Colorful molecular structure representing PROTACs in drug discovery.

Next-generation small molecules redefine targeted therapy by degrading, not inhibiting, disease proteins.

ImageFX (2025)

What are PROTACs? Mechanisms, advantages, and challenges

Discover how PROTACs redefine drug discovery by utilizing protein degradation to target previously 'undruggable' proteins.
| 4 min read
Register for free to listen to this article
Listen with Speechify
0:00
4:00

Introduction: Redefining drug discovery through targeted protein degradation

The concept of drugging the “undruggable” has long challenged pharmaceutical research. Conventional small-molecule inhibitors, while effective for many targets, depend on binding to active sites—leaving a large portion of the human proteome inaccessible.

Enter PROTACs, or proteolysis targeting chimeras—a new class of small-molecule therapeutics that harness the cell’s degradation machinery to selectively eliminate disease-causing proteins. Rather than inhibiting function, PROTACs remove the target protein entirely, representing a fundamental shift in targeted therapy design.


What are PROTACs?

PROTACs are heterobifunctional small molecules composed of three key parts:

  1. A ligand that binds the target protein.

  2. A ligand that recruits an E3 ubiquitin ligase.

  3. A chemical linker connecting the two.

Once inside the cell, a PROTAC acts as a molecular bridge between the target and an E3 ligase, inducing ubiquitination of the target and its subsequent degradation by the proteasome.

Because PROTACs catalytically trigger protein removal rather than continuously inhibit activity, a single molecule can degrade multiple targets—demonstrating an event-driven pharmacology model distinct from the occupancy-driven mechanisms of classical drugs.


Mechanism of action: Harnessing the ubiquitin-proteasome system

The ubiquitin-proteasome system (UPS) maintains protein homeostasis through a cascade involving E1 activating enzymes, E2 conjugating enzymes, and E3 ligases that attach ubiquitin to target proteins. The ubiquitinated proteins are then degraded by the 26S proteasome.

Continue reading below...
An illustration showing red cancer cells surrounded by white immune cells interacting on a dark textured background.
ExplainersWhy does immunotherapy work better for some cancers than others?
A powerful tool in modern oncology, immunotherapy doesn’t work the same for everyone. Researchers are exploring why and developing ways to improve its effectiveness.
Read More

PROTACs hijack this pathway in several coordinated steps:

  1. Target binding: The PROTAC ligand binds the protein of interest.

  2. E3 ligase recruitment: The other ligand binds an E3 ligase—commonly VHL (von Hippel–Lindau) or cereblon (CRBN).

  3. Ternary complex formation: The PROTAC promotes a transient complex between target and ligase.

  4. Ubiquitination: The E3 ligase catalyzes the transfer of ubiquitin to lysine residues on the target protein.

  5. Proteasomal degradation: The tagged protein is degraded by the proteasome, while the PROTAC is recycled to initiate another degradation cycle.

This catalytic cycle allows PROTACs to act at substoichiometric concentrations, achieving potent, durable effects.

(Sakamoto et al., PNAS 2001; Wurz et al., Nat Commun 2023)


Advantages of PROTACs over traditional small molecules

1. Expanding the druggable proteome

Because PROTACs rely on proximity rather than enzymatic inhibition, they can target proteins lacking active sites, such as transcription factors or scaffolding proteins—dramatically expanding the therapeutic landscape.

2. Catalytic efficiency

After initiating degradation, the PROTAC is released and reused, enabling potent effects at lower doses and potentially reducing off-target toxicity.

3. Overcoming resistance mechanisms

Drug resistance often emerges through mutations that reduce inhibitor binding or through protein overexpression. By eliminating the protein itself, PROTACs can bypass many of these pathways.
For example, BTK degraders such as NX-2127 degrade both wild-type and C481-mutant BTK, showing clinical activity in B-cell malignancies. However, new resistance mutations (e.g., BTK A428D) have also been observed, emphasizing the need for ongoing resistance monitoring.

4. Sustained pharmacological effects

Once a target is degraded, its resynthesis is required to restore function, providing prolonged target suppression even after the PROTAC has cleared.

Continue reading below...
A digital illustration showing a T cell attacking a cancer cell, symbolizing the promise of immune-based therapies in tackling disease.
Ebooks Advancing cell therapies with smarter strategies
Researchers are finding creative ways to make cell therapies safer and more effective.
Read More

Applications and emerging targets

Since the first demonstration of PROTACs in 2001, hundreds of preclinical programs have emerged, and several have entered human trials.

Oncology

Cancer remains the most active field for PROTAC development.

Neurological diseases

PROTACs targeting tau and α-synuclein are in preclinical development for Alzheimer’s and Parkinson’s disease, offering potential to clear misfolded or toxic proteins. Blood-brain barrier penetration remains a major challenge.

Inflammation and immune disorders

Programs are exploring degraders for NF-κB, STAT, and other transcription factors to modulate immune signaling in autoimmune diseases.


Design considerations and optimization

Designing PROTACs requires precise tuning of molecular properties:

  • E3 ligase selection: Although ~600 E3 ligases exist, only a few—VHL, CRBN, MDM2, and IAP—have been chemically exploited. Broadening this “E3 toolbox” will enable tissue-specific and condition-specific degradation.
  • Linker length and composition: The linker determines ternary complex geometry and cooperativity. Too short restricts alignment; too long can reduce selectivity or solubility.
  • Ternary complex cooperativity: Positive cooperativity between the target and E3 ligase increases degradation potency.
  • Pharmacokinetics: With molecular weights often >800 Da, optimizing permeability, solubility, and oral bioavailability remains critical.

(Bondeson et al., Nat Chem Biol 2015; Wurz et al., Nat Commun 2023)


Key challenges and limitations

1. Size and physicochemical properties

Many PROTACs violate Lipinski’s “Rule of Five.” Strategies such as macrocyclization, prodrugs, and conformational control aim to improve permeability and pharmacokinetics.

2. Selectivity and off-target degradation

Because E3 ligases can engage unintended proteins, proteomic profiling is required to confirm selectivity and mitigate toxicity.

3. E3 ligase expression and tissue specificity

Different tissues express distinct E3 ligases; ligase-dependent PROTACs may have variable efficacy. Developing ligands for new ligases could yield organ-specific degraders.

Continue reading below...
Red tumor cells are shown against a teal backdrop showing attachment to tissue.
WhitepaperDecoding the tumor microenvironment with immune profiling
Integrating multiplexed immunohistochemistry with spatial analysis offers a practical way to uncover tumor-immune dynamics.
Read More

4. Resistance and adaptive responses

Although PROTACs overcome many inhibitor-class resistances, adaptive responses—such as reduced E3 ligase expression—may limit durability.

5. Safety and immunogenicity

By perturbing protein networks, PROTACs can produce unanticipated downstream effects. Comprehensive preclinical assessment of proteomic impact is essential.


Beyond PROTACs: The expanding landscape of targeted protein degradation

PROTACs are part of a growing family of protein fate-modifying technologies:

  • Molecular glues: Small molecules that stabilize a target-E3 interaction without a linker (e.g., lenalidomide/thalidomide analogs).
  • LYTACs (lysosome-targeting chimeras): Developed by the Bertozzi lab (2020), these direct extracellular or membrane proteins to lysosomes via receptors such as CI-M6PR or ASGPR.
  • AUTACs/AUTOTACs/ATTECs: Autophagy-based degraders that recruit p62/SQSTM1 to clear cytosolic or aggregated proteins (Ji et al., Nat Commun 2022).
  • DUBTACs: Molecules that stabilize proteins by recruiting the deubiquitinase OTUB1 to remove ubiquitin chains (Henning et al., Nat Chem Biol 2022).

Together, these tools extend degradation beyond the proteasome, offering new ways to control protein stability.


Future outlook

The therapeutic potential of PROTACs continues to grow. Advances in AI-assisted design, computational modeling, and cryo-EM structural analysis are improving predictions of ternary complex formation and degradation kinetics.

As medicinal chemists refine linker architecture and expand ligase chemistry, future generations of PROTACs will likely feature enhanced selectivity, oral bioavailability, and tissue-specific control.

If late-stage trials such as VERITAC-2 translate into approved therapies, PROTACs will cement a new paradigm in drug discovery—one defined not by inhibiting protein function but by controlling protein fate.


This article was created with the assistance of Generative AI and has undergone editorial review before publishing.

About the Author

  • Trevor Henderson is the Creative Services Director for the Laboratory Products Group at LabX Media Group. With over two decades of experience, he specializes in scientific and technical writing, editing, and content creation. His academic background includes training in human biology, physical anthropology, and community health. Since 2013, he has been developing content to engage and inform scientists and laboratorians.

Related Topics

Loading Next Article...
Loading Next Article...
Subscribe to Newsletter

Subscribe to our eNewsletters

Stay connected with all of the latest from Drug Discovery News.

Subscribe

Sponsored

Portrait of Scott Weitze, Vice President of Research and Technical Standards at My Green Lab, beside text that reads “Tell us what you know: Bringing sustainability into scientific research,” with the My Green Lab logo.
Laboratories account for a surprising share of global emissions and plastic waste, making sustainability a priority for modern research.
A 3D rendering of red and yellow protein molecules floating in a fluid-like environment.
Discover approaches that shorten the path from DNA constructs to purified, functional proteins.
A 3D rendering of two DNA double helices in different colors, representing genetic diversity or molecular comparison on a light background.
By replacing conventional plasmid systems, cell-free DNA synthesis improves speed and quality in mRNA research.
Drug Discovery News September 2025 Issue
Latest IssueVolume 21 • Issue 3 • September 2025

September 2025

September 2025 Issue

Explore this issue