Artificial Intelligence for Early Drug Discovery Icon

Cambridge Healthtech Institute’s 4th Annual

Artificial Intelligence for Early Drug Discovery

Improving Speed and Precision for Drug Design, Lead Optimization and ADMET Predictions

April 19-20, 2022

 

The Artificial Intelligence for Early Drug Discovery conference brings together a diverse group of experts from chemistry, target discovery, pharmacology and bioinformatics, to talk about the increasing use of computational tools, artificial intelligence (AI) models, machine learning (ML) algorithms and data mining in preclinical drug development. The talks will highlight how AI/ML can help in drug design, target identification, lead optimization, PK/PD predictions, and early safety assessments. There will be discussions on the caveats and limitations of AI/ML-driven decision-making using relevant case studies and research findings.

Tuesday, April 19

7:00 am Registration Open and Morning Coffee (Sapphire West Foyer)

ROOM LOCATION: Sapphire P

ADVANCES IN AL/ML APPLICATIONS

8:00 am Welcome Remarks
8:05 am

Chairperson's Remarks

Ewa Lis, PhD, Founder & CTO, Koliber Biosciences
Alex Zhavoronkov, PhD, Founder & CEO, Insilico Medicine

Since the dawn of computing, medicinal and computational chemists attempted multi-parameter optimization of molecules utilizing a wide variety of methods. In this talk, we will present the history of generative chemistry primarily focusing on generative adversarial networks and reinforcement learning. We will review the evolution of these systems since the publication of the first peer-reviewed papers in 2016 to multiple experimental validation experiments and their applications in the recent past.

8:40 am

AI in Drug Discovery, From 2021 into 2022 – What Happened, Didn’t Happen, and What Still Needs to Happen?

Andreas Bender, PhD, Professor for Molecular Informatics and Group Leader for Data-Driven Drug Discovery, Department of Chemistry, University of Cambridge

In this contribution, we will review recent developments in the field of artificial intelligence in drug discovery, and discuss their likely impact on project outcomes in the near future. In particular, this will cover the aspect of model validation and the impact of 'AI' on the transition of compounds to the in vivo stages, which is their only raison d'être in the first place.

Elisabetta Micelotta, PhD, Science & Technology Team Leader, Nanoform

Poor solubility is arguably the biggest hurdle affecting drug development and the use of solubility enhancing technologies is commonplace throughout the industry.  The ability to predict a compound’s suitability for such technologies could be critical in drug discovery.  Nanoform’s Starmap2.0 uses sparse-data AI to augment experimental results from its CESS nanoparticle engineering process to predict the potential success of nanoforming drug molecules and enable the development of patient centric medications.

9:40 am Networking Coffee Break (Sapphire West Foyer)
10:05 am

Peptide Hit Identification and Lead Optimization Using Artificial Intelligence Approaches

Ewa Lis, PhD, Founder & CTO, Koliber Biosciences

Artificial Intelligence (AI) is becoming widely adopted for small molecule drug discovery, yet the methods for leveraging AI for peptide drug discovery are lagging far behind. These challenges are primarily driven by limited availability of peptide datasets, high dimensionality of the design space as well as poorly developed methods for encoding peptides for deep learning algorithms. In this presentation, we will discuss the advances that were made in developing feature encodings for peptides to enable development of high-performing models and efficient exploration of peptide design space. We will demonstrate how AI can be utilized to prioritize peptide variants for testing through in silico prediction of performance. Moreover, methods to explain the models and visualize feature importance will be presented. Lastly, results from wet-lab validation experiments will be presented in the areas of immunogenicity and antimicrobial peptide development.  

10:35 am

Discovering Leads with Defined Multi-Target Profile: Antivirals, Anti-Protozoan, and Anti-Cancer

Ruben Abagyan, PhD, Professor, Department of Molecular Biology, University of California, San Diego

A new approach that combines a heavy in silico front-end with the experimental back-end has emerged. The approach is based on full inclusion of quickly expanding structural and activity data incorporated in structural and AI in silico models. Examples of the application of this approach to finding new antiviral candidates, antiprotozoal candidates, neurological disorders, and aiming at new cancer targets, are presented.

