How many FDA approved Molecular glue are there?

17 March 2025
Introduction to Molecular Glues

Molecular glues represent a unique class of small molecules that modulate protein–protein interactions (PPIs) by inducing or stabilizing non-native contacts between proteins. Unlike traditional inhibitors, these molecules do not simply block an active site; rather, they facilitate the formation of ternary complexes that can lead to altered protein function, stabilization, or ubiquitination and subsequent degradation. This mechanism is particularly valuable when targeting proteins that are considered “undruggable” by classical approaches. As a result, molecular glues have emerged as a promising modality for therapeutic intervention in diseases where conventional drug discovery strategies have met with limited success.

Definition and Mechanism of Action

At its core, a molecular glue is a monovalent small molecule that enhances or stabilizes the interaction between two proteins that do not normally associate with high affinity. The key concept behind molecular glues is their ability to bind to one protein (often an E3 ubiquitin ligase) and remodel its surface to attract a target protein, thereby triggering either inhibition or degradation of the target. Such proximity-induced effects are usually achieved without the need for a linker, making these molecules smaller and often having more favorable pharmacokinetic properties than their bifunctional counterparts, such as PROTACs. The modulation of PPIs in this way can lead to therapeutic outcomes through mechanisms such as targeted protein degradation, complex stabilization, or even modulation of signaling cascades.

Historical Development and Significance

The concept of molecular glues traces back to early examples encountered in nature and serendipitous chemical discoveries. One of the earliest examples is the plant hormone auxin, which mediates the interaction between proteins involved in growth regulation. In the pharmaceutical realm, thalidomide was initially developed in the 1950s as a sedative and for the treatment of morning sickness. Despite its infamous history due to teratogenic effects, subsequent research revealed that thalidomide and its analogs act as molecular glues, recruiting the E3 ubiquitin ligase Cereblon (CRBN) to induce the degradation of specific transcription factors such as Ikaros (IKZF1) and Aiolos (IKZF3). This discovery not only rehabilitated the role of thalidomide but also paved the way for designing drugs that exploit induced proximity as a therapeutic strategy. Over the decades, incremental discoveries and structural studies have broadened our understanding and validated the significance of molecular glues in drug development.

FDA Approval Process for Molecular Glues

The pathway to FDA approval for any new therapeutic is multifaceted and stringent, ensuring that only agents with a well-documented safety and efficacy profile reach the market. For molecular glues, many of which have originally been discovered by serendipity, the path to approval necessitates a demonstration of both a novel mechanism of action and a clear clinical benefit over existing therapies.

Overview of FDA Approval Stages

The FDA approval process generally involves multiple stages:

1. Preclinical Studies: Molecular glue candidates are initially characterized in vitro and in animal models to assess their pharmacokinetic properties, mechanisms of action, and toxicity profiles. For molecular glues, studies often focus on demonstrating their ability to modulate PPIs and induce targeted protein degradation.

2. Investigational New Drug (IND) Application: Following promising preclinical results, sponsors file an IND application. This step includes detailed information on the candidate’s pharmacology, toxicology, and a plan for clinical trials. Given the novelty of molecular glues, a robust explanation of their induced proximity mechanisms is often required.

3. Clinical Trials: The clinical phase is typically divided into Phase I (safety and dosage), Phase II (efficacy and side effects), and Phase III (confirmation of effectiveness, monitoring of adverse reactions, and collection of information that will allow the drug to be used safely). Because molecular glues can target proteins traditionally considered “undruggable”, these trials not only evaluate traditional endpoints (such as overall survival and progression-free survival) but also biomarker modulation that confirms the mechanism of action.

4. New Drug Application (NDA) Submission: Once clinical trials demonstrate safety and efficacy, an NDA is submitted to the FDA for review. The application includes all preclinical, clinical, and manufacturing data. Given the unique mechanism of molecular glues, the NDA must also comprise detailed explanations of the induced ternary complex formation and its downstream biological effects.

5. FDA Review and Approval: During the review process, the FDA evaluates all submitted data, often requesting additional studies or information. For molecular glues, the review may focus on understanding the specifics of their degradation mechanism and assessing the risk–benefit profile.

