What are the different types of drugs available for Antibody toxin conjugate?

17 March 2025
Introduction to Antibody-Toxin Conjugates
Antibody-toxin conjugates (ATCs) represent a unique class of biopharmaceuticals that combine the exquisite specificity of antibodies with the potent cell‐killing ability of toxins. These compounds are designed to recognize and bind to specific cell surface antigens, internalize into the targeted cell, and subsequently release a cytotoxic payload that disrupts vital cellular processes. The ultimate goal is to achieve maximal killing of diseased cells (typically cancer cells) while minimizing systemic toxicity and off‐target effects.

Definition and Basic Principles
At their core, antibody-toxin conjugates consist of two main components:
1. A monoclonal antibody (or its fragment) engineered to recognize a particular antigen expressed on the surface of target cells. The antibody portion ensures high specificity and binding affinity, leading to selective delivery.
2. A toxin payload that is either derived from microbial sources (e.g., bacterial toxins) or engineered from plant toxins, synthetic cytotoxins, or recombinant protein toxins. These toxins are selected for their extreme potency and ability to interfere with essential cellular processes, such as protein synthesis or DNA replication.

The conjugation of the toxin to the antibody is typically facilitated by chemical linkers that play a critical role in ensuring that the toxin remains stably attached during circulation and is efficiently released upon internalization into the target cell. This “targeted” delivery not only improves the therapeutic index but also circumvents many dose-limiting toxicities associated with conventional chemotherapeutic agents.

Historical Development and Current Trends
The development of antibody-toxin conjugates has evolved over decades, starting from early attempts to harness the cell-killing potential of highly potent toxins without compromising antibody specificity. Initially, efforts focused on coupling full-length antibodies to bacterial toxins in a non-specific manner, often resulting in heterogeneous products with unpredictable pharmacokinetics. Over time, innovations in genetic engineering and conjugation chemistry have given rise to next-generation conjugates characterized by site-specific coupling techniques. These refined methods have increased homogeneity and improved both in vivo stability and safety profiles.

A major milestone in this field was the approval of agents like Moxetumomab Pasudotox in 2018 for the treatment of hairy cell leukemia, which demonstrated that immunotoxins can be successfully translated to the clinic. More recently, clinical candidates such as LMB-2, which targets IL2RA and employs a toxin payload, have advanced to phase II studies. Current trends focus on further refining conjugation methods, developing engineered antibody formats (such as single-chain variable fragments and antibody fragments), and exploring novel toxins with improved safety and potency profiles. These developments seek to expand the therapeutic applications beyond oncology, as newer targets and combination modalities emerge.

Classification of Antibody-Toxin Conjugates
Antibody-toxin conjugates can be classified from several perspectives. Two of the most useful classification schemes are based on the type of toxin payload attached and the type of antibody or antibody fragment used in the conjugate.

Based on Toxin Type
When classifying ATCs by toxin type, several categories emerge:

1. Bacterial Toxin-Based Conjugates
Pseudomonas Exotoxin-Based Conjugates: One prominent example is Moxetumomab Pasudotox-TDFK, which combines an antibody with a truncated form of Pseudomonas exotoxin A. This engineered toxin component is designed to inhibit protein synthesis once internalized, triggering cell death.
– Diphtheria Toxin-Based Conjugates: Similar strategies have been applied with diphtheria toxin. Although less common than Pseudomonas exotoxin conjugates, these agents have been explored for their potency and ability to induce rapid cell death by ADP-ribosylation of elongation factor 2.

2. Plant Toxin-Based Conjugates
– Ricin-Derived Conjugates: Ricin A chain is a well-known ribosome-inactivating protein that has been chemically linked to monoclonal antibodies to form immunotoxins. Despite its high potency, the clinical use of such conjugates has been limited by immunogenicity issues, although innovative strategies continue to address these challenges.
– Other Plant Toxins: Other plant-derived toxins, including certain ribosome-inactivating proteins, have been evaluated for inclusion in antibody conjugates. Their mechanism—usually through depurination of ribosomal RNA—leads to robust inhibition of protein synthesis in targeted cells.

