SLAS DiscoveryPub Date : 2024-04-18DOI: 10.1016/j.slasd.2024.100156
Alice Lanne , Catherine Bardelle , Gareth Davies , Antonia Turberville , Hannah Semple , Rachel Moore , Geoffrey A. Holdgate
{"title":"POLARISED views and FRETting about probe modulation assays: Learning from High Throughput Screening","authors":"Alice Lanne , Catherine Bardelle , Gareth Davies , Antonia Turberville , Hannah Semple , Rachel Moore , Geoffrey A. Holdgate","doi":"10.1016/j.slasd.2024.100156","DOIUrl":"https://doi.org/10.1016/j.slasd.2024.100156","url":null,"abstract":"<div><p>Fluorescent probe modulation assays are a widely used approach to monitor displacement or stabilisation of fluorescently labelled tool ligands by test compounds. These assays allow an optical read-out of probe-receptor binding and can be used to detect compounds that compete with the labelled ligand, either directly or indirectly. Probes for both orthosteric and allosteric sites are often employed. The method can also be used to identify test compounds that may stabilise the ternary complex, offering an opportunity to discover novel molecular glues. The utility of these fluorescence-based assays within high-throughput screening has been facilitated by the use of streptavidin labelled terbium as a donor and access to a range of different acceptor fluorophores. During 2023, the High-throughput Screening group at AstraZeneca carried out 8 high-throughput screens using these approaches. In this manuscript we will present the types of assays used, an overview of the timelines for assay development and screening, the application of orthogonal artefact methods to aid hit finding and the results of the screens in terms of hit rate and the number of compounds identified with IC<sub>50</sub> values of better than 30 µM. Learning across the development, execution and analysis of these screens will be presented.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2024-04-18","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2472555224000182/pdfft?md5=31508b5b2d14cbf4e0c02d5d7e69cecf&pid=1-s2.0-S2472555224000182-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140645364","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2024-04-01DOI: 10.1016/j.slasd.2024.100154
Inês C. Ferreira , Estefania Torrejón , Bernardo Abecasis , Bruno M. Alexandre , Ricardo A. Gomes , Chris Verslype , Jos van Pelt , Ana Barbas , Daniel Simão , Tiago M. Bandeiras , Alessio Bortoluzzi , Sofia P. Rebelo
{"title":"Aldehyde Dehydrogenase 2 (ALDH2): A novel sorafenib target in hepatocellular carcinoma unraveled by the proteome-wide cellular thermal shift assay","authors":"Inês C. Ferreira , Estefania Torrejón , Bernardo Abecasis , Bruno M. Alexandre , Ricardo A. Gomes , Chris Verslype , Jos van Pelt , Ana Barbas , Daniel Simão , Tiago M. Bandeiras , Alessio Bortoluzzi , Sofia P. Rebelo","doi":"10.1016/j.slasd.2024.100154","DOIUrl":"10.1016/j.slasd.2024.100154","url":null,"abstract":"<div><p>Sorafenib is a multikinase inhibitor indicated for first-line treatment of unresectable hepatocellular carcinoma. Despite its widespread use in the clinic, the existing knowledge of sorafenib mode-of-action remains incomplete. To build upon the current understanding, we used the Cellular Thermal Shift Assay (CETSA) coupled to Mass Spectrometry (CETSA-MS) to monitor compound binding to its target proteins in the cellular context on a proteome-wide scale. Among the potential sorafenib targets, we identified aldehyde dehydrogenase 2 (ALDH2), an enzyme that plays a major role in alcohol metabolism. We validated the interaction of sorafenib with ALDH2 by orthogonal methods using pure recombinant protein, proving that this interaction is not mediated by other cellular components. Moreover, we showed that sorafenib inhibits ALDH2 activity, supporting a functional role for this interaction. Finally, we were able to demonstrate that both ALDH2 protein expression and activity were reduced in sorafenib-resistant cells compared to the parental cell line. Overall, our study allowed the identification of ALDH2 as a novel sorafenib target and sheds light on its potential role in both hepatocellular carcinoma and sorafenib resistance condition.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2024-04-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2472555224000169/pdfft?md5=e39307bdcc31a11896db9724bd824787&pid=1-s2.0-S2472555224000169-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140195277","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2024-03-20DOI: 10.1016/j.slasd.2024.100153
Wenwen Zhou , Baoqing You , Xiaomeng Zhao , Shuyi Si , Yan Li , Jing Zhang
