AIDDISON: Empowering Drug Discovery with AI/ML and CADD Tools in a Secure, Web-Based SaaS PlatformClick to copy article linkArticle link copied!
- Andrew RusinkoAndrew RusinkoMilliporeSigma, 400 Summit Drive, Burlington, Massachusetts 01803, United StatesMore by Andrew Rusinko
- Mohammad RezaeiMohammad RezaeiMilliporeSigma, 400 Summit Drive, Burlington, Massachusetts 01803, United StatesMore by Mohammad Rezaei
- Lukas FriedrichLukas FriedrichMerck Healthcare KGaA, Medicinal Chemistry and Drug Design, Darmstadt 64293, GermanyMore by Lukas Friedrich
- Hans-Peter BuchstallerHans-Peter BuchstallerMerck Healthcare KGaA, Medicinal Chemistry and Drug Design, Darmstadt 64293, GermanyMore by Hans-Peter Buchstaller
- Daniel Kuhn*Daniel Kuhn*Email: [email protected]Merck Healthcare KGaA, Medicinal Chemistry and Drug Design, Darmstadt 64293, GermanyMore by Daniel Kuhn
- Ashwini Ghogare*Ashwini Ghogare*Email: [email protected]MilliporeSigma, 400 Summit Drive, Burlington, Massachusetts 01803, United StatesMore by Ashwini Ghogare
Abstract
The widespread proliferation of artificial intelligence (AI) and machine learning (ML) methods has a profound effect on the drug discovery process. However, many scientists are reluctant to utilize these powerful tools due to the steep learning curve typically associated with them. AIDDISON offers a convenient, secure, web-based platform for drug discovery, addressing the reluctance of scientists to adopt AI and ML methods due to the steep learning curve. By seamlessly integrating generative models, ADMET property predictions, searches in vast chemical spaces, and molecular docking, AIDDISON provides a sophisticated platform for modern drug discovery. It enables less computer-savvy scientists to utilize these powerful tools in their daily activities, as demonstrated by an example of identifying a valuable set of molecules for lead optimization. With AIDDISON, the benefits of AI/ML in drug discovery are accessible to all.
This publication is licensed under
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
License Summary*
You are free to share(copy and redistribute) this article in any medium or format and to adapt(remix, transform, and build upon) the material for any purpose, even commercially within the parameters below:
Creative Commons (CC): This is a Creative Commons license.
Attribution (BY): Credit must be given to the creator.
*Disclaimer
This summary highlights only some of the key features and terms of the actual license. It is not a license and has no legal value. Carefully review the actual license before using these materials.
Introduction
Figure 1
Figure 1. Workflows implemented in AIDDISON.
Methods
2D Similarity Search
2D Pharmacophore Search
De Novo Design (Generative Models)
Shape-Based Search
Figure 2
Figure 2. Shape-based alignment of XAV-939 against the crystal-bound conformation of Olaparib.
Molecular Docking
Figure 3
Figure 3. Visualization of results from Molecular Docking workflow.
Results
Case Study. Tankyrase Inhibitors
Figure 4
Figure 4. Case study: design and refinement of tankyrase inhibitors.

Discussion
Figure 5
Figure 5. Chemical space explored around XAV-939 using de novo design (green) and 2D pharmacophore search (gold) of virtual collections.
Summary
Data Availability
The AIDDISON platform is commercially available to the public (https://www.sigmaaldrich.com/US/en/services/software-and-digital-platforms/aiddison-ai-powered-drug-discovery). The platform incorporates REINVENT 3.2 which is publicly available from https://github.com/MolecularAI/Reinvent. The generative model is pretrained and publicly accessible from https://github.com/MolecularAI/ReinventCommunity/tree/master/notebooks/models as “random.prior.new”. The third-party software included in AIDDISON are the FTrees algorithm (v6.10) from BioSolveIT (https://www.biosolveit.de/products/#FTrees), Flare (v7.2) and pyFlare (v7) from Cresset (https://www.cresset-group.com/software/flare), and Synthia (v23.2) from MilliporeSigma, the U.S. and Canada Life Science business of Merck KGaA, Darmstadt, Germany (https://www.synthiaonline.com). All physicochemical properties and the clustering algorithms were implemented using RDKit (https://github.com/rdkit/rdkit). The starting molecules, the resulting compounds, and the parameters to run the workflows in the case study are shared at Zenodo (https://zenodo.org/record/10231008).
References
This article references 38 other publications.
- 1Baum, Z. J.; Yu, X.; Ayala, P. Y.; Zhao, Y.; Watkins, S. P.; Zhou, Q. Artificial Intelligence in Chemistry: Current Trends and Future Directions. J. Chem. Inf. Model. 2021, 61 (7), 3197– 3212, DOI: 10.1021/acs.jcim.1c00619Google Scholar1Artificial Intelligence in Chemistry: Current Trends and Future DirectionsBaum, Zachary J.; Yu, Xiang; Ayala, Philippe Y.; Zhao, Yanan; Watkins, Steven P.; Zhou, QiongqiongJournal of Chemical Information and Modeling (2021), 61 (7), 3197-3212CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)A review. The application of artificial intelligence (AI) to chem. has grown tremendously in recent years. In this paper, we studied the growth and distribution of AI-related chem. publications in the last two decades using the CAS Content Collection. The vol. of both journal and patent publications have increased dramatically, esp. since 2015. Study of the distribution of publications over various chem. research areas revealed that anal. chem. and biochem. are integrating AI to the greatest extent and with the highest growth rates. We also investigated trends in interdisciplinary research and identified frequently occurring combinations of research areas in publications. Furthermore, topic analyses were conducted for journal and patent publications to illustrate emerging assocns. of AI with certain chem. research topics. Notable publications in various chem. disciplines were then evaluated and presented to highlight emerging use cases. Finally, the occurrence of different classes of substances and their roles in AI-related chem. research were quantified, further detailing the popularity of AI adoption in the life sciences and anal. chem. In summary, this report offers a broad overview of how AI has progressed in various fields of chem. and aims to provide an understanding of its future directions.
- 2Meyers, J.; Fabian, B.; Brown, N. De novo Molecular Design and Generative Models. Drug Discovery Today 2021, 26 (11), 2707– 2715, DOI: 10.1016/j.drudis.2021.05.019Google Scholar2De novo molecular design and generative modelsMeyers, Joshua; Fabian, Benedek; Brown, NathanDrug Discovery Today (2021), 26 (11), 2707-2715CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Ltd.)A review. Mol. design strategies are integral to therapeutic progress in drug discovery. Computational approaches for de novo mol. design have been developed over the past three decades and, recently, thanks in part to advances in machine learning (ML) and artificial intelligence (AI), the drug discovery field has gained practical experience. Here, we review these learnings and present de novo approaches according to the coarseness of their mol. representation: i.e., whether mol. design is modeled on an atom-based, fragment-based, or reaction-based paradigm. Furthermore, we emphasize the value of strong benchmarks, describe the main challenges to using these methods in practice, and provide a viewpoint on further opportunities for exploration and challenges to be tackled in the upcoming years.
- 3Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; Bridgland, A.; Meyer, C.; Kohl, S. A. A.; Ballard, A. J.; Cowie, A.; Romera-Paredes, B.; Nikolov, S.; Jain, R.; Adler, J.; Back, T.; Petersen, S.; Reiman, D.; Clancy, E.; Zielinski, M.; Steinegger, M.; Pacholska, M.; Berghammer, T.; Bodenstein, S.; Silver, D.; Vinyals, O.; Senior, A. W.; Kavukcuoglu, K.; Kohli, P.; Hassabis, D. Highly accurate protein structure prediction with AlphaFold. Nature 2021, 596, 583– 589, DOI: 10.1038/s41586-021-03819-2Google Scholar3Highly accurate protein structure prediction with AlphaFoldJumper, John; Evans, Richard; Pritzel, Alexander; Green, Tim; Figurnov, Michael; Ronneberger, Olaf; Tunyasuvunakool, Kathryn; Bates, Russ; Zidek, Augustin; Potapenko, Anna; Bridgland, Alex; Meyer, Clemens; Kohl, Simon A. A.; Ballard, Andrew J.; Cowie, Andrew; Romera-Paredes, Bernardino; Nikolov, Stanislav; Jain, Rishub; Adler, Jonas; Back, Trevor; Petersen, Stig; Reiman, David; Clancy, Ellen; Zielinski, Michal; Steinegger, Martin; Pacholska, Michalina; Berghammer, Tamas; Bodenstein, Sebastian; Silver, David; Vinyals, Oriol; Senior, Andrew W.; Kavukcuoglu, Koray; Kohli, Pushmeet; Hassabis, DemisNature (London, United Kingdom) (2021), 596 (7873), 583-589CODEN: NATUAS; ISSN:0028-0836. (Nature Portfolio)Proteins are essential to life, and understanding their structure can facilitate a mechanistic understanding of their function. Through an enormous exptl. effort, the structures of around 100,000 unique proteins have been detd., but this represents a small fraction of the billions of known protein sequences. Structural coverage is bottlenecked by the months to years of painstaking effort required to det. a single protein structure. Accurate computational approaches are needed to address this gap and to enable large-scale structural bioinformatics. Predicting the three-dimensional structure that a protein will adopt based solely on its amino acid sequence-the structure prediction component of the 'protein folding problem'-has been an important open research problem for more than 50 years. Despite recent progress, existing methods fall far short of at. accuracy, esp. when no homologous structure is available. Here we provide the first computational method that can regularly predict protein structures with at. accuracy even in cases in which no similar structure is known. We validated an entirely redesigned version of our neural network-based model, AlphaFold, in the challenging 14th Crit. Assessment of protein Structure Prediction (CASP14), demonstrating accuracy competitive with exptl. structures in a majority of cases and greatly outperforming other methods. Underpinning the latest version of AlphaFold is a novel machine learning approach that incorporates phys. and biol. knowledge about protein structure, leveraging multi-sequence alignments, into the design of the deep learning algorithm.
- 4Wenzel, J.; Matter, H.; Schmidt, F. Predictive Multitask Deep Neural Network Models for ADMETox Properties: Learning from Large Data Sets. J. Chem. Inf. Model. 2019, 59 (3), 1253– 1268, DOI: 10.1021/acs.jcim.8b00785Google Scholar4Predictive multitask deep neural network models for ADME-tox properties: learning from large data setsWenzel, Jan; Matter, Hans; Schmidt, FriedemannJournal of Chemical Information and Modeling (2019), 59 (3), 1253-1268CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Successful drug discovery projects require control and optimization of compd. properties related to pharmacokinetics, pharmacodynamics, and safety. While vol. and chemotype coverage of public and corporate ADME-Tox (absorption, distribution, excretion, metab., and toxicity) databases are constantly growing, deep neural nets (DNN) emerged as transformative artificial intelligence technol. to analyze those challenging data. Relevant features are automatically identified, while appropriate data can also be combined to multitask networks to evaluate hidden trends among multiple ADME-Tox parameters for implicitly correlated data sets. Here we describe a novel, fully industrialized approach to parametrize and optimize the setup, training, application, and visual interpretation of DNNs to model ADME-Tox data. Investigated properties include microsomal lability in different species, passive permeability in Caco-2/TC7 cells, and logD. Statistical models are developed using up to 50 000 compds. from public or corporate databases. Both the choice of DNN hyperparameters and the type and quantity of mol. descriptors were found to be important for successful DNN modeling. Alternate learning of multiple ADME-Tox properties, resulting in a multitask approach, performs statistically superior on most studied data sets in comparison to DNN single-task models and also provides a scalable method to predict ADME-Tox properties from heterogeneous data. For example, predictive quality using external validation sets was improved from R2 of 0.6 to 0.7 comparing single-task and multitask DNN networks from human metabolic lability data. Besides statistical evaluation, a new visualization approach is introduced to interpret DNN models termed "response map", which is useful to detect local property gradients based on structure fragmentation and derivatization. This method is successfully applied to visualize fragmental contributions to guide further design in drug discovery programs, as illustrated by CRCX3 antagonists and renin inhibitors, resp.
- 5Klucznik, T.; Mikulak-Klucznik, B.; McCormack, M. P.; Lima, H.; Szymkuć, S.; Bhowmick, M.; Molga, K.; Zhou, Y.; Rickershauser, L.; Gajewska, E. P.; Toutchkine, A.; Dittwald, P.; Startek, M. P.; Kirkovits, G. J.; Roszak, R.; Adamski, A.; Sieredzińska, B.; Mrksich, M.; Trice, S. L. J.; Grzybowski, B. A. Efficient Syntheses of Diverse, Medicinally Relevant Targets Planned by Computer and Executed in the Laboratory. Chem. 2018, 4, 522– 532, DOI: 10.1016/j.chempr.2018.02.002Google Scholar5Efficient Syntheses of Diverse, Medicinally Relevant Targets Planned by Computer and Executed in the LaboratoryKlucznik, Tomasz; Mikulak-Klucznik, Barbara; McCormack, Michael P.; Lima, Heather; Szymkuc, Sara; Bhowmick, Manishabrata; Molga, Karol; Zhou, Yubai; Rickershauser, Lindsey; Gajewska, Ewa P.; Toutchkine, Alexei; Dittwald, Piotr; Startek, Michal P.; Kirkovits, Gregory J.; Roszak, Rafal; Adamski, Ariel; Sieredzinska, Bianka; Mrksich, Milan; Trice, Sarah L. J.; Grzybowski, Bartosz A.Chem (2018), 4 (3), 522-532CODEN: CHEMVE; ISSN:2451-9294. (Cell Press)The Chematica program was used to autonomously design synthetic pathways to eight structurally diverse targets, including seven com. valuable bioactive substances and one natural product. All of these computer-planned routes were successfully executed in the lab. and offer significant yield improvements and cost savings over previous approaches, provide alternatives to patented routes, or produce targets that were not synthesized previously.
- 6Hoffmann, T.; Gastreich, M. The Next Level in Chemical Space Navigation: Going Far Beyond Enumerable Compound Libraries. Drug Discovery Today 2019, 24 (5), 1148– 1156, DOI: 10.1016/j.drudis.2019.02.013Google Scholar6The next level in chemical space navigation: going far beyond enumerable compound librariesHoffmann, Torsten; Gastreich, MarcusDrug Discovery Today (2019), 24 (5), 1148-1156CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Ltd.)Recent innovations have brought pharmacophore-driven methods for navigating virtual chem. spaces, the size of which can reach into the billions of mols., to the fingertips of every chemist. There has been a paradigm shift in the underlying computational chem. that drives chem. space search applications, incorporating intelligent reaction knowledge into their core so that they can readily deliver com. available mols. as nearest neighbor hits from within giant virtual spaces. These vast resources enable medicinal chemists to execute rapid scaffold-hopping expts., rapid hit expansion, and structure-activity relationship (SAR) exploitation in largely intellectual property (IP)-free territory and at unparalleled low cost.
- 7Howes, L. Hunting for Drugs in Chemical Space. Chem. Eng. News 2022, 100 (23), 20, DOI: 10.47287/cen-10023-coverGoogle ScholarThere is no corresponding record for this reference.
- 8Daina, A.; Michielin, O.; Zoete, V. SwissADME: A Free Web Tool to Evaluate Pharmacokinetics, Drug-likeness and Medicinal Chemistry Friendliness of Small Molecules. Sci. Rep 2017, 7, 42717, DOI: 10.1038/srep42717Google Scholar8SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small moleculesDaina Antoine; Michielin Olivier; Zoete Vincent; Michielin Olivier; Michielin OlivierScientific reports (2017), 7 (), 42717 ISSN:.To be effective as a drug, a potent molecule must reach its target in the body in sufficient concentration, and stay there in a bioactive form long enough for the expected biologic events to occur. Drug development involves assessment of absorption, distribution, metabolism and excretion (ADME) increasingly earlier in the discovery process, at a stage when considered compounds are numerous but access to the physical samples is limited. In that context, computer models constitute valid alternatives to experiments. Here, we present the new SwissADME web tool that gives free access to a pool of fast yet robust predictive models for physicochemical properties, pharmacokinetics, drug-likeness and medicinal chemistry friendliness, among which in-house proficient methods such as the BOILED-Egg, iLOGP and Bioavailability Radar. Easy efficient input and interpretation are ensured thanks to a user-friendly interface through the login-free website http://www.swissadme.ch. Specialists, but also nonexpert in cheminformatics or computational chemistry can predict rapidly key parameters for a collection of molecules to support their drug discovery endeavours.
- 9ISO 27001 Certification . 2022; https://www.iso.org/obp/ui/#iso:std:iso-iec:27001:ed-3:v1:en (accessed 2023–04–21).Google ScholarThere is no corresponding record for this reference.
- 10Félix, E.; Landrum, G., Dalke, A. FPSim2: Simple package for fast molecular similarity searches . 2022; https://chembl.github.io/FPSim2/ (accessed 2023–04–20).Google ScholarThere is no corresponding record for this reference.
- 11Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B. A.; Thiessen, P. A.; Yu, B.; Zaslavsky, L.; Zhang, J.; Bolton, E. E. PubChem 2023 Update. Nucleic Acids Res. 2023, 51 (D1), D1373– D1380, DOI: 10.1093/nar/gkac956Google ScholarThere is no corresponding record for this reference.
- 12Mendez, D.; Gaulton, A.; Bento, A. P.; Chambers, J.; De Veij, M.; Félix, E.; Magariños, M. P.; Mosquera, J. F.; Mutowo, P.; Nowotka, M.; Gordillo-Marañón, M.; Hunter, F.; Junco, L.; Mugumbate, G.; Rodriguez-Lopez, M.; Atkinson, F.; Bosc, N.; Radoux, C. J.; Segura-Cabrera, A.; Leach, A. R.; Hersey, A. ChEMBL: Towards Direct Deposition of Bioassay Data. Nucleic Acids Res. 2019, 47 (D1), D930– D940, DOI: 10.1093/nar/gky1075Google Scholar12ChEMBL: towards direct deposition of bioassay dataMendez, David; Gaulton, Anna; Bento, A. Patricia; Chambers, Jon; De Veij, Marleen; Felix, Eloy; Magarinos, Maria Paula; Mosquera, Juan F.; Mutowo, Prudence; Nowotka, Michal; Gordillo-Maranon, Maria; Hunter, Fiona; Junco, Laura; Mugumbate, Grace; Rodriguez-Lopez, Milagros; Atkinson, Francis; Bosc, Nicolas; Radoux, Chris J.; Cabrera, Aldo Segura; Hersey, Anne; Leach, Andrew R.Nucleic Acids Research (2019), 47 (D1), D930-D940CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)ChEMBL is a large, open-access bioactivity database, previously described in the 2012, 2014 and 2017 Nucleic Acids Research Database Issues. In the last two years, several important improvements have been made to the database and are described here. These include more robust capture and representation of assay details; a new data deposition system, allowing updating of data sets and deposition of supplementary data; and a completely redesigned web interface, with enhanced search and filtering capabilities.
- 13Irwin, J. J.; Tang, K. G.; Young, J.; Dandarchuluun, C.; Wong, B. R.; Khurelbaatar, M.; Moroz, Y. S.; Mayfield, J.; Sayle, R. A. ZINC20-A Free Ultralarge-Scale Chemical Database for Ligand Discovery. J. Chem. Inf. Model. 2020, 60 (12), 6065– 6073, DOI: 10.1021/acs.jcim.0c00675Google Scholar13ZINC20-A Free Ultralarge-Scale Chemical Database for Ligand DiscoveryIrwin, John J.; Tang, Khanh G.; Young, Jennifer; Dandarchuluun, Chinzorig; Wong, Benjamin R.; Khurelbaatar, Munkhzul; Moroz, Yurii S.; Mayfield, John; Sayle, Roger A.Journal of Chemical Information and Modeling (2020), 60 (12), 6065-6073CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Identifying and purchasing new small mols. to test in biol. assays are enabling for ligand discovery, but as purchasable chem. space continues to grow into the tens of billions based on inexpensive make-on-demand compds., simply searching this space becomes a major challenge. We have therefore developed ZINC20, a new version of ZINC with two major new features: billions of new mols. and new methods to search them. As a fully enumerated database, ZINC can be searched precisely using explicit at.-level graph-based methods, such as SmallWorld for similarity and Arthor for pattern and substructure search, as well as 3D methods such as docking. Anal. of the new make-on-demand compd. sets by these and related tools reveals startling features. For instance, over 97% of the core Bemis-Murcko scaffolds in make-on-demand libraries are unavailable from "in-stock" collections. Correspondingly, the no. of new Bemis-Murcko scaffolds is rising almost as a linear fraction of the elaborated mols. Thus, an 88-fold increase in the no. of mols. in the make-on-demand vs. the in-stock sets is built upon a 16-fold increase in the no. of Bemis-Murcko scaffolds. The make-on-demand library is also more structurally diverse than phys. libraries, with a massive increase in disk- and sphere-like shaped mols. The new system is freely available at zinc20.docking.org.
- 14Rarey, M.; Dixon, J. S. Feature Trees: A New Molecular Similarity Measure Based on Tree Matching. J. Comput. Aided. Mol. Des. 1998, 12, 471– 490, DOI: 10.1023/A:1008068904628Google Scholar16Feature trees: a new molecular similarity measure based on tree matchingRarey, Matthias; Dixon, J. ScottJournal of Computer-Aided Molecular Design (1998), 12 (5), 471-490CODEN: JCADEQ; ISSN:0920-654X. (Kluwer Academic Publishers)In this paper we present a new method for evaluating mol. similarity between small org. compds. Instead of a linear representation like fingerprints, a more complex description, a feature tree, is calcd. for a mol. A feature tree represents hydrophobic fragments and functional groups of the mol. and the way these groups are linked together. Each node in the tree is labeled with a set of features representing chem. properties of the part of the mol. corresponding to the node. The comparison of feature trees is based on matching subtrees of two feature trees onto each other. Two algorithms for tackling the matching problem are described throughout this paper. On a dataset of about 1000 mols., we demonstrate the ability of our approach to identify mols. belonging to the same class of inhibitors. With a second dataset of 58 mols. with known binding modes taken from the Brookhaven Protein Data Bank, we show that the matchings produced by our algorithms are compatible with the relative orientation of the mols. in the active site in 61% of the test cases. The av. computation time for a pair comparison is about 50 ms on a current workstation.
- 15Boehm, M.; Wu, T.-Y.; Claussen, H.; Lemmen, C. Similarity Searching and Scaffold Hopping in Synthetically Accessible Combinatorial Chemistry Spaces. J. Med. Chem. 2008, 51, 2468– 2480, DOI: 10.1021/jm0707727Google Scholar17Similarity searching and scaffold hopping in synthetically accessible combinatorial chemistry spacesBoehm, Markus; Wu, Tong-Ying; Claussen, Holger; Lemmen, ChristianJournal of Medicinal Chemistry (2008), 51 (8), 2468-2480CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Large collections of combinatorial libraries are an integral element in today's pharmaceutical industry. It is of great interest to perform similarity searches against all virtual compds. that are synthetically accessible by any such library. Here we describe the successful application of a new software tool CoLibri on 358 combinatorial libraries based on validated reaction protocols to create a single chem. space contg. over 1012 possible products. Similarity searching with FTrees-FS allows the systematic exploration of this space without the need to enumerate all product structures. The search result is a set of virtual hits which are synthetically accessible by one or more of the existing reaction protocols. Grouping these virtual hits by their synthetic protocols allows the rapid design and synthesis of multiple follow-up libraries. Such library ideas support hit-to-lead design efforts for tasks like follow-up from high-throughput screening hits or scaffold hopping from one hit to another attractive series.
- 16Enamine REAL Space. https://enamine.net/compound-collections/real-compounds/real-space-navigator (accessed 2023-04–21).Google ScholarThere is no corresponding record for this reference.
- 17WuXi GalaXi. https://wuxibiology.com/drug-discovery-services/hit-finding-and-screening-services/virtual-screening/ (accessed 2023-04–21).Google ScholarThere is no corresponding record for this reference.
- 18Blaschke, T.; Arús-Pous, J.; Chen, H.; Margreitter, C.; Tyrchan, C.; Engkvist, O.; Papadopoulos, K.; Patronov, A. REINVENT 2.0: An AI Tool for De Novo Drug Design. J. Chem. Inf. Model. 2020, 60 (12), 5918– 5922, DOI: 10.1021/acs.jcim.0c00915Google Scholar18REINVENT 2.0: An AI Tool for De Novo Drug DesignBlaschke, Thomas; Arus-Pous, Josep; Chen, Hongming; Margreitter, Christian; Tyrchan, Christian; Engkvist, Ola; Papadopoulos, Kostas; Patronov, AtanasJournal of Chemical Information and Modeling (2020), 60 (12), 5918-5922CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)In the past few years, we have witnessed a renaissance of the field of mol. de novo drug design. The advancements in deep learning and artificial intelligence (AI) have triggered an avalanche of ideas on how to translate such techniques to a variety of domains including the field of drug design. A range of architectures have been devised to find the optimal way of generating chem. compds. by using either graph- or string (SMILES)-based representations. With this application note, we aim to offer the community a prodn.-ready tool for de novo design, called REINVENT. It can be effectively applied on drug discovery projects that are striving to resolve either exploration or exploitation problems while navigating the chem. space. It can facilitate the idea generation process by bringing to the researcher's attention the most promising compds. REINVENT's code is publicly available at https://github.com/MolecularAI/Reinvent.
- 19Bickerton, G. R.; Paolini, G. V.; Besnard, J.; Muresan, S.; Hopkins, A. L. Quantifying the Chemical Beauty of Drugs. Nat. Chem. 2012, 4, 90– 98, DOI: 10.1038/nchem.1243Google Scholar19Quantifying the chemical beauty of drugsBickerton, G. Richard; Paolini, Gaia V.; Besnard, Jeremy; Muresan, Sorel; Hopkins, Andrew L.Nature Chemistry (2012), 4 (2), 90-98CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)Drug-likeness is a key consideration when selecting compds. during the early stages of drug discovery. However, evaluation of drug-likeness in abs. terms does not reflect adequately the whole spectrum of compd. quality. More worryingly, widely used rules may inadvertently foster undesirable mol. property inflation as they permit the encroachment of rule-compliant compds. towards their boundaries. We propose a measure of drug-likeness based on the concept of desirability called the quant. est. of drug-likeness (QED). The empirical rationale of QED reflects the underlying distribution of mol. properties. QED is intuitive, transparent, straightforward to implement in many practical settings and allows compds. to be ranked by their relative merit. We extended the utility of QED by applying it to the problem of mol. target druggability assessment by prioritizing a large set of published bioactive compds. The measure may also capture the abstr. notion of aesthetics in medicinal chem.
- 20Cheeseright, T. Fragment hopping with Blaze. Shape Match 2022 (accessed 2023–04–26).Google ScholarThere is no corresponding record for this reference.
