What is Antibody-based PROTAC (AbTAC)? - A Beginner’s Guide

* Please be kindly noted that our services and products can only be used for research to organizations or companies and not intended for any clinical or individuals.

Introduction to Targeted Protein Degradation

What is Protein Degradation in Drug Discovery?

Targeted protein degradation has emerged as a cutting-edge strategy in the field of drug discovery. Unlike traditional drug development approaches that focus primarily on modulating protein function, protein degradation offers a novel mechanism by directly eliminating disease-causing proteins. This approach addresses pathological protein overexpression or dysfunction at the source, offering a transformative way to treat diseases.

By leveraging the cell's own degradation pathways to precisely recognize and remove target proteins, protein degradation enables the development of therapies against previously "undruggable" targets. This innovative strategy opens new avenues for drug discovery, enhances the precision and success rate of drug development, and holds great promise for treating complex conditions such as cancer and neurodegenerative diseases.

Traditional PROTACs vs. Emerging Protein Degradation Technologies

PROTACs (Proteolysis Targeting Chimeras) are a pioneering technology in the protein degradation field. These bifunctional molecules simultaneously recruit a target protein and an E3 ubiquitin ligase, leading to ubiquitination and subsequent proteasomal degradation of the target. Despite their groundbreaking potential, traditional PROTACs face limitations, including large molecular weight that hampers cell permeability and reduced degradation efficiency for certain targets.

Recent innovations are overcoming these challenges. Molecular glue degraders, for instance, promote conformational changes between E3 ligases and target proteins to induce degradation. These small molecules are easier to optimize for drug-like properties. Additionally, lysosome-based degradation strategies are expanding the toolkit beyond the proteasome pathway, enabling a broader range of targets.

Together, these new methods enhance the versatility and effectiveness of targeted protein degradation technologies. By addressing the shortcomings of classical PROTACs, they offer multiple therapeutic strategies to tackle difficult disease targets and accelerate the development of next-generation precision medicines.

What is AbTAC Technology?

Definition and Mechanism

AbTAC (Antibody-based PROTAC) is an emerging immunotherapy platform designed to enhance T cell-mediated tumor killing. This innovative technology works by linking a tumor-targeting antibody with a T cell-engaging domain, thereby precisely redirecting T cells toward tumor cells expressing specific antigens.

Once an AbTAC molecule binds to a tumor-specific antigen on the surface of cancer cells, it simultaneously engages a receptor on the T cell surface-typically CD3-bringing the T cell into close proximity with the tumor cell. This spatial connection activates the T cell's cytotoxic functions, leading to the release of immune effector molecules such as perforin and granzyme. As a result, the targeted tumor cells undergo apoptosis, achieving therapeutic effects in various cancers and potentially other immune-related diseases.

Key Components of AbTACs

  • Tumor-Specific Antibody Domain

The antibody component serves as the core recognition module of the AbTAC molecule. Typically composed of a high-affinity monoclonal antibody, it is designed to specifically recognize tumor-associated antigens such as carcinoembryonic antigen (CEA), CD19, or others. This specificity ensures that the AbTAC molecule accurately locates and binds to tumor cells, minimizing off-target effects.

  • T Cell Engaging Signal Domain

This portion interacts with specific receptors on T cells, most commonly via an anti-CD3ε antibody or other signaling domains capable of triggering T cell activation. It functions as a biological 'bridge,' connecting the T cell to the tumor cell and initiating the T cell's cytotoxic response.

The linker connects the antibody domain with the T cell engaging signal domain. Its length and flexibility are critical to the functional performance of the AbTAC molecule. An optimized linker ensures proper spatial orientation and conformational flexibility, allowing effective simultaneous binding to both the tumor antigen and the T cell receptor.

Differences Between PROTACs and AbTACs

Comparison ItemPROTACs (Proteolysis-Targeting Chimeras)AbTACs (Antibody-based PROTAC)
Mechanism of Action Induce degradation of intracellular target proteins by hijacking the ubiquitin-proteasome system. PROTACs link a target protein to an E3 ligase, leading to ubiquitination and subsequent proteasomal degradation.Activate T cells to eliminate tumor cells by recruiting them to the tumor microenvironment and stimulating their cytotoxic response.
Target Scope Capable of targeting almost any intracellular protein, including "undruggable" proteins that small molecules cannot modulate effectively.Primarily targets specific antigens expressed on the surface of tumor cells, relying on antibody-based recognition.
Design Complexity Requires careful design of both the target-binding ligand, E3 ligase ligand, and a suitable linker, all while ensuring cell permeability.Focuses on optimizing tumor-specific antibody domains and T cell-engaging sequences, balancing affinity, specificity, activation strength, and safety.
Site of Action Intracellular - mainly in the cytoplasm or nucleus, where the target protein is degraded by the proteasome.Extracellular -acts on the cell surface by facilitating immune synapse formation between T cells and tumor cells.
Onset of Action Protein degradation requires time; effects typically occur over several hours or longer, depending on degradation kinetics.T cell activation and cytotoxicity can occur rapidly upon cell engagement, though therapeutic efficacy may vary depending on tumor and immune context.
Disease Applicability Used for diseases driven by abnormal protein expression, such as certain cancers and neurodegenerative disorders-especially where targets are inaccessible to traditional drugs.Best suited for cancer treatment, especially tumors with well-defined surface antigens that can be recognized by monoclonal antibodies.

