AI in lead optimization: From Trial-and-Error to First-Try Precision
***If you're already familiar with lead optimization, I suggest skipping directly to How AI Is Reshaping Lead Optimization.
Before AI began reshaping hit-to-lead workflows, lead optimization was-and still is-a domain where experience, precision, and patience converge, guided by the expertise of medicinal chemists. This critical phase in drug discovery refines promising hits into viable candidates through iterative cycles of design, synthesis, and testing. At its core, lead optimization juggles three imperatives: increase a compound’s potency and selectivity, fine-tune its pharmacokinetics, and reduce its toxic liabilities, all without tipping the balance too far in any direction.
In this section, I revisit the traditional toolkit of lead optimization, still essential today, structured around four core strategies: medicinal chemistry, pharmacokinetics, safety profiling, and early computational methods.
1. Medicinal Chemistry: Iterating Toward the Ideal
Medicinal chemistry is the craft behind the compound. Through careful structural modification, chemists sculpt molecules for better fit, function, and fate. Several foundational strategies guide this process:
- SAR (Structure-Activity Relationships): By systematically tweaking substituents and observing changes in activity, SAR helps identify which molecular features are critical and which are tunable. Quantitative SAR (QSAR) further links structural descriptors to potency, allowing for predictive modeling of analogs before synthesis.
- Functional Group Modifications: Small tweaks often yield big gains. Adding a methyl can fill a hydrophobic pocket ("magic methyl"), while introducing polar groups can enhance solubility and absorption. Fluorine atoms may block metabolic weak spots, and prodrugs can temporarily mask polarity to enhance permeability.
- Bioisosteric Replacements: Swapping problematic groups for functionally similar ones-like replacing an ester with an amide or a carboxylic acid with a tetrazole-can improve metabolic stability, retain activity, and navigate around IP constraints.
- Molecular Rigidification: Reducing conformational flexibility by introducing rings or double bonds can lock a molecule into its bioactive form, often improving target selectivity and reducing off-target binding. This strategy reduces the entropic penalty of binding by pre-organizing the ligand, though it may come at the cost of reduced flexibility in the unbound state.
- Scaffold Hopping and Hybrid Design: When a series hits a wall, scaffold hopping can reboot the campaign with a new chemotype that retains key interactions. Hybrid molecules merge the best features from multiple leads, often solving conflicting issues like potency versus solubility in a single stroke.
These tools are often used in concert, guided by multiparameter optimization frameworks to balance potency, physicochemical properties, and safety. Lead optimization is not just about getting a molecule to bind; it's about shaping it to survive and thrive in a biological system.
2. Pharmacokinetics: Shaping Exposure, Not Just Effect
A drug’s journey through the body-how it’s absorbed, distributed, metabolized, and excreted (ADME)-often determines whether it succeeds in the clinic. Many potent leads fail not because they don’t work, but because they can’t get where they need to go, or they don’t stay there long enough. In traditional lead optimization, ADME considerations are not an afterthought; they are a core axis of design.
Absorption and Solubility For oral drugs, solubility is step zero. A compound must dissolve in the gastrointestinal tract to be absorbed, and poor aqueous solubility can cap exposure regardless of how potent the molecule is. Medicinal chemists often address this by adding polar or ionizable groups, designing salt forms, or developing prodrugs that boost solubility before conversion to the active form in vivo.
Permeability Molecules that are too polar or too large may struggle to cross lipid membranes. Lead series are typically designed to align with Lipinski’s Rule of Five, staying within a range of molecular weight, hydrogen bonding, and lipophilicity that favors oral absorption. Tools like Caco-2 or PAMPA assays help flag permeability issues early, guiding structural tweaks such as reducing hydrogen bond donors or tuning logP.
Distribution and Plasma Protein Binding After absorption, the next hurdle is distribution-reaching the target tissue in the right concentration. Distribution depends on lipophilicity, molecular size, and plasma protein binding (PPB). High PPB can limit free drug levels, while excessive tissue partitioning can complicate dosing and clearance. Chemists adjust polarity and reduce hydrophobic moieties to modulate distribution volume and achieve a more predictable exposure profile.
Metabolic Stability and CYP Interactions Rapid metabolic clearance is a classic failure mode in early leads. The liver’s CYP450 enzymes frequently oxidize “soft spots” such as benzylic positions or heteroatom linkers. Traditional med chem responses include blocking these sites with fluorine or methyl groups, or swapping out labile moieties for bioisosteres that resist metabolism.
CYP interactions are also monitored to prevent drug-drug interactions (DDIs). Compounds that inhibit or strongly induce key CYP enzymes (like CYP3A4 or CYP2D6) may need to be deprioritized or structurally modified to reduce binding affinity to the enzyme active site.