- 21Narwal, M.; Venkannagari, H.; Lehtiö, L. Structural Basis of Selective Inhibition of Human Tankyrases. J. Med. Chem. 2012, 55 (3), 1360– 1367, DOI: 10.1021/jm201510pGoogle Scholar21Structural Basis of Selective Inhibition of Human TankyrasesNarwal, Mohit; Venkannagari, Harikanth; Lehtio, LariJournal of Medicinal Chemistry (2012), 55 (3), 1360-1367CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Tankyrases are poly(ADP-ribose) polymerases that have many cellular functions. They play pharmaceutically important roles, at least in telomere homeostasis and Wnt signaling, by covalently ADP-ribosylating target proteins and consequently regulating their functions. These features make tankyrases potential targets for treatment of cancer. We report here crystal structures of human tankyrase 2 catalytic fragment in complex with a byproduct, nicotinamide, and with selective inhibitors of tankyrases (IWR-1) and PARPs 1 and 2 (olaparib). Binding of these inhibitors to tankyrase 2 induces specific conformational changes. The crystal structures explain the selectivity of the inhibitors, reveal the flexibility of a substrate binding loop, and explain existing structure-activity relationship data. The first crystal structure of a PARP enzyme in complex with a potent inhibitor, IWR-1, that does not bind to the widely utilized nicotinamide-binding site makes the structure valuable for development of PARP inhibitors in general.
- 22Berman, H. M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T. N.; Weissig, H.; Shindyalov, I. N.; Bourne, P. E. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235– 242, DOI: 10.1093/nar/28.1.235Google Scholar22The Protein Data BankBerman, Helen M.; Westbrook, John; Feng, Zukang; Gilliland, Gary; Bhat, T. N.; Weissig, Helge; Shindyalov, Ilya N.; Bourne, Philip E.Nucleic Acids Research (2000), 28 (1), 235-242CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The Protein Data Bank (PDB; http://www.rcsb.org/pdb/)is the single worldwide archive of structural data of biol. macromols. This paper describes the goals of the PDB, the systems in place for data deposition and access, how to obtain further information, and near-term plans for the future development of the resource.
- 23Volkamer, A.; Kuhn, D.; Rippmann, F.; Rarey, M. DoGSiteScorer: A Web Server for Automatic Binding Site Prediction, Analysis and Druggability Assessment, Analysis and Druggability Assessment. Bioinformatics 2012, 28 (15), 2074– 5, DOI: 10.1093/bioinformatics/bts310Google Scholar23DoGSiteScorer: a web server for automatic binding site prediction, analysis and druggability assessmentVolkamer, Andrea; Kuhn, Daniel; Rippmann, Friedrich; Rarey, MatthiasBioinformatics (2012), 28 (15), 2074-2075CODEN: BOINFP; ISSN:1367-4803. (Oxford University Press)Motivation: Many drug discovery projects fail because the underlying target is finally found to be undruggable. Progress in structure elucidation of proteins now opens up a route to automatic structure-based target assessment. DoGSiteScorer is a newly developed automatic tool combining pocket prediction, characterization and druggability estn. and is now available through a web server. Availability: The DoGSiteScorer web server is freely available for academic use at http://dogsite.zbh.uni-hamburg.de Contact: [email protected].
- 24Cresset Flare Docking . 2022; https://www.cresset-group.com/software/flare-docking/ (accessed 2023–04–21).Google ScholarThere is no corresponding record for this reference.
- 25Shang, S.; Hua, F.; Hu, A.-W. The Regulation of β-Catenin Activity and Function in Cancer: Therapeutic Opportunities. Oncotarget 2017, 8 (20), 33972– 33989, DOI: 10.18632/oncotarget.15687Google Scholar25The regulation of β-catenin activity and function in cancer: therapeutic opportunitiesShang Shuang; Hua Fang; Hu Zhuo-WeiOncotarget (2017), 8 (20), 33972-33989 ISSN:.Wnt/β-catenin signaling is an evolutionarily conserved and versatile pathway that is known to be involved in embryonic development, tissue homeostasis and a wide variety of human diseases. Aberrant activation of this pathway gives rise to the accumulation of β-catenin in the nucleus and promotes the transcription of many oncogenes such as c-Myc and CyclinD-1. As a result, it contributes to carcinogenesis and tumor progression of several cancers, including colon cancer, hepatocellular carcinoma, pancreatic cancer, lung cancer and ovarian cancer. β-Catenin is a pivotal component of the Wnt signaling pathway and it is tightly regulated at three hierarchical levels: protein stability, subcellular localization and transcriptional activity. Uncovering the regulatory mechanisms of β-catenin will provide new insights into the pathogenesis of cancer and other diseases, as well as new therapeutic strategies against these diseases. In this review we dissect the concrete regulatory mechanisms of β-catenin from three aspects mentioned above. Then we focus on the role of β-catenin in cancer initiation, progression, dormancy, immunity and cancer stem cell maintenance. At last, we summarize the recent progress in the development of agents for the pharmacological modulation of β-catenin activity in cancer therapy.
- 26Wong, C. M.; Fan, S. T.; Ng, I. O. beta-Catenin mutation and overexpression in hepatocellular carcinoma: clinicopathologic and prognostic significance. Cancer 2001, 92 (1), 136– 145, DOI: 10.1002/1097-0142(20010701)92:1<136::AID-CNCR1301>3.0.CO;2-RGoogle Scholar26β-catenin mutation and overexpression in hepatocellular carcinoma: Clinicopathologic and prognostic significanceWong, Chun M.; Fan, Sheung T.; Ng, Irene O. L.Cancer (New York, NY, United States) (2001), 92 (1), 136-145CODEN: CANCAR; ISSN:0008-543X. (John Wiley & Sons, Inc.)β-Catenin has been recognized as a crit. member of the Wnt signaling pathway, and inappropriate activation of this pathway has been implicated in carcinogenesis. To det. the clin. significance of β-catenin in hepatocellular carcinoma (HCC), we performed mutational anal. at exon 3 of the gene, investigated the subcellular protein expression, and analyzed their clinicopathol. and prognostic significance in 60 patients with resected primary HCC. By direct DNA sequencing, somatic mutations of the β-catenin gene were detected in 7 (12%) HCCs. All the mutations were found at the region (exon 3) responsible for phosphorylation and ubiquitination, therefore likely to result in stabilization of free cytoplasmic β-catenin. Nuclear accumulation of the β-catenin protein, similar to the response to the Wnt signal, was found in 10 (17%) HCCs and was closely assocd. with gene mutation (P < 0.001). In the remaining cases, nonnuclear type overexpression, i.e., overexpression in the cytoplasm and/or cytoplasmic membrane, was obsd. in 31 (62%) HCCs, thus suggesting that the mechanisms leading to β-catenin overexpression may be heterogeneous. HCCs with a nonnuclear type of β-catenin overexpression were more frequently larger than 5 cm in diam. (P = 0.022) and had poorer cellular differentiation (P = 0.037), and the patients had significantly shorter disease-free survival lengths (P = 0.041). Review of the data from previous studies in HCC showed that β-catenin mutations were more frequent in HCV-assocd. HCC than in HBV-assocd. ones. β-Catenin mutation and deregulation may play an important role in hepatocarcinogenesis. Nonnuclear type β-catenin overexpression appeared to have pathol. and prognostic significance.
- 27Brabletz, T.; Jung, A.; Hermann, K.; Günther, K.; Hohenberger, W.; Kirchner, T. Nuclear overexpression of the oncoprotein beta-catenin in colorectal cancer is localized predominantly at the invasion front. Pathology, research and practice 1998, 194 (10), 701– 704, DOI: 10.1016/S0344-0338(98)80129-5Google Scholar27Nuclear overexpression of the oncoprotein β-catenin in colorectal cancer is localized predominantly at the invasion frontBrabletz, Thomas; Jung, Andreas; Hermann, Kathrin; Guenther, Klaus; Hohenberger, Werner; Kirchner, ThomasPathology, Research and Practice (1998), 194 (10), 701-704CODEN: PARPDS; ISSN:0344-0338. (Gustav Fischer Verlag)The distribution of overexpressed β-catenin within individual colorectal carcinomas was investigated using immunohistochem. In the most tumors a strong nuclear expression of β-catenin was found, predominantly localized at the invasion front with strongest nuclear staining of isolated, scattered tumor cells. In contrast, cells in the tumor center often showed a membranous expression of β-catenin, comparable to normal colon epithelium. It is therefore likely, that in addn. to the overexpression of β-catenin caused by defects in the adenomatous polyposis coli (APC) locus, regulatory events in the tumor itself lead to a different distribution of this oncoprotein.
- 28Shigemitsu, K.; Sekido, Y.; Usami, N.; Mori, S.; Sato, M.; Horio, Y.; Hasegawa, Y.; Bader, S.; Gazdar, A.; Minna, J.; Hida, T.; Yoshioka, H.; Imaizumi, M.; Ueda, Y.; Takahashi, M.; Shimokata, K. Genetic alteration of the beta-catenin gene (CTNNB1) in human lung cancer and malignant mesothelioma and identification of a new 3p21.3 homozygous deletion. Oncogene 2001, 20 (31), 4249– 4257, DOI: 10.1038/sj.onc.1204557Google Scholar28Genetic alteration of the β-catenin gene (CTNNB1) in human lung cancer and malignant mesothelioma and identification of a new 3p21.3 homozygous deletionShigemitsu, Kikuo; Sekido, Yoshitaka; Usami, Noriyasu; Mori, Shoichi; Sato, Mitsuo; Horio, Yoshitsugu; Hasegawa, Yoshinori; Bader, Scott A.; Gazdar, Adi F.; Minna, John D.; Hida, Toyoaki; Yoshioka, Hiromu; Imaizumi, Munehisa; Ueda, Yuichi; Takahashi, Masahide; Shimokata, KaoruOncogene (2001), 20 (31), 4249-4257CODEN: ONCNES; ISSN:0950-9232. (Nature Publishing Group)The β-catenin gene (CTNNB1) has been shown to be genetically mutated in various human malignancies. To det. whether the β-catenin gene is responsible for oncogenesis in thoracic malignancies, the authors searched for the mutation in 166 lung cancers (90 primary tumors and 76 cell lines), one blastoma and 10 malignant mesotheliomas (two primary tumors and eight cell lines). Among the lung cancers, including 43 small cell lung cancers (SCLCs) and 123 non-small cell lung cancers (NSCLCs), the authors identified four alterations in exon 3, which is the target region of mutation for stabilizing β-catenin. One primary adenocarcinoma had a somatic mutation from C to G, leading to an amino acid substitution from Ser to Cys at codon 37. Among the cell lines, SCLC NCI-H1092 had a mutation from A to G, leading to an Asp to Gly substitution at codon 6, NSCLC HCC15 had a mutation from C to T, leading to a Ser to Phe substitution at codon 45, and NSCLC NCI-H358 had a mutation from A to G, leading to a Thr to Ala substitution at codon 75. One blastoma also had a somatic mutation from C to G, leading to a Ser to Cys substitution at codon 37. Among the 10 malignant mesotheliomas, the authors identified a homozygous deletion in the NCI-H28 cell line. Cloning of the rearranged fragment from NCI-H28 indicated that all the exons except exon 1 of the β-catenin gene are deleted and that the deletion junction is 13 kb downstream from exon 1. Furthermore, Northern blot anal. of 26 lung cancer and eight mesothelioma cell line RNAs detected ubiquitous expression of the β-catenin messages except NCI-H28, although Western blot anal. showed that relatively less amts. of protein products were expressed in some of lung cancer cell lines. The authors' findings suggest that the β-catenin gene is infrequently mutated in lung cancer and that the NCI-H28 homozygous deletion of the β-catenin gene might indicate the possibility of a new tumor suppressor gene residing in this region at 3p21.3, where various types of human cancers show frequent allelic loss.
- 29Li, K.; Pan, W.-T.; Ma, Y.-B.; Xu, X.-L.; Gao, Y.; He, Y.-Q.; Wei, L.; Zhang, J.-W. BMX activates Wnt/β-catenin signaling pathway to promote cell proliferation and migration in breast cancer. Breast Cancer 2020, 27 (3), 363– 371, DOI: 10.1007/s12282-019-01024-8Google Scholar29BMX activates Wnt/β-catenin signaling pathway to promote cell proliferation and migration in breast cancerLi Kai; Pan Wen-Ting; Ma Yan-Bin; Xu Xiao-Long; Gao Yang; He Yan-Qi; Wei Lei; Zhang Jing-WeiBreast cancer (Tokyo, Japan) (2020), 27 (3), 363-371 ISSN:.BACKGROUND: Breast cancer has become a dangerous killer for the female, which seriously threatened women's life, leading to huge pressures to society. The present study assessed the mechanism underlying the involvement of bone marrow tyrosine kinase on chromosome X (BMX) in breast cancer development. METHODS: The expression of BMX was examined by qPCR and immunohistochemistry. The effect of BMX on cell proliferation and migration was detected by Clone formation assay and Transwell assay. In vitro study, the correlation of BMX with Wnt/β-catenin pathway was explored by western blot and TOP/FOP flash assay. RESULTS: In the present study, we found that BMX was up-regulated in breast cancer, which was associated with the tumor differentiation and TNM stage. Oncogenic BMX enhanced the ability of breast cancer cell proliferation and migration. Furthermore, BMX could up-regulate the protein expression levels of p-β-catenin (Y142), p-β-catenin(Y654) and inhibit the expression level of p-β-catenin (S33/37), thus activating Wnt/β-catenin pathway in MCF-7 and MDA-MB-231 cells. In addition, we revealed that BMX promoted GSK3β phosphorylation, which suppressed the degradation of β-catenin. CONCLUSIONS: In this study, we identified that BMX-activated Wnt/β-catenin signaling pathway, playing an oncogenic role in breast cancer, suggesting that BMX could become a potential treatment target of breast cancer.
