SwePub
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "WFRF:(Bjerrum Esben Jannik) "

Sökning: WFRF:(Bjerrum Esben Jannik)

  • Resultat 1-11 av 11
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Fialková, Vendy, et al. (författare)
  • LibINVENT: Reaction-based Generative Scaffold Decoration for in Silico Library Design
  • 2022
  • Ingår i: Journal of Chemical Information and Modeling. - : American Chemical Society (ACS). - 1549-960X .- 1549-9596. ; 62:9, s. 2046-2063
  • Tidskriftsartikel (refereegranskat)abstract
    • Because of the strong relationship between the desired molecular activity and its structural core, the screening of focused, core-sharing chemical libraries is a key step in lead optimization. Despite the plethora of current research focused on in silico methods for molecule generation, to our knowledge, no tool capable of designing such libraries has been proposed. In this work, we present a novel tool for de novo drug design called LibINVENT. It is capable of rapidly proposing chemical libraries of compounds sharing the same core while maximizing a range of desirable properties. To further help the process of designing focused libraries, the user can list specific chemical reactions that can be used for the library creation. LibINVENT is therefore a flexible tool for generating virtual chemical libraries for lead optimization in a broad range of scenarios. Additionally, the shared core ensures that the compounds in the library are similar, possess desirable properties, and can also be synthesized under the same or similar conditions. The LibINVENT code is freely available in our public repository at https://github.com/MolecularAI/Lib-INVENT. The code necessary for data preprocessing is further available at: https://github.com/MolecularAI/Lib-INVENT-dataset.
  •  
2.
  • Gummesson Svensson, Hampus, 1996, et al. (författare)
  • Autonomous Drug Design with Multi-Armed Bandits
  • 2022
  • Ingår i: Proceedings - 2022 IEEE International Conference on Big Data, Big Data 2022. ; , s. 5584-5592
  • Konferensbidrag (refereegranskat)abstract
    • Recent developments in artificial intelligence and automation support a new drug design paradigm: autonomous drug design. Under this paradigm, generative models can provide suggestions on thousands of molecules with specific properties, and automated laboratories can potentially make, test and analyze molecules with minimal human supervision. However, since still only a limited number of molecules can be synthesized and tested, an obvious challenge is how to efficiently select among provided suggestions in a closed-loop system. We formulate this task as a stochastic multi-armed bandit problem with multiple plays, volatile arms and similarity information. To solve this task, we adapt previous work on multi-armed bandits to this setting, and compare our solution with random sampling, greedy selection and decaying-epsilon-greedy selection strategies. According to our simulation results, our approach has the potential to perform better exploration and exploitation of the chemical space for autonomous drug design.
  •  
3.
  • He, Jiazhen, et al. (författare)
  • Molecular optimization by capturing chemist’s intuition using deep neural networks
  • 2021
  • Ingår i: Journal of Cheminformatics. - : BioMed Central. - 1758-2946. ; 13:1
  • Tidskriftsartikel (refereegranskat)abstract
    • A main challenge in drug discovery is finding molecules with a desirable balance of multiple properties. Here, we focus on the task of molecular optimization, where the goal is to optimize a given starting molecule towards desirable properties. This task can be framed as a machine translation problem in natural language processing, where in our case, a molecule is translated into a molecule with optimized properties based on the SMILES representation. Typically, chemists would use their intuition to suggest chemical transformations for the starting molecule being optimized. A widely used strategy is the concept of matched molecular pairs where two molecules differ by a single transformation. We seek to capture the chemist’s intuition from matched molecular pairs using machine translation models. Specifically, the sequence-to-sequence model with attention mechanism, and the Transformer model are employed to generate molecules with desirable properties. As a proof of concept, three ADMET properties are optimized simultaneously: logD, solubility, and clearance, which are important properties of a drug. Since desirable properties often vary from project to project, the user-specified desirable property changes are incorporated into the input as an additional condition together with the starting molecules being optimized. Thus, the models can be guided to generate molecules satisfying the desirable properties. Additionally, we compare the two machine translation models based on the SMILES representation, with a graph-to-graph translation model HierG2G, which has shown the state-of-the-art performance in molecular optimization. Our results show that the Transformer can generate more molecules with desirable properties by making small modifications to the given starting molecules, which can be intuitive to chemists. A further enrichment of diverse molecules can be achieved by using an ensemble of models.
  •  
4.
