VLI-Research

VLI-ResearchVLI-ResearchVLI-Research

VLI-Research

VLI-ResearchVLI-ResearchVLI-Research
  • Sign In
  • Create Account

  • Bookings
  • My Account
  • Signed in as:

  • filler@godaddy.com


  • Bookings
  • My Account
  • Sign out

Signed in as:

filler@godaddy.com

  • Home
  • About
  • Contact
  • Our Research
  • Drug Discovery
  • TPD
  • Protein Expression
  • Mass Spec Standards
  • Solid State NMR

Account


  • Bookings
  • My Account
  • Sign out


  • Sign In
  • Bookings
  • My Account

Targeted Protein Degradation

Redefining Druggability: Eliminating ‘Un-targetable’ Proteins

Catalytic Efficiency & Durability: Beyond Occupancy-Driven Pharmacology

Catalytic Efficiency & Durability: Beyond Occupancy-Driven Pharmacology

 TPD overcomes a fundamental limitation of traditional therapeutics: its ability to degrade disease-causing proteins lacking enzymatic activity or functional pockets. By using bifunctional molecules like PROTACs® (PROteolysis-TArgeting Chimeras) or molecular glues, TPD redirects E3 ubiquitin ligases (e.g., VHL, CRBN) to tag target proteins with destructive K48-linked polyubiquitin chains. This triggers their proteasomal degradation — enabling intervention against transcription factors, scaffold proteins, and aggregated toxins in neurodegeneration or oncology that resist conventional inhibition. Over 80% of the human proteome, previously deemed "undruggable," is now pharmacologically accessible.

Catalytic Efficiency & Durability: Beyond Occupancy-Driven Pharmacology

Catalytic Efficiency & Durability: Beyond Occupancy-Driven Pharmacology

Catalytic Efficiency & Durability: Beyond Occupancy-Driven Pharmacology

 Unlike inhibitors requiring sustained target binding, TPD operates catalytically: a single degrader molecule can eliminate hundreds of target proteins. This efficiency translates to lower dosing and reduced off-target effects. Critically, TPD achieves long-lasting effects — degrading a protein resets cellular homeostasis, whereas inhibitors lose efficacy upon discontinuation. Clinical successes, like the ERα degrader ARV-471 achieving tumor regression in advanced breast cancer patients whose tumors resisted hormone therapies and CDK4/6 inhibitors, highlight this durability. When paired with ubiquitin-proteomics (e.g., LifeSensors’ TUBE-MS), degraders like this can be validated based on their specific K48 vs. K63 ubiquitination signatures, revealing mechanism-of-action with preclinical precision. 

Expanding Horizons: From Oncology to Neurodegeneration and Beyond

Catalytic Efficiency & Durability: Beyond Occupancy-Driven Pharmacology

Expanding Horizons: From Oncology to Neurodegeneration and Beyond

 While oncology dominates current TPD pipelines (24+ PROTACs/glues in human trials at the time of writing), the field is rapidly diversifying. Molecular glues like immunomodulatory drugs (e.g., lenalidomide) degrade IKZF1/3 in hematologic cancers, but newer glue designs degrade tau aggregates implicated in Alzheimer’s. Emerging strategies include: Lysosome-Targeting Chimeras (LYTACs/AUTACs) degrading extracellular/pathogenic proteins, Tissue-restricted degraders using neuron-specific E3 ligases (e.g., FBW7) to avoid systemic toxicity, Peptide-PROTACs disrupting oncogenic transcription factors. As TPD targets expand to include viral proteins, prions, and lipid kinases, the field is poised to treat multiple conditions with a single modality: 

eliminating pathogenesis at the protein source.

Oncology

Other Applications

Expanding Horizons: From Oncology to Neurodegeneration and Beyond

Clinical-stage PROTACs like ARV-110 and ARV-471 demonstrate remarkable efficacy against recalcitrant cancers by catalytically degrading disease drivers that evade inhibition. ARV-110, currently in Phase III trials for metastatic castration-resistant prostate cancer, eliminates androgen receptor (AR) variants with T878X/H875Y mutations – variants that resist enzalutamide and other anti-androgens. Similarly, ARV-471 achieves tumor regression in 88% of heavily pretreated ER+/HER2- metastatic breast cancer patients whose tumors progressed on CDK4/6 inhibitors and hormonal therapies. Beyond hormone receptors, BTK degraders (e.g., NX-2127) effectively clear the C481S-mutant BTK protein in chronic lymphocytic leukemia patients who relapsed on covalent BTK inhibitors like ibrutinib. Molecular glues further expand this arsenal; agents like lenalidomide and pomalidomide degrade IKZF1/3 transcription factors in multiple myeloma, altering gene expression programs impossible to achieve with receptor blockade alone. This capacity to eliminate—rather than merely inhibit—resistant targets positions TPD as a transformative strategy in precision oncology. 

Neurodegeneration

Other Applications

Other Applications

  For Alzheimer's disease, pioneering degrader platforms are being developed to clear tau tangles – including photo-activated PHoTACs that enable spatiotemporal control of tau degradation, and CEREBON-based molecular glues designed to selectively ubiquitinate hyperphosphorylated tau conformers for proteasomal clearance. In Parkinson's disease, LRRK2 degraders target the G2019S-mutant kinase implicated in familial cases, while α-synuclein-directed PROTACs reduce Lewy body burden in preclinical models by hijacking E3 ligases to tag fibrillar aggregates. Unlike antibody therapies that primarily intercept extracellular species, TPD agents can access intracellular aggregates and exploit the ubiquitin-proteasome system to dismantle existing pathology. This strategy addresses a fundamental gap in neurodegenerative therapeutics: the urgent need to reduce the burden of proteotoxic species that drive neuronal dysfunction beyond what passive immunization can achieve. Early-stage clinical programs for tau and α-synuclein degraders are expected to enter human trials within the next 1-2 years, marking a significant expansion beyond oncology. 

Other Applications

Other Applications

Other Applications

 In autoimmune disorders such as rheumatoid arthritis and lupus, BTK degraders like NX-5948 suppress aberrant B-cell activation more comprehensively than reversible inhibitors by eliminating both enzymatic and scaffolding functions of BTK. IRAK4 degraders (e.g., KT-413) potently disrupt toll-like receptor (TLR) and interleukin-1 receptor signaling implicated in inflammatory bowel diseases, offering potential advantages over anticytokine biologics through broader pathway suppression and oral bioavailability. For infectious diseases, degrader-antibody conjugates targeting viral proteins represent an emerging frontier; the SARS-CoV-2 main protease (Nsp5) degrader ALG-097161 accelerates viral clearance by co-opting host ubiquitin machinery, while HIV-focused programs aim to degrade the Tat protein to prevent reactivation from latent reservoirs. These applications highlight TPD’s versatility across therapeutic areas, though challenges remain in mitigating on-target toxicities (e.g., CRBN-mediated teratogenicity) and achieving tissue-selective degradation. Current advancements include engineered degraders with pH-sensitive linkers for gut-restricted activity in inflammatory conditions and nanocarrier-encapsulated PROTACs to enhance brain penetration – innovations poised to expand TPD’s clinical footprint dramatically over the next decade. 

Copyright © 2025 VLI-Research - All Rights Reserved.


Powered by

This website uses cookies.

We use cookies to analyze website traffic and optimize your website experience. By accepting our use of cookies, your data will be aggregated with all other user data.

Accept