- 30Zyla, R. E.; Olkhov-Mitsel, E.; Amemiya, Y.; Bassiouny, D.; Seth, A.; Djordjevic, B.; Nofech-Mozes, S.; Parra-Herran, C. CTNNB1Mutations and Aberrant β-Catenin Expression in Ovarian Endometrioid Carcinoma: Correlation With Patient Outcome. American journal of surgical pathology 2021, 45 (1), 68– 76, DOI: 10.1097/PAS.0000000000001553Google Scholar30CTNNB1 Mutations and Aberrant β-Catenin Expression in Ovarian Endometrioid Carcinoma: Correlation With Patient OutcomeZyla Roman E; Seth Arun; Djordjevic Bojana; Nofech-Mozes Sharon; Parra-Herran Carlos; Zyla Roman E; Olkhov-Mitsel Ekaterina; Bassiouny Dina; Seth Arun; Djordjevic Bojana; Nofech-Mozes Sharon; Parra-Herran Carlos; Amemiya Yutaka; Seth Arun; Bassiouny DinaThe American journal of surgical pathology (2021), 45 (1), 68-76 ISSN:.CTNNB1 mutations and aberrant β-catenin expression have adverse prognosis in endometrial endometrioid carcinoma, and recent evidence suggests a prognostic role of β-catenin in ovarian endometrioid carcinoma. Thus, we aimed to determine the prognostic value of the CTNNB1 mutational status, and its correlation with β-catenin expression, in a well-annotated cohort of 51 ovarian endometrioid carcinomas. We performed immunohistochemistry for β-catenin and developed an 11-gene next-generation sequencing panel that included whole exome sequencing of CTNNB1 and TP53. Results were correlated with clinicopathologic variables including disease-free and disease-specific survival. Tumor recurrence was documented in 14 patients (27%), and cancer-related death in 8 patients (16%). CTNNB1 mutations were found in 22 cases (43%), and nuclear β-catenin in 26 cases (51%). CTNNB1 mutation highly correlated with nuclear β-catenin (P<0.05). Mutated CTNNB1 status was statistically associated with better disease-free survival (P=0.04, log-rank test) and approached significance for better disease-specific survival (P=0.07). It also correlated with earlier International Federation of Gynecology and Obstetrics stage (P<0.05). Nuclear β-catenin, TP53 mutations, age, ProMisE group, surface involvement, tumor grade and stage also correlated with disease-free survival. There was no association between membranous β-catenin expression and disease-free or disease-specific survival. CTNNB1 mutations and nuclear β-catenin expression are associated with better progression-free survival in patients with OEC. This relationship may be in part due to a trend of CTNNB1-mutated tumors to present at early stage. β-catenin immunohistochemistry may serve as a prognostic biomarker and a surrogate for CTNN1B mutations in the evaluation of patients with ovarian endometrioid neoplasia, particularly those in reproductive-age or found incidentally without upfront staging surgery.
- 31Ferri, M.; Liscio, P.; Carotti, A.; Asciutti, S.; Sardella, R.; Macchiarulo, A.; Camaioni, E. Targeting Wnt-driven Cancers: Discovery of Novel Tankyrase Inhibitors. Eur. J. Med. Chem. 2017, 142, 506– 522, DOI: 10.1016/j.ejmech.2017.09.030Google Scholar31Targeting Wnt-driven cancers: Discovery of novel tankyrase inhibitorsFerri, Martina; Liscio, Paride; Carotti, Andrea; Asciutti, Stefania; Sardella, Roccaldo; Macchiarulo, Antonio; Camaioni, EmidioEuropean Journal of Medicinal Chemistry (2017), 142 (), 506-522CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)A review. Recent years have seen substantially heightened interest in the discovery of tankyrase inhibitors (TNKSi) as new promising anticancer agents. In this framework, the aim of this review article is focused on the description of potent TNKSi also endowed with disruptor activity toward the Wnt/β-catenin signaling pathway. Beginning with an overview of the most characterized TNKSi deriving from several drug design approaches and classifying them on the basis of the mol. interactions with the target, the authors discuss only those ones acting against Wnt cancer cell lines. In addn., comprehensive structure property relationships (SPR) emerging from the hit evolution processes and preclin. results are provided. The authors then review the most promising TNKSi hitherto reported in literature, acting in vivo models of Wnt-driven cancers. Some out-looks on current issues and future directions in this field are also discussed.
- 32Buchstaller, H. P.; Anlauf, U.; Dorsch, D.; Kögler, S.; Kuhn, D.; Lehmann, M.; Leuthner, B.; Lodholz, S.; Musil, D.; Radtki, D.; Rettig, C.; Ritzert, C.; Rohdich, F.; Schneider, R.; Wegener, A.; Weigt, S.; Wilkinson, K.; Esdar, C. Optimization of a Screening Hit toward M2912, an Oral Tankyrase Inhibitor with Antitumor Activity in Colorectal Cancer Models. J. Med. Chem. 2021, 64 (14), 10371– 10392, DOI: 10.1021/acs.jmedchem.1c00800Google Scholar32Optimization of a Screening Hit toward M2912, an Oral Tankyrase Inhibitor with Antitumor Activity in Colorectal Cancer ModelsBuchstaller, Hans-Peter; Anlauf, Uwe; Dorsch, Dieter; Koegler, Sarah; Kuhn, Daniel; Lehmann, Martin; Leuthner, Birgitta; Lodholz, Sara; Musil, Djordje; Radtki, Daniela; Rettig, Corinna; Ritzert, Claudio; Rohdich, Felix; Schneider, Richard; Wegener, Ansgar; Weigt, Stefan; Wilkinson, Kai; Esdar, ChristinaJournal of Medicinal Chemistry (2021), 64 (14), 10371-10392CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The identification of a screening hit series and its optimization through scaffold hopping and SAR exploration was described. The systematic assessment delivered M2912 I a compd. with an optimal balance between excellent TNKS potency, exquisite PARP selectivity, and a predicted human PK compatible with once daily oral dosing. Modulation of cellular Wnt pathway activity and significant tumor growth inhibition was demonstrated with this compd. in colorectal xenograft models in vivo.
- 33Buchstaller, H. P.; Anlauf, U.; Dorsch, D.; Kuhn, D.; Lehmann, M.; Leuthner, B.; Musil, D.; Radtki, D.; Ritzert, C.; Rohdich, F.; Schneider, R.; Esdar, C. Discovery and Optimization of 2-Arylquinazolin-4-ones into a Potent and Selective Tankyrase Inhibitor Modulating Wnt Pathway Activity. J. Med. Chem. 2019, 62 (17), 7897– 7909, DOI: 10.1021/acs.jmedchem.9b00656Google Scholar33Discovery and Optimization of 2-Arylquinazolin-4-ones into a Potent and Selective Tankyrase Inhibitor Modulating Wnt Pathway ActivityBuchstaller, Hans-Peter; Anlauf, Uwe; Dorsch, Dieter; Kuhn, Daniel; Lehmann, Martin; Leuthner, Birgitta; Musil, Djordje; Radtki, Daniela; Ritzert, Claudio; Rohdich, Felix; Schneider, Richard; Esdar, ChristinaJournal of Medicinal Chemistry (2019), 62 (17), 7897-7909CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Tankyrases 1 and 2 (TNKS1/2) are promising pharmacol. targets which recently gained interest for anticancer therapy in Wnt pathway dependent tumors. 2-Aryl-quinazolinones were identified and optimized into potent tankyrase inhibitors through SAR exploration around the quinazolinone core and the 4'-position of the Ph residue. These efforts were supported by anal. of TNKS X-ray and Watermap structures and resulted in compd. 5k(I), a potent, selective tankyrase inhibitor with favorable pharmacokinetic properties. The X-ray structure of I in complex with TNKS1 was solved and confirmed the design hypothesis. Modulation of Wnt pathway activity was demonstrated with this compd. in a colorectal xenograft model in vivo.
- 34Mehta, C. C.; Bhatt, H. G. Tankyrase inhibitors as Antitumor Agents: a Patent Update (2013–2020). Expert Opinion on Therapeutic Patents 2021, 31 (7), 645– 661, DOI: 10.1080/13543776.2021.1888929Google Scholar34Tankyrase inhibitors as antitumor agents: a patent update (2013 - 2020)Mehta, Chirag C.; Bhatt, Hardik G.Expert Opinion on Therapeutic Patents (2021), 31 (7), 645-661CODEN: EOTPEG; ISSN:1354-3776. (Taylor & Francis Ltd.)IntroductionTankyrase inhibitors gained significant attention as therapeutic targets in oncol. because of their potency. Their primary role in inhibiting the Wnt signaling pathway makes them an important class of compds. with the potential to be used as a combination therapy in future treatments of colorectal cancer. Areas coveredThis review describes pertinent work in the development of tankyrase inhibitors with a great emphasis on the recently patented TNKS inhibitors published from 2013 to 2020. This article also highlights a couple of promising candidates having tankyrase inhibitory effects and are currently undergoing clin. trials. Expert opinionFollowing the successful clin. applications of PARP inhibitors, tankyrase inhibition has gained significant attention in the research community as a target with high therapeutic potential. The ubiquitous role of tankyrase in cellular homeostasis and Wnt-dependent tumor proliferation brought difficulties for researchers to strike the right balance between potency and on-target toxicity. The need for novel tankyrase inhibitors with a better ADMET profile can introduce an addnl. regimen in treating various malignancies in monotherapy or adjuvant therapy. The development of combination therapies, including tankyrase inhibitors with or without PARP inhibitory properties, can potentially benefit the larger population of patients with unmet medical needs.
- 35Karlberg, T.; Markova, N.; Johansson, I.; Hammarström, M.; Schütz, P.; Weigelt, J.; Schüler, H. Structural Basis for the Interaction between Tankyrase-2 and a Potent Wnt-Signaling Inhibitor. J. Med. Chem. 2010, 53 (14), 5352– 5355, DOI: 10.1021/jm100249wGoogle Scholar35Structural Basis for the Interaction between Tankyrase-2 and a Potent Wnt-Signaling InhibitorKarlberg, Tobias; Markova, Natalia; Johansson, Ida; Hammarstrom, Martin; Schutz, Patrick; Weigelt, Johan; Schuler, HerwigJournal of Medicinal Chemistry (2010), 53 (14), 5352-5355CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We report two crystal structures of the PARP domain of human tankyrase-2 (TNKS2). Tankyrases are involved in fundamental cellular processes such as telomere homeostasis and Wnt signaling. The complex of TNKS2 with the potent inhibitor XAV939 provides insights into the mol. basis of the strong interaction and suggests routes for further development of tankyrase inhibitors.
- 36Narwal, M.; Haikarainen, T.; Fallarero, A.; Vuorela, P. M.; Lehtiö, L. Screening and Structural Analysis of Flavones Inhibiting Tankyrases. J. Med. Chem. 2013, 56 (9), 3507– 3517, DOI: 10.1021/jm3018783Google Scholar36Screening and Structural Analysis of Flavones Inhibiting TankyrasesNarwal, Mohit; Haikarainen, Teemu; Fallarero, Adyary; Vuorela, Pia M.; Lehtio, LariJournal of Medicinal Chemistry (2013), 56 (9), 3507-3517CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Flavonoids are known for their beneficial effects on human health, and therefore the therapeutic potential of these compds. have been extensively studied. Flavone has been previously identified as a tankyrase inhibitor, and to further elucidate whether tankyrases would be inhibited by other flavonoids, the authors performed a systematic screening of tankyrase 2 inhibitory activity using 500 natural and naturally derived flavonoids covering nine different flavonoid classes. All identified tankyrase inhibitors were flavones. The authors report crystal structures of all the hit compds. in complex with the catalytic domain of human tankyrase 2. Flavone derivs. in all 10 crystal structures bind to the nicotinamide binding site of tankyrase 2. Potencies of the active flavones toward tankyrases vary between 50 nM and 1.1 μM, and flavones show up to 200-fold selectivity for tankyrases over ARTD1. The mol. details of the interactions revealed by cocrystal structures efficiently describe the properties of potent flavone derivs. inhibiting tankyrases.
- 37Narwal, M.; Koivunen, J.; Haikarainen, T.; Obaji, E.; Legala, O. E.; Venkannagari, H.; Joensuu, P.; Pihlajaniemi, T.; Lehtiö, L. Discovery of Tankyrase Inhibiting Flavones with Increased Potency and Isoenzyme Selectivity. J. Med. Chem. 2013, 56 (20), 7880– 7889, DOI: 10.1021/jm401463yGoogle Scholar37Discovery of Tankyrase Inhibiting Flavones with Increased Potency and Isoenzyme SelectivityNarwal, Mohit; Koivunen, Jarkko; Haikarainen, Teemu; Obaji, Ezeogo; Legala, Ongey E.; Venkannagari, Harikanth; Joensuu, Paivi; Pihlajaniemi, Taina; Lehtio, LariJournal of Medicinal Chemistry (2013), 56 (20), 7880-7889CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Tankyrases are ADP-ribosyltransferases that play key roles in various cellular pathways, including the regulation of cell proliferation, and thus, they are promising drug targets for the treatment of cancer. Flavones have been shown to inhibit tankyrases and we report here the discovery of more potent and selective flavone derivs. Com. available flavones with single substitutions were used for structure-activity relation studies, and cocrystal structures of the 18 hit compds. were analyzed to explain their potency and selectivity. The most potent inhibitors were also tested in a cell-based assay, which demonstrated that they effectively antagonize Wnt signaling. To assess selectivity, they were further tested against a panel of homologous human ADP-ribosyltransferases. The most effective compd.(I; MN-64), showed 6 nM potency against tankyrase 1, isoenzyme selectivity, and Wnt signaling inhibition. This work forms a basis for rational development of flavones as tankyrase inhibitors and guides the development of other structurally related inhibitors.
- 38Sorkun, M. C.; Mullaj, D.; Koelman, J. M. V. A.; Er, S. ChemPlot, A Python Library for Chemical Space Visualization. Chemistry-Europe 2022, 2 (7), e202200005 DOI: 10.1002/cmtd.202200005Google ScholarThere is no corresponding record for this reference.
Cited By
Smart citations by scite.ai include citation statements extracted from the full text of the citing article. The number of the statements may be higher than the number of citations provided by ACS Publications if one paper cites another multiple times or lower if scite has not yet processed some of the citing articles.
This article is cited by 9 publications.
