Science Pool

Advances in Cardiotoxicity Prediction using Transcriptomics and Machine Learning

Posted by Evotec on Apr 8, 2024 12:26:35 PM

Cardiotoxicity is one of the leading causes of drug attrition. To address this, there is a need for improved predictive screens which can be applied at an early stage in drug development to ensure only safe compounds progress to the clinic.

Drug-induced cardiotoxicity can manifest itself via various different mechanisms which makes detection challenging. Either functional or structural changes can occur, and these effects can be direct or indirect. Functional cardiotoxicity results from acute alteration in the heart function often as a consequence of electrophysiological effects. Structural cardiotoxicity is associated with alterations in cell and tissue morphology and can manifest itself as cell death, inflammatory changes and fibrosis. These morphological changes can take time to present themselves clinically and, therefore, sensitive techniques which can detect early molecular changes are valuable from a predictive perspective.

Combining transcriptomics with artificial intelligence is showing great potential in providing this transformative improvement in predictive safety assessment. In fact, the value of this approach in drug-induced liver injury (DILI) has been demonstrated in a recent poster presented by Cyprotex. At SOT in Salt Lake City from 10-14 March 2024, Cyprotex showcased how this technique can also be applied to cardiotoxicity. The research evaluated 42 compounds (33 cardiotoxicants and 9 non-cardiotoxicants) used in a variety of therapeutic indications. The compounds were assessed at 2 time points and 8 concentrations in a beating cardiac organ model using human iPSC-derived cardiomyocytes. At the end of the incubation period, three different analytical methods were compared; high content screening (HCS) (cell count, cellular ATP, mitochondrial mass, mitochondrial membrane potential, cellular calcium levels, DNA structure and nuclear size), calcium transience (wave amplitude, frequency, full peak width and full decay time) and transcriptomics (high-throughput RNA sequencing).

The HCS and calcium transience assays were valuable in detecting structural and functional cardiotoxicants, respectively. However, the synergism of combining these assays with transcriptomics was demonstrated by an overall improved cardiotoxicity risk prediction. Additionally, transcriptomics analysis provided detailed mechanistic information and identified specific pathway responses. Combined, the three approaches (HCS, calcium transience and transcriptomics analysis) gave excellent cardiotoxicity prediction metrics of 100% specificity, 82% sensitivity and 86% accuracy at 10x Cmax and 89% specificity, 91% sensitivity and 90% accuracy at 25x Cmax. The transcriptomics analysis improved the overall sensitivity by identifying various molecular mechanisms of structural toxicity such as alterations in cardiac pathways, genotoxicity, ER stress and mitochondrial toxicity.

It is not just cardiomyocytes which can be affected by drug induced toxicity, non-cardiomyocytes such as fibroblasts and endothelial cells may also be impacted. Organotypic models developed using different cell types, therefore, are likely to be more representative both structurally and functionally. In the future, Cyprotex is extending its research to evaluate a number of different cell-based models and different organ-specific toxicities using the transcriptomics approach. Our cardiac safety database is also growing and we now have identified approximately 140 compounds for testing in our models. The use of transcriptomics and artificial intelligence is accelerating the development of new cell-based models in the field of drug-induced toxicity leading to a new paradigm in in vitro safety testing.

Interested in learning more?

CONTACT US to discuss your project

READ OUR PUBLICATION

Tags: Blog, Toxicology & Safety, Modelling and Simulation

Toxicogenomics and AI: A Breakthrough in DILI Prediction

Posted by Evotec on Mar 27, 2024 1:18:44 PM

Predicting DILI (drug-induced liver injury) is challenging compared to other organ-specific toxicities. Translation from animals to humans is poor and, mechanistically, DILI can be complex. As a consequence, DILI continues to be one of the leading causes of attrition during drug development. Better human relevant models are required to improve early stage DILI prediction. Cyprotex is committed to researching and developing approaches to improve the prediction of DILI using human cell-based models in combination with novel techniques such as toxicogenomics and artificial intelligence (AI).