Elimination and Multi-Parameter Balancing Elimination routes (renal vs. biliary) depend on polarity and metabolic processing. Very polar drugs may rely on active transport, which introduces variability; highly lipophilic ones may face accumulation risks. Often, the goal is moderate polarity-enough for solubility and excretion, but not so much that permeability is lost.
Throughout optimization, teams balance conflicting PK parameters-logP, solubility, permeability, metabolic stability, and volume of distribution-using iterative SAR, ADME assays, and in vivo pharmacokinetic profiling in rodent models. Metrics like Lipophilic Efficiency (LipE) help evaluate trade-offs between potency and drug-likeness. The goal is a compound that delivers sufficient, sustained exposure at the target site without triggering off-target effects or metabolic instability.
3. Safety Profiling: Eliminating Risk Before It’s Real
Potency without safety is a dead end. Traditional lead optimization pays close attention to early toxicity flags-not just to protect future trial subjects, but to avoid investing in doomed candidates. This means designing out liabilities before they become expensive failures.
Off-Target Activity and Selectivity Selectivity is a first-order filter. The ideal lead binds tightly to its intended target and ignores everything else. Lack of selectivity can lead to side effects or mechanism-unrelated toxicity. Early in optimization, leads are screened across broad target panels (e.g., kinases, receptors, ion channels) to catch unintended interactions. Analog design then focuses on removing functional groups or conformations that drive these off-target effects.
Tuning selectivity often requires trade-offs-sometimes sacrificing a bit of potency to gain a cleaner profile. Strategies like molecular rigidification or removing promiscuous features (e.g., large flat aromatic surfaces) help reduce binding to unrelated proteins.
Genotoxicity and Structural Alerts Certain chemical groups raise red flags by default. Nitroaromatics, anilines, Michael acceptors, and epoxides are all associated with DNA damage and mutagenicity. These features are evaluated using the Ames test and in silico toxicophore screening. If an alert is flagged, the chemist’s next move is clear: modify, cap, or eliminate the liability.
Even metabolites can pose a genotoxic risk, so metabolic stability work dovetails with safety design. Avoiding bioactivation (conversion to reactive intermediates) is key, often requiring the use of bioisosteres or shielding groups.
Cardiotoxicity and hERG Inhibition The hERG potassium channel is notorious: blocking it can lead to QT prolongation and fatal arrhythmias. In early optimization, compounds are screened for hERG binding, often using patch-clamp assays or cell-based flux systems.
Structure drives risk. Highly lipophilic, basic, and aromatic compounds tend to bind hERG. Medicinal chemists mitigate this by lowering pKa, adding polar groups, or disrupting pi-stacking interactions. The goal is to reduce channel affinity while preserving on-target activity-a balancing act that often requires multiple design cycles.
Early Tox Screens and Therapeutic Index General cytotoxicity is also assessed in vitro, using human cell lines and assays for mitochondrial function or membrane integrity. In vivo, rodent studies evaluate tolerability, behavior (e.g., Irwin’s test), and early biomarkers like liver enzymes.
Throughout, the emphasis is on widening the therapeutic window: raising the exposure threshold for toxicity while lowering the dose needed for efficacy. That’s rarely achieved with a single modification; it requires multiparameter optimization across potency, ADME, and off-target risk.
While traditional lead optimization relies on expert intuition, iterative synthesis, and well-established rules of medicinal chemistry, it is ultimately constrained by human bandwidth and the complexity of multi-parameter trade-offs. Each design cycle can take weeks, and subtle patterns in structure-property relationships may go unnoticed. This is where AI steps in-not to replace the foundational methods, but to amplify them. By learning from historical data, predicting outcomes, and even proposing novel structures, AI transforms lead optimization from a largely manual craft into a more scalable, predictive, and hypothesis-generating process. What once required hundreds of compounds and dozens of analogs can now begin with a well-trained model and a few powerful prompts.
Recommended by LinkedIn
How AI Is Reshaping Lead Optimization
If traditional lead optimization is where chemistry meets judgment, then AI-driven lead optimization is where judgment meets scale. AI is not replacing the medicinal chemist; it is augmenting them with tools that can learn from millions of compounds, model complex relationships, and propose candidates with precision, speed, and novelty.
From Manual to Model-Driven Design
AI enables more efficient multiparameter optimization than ever before. Traditionally, improving a molecule’s potency, selectivity, and drug-like properties required laborious iterative chemistry and testing. Now, AI models can:
- Predict biological activity and ADMET properties (Absorption, Distribution, Metabolism, Excretion, Toxicity) in silico
- Design novel molecules using generative models trained on massive compound libraries
- Optimize lead series using closed-loop feedback from lab assays
Machine learning-based QSAR models like Chemprop from Charles McGill and collaborators make it easier to prioritize analogs based on potency and safety. Graph neural networks (GNNs) capture nuanced molecular features directly from chemical graphs. In frameworks like DeepChem from Bharath Ramsundar and his team, GNNs outperform classical fingerprints in many property prediction tasks.