- Ayesh Madushanka, Eli Laird, Corey Clark, Elfi Kraka. SmartCADD: AI-QM Empowered Drug Discovery Platform with Explainability. Journal of Chemical Information and Modeling 2024, 64
(17)
, 6799-6813. https://doi.org/10.1021/acs.jcim.4c00720
- Chao Shen, Jianfei Song, Chang-Yu Hsieh, Dongsheng Cao, Yu Kang, Wenling Ye, Zhenxing Wu, Jike Wang, Odin Zhang, Xujun Zhang, Hao Zeng, Heng Cai, Yu Chen, Linkang Chen, Hao Luo, Xinda Zhao, Tianye Jian, Tong Chen, Dejun Jiang, Mingyang Wang, Qing Ye, Jialu Wu, Hongyan Du, Hui Shi, Yafeng Deng, Tingjun Hou. DrugFlow: An AI-Driven One-Stop Platform for Innovative Drug Discovery. Journal of Chemical Information and Modeling 2024, 64
(14)
, 5381-5391. https://doi.org/10.1021/acs.jcim.4c00621
- Xu Gong, Qun Liu, Rui Han, Yike Guo, Guoyin Wang. MIFS: An adaptive multipath information fused self-supervised framework for drug discovery. Neural Networks 2025, 184 , 107088. https://doi.org/10.1016/j.neunet.2024.107088
- Qi An, Liang Huang, Chuan Wang, Dongmei Wang, Yalan Tu. New strategies to enhance the efficiency and precision of drug discovery. Frontiers in Pharmacology 2025, 16 https://doi.org/10.3389/fphar.2025.1550158
- Olga Kapustina, Polina Burmakina, Nina Gubina, Nikita Serov, Vladimir Vinogradov. User-friendly and industry-integrated AI for medicinal chemists and pharmaceuticals. Artificial Intelligence Chemistry 2024, 2
(2)
, 100072. https://doi.org/10.1016/j.aichem.2024.100072
- Negar Mottaghi-Dastjerdi, Mohammad Soltany-Rezaee-Rad. Advancements and Applications of Artificial Intelligence in Pharmaceutical Sciences: A Comprehensive Review. Iranian Journal of Pharmaceutical Research 2024, 23
(1)
https://doi.org/10.5812/ijpr-150510
- Amol D. Gholap, Md Jasim Uddin, Md Faiyazuddin, Abdelwahab Omri, S. Gowri, Mohammad Khalid. Advances in artificial intelligence for drug delivery and development: A comprehensive review. Computers in Biology and Medicine 2024, 178 , 108702. https://doi.org/10.1016/j.compbiomed.2024.108702
- Bjoern Wimmer, Jan Schernthaner, Genevieve Edobor, Andreas Friedrich, Katharina Poeltner, Gazmend Temaj, Marlies Wimmer, Elli Kronsteiner, Mara Pichler, Hanna Gercke, Ronald Huber, Niklas Kaefer, Mark Rinnerthaler, Thomas Karl, Jan Krauß, Thomas Mohr, Christopher Gerner, Helmut Hintner, Michael Breitenbach, Johann W. Bauer, Christin Rakers, Daniel Kuhn, Joerg von Hagen, Norbert Müller, Adriana Rathner, Hannelore Breitenbach-Koller. RiboScreenTM Technology Delivers a Ribosomal Target and a Small-Molecule Ligand for Ribosome Editing to Boost the Production Levels of Tropoelastin, the Monomeric Unit of Elastin. International Journal of Molecular Sciences 2024, 25
(15)
, 8430. https://doi.org/10.3390/ijms25158430
- Anand Kumar Pandey, Shalja Verma. Computational Approaches for Structure-Assisted Drug Discovery and Repurposing. 2024, 163-192. https://doi.org/10.1007/978-981-97-7123-3_9
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
Abstract
Figure 1
Figure 1. Workflows implemented in AIDDISON.
Figure 2
Figure 2. Shape-based alignment of XAV-939 against the crystal-bound conformation of Olaparib.
Figure 3
Figure 3. Visualization of results from Molecular Docking workflow.
Figure 4
Figure 4. Case study: design and refinement of tankyrase inhibitors.
Figure 5
Figure 5. Chemical space explored around XAV-939 using de novo design (green) and 2D pharmacophore search (gold) of virtual collections.
References
This article references 38 other publications.
- 1Baum, Z. J.; Yu, X.; Ayala, P. Y.; Zhao, Y.; Watkins, S. P.; Zhou, Q. Artificial Intelligence in Chemistry: Current Trends and Future Directions. J. Chem. Inf. Model. 2021, 61 (7), 3197– 3212, DOI: 10.1021/acs.jcim.1c006191Artificial Intelligence in Chemistry: Current Trends and Future DirectionsBaum, Zachary J.; Yu, Xiang; Ayala, Philippe Y.; Zhao, Yanan; Watkins, Steven P.; Zhou, QiongqiongJournal of Chemical Information and Modeling (2021), 61 (7), 3197-3212CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)A review. The application of artificial intelligence (AI) to chem. has grown tremendously in recent years. In this paper, we studied the growth and distribution of AI-related chem. publications in the last two decades using the CAS Content Collection. The vol. of both journal and patent publications have increased dramatically, esp. since 2015. Study of the distribution of publications over various chem. research areas revealed that anal. chem. and biochem. are integrating AI to the greatest extent and with the highest growth rates. We also investigated trends in interdisciplinary research and identified frequently occurring combinations of research areas in publications. Furthermore, topic analyses were conducted for journal and patent publications to illustrate emerging assocns. of AI with certain chem. research topics. Notable publications in various chem. disciplines were then evaluated and presented to highlight emerging use cases. Finally, the occurrence of different classes of substances and their roles in AI-related chem. research were quantified, further detailing the popularity of AI adoption in the life sciences and anal. chem. In summary, this report offers a broad overview of how AI has progressed in various fields of chem. and aims to provide an understanding of its future directions.
- 2Meyers, J.; Fabian, B.; Brown, N. De novo Molecular Design and Generative Models. Drug Discovery Today 2021, 26 (11), 2707– 2715, DOI: 10.1016/j.drudis.2021.05.0192De novo molecular design and generative modelsMeyers, Joshua; Fabian, Benedek; Brown, NathanDrug Discovery Today (2021), 26 (11), 2707-2715CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Ltd.)A review. Mol. design strategies are integral to therapeutic progress in drug discovery. Computational approaches for de novo mol. design have been developed over the past three decades and, recently, thanks in part to advances in machine learning (ML) and artificial intelligence (AI), the drug discovery field has gained practical experience. Here, we review these learnings and present de novo approaches according to the coarseness of their mol. representation: i.e., whether mol. design is modeled on an atom-based, fragment-based, or reaction-based paradigm. Furthermore, we emphasize the value of strong benchmarks, describe the main challenges to using these methods in practice, and provide a viewpoint on further opportunities for exploration and challenges to be tackled in the upcoming years.
- 3Jumper, J.; Evans, R.; Pritzel, A.; Green, T.; Figurnov, M.; Ronneberger, O.; Tunyasuvunakool, K.; Bates, R.; Žídek, A.; Potapenko, A.; Bridgland, A.; Meyer, C.; Kohl, S. A. A.; Ballard, A. J.; Cowie, A.; Romera-Paredes, B.; Nikolov, S.; Jain, R.; Adler, J.; Back, T.; Petersen, S.; Reiman, D.; Clancy, E.; Zielinski, M.; Steinegger, M.; Pacholska, M.; Berghammer, T.; Bodenstein, S.; Silver, D.; Vinyals, O.; Senior, A. W.; Kavukcuoglu, K.; Kohli, P.; Hassabis, D. Highly accurate protein structure prediction with AlphaFold. Nature 2021, 596, 583– 589, DOI: 10.1038/s41586-021-03819-23Highly accurate protein structure prediction with AlphaFoldJumper, John; Evans, Richard; Pritzel, Alexander; Green, Tim; Figurnov, Michael; Ronneberger, Olaf; Tunyasuvunakool, Kathryn; Bates, Russ; Zidek, Augustin; Potapenko, Anna; Bridgland, Alex; Meyer, Clemens; Kohl, Simon A. A.; Ballard, Andrew J.; Cowie, Andrew; Romera-Paredes, Bernardino; Nikolov, Stanislav; Jain, Rishub; Adler, Jonas; Back, Trevor; Petersen, Stig; Reiman, David; Clancy, Ellen; Zielinski, Michal; Steinegger, Martin; Pacholska, Michalina; Berghammer, Tamas; Bodenstein, Sebastian; Silver, David; Vinyals, Oriol; Senior, Andrew W.; Kavukcuoglu, Koray; Kohli, Pushmeet; Hassabis, DemisNature (London, United Kingdom) (2021), 596 (7873), 583-589CODEN: NATUAS; ISSN:0028-0836. (Nature Portfolio)Proteins are essential to life, and understanding their structure can facilitate a mechanistic understanding of their function. Through an enormous exptl. effort, the structures of around 100,000 unique proteins have been detd., but this represents a small fraction of the billions of known protein sequences. Structural coverage is bottlenecked by the months to years of painstaking effort required to det. a single protein structure. Accurate computational approaches are needed to address this gap and to enable large-scale structural bioinformatics. Predicting the three-dimensional structure that a protein will adopt based solely on its amino acid sequence-the structure prediction component of the 'protein folding problem'-has been an important open research problem for more than 50 years. Despite recent progress, existing methods fall far short of at. accuracy, esp. when no homologous structure is available. Here we provide the first computational method that can regularly predict protein structures with at. accuracy even in cases in which no similar structure is known. We validated an entirely redesigned version of our neural network-based model, AlphaFold, in the challenging 14th Crit. Assessment of protein Structure Prediction (CASP14), demonstrating accuracy competitive with exptl. structures in a majority of cases and greatly outperforming other methods. Underpinning the latest version of AlphaFold is a novel machine learning approach that incorporates phys. and biol. knowledge about protein structure, leveraging multi-sequence alignments, into the design of the deep learning algorithm.
- 4Wenzel, J.; Matter, H.; Schmidt, F. Predictive Multitask Deep Neural Network Models for ADMETox Properties: Learning from Large Data Sets. J. Chem. Inf. Model. 2019, 59 (3), 1253– 1268, DOI: 10.1021/acs.jcim.8b007854Predictive multitask deep neural network models for ADME-tox properties: learning from large data setsWenzel, Jan; Matter, Hans; Schmidt, FriedemannJournal of Chemical Information and Modeling (2019), 59 (3), 1253-1268CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Successful drug discovery projects require control and optimization of compd. properties related to pharmacokinetics, pharmacodynamics, and safety. While vol. and chemotype coverage of public and corporate ADME-Tox (absorption, distribution, excretion, metab., and toxicity) databases are constantly growing, deep neural nets (DNN) emerged as transformative artificial intelligence technol. to analyze those challenging data. Relevant features are automatically identified, while appropriate data can also be combined to multitask networks to evaluate hidden trends among multiple ADME-Tox parameters for implicitly correlated data sets. Here we describe a novel, fully industrialized approach to parametrize and optimize the setup, training, application, and visual interpretation of DNNs to model ADME-Tox data. Investigated properties include microsomal lability in different species, passive permeability in Caco-2/TC7 cells, and logD. Statistical models are developed using up to 50 000 compds. from public or corporate databases. Both the choice of DNN hyperparameters and the type and quantity of mol. descriptors were found to be important for successful DNN modeling. Alternate learning of multiple ADME-Tox properties, resulting in a multitask approach, performs statistically superior on most studied data sets in comparison to DNN single-task models and also provides a scalable method to predict ADME-Tox properties from heterogeneous data. For example, predictive quality using external validation sets was improved from R2 of 0.6 to 0.7 comparing single-task and multitask DNN networks from human metabolic lability data. Besides statistical evaluation, a new visualization approach is introduced to interpret DNN models termed "response map", which is useful to detect local property gradients based on structure fragmentation and derivatization. This method is successfully applied to visualize fragmental contributions to guide further design in drug discovery programs, as illustrated by CRCX3 antagonists and renin inhibitors, resp.
- 5Klucznik, T.; Mikulak-Klucznik, B.; McCormack, M. P.; Lima, H.; Szymkuć, S.; Bhowmick, M.; Molga, K.; Zhou, Y.; Rickershauser, L.; Gajewska, E. P.; Toutchkine, A.; Dittwald, P.; Startek, M. P.; Kirkovits, G. J.; Roszak, R.; Adamski, A.; Sieredzińska, B.; Mrksich, M.; Trice, S. L. J.; Grzybowski, B. A. Efficient Syntheses of Diverse, Medicinally Relevant Targets Planned by Computer and Executed in the Laboratory. Chem. 2018, 4, 522– 532, DOI: 10.1016/j.chempr.2018.02.0025Efficient Syntheses of Diverse, Medicinally Relevant Targets Planned by Computer and Executed in the LaboratoryKlucznik, Tomasz; Mikulak-Klucznik, Barbara; McCormack, Michael P.; Lima, Heather; Szymkuc, Sara; Bhowmick, Manishabrata; Molga, Karol; Zhou, Yubai; Rickershauser, Lindsey; Gajewska, Ewa P.; Toutchkine, Alexei; Dittwald, Piotr; Startek, Michal P.; Kirkovits, Gregory J.; Roszak, Rafal; Adamski, Ariel; Sieredzinska, Bianka; Mrksich, Milan; Trice, Sarah L. J.; Grzybowski, Bartosz A.Chem (2018), 4 (3), 522-532CODEN: CHEMVE; ISSN:2451-9294. (Cell Press)The Chematica program was used to autonomously design synthetic pathways to eight structurally diverse targets, including seven com. valuable bioactive substances and one natural product. All of these computer-planned routes were successfully executed in the lab. and offer significant yield improvements and cost savings over previous approaches, provide alternatives to patented routes, or produce targets that were not synthesized previously.
