Artificial Intelligence for Early Drug Discovery Part 2 Icon

Cambridge Healthtech Institute’s 5th Annual

Artificial Intelligence for Early Drug Discovery - Part 2

Innovative Use of Computational Tools for Finding New Targets and Drugs

APRIL 12 - 13, 2023

 

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 the opportunities and limitations of AI/ML-driven decision-making, using relevant case studies and research findings. The first part of the conference will focus on how AI/ML can help in drug design, hit identification, lead optimization, PK/PD predictions, and early safety assessments. The second part will focus on emerging computational tools and models to identify new drug targets, prioritize compounds for development and to help drive niche applications, such as predictions for targeted protein degradation.

Wednesday, April 12

Registration Open (Indigo West Foyer)12:00 pm

Dessert Break in the Exhibit Hall with Poster Viewing (Indigo A-G)12:45 pm

Welcome Remarks1:30 pm

ROOM LOCATION: Indigo 204

AI FOR SCREENING & DEGRADATION

1:35 pm

Chairperson's Remarks

Scott Bembenek, PhD, Founder, CEO & CSO, Denovicon Therapeutics

1:40 pm

Differentiable Design: Dynamic Ternary Complex Structure Prediction with Multiscale Generative Diffusion Models

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

Designing bioactive molecules that serve a purpose – such as binding specifically to a protein – is central to medicinal chemistry and a common practice in drug discovery. Although automating design has tremendous promise, general-purpose methods do not yet exist. Here we explore a simple, fast, and robust approach to inverse design which combines learned forward simulators based on graph neural networks with gradient-based design optimization. Our approach solves high-dimensional problems with complex physical dynamics, including designing molecules that induce proximity between proteins forming a ternary complex.

2:10 pm

In silico Screening for Heterobifunctional Small Molecule (hSM) Linkers

Shu-Ching Ou, PhD, Scientist, Center for Research Acceleration by Digital Innovation, Amgen, Inc.

With more Heterobifunctional Small Molecules (hSM) being developed, the importance of linker design and optimization has been broadly recognized. Here we report an approach to virtually screen the linker designs by proactively using the available hSM ternary structures. Combined with expansive chemical building blocks, this approach helps us rapidly triage design ideas and expedite the discovery process.

2:40 pm Synthesizability Orient Mega-size Interactive Chemical Space (SyOMics)

Liu Liu, Dr., Vice President, Drug Discovery Business Unit, PharmaBlock

Hits identified from recent AIDD campaigns are often lacking structural novelty, particularly synthesizability, with existing chemical spaces. In PharmaBlock, taking advantage of our 160k novel building block inventory and proved synthesis route collections, we established a megasize (1014) interactive virtual chemical space. Supported by our proprietary searching algorithm, we are able to generate structurally diverse hits, which are ready to be synthesized with our building blocks in stock.

Refreshment Break in the Exhibit Hall with Poster Viewing (Indigo A-G)3:10 pm

4:00 pm

Machine Learning-based Identification of Targets in Cancer for Protein Degradation

Collin Tokheim, PhD, Senior Scientist, Early Oncology Data Science, AstraZeneca Pharmaceuticals

While Targeted Protein Degradation (TPD) can expand druggable targets, how protein degradation is dysregulated in cancer and how TPD drugs counteract this effect is incompletely understood. First, we developed a deep-learning model (deepDegron) to identify mutations that result in loss of protein degradation signals. Second, we developed a machine learning model (MAPD) to predict which protein targets are likely degradable by TPD compounds from unbiased proteomic experiments of the kinome.

4:30 pm

Enhanced Active Learning by Combining Machine Learning and Structure-Based Methods

Scott Bembenek, PhD, Founder, CEO & CSO, Denovicon Therapeutics

We will discuss an approach that applies AI and ML to design, test, and optimize lead molecules rapidly in silico and to suggest what compounds to synthesize and screen next in an ‘active learning’ process. A comprehensive platform approach offers tight integration between the virtual and real cycles (V+R). 3D modeling and simulation methods enhance the accuracy of predictions for drug potency, efficacy, and selectivity, while also addressing multi-target effects.

In-Person Group Discussions (All Session Rooms)5:00 pm

In-Person Group 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 In-Person Group Discussions page on the conference website for a complete listing of topics and descriptions.

IN-PERSON GROUP DISCUSSION:

Emerging Applications of AI/ML Tools in Drug Discovery

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

Collin Tokheim, PhD, Senior Scientist, Early Oncology Data Science, AstraZeneca Pharmaceuticals

  • Designing molecules that induce proximity between proteins 
  • Integrating fragment-based screening with molecular dynamics
  • Virtually screening linker designs using available PROTAC ternary structures
  • Machine learning to predict which protein targets are likely degradable
  • AI and ML models to design, test, and optimize lead molecules rapidly in silico ​

Close of Day5:45 pm

Dinner Short Courses*6:15 pm

*Premium Pricing or separate registration required. See Short Courses page for details.

