What are the different types of drugs available for Protein drug conjugate?

17 March 2025
Introduction to Protein Drug Conjugates (PDCs)

Definition and Basic Concepts
Protein drug conjugates (PDCs) are advanced therapeutic constructs where a bioactive protein is chemically linked to a drug molecule via a well‐designed linker. In these systems, the protein component usually serves to target specific tissues or cells and to improve the pharmacokinetic profile of the drug, whereas the drug moiety—often highly potent—exerts the desired pharmacologic effect once delivered at the target site. This approach stems from approaches used in antibody–drug conjugates (ADCs) but extends the concept to a broader variety of targeting proteins including engineered protein scaffolds, peptides, and other protein-based vectors. The conjugation chemistry is often optimized to occur in a site-specific manner, thereby producing homogeneous products that avoid pitfalls such as reduced bioactivity or unpredictable pharmacological properties, which are sometimes encountered with random conjugation methods.

Advantages and Applications in Medicine
The advantages of PDCs are considerable. First, by combining the targeting capability of a protein with the potent pharmacological effect of a conjugated drug, these systems can significantly enhance therapeutic indices while reducing off-target toxicity. The protein component can improve the solubility, circulatory half-life, and tissue specificity of the drug by acting as both a carrier and a targeting agent. Often, such conjugates display targeted drug delivery, thereby sparing healthy tissues and reducing systemic side effects. For example, the BC2-IR700 conjugate (a protein drug conjugate currently in preclinical development) uses a protein moiety derived from the University of Tsukuba to target proteoglycans in neoplasms and digestive system disorders, with the IR700 component acting as a photosensitizer that is activated upon irradiation, harnessing the specificity of protein binding and the cytotoxic effect of photodynamic activation. Applications of PDCs cover cancer therapies, endocrine and metabolic diseases, and even inflammatory disorders. As research in proteomics, recombinant DNA technology, and selective conjugation chemistry evolves, the use of PDCs is poised to expand into diagnostics and theranostic applications, integrating both imaging and therapeutic functionalities into a single molecule.

Types of Drugs Used in Protein Drug Conjugates

The types of drugs available for conjugation to proteins vary broadly and can be classified from several perspectives. In the context of PDCs, the drugs are usually categorized based on their molecular size, mechanism of therapeutic activity, and the nature of their interaction with the protein carrier. Below is a detailed exploration of different drug types available for PDCs.

Small Molecule Drugs
Small molecule drugs are by far the most commonly used payloads in protein drug conjugate systems. They typically have low molecular weights, high potency, and can exert their effects by interfering with essential cellular processes such as microtubule dynamics, DNA synthesis, or enzymatic activity. In many PDCs, small molecule drugs are cytotoxic agents derived from natural sources or synthetically optimized for their potency.

1. Cytotoxic Agents (Toxins):
- Auristatins and Maytansine Derivatives: A classic example is the AFP-maytansine conjugate, which is still in preclinical development. In this type of construct, the potent cytotoxic activity of maytansine (a highly toxic antimitotic agent) is harnessed and delivered specifically to tumor cells via a targeting protein. These drugs are designed to inhibit microtubule assembly, thereby halting cell division.
- Auristatin-Based Conjugates: Several patents describe conjugates that incorporate auristatins using specific linker technologies that overcome limitations seen with conventional ADCs. These small molecule toxins are particularly beneficial because they can deliver potent cytotoxic effects even in cases where the parent drug would be too toxic for systemic use.

2. Photodynamic Agents:
- IR700-Based Drugs: BC2-IR700 is an excellent example where a protein is conjugated to a photosensitizer (IR700) that, upon irradiation, generates cytotoxic reactive oxygen species. Photodynamic therapy (PDT) relies on the light-triggered activation of these drugs to achieve spatially controlled toxicity, making them ideal for treating localized tumors with minimal systemic side effects.