- 6Hoffmann, T.; Gastreich, M. The Next Level in Chemical Space Navigation: Going Far Beyond Enumerable Compound Libraries. Drug Discovery Today 2019, 24 (5), 1148– 1156, DOI: 10.1016/j.drudis.2019.02.0136The next level in chemical space navigation: going far beyond enumerable compound librariesHoffmann, Torsten; Gastreich, MarcusDrug Discovery Today (2019), 24 (5), 1148-1156CODEN: DDTOFS; ISSN:1359-6446. (Elsevier Ltd.)Recent innovations have brought pharmacophore-driven methods for navigating virtual chem. spaces, the size of which can reach into the billions of mols., to the fingertips of every chemist. There has been a paradigm shift in the underlying computational chem. that drives chem. space search applications, incorporating intelligent reaction knowledge into their core so that they can readily deliver com. available mols. as nearest neighbor hits from within giant virtual spaces. These vast resources enable medicinal chemists to execute rapid scaffold-hopping expts., rapid hit expansion, and structure-activity relationship (SAR) exploitation in largely intellectual property (IP)-free territory and at unparalleled low cost.
- 7Howes, L. Hunting for Drugs in Chemical Space. Chem. Eng. News 2022, 100 (23), 20, DOI: 10.47287/cen-10023-coverThere is no corresponding record for this reference.
- 8Daina, A.; Michielin, O.; Zoete, V. SwissADME: A Free Web Tool to Evaluate Pharmacokinetics, Drug-likeness and Medicinal Chemistry Friendliness of Small Molecules. Sci. Rep 2017, 7, 42717, DOI: 10.1038/srep427178SwissADME: a free web tool to evaluate pharmacokinetics, drug-likeness and medicinal chemistry friendliness of small moleculesDaina Antoine; Michielin Olivier; Zoete Vincent; Michielin Olivier; Michielin OlivierScientific reports (2017), 7 (), 42717 ISSN:.To be effective as a drug, a potent molecule must reach its target in the body in sufficient concentration, and stay there in a bioactive form long enough for the expected biologic events to occur. Drug development involves assessment of absorption, distribution, metabolism and excretion (ADME) increasingly earlier in the discovery process, at a stage when considered compounds are numerous but access to the physical samples is limited. In that context, computer models constitute valid alternatives to experiments. Here, we present the new SwissADME web tool that gives free access to a pool of fast yet robust predictive models for physicochemical properties, pharmacokinetics, drug-likeness and medicinal chemistry friendliness, among which in-house proficient methods such as the BOILED-Egg, iLOGP and Bioavailability Radar. Easy efficient input and interpretation are ensured thanks to a user-friendly interface through the login-free website http://www.swissadme.ch. Specialists, but also nonexpert in cheminformatics or computational chemistry can predict rapidly key parameters for a collection of molecules to support their drug discovery endeavours.
- 9ISO 27001 Certification . 2022; https://www.iso.org/obp/ui/#iso:std:iso-iec:27001:ed-3:v1:en (accessed 2023–04–21).There is no corresponding record for this reference.
- 10Félix, E.; Landrum, G., Dalke, A. FPSim2: Simple package for fast molecular similarity searches . 2022; https://chembl.github.io/FPSim2/ (accessed 2023–04–20).There is no corresponding record for this reference.
- 11Kim, S.; Chen, J.; Cheng, T.; Gindulyte, A.; He, J.; He, S.; Li, Q.; Shoemaker, B. A.; Thiessen, P. A.; Yu, B.; Zaslavsky, L.; Zhang, J.; Bolton, E. E. PubChem 2023 Update. Nucleic Acids Res. 2023, 51 (D1), D1373– D1380, DOI: 10.1093/nar/gkac956There is no corresponding record for this reference.
- 12Mendez, D.; Gaulton, A.; Bento, A. P.; Chambers, J.; De Veij, M.; Félix, E.; Magariños, M. P.; Mosquera, J. F.; Mutowo, P.; Nowotka, M.; Gordillo-Marañón, M.; Hunter, F.; Junco, L.; Mugumbate, G.; Rodriguez-Lopez, M.; Atkinson, F.; Bosc, N.; Radoux, C. J.; Segura-Cabrera, A.; Leach, A. R.; Hersey, A. ChEMBL: Towards Direct Deposition of Bioassay Data. Nucleic Acids Res. 2019, 47 (D1), D930– D940, DOI: 10.1093/nar/gky107512ChEMBL: towards direct deposition of bioassay dataMendez, David; Gaulton, Anna; Bento, A. Patricia; Chambers, Jon; De Veij, Marleen; Felix, Eloy; Magarinos, Maria Paula; Mosquera, Juan F.; Mutowo, Prudence; Nowotka, Michal; Gordillo-Maranon, Maria; Hunter, Fiona; Junco, Laura; Mugumbate, Grace; Rodriguez-Lopez, Milagros; Atkinson, Francis; Bosc, Nicolas; Radoux, Chris J.; Cabrera, Aldo Segura; Hersey, Anne; Leach, Andrew R.Nucleic Acids Research (2019), 47 (D1), D930-D940CODEN: NARHAD; ISSN:1362-4962. (Oxford University Press)ChEMBL is a large, open-access bioactivity database, previously described in the 2012, 2014 and 2017 Nucleic Acids Research Database Issues. In the last two years, several important improvements have been made to the database and are described here. These include more robust capture and representation of assay details; a new data deposition system, allowing updating of data sets and deposition of supplementary data; and a completely redesigned web interface, with enhanced search and filtering capabilities.
- 13Irwin, J. J.; Tang, K. G.; Young, J.; Dandarchuluun, C.; Wong, B. R.; Khurelbaatar, M.; Moroz, Y. S.; Mayfield, J.; Sayle, R. A. ZINC20-A Free Ultralarge-Scale Chemical Database for Ligand Discovery. J. Chem. Inf. Model. 2020, 60 (12), 6065– 6073, DOI: 10.1021/acs.jcim.0c0067513ZINC20-A Free Ultralarge-Scale Chemical Database for Ligand DiscoveryIrwin, John J.; Tang, Khanh G.; Young, Jennifer; Dandarchuluun, Chinzorig; Wong, Benjamin R.; Khurelbaatar, Munkhzul; Moroz, Yurii S.; Mayfield, John; Sayle, Roger A.Journal of Chemical Information and Modeling (2020), 60 (12), 6065-6073CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)Identifying and purchasing new small mols. to test in biol. assays are enabling for ligand discovery, but as purchasable chem. space continues to grow into the tens of billions based on inexpensive make-on-demand compds., simply searching this space becomes a major challenge. We have therefore developed ZINC20, a new version of ZINC with two major new features: billions of new mols. and new methods to search them. As a fully enumerated database, ZINC can be searched precisely using explicit at.-level graph-based methods, such as SmallWorld for similarity and Arthor for pattern and substructure search, as well as 3D methods such as docking. Anal. of the new make-on-demand compd. sets by these and related tools reveals startling features. For instance, over 97% of the core Bemis-Murcko scaffolds in make-on-demand libraries are unavailable from "in-stock" collections. Correspondingly, the no. of new Bemis-Murcko scaffolds is rising almost as a linear fraction of the elaborated mols. Thus, an 88-fold increase in the no. of mols. in the make-on-demand vs. the in-stock sets is built upon a 16-fold increase in the no. of Bemis-Murcko scaffolds. The make-on-demand library is also more structurally diverse than phys. libraries, with a massive increase in disk- and sphere-like shaped mols. The new system is freely available at zinc20.docking.org.
- 14Rarey, M.; Dixon, J. S. Feature Trees: A New Molecular Similarity Measure Based on Tree Matching. J. Comput. Aided. Mol. Des. 1998, 12, 471– 490, DOI: 10.1023/A:100806890462816Feature trees: a new molecular similarity measure based on tree matchingRarey, Matthias; Dixon, J. ScottJournal of Computer-Aided Molecular Design (1998), 12 (5), 471-490CODEN: JCADEQ; ISSN:0920-654X. (Kluwer Academic Publishers)In this paper we present a new method for evaluating mol. similarity between small org. compds. Instead of a linear representation like fingerprints, a more complex description, a feature tree, is calcd. for a mol. A feature tree represents hydrophobic fragments and functional groups of the mol. and the way these groups are linked together. Each node in the tree is labeled with a set of features representing chem. properties of the part of the mol. corresponding to the node. The comparison of feature trees is based on matching subtrees of two feature trees onto each other. Two algorithms for tackling the matching problem are described throughout this paper. On a dataset of about 1000 mols., we demonstrate the ability of our approach to identify mols. belonging to the same class of inhibitors. With a second dataset of 58 mols. with known binding modes taken from the Brookhaven Protein Data Bank, we show that the matchings produced by our algorithms are compatible with the relative orientation of the mols. in the active site in 61% of the test cases. The av. computation time for a pair comparison is about 50 ms on a current workstation.
- 15Boehm, M.; Wu, T.-Y.; Claussen, H.; Lemmen, C. Similarity Searching and Scaffold Hopping in Synthetically Accessible Combinatorial Chemistry Spaces. J. Med. Chem. 2008, 51, 2468– 2480, DOI: 10.1021/jm070772717Similarity searching and scaffold hopping in synthetically accessible combinatorial chemistry spacesBoehm, Markus; Wu, Tong-Ying; Claussen, Holger; Lemmen, ChristianJournal of Medicinal Chemistry (2008), 51 (8), 2468-2480CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Large collections of combinatorial libraries are an integral element in today's pharmaceutical industry. It is of great interest to perform similarity searches against all virtual compds. that are synthetically accessible by any such library. Here we describe the successful application of a new software tool CoLibri on 358 combinatorial libraries based on validated reaction protocols to create a single chem. space contg. over 1012 possible products. Similarity searching with FTrees-FS allows the systematic exploration of this space without the need to enumerate all product structures. The search result is a set of virtual hits which are synthetically accessible by one or more of the existing reaction protocols. Grouping these virtual hits by their synthetic protocols allows the rapid design and synthesis of multiple follow-up libraries. Such library ideas support hit-to-lead design efforts for tasks like follow-up from high-throughput screening hits or scaffold hopping from one hit to another attractive series.
- 16Enamine REAL Space. https://enamine.net/compound-collections/real-compounds/real-space-navigator (accessed 2023-04–21).There is no corresponding record for this reference.
- 17WuXi GalaXi. https://wuxibiology.com/drug-discovery-services/hit-finding-and-screening-services/virtual-screening/ (accessed 2023-04–21).There is no corresponding record for this reference.
- 18Blaschke, T.; Arús-Pous, J.; Chen, H.; Margreitter, C.; Tyrchan, C.; Engkvist, O.; Papadopoulos, K.; Patronov, A. REINVENT 2.0: An AI Tool for De Novo Drug Design. J. Chem. Inf. Model. 2020, 60 (12), 5918– 5922, DOI: 10.1021/acs.jcim.0c0091518REINVENT 2.0: An AI Tool for De Novo Drug DesignBlaschke, Thomas; Arus-Pous, Josep; Chen, Hongming; Margreitter, Christian; Tyrchan, Christian; Engkvist, Ola; Papadopoulos, Kostas; Patronov, AtanasJournal of Chemical Information and Modeling (2020), 60 (12), 5918-5922CODEN: JCISD8; ISSN:1549-9596. (American Chemical Society)In the past few years, we have witnessed a renaissance of the field of mol. de novo drug design. The advancements in deep learning and artificial intelligence (AI) have triggered an avalanche of ideas on how to translate such techniques to a variety of domains including the field of drug design. A range of architectures have been devised to find the optimal way of generating chem. compds. by using either graph- or string (SMILES)-based representations. With this application note, we aim to offer the community a prodn.-ready tool for de novo design, called REINVENT. It can be effectively applied on drug discovery projects that are striving to resolve either exploration or exploitation problems while navigating the chem. space. It can facilitate the idea generation process by bringing to the researcher's attention the most promising compds. REINVENT's code is publicly available at https://github.com/MolecularAI/Reinvent.
- 19Bickerton, G. R.; Paolini, G. V.; Besnard, J.; Muresan, S.; Hopkins, A. L. Quantifying the Chemical Beauty of Drugs. Nat. Chem. 2012, 4, 90– 98, DOI: 10.1038/nchem.124319Quantifying the chemical beauty of drugsBickerton, G. Richard; Paolini, Gaia V.; Besnard, Jeremy; Muresan, Sorel; Hopkins, Andrew L.Nature Chemistry (2012), 4 (2), 90-98CODEN: NCAHBB; ISSN:1755-4330. (Nature Publishing Group)Drug-likeness is a key consideration when selecting compds. during the early stages of drug discovery. However, evaluation of drug-likeness in abs. terms does not reflect adequately the whole spectrum of compd. quality. More worryingly, widely used rules may inadvertently foster undesirable mol. property inflation as they permit the encroachment of rule-compliant compds. towards their boundaries. We propose a measure of drug-likeness based on the concept of desirability called the quant. est. of drug-likeness (QED). The empirical rationale of QED reflects the underlying distribution of mol. properties. QED is intuitive, transparent, straightforward to implement in many practical settings and allows compds. to be ranked by their relative merit. We extended the utility of QED by applying it to the problem of mol. target druggability assessment by prioritizing a large set of published bioactive compds. The measure may also capture the abstr. notion of aesthetics in medicinal chem.
- 20Cheeseright, T. Fragment hopping with Blaze. Shape Match 2022 (accessed 2023–04–26).There is no corresponding record for this reference.