Why AbTACs Matter in Therapeutics?

Targeting "Undruggable" Proteins

In traditional drug discovery, a large number of disease-relevant proteins have been deemed "undruggable" due to the absence of accessible binding pockets or the inability of conventional drugs to effectively engage them. This presents a significant challenge in developing targeted therapies.

AbTAC (Antibody-based PROTAC) technology offers a breakthrough solution through its unique mechanism of action. By leveraging the high specificity of antibodies, AbTACs can precisely recognize and bind to these elusive targets-even those previously considered undruggable. Once bound, the T cell-engaging sequence within the AbTAC molecule recruits and activates cytotoxic T cells at the tumor site, redirecting the immune system to destroy the tumor cells harboring the target protein.

This innovative approach not only overcomes long-standing limitations in drug development but also expands the therapeutic landscape. AbTACs open new possibilities for treating cancers and diseases previously out of reach, offering renewed hope for patients facing limited or no treatment options.

AbTACs vs. Small Molecule PROTACs

Comparison CriteriaAbTACs (Antibody-based PROTAC)Small Molecule PROTACs (Proteolysis Targeting Chimeras)
Target Expandability Broad targetability including "undruggable" intracellular proteins and cell surface antigens. Antibodies can recognize subtle conformational epitopes within complex 3D structures, enabling access to a wide range of therapeutic targets.Limited by the specificity and affinity of small molecule ligands. Primarily effective against intracellular proteins with well-defined binding pockets.
Mechanism of Action Engages and redirects T cells to tumor cells, triggering direct immune-mediated killing. May establish immune memory, offering long-term surveillance and reducing tumor recurrence.Relies on the intracellular ubiquitin-proteasome system to degrade target proteins. Acts within cells without generating immune memory; therapeutic effects may wane after treatment stops.
Drug Properties As biologic macromolecules, AbTACs have relatively complex in vivo metabolism but benefit from extended half-life, reduced dosing frequency, and manageable immunogenicity.As small chemical entities, PROTACs exhibit good tissue penetration but often require frequent dosing due to short half-lives and rapid metabolism and clearance.

Summary & Future Outlook

Opportunities in Oncology and Beyond

AbTACs (Antibody-based PROTAC) offer unique opportunities in oncology by precisely targeting tumor-associated surface antigens and activating T cell-mediated cytotoxicity. This immune redirection strategy provides a novel therapeutic approach for effectively eliminating cancer cells, especially in tumors expressing well-defined surface markers.

Beyond oncology, AbTACs hold promise in the treatment of other immune-related conditions. In autoimmune diseases, AbTACs may help modulate immune cell function and restore immune homeostasis. In organ transplantation, they can be employed to regulate T cell activity, reducing the risk of graft rejection and improving transplant outcomes.

Challenges in Development

Despite their potential, the development of AbTACs faces several technical and clinical hurdles. The molecular design of AbTACs is complex, requiring careful optimization of antibody specificity, binding affinity, and linker architecture to improve stability and functional activity. Moreover, the manufacturing process is intricate and costly, necessitating advances in bioproduction technologies to ensure scalability and cost-efficiency.

From a safety perspective, immune-related adverse effects such as cytokine release syndrome and organ-specific inflammation pose significant concerns. Deeper insights into immune mechanisms are essential to enhance safety profiles. Additionally, AbTACs face stringent regulatory oversight during clinical translation, requiring robust multidisciplinary collaboration for successful development and approval.

Related Services

  • Custom AbTAC design and feasibility assessment
  • Antibody development and humanization services
  • Antibody-drug conjugation (ADC) platform adaptation for AbTAC
  • Linker synthesis for lysosome-targeting payloads
  • Target protein expression and purification for conjugation
  • Cell-based assays for AbTAC validation (internalization, degradation)
Request a Consultation
BOC Sciences Support

Please contact us with any specific requirements and we will get back to you as soon as possible.


  • Verification code

We invite you to contact us at or through our contact form above for more information about our services and products.

USA
  • International:
  • US & Canada (Toll free):
  • Email:
  • Fax:
UK
  • Email:
Inquiry Basket