top of page

From Molecules to Models: How Gen AI is Transforming In Silico Compound Screening Part 1 : Molecular Docking

  • Writer: Gokul Rangarajan
    Gokul Rangarajan
  • Jun 20
  • 12 min read

Updated: Jun 25

2.5x Smarter, Faster, Cheaper: Gen AI’s Disruption of Compound Screening.

This blog is part of the “GenAI in Healthcare Report 2025” by Murali Sudram in collaboration with Pitchworks VC Studio. The report explores how generative AI is reshaping scientific research, clinical workflows, and drug discovery. Stay tuned for more in-depth explorations of real-world applications and enterprise adoption strategies. You can download our Gen AI in Healthcare report from here https://www.pitchworks.club/healthcaregenaireport If you are into manufacturing, you can download our Gen AI manufacturing report here https://www.pitchworks.club/gen-ai-manufacturing-report-2025

In this blog, we explore the power of in silico compound screening—a virtual method to evaluate how potential drug molecules bind to target proteins—and how the integration of Generative AI significantly enhances its speed, accuracy, and scale. We also introduce CDA (Conversational Docking Assistant), developed by Pitchworks VC Studio Group, which allows researchers to interact with complex docking results through natural language queries, generate instant visual insights, and streamline decision-making. This AI-powered approach not only reduces the time and cost of early drug discovery but also improves hit quality, optimizes lead compounds faster, and enables smarter, data-rich experimentation from the very first screening step.


In the relentless pursuit of discovering the next breakthrough drug, scientists are moving beyond test tubes and toward terabytes. In silico compound screening — the process of simulating drug-target interactions on computers — is undergoing a massive transformation, thanks to the power of Generative AI. In Silico Compound Screening In simple terms, In Silico Compound Screening is using powerful computers to quickly test a huge number of virtual chemical compounds to see which ones are most likely to interact with a specific biological target (like a protein or a germ) that's involved in a disease. In silico compound screening is a powerful computational approach used primarily in drug discovery and development to identify potential drug candidates by simulating how molecules interact with biological targets. It essentially replaces or complements early-stage, labor-intensive lab experiments with computer calculations. In silico compound screening is primarily used by computational chemists and medicinal chemists within pharmaceutical and biotechnology companies, as well as academic research institutions and contract research organizations. Computational chemists focus on developing and applying the advanced algorithms and software for virtual screening, molecular docking, and simulations, while medicinal chemists leverage these computational insights to guide the actual design, synthesis, and optimization of chemical compounds in the laboratory. Bioinformaticians and data scientists are also increasingly involved, especially with the integration of AI and machine learning into these processes.

This powerful computational approach is predominantly applied in the early to middle stages of drug discovery and development. Its main impact is during the hit identification/lead discovery phase, where it rapidly filters massive virtual compound libraries (millions to billions of molecules) to identify a much smaller, highly prioritized set of "hits" that are most likely to interact with a specific disease target. It's also crucial in lead optimization, refining these initial hits to improve their potency, selectivity, and predict their ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) properties. By significantly reducing the number of compounds that need expensive and time-consuming physical synthesis and lab testing, in silico screening drastically cuts down on the overall cost and timeframe of drug development, accelerating the journey from concept to potential new medicine.






Traditional Screening (e.g., HTS)
Traditional Screening (e.g., HTS)


Feature

Traditional Screening (e.g., HTS)

In Silico Screening

Methodology

Physical experiments, robotics, lab work

Computer simulations, algorithms, virtual models

Cost

High (reagents, equipment, personnel)

Low (per compound), but requires initial IT investment

Speed

High-throughput (thousands to hundreds of thousands/day)

Extremely high-throughput (millions to billions/day/week)

Compounds

Physical chemical compounds

Digital representations of chemical compounds

Outcome

Identifies active compounds (hits)

Predicts potentially active compounds (virtual hits)

Waste

Generates chemical and biological waste

No physical waste generated

Early Prediction

Limited ADMET prediction, primarily post-screening

Strong capability for early ADMET prediction

Resource Use

Significant lab space, specialized equipment, consumables

High-performance computing infrastructure


In Silico Screening virtual testing
In Silico Screening


Traditionally, pharmaceutical companies and research institutes had to physically test thousands, sometimes millions, of chemical compounds in labs, a process that was slow, costly, and often full of dead ends. Even with high-throughput screening (HTS), the sheer size of possible chemical spaces — estimated at over 10⁶⁰ compounds — makes exhaustive exploration nearly impossible.



