Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters











Publication year range
1.
Clin Pathol ; 17: 2632010X241248909, 2024.
Article in English | MEDLINE | ID: mdl-38645837

ABSTRACT

Appendiceal diverticulitis is an uncommon condition that clinically resembles acute appendicitis. However, it is an incidental finding in histopathological studies and is rarely diagnosed preoperatively by imaging studies. In this article, we present the clinical and imaging findings of a male patient presenting with right upper quadrant pain with a preoperative imaging diagnosis of appendiceal diverticulitis. He underwent laparoscopic appendectomy and confirmed the diagnosis of appendiceal diverticulitis. This is a rare preoperative diagnosis. The management is often like typical appendicitis which is appendectomy. It is important to differentiate it from diverticulitis of the small intestine or colon because these diseases usually require only conservative treatment.

2.
J Phys Chem Lett ; 14(49): 10870-10879, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38032742

ABSTRACT

Accurate prediction of binding free energy changes upon mutations is vital for optimizing drugs, designing proteins, understanding genetic diseases, and cost-effective virtual screening. While machine learning methods show promise in this domain, achieving accuracy and generalization across diverse data sets remains a challenge. This study introduces Geometric Graph Learning for Protein-Protein Interactions (GGL-PPI), a novel approach integrating geometric graph representation and machine learning to forecast mutation-induced binding free energy changes. GGL-PPI leverages atom-level graph coloring and multiscale weighted colored geometric subgraphs to capture structural features of biomolecules, demonstrating superior performance on three standard data sets, namely, AB-Bind, SKEMPI 1.0, and SKEMPI 2.0 data sets. The model's efficacy extends to predicting protein thermodynamic stability in a blind test set, providing unbiased predictions for both direct and reverse mutations and showcasing notable generalization. GGL-PPI's precision in predicting changes in binding free energy and stability due to mutations enhances our comprehension of protein complexes, offering valuable insights for drug design endeavors.


Subject(s)
Machine Learning , Proteins , Mutation , Proteins/chemistry , Thermodynamics , Protein Binding
3.
Appl Intell (Dordr) ; 53(12): 15727-15746, 2023 Jun.
Article in English | MEDLINE | ID: mdl-38031564

ABSTRACT

Machine learning has greatly influenced many fields, including science. However, despite of the tremendous accomplishments of machine learning, one of the key limitations of most existing machine learning approaches is their reliance on large labeled sets, and thus, data with limited labeled samples remains a challenge. Moreover, the performance of machine learning methods often severely hindered in case of diverse data, usually associated with smaller data sets or data associated with areas of study where the size of the data sets is constrained by high experimental cost and/or ethics. These challenges call for innovative strategies for dealing with these types of data. In this work, the aforementioned challenges are addressed by integrating graph-based frameworks, semi-supervised techniques, multiscale structures, and modified and adapted optimization procedures. This results in two innovative multiscale Laplacian learning (MLL) approaches for machine learning tasks, such as data classification, and for tackling data with limited samples, diverse data, and small data sets. The first approach, multikernel manifold learning (MML), integrates manifold learning with multikernel information and incorporates a warped kernel regularizer using multiscale graph Laplacians. The second approach, the multiscale MBO (MMBO) method, introduces multiscale Laplacians to the modification of the famous classical Merriman-Bence-Osher (MBO) scheme, and makes use of fast solvers. We demonstrate the performance of our algorithms experimentally on a variety of benchmark data sets, and compare them favorably to the state-of-art approaches.

4.
Comput Biol Med ; 164: 107250, 2023 09.
Article in English | MEDLINE | ID: mdl-37515872

ABSTRACT

Understanding and accurately predicting protein-ligand binding affinity are essential in the drug design and discovery process. At present, machine learning-based methodologies are gaining popularity as a means of predicting binding affinity due to their efficiency and accuracy, as well as the increasing availability of structural and binding affinity data for protein-ligand complexes. In biomolecular studies, graph theory has been widely applied since graphs can be used to model molecules or molecular complexes in a natural manner. In the present work, we upgrade the graph-based learners for the study of protein-ligand interactions by integrating extensive atom types such as SYBYL and extended connectivity interactive features (ECIF) into multiscale weighted colored graphs (MWCG). By pairing with the gradient boosting decision tree (GBDT) machine learning algorithm, our approach results in two different methods, namely sybylGGL-Score and ecifGGL-Score. Both of our models are extensively validated in their scoring power using three commonly used benchmark datasets in the drug design area, namely CASF-2007, CASF-2013, and CASF-2016. The performance of our best model sybylGGL-Score is compared with other state-of-the-art models in the binding affinity prediction for each benchmark. While both of our models achieve state-of-the-art results, the SYBYL atom-type model sybylGGL-Score outperforms other methods by a wide margin in all benchmarks. Finally, the best-performing SYBYL atom-type model is evaluated on two test sets that are independent of CASF benchmarks.


Subject(s)
Algorithms , Proteins , Ligands , Proteins/chemistry , Protein Binding , Drug Design
5.
J Chem Inf Model ; 63(9): 2656-2666, 2023 05 08.
Article in English | MEDLINE | ID: mdl-37075324

ABSTRACT

Advances in deep neural networks (DNNs) have made a very powerful machine learning method available to researchers across many fields of study, including the biomedical and cheminformatics communities, where DNNs help to improve tasks such as protein performance, molecular design, drug discovery, etc. Many of those tasks rely on molecular descriptors for representing molecular characteristics in cheminformatics. Despite significant efforts and the introduction of numerous methods that derive molecular descriptors, the quantitative prediction of molecular properties remains challenging. One widely used method of encoding molecule features into bit strings is the molecular fingerprint. In this work, we propose using new Neumann-Cayley Gated Recurrent Units (NC-GRU) inside the Neural Nets encoder (AutoEncoder) to create neural molecular fingerprints (NC-GRU fingerprints). The NC-GRU AutoEncoder introduces orthogonal weights into widely used GRU architecture, resulting in faster, more stable training, and more reliable molecular fingerprints. Integrating novel NC-GRU fingerprints and Multi-Task DNN schematics improves the performance of various molecular-related tasks such as toxicity, partition coefficient, lipophilicity, and solvation-free energy, producing state-of-the-art results on several benchmarks.


