Pages
Products

AI-Driven Protein Degrader Drug Development

Service OverviewDetailed Technical ModulesWet Lab VerificationAdvantagesContact Us

Protein Degrader technology represents a paradigm shift in drug discovery by leveraging the cell's ubiquitin-proteasome system (UPS) to specifically degrade target proteins, successfully breaking through "undruggable" targets that traditional small molecule drugs cannot reach. This technology has transformed the drug discovery landscape, making previously untargetable proteins in the human proteome potentially druggable. However, successful Protein Degrader design involves multidimensional challenges including complex ternary complex conformation prediction, precise linker design, and E3 ligase selection, requiring interdisciplinary innovative approaches.

Creative Biogene integrates artificial intelligence (AI), molecular dynamics simulations, chemoinformatics, and high-throughput computational technologies to provide one-stop services from target validation to preclinical optimization, accelerating the translation of protein degradation drugs from concept to clinic.

AI-Driven Protein Degrader Drug Development Service

We provide a comprehensive AI-driven Protein Degrader drug development service, integrating reinforcement learning modeling, intelligent linker design, virtual screening, molecular dynamics simulations, and data-driven molecular optimization to accelerate the entire process from hit discovery to candidate drug optimization.

Client Requirements

To ensure an efficient and precise project workflow, we recommend that clients provide the following key information:

  • Target Protein (POI) Information: Amino acid sequence, known crystal structures (if available), protein function, and mechanism of action
  • E3 Ligase Selection: Preferred E3 ligases (e.g., VHL, CRBN, IAP, DCAF15) or indication areas for recommendation
  • Existing Protein Degrader Molecules (if available): Any known Protein Degrader analogs or ligand data
  • Drug Development Requirements: Solubility, membrane permeability, metabolic stability, and other pharmacokinetic properties

Service Workflow

Part 01

Data Preparation

  • Pretraining & Fine-Tuning: Optimizing molecular design for drug-like properties
  • Dataset Construction: Integrating PROTAC and quasi-PROTAC molecules
  • Data Processing: Optimizing molecular structures for AI modeling
Part 03

Conditional Molecular Design

  • Ensuring Diversity & Drug-Likeness: Screening 100+ high-potential molecules
Part 05

Molecular Dynamics Simulations

  • Long-Timescale Simulations: Assessing binding stability and resistance risks
  • Free Energy Calculations: Optimizing molecular affinity
Part 07

Chemical Synthesis & Biological Validation

  • Synthesis of 5-10 Candidates
  • In Vitro Testing: Degradation activity, permeability, and metabolic stability
Part 02

Reinforcement Learning Modeling

  • Objective Function Optimization: Enhancing PK, molecular length, and solubility
  • Generating 5,000 Protein Degrader Candidates
Part 04

AI-Based Screening

  • Evaluating bioactivity, binding free energy (GBSA), and synthetic feasibility
  • Selecting 5-10 Candidate Molecules
Part 06

Data-Driven Optimization

  • AI-Guided Pharmacophore & QSAR Modeling: Refining selectivity and potency
  • Scaffold Hopping: Ensuring compound diversity and patentability

Detailed Technical Modules

01 Ternary Complex Structure Prediction

Predicts the stable ternary complex structure between the POI, degradation molecule, and E3 ligase using multi-scale computational simulations, providing the theoretical basis for protein degradation mechanisms.

  • Target-E3 Compatibility
  • Docking & Binding Energy
  • Stability Analysis

02 Intelligent Linker Design

Designs efficient linker structures with optimized drug-like properties to enhance degradation efficiency and avoid the "Hook effect."

  • Spatial Optimization
  • AI-Driven Design
  • Energetic Evaluation

03 Virtual Screening

Integrates structural scoring and AI models to rapidly screen potential protein degrader candidates from large compound libraries, accelerating early-stage discovery.

  • Hybrid Scoring
  • Similarity Clustering
  • ADMET Filtering

04 Molecular Dynamics Simulation

Performs microsecond-level molecular dynamics simulations to quantitatively assess the binding stability and dynamic characteristics of protein degraders with their targets.

  • High-Resolution MD
  • Free Energy Profiling
  • Resistance Prediction

05 Data-Driven Molecular Optimization

Uses deep learning and chemoinformatics to optimize the structure and activity of protein degraders, enhancing selectivity and drug-like properties.

  • Pharmacophore Modeling
  • Deep QSAR
  • Library Management

Figure 1. The general workflow for the design of lead Protein Degraders.

Wet Lab Verification

MethodDescription
Chemical Synthesis & Quality ControlSynthetic Route DesignDesigning efficient synthetic routes based on molecular structural features, optimizing reaction conditions.
PurityEnsuring compound purity using HPLC, mass spectrometry, and other technologies.
In Vitro Biological EvaluationCellular Activity VerificationDetecting target protein degradation efficiency (DC50) via Western Blot using target protein-related cell lines.
Ternary Complex Formation VerificationQuantitatively evaluating Protein Degrader-induced ternary complex formation capabilities using FP or TR-FRET.
Pharmacokinetics & Safety AssessmentIn Vitro ADMET TestingEvaluating candidate molecule solubility, liver microsomal stability, CYP450 inhibitory activity, and hERG toxicity risks.
In Vivo Pharmacokinetic StudiesDetermining blood concentration-time curves (AUC, Cmax, T1/2) through mouse models, assessing oral bioavailability and half-life.

Our Core Advantages

Dual-engine technology combining deep learning with quantum mechanics-optimized molecular simulations, breaking through traditional computational precision limitations and reducing false positive rates

AI & Physical Model Integration

Utilizing large-scale screening technologies to discover and validate globally important cancer driver gene targets, ensuring precision in targeting effects.

Target Selection & Customized Design

Developing unique linker libraries, optimizing oral bioavailability, enhancing degradation efficiency and drugability of compounds.

Unique Linker Library & High Oral Bioavailability

Providing end-to-end solutions from target validation, small molecule design, and activity screening to pharmacodynamic/pharmacokinetic evaluation, seamlessly connecting all R&D stages

Full Process Integration

Standardized parallel workflows and automated high-throughput screening platforms significantly shorten development time, reducing candidate drug confirmation cycles from the traditional 18 months to 5-6 months on average

Efficient R&D Cycle

Electronic record systems compliant with FDA/NMPA/EMA regulatory requirements, ensuring data integrity, traceability, and intellectual property protection, supporting IND submission material preparation.

Data Security Guarantee

Contact Us

Our professional technical team is ready to provide you with customized technical solutions and quotations within 24 hours to support your innovative drug development.

FAQ

Q: What are the main technical challenges in Protein Degrader design?

A: Ternary complex conformation prediction, linker optimization, and E3 ligase compatibility are the three core challenges requiring the integration of AI and molecular simulation technologies.

Q: How is the accuracy of virtual screening guaranteed?

A: We adopt a multidimensional scoring system (physical force fields + deep learning) and continuously optimize our models through experimental validation data.

Q: How long does the service cycle typically take?

A: Standard project cycles are 8-12 weeks, depending on target complexity and data availability.

Reference:

  1. Ma B, Liu D, Wang Z, et al. A Top-Down Design Approach for Generating a Peptide PROTAC Drug Targeting Androgen Receptor for Androgenetic Alopecia Therapy. J Med Chem. 2024;67(12):10336-10349.
* For research use only. Not intended for any clinical use.
Quick Inquiry