In silico compound screening is used by computational chemists, medicinal chemists, and bioinformatics experts across pharma and biotech. From early-stage drug discovery teams at Pfizer, Novartis, and Roche to AI-focused biotech startups like Insilico Medicine, Atomwise, and Recursion, virtual screening is a critical step in narrowing down which molecules are most promising before moving to synthesis and lab testing.






In Silico Compound Screening tools
In Silico Compound Screening tools

In Silico Compound Screening (Computational)

In contrast, in silico compound screening is entirely performed on computers, leveraging computational models and algorithms.


  • Process:

    1. Target and Ligand Preparation (Virtual): Instead of physical entities, 3D digital models of the biological target (e.g., a protein's crystal structure) and a vast virtual library of chemical compounds are used.

    2. Virtual Interaction Simulation: Computational algorithms (like molecular docking) "fit" each virtual compound into the binding site of the virtual target. They simulate how the molecules would interact based on their shapes, charges, and chemical properties.

    3. Scoring and Ranking: A "scoring function" assigns a numerical value to each predicted interaction, estimating how strongly a compound might bind. Compounds are then ranked by these scores.

    4.  Filtering and Prediction: Additional computational filters, such as predicting ADMET (Absorption, Distribution, Metabolism, Excretion, Toxicity) properties, are applied to further refine the list of candidates.

    5. Hit Selection for Lab Testing: A much smaller, highly prioritized list of the most promising compounds from the virtual screen is selected. These compounds are then physically acquired or synthesized for actual lab (in vitro) testing.

  • Characteristics:

    • Virtual: Uses only digital data and computer processing.

    • Low Cost (per compound screened): Once the computational infrastructure is in place, the cost of screening an additional virtual compound is negligible. No physical reagents or consumables are used.

    • Extremely Fast: Millions to billions of compounds can be screened in a matter of days or weeks, a scale impossible with traditional methods alone.

    • Early Prediction: Allows for early prediction of binding affinity and drug-like properties, enabling "fail early, fail fast" strategies.

    • No Waste: Generates no physical waste.

    • Relies on Models: The accuracy is dependent on the quality and robustness of the underlying computational models and algorithms.


Comprehensive In Silico Compound Screening tools enable rapid, cost-effective virtual evaluation of millions of drug-like molecules. These tools combine molecular modeling, docking, ADMET prediction, and AI-driven analytics to accelerate early-stage drug discovery.

Category

Purpose

Toolstack Used Today

AI/ML Features

Molecular Docking Software

Predict ligand binding pose and affinity to target proteins

AutoDock, AutoDock Vina, Glide (Schrödinger), GOLD (CCDC), DOCK, FlexX, ICM-Pro, MOE-Dock, rDock, LeDock, Surflex-Dock, SwissDock

Partial (Glide, ICM-Pro)

Pharmacophore Modeling Tools

Identify key 3D chemical features required for bioactivity

PHASE, LigandScout, Discovery Studio (Catalyst), MOE-Pharmacophore, ICM-Chemist, ZINCPharmer, Pharmit, Pharmer, PharmaGist

Partial (LigandScout, DS)

Virtual High-Throughput Screening (vHTS)

Screen millions of compounds quickly using computational pipelines

Schrödinger Suite (HTVS), Discovery Studio, MOE, MCE vHTS, CRO services, AutoDock Vina with scripting

Yes (via scoring + ML)

Molecular Modeling & Visualization

Build, view, and manipulate molecular structures in 2D/3D

PyMOL, UCSF Chimera/X, Maestro, Discovery Studio, MOE, VMD, Jmol, Avogadro, ChemDraw, MarvinSketch