{"title":"Establishment, optimization and validation of a fluorescence polarization-based high-throughput screening assay targeting cathepsin L inhibitors","authors":"Wenwen Zhou , Baoqing You , Xiaomeng Zhao , Shuyi Si , Yan Li , Jing Zhang","doi":"10.1016/j.slasd.2024.100153","DOIUrl":"10.1016/j.slasd.2024.100153","url":null,"abstract":"<div><p>Cathepsin L (CTSL), a lysosomal cysteine proteinase, is primarily dedicated to the metabolic turnover of intracellular proteins. It is involved in various physiological processes and contributes to pathological conditions such as viral infection, tumor invasion and metastasis, inflammatory status, atherosclerosis, renal disease, diabetes, bone diseases, and other ailments. The coronavirus disease 2019 (COVID-19), with its rapid global spread and significant mortality, has been a worldwide epidemic since the late 2019s. Notably, CTSL plays a role in the processing of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein, providing a potential avenue to block coronavirus host cell entry and thereby inhibit SARS-CoV-2 infection in humans. In this study, we have developed a novel method using fluorescence polarization (FP) for screening CTSL inhibitors in a high-throughput format. The optimized assay demonstrated its appropriateness for high-throughput screening (HTS) with a Z-factor of 0.9 in a 96-well format. Additionally, the IC<sub>50</sub> of the known inhibitor, Z-Phe-Tyr-CHO, was determined to be 188.50 ± 46.88 nM. Upon screening over 2000 small molecules, we identified, for the first time, the anti-CTSL properties of a benzothiazoles derivative named IMB 8015. This work presents a novel high-throughput approach and its application in discovering and evaluating CTSL inhibitors.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2024-03-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2472555224000157/pdfft?md5=531dea274a50b4826ec3758fe007d67d&pid=1-s2.0-S2472555224000157-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140190478","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2024-03-20DOI: 10.1016/j.slasd.2024.100155
Wenyu Wang , Jing Tang , Andrea Zaliani
{"title":"Outline and background for the EU-OS solubility prediction challenge","authors":"Wenyu Wang , Jing Tang , Andrea Zaliani","doi":"10.1016/j.slasd.2024.100155","DOIUrl":"10.1016/j.slasd.2024.100155","url":null,"abstract":"<div><p>In June 2022, EU-OS came to the decision to make public a solubility data set of 100+K compounds obtained from several of the EU-OS proprietary screening compound collections. Leveraging on the interest of SLAS for screening scientific development it was decided to launch a joint EUOS-SLAS competition within the chemoinformatics and machine learning (ML) communities. The competition was open to real world computation experts, for the best, most predictive, classification model of compound solubility. The aim of the competition was multiple: from a practical side, the winning model should then serve as a cornerstone for future solubility predictions having used the largest training set so far publicly available. From a higher project perspective, the intent was to focus the energies and experiences, even if professionally not precisely coming from Pharma R&D; to address the issue of how to predict compound solubility. Here we report how the competition was ideated and the practical aspects of conducting it within the Kaggle framework, leveraging of the versatility and the open-source nature of this data science platform. Consideration on results and challenges encountered have been also examined.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2024-03-20","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2472555224000170/pdfft?md5=31adf40156a4682f7a09408654d5d462&pid=1-s2.0-S2472555224000170-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140190479","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2024-03-06DOI: 10.1016/j.slasd.2024.100149
Jose Brea , Maria J. Varela , Geert A. Daudey , Maria I. Loza
{"title":"High-throughput nephelometry methodology for qualitative determination of aqueous solubility of chemical libraries","authors":"Jose Brea , Maria J. Varela , Geert A. Daudey , Maria I. Loza","doi":"10.1016/j.slasd.2024.100149","DOIUrl":"10.1016/j.slasd.2024.100149","url":null,"abstract":"<div><p>The purpose of the protocol reported in this work is the solubility profiling of large chemical libraries using nephelometry. This technique allows the qualitative classification of compounds as highly, moderately, or poorly water-soluble. The described methodology is not intended to yield quantitative solubility values of the studied compounds but can be used as a primary solubility assessment of large chemical libraries, to guide hit prioritization after High Throughput Screening (HTS) campaigns.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2024-03-06","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S247255522400011X/pdfft?md5=a9b754a03190a54dd2fb6bb4cd348956&pid=1-s2.0-S247255522400011X-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140084712","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2024-03-02DOI: 10.1016/j.slasd.2024.100148