Criteria for Approval

For a molecular glue to gain FDA approval, several criteria must be met:
- Safety: Extensive toxicity data must be provided to ensure that the molecule does not exert off-target effects that could result in adverse clinical events.
- Efficacy: The drug must demonstrate a clear clinical benefit over existing therapies. In the context of molecular glues, this often involves showing significant reduction in target protein levels and corresponding improvement in clinical outcomes.
- Mechanism of Action: A clear elucidation of the molecular glue mechanism is essential. This includes structural data showing the formation of a ternary complex and biochemical evidence that the intended pathway is engaged.
- Manufacturing and Pharmacokinetics: The candidate must have adequate bioavailability, stability, and acceptable pharmacokinetics/pharmacodynamics (PK/PD) profiles for clinical use.

The rigorous nature of these requirements, coupled with the novelty of molecular glues as a therapeutic class, necessitates a detailed and transparent approach in the regulatory submission process.

Current FDA Approved Molecular Glues

When discussing FDA-approved molecular glues, the literature and available databases consistently indicate that the current approved portfolio is limited to a small set of molecules. In fact, the only FDA-approved molecular glues as defined by their mechanism of action in targeted protein degradation are thalidomide and its analogs, lenalidomide and pomalidomide.

List and Description of Approved Drugs

1. Thalidomide:
Initially infamous for its teratogenic effects, thalidomide was later repurposed after its molecular glue properties were elucidated. Thalidomide binds to CRBN and thereby modulates the recruitment of transcription factors for degradation. Despite its rocky past, its efficacy in certain hematological malignancies has been validated, leading to its re-approval and establishment as a molecular glue with a novel mechanism of action.

2. Lenalidomide (Revlimid):
Developed as a safer analog of thalidomide, lenalidomide enhances the degradation of specific neosubstrates via its interaction with CRBN. It has been approved for multiple indications, including multiple myeloma, myelodysplastic syndrome (specifically deletion 5q MDS), and mantle cell lymphoma. The improved safety and efficacy profile compared to thalidomide has enabled lenalidomide to become one of the cornerstone drugs in the treatment of hematological cancers.

3. Pomalidomide (Pomalyst):
As a third-generation immunomodulatory derivative, pomalidomide further refines the molecular glue mechanism by more effectively recruiting CRBN to target proteins for degradation. It has also received FDA approval for the treatment of multiple myeloma, particularly in patients who have failed previous lines of therapy. Its clinical success reinforces the utility of modulating PPIs through a molecular glue mechanism.

Across multiple sources, including structured analyses from the Synapse database, it is consistently reported that these three agents—thalidomide, lenalidomide, and pomalidomide—comprise the complete list of FDA-approved molecular glues. No additional drugs have received approval strictly under the molecular glue designation despite ongoing research and emerging pipelines aiming to expand beyond the well-characterized CRBN-mediated mechanism.

Therapeutic Areas and Indications

The approved molecular glues primarily target hematological malignancies and related diseases:
- Multiple Myeloma:
All three agents have been extensively used in the clinical management of multiple myeloma. Their ability to modulate the degradation of key transcription factors and regulatory proteins is directly linked to the suppression of myeloma cell proliferation.
- Myelodysplastic Syndrome (MDS):
Lenalidomide, in particular, has demonstrated efficacy in deletion 5q MDS. The drug’s molecular glue properties contribute to the correction of dysregulated protein interactions that are essential for the survival of malignant clones in MDS.
- Lymphomas:
In addition to multiple myeloma, lenalidomide is also indicated for use in certain types of lymphomas, such as mantle cell lymphoma. Its immunomodulatory and protein degradation mechanisms contribute to its clinical benefits in such indications.

The broad therapeutic benefits, especially in oncology and hematology, underscore the importance of these molecular glues and provide a rationale for further expanding this drug class to other undruggable targets in different disease areas.

Future Prospects and Research Directions

Despite the clear clinical success of thalidomide, lenalidomide, and pomalidomide, the future of molecular glues as a therapeutic modality is both promising and challenging. Research is actively focused on understanding the detailed mechanisms of action and on overcoming limitations in the rational design of these compounds.