At the Society of Toxicology (SOT) conference on March 10-14, 2024, Cyprotex presented a poster titled, ‘An AI Approach to Drug-Induced Liver Injury Risk: Prediction of Safe Maximum Doses from Toxicogenomics Profiles’. The research evaluated 128 compounds from the FDA Liver Toxicity Knowledge Base – 68 of these compounds were associated with DILI and 60 of these compounds were not associated with DILI. Transcriptomics profiles were generated after dosing primary human hepatocytes in triplicate at 8 concentrations over 24 hr.

Machine learning is a subset of artificial intelligence which is used to find patterns, make decisions and optimise outcomes. In this study, the high throughput transcriptomics profiles of a set of known DILI-positive and DILI-negative compounds were used to train a supervised machine learning model to predict a safe maximum Cmax for novel compounds. When interpreting the results, a compound was predicted as DILI-positive if the true Cmax was above the predicted safe Cmax, and a compound was predicted as DILI-negative if the true Cmax was below the predicted safe Cmax. The model achieved the following metrics on the test set (assuming 40x Cmax level and 90% DILI score threshold):

The poster provides a detailed insight into two DILI-positive (TAK-875 and bosentan) and two DILI-negative compounds (dopamine and caffeine) to demonstrate the power of the transcriptomics and AI in predicting DILI as well as identifying specific mechanisms of toxicity. The model was able to capture the importance of cholestasis-associated genes in DILI.

To learn more about the use of transcriptomics and AI in DILI prediction:

READ THE POSTER

Tags: Blog, Toxicology & Safety, Modelling and Simulation

Toxicogenomics and AI: A Breakthrough in DILI Prediction

Posted by Evotec on Mar 27, 2024 12:49:09 PM

Prediction of drug-induced liver injury (DILI) is challenging. Translation from animals to humans is poor and manifestation of DILI can be complex mechanistically. 

In this poster, we focus on:

  • transcriptomics analysis of 128 compounds form the FDA Liver Toxicity Knowledge Base (68 associated with DILI and 60 not associated with DILI)
  • the use of machine learning in accurately predicting DILI and providing an insight into the mechanism of toxicity

Read our poster to learn more about our research!

LEARN MORE

 

Tags: Posters, Toxicology & Safety, Modelling and Simulation

PanHunter Multi-Omics Data Analysis DDup #13

Posted by Evotec on Nov 3, 2022 10:31:09 AM

The 13th edition of our Drug Discovery Update (DDup) provides insights into our unique PanHunter next generation multi-omics data analysis platform.

In this edition, it covers:

  • an introduction to PanHunter and multi-omics data analysis including:
    • the importance of multi-omics data (genomics, transcriptomics, proteomics and metabolomics) in understanding the complexity of disease and treatments
    • how the versatile and interactive multi-omics analysis platform, PanHunter, can greatly simplify the data analysis and interpretation process and so reduce time, improve visualisation and contextualisation, and inform decisions
    • how PanHunter can access additional meta information, reference data, chemical and structural information and clinical data to  provide further relevance and perspective
  • an interview with Dr John Szilagyi at Bristol Myers Squibb on how he uses PanHunter and the benefits it brings
  • a case study on how PanHunter has been applied in rapid candidate biomarker discovery
  • user perspectives from different functions within Evotec (wet lab biologists, computational biologists and bioinformaticians) on how PanHunter is making an impact on their research

 

LEARN MORE

Tags: Articles & Whitepapers, Proteomics, Metabolomics & Biomarkers, Modelling and Simulation

Functional Genomics for Infectious Disease

Posted by Evotec on Oct 27, 2022 10:18:30 AM

Bioinformatics supporting Infectious Disease: Creating knowledge for efficient discovery and development of better medicines.​

Pathogen functional genomics analyses for unbiased, multidimensional small molecule effect characterisation (RNA-, DNA-seq analyses to characterise pathogen genome and transcriptome)

Genome assembly and annotation with a focus on resistance and virulence (Illumina, ONT, hybrid)

  • Short- (SNP, short indels) and long-range (insertion, deletion, inversion, translocation, duplication) variant analysis associated with resistance (Illumina, ONT)
  • Bacterial genome wide association study (GWAS)
  • Transcriptome analysis (RNAseq) and functional interpretation
  • Bacterial population sequencing and variant characterisation (PoolSeq)
  • Molecular phylogeny
  • Network analysis (enrichment analysis, pathway mapping)