Emery Smith , Louise Dickson , Philip Pickford , Anna Rowland , Justin Shumate , Katherine Perez , Louis Scampavia , Derek Hernandez , Timothy P. Spicer
{"title":"Protocol for kinetic mode potassium channel assays on common plate readers and microscopes","authors":"Emery Smith , Louise Dickson , Philip Pickford , Anna Rowland , Justin Shumate , Katherine Perez , Louis Scampavia , Derek Hernandez , Timothy P. Spicer","doi":"10.1016/j.slasd.2024.100148","DOIUrl":"https://doi.org/10.1016/j.slasd.2024.100148","url":null,"abstract":"<div><p>Fluorescence-based potassium channel assays are typically run on expensive, hard to obtain, fluorescence imaging kinetic plate readers that are uncommon in most laboratories. Here we describe the use of the Brilliant Thallium Snapshot assay to conduct an endpoint potassium channel assay, so that it can be used across multiple plate reader platforms that are more common in many labs. These methods will allow users to identify modulators of potassium channels. For this work, we have taken a kinetic mode Molecular Devices FLIPR based protocol and adapted it to be utilized on endpoint plate readers, such as the BMG Labtech PHERAstar, to identify activators of GIRK channels in CHO cells. We demonstrate that both plate readers are functionally competent at generating excellent Z’ values which makes them ideally suited to finding corollary hits from the Sigma LOPAC 1,280 screening collection. Importantly, this assay has also been validated using a high content reader, demonstrating the possibility of spatially resolving signals from individual cells within a mixed cell population. The compendium of these results shows the flexibility, accessibility and functionality of endpoint-compatible potassium channel assay readouts on more common plate readers.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2024-03-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2472555224000108/pdfft?md5=fb5fd387899cb3c039a7cbf52f959893&pid=1-s2.0-S2472555224000108-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140052681","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2024-03-01DOI: 10.1016/j.slasd.2023.12.009
Lamiaa Bahnassawy , Nathalie Nicolaisen , Christopher Untucht , Benjamin Mielich-Süss , Lydia Reinhardt , Janina S. Ried , Martina P. Morawe , Daniela Geist , Anja Finck , Elke Käfer , Jürgen Korffmann , Matthew Townsend , Brinda Ravikumar , Viktor Lakics , Miroslav Cik , Peter Reinhardt
{"title":"Establishment of a high-content imaging assay for tau aggregation in hiPSC-derived neurons differentiated from two protocols to routinely evaluate compounds and genetic perturbations","authors":"Lamiaa Bahnassawy , Nathalie Nicolaisen , Christopher Untucht , Benjamin Mielich-Süss , Lydia Reinhardt , Janina S. Ried , Martina P. Morawe , Daniela Geist , Anja Finck , Elke Käfer , Jürgen Korffmann , Matthew Townsend , Brinda Ravikumar , Viktor Lakics , Miroslav Cik , Peter Reinhardt","doi":"10.1016/j.slasd.2023.12.009","DOIUrl":"10.1016/j.slasd.2023.12.009","url":null,"abstract":"<div><p>Aberrant protein aggregation is a pathological cellular hallmark of many neurodegenerative diseases, such as Alzheimer's disease (AD) and frontotemporal dementia (FTD), where the tau protein is aggregating, forming neurofibrillary tangles (NFTs), and propagating from neuron to neuron. These processes have been linked to disease progression and a decline in cognitive function. Various therapeutic approaches aim at the prevention or reduction of tau aggregates in neurons. Human induced pluripotent stem cells (hiPSCs) are a very valuable tool in neuroscience discovery, as they offer access to potentially unlimited amounts of cell types that are affected in disease, including cortical neurons, for <em>in vitro</em> studies. We have generated an <em>in vitro</em> model for tau aggregation that uses hiPSC – derived neurons expressing an aggregation prone, fluorescently tagged version of the human tau protein after lentiviral transduction. Upon addition of tau seeds in the form of recombinant