11:05 am

Enterprise-Wide Predictive Model for Early Absorption, Distribution, Metabolism, and Excretion Properties

Renee DesJarlais, PhD, Scientific Director & Fellow, Janssen R&D LLC

Accurate prediction of absorption, distribution, metabolism, and excretion (ADME) properties can facilitate the identification of promising drug candidates. At Janssen we have developed and implemented generic Target Product Profile (gTPP) models, which predict 15 early ADME properties, at various thresholds and different species where applicable.  Logging of predictions allows monitoring of usage and assessment of impact on decision-making in chemistry.  Model development, performance and benefit will be discussed.

11:35 am Session Break
Paul Belcher, PhD, Product Strategy Manager, Biacore, Cytiva

Higher throughput optical biosensors have dramatically reduced the time to run label free binding assays, allowing the more routine analysis of increasing numbers of compounds.  But the innovation in analysis of data has not kept pace with instrument throughput creating a significant burden in time and expertise.  In this presentation we will present a new path that allows the efficient and robust analysis of large data sets from optical biosensors

12:25 pm Session Break

AL/ML PREDICTIONS FOR HIT TO LEAD IDENTIFICATION

1:10 pm

Chairperson's Remarks

Aaron Daugherty, PhD, Vice President, Discovery Science, Aria Pharmaceuticals
1:15 pm

Contrastive Learned Molecular Representations: Accessible and Accurate AI Methods

David Huang, CEO, Oloren AI
Raunak Chowdhuri, CTO, Oloren AI

Artificial intelligence models are all the hype, but its practical application is extremely complicated, because current methods are difficult to deploy given the amount of training data required, quick timelines, and practitioner expertise. We introduce an approach where AI is trained by contrastive learning to create a molecular representation which clearly identifies chemically and biologically significant differences between compounds. This representation can be easily used as traditional descriptors would be used, and we show their superiority over popular descriptors and fingerprints.

1:45 pm

Developing Generative Machine Learning Platforms for Drug Discovery

Sean Ekins, PhD, Founder & CEO, Collaborations Pharmaceuticals, Inc.

We have developed a suite of automated tools called MegaSyn (representing 3 components: a new hill-climb algorithm which makes use of SMILES-based RNN generative models, analog generation software, and retrosynthetic analysis coupled with fragment analysis to score molecules for their synthetic feasibility) for designing new molecules. We will describe how it might be used to optimize molecules or prioritize promising lead compounds using test case examples for drug discovery.

2:15 pm

DeepFrag: A Deep-Learning Method for Structure-Based Lead Optimization

Jacob Durrant, PhD, Assistant Professor, Department of Biological Sciences, University of Pittsburgh

Machine learning has been applied with great success to a variety of drug-design problems including binding-affinity prediction, virtual screening, and QSAR, but it is less often used for lead optimization. DeepFrag is a deep convolutional neural network that fills this gap. Given the structure of a protein/ligand complex, DeepFrag (1) predicts the structural properties of a suitable optimizing chemical moiety and (2) searches a database of known moieties to identify those with properties that most closely match the prediction. In this talk, I will discuss recent DeepFrag development and applications.

2:45 pm

Leveraging AI for Drug Discovery

Aaron Daugherty, PhD, Vice President, Discovery Science, Aria Pharmaceuticals

Aria Pharmaceutical’s unique AI-enabled drug discovery approach can save ~3 years from project initiation to in vivo results while generating a 30x hit rate at those milestones, compared to traditional methods. Aria’s AI platform builds novel pathogenesis models to identify molecules with unique first-in-class MOAs. Using their platform, Aria has developed a pipeline of treatments in 18+ programs across a range of therapeutic areas, including SLE, CKD, NASH, and IPF.

Marie-Aude Guié, Vice President, Scientific Computing, X-Chem Inc.

X-Chem’s DNA-Encoded Chemical Library (DEL) platform generates a large amount of binding data for each target it is applied to. In this presentation, we will discuss the application of predictive machine learning to DNA-Encoded Chemical Library technology towards the identification of inhibitors of ERα, one of two main types of receptors activated by estrogen. We will also introduce other applications of machine learning to DEL data currently being developed at X-Chem.

3:30 pm Refreshment Break in the Exhibit Hall with Poster Viewing (Sapphire Ballroom B-O)

PLENARY KEYNOTE LOCATION: Sapphire D

PLENARY KEYNOTE SESSION

4:30 pm Plenary Welcome Remarks from Lead Content Director with Poster Finalists Announced

Anjani Shah, PhD, Senior Conference Director, Cambridge Healthtech Institute

Jordan Kovacev, PhD, Executive Director - University of California Drug Discovery Consortium (UCDDC), Director of Business Development – California NanoSystems Institute at UCLA (CNSI), University of CA Drug Discovery Consortium (UCDDC)

The University of California Drug Discovery Consortium (UCDDC) strives to leverage the biomedical research and commercialization strengths of the University of California (UC) system to accelerate the development of life-saving therapies and to translate discoveries into knowledge driven commercial enterprises that stimulate California’s economy. The UCDDC Executive Committee works with each UC-campus to advance drug development by providing expertise in drug discovery and by building relationships between industry and academics.

4:55 pm

PLENARY: Using Cryo-EM to Explore the Allosteric Regulation of Molecular Glues

Gabriel Lander, PhD, Professor, Department of Integrative Structural and Computational Biology, Scripps Research Institute

Crystallographic studies previously defined the binding site of anti-cancer immunomodulatory imide drugs within Cereblon’s Thalidomide Binding Domain (TBD), but questions surrounding the allostery of drug-induced substrate-binding remain. We performed cryo-EM analyses of the complex in the presence or absence of drugs and substrates to show that association of an IMiD to the TBD is both necessary and sufficient for triggering an allosteric rearrangement from a basally “open” conformation of Cereblon to the canonical “closed” conformation.

5:40 pm Welcome Reception in the Exhibit Hall with Poster Viewing (Sapphire Ballroom B-O)
6:40 pm Close of Day

Wednesday, April 20

7:00 am Registration Open (Sapphire West Foyer)

ROOM LOCATION: Sapphire P

7:30 am Continental Breakfast Interactive Discussions

Interactive Discussions are informal, moderated discussions, allowing participants to exchange ideas and experiences and develop future collaborations around a focused topic. Each discussion will be led by a facilitator who keeps the discussion on track and the group engaged. To get the most out of this format, please come prepared to share examples from your work, be a part of a collective, problem-solving session, and participate in active idea sharing. Please visit the Interactive Discussion page on the conference website for a complete listing of topics and descriptions.

IN-PERSON INTERACTIVE DISCUSSION: Challenges with AI Adoption and Implementation for Drug Discovery

Ruben Abagyan, PhD, Professor, Department of Molecular Biology, University of California, San Diego
Sean Ekins, PhD, Founder & CEO, Collaborations Pharmaceuticals, Inc.
Renee DesJarlais, PhD, Scientific Director & Fellow, Janssen R&D LLC
  • Current trends for the application of AI towards preclinical drug discovery
  • Understanding the caveats of AI-driven predictions 
  • The challenge of continuous evolution of models in response to growth of big data, data types, and computational platforms  
  • What measures should be taken to invest in and effectively use AI at various stages of drug development?

UPDATES ON AI INITIATIVES

8:15 am

Chairperson's Remarks

Bryce Allen, PhD, Co-Founder & CEO, Differentiated Therapeutics
8:20 am

AI/ML Applications: Updates from the ATOM Consortium

Amanda Paulson, PhD, Specialist, Pharmaceutical Chemistry, University of California, San Francisco

The ATOM Consortium aims to accelerate and democratize drug discovery by partnering an open-source molecular design platform with active learning to experimentally validate candidate molecules. We apply this platform to design brain penetrant small molecule inhibitors. Property prediction models are built for blood-brain barrier permeability and efflux. These models are integrated into our generative molecular design software to propose novel molecular entities that achieve our desired design criteria.