- 21Narwal, M.; Venkannagari, H.; Lehtiö, L. Structural Basis of Selective Inhibition of Human Tankyrases. J. Med. Chem. 2012, 55 (3), 1360– 1367, DOI: 10.1021/jm201510p21Structural Basis of Selective Inhibition of Human TankyrasesNarwal, Mohit; Venkannagari, Harikanth; Lehtio, LariJournal of Medicinal Chemistry (2012), 55 (3), 1360-1367CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Tankyrases are poly(ADP-ribose) polymerases that have many cellular functions. They play pharmaceutically important roles, at least in telomere homeostasis and Wnt signaling, by covalently ADP-ribosylating target proteins and consequently regulating their functions. These features make tankyrases potential targets for treatment of cancer. We report here crystal structures of human tankyrase 2 catalytic fragment in complex with a byproduct, nicotinamide, and with selective inhibitors of tankyrases (IWR-1) and PARPs 1 and 2 (olaparib). Binding of these inhibitors to tankyrase 2 induces specific conformational changes. The crystal structures explain the selectivity of the inhibitors, reveal the flexibility of a substrate binding loop, and explain existing structure-activity relationship data. The first crystal structure of a PARP enzyme in complex with a potent inhibitor, IWR-1, that does not bind to the widely utilized nicotinamide-binding site makes the structure valuable for development of PARP inhibitors in general.
- 22Berman, H. M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T. N.; Weissig, H.; Shindyalov, I. N.; Bourne, P. E. The Protein Data Bank. Nucleic Acids Res. 2000, 28, 235– 242, DOI: 10.1093/nar/28.1.23522The Protein Data BankBerman, Helen M.; Westbrook, John; Feng, Zukang; Gilliland, Gary; Bhat, T. N.; Weissig, Helge; Shindyalov, Ilya N.; Bourne, Philip E.Nucleic Acids Research (2000), 28 (1), 235-242CODEN: NARHAD; ISSN:0305-1048. (Oxford University Press)The Protein Data Bank (PDB; http://www.rcsb.org/pdb/)is the single worldwide archive of structural data of biol. macromols. This paper describes the goals of the PDB, the systems in place for data deposition and access, how to obtain further information, and near-term plans for the future development of the resource.
- 23Volkamer, A.; Kuhn, D.; Rippmann, F.; Rarey, M. DoGSiteScorer: A Web Server for Automatic Binding Site Prediction, Analysis and Druggability Assessment, Analysis and Druggability Assessment. Bioinformatics 2012, 28 (15), 2074– 5, DOI: 10.1093/bioinformatics/bts31023DoGSiteScorer: a web server for automatic binding site prediction, analysis and druggability assessmentVolkamer, Andrea; Kuhn, Daniel; Rippmann, Friedrich; Rarey, MatthiasBioinformatics (2012), 28 (15), 2074-2075CODEN: BOINFP; ISSN:1367-4803. (Oxford University Press)Motivation: Many drug discovery projects fail because the underlying target is finally found to be undruggable. Progress in structure elucidation of proteins now opens up a route to automatic structure-based target assessment. DoGSiteScorer is a newly developed automatic tool combining pocket prediction, characterization and druggability estn. and is now available through a web server. Availability: The DoGSiteScorer web server is freely available for academic use at http://dogsite.zbh.uni-hamburg.de Contact: [email protected].
- 24Cresset Flare Docking . 2022; https://www.cresset-group.com/software/flare-docking/ (accessed 2023–04–21).There is no corresponding record for this reference.
- 25Shang, S.; Hua, F.; Hu, A.-W. The Regulation of β-Catenin Activity and Function in Cancer: Therapeutic Opportunities. Oncotarget 2017, 8 (20), 33972– 33989, DOI: 10.18632/oncotarget.1568725The regulation of β-catenin activity and function in cancer: therapeutic opportunitiesShang Shuang; Hua Fang; Hu Zhuo-WeiOncotarget (2017), 8 (20), 33972-33989 ISSN:.Wnt/β-catenin signaling is an evolutionarily conserved and versatile pathway that is known to be involved in embryonic development, tissue homeostasis and a wide variety of human diseases. Aberrant activation of this pathway gives rise to the accumulation of β-catenin in the nucleus and promotes the transcription of many oncogenes such as c-Myc and CyclinD-1. As a result, it contributes to carcinogenesis and tumor progression of several cancers, including colon cancer, hepatocellular carcinoma, pancreatic cancer, lung cancer and ovarian cancer. β-Catenin is a pivotal component of the Wnt signaling pathway and it is tightly regulated at three hierarchical levels: protein stability, subcellular localization and transcriptional activity. Uncovering the regulatory mechanisms of β-catenin will provide new insights into the pathogenesis of cancer and other diseases, as well as new therapeutic strategies against these diseases. In this review we dissect the concrete regulatory mechanisms of β-catenin from three aspects mentioned above. Then we focus on the role of β-catenin in cancer initiation, progression, dormancy, immunity and cancer stem cell maintenance. At last, we summarize the recent progress in the development of agents for the pharmacological modulation of β-catenin activity in cancer therapy.
- 26Wong, C. M.; Fan, S. T.; Ng, I. O. beta-Catenin mutation and overexpression in hepatocellular carcinoma: clinicopathologic and prognostic significance. Cancer 2001, 92 (1), 136– 145, DOI: 10.1002/1097-0142(20010701)92:1<136::AID-CNCR1301>3.0.CO;2-R26β-catenin mutation and overexpression in hepatocellular carcinoma: Clinicopathologic and prognostic significanceWong, Chun M.; Fan, Sheung T.; Ng, Irene O. L.Cancer (New York, NY, United States) (2001), 92 (1), 136-145CODEN: CANCAR; ISSN:0008-543X. (John Wiley & Sons, Inc.)β-Catenin has been recognized as a crit. member of the Wnt signaling pathway, and inappropriate activation of this pathway has been implicated in carcinogenesis. To det. the clin. significance of β-catenin in hepatocellular carcinoma (HCC), we performed mutational anal. at exon 3 of the gene, investigated the subcellular protein expression, and analyzed their clinicopathol. and prognostic significance in 60 patients with resected primary HCC. By direct DNA sequencing, somatic mutations of the β-catenin gene were detected in 7 (12%) HCCs. All the mutations were found at the region (exon 3) responsible for phosphorylation and ubiquitination, therefore likely to result in stabilization of free cytoplasmic β-catenin. Nuclear accumulation of the β-catenin protein, similar to the response to the Wnt signal, was found in 10 (17%) HCCs and was closely assocd. with gene mutation (P < 0.001). In the remaining cases, nonnuclear type overexpression, i.e., overexpression in the cytoplasm and/or cytoplasmic membrane, was obsd. in 31 (62%) HCCs, thus suggesting that the mechanisms leading to β-catenin overexpression may be heterogeneous. HCCs with a nonnuclear type of β-catenin overexpression were more frequently larger than 5 cm in diam. (P = 0.022) and had poorer cellular differentiation (P = 0.037), and the patients had significantly shorter disease-free survival lengths (P = 0.041). Review of the data from previous studies in HCC showed that β-catenin mutations were more frequent in HCV-assocd. HCC than in HBV-assocd. ones. β-Catenin mutation and deregulation may play an important role in hepatocarcinogenesis. Nonnuclear type β-catenin overexpression appeared to have pathol. and prognostic significance.
- 27Brabletz, T.; Jung, A.; Hermann, K.; Günther, K.; Hohenberger, W.; Kirchner, T. Nuclear overexpression of the oncoprotein beta-catenin in colorectal cancer is localized predominantly at the invasion front. Pathology, research and practice 1998, 194 (10), 701– 704, DOI: 10.1016/S0344-0338(98)80129-527Nuclear overexpression of the oncoprotein β-catenin in colorectal cancer is localized predominantly at the invasion frontBrabletz, Thomas; Jung, Andreas; Hermann, Kathrin; Guenther, Klaus; Hohenberger, Werner; Kirchner, ThomasPathology, Research and Practice (1998), 194 (10), 701-704CODEN: PARPDS; ISSN:0344-0338. (Gustav Fischer Verlag)The distribution of overexpressed β-catenin within individual colorectal carcinomas was investigated using immunohistochem. In the most tumors a strong nuclear expression of β-catenin was found, predominantly localized at the invasion front with strongest nuclear staining of isolated, scattered tumor cells. In contrast, cells in the tumor center often showed a membranous expression of β-catenin, comparable to normal colon epithelium. It is therefore likely, that in addn. to the overexpression of β-catenin caused by defects in the adenomatous polyposis coli (APC) locus, regulatory events in the tumor itself lead to a different distribution of this oncoprotein.
- 28Shigemitsu, K.; Sekido, Y.; Usami, N.; Mori, S.; Sato, M.; Horio, Y.; Hasegawa, Y.; Bader, S.; Gazdar, A.; Minna, J.; Hida, T.; Yoshioka, H.; Imaizumi, M.; Ueda, Y.; Takahashi, M.; Shimokata, K. Genetic alteration of the beta-catenin gene (CTNNB1) in human lung cancer and malignant mesothelioma and identification of a new 3p21.3 homozygous deletion. Oncogene 2001, 20 (31), 4249– 4257, DOI: 10.1038/sj.onc.120455728Genetic alteration of the β-catenin gene (CTNNB1) in human lung cancer and malignant mesothelioma and identification of a new 3p21.3 homozygous deletionShigemitsu, Kikuo; Sekido, Yoshitaka; Usami, Noriyasu; Mori, Shoichi; Sato, Mitsuo; Horio, Yoshitsugu; Hasegawa, Yoshinori; Bader, Scott A.; Gazdar, Adi F.; Minna, John D.; Hida, Toyoaki; Yoshioka, Hiromu; Imaizumi, Munehisa; Ueda, Yuichi; Takahashi, Masahide; Shimokata, KaoruOncogene (2001), 20 (31), 4249-4257CODEN: ONCNES; ISSN:0950-9232. (Nature Publishing Group)The β-catenin gene (CTNNB1) has been shown to be genetically mutated in various human malignancies. To det. whether the β-catenin gene is responsible for oncogenesis in thoracic malignancies, the authors searched for the mutation in 166 lung cancers (90 primary tumors and 76 cell lines), one blastoma and 10 malignant mesotheliomas (two primary tumors and eight cell lines). Among the lung cancers, including 43 small cell lung cancers (SCLCs) and 123 non-small cell lung cancers (NSCLCs), the authors identified four alterations in exon 3, which is the target region of mutation for stabilizing β-catenin. One primary adenocarcinoma had a somatic mutation from C to G, leading to an amino acid substitution from Ser to Cys at codon 37. Among the cell lines, SCLC NCI-H1092 had a mutation from A to G, leading to an Asp to Gly substitution at codon 6, NSCLC HCC15 had a mutation from C to T, leading to a Ser to Phe substitution at codon 45, and NSCLC NCI-H358 had a mutation from A to G, leading to a Thr to Ala substitution at codon 75. One blastoma also had a somatic mutation from C to G, leading to a Ser to Cys substitution at codon 37. Among the 10 malignant mesotheliomas, the authors identified a homozygous deletion in the NCI-H28 cell line. Cloning of the rearranged fragment from NCI-H28 indicated that all the exons except exon 1 of the β-catenin gene are deleted and that the deletion junction is 13 kb downstream from exon 1. Furthermore, Northern blot anal. of 26 lung cancer and eight mesothelioma cell line RNAs detected ubiquitous expression of the β-catenin messages except NCI-H28, although Western blot anal. showed that relatively less amts. of protein products were expressed in some of lung cancer cell lines. The authors' findings suggest that the β-catenin gene is infrequently mutated in lung cancer and that the NCI-H28 homozygous deletion of the β-catenin gene might indicate the possibility of a new tumor suppressor gene residing in this region at 3p21.3, where various types of human cancers show frequent allelic loss.
- 29Li, K.; Pan, W.-T.; Ma, Y.-B.; Xu, X.-L.; Gao, Y.; He, Y.-Q.; Wei, L.; Zhang, J.-W. BMX activates Wnt/β-catenin signaling pathway to promote cell proliferation and migration in breast cancer. Breast Cancer 2020, 27 (3), 363– 371, DOI: 10.1007/s12282-019-01024-829BMX activates Wnt/β-catenin signaling pathway to promote cell proliferation and migration in breast cancerLi Kai; Pan Wen-Ting; Ma Yan-Bin; Xu Xiao-Long; Gao Yang; He Yan-Qi; Wei Lei; Zhang Jing-WeiBreast cancer (Tokyo, Japan) (2020), 27 (3), 363-371 ISSN:.BACKGROUND: Breast cancer has become a dangerous killer for the female, which seriously threatened women's life, leading to huge pressures to society. The present study assessed the mechanism underlying the involvement of bone marrow tyrosine kinase on chromosome X (BMX) in breast cancer development. METHODS: The expression of BMX was examined by qPCR and immunohistochemistry. The effect of BMX on cell proliferation and migration was detected by Clone formation assay and Transwell assay. In vitro study, the correlation of BMX with Wnt/β-catenin pathway was explored by western blot and TOP/FOP flash assay. RESULTS: In the present study, we found that BMX was up-regulated in breast cancer, which was associated with the tumor differentiation and TNM stage. Oncogenic BMX enhanced the ability of breast cancer cell proliferation and migration. Furthermore, BMX could up-regulate the protein expression levels of p-β-catenin (Y142), p-β-catenin(Y654) and inhibit the expression level of p-β-catenin (S33/37), thus activating Wnt/β-catenin pathway in MCF-7 and MDA-MB-231 cells. In addition, we revealed that BMX promoted GSK3β phosphorylation, which suppressed the degradation of β-catenin. CONCLUSIONS: In this study, we identified that BMX-activated Wnt/β-catenin signaling pathway, playing an oncogenic role in breast cancer, suggesting that BMX could become a potential treatment target of breast cancer.