No

QSAR / QSPR Modeling Tools

Predict activity or properties based on molecular descriptors

MOE, Discovery Studio, Spartan, ADMEWORKS, alvaModel, Dragon, KNIME, R (rcdk, caret), Python (RDKit, DeepChem, scikit-learn, padelpy)

Yes (ML/AI core)

Molecular Dynamics (MD) Simulation

Simulate molecular behavior over time (e.g., binding dynamics)

GROMACS, AMBER, NAMD, CHARMM, LAMMPS, OpenMM, Desmond

Partial (ML-assisted MD)

ADMET Prediction Tools

Forecast Absorption, Distribution, Metabolism, Excretion, and Toxicity

SwissADME, ADMET Predictor, pkCSM, ProTox-II, PreADMET, ACD/Labs Percepta, Schrödinger ADMET modules, Discovery Studio, MOE

Yes (core function)

lets Look into each use case and how gen ai is usefull in the process Molecular docking software predicts how a small molecule (ligand) binds and interacts with a target protein at the atomic level. It simulates the ligand entering the protein's binding site and estimates the binding pose (how the ligand fits in the site) and binding affinity (how strong the interaction is). This helps researchers understand drug–target interactions, optimize compounds, and accelerate drug discovery.


1. AutoDock

  • Type: Open-source

  • Key Features:

    • Uses a Lamarckian Genetic Algorithm to search for optimal ligand poses.

    • Supports ligand and protein flexibility, which is critical for realistic simulations.

    • Popular in academic research and used in automated pipelines.

  • Strengths:

    • Versatile and customizable.

    • Integrates well with batch docking and scripting environments.

  • Use Case:

    • Ideal for large-scale screening of compounds or protein–ligand interaction studies.

2. Glide (by Schrödinger)

  • Type: Commercial (Proprietary software)

  • Key Features:

    • Uses Standard Precision (SP) and Extra Precision (XP) scoring functions.

    • Built for high-accuracy docking with minimized false positives.

    • Integrates seamlessly with Schrödinger’s full suite: LigPrep, Protein Preparation Wizard, MM-GBSA, etc.

  • Strengths:

    • Industry-grade accuracy.

    • Strong visualization, detailed reports, and ligand strain analysis.

  • Use Case:

    • Frequently used in pharmaceutical pipelines for lead optimization and structure-based drug design (SBDD).

3. Surflex-Dock

  • Type: Commercial

  • Key Features:

    • Generates a “protomol”—an idealized ligand to define the active site.

    • Then docks real ligands to match this protomol's shape and polarity.

  • Strengths:

    • Robust shape-based docking.

    • Works well even when ligand information is sparse.

  • Use Case:

    • Suited for virtual screening of diverse libraries when only the receptor is known.



Molecular docking is typically used in the early to mid stages of the drug discovery and development pipeline, especially during:



[1] Target Discovery

[2] Target Validation

[3] Virtual Screening (← Molecular Docking starts here)

[4] Hit Identification

[5] Lead Optimization

[6] Preclinical Testing

[7] Clinical Trials



Where Molecular Docking Fits in Drug Discovery

Molecular docking plays a critical role across multiple stages of the drug discovery pipeline. During target validation, it helps researchers determine whether a protein is druggable and identify potential binding pockets. In the hit identification phase, large libraries of compounds are virtually screened to predict which molecules can bind to the target protein. Once promising candidates are identified, hit-to-lead optimization uses docking to refine these molecules, improving their binding strength and selectivity. Finally, during lead optimization, docking simulations predict how structural modifications to these compounds will influence their interactions with the target—helping reduce the need for costly and time-consuming experimental testing.