The most efficient optimization strategy today uses these models to virtually assess compounds before they are synthesized, freeing up chemists to focus on the highest-value molecules.
Foundation Models, Reinforcement Learning, and Generative Design
Emerging foundation models such as MolE and ChemBERTa have been trained on hundreds of millions of compounds. MolE, a molecular transformer model, recently ranked first in 10 of 22 ADMET prediction tasks in the Therapeutic Data Commons benchmark. It learns directly from molecular graphs using a two-stage pretraining pipeline: self-supervised learning on over 800 million unlabeled compounds, followed by fine-tuning on 22 property prediction tasks.
Meanwhile, generative approaches like diffusion models and variational autoencoders (VAEs) are producing high-quality, structurally diverse molecules tailored to complex objectives. Diffusion models are particularly promising for 3D structure-based design—generating new compounds directly inside protein binding pockets.
Reinforcement learning tools such as REINVENT by AstraZeneca ( Atanas Patronov and collaborators) and and MolDQN (collaboration between Zhenpeng Zhou & Richard Zare from Stanford and Steven Kearnes , Li Li & Patrick Riley from Google Research Applied science) allow AI to learn how to make effective structural modifications, optimizing leads based on compound-specific reward functions. These approaches have been successfully used to generate novel kinase inhibitors and other drug-like scaffolds in timelines as short as a few weeks.
An especially inspiring example of new talent I came across while researching this issue: a recent paper by Akshat Santhana Gopalan , a high school researcher at the Rayan H. Assaad, Ph.D. Lab, applies Generative Flow Networks (GFlowNets) to optimize drug leads for Caco-2 cell permeability. The method uses a machine learning model to predict how well a molecule passes through Caco-2 cells—a standard in vitro assay for intestinal absorption—and guides the GFlowNet to make smart edits while preserving the molecule’s core structure. The result: diverse, valid analogs with improved predicted permeability. The first time I heard of the Caco assay was just last year. And now, high schoolers are out here using AI to tackle real-world ADMET bottlenecks. Big shout out.
From Code to Clinic: Real-World Progress
AI-powered lead optimization is now delivering clinical-stage candidates:
- Insilico Medicine ’s INS018_055, a novel fibrosis drug, reached Phase II in just 30 months—half the time of a traditional path.
- Schrödinger contributed to the design of MORF-057, an oral integrin inhibitor now in Phase II trials.
- GSK ’s abaucin, an antibiotic candidate against drug-resistant A. baumannii, emerged from an AI-led screen and optimization loop.
Tools in the Wild: How Industry Uses AI
Drug discovery teams now have access to a robust suite of AI-driven platforms:
- Exscientia (now part of Recursion) developed the Centaur Chemist platform to design compounds meeting multiple pharmacological criteria on accelerated timelines.
- Insilico Medicine integrates Chemistry42 (for de novo design) and PandaOmics (for target discovery) into a seamless discovery engine.
- Schrödinger combines ML-based property prediction with physics-based binding affinity tools for structure-guided design.
- BenevolentAI uses a biomedical knowledge graph to drive multi-parametric lead generation.
- Atomwise applies deep learning to predict binding affinity from 3D structures, tackling targets previously considered undruggable.
Infrastructure and Investment
Progress in AI-driven lead optimization is being propelled by significant infrastructure and funding:
- The ATOM Consortium developed an end-to-end multiparameter optimization pipeline using supercomputers.
- Open Targets and MELLODDY support data-sharing and federated learning to unlock insights across pharma datasets.
- Inductive Bio last week raised $25M (ed by Obvious Ventures with participation from a16z Bio + Health, Lux Capital, S32, Character Capital, Amino Collective, and leading angel investors) to expand its Compass platform, which predicts ADMET properties pre-synthesis. Their approach combining a precompetitive data consortium with powerful predictive models helps chemists focus only on the most promising leads.
The Road Ahead
AI is not a magic wand, but it is a force multiplier. By compressing timelines, exploring larger chemical spaces, and reducing false starts, AI is transforming lead optimization from a guessing game into a guided search.
As foundation models mature and experimental feedback loops tighten, we are approaching a future where designing an IND-ready compound may be as much a software challenge as a synthetic one. The next leap will come from integrating these tools into lab workflows, validating predictions, and co-developing hits with algorithms as active collaborators.
Tomorrow’s leads will not just be optimized, they will be co-created.
If you found this useful, consider subscribing and sharing it with a colleague who might, too: https://www.linkedin.com/build-relation/newsletter-follow?entityUrn=7312639268911226880