DOWNLOAD

 

Tags: Fact Sheets, Anti-Infectives, Modelling and Simulation

Transcriptomics Brings New Era of Toxicology Prediction

Posted by Evotec on Jul 22, 2022 11:57:59 AM

Featured in Nature, this article interviews experts, Paul Walker, Rüdiger Fritsch, and Carla Tameling, and provides the latest insights into transcriptomics and how the technology is transforming toxicology prediction.

It includes:

  • an overview of the current challenges in predicting organ-specific toxicity especially DILI
  • the advantages of using human cell-based models with a particular focus on 3D organoid models
  • a background to transcriptomics and how advances in throughput have widened its application 
  • how combining transcriptomics with machine learning and artificial intelligence has increased the power of this technology in toxicology prediction

LEARN MORE

Tags: Articles & Whitepapers, Toxicology & Safety, Modelling and Simulation

Improving Drug Safety with Transcriptomics

Posted by Evotec on Jul 19, 2022 5:09:05 PM

According to a study from 2020, a total of 133 drugs were withdrawn from the market due to safety reasons between 1990 and 2010. Major causes were hepatotoxicity (27.1%), cardiac disorders (18.8%), hypersensitivity (12.8%), and nephrotoxicity (9.8%), accounting for 69.2% of all drugs withdrawn. In most cases, these withdrawals were initiated because of spontaneous reports and/or case reports. Another study looking into drug withdrawals between 1953 and 2013 revealed that 18% of drug withdrawals from the market in this period were due to liver damage.

Add to these withdrawals of marketed drugs the attrition rate of drug candidates in clinical trials: 90 percent of all drug candidates fail in clinical trials, and 30 percent of these failures are due to unmanageable toxicity issues.

These failures occur despite thorough preclinical work and intensive animal studies. It is estimated that only 50% of the compounds that cause liver toxicity in humans are detected by animal studies. Furthermore, some adverse reactions or idiosyncratic toxic effects are typically not detected until the drug in question has gained large exposure in a broad patient population.

Interestingly, a study evaluating the attrition of drug candidates from AstraZeneca, Eli Lilly and Company, GlaxoSmithKline and Pfizer came to the conclusion that there is a strong link between physicochemical properties of compounds and clinical failure due to safety issues. The results also suggest that further control of physicochemical properties is unlikely to have a significant effect on attrition rates and that additional work is required to address safety-related failures.

These failures are not only costly (according to the FDA, drug development takes over 10–15 years with an average cost of over $1–2 billion for each new drug to be approved), but are also putting the health and the life of patients in danger.

Consequently, Cyprotex and its parent company Evotec are very focused on assessing toxicology issues from the very beginning of its drug R&D process and have invested a significant amount of time and resources to expand its technologies for the toxicological evaluation of drug candidates.

“The idea is to make better informed decisions earlier in your discovery campaign when you can select potentially safer compounds, rather than finding a safety liability later on,” says Paul Walker PhD, Vice President, Head of Toxicology at Cyprotex, in Cheshire, UK.

This improved discovery and selection is implemented by Cyprotex by using the unbiased view of transcriptomics and its potential to predict drug-induced toxicity. Transcriptomics involves sequencing thousands of mRNA molecules to identify which processes are active in the cell and allows for a better understanding of the cell’s reaction to known and novel drugs.

This is by no means a purely academic endeavour. As an example, the Cyprotex team demonstrated via transcriptomics it was able to identify problems in liver cells treated with fasiglifam, a promising diabetes drug candidate, which was withdrawn from late-stage clinical trials by its developer, following signs of liver damage in trial participants. This example proves that transcriptomics could have raised a red flag during preclinical development and might have saved hundreds of millions of dollars.

“Our studies have found potential effects on mitochondrial function, which were previously missed in preclinical studies” says Walker.

Therefore, transcriptomics has the potential to supplement or reduce in vivo toxicology studies by effectively identifying safety issues early in drug development, saving time and money — and animal testing.