- 30Zyla, R. E.; Olkhov-Mitsel, E.; Amemiya, Y.; Bassiouny, D.; Seth, A.; Djordjevic, B.; Nofech-Mozes, S.; Parra-Herran, C. CTNNB1Mutations and Aberrant β-Catenin Expression in Ovarian Endometrioid Carcinoma: Correlation With Patient Outcome. American journal of surgical pathology 2021, 45 (1), 68– 76, DOI: 10.1097/PAS.000000000000155330CTNNB1 Mutations and Aberrant β-Catenin Expression in Ovarian Endometrioid Carcinoma: Correlation With Patient OutcomeZyla Roman E; Seth Arun; Djordjevic Bojana; Nofech-Mozes Sharon; Parra-Herran Carlos; Zyla Roman E; Olkhov-Mitsel Ekaterina; Bassiouny Dina; Seth Arun; Djordjevic Bojana; Nofech-Mozes Sharon; Parra-Herran Carlos; Amemiya Yutaka; Seth Arun; Bassiouny DinaThe American journal of surgical pathology (2021), 45 (1), 68-76 ISSN:.CTNNB1 mutations and aberrant β-catenin expression have adverse prognosis in endometrial endometrioid carcinoma, and recent evidence suggests a prognostic role of β-catenin in ovarian endometrioid carcinoma. Thus, we aimed to determine the prognostic value of the CTNNB1 mutational status, and its correlation with β-catenin expression, in a well-annotated cohort of 51 ovarian endometrioid carcinomas. We performed immunohistochemistry for β-catenin and developed an 11-gene next-generation sequencing panel that included whole exome sequencing of CTNNB1 and TP53. Results were correlated with clinicopathologic variables including disease-free and disease-specific survival. Tumor recurrence was documented in 14 patients (27%), and cancer-related death in 8 patients (16%). CTNNB1 mutations were found in 22 cases (43%), and nuclear β-catenin in 26 cases (51%). CTNNB1 mutation highly correlated with nuclear β-catenin (P<0.05). Mutated CTNNB1 status was statistically associated with better disease-free survival (P=0.04, log-rank test) and approached significance for better disease-specific survival (P=0.07). It also correlated with earlier International Federation of Gynecology and Obstetrics stage (P<0.05). Nuclear β-catenin, TP53 mutations, age, ProMisE group, surface involvement, tumor grade and stage also correlated with disease-free survival. There was no association between membranous β-catenin expression and disease-free or disease-specific survival. CTNNB1 mutations and nuclear β-catenin expression are associated with better progression-free survival in patients with OEC. This relationship may be in part due to a trend of CTNNB1-mutated tumors to present at early stage. β-catenin immunohistochemistry may serve as a prognostic biomarker and a surrogate for CTNN1B mutations in the evaluation of patients with ovarian endometrioid neoplasia, particularly those in reproductive-age or found incidentally without upfront staging surgery.
- 31Ferri, M.; Liscio, P.; Carotti, A.; Asciutti, S.; Sardella, R.; Macchiarulo, A.; Camaioni, E. Targeting Wnt-driven Cancers: Discovery of Novel Tankyrase Inhibitors. Eur. J. Med. Chem. 2017, 142, 506– 522, DOI: 10.1016/j.ejmech.2017.09.03031Targeting Wnt-driven cancers: Discovery of novel tankyrase inhibitorsFerri, Martina; Liscio, Paride; Carotti, Andrea; Asciutti, Stefania; Sardella, Roccaldo; Macchiarulo, Antonio; Camaioni, EmidioEuropean Journal of Medicinal Chemistry (2017), 142 (), 506-522CODEN: EJMCA5; ISSN:0223-5234. (Elsevier Masson SAS)A review. Recent years have seen substantially heightened interest in the discovery of tankyrase inhibitors (TNKSi) as new promising anticancer agents. In this framework, the aim of this review article is focused on the description of potent TNKSi also endowed with disruptor activity toward the Wnt/β-catenin signaling pathway. Beginning with an overview of the most characterized TNKSi deriving from several drug design approaches and classifying them on the basis of the mol. interactions with the target, the authors discuss only those ones acting against Wnt cancer cell lines. In addn., comprehensive structure property relationships (SPR) emerging from the hit evolution processes and preclin. results are provided. The authors then review the most promising TNKSi hitherto reported in literature, acting in vivo models of Wnt-driven cancers. Some out-looks on current issues and future directions in this field are also discussed.
- 32Buchstaller, H. P.; Anlauf, U.; Dorsch, D.; Kögler, S.; Kuhn, D.; Lehmann, M.; Leuthner, B.; Lodholz, S.; Musil, D.; Radtki, D.; Rettig, C.; Ritzert, C.; Rohdich, F.; Schneider, R.; Wegener, A.; Weigt, S.; Wilkinson, K.; Esdar, C. Optimization of a Screening Hit toward M2912, an Oral Tankyrase Inhibitor with Antitumor Activity in Colorectal Cancer Models. J. Med. Chem. 2021, 64 (14), 10371– 10392, DOI: 10.1021/acs.jmedchem.1c0080032Optimization of a Screening Hit toward M2912, an Oral Tankyrase Inhibitor with Antitumor Activity in Colorectal Cancer ModelsBuchstaller, Hans-Peter; Anlauf, Uwe; Dorsch, Dieter; Koegler, Sarah; Kuhn, Daniel; Lehmann, Martin; Leuthner, Birgitta; Lodholz, Sara; Musil, Djordje; Radtki, Daniela; Rettig, Corinna; Ritzert, Claudio; Rohdich, Felix; Schneider, Richard; Wegener, Ansgar; Weigt, Stefan; Wilkinson, Kai; Esdar, ChristinaJournal of Medicinal Chemistry (2021), 64 (14), 10371-10392CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)The identification of a screening hit series and its optimization through scaffold hopping and SAR exploration was described. The systematic assessment delivered M2912 I a compd. with an optimal balance between excellent TNKS potency, exquisite PARP selectivity, and a predicted human PK compatible with once daily oral dosing. Modulation of cellular Wnt pathway activity and significant tumor growth inhibition was demonstrated with this compd. in colorectal xenograft models in vivo.
- 33Buchstaller, H. P.; Anlauf, U.; Dorsch, D.; Kuhn, D.; Lehmann, M.; Leuthner, B.; Musil, D.; Radtki, D.; Ritzert, C.; Rohdich, F.; Schneider, R.; Esdar, C. Discovery and Optimization of 2-Arylquinazolin-4-ones into a Potent and Selective Tankyrase Inhibitor Modulating Wnt Pathway Activity. J. Med. Chem. 2019, 62 (17), 7897– 7909, DOI: 10.1021/acs.jmedchem.9b0065633Discovery and Optimization of 2-Arylquinazolin-4-ones into a Potent and Selective Tankyrase Inhibitor Modulating Wnt Pathway ActivityBuchstaller, Hans-Peter; Anlauf, Uwe; Dorsch, Dieter; Kuhn, Daniel; Lehmann, Martin; Leuthner, Birgitta; Musil, Djordje; Radtki, Daniela; Ritzert, Claudio; Rohdich, Felix; Schneider, Richard; Esdar, ChristinaJournal of Medicinal Chemistry (2019), 62 (17), 7897-7909CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Tankyrases 1 and 2 (TNKS1/2) are promising pharmacol. targets which recently gained interest for anticancer therapy in Wnt pathway dependent tumors. 2-Aryl-quinazolinones were identified and optimized into potent tankyrase inhibitors through SAR exploration around the quinazolinone core and the 4'-position of the Ph residue. These efforts were supported by anal. of TNKS X-ray and Watermap structures and resulted in compd. 5k(I), a potent, selective tankyrase inhibitor with favorable pharmacokinetic properties. The X-ray structure of I in complex with TNKS1 was solved and confirmed the design hypothesis. Modulation of Wnt pathway activity was demonstrated with this compd. in a colorectal xenograft model in vivo.
- 34Mehta, C. C.; Bhatt, H. G. Tankyrase inhibitors as Antitumor Agents: a Patent Update (2013–2020). Expert Opinion on Therapeutic Patents 2021, 31 (7), 645– 661, DOI: 10.1080/13543776.2021.188892934Tankyrase inhibitors as antitumor agents: a patent update (2013 - 2020)Mehta, Chirag C.; Bhatt, Hardik G.Expert Opinion on Therapeutic Patents (2021), 31 (7), 645-661CODEN: EOTPEG; ISSN:1354-3776. (Taylor & Francis Ltd.)IntroductionTankyrase inhibitors gained significant attention as therapeutic targets in oncol. because of their potency. Their primary role in inhibiting the Wnt signaling pathway makes them an important class of compds. with the potential to be used as a combination therapy in future treatments of colorectal cancer. Areas coveredThis review describes pertinent work in the development of tankyrase inhibitors with a great emphasis on the recently patented TNKS inhibitors published from 2013 to 2020. This article also highlights a couple of promising candidates having tankyrase inhibitory effects and are currently undergoing clin. trials. Expert opinionFollowing the successful clin. applications of PARP inhibitors, tankyrase inhibition has gained significant attention in the research community as a target with high therapeutic potential. The ubiquitous role of tankyrase in cellular homeostasis and Wnt-dependent tumor proliferation brought difficulties for researchers to strike the right balance between potency and on-target toxicity. The need for novel tankyrase inhibitors with a better ADMET profile can introduce an addnl. regimen in treating various malignancies in monotherapy or adjuvant therapy. The development of combination therapies, including tankyrase inhibitors with or without PARP inhibitory properties, can potentially benefit the larger population of patients with unmet medical needs.
- 35Karlberg, T.; Markova, N.; Johansson, I.; Hammarström, M.; Schütz, P.; Weigelt, J.; Schüler, H. Structural Basis for the Interaction between Tankyrase-2 and a Potent Wnt-Signaling Inhibitor. J. Med. Chem. 2010, 53 (14), 5352– 5355, DOI: 10.1021/jm100249w35Structural Basis for the Interaction between Tankyrase-2 and a Potent Wnt-Signaling InhibitorKarlberg, Tobias; Markova, Natalia; Johansson, Ida; Hammarstrom, Martin; Schutz, Patrick; Weigelt, Johan; Schuler, HerwigJournal of Medicinal Chemistry (2010), 53 (14), 5352-5355CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)We report two crystal structures of the PARP domain of human tankyrase-2 (TNKS2). Tankyrases are involved in fundamental cellular processes such as telomere homeostasis and Wnt signaling. The complex of TNKS2 with the potent inhibitor XAV939 provides insights into the mol. basis of the strong interaction and suggests routes for further development of tankyrase inhibitors.
- 36Narwal, M.; Haikarainen, T.; Fallarero, A.; Vuorela, P. M.; Lehtiö, L. Screening and Structural Analysis of Flavones Inhibiting Tankyrases. J. Med. Chem. 2013, 56 (9), 3507– 3517, DOI: 10.1021/jm301878336Screening and Structural Analysis of Flavones Inhibiting TankyrasesNarwal, Mohit; Haikarainen, Teemu; Fallarero, Adyary; Vuorela, Pia M.; Lehtio, LariJournal of Medicinal Chemistry (2013), 56 (9), 3507-3517CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Flavonoids are known for their beneficial effects on human health, and therefore the therapeutic potential of these compds. have been extensively studied. Flavone has been previously identified as a tankyrase inhibitor, and to further elucidate whether tankyrases would be inhibited by other flavonoids, the authors performed a systematic screening of tankyrase 2 inhibitory activity using 500 natural and naturally derived flavonoids covering nine different flavonoid classes. All identified tankyrase inhibitors were flavones. The authors report crystal structures of all the hit compds. in complex with the catalytic domain of human tankyrase 2. Flavone derivs. in all 10 crystal structures bind to the nicotinamide binding site of tankyrase 2. Potencies of the active flavones toward tankyrases vary between 50 nM and 1.1 μM, and flavones show up to 200-fold selectivity for tankyrases over ARTD1. The mol. details of the interactions revealed by cocrystal structures efficiently describe the properties of potent flavone derivs. inhibiting tankyrases.
- 37Narwal, M.; Koivunen, J.; Haikarainen, T.; Obaji, E.; Legala, O. E.; Venkannagari, H.; Joensuu, P.; Pihlajaniemi, T.; Lehtiö, L. Discovery of Tankyrase Inhibiting Flavones with Increased Potency and Isoenzyme Selectivity. J. Med. Chem. 2013, 56 (20), 7880– 7889, DOI: 10.1021/jm401463y37Discovery of Tankyrase Inhibiting Flavones with Increased Potency and Isoenzyme SelectivityNarwal, Mohit; Koivunen, Jarkko; Haikarainen, Teemu; Obaji, Ezeogo; Legala, Ongey E.; Venkannagari, Harikanth; Joensuu, Paivi; Pihlajaniemi, Taina; Lehtio, LariJournal of Medicinal Chemistry (2013), 56 (20), 7880-7889CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Tankyrases are ADP-ribosyltransferases that play key roles in various cellular pathways, including the regulation of cell proliferation, and thus, they are promising drug targets for the treatment of cancer. Flavones have been shown to inhibit tankyrases and we report here the discovery of more potent and selective flavone derivs. Com. available flavones with single substitutions were used for structure-activity relation studies, and cocrystal structures of the 18 hit compds. were analyzed to explain their potency and selectivity. The most potent inhibitors were also tested in a cell-based assay, which demonstrated that they effectively antagonize Wnt signaling. To assess selectivity, they were further tested against a panel of homologous human ADP-ribosyltransferases. The most effective compd.(I; MN-64), showed 6 nM potency against tankyrase 1, isoenzyme selectivity, and Wnt signaling inhibition. This work forms a basis for rational development of flavones as tankyrase inhibitors and guides the development of other structurally related inhibitors.
- 38Sorkun, M. C.; Mullaj, D.; Koelman, J. M. V. A.; Er, S. ChemPlot, A Python Library for Chemical Space Visualization. Chemistry-Europe 2022, 2 (7), e202200005 DOI: 10.1002/cmtd.202200005There is no corresponding record for this reference.