preclinical data generated with artesunate-aptamer conjugates (targeting PTK7-overexpressing cells) have led to proposals for clinical evaluation. Researchers are particularly interested in their improved water solubility and selective cytotoxicity, which may translate into better patient outcomes with less off-target toxicity.

- Direct Incorporation and One-Pot Synthesized Conjugates:
The innovative technique of incorporating drug nucleoside analogs directly during aptamer synthesis is being validated in cell-based assays and animal models. Early studies have indicated that these conjugates have high drug loading, controlled intracellular release profiles, and potent anticancer effects, setting the stage for future clinical trials.

- Mitomycin C Aptamer Conjugates:
Early-stage clinical research in mitomycin C-based ApDCs has shown that the targeted delivery provided by the aptamer significantly reduces systemic toxicity while retaining the cytotoxic potency against tumor cells. Such outcomes are particularly promising for cancers where chemotherapeutic resistance and off-target effects are major obstacles.

- 5′-Modified Aptamer Conjugates:
Clinical studies investigating 5′-modified aptamers demonstrate improved drug stability and enhanced tumor cell uptake. These conjugates are currently being evaluated for their pharmacodynamic properties and overall therapeutic index in phase I/II clinical trials.

- Aptamer-toxin Conjugates:
Toxin-based ApDCs are under preclinical development with several candidates showing impressive selectivity and cytotoxicity in in vitro models. These candidates are being taken forward with plans for future clinical trials, particularly for cancers that are refractory to conventional treatments.

Overall, the body of research, including both preclinical models and early clinical trials, indicates that aptamer drug conjugates are progressing from concept to clinical application. Numerous scientific articles and reviews have underscored the steady improvement in synthesis, modification, and conjugation techniques that are driving this advancement. The results from both in vitro and in vivo studies confirm that ApDCs can deliver multiple types of therapeutics with high specificity and minimal collateral damage.

Challenges and Future Prospects

Current Development Challenges
Despite the promising results, several challenges must be addressed before ApDCs become standard therapeutic modalities in clinical settings:

- Stability and Pharmacokinetics:
Although chemical modifications such as phosphorothioate backbones, PEGylation, and sugar modifications considerably improve aptamer stability, even slight degradation in vivo can reduce the efficacy of the ApDC. Balancing the need for prolonged circulation with rapid cleavage at the target site remains a substantial challenge.
- Manufacturing and Scale-Up:
While aptamers are produced by chemical synthesis on a scalable platform, the high cost of modified nucleotides and custom linkers may pose limitations to large-scale manufacture, particularly for ApDCs that require multiple chemical modifications.
- Target Specificity and Off-Target Effects:
Although aptamers are selected for high specificity, the in vivo milieu presents challenges such as nonspecific binding or unintended uptake by normal cells. Furthermore, there is a necessity to validate that the aptamer’s structure and binding affinity are maintained under physiological conditions.
- Drug Payload and Release Mechanisms:
Ensuring a high drug loading capacity and controlled release upon target binding are critical. Strategies like incorporating photocleavable linkers or enzymatically cleavable groups have shown promise; however, optimizing these release mechanisms to prevent premature drug release in circulation is still an area of active research.
- Immunogenicity and Toxicity:
Although aptamers are largely non-immunogenic, the conjugated drugs and the modifications themselves might elicit an immune response or have unforeseen toxicities. Extensive preclinical studies are needed to verify the safety of each ApDC formulation.

Future Research Directions and Potential Applications
Future directions in ApDC research will likely focus on several key areas that will further refine and expand their clinical use:

- Enhancement of Stability and Controlled Release:
Future research will continue to explore novel chemical modifications and linker technologies that enable precise control over the release of a cytotoxic agent in response to specific intracellular triggers (e.g., pH changes, redox conditions, or enzymatic activity). Innovations in “smart” linkers that respond dynamically to the tumor microenvironment are anticipated to improve therapeutic efficacy.
- Integration with Nanotechnology and Hybrid Systems:
Combinatorial systems integrating aptamers with nanoparticles, dendrimers, and polymeric carriers are an emerging trend. For example, gated nanoparticles with aptamer “switches” have been developed for stimuli-responsive drug release, thereby optimizing the localized delivery of high doses of chemotherapy while minimizing systemic exposure.
- Multifunctional Aptamer Conjugates:
Avenues such as bispecific or multifunctional aptamer drug conjugates that couple targeting ligands with additional therapeutic modalities (e.g., siRNA, immunotherapeutics, or photosensitizers) are under exploration. Such hybrid conjugates may yield synergistic effects, address tumor heterogeneity, and overcome resistance mechanisms.
- Personalized Medicine and Precision Targeting:
Advances in cell-based SELEX and next-generation sequencing may lead to the development of personalized aptamer-based treatments, where aptamer selection is tailored to the specific molecular profile of an individual’s tumor. This approach would enable high precision in drug delivery and maximize therapeutic benefits relative to side effects.
- Expanded Therapeutic Indications:
Although much of the current focus has been on anticancer therapies, aptamer drug conjugates are also being explored for other conditions, including cardiovascular diseases, autoimmune disorders, infections, and ophthalmological applications. For instance, aptamer-based antagonists targeting vascular endothelial growth factor (VEGF) have been successfully used in age-related macular degeneration, and similar principles can be adopted for other systemic diseases.
- Regulatory and Clinical Trial Optimization:
With ongoing and future clinical trials, it will be important to develop standardized protocols for evaluating ApDC safety, efficacy, dosages, and pharmacokinetic profiles. Harmonization of regulatory guidelines and robust clinical study designs are essential for translating preclinical success into approved clinical therapies.
- Cost-Effectiveness and Manufacturing Innovations:
As improvements in synthesis techniques and automation become more widespread, the overall cost of producing ApDCs is expected to drop significantly. Innovations such as PCR-based amplification for aptamer synthesis and solid-phase automated drug conjugation processes are potential game-changers in this regard.

Conclusion
In summary, a wide array of aptamer drug conjugates are being developed with the overarching goal of combining the superb targeting properties of aptamers with potent therapeutic payloads. These conjugates include fully phosphorothioate-modified nucleic acid aptamers linked with agents such as mitomycin C, polypeptide–aptamer chimeras incorporating peptide fragments for improved cytotoxicity, artemisinin derivative conjugates targeting PTK7, direct drug incorporation through phosphoramidite chemistry, and 5′-modified aptamers that enhance serum stability and intracellular delivery. Additionally, aptamer-toxin conjugates and multifunctional constructs combining aptamers with antibodies or siRNA are being investigated to broaden the scope of targeted therapies.

These developments demonstrate, in a general-specific-general structure, that the field of aptamer-based drug conjugates has matured from basic principles of molecular recognition into sophisticated therapeutic systems capable of precise drug delivery. On the one hand, the general advantages of aptamers—such as their small size, ease of chemical modification, high specificity, and low immunogenicity—set the stage for developing next-generation therapeutic agents. On the other hand, the specific designs, including advanced chemical modifications and smart linker technologies, are addressing critical issues like stability, controlled release, and off-target toxicity. Finally, looking at the broader perspective, the intensive preclinical and emerging clinical research signifies a promising future for ApDCs in oncology and beyond, with anticipated applications in cardiovascular, autoimmune, and ophthalmologic disorders.

Despite these promising advances, several challenges remain. Issues related to enhanced stability in vivo, cost-effective manufacturing, precise control over drug release, and comprehensive safety evaluation must be overcome to fully harness the potential of aptamer drug conjugates. Future research is expected to focus on integrating advanced nanotechnology, developing multifunctional hybrid systems, targeting personalization, and refining regulatory frameworks to facilitate clinical translation.

In conclusion, aptamer drug conjugates represent a rapidly evolving field that promises high precision in drug delivery with reduced systemic toxicity and improved patient outcomes. With ongoing innovations in chemical synthesis, molecular engineering, and clinical research, these systems are likely to play a vital role in the future of targeted therapeutics, ushering in a new era of personalized and effective treatments for a myriad of diseases.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成