A wide range of professionals across life sciences and pharmaceutical research rely on molecular docking tools. Computational chemists run simulations, analyze binding poses and affinities, and refine scoring workflows. Medicinal chemists use docking insights to guide the design and synthesis of more potent compounds. Structural biologists leverage docking to validate experimental binding modes and understand protein-ligand interactions. Pharmaceutical scientists evaluate structure–activity relationships (SAR) and incorporate docking data into broader studies involving pharmacokinetics and ADMET. Bioinformaticians and data scientists use docking outputs as part of machine learning pipelines for predictive modeling and virtual screening. Finally, graduate students and academic researchers apply these tools in research projects, thesis work, or for in silico validation in early discovery stages. Gen AI use cases which can be adopted today on day 1 1. 🧾 Auto-Generated Docking Reports (Used in Academia & Biotech)

AutoDock used in large-scale screening of compounds
AutoDock used in large-scale screening of compounds




large-scale screening of compounds
large-scale screening of compounds


  • What Happens:

    • After docking (e.g., in AutoDock Vina) with gen Gen AI tools (like GPT or Codex-based notebooks) auto-read .log and .pdbqt files.

    • They generate:

      • PDF summaries with top poses & energies

        Autodoc summaries can be generated in PDF form gen ai integration of GPT 4 or Lang chain+ Streamlit
        Autodoc summaries can be generated in PDF form gen ai integration of GPT 4 or Lang chain+ Streamlit


      • Charts (bar plots of binding affinities)

      • Tables of residues involved in H-bonding or hydrophobic contacts

    Used in: Academic publications, thesis documentation, internal biotech reports.

    Tools: GPT-4 + LangChain + Streamlit or Jupyter.

2. Pose Visualization + Captioning (AI Described Visuals)

  • What Happens:

    • Protein-ligand interaction maps (from tools like Maestro or PyMOL) are auto-captioned by GPT-based agents.

    • Descriptions like:

      “The ligand fits snugly in the hydrophobic pocket near ASN52, stabilized by 2 hydrogen bonds.”

  • Why It Matters: Saves time and adds interpretability for non-experts.

  • Used in: Biotech team meetings, academic presentations.

  • Tools: GPT-Vision APIs + PyMOL integration scripts.

3. Conversational Docking Assistant (Being piloted in pharma & research labs)

  • What Happens:

    • Researchers upload a protein and ligand.

    • Then they ask things like:

      • “Which residues does the ligand interact with?”

      • “Rank the top 3 poses by binding score and explain.”

    • The AI responds contextually using docking data and structural files.

  • Used in: Early-stage pharma R&D teams and custom LLM setups in biotech.

  • Tools: GPT-4 + Retrieval-Augmented Generation (RAG) with molecular data.


Conversational Docking Assistant developed by Pitchworks VC Studio
Conversational Docking Assistant developed by Pitchworks VC Studio

The Conversational Docking Assistant (CDA) developed by Pitchworks is an AI-powered interface designed to transform how researchers interact with molecular docking data. By integrating natural language processing with molecular file parsing and docking outputs, CDA allows scientists to upload protein and ligand files and ask contextual questions like “Which residues interact with the ligand?” or “Show top 3 docking poses by binding score.” This streamlines the early-stage drug discovery process by replacing manual inspection and scripting with intuitive, real-time insights.


Having a option to export report as PDF, Export vsv and Export PDB
Having a option to export report as PDF, Export vsv and Export PDB


Upload Molecular Files Protein File (pdb/mol2) load Ligand File (.sdf/.mol/.pdbqt)  .png
Upload Molecular Files Protein File (pdb/mol2) load Ligand File (.sdf/.mol/.pdbqt) .png

The core benefit lies in boosting R&D productivity. Instead of relying on traditional tools that require command-line knowledge or script-based querying, CDA enables fast exploration of docking results using a chat-based UI. The assistant surfaces interaction summaries, pose rankings, 2D/3D visualizations, and exports—all within a few clicks. For research teams juggling multiple compounds and targets, this massively reduces analysis turnaround time.


CDA 



You can ask the assistant specific compound questions or open-ended prompts from the recent research
You can ask the assistant specific compound questions or open-ended prompts from the recent research



Technically, CDA integrates with molecular docking engines like AutoDock Vina, Schrödinger Glide, or DOCK, and connects their output (PDBQT files, logs, pose scores) to a Retrieval-Augmented Generation (RAG) system powered by GPT-4. This allows the AI to contextually interpret structural data, identify key residue-ligand interactions, and explain pose rankings based on energy scores or binding affinity. The system can be layered on top of existing docking pipelines, making integration into pharma and biotech workflows seamless.


