What are the different types of drugs available for Virus-like Drug Conjugates (VDCs)?

17 March 2025
Introduction to Virus-like Drug Conjugates (VDCs)

Definition and Basic Concepts
Virus-like Drug Conjugates (VDCs) represent an innovative class of drug delivery systems that leverage the inherent properties of virus-like particles (VLPs) to transport therapeutic agents to targeted sites. VLPs mimic the structural features of real viruses without carrying the infectious genetic material, thereby offering a safe, non-replicative scaffold for drug delivery. The conjugation process involves covalently attaching drugs to the capsid proteins of these particles, which allows for multivalent drug display, high payload capacity, and unique bio-distribution characteristics. The formulation is designed so that the drug-carrier complex, while retaining the advantageous features of viral binding, can be administered systemically with reduced off-target toxicity. This concept is underpinned by decades of research in nanotechnology, virology, and medicinal chemistry, which collectively enable scientists to harness the natural targeting mechanisms of viruses without exposing patients to the risks associated with viral replication.

Overview of VDCs in Drug Delivery
VDCs have emerged as a promising modality particularly in oncology, where conventional chemotherapeutic regimens are often limited by non-specific toxicity and suboptimal biodistribution. By employing VLPs as the foundation, VDCs enable targeted drug delivery by exploiting the preferential binding of viral capsids to receptors overexpressed on tumor cells – such as modified heparan sulfate proteoglycans (HSPGs). The dual mandate of these conjugates is not only to deliver cytotoxic agents directly into cancer cells but also to initiate subsequent cell death mechanisms that may include immune-mediated responses. Recent clinical studies underscore the potential of VDCs: for example, Belzupacap sarotalocan has achieved Phase 3 status in clinical development, and MLT-103 is still in Phase 1, providing early evidence that VDCs can safely and effectively deliver payloads in vivo. This rapid evolution from concept to clinical development highlights the translational promise of VDCs as a next-generation therapeutic platform.

Types of Drugs Used in VDCs

The broad spectrum of therapeutics available for conjugation with virus-like particles can be categorized into three main types: small molecule drugs, biologics, and nucleic acid-based drugs. Each category presents its own set of physicochemical characteristics, therapeutic profiles, challenges in conjugation, and mechanisms of action.

Small Molecule Drugs
Small molecule drugs constitute the most traditional and widely used payload in VDCs. Their low molecular weight and solubility profiles make them amenable to chemical modifications that allow for covalent coupling onto VLPs. In the context of VDCs:
- Cytotoxic Agents: Many VDCs are designed to deliver potent anticancer drugs such as microtubule disruptors or DNA-damaging agents. These agents, when conjugated to VLPs, maintain their intrinsic cytotoxicity while gaining improved targeting and reduced systemic exposure. For instance, Belzupacap sarotalocan is an example of a small molecule payload specifically optimized for precision delivery to tumors, thereby reducing traditional side effects associated with chemotherapeutic agents.
- Chemotherapeutic Modulators: Some small molecules in VDCs are designed to overcome multidrug resistance mechanisms by directly interfering with cellular drug efflux pathways or altering intracellular drug trafficking. Such molecules often feature modifications that enhance their retention when delivered via VLPs, thereby increasing their intracellular concentration and therapeutic efficacy.
- Advantages and Challenges: Small molecules offer the benefits of rapid diffusion, well-defined structure, and ease of synthesis. However, their small size may sometimes result in premature dissociation from the VLP scaffold or off-target toxicity if not adequately stabilized by the linker chemistry. The precise engineering of linkers that are responsive to tumor-specific cues (for instance, pH or enzymatic activity) is critical in ensuring that the active drug is released at the intended site.