Sophisticated Human Cell-Based Models

A key advantage of transcriptomics is its use of human cells and Evotec as well as Cyprotex are not just looking at 2D cell cultures, but investigating 3D organoids. These structures formed of thousands of cells that mimic organ-specific tissues are much closer to the real organ and have valuable features: For example, 3D-organoids of the heart exhibit regular contractions, beating like a living heart, and liver organoids secrete typical liver enzymes for days.

“On top of that, a 3D system allows repeat dosing, mimicking dosing regimens in vivo and potentially helps to detect effects due to toxic metabolites,” says Walker.

As they are small, the organoids can be placed in 384-well plates and individually molecular barcoded for simultaneous sequencing. This combination of miniaturization and high-throughput screening is implemented in Evotec’s EVOpanOmics platform and allows a wider adoption of transcriptomics in preclinical toxicology studies allowing for the repeat testing of dozens or even hundreds of compounds at several doses and in multiple organs.

“People have thought about using transcriptomics for toxicology before, but it was always a numbers game,” explains Rüdiger Fritsch PhD, Principal Scientist and Project Lead for EVOpanOmics. “For any compound that’s a real troublemaker, the evidence will show up in the transcriptomics data if you profile it in a relevant model. You just need to test appropriate dosing scenarios with the breadth of genome-wide off-target effects so that you have a chance to find it.”

Complex Analysis of Transcriptomics Data

Evotec, in conjunction with Cyprotex, offers transcriptomics services to drug developers and carries out the entire process in-house, from growing the organoids to sequencing and analysis. This streamlined process allows its researchers to screen hundreds of compounds a day, each delivering tens of thousands of data points on RNA levels. To analyze all of these vast amounts of data, Evotec has developed a software platform called EVOpanHunter that allows among others the analysis of these transcriptomics in an interactive manner.

“We want to democratize data analysis for the biologists who know the biological pathways and processes, without them needing to rely on additional experts from the bioinformatics department for routine tasks,” says Carla Tameling PhD, Head of Sales and Application for EVOpanHunter at Evotec.

On top of the interactive multi-omics analysis platform machine learning is used to trawl through this immense amount of data in order to find specific patterns hinting for toxicological effects and alert the researchers to dig deeper. “The more data we get, the harder it is for a human to dig through it all,” adds Tameling. “Transcriptomics is an unbiased view. You don’t need to define what to look at prior to your studies — you get all the data, and you might see things that you didn’t think would be relevant initially.”

From publically available sources, Cyprotex has compiled a broad and highly valuable transcriptomics reference database for drug-induced liver injuries.. Machine learning is being applied to predict whether a compound is likely to have issues by comparing the observed pattern of gene activity to the activity patterns of known toxic molecules. Furthermore, this is not restricted to hepotoxicity. Cyprotex is already building databases of other organs, such as heart, kidney and brain, using publicly available drug development trial results to select a broad space of reference comounds. “We’re running reference compounds from all kinds of sources where we know there are either late-stage clinical findings or withdrawals from the market,” states Walker.

Given the rapid advancements of the technology, it may be only a matter of time before transcriptomics and other omics technologies become a regulatory standard approach for preclinical toxicity testing.

Contact Us for More Information

Learn More

Tags: Blog, In vitro Biology, Toxicology & Safety, Modelling and Simulation

Targeted Protein Degradation as a Promising Tool in Drug Discovery

Posted by Evotec on Jun 21, 2022 3:13:45 PM

How to knock down proteins driving disease processes in a cell

Many diseases are caused by the overproduction of certain proteins. The traditional approach to interfere with these proteins is based on small molecules or antibodies blocking these proteins or their corresponding target, e.g., receptors. Thanks to the recent progress of nucleic acid research, there are several new approaches today which intervene at different stages, from gene regulation to transcription to translation: CRISPR-Cas9 methods to target the DNA, zinc finger repressors targeting gene transcription, or RNA-molecules (antisense oligonucleotides, RNA interference, micro RNAs, etc.) to inactivate the mRNA or to suppress the translation. All approaches come with advantages and disadvantages. The main problem with DNA- and RNA-based medicine is delivery, followed by off-target-effects.