3. Recombinant Protein Toxins and Engineered Toxins
– Genetically Engineered Toxins: Advances in recombinant DNA technology have enabled the production of engineered toxins with reduced immunogenicity and improved pharmacokinetic properties. For instance, truncated bacterial toxins can be fused seamlessly to antibody fragments, resulting in more predictable behavior and easier manufacturing.
– Fusion Proteins: In some cases, the toxin is not chemically conjugated but genetically fused to the antibody fragment to produce a single-chain immunotoxin. This strategy has been used in early-phase clinical agents and research prototypes, capitalizing on molecular design to optimize stability and efficacy.

4. Synthetic Cytotoxic Payloads (Chemical Toxins)
Although not “toxins” in the classical sense derived from bacterial or plant sources, many antibody-drug conjugates incorporate highly potent small-molecule cytotoxins such as maytansinoids, auristatins, tubulysins, and camptothecin analogues. These agents act in a manner akin to protein toxins by interfering with tubulin dynamics or DNA integrity, leading to apoptosis. Their inclusion in ADCs has revolutionized cancer treatment by significantly enhancing the therapeutic index when delivered in a targeted fashion.

Based on Antibody Type
The antibody component of a toxin conjugate is equally critical. Classification based on antibody type includes:

1. Full-Length IgG Conjugates
– These ATCs utilize whole monoclonal antibodies (typically IgG1) that have the benefit of long serum half-life, robust effector functions, and established manufacturing processes. However, their large size can sometimes impair tissue penetration.

2. Fragment-Based Conjugates
– scFv (Single-Chain Variable Fragment) Conjugates: By engineering the antigen-binding region into a single-chain format, these conjugates can improve tumor penetration and reduce potential immunogenicity due to their smaller size.
– Fab or F(ab')₂ Conjugates: These fragments maintain a substantial portion of the antigen-binding region while eliminating the Fc portion, which can be beneficial in reducing non-specific interactions and improving biodistribution.

3. Engineered and Bispecific Antibody Conjugates
– The field is increasingly exploring bispecific formats that can simultaneously target two different antigens or engage additional cellular processes (e.g., immune cell recruitment) along with toxin delivery. This approach can enhance specificity and potentiate cell killing even further.

4. Recombinant Fusion Proteins
– Rather than relying on chemical conjugation, some strategies involve the direct genetic fusion of the toxin to the antibody or its fragment. This method offers advantages in terms of product homogeneity and consistency but may require more complex engineering to ensure proper folding and function.

Examples of Antibody-Toxin Conjugate Drugs
A number of antibody-toxin conjugates have reached various stages of clinical development, from early-phase clinical trials to FDA approval. These examples illustrate both the promise and the challenges of the approach.

FDA-Approved Drugs
One of the landmark achievements in this field has been the regulatory approval of certain immunotoxins:

– Moxetumomab Pasudotox-TDFK:
  Moxetumomab Pasudotox is a prime example of a bacterial toxin-based immunotoxin. It is designed to target CD22 on hairy cell leukemia cells and is conjugated to a truncated form of Pseudomonas exotoxin A. Its approval in 2018 marked a significant milestone, demonstrating that a carefully engineered antibody-toxin conjugate can safely deliver a potent toxin to malignant cells with manageable toxicity.

– Other Examples:
  While Moxetumomab Pasudotox is one of the few FDA-approved ATCs, additional drugs have been approved under the broader category of antibody-drug conjugates in oncology—often incorporating synthetic cytotoxins rather than protein toxins. For instance, ADCs such as trastuzumab emtansine (T-DM1) and brentuximab vedotin, although traditionally considered ADCs with small-molecule payloads, share many mechanistic similarities with immunotoxins and continue to inspire development in toxin-based designs.