Biologics
Biologics refer to larger, complex molecules such as peptides, proteins, or even antibodies, which can be employed either as the active payload or as part of a therapeutic strategy in conjunction with VDCs. Key aspects of using biologics in VDCs include:
- Direct Therapeutic Proteins: These can include enzymes, receptors, or even antibody fragments that are delivered directly to modulate intracellular signaling or immune responses. The protein component benefits from the VLP’s ability to shield it from proteolytic degradation while enhancing its circulation time and uptake into target cells.
- Immunomodulatory Peptides: In some cases, VDCs are designed to trigger an immune response by delivering peptides that serve as antigens or immune adjuvants. These peptides can stimulate both the innate and adaptive arms of the immune system, thus working synergistically with the cytotoxic payload.
- Advantages and Challenges: Biologics are inherently specific, given their larger size and complex tertiary structures, which allow them to interact with cellular receptors with high affinity. However, they also face higher production costs, a higher risk of immunogenicity, and challenges associated with maintaining structural integrity during conjugation and delivery. The careful design of the conjugation chemistry, sometimes requiring site-specific attachment strategies, is paramount to preserve biologics’ activity when coupled to VLPs.

Nucleic Acid-based Drugs
Nucleic acid-based drugs, which include DNA, messenger RNA (mRNA), small interfering RNA (siRNA), and antisense oligonucleotides (ASOs), represent a rapidly evolving frontier in therapeutic development. Their integration into VDCs offers several potential advantages:
- Gene Replacement and Expression: Plasmid DNA or mRNA payloads can be delivered to restore the expression of deficient proteins, making them especially useful in genetic disorders and certain cancers where the restoration of tumor suppressor genes may be beneficial.
- Gene Silencing: The use of siRNA or ASOs enables the downregulation of specific target genes implicated in disease progression. When delivered by VLPs, these nucleic acid compounds are more stable, have enhanced uptake, and can overcome limitations related to nuclease degradation that often plague free nucleic acid therapeutics.
- Advantages and Challenges: Nucleic acid drugs offer precise sequence specificity and the promise to modulate gene expression at the molecular level. However, their large size, negative charge, and susceptibility to enzymatic degradation require sophisticated delivery strategies. VLPs provide an ideal shield for these molecules; nevertheless, the conjugation process must be optimized to ensure that the nucleic acid payload remains functionally intact after release. Moreover, the cellular uptake and endosomal escape of nucleic acids delivered via VDCs remain active areas of research, with ongoing studies focusing on improving intracellular release mechanisms.

Mechanisms of Action

The effectiveness of VDCs lies in their dual ability to deliver the therapeutic payload and ensure its subsequent release at the specific target site. Two overarching mechanisms shape the action of these conjugates: drug delivery mechanisms and targeting strategies.

Drug Delivery Mechanisms
VDCs harness the natural capability of virus-like particles to interact with and penetrate cell membranes. The underlying mechanisms include:
- Multivalent Targeting: VDCs can display multiple copies of the drug on their surface due to the repetitive nature of viral capsid proteins. This multivalent display enhances the binding avidity to cell surface receptors and facilitates uptake by receptor-mediated endocytosis.
- Controlled Drug Release: The linker chemistry used in VDCs is often designed to be responsive to specific intracellular stimuli – such as low pH, specific enzymes, or redox conditions – ensuring that once the conjugate is internalized, the drug is released in a controlled manner. For example, acid-labile hydrazone linkers are frequently used to trigger drug release in the acidic environment of the endosome or lysosome.
- Protection and Stability: One of the advantages of coupling a drug to a VLP is the protection afforded to the drug from premature degradation in the bloodstream. The VLP acts as a nano-shield, reducing the impact of serum enzymes and immune recognition, and thereby prolonging circulation time.
- Enhanced Permeability and Retention (EPR) Effect: Particularly in tumor tissues, the leaky vasculature combined with poor lymphatic drainage allows for an enhanced accumulation of VDCs at the target site via the EPR effect. This phenomenon further amplifies the payload delivered by the VDCs into the tumor microenvironment.