There are, however other new knockdown strategies as well, e.g., enhancing the protein clearance pathways to speed up the degradation of unwanted proteins, such as the autophagy-lysosome pathway and the ubiquitin-proteasome system (UPS).

Transcriptomics, data analysis, and AI/ML platforms as basis for partnership with BMS in targeted protein degradation

Since 2018, the latter technology of targeted protein degradation is also being used in a cooperation with Bristol Myers Squibb (BMS) to identify first-in-class drug candidates in oncology to treat solid tumors. For this collaboration, Evotec uses its PanOmics platform, EVOpanOmics, which combines enhanced throughput proteomics, high-throughput transcriptomics, and cell imaging with the integrated data analysis platform EVOpanHunter and Evotec’s AI/ML-based drug discovery and development platforms.

This research has led to the discovery of novel first-in-class molecular glue degraders. These small, drug-like compounds induce interactions between an E3 ubiquitin ligase and a molecular target, leading to ubiquitination and subsequent degradation of the recruited protein. The resulting therapeutic effect is long-lasting as the molecular glue degraders themselves are not degraded in the process and can initiate the degradation process through several iterations. BMS is a leader in this field based in particular on its unique library of cereblon E3 ligase modulators (CELMoD®) with specific protein-binding properties. Based on the needs of this project, Evotec focused on the development of dedicated and innovative software solutions that greatly helped to accelerate not only the project’s progress but also contributed to the overall progression of Evotec’s PanHunter platform.

The approach has generated a pipeline of novel first-in-class programs, two of which have transitioned successfully into lead optimization after completing respective validation processes on Evotec’s platforms. In this context, Evotec´s integrated data analysis platform panHunter and the Company’s AI and machine learning tools are used to quickly screen, share, and validate results – not only by Evotec, but also by BMS scientists. In May 2022, the partnership was expanded even further for an additional 8 years with the goal to once again broaden and deepen the strategic alliance.

Targeted protein degradation is not only useful in oncology - a number of other diseases, e.g. Alzheimer’s, bacterial and viral infections lead to the presence of unwanted proteins inside cells that may be marked for destruction by this powerful technology. Evotec therefore is welcoming partners interested in exploring this approach in collaborations.

To learn more about the BMS collaboration and the use of targeted protein degradation technology read the official press release.

Learn More

Tags: Oncology, Blog, Modelling and Simulation

AAV Gene Therapy: Closing the Translational Gap

Posted by Evotec on Mar 31, 2022 10:26:55 AM

About the Webinar

This exciting collaboration between Evotec and Professor Dirk Grimm from the University of Heidelberg will review the current challenges facing the field of gene therapy.

The webinar includes:

  • A short introduction to AAV gene therapy
  • Recent clinical findings
  • Lessons we have already learned regarding improving AAV-based gene therapies
  • Strategies for next generation canidates
  • Efficacy and safety evaluation in preclinical models and their translatability into humans

 


About the Speakers

 

WernerHöllriegl

Werner Höllriegl

VP, Head of In Vivo Gene Therapy | Evotec

Werner Höllriegl is heading In Vivo Gene Therapy at Evotec GT in Orth an der Donau, Austria. He received his degree in Veterinary Medicine from the University of Veterinary Medicine Vienna, Austria. After spending a couple of years as Assistant Professor at the Department of Anesthesiology, he moved into industrial research spending more than 15 years in different Pharma Companies (Baxter, AstraZeneca, Novartis Institutes for BioMedical Research, Baxalta, Shire, Takeda) in Research and Nonclinical Development, focussing on biologics and in vivo gene therapy.

HanspeterRottensteiner_evotec

Hanspeter Rottensteiner

VP, Head of In Vitro Gene Therapy | Evotec

Hanspeter Rottensteiner is heading In Vitro Gene Therapy at Evotec GT in Austria. Biochemist by training, Hanspeter has more than 10 years in academia, and 15 years of experience in Pharma (Baxter, Takeda) in senior positions. He brings significant expertise in drug development of biologics and gene therapies, with a strong focus on rare diseases. He received his degree in Biochemistry from the University of Vienna, Austria.