Drugs in Clinical Trials
Several promising candidates are in various stages of clinical development:

– LMB-2:
  LMB-2 is an immunotoxin that targets IL2RA (interleukin-2 receptor alpha) and employs a toxin moiety that acts as an IL2RA inhibitor. Currently in Phase II clinical trials, LMB-2 exemplifies the potential of engineered immunotoxins for hematologic malignancies, building on the lessons learned from earlier generations of ADCs.

– Other Experimental Agents:
  Many research programs are investigating a diverse range of toxin payloads, including plant-derived toxins, modified bacterial toxins, and novel engineered toxin variants. Preclinical studies and early-phase clinical trials are exploring conjugates that incorporate these payloads alongside novel antibody formats. Some of these agents are designed with specific linker chemistries to improve in vivo stability, release kinetics, and overall therapeutic efficacy.

– Hybrid and Next-Generation Conjugates:
  Beyond the classical immunotoxin concept, hybrid conjugates that combine bacterial toxins with synthetic molecules or dual payloads are under investigation. Such constructs aim to exploit the advantages of both biological and chemical cytotoxic agents. Recent advancements in site-specific conjugation, for instance, using enzymatic strategies or unnatural amino acids, have enabled the production of homogenous and highly potent antibody-toxin conjugates that are now entering clinical evaluation.

Mechanisms of Action
Understanding how antibody-toxin conjugates exert their effect is key to appreciating both their efficacy and the challenges that must be overcome to optimize their therapeutic window.

Cellular Uptake and Internalization
The mechanism of action of ATCs begins with selective binding to a target antigen expressed on the cell surface. Once bound, the antibody engages with the cellular recognition machinery, initiating internalization via receptor-mediated endocytosis. For fully effective conjugates, the following steps are critical:

– Receptor Binding:
  The antibody portion of the conjugate recognizes and binds to a specific antigen (e.g., CD22 or IL2RA) on the target cell. High affinity and specificity of this interaction are essential to minimize binding to non-target cells and ensure effective delivery of the toxin.

– Internalization:
  Upon binding, the conjugate is internalized via endocytosis. The mechanism may vary depending on the antigen and cell type; some may exploit clathrin-mediated endocytosis while others may utilize caveolar or macropinocytic pathways. The rate and efficiency of internalization are pivotal determinants of the conjugate’s therapeutic efficacy.

– Trafficking to Lysosomes:
  Once inside the cell, the conjugate is trafficked to the lysosomal compartment, where the acidic environment and enzymatic activity facilitate the cleavage of the linker. This step is essential for ensuring that the toxin payload is released in a controlled fashion only within the target cell.

Toxin Release and Action
Following internalization, the toxin payload must be liberally released to exert its cytotoxic effect. This process involves several key steps:

– Linker Cleavage:
  Linkers are engineered to be stable in the bloodstream but labile within the intracellular environment. Cleavable linkers typically respond to the acidic pH within lysosomes or to specific proteases that are overexpressed in certain tumor cells.
  Once cleaved, the released toxin becomes active. For example, the truncated Pseudomonas exotoxin A in Moxetumomab Pasudotox undergoes proteolytic processing to generate an active fragment that inactivates elongation factor‑2, halting protein synthesis and leading to cell death.

– Toxin Action:
  Different toxins have different mechanisms of action. Bacterial toxins such as Pseudomonas exotoxin A and diphtheria toxin modify elongation factors by ADP-ribosylation, effectively shutting down protein synthesis. Plant toxins like ricin A chain disable ribosomes by depurinating ribosomal RNA. Engineered toxins may also disrupt other cellular functions such as inducing apoptosis or damaging DNA. Regardless of the specific mechanism, the payload must be potent enough to kill the target cell at very low concentrations given the limited number of molecules delivered per cell.