  • He, Jiazhen, et al. (författare)
  • Transformer-based molecular optimization beyond matched molecular pairs
  • 2022
  • Ingår i: Journal of Cheminformatics. - : Springer Science and Business Media LLC. - 1758-2946 .- 1758-2946. ; 14:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Molecular optimization aims to improve the drug profile of a starting molecule. It is a fundamental problem in drug discovery but challenging due to (i) the requirement of simultaneous optimization of multiple properties and (ii) the large chemical space to explore. Recently, deep learning methods have been proposed to solve this task by mimicking the chemist's intuition in terms of matched molecular pairs (MMPs). Although MMPs is a widely used strategy by medicinal chemists, it offers limited capability in terms of exploring the space of structural modifications, therefore does not cover the complete space of solutions. Often more general transformations beyond the nature of MMPs are feasible and/or necessary, e.g. simultaneous modifications of the starting molecule at different places including the core scaffold. This study aims to provide a general methodology that offers more general structural modifications beyond MMPs. In particular, the same Transformer architecture is trained on different datasets. These datasets consist of a set of molecular pairs which reflect different types of transformations. Beyond MMP transformation, datasets reflecting general structural changes are constructed from ChEMBL based on two approaches: Tanimoto similarity (allows for multiple modifications) and scaffold matching (allows for multiple modifications but keep the scaffold constant) respectively. We investigate how the model behavior can be altered by tailoring the dataset while using the same model architecture. Our results show that the models trained on differently prepared datasets transform a given starting molecule in a way that it reflects the nature of the dataset used for training the model. These models could complement each other and unlock the capability for the chemists to pursue different options for improving a starting molecule.
  •  
5.
  • Irwin, Ross, et al. (författare)
  • Chemformer : a pre-trained transformer for computational chemistry
  • 2022
  • Ingår i: Machine Learning. - : IOP Publishing Ltd. - 2632-2153. ; 3:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Transformer models coupled with a simplified molecular line entry system (SMILES) have recently proven to be a powerful combination for solving challenges in cheminformatics. These models, however, are often developed specifically for a single application and can be very resource-intensive to train. In this work we present the Chemformer model-a Transformer-based model which can be quickly applied to both sequence-to-sequence and discriminative cheminformatics tasks. Additionally, we show that self-supervised pre-training can improve performance and significantly speed up convergence on downstream tasks. On direct synthesis and retrosynthesis prediction benchmark datasets we publish state-of-the-art results for top-1 accuracy. We also improve on existing approaches for a molecular optimisation task and show that Chemformer can optimise on multiple discriminative tasks simultaneously. Models, datasets and code will be made available after publication.
  •  
6.
  • Menke, Janosch, 1995, et al. (författare)
  • Metis: a python-based user interface to collect expert feedback for generative chemistry models
  • 2024
  • Ingår i: Journal of Cheminformatics. - 1758-2946 .- 1758-2946. ; 16:1
  • Tidskriftsartikel (refereegranskat)abstract
    • One challenge that current de novo drug design models face is a disparity between the user’s expectations and the actual output of the model in practical applications. Tailoring models to better align with chemists’ implicit knowledge, expectation and preferences is key to overcoming this obstacle effectively. While interest in preference-based and human-in-the-loop machine learning in chemistry is continuously increasing, no tool currently exists that enables the collection of standardized and chemistry-specific feedback. Metis is a Python-based open-source graphical user interface (GUI), designed to solve this and enable the collection of chemists’ detailed feedback on molecular structures. The GUI enables chemists to explore and evaluate molecules, offering a user-friendly interface for annotating preferences and specifying desired or undesired structural features. By providing chemists the opportunity to give detailed feedback, allows researchers to capture more efficiently the chemist’s implicit knowledge and preferences. This knowledge is crucial to align the chemist’s idea with the de novo design agents. The GUI aims to enhance this collaboration between the human and the “machine” by providing an intuitive platform where chemists can interactively provide feedback on molecular structures, aiding in preference learning and refining de novo design strategies. Metis integrates with the existing de novo framework REINVENT, creating a closed-loop system where human expertise can continuously inform and refine the generative models. Scientific contribution We introduce a novel Graphical User Interface, that allows chemists/researchers to give detailed feedback on substructures and properties of small molecules. This tool can be used to learn the preferences of chemists in order to align de novo drug design models with the chemist’s ideas. The GUI can be customized to fit different needs and projects and enables direct integration into de novo REINVENT runs. We believe that Metis can facilitate the discussion and development of novel ways to integrate human feedback that goes beyond binary decisions of liking or disliking a molecule.
  •  
7.
  • Mercado, Rocio, 1992, et al. (författare)
  • Exploring Graph Traversal Algorithms in Graph-Based Molecular Generation
  • 2022
  • Ingår i: Journal of Chemical Information and Modeling. - : American Chemical Society (ACS). - 1549-960X .- 1549-9596. ; 62:9, s. 2093-2100
  • Tidskriftsartikel (refereegranskat)abstract
    • Here, we explore the impact of different graph traversal algorithms on molecular graph generation. We do this by training a graph-based deep molecular generative model to build structures using a node order determined via either a breadth- or depth-first search algorithm. What we observe is that using a breadth-first traversal leads to better coverage of training data features compared to a depth-first traversal. We have quantified these differences using a variety of metrics on a data set of natural products. These metrics include percent validity, molecular coverage, and molecular shape. We also observe that by using either a breadth- or depth-first traversal it is possible to overtrain the generative models, at which point the results with either graph traversal algorithm are identical.