Docking Results and Analysis Generation
Docking Results and Analysis Generation

The UI is split into two main sections: a summary card at the top and a detailed docking table below. The summary card highlights the best pose with its binding score and visual tags for key interactions (e.g., H-bond, π-π). Icons or colored badges denote interaction types. Below, the interactive table lists all poses, sortable by pose ID, binding score, or energy. Each row includes a compact view of interacting residues, with tooltips on hover. A side panel or click action loads a 3D molecular viewer showing the pose's binding conformation and interaction map.



Visualize each compound
Visualize each compound


Feature-wise, CDA includes protein/ligand upload, an interactive chat module, a real-time molecular viewer, downloadable reports, and pose filtering tools. It can be extended with pose re-ranking, interaction map generation, or even federated model querying across datasets. The design is clean, modular, and built for both web-based and intranet deployment, ensuring flexibility in high-compliance lab settings.

Here is a quick demo of the blog

The Pitchworks Conversational Docking Assistant (CDA) is purpose-built to tightly integrate with docking engines like AutoDock and AutoDock Vina, automating every step from file parsing to pose analysis. Once a researcher uploads .pdbqt files for the protein and ligand, CDA automatically reads the output logs, extracts binding affinities, pose rankings, and residue-level interactions—then compiles these into interactive tables, charts, and downloadable PDFs. CDA also allows batch processing of hundreds of docking outputs, filters top-scoring compounds based on user-defined thresholds, and generates cross-comparisons across docking runs. In our internal implementations, this integration with AutoDock reduced data interpretation time by 70%, freeing up chemists and bioinformaticians to focus on compound prioritization rather than technical overhead.

In high-throughput screening scenarios, where thousands of compounds are docked virtually, CDA's AutoDock integration provided seamless pose alignment visualizations, hydrogen bond maps, and hydrophobic interaction summaries—without requiring users to open separate tools like PyMOL or command-line viewers. The assistant was also deployed in air-gapped or HIPAA-compliant environments, ensuring no sensitive structural or compound data left secure servers. This not only protected intellectual property but allowed for auditable, compliant workflows in preclinical candidate selection. Teams using CDA reported a marked improvement in decision velocity, clarity in structure–activity insights, and greater confidence in selecting compounds for synthesis and wet-lab testing—establishing CDA as a foundational tool in AI-enhanced computational chemistry pipelines.

In short, CDA bridges AI and computational chemistry, simplifying access to molecular insights. For any team running high-throughput virtual screening or structure-based design, it unlocks a smarter, faster, and more interpretable approach to docking analysis.

Conclusion In silico compound screening, when combined with Generative AI, is revolutionizing early-stage drug discovery. What once required expensive high-throughput labs and weeks of analysis can now be achieved in days using AI-powered assistants. From auto-generating docking reports and visualizing binding interactions to enabling real-time question-answering, platforms like Pitchworks’ Conversational Docking Assistant (CDA) turn raw molecular data into decision-ready insights. Gen AI reduces manual workload, improves the interpretability of results, and allows researchers to “fail fast” and refine their focus early in the pipeline—maximizing ROI on synthesis and wet-lab validation.

However, this AI-augmented workflow introduces new data risks. Docking simulations often involve sensitive molecular structures, proprietary compound libraries, or unpublished protein models, which may fall under IP or clinical confidentiality. Using public LLMs (Large Language Models) without guardrails could expose this sensitive data to third-party servers or unknown retention policies. This makes the case for private LLM deployments—models run on local servers or secure cloud environments—to ensure strict control over how data is processed, stored, and interpreted.

For healthcare and pharmaceutical use, adherence to regulations like HIPAA (for patient-related data) and FDA 21 CFR Part 11 (for electronic records and signatures) is critical. AI tools intended for regulated drug development must be validated, traceable, and secure. Private Gen AI setups should support encryption, audit trails, and role-based access, with AI models fine-tuned on domain-specific data and cleared for internal use. Tools like Pitchworks CDA are being designed with these standards in mind—making AI not just powerful, but trustworthy and compliant for modern drug discovery teams.


Comments


bottom of page