– Bystander Effects:
  Some constructs are designed so that the released toxin can diffuse to adjacent cells, contributing to a bystander effect. This mechanism can be particularly useful in tumors with heterogenous antigen expression, ensuring that even cells lacking high antigen density can be affected.

Challenges and Future Perspectives
Despite the significant promise of antibody-toxin conjugates, several challenges remain that limit their therapeutic window and broader clinical application. Addressing these challenges is a major focus of current research and innovation in the field.

Current Limitations
Antibody-toxin conjugates face multiple hurdles that researchers are actively working to overcome:

1. Heterogeneity of Conjugates:
  Early-generation conjugates suffered from heterogeneous coupling sites and variable drug-to-antibody ratios (DARs). This heterogeneity complicates production, quality control, and leads to unpredictable pharmacokinetic profiles. Although site-specific conjugation methods and engineered antibody formats have improved product uniformity, ensuring consistency remains a technical challenge.

2. Immunogenicity:
  The toxin payloads, especially those derived from bacterial or plant sources, are highly immunogenic. Repeated administration of these conjugates can trigger neutralizing antibody responses, thereby reducing their efficacy and increasing the risk of hypersensitivity reactions. Efforts to humanize or modify toxin structures while maintaining potency are ongoing.

3. Stability of the Linker:
  The linker connects the antibody to the toxin and must balance stability in circulation with efficient cleavage upon internalization. If the linker is too labile, premature toxin release may occur, leading to systemic toxicity. Conversely, overly stable linkers may hinder intracellular release and reduce efficacy.

4. Limited Tumor Penetration:
  Large, full-length antibody-toxin conjugates may have difficulty penetrating dense tumor masses. The size and steric hindrance can reduce their ability to reach all malignant cells, particularly in solid tumors with a complex microenvironment. Emerging strategies involving smaller antibody fragments or bispecific formats are being explored to mitigate this issue.

5. Resistance Mechanisms:
  Tumor cells may develop resistance to the toxin payload or downregulate antigen expression. Such adaptive mechanisms may limit the long-term efficacy of these agents and necessitate combination therapies or sequential treatment strategies.

Innovations and Future Research Directions
Ongoing research in antibody-toxin conjugates is focused on overcoming current limitations and expanding the utility of these agents:

1. Advanced Conjugation Techniques:
  Innovation in site-specific conjugation methods—such as enzymatic conjugation and the use of unnatural amino acids—has greatly enhanced the homogeneity and stability of ATCs. Continued optimization of these technologies is expected to improve manufacturing consistency and safety profiles while reducing off-target toxicity.

2. Engineered Toxins with Reduced Immunogenicity:
  Efforts are underway to engineer toxin payloads that retain high potency but are less likely to trigger immune responses. Approaches include gene shuffling, epitope masking, and the development of truncated or modified toxin variants that maintain their cytotoxic function. These strategies are essential for allowing repeated dosing and achieving durable clinical responses.

3. Optimization of Linker Chemistry:
  The development of cleavable linkers that respond to specific intracellular conditions (such as pH, protease activity, or redox potential) is a major area of focus. Optimized linkers will help ensure that the toxin is released only within the target cell, thereby reducing systemic adverse effects.
  Future research is looking at novel chemical moieties for linkers that can offer even greater precision in payload release.

4. Novel Antibody Formats and Payload Combinations:
  The exploration of different antibody formats—such as scFv, Fab, and multispecific constructs—offers the promise of improved tumor penetration and selective targeting. Additionally, combining traditional toxin payloads with innovative cytotoxic agents (including dual payloads or payloads that exert two complementary mechanisms of action) is an emerging strategy to overcome tumor heterogeneity and resistance.

5. Combination Therapies and Personalized Medicine:
  Given the challenges associated with monotherapy using ATCs, there is growing interest in combining antibody-toxin conjugates with other therapeutic modalities, such as immune checkpoint inhibitors, targeted small molecules, or even other antibody constructs (e.g., bispecific antibodies). This combinatorial approach not only improves the likelihood of tumor eradication but also may enable lower dosing of toxic payloads, thereby reducing side effects.
  Furthermore, advances in genomics and biomarker development are driving personalized medicine approaches. Tailoring antibody-toxin conjugates to patient-specific tumor profiles will be key to maximizing therapeutic efficacy while minimizing toxicity.