8:50 am

CACHE: An Open-Science Competition to Define the State-of-the-Art in Computational Prospective Hit Finding

Matthieu Schapira, PhD, Principal Investigator, Structural Genomics Consortium

The Critical Assessment of Computational Hit-finding Experiment (CACHE), is a new competition to guide the development of virtual screening methods to predict bioactive small molecules. Compounds predicted by participants for pre-selected protein targets will be purchased and tested experimentally at the CACHE experimental hub. All data will be publicly released. Participants will have a choice to remain anonymous. Computational methods will be explained but algorithms may remain proprietary. A new challenge/target will start every 3 months. 

John Wai, Vice President, WuXi AppTec, WuXi AppTec

Accelerating the synthesis of newly designed molecules means investing only in synthetic sequences that are more likely to work. Structural diversifications, different substitution patterns, unique heterocyclic systems and arrays of functionalities can lead to unexpected reactivity. Integrating Machine Learning retrosynthetic tools with quantum mechanics calculations for prospective analyses significantly improve success rate in our MedChem syntheses.

Quentin Perron, PhD, Chief Scientific Officier & Co-founder, Iktos

Multi-parameter optimization (MPO) is a major challenge confronting the drug discovery process, making it difficult to identify promising synthetically accessible molecules meeting the TPP. To overcome this, we have developed an AI-driven pipeline (Makya and Spaya) for generative de novo drug design to enhance chemical space exploration while tackling the MPO challenge under synthetic constraints. We will discuss the implementation and impact of this pipeline to real-life drug discovery projects.

10:05 am Coffee Break in the Exhibit Hall with Poster Awards Announced (Sapphire Ballroom B-O)

AI/ML MODELING FOR PROTEIN DEGRADATION

10:30 am

Atomic-Resolution Prediction of Degrader-Mediated Ternary Complex Structures by Combining Molecular Simulations with HDX-MS

Woody Sherman, PhD, Chief Computational Scientist, Roivant Sciences

We describe a novel method that combines experimental biophysical data (HDX-MS) with weighted ensemble simulations (WES) to accurately predict ternary complex structures at atomic resolution. We show that the WES+HDX approach generates accurate structures (RMSD below 2.0 Å to x-ray) and can reproduce solution-state dynamic behavior of the ternary complex. We also show how we are extending this approach to predict degradation propensity of different heterobifunctional and glue molecules.

11:00 am

Assessing Ternary Complex Formation for Protein Degradation

Victor Guallar, PhD, Professor, Barcelona Supercomputing Center and Nostrum Biodiscovery

The potential of providing realistic protein-protein interaction models, and of their assembly with a third (smaller) molecule, such as PROTACs or molecular glue, is, no doubt, a very hot topic. Current molecular modeling approaches have too much bias (and correlation) with the linker degrees of freedom when modeling a PROTAC. At the same time, the lack of large amounts of data severely hinders the application of machine learning techniques. In this talk we will showcase the development of our mixed models, taking advantage of molecular modeling, a consensus bioinformatics approach, and machine learning techniques using data augmentation from modeling. Such a combined approach is capable of enhancing the degrader selection, providing accurate structural interaction models, and screening hundreds of ternary complexes formations.

11:30 am

Differentiable Portfolio Planning: A Graph-Based Recommendation Framework Applied to Targeted Protein Degradation

Bryce Allen, PhD, Co-Founder & CEO, Differentiated Therapeutics

Targeted Protein Degradation (TPD) is complex and only partially understood. In contrast to orthosteric or allosteric target inhibition, TPD approaches have emerged as novel therapeutic modalities that promote distinct pharmacological profiles and enable modulation of previously undruggable targets. To systematically assess which potential drug targets might be most amenable to TPD therapeutic development, we present a deep learning approach based on a differentiable heterogeneous biomedical knowledge graph built to support data-driven decision-making for TPD portfolio planning and prioritization. 

12:00 pm Close of Artificial Intelligence for Early Drug Discovery Conference