3. Enzyme Inhibitors and Other Modulators:
- Some small molecules act as modulators by inhibiting specific enzymes or receptors. For instance, TSDC-01 is designed as a protein drug conjugate that targets multiple receptors (EGFR, HER2, MSLN) using antagonistic and inhibitory mechanisms. Here, the small molecule drug component may work by blocking signal transduction pathways crucial for cancer cell survival.
- Moreover, small molecule drugs can be designed to act as molecular glues, stabilizing protein–protein interactions in a cooperative manner, as discussed in several recent reviews. These molecules, although not strictly cytotoxic in all cases, modulate protein function and potentially can be conjugated to protein carriers for precise therapeutic interventions.

4. Other Small Molecule Approaches:
- Dual-Action and Bifunctional Molecules: Some PDCs are now being designed to target multiple pathways simultaneously. In these cases, bifunctional molecules combine two distinct mechanisms such as kinase inhibition and receptor antagonism into a single small molecule that is linked to a protein. This dual action approach is particularly promising in overcoming drug resistance and enhancing efficacy.

Biologics
Biologic drugs include molecules that are typically produced using recombinant DNA technology. In the context of PDCs, biologics usually refer to peptides, proteins, and even antibodies that are used either as the targeting moiety or as payloads in their own right.

1. Peptide Drugs:
- Therapeutic Peptides: Due to their smaller size, peptides are easier to synthesize and potentially offer better tissue penetration compared to larger proteins. In some drug conjugate strategies, peptides are incorporated to target cell surface receptors or interfere with protein–protein interactions. For example, peptide-drug conjugates often combine a targeting peptide with a cytotoxic drug to enhance cell-specific delivery.
- Peptide Modulators: These peptides can also be engineered to modulate receptor activities by assisting the internalization of the conjugate; once internalized, the cytotoxic payload is released, thereby increasing the efficacy of the therapeutic molecule.

2. Protein Drugs (Enzymes, Growth Factors, and Antibodies):
- Therapeutic Proteins: In some cases, the conjugate itself is a fusion of two protein units. One common strategy is to attach a cytotoxic drug to an antibody—a strategy widely used in antibody–drug conjugates (ADCs). Even though these are traditionally considered a subset of PDCs, the underlying principle is similar: the protein is used to target cancer cells and deliver the payload precisely, thereby increasing efficacy while reducing systemic toxicity.
- Antibody Fragments and Mimetics: Engineered proteins such as single-chain variable fragments (scFvs) and nanobodies are also used in drug conjugates. These smaller antibody formats maintain the antigen-binding specificity while potentially offering advantages in terms of tissue penetration and clearance rates when conjugated with drugs.
- Enzyme-Based Payloads: Although less common, biologics can also serve as payloads. For instance, enzymes that can trigger prodrug activation or therapeutically modulate a disease pathway are being integrated into conjugate designs. Conjugating these enzymes to targeting proteins can help localize their activity to desired sites, enhancing their therapeutic potential.

3. Combination of Biologic and Small Molecule Features:
- Multifunctional Conjugates: Recent trends involve assembling conjugates that incorporate both biologics and small molecule drugs. This “dual modality” enables the simultaneous delivery of a protein-based targeting moiety along with a small molecule drug. For example, certain antibody or peptide conjugates are engineered to deliver both a therapeutic protein and a small molecule cytotoxin, thereby acting on multiple targets or pathways simultaneously.

Other Novel Drug Types
Beyond the traditional small molecules and biologics, the scope of drug payloads used in protein drug conjugation is expanding thanks to innovative conjugation technologies and emerging therapeutic modalities.

1. Nucleic Acid-Based Agents:
- siRNA and Oligonucleotides: Although traditionally challenging due to issues of stability and delivery, conjugation of nucleic acid drugs such as siRNA to targeting proteins is emerging as a promising strategy. By protecting the nucleic acid with a protein carrier or by attaching a targeting moiety, these conjugates can improve the localized gene silencing effects, thereby treating diseases at the genetic level.