Challenges in Development

One of the major challenges in the development of molecular glues is the reliance on serendipitous discovery. Traditionally, many molecular glues were identified by chance in high-throughput screening experiments rather than through a rational design strategy. This unpredictable nature of discovery presents several hurdles:
- Rational Design Limitations:
While significant progress has been made in understanding the structural prerequisites for molecular glue function, the prediction of effective glue-like interactions remains difficult. Detailed structural and computational studies are required to accurately model the induced proximity mechanisms, and current computational tools such as docking and molecular dynamics simulations are still being refined for this purpose.

- Expanding the Range of E3 Ligases:
To date, nearly all approved molecular glues function via interaction with the CRBN E3 ligase. However, the human genome encodes more than 600 E3 ligases, many of which remain untapped as potential targets for molecular glue agents. Expanding the recruitment of different E3 ligases could potentially broaden the therapeutic scope of molecular glues to target a wider array of disease-relevant proteins.

- Pharmacokinetic Challenges:
Although molecular glues tend to have favorable physicochemical properties compared to larger bifunctional molecules, ensuring optimal bioavailability and minimizing off-target effects remain crucial challenges. This is particularly important for molecules that operate in a substoichiometric manner in vivo.

Emerging Trends and Innovations

Innovations in both the discovery and design of molecular glues are rapidly advancing:
- Structure-Based Drug Design:
Recent advances in structural biology, including cryo-electron microscopy and high-resolution X-ray crystallography, are providing unprecedented insights into the ternary complexes formed by molecular glues. These insights are fueling the development of rational design strategies, enabling scientists to predict and optimize glue-like interactions more systematically rather than relying solely on chance.

- Computational Modeling and High-Throughput Screening:
Integration of computational techniques with innovative high-throughput screening methods is now being used to identify novel molecular glues with better precision. Advanced algorithms that simulate the dynamic conformational changes in protein–protein interfaces are poised to accelerate the discovery of new glue candidates.

- Expanding Therapeutic Applications:
While the current FDA-approved molecular glues are confined to hematological malignancies, there is significant interest in exploring these compounds for applications in other disease areas such as solid tumors, autoimmune diseases, and even neurodegenerative disorders. Several biotechnology companies are investing in platforms aimed at expanding the molecular glue landscape beyond the CRBN paradigm.

- Synergistic Modalities:
There is also growing interest in combining molecular glue therapeutics with other treatment strategies, such as immunotherapies and conventional chemotherapies. Such combination approaches could enhance overall efficacy and overcome drug resistance by exploiting multiple cellular pathways concurrently.

Conclusion

In summary, current evidence from multiple structured synapse references indicates that there are exactly three FDA-approved molecular glues: thalidomide, lenalidomide, and pomalidomide. Their approval is based on a robust mechanism of action that involves binding to the E3 ubiquitin ligase Cereblon (CRBN) and promoting ubiquitination and degradation of disease-driving proteins. These drugs have been transformative in treating multiple myeloma, certain subtypes of myelodysplastic syndrome, and specific lymphomas.

The FDA approval process for these drugs involved rigorous multi-phase clinical trials and thorough mechanistic studies. As a result, the safety and efficacy profiles of these agents have been well established. However, despite their success, the process of discovering and designing new molecular glues remains challenging due to the serendipitous nature of their initial identification and the intricate details of induced proximity dynamics.

Looking to the future, ongoing research is focused on developing rational design frameworks, leveraging advanced structural and computational methods, and expanding the targetable E3 ligase repertoire. These efforts are expected to pave the way for a new generation of molecular glues that could potentially target a broader range of diseases, moving beyond hematological cancers and enhancing treatment options across a wide spectrum of clinical indications.

The detailed analysis presented here—spanning from an introduction to the nature of molecular glues, through the FDA approval process, to future research directions—illustrates not only the current state of the art but also the promising future outlook for this innovative therapeutic approach. In conclusion, with only three FDA-approved molecular glues to date, the field is ripe for innovation, and new discoveries in this area may revolutionize how we approach previously undruggable targets in the coming years.

For an experience with the large-scale biopharmaceutical model Hiro-LS, please click here for a quick and free trial of its features

图形用户界面, 图示

描述已自动生成