Targeting Strategies
Targeting is a critical dimension of VDC function, ensuring selective drug delivery with minimal systemic side effects. The targeting strategies employed include:
- Receptor-mediated Binding: VLPs possess innate capabilities, often derived from their natural viral origins, to bind specifically to receptors on target cells. For instance, VLPs can recognize and bind to modified HSPGs, which are overexpressed on a variety of cancer cells. This selective binding ensures that the drug conjugate accumulates preferentially in tumor tissues.
- Ligand Attachment and Dual Targeting: In some VDC designs, additional targeting ligands, such as peptides or antibodies, are conjugated to the viral capsid. These ligands can offer secondary binding interactions, thereby increasing specificity and improving the internalization of the conjugate.
- Stimuli-responsive Targeting: Some VDC systems incorporate switchable components – such as photosensitive groups or magnetic nanoparticles – that allow for externally triggered targeting or drug release. Although the majority of VDCs rely on endogenous triggers, these advanced strategies open avenues for remote control and precision delivery.
- Immune-mediated Targeting: Certain VDCs are engineered to evoke an immune response by exposing tumor neoantigens upon drug release. The resulting local immune activation can further facilitate the targeting and destruction of tumor cells, thereby enhancing therapeutic efficacy.

Current Research and Examples

The practical application of VDCs spans a wide spectrum of clinical areas, particularly in oncology. Research has evolved from in vitro characterization to advanced clinical investigations that showcase promising therapeutic outcomes.

Case Studies of VDCs
Several notable examples of VDCs have been documented in recent literature and clinical pipelines:
- Belzupacap sarotalocan: Currently in Phase 3 clinical development, this VDC candidate exemplifies the coupling of a potent small molecule drug with VLPs to treat specific neoplastic conditions. Its development demonstrates that VDCs can achieve a favorable therapeutic index through the optimized conjugation of prognostic small molecule drugs.
- MLT-103: As a Phase 1 candidate, MLT-103 represents another instance of a small molecule VDC that is being evaluated for its safety and efficacy. Although in the early stages of clinical testing, the progress of MLT-103 reinforces the promise of small molecule payloads delivered via VLPs in various therapeutic areas including infectious and respiratory diseases.
- AU-011: Developed by Aura Biosciences, AU-011 targets ocular oncology, particularly choroidal melanoma. This VDC candidate is designed to conjugate numerous cytotoxic drugs—indeed, one of its unique advantages is the high drug loading capacity, with up to 400 drug molecules per VLP—thereby offering a highly potent treatment modality with minimized collateral damage to healthy tissues. These cases highlight the clinical viability of VDCs across diverse indications, emphasizing both small molecule and biologic approaches within the VDC framework.

Recent Advances and Innovations
Research in the VDC arena is rapidly advancing on multiple fronts, influenced by improvements in both chemical conjugation strategies and an enhanced mechanistic understanding of virus-like particle biology:
- Optimization of Linker Chemistry: Recent innovations have focused on the development of stimuli-responsive linkers that ensure rapid and selective drug release upon encountering the target intracellular environment. For instance, pH-sensitive linkers and enzyme-cleavable bonds have been engineered to facilitate high-fidelity drug release inside tumor cells, significantly reducing off-target effects.
- Dual Payload Conjugates: An exciting advancement in the field involves the conjugation of dual payloads – combining a small molecule with a biologic or nucleic acid – onto a single VLP. This strategy enables a multipronged attack on tumor cells by simultaneously perturbing multiple pathways; one payload might induce direct cytotoxicity while the other modulates the tumor microenvironment or sensitizes the cell to immune attack.
- Enhanced Targeting via Engineered CAPSID Proteins: Advances in protein engineering have allowed the modification of viral capsid proteins to improve targeting specificity. By genetically inserting binding motifs or chemically modifying surface residues, researchers can tailor VDCs to recognize unique molecular markers on cancer cells, thereby enhancing the delivery precision.
- Combination with Immunotherapy: There is an emergent trend in combining VDCs with immunotherapeutic agents. In such systems, the immediate cytotoxic effects of the drug payload are supplemented by a robust immune response, which is partly triggered by the immunogenic nature of the VLP itself. This synergistic effect has been observed in preclinical studies and holds promise for transforming treatment paradigms in oncology.
- Integration with Nanotechnology Platforms: Recent developments have also seen the integration of VDCs with nanotechnology platforms, such as incorporating magnetic nanoparticles or fluorescence tags for real-time tracking of biodistribution. This convergence of nanotechnology and VDC development not only advances our understanding of in vivo performance but also paves the way for personalized medicine approaches, where drug release and distribution are monitored and adjusted in real time.