2. Polymeric and Hybrid Conjugates:
- Polymer-Drug Conjugates: In some PDCs, the drug payload is part of a larger polymeric carrier that may be conjugated to proteins. The polymeric component helps improve solubility and stability and can modulate the release kinetics of the drug, ensuring that the therapeutic moiety is released in a controlled manner at the target site. Examples include PEGylated drugs or polymer-drug constructs that impart desirable pharmacokinetic properties.
- Hybrid Molecules: Another novel approach involves the creation of drug hybrids where two or more drugs are chemically linked together via a protein carrier. This strategy is particularly useful in addressing multidrug resistance and in diseases such as cancer, where the simultaneous inhibition of multiple pathways can result in synergistic therapeutic outcomes.

3. Theranostic Agents:
- Imaging and Therapeutic Conjugates: Novel PDCs that integrate diagnostic as well as therapeutic functions are emerging. These conjugates are designed to provide not only a therapeutic effect but also allow for imaging of the target tissue. For example, conjugates containing imaging probes (e.g., fluorophores, radionuclides) alongside therapeutic drugs allow for real-time monitoring of drug targeting and distribution in the body.
- Radioimmunoconjugates: A specific subset of theranostic agents, radioimmunoconjugates, combine radioactive isotopes with antibodies or other protein carriers. The radioactive payload can serve dual roles: delivering a cytotoxic effect and enabling imaging via positron-emission tomography (PET) or single photon-emission computed tomography (SPECT).

Mechanisms of Action

How PDCs Work
The general mechanism of action for protein drug conjugates involves a three-component system: the targeting protein, the linker, and the drug payload. This structured design ensures that each component plays a distinct role in improving the therapeutic outcome.

1. Target Recognition and Binding:
The protein part, often an antibody, peptide, or engineered protein mimic, is designed to recognize a specific antigen or receptor on the surface of the targeted cells. Once injected, the conjugate circulates until it encounters its target. The high specificity is conferred by the natural binding affinity of the protein or peptide, ensuring that the majority of the conjugate accumulates in the diseased tissue—for example, tumor cells expressing specific cell surface markers.

2. Internalization and Activation:
After binding to the target cell’s surface receptor, the conjugate is internalized, typically through receptor-mediated endocytosis. Different strategies exist for release of the drug payload from the conjugate:
- Enzymatic Cleavage: Linkers sensitive to intracellular proteases or esterases are designed to be cleaved within the endosome or lysosome, releasing the active drug.
- pH-Sensitive Linkers: Some linkers are engineered to be cleaved under the acidic conditions present in intracellular compartments such as lysosomes. This pH-triggered release is common in many ADCs and other PDCs.
- Photoactivation: In photodynamic therapeutic conjugates like BC2-IR700, drug activation occurs upon exposure to a specific wavelength of light. This approach allows for spatial control over the activation of the drug at the tumor site.

3. Pharmacologic Activity:
Once released, the small molecule or biologic drug exerts its pharmacological effect on intracellular targets. This can involve inhibiting key enzymes, interfering with microtubule dynamics (in the case of cytotoxic agents), or modulating signaling pathways. The specificity of the initial binding ensures that the drug has minimal off-target effects, thereby enhancing its therapeutic index.

Targeting Strategies
Different targeting strategies are employed in the design of PDCs to maximize the efficacy and specificity of the drug conjugate:

1. Receptor-Mediated Targeting:
The most common targeting strategy is the use of receptor recognition. In many cancers, overexpressed receptors provide an ideal target. By conjugating a drug to a molecule that has a high affinity for such receptors, the payload can be delivered preferentially to the cancer cells. Examples include targeting HER2 in breast cancer or EGFR in various solid tumors.

2. Tissue-Specific Ligands:
Beyond antibodies, small peptides or aptamers can serve as targeting components. These ligands are chosen based on the specific expression patterns of receptors in diseased tissues. Their smaller size compared to antibodies may result in better tissue penetration and faster clearance from non-target tissues.