Detailed Conclusion
In summary, antibody-toxin conjugates are a powerful class of therapeutics that synergistically combine the targeting specificity of antibodies with the potent cell-killing ability of toxin payloads. Their development has followed a fascinating trajectory from early, heterogeneous constructs with unpredictable clinical performance toward advanced, site-specifically engineered conjugates with markedly improved safety and efficacy profiles.

When classifying these conjugates by toxin type, we see several categories: bacterial toxin-based conjugates (such as those utilizing truncated Pseudomonas exotoxin as in Moxetumomab Pasudotox and those based on diphtheria toxin), plant toxin-based conjugates (including those derived from ricin A chain), recombinant/engineered protein toxins that have been optimized for reduced immunogenicity and improved pharmacokinetics, and even synthetic cytotoxic payloads that, while not natural toxins, function in a similarly devastating manner against tumor cells. These payloads may be coupled with full-length IgG antibodies, antibody fragments (e.g., scFvs, Fabs), or increasingly sophisticated bispecific or recombinant fusion proteins that offer additional functional advantages.

Examples of antibody-toxin conjugate drugs in the clinical realm include the FDA-approved Moxetumomab Pasudotox—a bacterial toxin-derived immunotoxin used for hairy cell leukemia—and agents such as LMB-2 currently in clinical development for hematologic malignancies. Emerging candidates benefit from advances in linker chemistry (ensuring efficient intracellular toxin release), site-specific conjugation (enhancing product homogeneity), and novel toxin engineering strategies designed to minimize immunogenicity while preserving potent cytotoxicity.

Mechanistically, these conjugates function by binding to target antigens on the cancer cell surface, undergoing receptor-mediated internalization, and then releasing the toxin payload within the lysosomal compartment of the cell. The released toxin then disrupts vital intracellular processes—often by inhibiting protein synthesis or damaging essential cellular structures—leading to cell death. This intricate mechanism is under continuous refinement with the aim of overcoming challenges such as limited tumor penetration, heterogeneous antigen expression, immune responses, and resistance mechanisms.

Despite significant advances, challenges remain. Heterogeneity in conjugation, potential immunogenicity of toxin payloads, the delicate balance required in linker design, and issues related to tumor microenvironment penetration are all areas of active research. Innovations aimed at improving conjugate uniformity, reducing immunogenicity via toxin engineering, optimizing linker stability, and devising combination treatment strategies have the potential to further broaden the therapeutic window of these agents.

Future directions in antibody-toxin conjugate development are poised to benefit from personalized medicine approaches, where patient-specific biomarkers guide treatment decisions, and from combination therapies that harness synergistic effects between targeted toxins and other therapeutic modalities. As our understanding of both tumor biology and conjugate pharmacology continues to evolve, it is anticipated that next-generation ATCs will overcome current limitations, leading to more effective and safer therapies for cancer patients and beyond.

In conclusion, the landscape of drugs available for antibody-toxin conjugates is diverse and multifaceted. It includes bacterial toxin-based conjugates, plant toxin-based conjugates, recombinant engineered toxins, and synthetic cytotoxic agents—all coupled to various antibody formats. While FDA-approved drugs such as Moxetumomab Pasudotox have paved the way, numerous candidates in clinical trials demonstrate the vibrant innovation in the field. The progress in precise conjugation techniques, toxin engineering, and novel antibody formats is set to revolutionize targeted therapies in oncology, promising higher efficacy with managed toxicity. The future of antibody-toxin conjugates relies on overcoming existing challenges through innovative research and a personalized approach to cancer therapy, offering a beacon of hope for improved 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.