Detailed Conclusion

In summary, Virus-like Drug Conjugates (VDCs) represent a transformative drug delivery system that combines the natural cell-interactive properties of virus-like particles with a broad array of therapeutic payloads. The different types of drugs available for VDCs can be broadly categorized into three groups:

1. Small Molecule Drugs:
These are traditionally used chemotherapeutic agents and other low-molecular weight drugs that can be chemically modified for stable conjugation to VLPs. They offer the advantages of well-characterized pharmacokinetics and potential to overcome resistance mechanisms when delivered in a targeted manner. Clinical examples such as Belzupacap sarotalocan and MLT-103 highlight the promise of small molecule VDCs in reducing systemic toxicity while maximizing on-target effects.

2. Biologics:
Biologics include larger macromolecules such as peptides, proteins, and antibody fragments. Due to their inherent specificity and complex structure, they can directly modulate cellular pathways or serve as immune stimulators when delivered via VLP platforms. The challenges in conjugating biologics while preserving their active configuration are being addressed through advanced site-specific conjugation techniques. Such biologics can be exploited either for direct therapeutic action or as complementary agents in combination therapies.

3. Nucleic Acid-based Drugs:
This category encompasses gene-based therapeutics, such as plasmid DNA, mRNA, siRNA, and antisense oligonucleotides, that aim to modulate gene expression. When integrated into VDCs, these nucleic acid drugs benefit from the VLP’s protective envelope, which minimizes degradation and enhances cellular uptake. The precision afforded by nucleic acid therapeutics, along with the potential for gene silencing or expression restoration, makes them highly attractive payloads, despite the delivery challenges that are gradually being overcome.

From a mechanistic standpoint, VDCs operate by combining the drug delivery mechanics of VLPs with sophisticated targeting strategies. The multivalent and stimuli-responsive nature of these systems promotes efficient receptor-mediated uptake, controlled intracellular drug release, and, in some cases, simultaneous engagement of the immune system. Advances in linker chemistry, capsid engineering, and dual payload strategies further optimize these mechanisms, ensuring that drugs are released in a controlled, site-specific manner.

Current research and clinical case studies have provided tangible examples of how VDCs can be tailored for specific applications. Case studies such as AU-011 for ocular oncology show that high payload capacity and precise targeting can be achieved, setting the stage for further clinical innovations. Meanwhile, ongoing improvements in targeting precision, stimulus-responsive drug release, and combination therapies underscore a vibrant and rapidly advancing field that promises to overcome the limitations of conventional drug delivery systems.

In conclusion, the landscape of drugs available for Virus-like Drug Conjugates is diverse and multifaceted. Small molecule drugs continue to lead the way in conventional cytotoxic agent delivery, whereas biologics and nucleic acid-based drugs open new avenues for precision medicine and gene-targeted therapies. By merging these payloads with highly engineered virus-like particles, VDCs offer an unparalleled level of specificity, control, and efficacy in drug delivery. The evolving research, as documented in recent studies and clinical advancements, highlights a future where VDCs not only transform cancer treatment but also expand into other disease areas with similar unmet therapeutic needs. This integrative approach, which encompasses both chemical precision and biological targeting, sets the stage for a new era of drug delivery systems that can be tailored to a wide range of clinical applications, providing hope for improved patient outcomes through enhanced efficacy and reduced side effects.

Discover Eureka LS: AI Agents Built for Biopharma Efficiency

Stop wasting time on biopharma busywork. Meet Eureka LS - your AI agent squad for drug discovery.

▶ See how 50+ research teams saved 300+ hours/month

From reducing screening time to simplifying Markush drafting, our AI Agents are ready to deliver immediate value. Explore Eureka LS today and unlock powerful capabilities that help you innovate with confidence.