sonicated paired helical filaments (sPHFs), the neurons show robust, disease-like aggregation of the tau protein. The model was developed as a plate-based high content screening assay coupled with an image analysis algorithm to evaluate the impact of small molecules or genetic perturbations on tau. We show that the assay can be used to evaluate small molecules or screen targeted compound libraries. Using siRNA-based gene knockdown, genes of interest can be evaluated, and we could show that a targeted gene library can be screened, by screening nearly 100 deubiquitinating enzymes (DUBs) in that assay. The assay uses an imaging-based readout, a relatively short timeline, quantifies the extent of tau aggregation, and also allows the assessment of cell viability. Furthermore, it can be easily adapted to different hiPSC lines or neuronal subtypes. Taken together, this complex and highly relevant approach can be routinely applied on a weekly basis in the screening funnels of several projects and generates data with a turnaround time of approximately five weeks.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2472555223000990/pdfft?md5=34d92141e506ff12fc2f7f9b45f750eb&pid=1-s2.0-S2472555223000990-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138833439","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2024-03-01DOI: 10.1016/j.slasd.2023.12.007
Shitalben Patel , Marie Karlsson , Joseph T. Klahn , Frank Gambino Jr. , Helena Costa , Kathleen A. McGuire , Christina K. Baumgartner , Jon Williams , Sarah Sandoz , James E. Kath
{"title":"Quantitative target engagement of RIPK1 in human whole blood via the cellular thermal shift assay for potential pre-clinical and clinical applications","authors":"Shitalben Patel , Marie Karlsson , Joseph T. Klahn , Frank Gambino Jr. , Helena Costa , Kathleen A. McGuire , Christina K. Baumgartner , Jon Williams , Sarah Sandoz , James E. Kath","doi":"10.1016/j.slasd.2023.12.007","DOIUrl":"10.1016/j.slasd.2023.12.007","url":null,"abstract":"<div><p>The cellular thermal shift assay (CETSA®) is a target engagement method widely used for preclinical characterization of small molecule compounds. CETSA® has been used for semi-quantitative readouts in whole blood with PBMC isolation, and quantitative, plate-based readouts using cell lines. However, there has been no quantitative evaluation of CETSA® in unprocessed human whole blood, which is preferred for clinical applications. Here we report two separate assay formats – Alpha CETSA® and MSD CETSA® – that require less than 100 μL of whole blood per sample without PBMC isolation. We chose RIPK1 as a proof-of-concept target and, by measuring engagement of seven different inhibitors, demonstrate high assay sensitivity and robustness. These quantitative CETSA® platforms enable possible applications in preclinical pharmacokinetic-pharmacodynamic studies, and direct target engagement with small molecules in clinical trials.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2472555223000977/pdfft?md5=c695ee332c2a10c9bcaef4c168483824&pid=1-s2.0-S2472555223000977-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138693365","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
{"title":"Screening approaches for the identification of Nrf2-Keap1 protein-protein interaction inhibitors targeting hot spot residues","authors":"Wataru Asano , Rie Hantani , Toru Uhara , François Debaene , Akihiro Nomura , Keishi Yamaguchi , Tsuyoshi Adachi , Kazuki Otake , Kazuhito Harada , Yoshiji Hantani","doi":"10.1016/j.slasd.2023.11.001","DOIUrl":"10.1016/j.slasd.2023.11.001","url":null,"abstract":"<div><p>Protein-protein interactions (PPIs) play a crucial role in most biological processes and are important targets in the development of therapeutic agents. However, small molecule drug discovery that targets PPIs remains very challenging. Targeting hot spot residues is considered the best option for inhibiting such interactions, but there are few examples of how knowledge of hot spots can be used in high throughput screening to find hit compounds. A substrate adaptor protein for a ubiquitin ligase complex, Kelch-like ECH-associated protein 1 (Keap1), negatively modulates the expression of genes involved in cellular protection against oxidative stress. Here, we focused on three arginine hot spot residues in the Keap1 substrate binding pocket (Arg380, Arg415, and Arg483), and screened the carboxylic acid library owned by Japan Tobacco Inc. for compounds that interact with the arginine residues in differential scanning fluorescence assays. Furthermore, we identified several small molecule compounds that