Thursday, April 13

Registration Open7:15 am

Diversity in Chemistry Breakfast Discussion (Indigo 202)7:45 am

IN-PERSON GROUP DISCUSSION:

Diversity in Chemistry beyond Molecules: Gender and More

Michelle Arkin, PhD, Chair and Distinguished Professor, Pharmaceutical Chemistry & Director, Small Molecule Discovery Center, University of California, San Francisco

Thomas P. Garner, PhD, Principal Scientist, Biophysics, Genentech, Inc.

Justyna Sikorska, PhD, Associate Principal Scientist, Mass Spectrometry & Biophysics, Merck

We encourage all to attend this moderated, audience-interactive discussion session. When it comes to increasing diversity among scientists, there continues to be a drop-off as one moves higher in leadership. Where do systemic challenges remain, what is your experience, and how can we continue to equalize the system?   

Topics may include below, but will be guided by audience input: 

  • Where does the 'drop-off' of women in the chemistry career progression pipeline occur and why? 
  • How did the pandemic and other sea changes in the past three years bring us closer to or further from equality? 
  • What issues arose that you thought were solved? 
  • Diversity in life paths should include us all – how are men and nonbinary scientists being included? 
  • Intersectionality and equality – what is the experience of women of color, first-generation women scientists, and others?​

ROOM LOCATION: Indigo D+H

PLENARY KEYNOTE SESSION

8:30 am

Plenary Welcome Remarks from Lead Content Director with Poster Finalists Announced

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

8:35 am

PLENARY: Reflections on a Career as a Medicinal Chemist in Drug Discovery

Nicholas A. Meanwell, PhD, Vice President (recently retired), Small Molecule Drug Discovery, Bristol Myers Squibb Co.

A successful drug candidate depends on many factors: creativity of scientists involved, effective collaboration and commitment by the team, and the quality of the compound advanced. I reflect on a 40-year career pursuing the discovery of drug candidates designed to address unmet medical need in the cardiovascular, CNS, and viral diseases therapeutic areas and share undervalued strategies and other synthetic chemistry approaches for overcoming specific medicinal chemistry challenges.

Coffee Break in the Exhibit Hall with Poster Viewing9:30 am

ROOM LOCATION: Indigo 204

EMERGING AI APPLICATIONS

10:20 am

Chairperson's Remarks

Ewa Lis, PhD, Founder & CTO, Koliber Biosciences

10:25 am

Improving Machine Learning Predictions: Focus Is All You Need

Anton Filikov, PhD, Associate Director, Computational Drug Discovery, Arrakis Therapeutics

The mainstream view in improving model predictions is identifying better modeling algorithms and assembling larger training sets curated to get more reliable data. In addition to these approaches we use other measures that are very effective: (1) eliminating data points that confuse the model algorithm, (2) biasing property distributions in training set with the goal to maximize performance metrics. Examples will be given with demonstrated material improvements.

10:55 am

Fingerprinting Drug Effects and Disease Phenotypes of the CNS Using Deep Functional Readouts of Human iPSC-Derived Neurons

Benjamin Harwood, PhD, Senior Scientist, Q State Biosciences

We built a map of the electrophysiology of human excitatory neurons, with applications to high-throughput screening, drug repurposing, and target deconvolution. First, we measured hundreds of parameters from millions of iPSC-derived neurons perturbed by tool compounds, yielding a database of activity profiles. Then, we used representation learning to distill these data down to concise fingerprints, where similar compounds form neighborhoods and several important tasks (e.g. drug repurposing, target deconvolution) reduce to path-matching problems.  

11:25 am Pragmatic Drug Discovery Acceleration in Place of Dogmatic Automation

Nathan Allen, PhD, Research Fellow and Director of Business Development, Automated Synthetic Chemistry, XtalPi Inc.

Medicinal chemistry synthesis for drug discovery is an expensive and laborious aspect of drug development, often requiring years of effort by skilled chemists.  XtalPi is developing a custom robotics and human chemist platform.  The goal-deliver productivity and reduce downtime in the development cycle.  Real-world complex molecule applications will be presented to demonstrate the progress toward this goal.

11:40 am

Protein Flexibility and Binding Affinity: What Can AI Predict?

Ponni Rajagopal, PhD, Founder & President, NstructuredesignS, LLC

Accurately predicted binding affinities can be a powerful tool for virtual screening of ligands. Protein flexibility plays an important role in ligand binding. Hence, using parameters that determine protein flexibility will lead to better prediction of binding affinities in ML algorithms. This work will evaluate the use of such independent variables to determine binding affinities.