3. Dual-Targeting Approaches:
Some innovative PDCs are being designed to accommodate dual-targeting strategies, in which one molecule binds two different targets. This approach can be particularly useful in complex diseases such as cancer, where multiple signaling pathways may need to be inhibited simultaneously. Conjugation strategies that allow for the incorporation of two different drug modalities within one molecule are being explored to overcome drug resistance and to enhance synergistic therapeutic effects.

4. Enzyme-Responsive and Environment-Sensitive Systems:
Specific linkers that are sensitive to the biochemical environment (for example, high glutathione concentrations or specific enzymatic profiles) in diseased tissue are being developed. These linkers ensure that drug release only occurs under conditions specific to the target area, further increasing the specificity of the conjugate.

Current Challenges and Future Trends

Challenges in Development and Approval
Despite the promising advantages of PDCs, several challenges exist in their development and regulatory approval.

1. Heterogeneity and Conjugation Efficiency:
One of the key challenges is achieving site-specific conjugation. Non-selective chemical modification can produce heterogeneous conjugate populations, leading to variability in efficacy and safety. Advanced bioorthogonal and enzymatic conjugation techniques have been developed to address these issues; however, ensuring consistency during large-scale manufacturing remains a critical challenge.

2. Stability and Pharmacokinetic Issues:
Stability of the conjugate both during manufacturing and in vivo is of paramount importance. Proteins, particularly when chemically modified, may undergo denaturation or rapid clearance by renal filtration. Additionally, the linker chemistry must be robust enough to prevent premature drug release in circulation, yet sensitive enough to release the drug once internalized by the target cell.
Immunogenicity is another concern; the introduction of non-native chemical groups or modifications can trigger immune reactions that may diminish the therapeutic’s efficacy or cause adverse side effects.

3. Regulatory and Manufacturing Challenges:
The complex nature of PDCs means that the regulatory approval pathway is more demanding than for drugs or proteins alone. Regulatory agencies require extensive data on the manufacturing process, reproducibility, and long-term safety of these novel constructs. Integrated approaches that combine multiple regulatory guidelines—spanning quality control, stability testing, and clinical efficacy—are essential to facilitate approval.

4. Cost and Scalability:
The production of PDCs often necessitates high-cost manufacturing techniques, advanced purification processes, and stringent quality control. Scaling these processes from the laboratory to commercial production while maintaining product homogeneity is a significant challenge.

Innovations and Future Prospects
Despite the challenges, ongoing innovations and technological advances promise a bright future for PDCs.

1. Advanced Conjugation Techniques:
Emerging strategies, such as the incorporation of noncanonical amino acids via genetic code expansion, offer unprecedented control over the site of conjugation. These techniques have enabled the creation of homogeneous PDCs with defined drug-to-protein ratios that have improved therapeutic indices.
Enzymatic conjugation methods, including those utilizing sortase and transglutaminase, are leading the charge toward more controlled and scalable PDC production. These methods allow for efficient, site-specific labeling under mild conditions, preserving protein structure and function.

2. Multifunctional and Theranostic Conjugates:
Future PDCs are expected to integrate diagnostic and therapeutic functions into a single molecule. The development of theranostic agents—conjugates that combine imaging probes with therapeutic drugs—will facilitate real-time monitoring of bio-distribution and target engagement. This dual capability could greatly enhance personalized medicine, allowing clinicians to tailor treatments based on direct feedback from the target site.

3. Multi-Payload Conjugates:
There is growing interest in the development of PDCs that carry more than one payload. Such conjugates may involve the simultaneous delivery of a small molecule drug and a biologic, or even two different small molecules designed to act synergistically. This approach can potentially overcome drug resistance mechanisms by simultaneously targeting multiple signaling pathways.