  •  
8.
  • Mercado, Rocio, 1992, et al. (författare)
  • Graph networks for molecular design
  • 2021
  • Ingår i: Machine Learning: Science and Technology. - : IOP Publishing. - 2632-2153. ; 2:2
  • Tidskriftsartikel (refereegranskat)abstract
    • Deep learning methods applied to chemistry can be used to accelerate the discovery of new molecules. This work introduces GraphINVENT, a platform developed for graph-based molecular design using graph neural networks (GNNs). GraphINVENT uses a tiered deep neural network architecture to probabilistically generate new molecules a single bond at a time. All models implemented in GraphINVENT can quickly learn to build molecules resembling the training set molecules without any explicit programming of chemical rules. The models have been benchmarked using the MOSES distribution-based metrics, showing how GraphINVENT models compare well with state-of-the-art generative models. This work compares six different GNN-based generative models in GraphINVENT, and shows that ultimately the gated-graph neural network performs best against the metrics considered here.
  •  
9.
  • Mercado, Rocio, 1992, et al. (författare)
  • Practical notes on building molecular graph generative models
  • 2020
  • Ingår i: Applied AI Letters. - : Wiley. - 2689-5595. ; 1:2
  • Tidskriftsartikel (övrigt vetenskapligt/konstnärligt)abstract
    • Here are presented technical notes and tips on developing graph generative models for molecular design. Although this work stems from the development of GraphINVENT, a Python platform for iterative molecular generation using graph neural networks, this work is relevant to researchers studying other architectures for graph-based molecular design. In this work, technical details that could be of interest to researchers developing their own molecular generative models are discussed, including an overview of previous work in graph-based molecular design and strategies for designing new models. Advice on development and debugging tools which are helpful during code development is also provided. Finally, methods that were tested but which ultimately did not lead to promising results in the development of GraphINVENT are described here in the hope that this will help other researchers avoid pitfalls in development and instead focus their efforts on more promising strategies for graph-based molecular generation.
  •  
10.
  • Shevtsov, Oleksii, 1988, et al. (författare)
  • A de novo molecular generation method using latent vector based generative adversarial network
  • 2019
  • Ingår i: Journal of Cheminformatics. - : Springer Science and Business Media LLC. - 1758-2946 .- 1758-2946. ; 11:1
  • Tidskriftsartikel (refereegranskat)abstract
    • Deep learning methods applied to drug discovery have been used to generate novel structures. In this study, we propose a new deep learning architecture, LatentGAN, which combines an autoencoder and a generative adversarial neural network for de novo molecular design. We applied the method in two scenarios: One to generate random drug-like compounds and another to generate target-biased compounds. Our results show that the method works well in both cases. Sampled compounds from the trained model can largely occupy the same chemical space as the training set and also generate a substantial fraction of novel compounds. Moreover, the drug-likeness score of compounds sampled from LatentGAN is also similar to that of the training set. Lastly, generated compounds differ from those obtained with a Recurrent Neural Network-based generative model approach, indicating that both methods can be used complementarily.[Figure not available: See fulltext.]
  •  
11.
  • Sundin, Iiris, et al. (författare)
  • Human-in-the-loop assisted de novo molecular design
  • 2022
  • Ingår i: Journal of Cheminformatics. - : Springer Science and Business Media LLC. - 1758-2946 .- 1758-2946. ; 14:1
  • Tidskriftsartikel (refereegranskat)abstract
    • A de novo molecular design workflow can be used together with technologies such as reinforcement learning to navigate the chemical space. A bottleneck in the workflow that remains to be solved is how to integrate human feedback in the exploration of the chemical space to optimize molecules. A human drug designer still needs to design the goal, expressed as a scoring function for the molecules that captures the designer’s implicit knowledge about the optimization task. Little support for this task exists and, consequently, a chemist usually resorts to iteratively building the objective function of multi-parameter optimization (MPO) in de novo design. We propose a principled approach to use human-in-the-loop machine learning to help the chemist to adapt the MPO scoring function to better match their goal. An advantage is that the method can learn the scoring function directly from the user’s feedback while they browse the output of the molecule generator, instead of the current manual tuning of the scoring function with trial and error. The proposed method uses a probabilistic model that captures the user’s idea and uncertainty about the scoring function, and it uses active learning to interact with the user. We present two case studies for this: In the first use-case, the parameters of an MPO are learned, and in the second use-case a non-parametric component of the scoring function to capture human domain knowledge is developed. The results show the effectiveness of the methods in two simulated example cases with an oracle, achieving significant improvement in less than 200 feedback queries, for the goals of a high QED score and identifying potent molecules for the DRD2 receptor, respectively. We further demonstrate the performance gains with a medicinal chemist interacting with the system. Graphical Abstract: [Figure not available: see fulltext.].
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-11 av 11

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy