Product News | Molecular Glue vs PROTAC: Which is Better in Drug Design?

Jan.31.2024

Targeted Protein Degradation (TPD) mainly involves the degradation of target proteins through the ubiquitin-proteasome and lysosome systems. There are nearly ten different technological routes within TPD, among which molecular glue and PROTAC are the fastest growing ones.

 

BMS's molecular glue Lenalidomide has annual sales of US$12.891 billion. Arvinas' PROTAC molecule ARV-471 has entered Phase III clinical trials, as lysosome-based degradation is relatively new and still in the preclinical stage. 

At present, conventional targets are exhausted and new targets are difficult to discover in drug development. TPD technology provides a new path for drug development and expands the range of targetable proteins, and is one of the most promising technologies for future development. Next, let’s introduce PROTAC and molecular glue.

 


01 Differences between PROTAC and Molecular Glue

 

Proteolysis Targeting Chimeras(PROTACs)and molecular glue are two technologies rapidly developed in recent years in targeted protein degradation. They result in protein degradation for treatment of diseases through different mechanisms, and both methods have the potential to change the traditional way of drug development.

 

① Structural Differences: PROTACs typically consist of three parts: a ligand of a target protein, a linker, and an E3 ligase-binding warhead. This structural design allows PROTAC molecules to interact with both the target protein and the E3 ligase simultaneously.

Whereas, molecular glue is usually a single small molecule that alters the conformation of the target protein or E3 ubiquitin ligase, facilitating their interaction without requiring a specific linker or high-affinity ligand for the target protein (Figure 1).

 

Figure 1. Mode of action and structural features of molecular glue and PROTAC.

 

② Mechanism Differences: The main difference between PROTACs and molecular glue lies in their mechanisms of action. PROTACs induce TPD by connecting the target protein with the ligand for E3 ubiquitin ligase, allowing the ubiquitin attached to the target protein and subsequent degradation in the proteasome. This approach can completely eliminate pathological proteins and counteract their disease activity. In contrast, molecular glue forms stable complexes through stabilization and affinity interactions with the pathological protein, inhibiting its activity or promoting the degradation of the target protein.

 

③ Efficiency Differences: PROTACs can achieve selective degradation of the target protein, and their efficiency depends on the affinity between the PROTAC molecule and the target protein as well as the E3 ligase. Unlike conventional enzyme inhibitors or receptor blockers, PROTACs not only inhibit protein function but also reduce the amount of the protein.

Similarly, molecular glue can induce TPD by directly binding the target protein to the E3 ligase. The efficiency of molecular glue also depends on the affinity between the molecule and the target protein as well as the E3 ligase (Table 1).

 

 

Molecular glue

PROTAC

Mechanism

Binds E3 or target protein induces PPI

Binds target and E3

Target protein (POI)

To be determined

Predictable

Discovery strategy

Historically serendipitous discovery

Rational design

Feature

Monovalent

Bivalent

Linker

Without linker

With linker

Molecular weight

Lower

Higher

Lipinski’s rule of five

Typically within

Beyond

Binding pocket in the target protein

Nonessential

Required

Binding affinity

Weaker binding affinities for either E3 ligase
or target protein

Stonger binding affinities for either E3 ligase
or target protein

Table 1. Comparison of Molecular Glues and PROTACs

 

02 Frontier Developments of PROTAC and Molecular Glue

 

PROTACs have been successfully employed in research of various protein-related diseases and have shown strong therapeutic effects in clinical trials. Examples include ARV-471 targeting estrogen receptor and MT-802 targeting BTK, etc. However, no related products of PROTAC drugs have been approved globally. Some overseas companies, such as Arvinas, C4 Therapeutics, Kymera Therapeutics, and Nurix, have made significant progress.

With Arvinas' two candidates, ARV-110 and ARV-471 obtaining positive clinical data, this field has experienced vigorous growth in the past two years. Currently, multiple PROTAC drugs have entered clinical stages, with targets including AR, ER, BCL-XL, IKZF1/3, STAT3, BTK, TRK, BRD9, etc. In addition, Kymera Therapeutics, C4 Therapeutics, and Nurix Therapeutics have molecules in clinical stages. Chinese pharmaceutical companies like Lynk Pharmaceuticals, Kintor Pharmaceutical, Haisco Pharmaceutical, Meizer Pharma, Cullgen, Hinova Pharmaceuticals, and Seed Therapeutics, a subsidiary of BeyondSpring Pharmaceuticals, have already deployed in the PROTAC field. Among them, Lynk Pharmaceuticals, Kintor Pharmaceutical, and Haisco Pharmaceutical have PROTAC molecules in clinical stages (Table 2).

 

PROTAC

Target

Indications

Phase

Company

ARV-471

Estrogen receptor (ER)

Breast cancer (ER+/HER2-Breast Cancer)

Phase III

Arvinas

ARV-110

Androgen receptor (AR)

Metastatic castration-resistant prostate cancer (mCRPC)

Phase II

Arvinas

ARV-766

Androgen receptor (AR)

Metastatic castration-resistant prostate cancer (mCRPC)

Phase II

Arvinas

KT-474

Interleukin-1 receptor-associated kinase 4 (IRAK-4)

Atopic dermatitis, hidradenitis suppurativa, rheumatoid

arthritis

Phase II

Kymera Therapeutics

LNK01001

-

Rheumatoid arthritis, atopic dermatitis, ankylosing spondylitis

Phase II

Lynk Pharmaceuticals

GT20029

AR

Androgenic alopecia

Phase II

Lynk Pharmaceuticals

CFT7455

IKZF1/3

Relapsed/refractory non-Hodgkin lymphoma or multiple myeloma

Phase I/ II

C4 Therapeutics

CFT1946

BRAF V600

BRAF V600 mutated solid tumors

Phase I/ II

C4 Therapeutics

NX-2127

BIK + IKZF

B cell malignancies

Phase I

Nurix Therapeutics

NX-5948

BTK

B cell malignancies and autoimmune diseases

Phase I

Nurix Therapeutics

UNK01002

-

Blood cancer

Phase I

Lynk Pharmaceuticals

LNK01003

-

Immunity, inflammation

Phase I

Lynk Pharmaceuticals

HSK29116

BTK

B cell malignancies

Phase I

Haisco Pharmaceutical

MZ-001

BTK

B-cell malignancies and autoimmune diseases

Phase I

Meizer Pharma

HP518

AR

mCRPC that fails standard therapy

Phase I

Hinova Pharmaceuticals

CFT8919

EGFR L858R

Resistant EGFR-mutated non-small cell lung cancer (NSCLC)

Phase I

C4 Therapeutics

CFT8634

BRD9

Synovial sarcoma and smarcb1-deficient solid tumors

Phase I

C4 Therapeutics

KYM-001

KYM-001

MYD88 mutated B-cell lymphoma

IND

Kymera Therapeutics

CG416

Tyrosine kinase receptor (TRK)

-

IND

Cullgen

CG428

TRK

-

Preclinical

Cullgen

CG001419

TRK

-

Preclinical

Cullgen

HC-X029

AR-SV

End-line treatment of mCRPC after failure of standard therapy

Preclinical

Hinova Pharmaceuticals

HC-X035

Src-homology 2 domain-containing protein tyrosine phosphatase (SHP2)

KRAS mutated cancer

Preclinical

Hinova Pharmaceuticals

AR-V7

AR-FL & AR-V7

Metastatic castration-resistant prostate cancer (mCRPC)

Preclinical

Arvinas

Table 2. PROTACs in Clinical Trials

 

Another relatively new therapy, molecular glue is still in early research and development.It has shown therapeutic potential in some specific cases, and currently approved molecular glues mainly consist of immunomodulators, namely thalidomide, lenalidomide, and pomalidomide, which are used to treat multiple myeloma and myelodysplastic syndrome, etc. These three molecular glue degraders have molecular weights below 300 Da and degrade target proteins including the transcription factor IKZF1/3 by recruiting the E3 ubiquitin ligase CRBN. Additionally, thalidomide analogs are commonly used as ligands for the E3 ubiquitin ligase CRBN in many PROTAC molecules. For instance, the E3 ligand for ARV-471, which has entered Phase III clinical trials, is based on (R)-thalidomide.

 

Apart from three approved ones, there are currently over twenty molecular glue degraders in clinical or preclinical stages. Overseas companies such as BMS, C4 Therapeutics, Nurix, and Monte Rosa Therapeutics, etc. have made significant progress. Undoubtedly, BMS holds a leading position in this field. The molecular glue pipeline mainly comes from the acquisition of Celgene products in 2019, including Lenalidomide, CC-92480, CC-99282, CC-220, and others. Nurix and C4 Therapeutics also have molecular glue degraders in clinical trials. Preclinical molecular glue development companies include Monte Rosa Therapeutics, Ambagon Therapeutics which targets degradation of intrinsically disordered proteins, and Ranok Therapeutics, which was the first to report a BRD4 degrader. The targets of molecular glue degraders include IKZF1/2/3, RBM39, GSPT1, CK1α, BCL6, Cyclin K, NEK7, primarily for hematologic malignancies, and some solid tumors. Chinese companies like InnoCare Pharma and Gluetacs Therapeutics have also conducted clinical research on molecular glues. InnoCare Pharma’s ICP-490 has already in Phase II clinical trials for multiple myeloma (Table 3).

 

Molecular glue

Target

Indications

Phase

Company

Lenalidomide

1KZF1/3

Multiple myeloma

Approved

BMS/Celgene

Thalidomide

1KZF1/3

Multiple myeloma

Approved

BMS/Celgene

Pomalidomide

1KZF1/3

Multiple myeloma

Approved

BMS/Celgene

CC-92480

1KZF1/3

Multiple myeloma

Phase l/ II/ III

BMS/Celgene

CC-220

FP91/98;1KZF1

Solid tumors

Phase l/ II/ III

BMS/Celgene

E7820

RBM39

Acute myeloid leukemia

Phase II

Eisai

ICP-490

1KZF1/3

Multiple myeloma

Phase II

InnoCare Pharma

CC-99282

1KZF1/3

Lymphoma

Phase I/ II

BMS/Celgene

MRT-2359

GSPTI

Lung cancer and other solid tumors

Phase I/ II

Monte Rosa

CFT-7455

1KZF1/3

Multiple myeloma

Phase I/ II

C4 Therapeutics

DKY-709

1KZF2

NSCLC, melanoma

Phase l/ lb

Novartis

CC-90009

GSPTI

Acute myeloid leukemia

Phase I

BMS/Celgene

GT-919

1KZF1/3

Multiple myeloma

Phase I

Gluetacs Therapeutics

BTX-1188

GSPT1; IKZF1/3

Hematoma, solid tumor

Phase I

BioTheryX

BAY-2666605

PDE3A/SLFN12

Melanoma

Phase I

Bayer

TMX-4116

CK1α

Multiple myeloma

Preclinical

Dana-Farber

(R)-CR8

Cyclin K

Lung cancer and other solid tumors

Preclinical

Broad Institute

BI-3802

Bcl-6

Lymphoma

Preclinical

Boehringer Ingelheim

NRX-252114

β-Catenin mutant

Lung cancer

Preclinical

Nurix Therapeutics

MGD molecule

NEK7

Inflammatory diseases

Preclinical

Monte Rosa

 

Table 3. Molecular glues in Clinical Trials

 

03 Future Trends of PROTAC and Molecular Glue

 

Though there are no PROTAC drugs on the market, several candidates demonstrate preliminary clinical data with significant degradation of intracellular proteins and promising therapeutic effects.However, the validation process is gradual and requires larger sample sizes to confirm these findings. Due to its unique mechanism of action, PROTAC technology is attracting more and more biomedical innovators and entrepreneurs to compete on this new track. Through twenty years of development, PROTAC has broken through the rules of established medicines that have always been widely recognized, and opened a new chapter for the development of new drugs in the continuous exploration and advancement. It is expected that PROTAC technology will find even broader applications in future drug development. Meanwhile, as technology advances, the design and preparation processes of PROTAC are being more precise and controllable.

 

Currently, over 600 E3 ligases have been reported, but only five have been used for molecular glue-mediated degradation, namely CRBN, DDB1, β-TrCP, DCAF15 and SIAH1. The E3 ligase library still holds huge potential for further exploration, and the identification of new ligands for E3 ligases will expand our range of degradable target proteins. Additionally, there is a need for more exploration of the chemical space of molecular glue molecules. Most reported molecular glues so far are still highly similar to thalidomide and its derivatives. This would be indeed a considerable challenge for drug developers. We need to deepen our insights and understanding of protein-protein interaction interface, make more reasonable structure-guided molecular glue designs, in order to truly promote molecular glues into clinical applications and help treat a wider range of diseases.

 

 

Source:

[1] Dong, G.; Ding, Y.; He, S.; Sheng, C. Molecular Glues for Targeted Protein Degradation: From Serendipity to Rational Discovery. J Med Chem. 2021, 64 (15), 10606-10620.

[2] Zhao, L.; Zhao, J.; Zhong K.; Tong, A.; Jia, D. Targeted protein degradation: mechanisms, strategies and application. Signal Transduct Target Ther. 2022, 7 (1), 113.

[3] CAS Insights. Targeted Protein Degradation and Induced Proximity: Molecular Glues Landscape in Drug Discovery. https://www.cas.org/resources/cas-insights/drug-discovery/targeted-protein-induced-proximity (accessed 2024-01-31).

 

Please contact us to remove any infringement.