4. Responsive and Smart Linkers:
Innovations in linker chemistry continue to evolve. New smart linkers that respond to multiple stimuli—such as pH, specific enzyme activity, or redox conditions—are in development. These linkers can greatly enhance the control over drug release, ensuring that the payload is released only under the desired conditions at the target site.
Additionally, research into reversible and cleavable linkers is progressing, which might ultimately allow for tunable pharmacokinetic profiles and minimize systemic toxicity.

5. Integration with Systems Biology and Computational Tools:
Systems biology approaches are increasingly being used to model the interactions of PDCs with cellular networks. In silico models can predict protein–drug interactions and the systemic effects of conjugates, guiding the design of more efficient drug conjugates with fewer off-target effects. Computational tools also enable virtual screening of conjugate candidates, expediting the drug discovery process by predicting pharmacokinetic and pharmacodynamic properties early in the development pipeline.

6. Tailored Approaches for Patient-Specific Therapies:
Precision medicine is poised to benefit significantly from advancements in PDC technology. By linking drugs to targeting proteins that are specific to a patient’s tumor profile or disease phenotype, it becomes possible to devise highly individualized treatment regimens. The integration of biomarkers and genomics data in the design of PDCs will further refine patient selection, maximizing efficacy while reducing adverse effects.

Conclusion

In summary, protein drug conjugates represent one of the most promising arenas in modern drug discovery and targeted therapeutic delivery. The concept leverages the selectivity and favorable pharmacokinetic properties of proteins to deliver potent drug payloads specifically to diseased tissues. The different types of drugs available for incorporation into PDCs can broadly be categorized into small molecule drugs, biologics, and other novel drug types such as nucleic acids and polymer-drug hybrids.

Small molecule drugs, including cytotoxic agents like auristatins, maytansine derivatives, and photodynamic agents such as IR700, have been extensively used in PDCs to provide potent, targeted cell killing. Biologic drugs include a range of therapeutic peptides, engineered proteins, and antibody fragments that serve either as targeting moieties or as payloads themselves, offering a high degree of specificity and versatility. In addition, novel payloads—including siRNA, oligonucleotides, and polymer-based drugs—are emerging as innovative approaches that expand the range of therapeutic applications for PDCs.

Mechanistically, these conjugates work by combining a targeting protein with a drug payload via a carefully engineered linker that responds to specific intracellular triggers such as pH changes, enzymatic activity, or even photoactivation. This targeted release mechanism not only ensures maximum drug efficacy at the pathological site but also minimizes systemic side effects and off-target toxicity. Multiple targeting strategies are applied, ranging from receptor-mediated binding and internalization to dual-targeting systems that address complex disease mechanisms, such as cancer.

Despite the significant promise, the development of PDCs faces several challenges—most notably issues of heterogeneity, stability, immunogenicity, and scaling up the manufacturing processes. However, innovations in site-specific conjugation chemistry, smart linker designs, and the integration of computational predictive models are paving the way for overcoming these obstacles. There is also an increasing trend toward the development of multifunctional and theranostic conjugates that not only treat but also diagnose disease conditions in real time.

Looking ahead, the future of PDCs appears bright with ongoing research focused on refining conjugation methodologies, enhancing payload diversity, and personalizing therapeutic approaches. As systems biology and advanced computational chemistry merge with traditional biochemical methods, the next generation of PDCs will likely offer improved therapeutic indices, fewer side effects, and ultimately a higher success rate in clinical applications. The continued collaboration among academic researchers, biotechnology companies, and regulatory bodies will be essential in translating these innovations into clinically approved therapies.

In conclusion, understanding the different types of drugs available for protein drug conjugates—from small molecule toxins and photodynamic agents to biologics and emerging nucleic acid-based therapies—provides a comprehensive overview of the current state and future potential of this therapeutic modality. The detailed insights from multiple perspectives, including molecular design, targeting strategies, challenges in development, and innovative future prospects, confirm that PDCs are a multifaceted and evolving field. These insights, as evidenced by numerous high-quality studies and patent literature, underscore the importance of an integrated approach to developing next-generation therapeutics with improved efficacy, safety, and patient outcomes.

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.