specifically bind to the Keap1 Kelch domain hot spots by comparing binding to alanine mutant proteins (R380A, R415A, and R483A) with binding to the wild-type protein using surface plasmon resonance (SPR) screening. These compounds inhibited the protein-protein interaction between the Keap1 Kelch domain and the nuclear factor erythroid 2-related factor 2 (Nrf2) peptide, and the ubiquitination of Nrf2 catalyzed by the Cul3/RINGBox 1 E3 ligase. In addition, the binding mode of one compound (Compound 4) was determined by X-ray crystallography after validation of binding by isothermal titration calorimetry, native mass spectrometry, and nuclear magnetic resonance. Compound 4 had favorable thermodynamic properties, and noncovalently bound to Keap1 with a stoichiometry of 1:1. Our results suggest that Compound 4 could potentially be developed into effective therapeutic or preventive agents for a variety of diseases and conditions such as oxidative stress response, inflammation, and carcinogenesis. We believe that the use of a set of complementary biophysical techniques including the SPR assay with single alanine mutant of hot spots provides opportunities to identify hit compounds for developing inhibitors of PPIs.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2472555223000795/pdfft?md5=15c038b4f2ad2614e505129c72ef0593&pid=1-s2.0-S2472555223000795-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"71489819","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
SLAS DiscoveryPub Date : 2024-03-01DOI: 10.1016/j.slasd.2023.12.001
Kamran A. Ali , Reecha D. Shah , Anukriti Dhar , Nina M. Myers , Cameron Nguyen , Arisa Paul , Jordan E. Mancuso , A. Scott Patterson , James P. Brody , Diane Heiser
{"title":"Ex vivo discovery of synergistic drug combinations for hematologic malignancies","authors":"Kamran A. Ali , Reecha D. Shah , Anukriti Dhar , Nina M. Myers , Cameron Nguyen , Arisa Paul , Jordan E. Mancuso , A. Scott Patterson , James P. Brody , Diane Heiser","doi":"10.1016/j.slasd.2023.12.001","DOIUrl":"10.1016/j.slasd.2023.12.001","url":null,"abstract":"<div><p>Combination therapies have improved outcomes for patients with acute myeloid leukemia (AML). However, these patients still have poor overall survival. Although many combination therapies are identified with high-throughput screening (HTS), these approaches are constrained to disease models that can be grown in large volumes (e.g., immortalized cell lines), which have limited translational utility. To identify more effective and personalized treatments, we need better strategies for screening and exploring potential combination therapies. Our objective was to develop an HTS platform for identifying effective combination therapies with highly translatable ex vivo disease models that use size-limited, primary samples from patients with leukemia (AML and myelodysplastic syndrome). We developed a system, ComboFlow, that comprises three main components: MiniFlow, ComboPooler, and AutoGater. MiniFlow conducts ex vivo drug screening with a miniaturized flow-cytometry assay that uses minimal amounts of patient sample to maximize throughput. ComboPooler incorporates computational methods to design efficient screens of pooled drug combinations. AutoGater is an automated gating classifier for flow cytometry that uses machine learning to rapidly analyze the large datasets generated by the assay. We used ComboFlow to efficiently screen more than 3000 drug combinations across 20 patient samples using only 6 million cells per patient sample. In this screen, ComboFlow identified the known synergistic combination of bortezomib and panobinostat. ComboFlow also identified a novel drug combination, dactinomycin and fludarabine, that synergistically killed leukemic cells in 35 % of AML samples. This combination also had limited effects in normal, hematopoietic progenitors. In conclusion, ComboFlow enables exploration of massive landscapes of drug combinations that were previously inaccessible in ex vivo models. We envision that ComboFlow can be used to discover more effective and personalized combination therapies for cancers amenable to ex vivo models.</p></div>","PeriodicalId":21764,"journal":{"name":"SLAS Discovery","volume":null,"pages":null},"PeriodicalIF":3.1,"publicationDate":"2024-03-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2472555223000849/pdfft?md5=456103813a56aee8d81306efba4130ea&pid=1-s2.0-S2472555223000849-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138693366","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":4,"RegionCategory":"生物学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}