12:10 pm

ASPIRE (A Specialized Platform for Innovative Research Exploration): Lowering the Barrier to Drug Development by Applying Automation, Data Analytics, and AI/Machine Learning to Chemistry and Biology

Sean Gardner, MS, Scientific Program Manager, Office of Special Initiatives, NCATS, National Institutes of Health

The gap between drug discovery and information science continues to close, hence there has never been a better time to leverage the power of AI/ML techniques to advance our understanding of the relationships between chemical and biological space. NCATS has identified, through the input of the greater scientific community, focus areas that need to be addressed in order to transform the design-synthesize-test cycle to transition to be more data-driven. The ASPIRE Program was created to support the development of AI/ML tools to process captured data to inform the next iteration of the process.

Enjoy Lunch on Your Own12:50 pm

Refreshment Break in the Exhibit Hall with Poster Awards Announced1:20 pm

Poster Award (Sponsorship Opportunity Available)

AI FOR HIT IDENTIFICATION

2:00 pm

Chairperson's Remarks

Tonglei Li, PhD, Allen Chao Chair & Professor, Industrial & Physical Pharmacy, Purdue University

2:05 pm

Manifold Embedding of Molecular Surface: A New Dimension in Chemical Deep Learning

Tonglei Li, PhD, Allen Chao Chair & Professor, Industrial & Physical Pharmacy, Purdue University

To effectively differentiate huge quantities of molecules in data-driven drug research, we have developed a low-dimensional representation of molecules and utilize it in deep learning for predicting molecular properties and designing new drugs. The concept centers on transforming a molecule's 3D electronic attributes of local hardness and softness to lower-dimensional manifold embeddings. The representation carries the inherent information of intermolecular interaction strength and specificity. Both predictive and generative deep learning models have been developed out of the concept of data-driven drug research.

2:35 pm

The CACHE Computational Hit-Finding Competition: Lessons Learned So Far

Matthieu Schapira, PhD, Principal Investigator, Structural Genomics Consortium

CACHE is a benchmarking exercise modeled after CASP where every four months, computational chemistry and AI experts predict up to 100 compounds for a predefined protein target. Hit candidates are then procured, tested experimentally at CACHE, and all data and a generic description of the methods are released publicly. The emerging landscape of the most successful computational hit-finding approaches so far will be outlined.

3:05 pm

A Unified System for Molecular Property Predictions

David Huang, CEO, Oloren AI

There is no unified API for molecular property predictors (MPPs), which makes it difficult to share, distribute, version, retrain and manage predictors. We present Oloren ChemEngine (OCE), an open-source Python library with a unified, reproducible, and easy-to-integrate API for MPPs. Using OCE, we create models with the best leaderboard performances on 19 ADME/Tox benchmarks with MPP ensembling strategies. Using such API, we integrate model-agnostic uncertainty quantification and interpretability methods.

Networking Refreshment Break3:35 pm

AI FOR PROTEIN THERAPEUTICS

3:50 pm

Chairperson's Remarks

Ewa Lis, PhD, Founder & CTO, Koliber Biosciences

3:55 pm

FEATURED PRESENTATION: Protein Design Using Deep Learning 

David A. Baker, PhD, Henrietta & Aubrey David Endowed Professor, Biochemistry, University of Washington

Proteins mediate the critical processes of life and beautifully solve the challenges faced during the evolution of modern organisms. Our goal is to design a new generation of proteins that address current-day problems not faced during evolution. In this talk, I will describe recent advances in protein design using both traditional physics-based approaches as well as deep learning methods to design sequences predicted to fold into desired structures.

4:25 pm

Designing Therapeutic Antibodies with Synthetic Biology and Machine Learning

Peyton Greenside, PhD, Co-Founder & CSO, BigHat Biosciences

BigHat Biosciences is designing safer, more effective antibody therapies for patients using machine learning and synthetic biology. Machine learning guides the search for better molecules by directing and learning from each cycle of our high-speed, automated wet lab that synthesizes and characterizes hundreds of antibodies each week. We’ll highlight key features of our platform and share several case studies of protein engineering using this novel platform.

4:55 pm

Peptide Discovery and Optimization Using Artificial Intelligence Approaches

Ewa Lis, PhD, Founder & CTO, Koliber Biosciences

Successful peptide drug discovery programs today require attainment of multiple performance metrics to progress a compound to clinical stage. To aid decision-making, Koliber has developed an AI peptide platform based on state-of-the-art machine learning methods to analyze peptide properties, profile positions, and predict new variants. The capabilities and wet-lab validation of the AI platform will be demonstrated with examples from immunology and antimicrobial peptide discovery and optimization.

Close of Conference5:25 pm