 

Rudiger

Rüdiger Fritsch

Principle Scientist, Metabolic Disease | Evotec

Rüdiger Fritsch is heading a transcriptomics team at Evotec. He has recieved his PhD from the Max Planck Institute for Biophysical Chemistry. With over 15 years of experience as a biologist, Rüdiger has a strong expertise in omics.

 

Dirk Grimm

Dirk Grimm

Professor of Viral Vector Technologies Medical Faculty and the Department of Infectious Diseases/Virology | Heidelberg University

Dirk Grimm is heading the "Virus-host interactions" research group within the Department of Infectious Diseases/Virology. He has over 25 years of experience in AAV capsid and genome engineering as well as  in human gene therapy. Dirk has trained under AAV experts Jürgen Kleinschmidt (German Cancer Research Center Heidelberg) and Mark Kay (Stanford University School of Medicine). 

Watch the webinar to learn more!

[hs-form id="6827abd7-94a5-4df5-8beb-3d1da436ad90" show="hide on login" url="https://news.evotecsciencepool.com/hubfs/Webinars/AAV%20GENE%20THERAPY_2022/AAV%20Gene%20Therapy%20Webinar%20WoD.mp4"]

Tags: Videos & Webinars, ADME/DMPK, Modelling and Simulation

PK Prediction for Early Drug Discovery- Simon Thomas, Cyprotex

Posted by Evotec on Jan 31, 2022 1:17:56 PM

Learn about exciting new developments in pharmacokinetic prediction, and how to transform your use of ADME data.

 


About the Webinar

 

Whilst early in vitro screening for absorption, distribution metabolism and elimination (ADME) properties has significantly transformed early drug screening in many companies, the corresponding ability to quickly, and reliably, predict pharmacokinetics (PK) from these data has lagged behind. In vivo human prediction still largely relies on scaling from animal in vivo PK data, precluding its use for mass PK screening in early discovery.

In this webinar our expert, Simon Thomas, outlines Cyprotex's most recent work in the prediction of human PK from early ADME data. Combining ADME data with physicochemical and structural information within a novel physiologically based pharmacokinetic (PBPK) model, we have developed a service that returns a comprehensive array of reliable PK data and metrics: summary PK parameters and plasma concentrations are predicted for oral, intravenous bolus and intravenous infusion administration, whether single- or repeat-dose regimes. Generation of these predictions greatly enhances the value of the ADME data, and facilitates multiple options for directing compound progression: use of the predicted PK parameters enables compounds with desirable properties to be identified, whilst reliable plasma concentration prediction enables the implementation of pharmacokinetic/pharmacodynamic (PK/PD) modelling for early assessment of in vivo potential. High throughput and rapid turnaround maximally facilitate the make-test-analyse process.

 


About the Speaker

 

Simon Thomas resize bw

Simon Thomas PhD

Head of Modelling and Simulation | Cyprotex

Dr Simon Thomas is the Head of Modelling and Simulation at Cyprotex where he is responsible for the development of mathematical models for predicting ADME properties, pharmacokinetics, toxicity and clinical efficacy. Simon studied chemistry at the University of Oxford, as a final year student writing his first computer models, on the emission of electrons via the photoelectric effect. He obtained his Ph.D. and carried out post-doctoral work in the nascent field of systems biology at Oxford Brookes University, modelling the regulation of biochemical pathways in plants and animals, particularly with respect to pathways of energy metabolism. He joined Cyprotex in 1999, initially leading the company’s modelling efforts in PK prediction, over time extending the company’s capabilities into prediction of toxicity and pharmacological activity.

Watch the webinar to learn more!

[hs-form id="65ebef6f-b35d-4dff-a609-c8f1b77e39c4" show="hide on login" url="https://news.evotecsciencepool.com/hubfs/Sciencepool/Videos%20and%20Webinars/Cyprotex%20webinars%20and%20videos/pk-prediction-in-early-drug-discovery-webinar-nov2021.mp4"]

Tags: Videos & Webinars, ADME/DMPK, Modelling and Simulation