Subject(s)
Neural Networks, Computer , Proteins , Drug Discovery , Cheminformatics
6.
J Chem Inf Model ; 62(18): 4329-4341, 2022 09 26.
Article in English | MEDLINE | ID: mdl-36108270

ABSTRACT

Molecular surface representations have been advertised as a great tool to study protein structure and functions, including protein-ligand binding affinity modeling. However, the conventional surface-area-based methods fail to deliver a competitive performance on the energy scoring tasks. The main reason is the lack of crucial physical and chemical interactions encoded in the molecular surface generations. We present novel molecular surface representations embedded in different scales of the element interactive manifolds featuring the dramatically dimensional reduction and accurately physical and biological properties encoders. Those low-dimensional surface-based descriptors are ready to be paired with any advanced machine learning algorithms to explore the essential structure-activity relationships that give rise to the element interactive surface area-based scoring functions (EISA-score). The newly developed EISA-score has outperformed many state-of-the-art models, including various well-established surface-related representations, in standard PDBbind benchmarks.


Subject(s)
Machine Learning , Proteins , Algorithms , Ligands , Protein Binding , Proteins/chemistry
7.
Article in English | WPRIM (Western Pacific) | ID: wpr-1032031

ABSTRACT

@#As authorities braced for the arrival of the Omicron variant of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), infrastructure investments and government directives prompted action in central Viet Nam to establish capacity for genomic surveillance sequencing. From 17 November 2021 to 7 January 2022, the Pasteur Institute in Nha Trang sequenced 162 specimens from 98 150 confirmed SARS-CoV-2 cases in the region collected from 8 November to 31 December 2021. Of these, all 127 domestic cases were identified as the B.1.617.2 (Delta) variant, whereas 92% (32/35) of imported cases were identified as the B.1.1.529 (Omicron) variant, all among international flight passengers. Patients were successfully isolated, enabling health-care workers to prepare for additional cases. Most (78%) of the 32 Omicron cases were fully vaccinated, suggesting continued importance of public health and social measures to control the spread of new variants.

8.
Nat Commun ; 12(1): 3521, 2021 06 10.
Article in English | MEDLINE | ID: mdl-34112777

ABSTRACT

The ability of molecular property prediction is of great significance to drug discovery, human health, and environmental protection. Despite considerable efforts, quantitative prediction of various molecular properties remains a challenge. Although some machine learning models, such as bidirectional encoder from transformer, can incorporate massive unlabeled molecular data into molecular representations via a self-supervised learning strategy, it neglects three-dimensional (3D) stereochemical information. Algebraic graph, specifically, element-specific multiscale weighted colored algebraic graph, embeds complementary 3D molecular information into graph invariants. We propose an algebraic graph-assisted bidirectional transformer (AGBT) framework by fusing representations generated by algebraic graph and bidirectional transformer, as well as a variety of machine learning algorithms, including decision trees, multitask learning, and deep neural networks. We validate the proposed AGBT framework on eight molecular datasets, involving quantitative toxicity, physical chemistry, and physiology datasets. Extensive numerical experiments have shown that AGBT is a state-of-the-art framework for molecular property prediction.


Subject(s)
Drug Discovery/methods , Machine Learning , Molecular Conformation , Neural Networks, Computer , Algorithms , Blood-Brain Barrier/drug effects , Computer Simulation , Databases, Chemical , Drug-Related Side Effects and Adverse Reactions , Hydrophobic and Hydrophilic Interactions , Pharmaceutical Preparations/chemistry
9.
Comput Biol Med ; 134: 104460, 2021 07.
Article in English | MEDLINE | ID: mdl-34020133

ABSTRACT

While automated feature extraction has had tremendous success in many deep learning algorithms for image analysis and natural language processing, it does not work well for data involving complex internal structures, such as molecules. Data representations via advanced mathematics, including algebraic topology, differential geometry, and graph theory, have demonstrated superiority in a variety of biomolecular applications, however, their performance is often dependent on manual parametrization. This work introduces the auto-parametrized weighted element-specific graph neural network, dubbed AweGNN, to overcome the obstacle of this tedious parametrization process while also being a suitable technique for automated feature extraction on these internally complex biomolecular data sets. The AweGNN is a neural network model based on geometric-graph features of element-pair interactions, with its graph parameters being updated throughout the training, which results in what we call a network-enabled automatic representation (NEAR). To enhance the predictions with small data sets, we construct multi-task (MT) AweGNN models in addition to single-task (ST) AweGNN models. The proposed methods are applied to various benchmark data sets, including four data sets for quantitative toxicity analysis and another data set for solvation prediction. Extensive numerical tests show that AweGNN models can achieve state-of-the-art performance in molecular property predictions.


Subject(s)
Algorithms , Neural Networks, Computer , Image Processing, Computer-Assisted
10.
Annu Rev Biophys ; 50: 1-30, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33064571

ABSTRACT

In the global health emergency caused by coronavirus disease 2019 (COVID-19), efficient and specific therapies are urgently needed. Compared with traditional small-molecular drugs, antibody therapies are relatively easy to develop; they are as specific as vaccines in targeting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2); and they have thus attracted much attention in the past few months. This article reviews seven existing antibodies for neutralizing SARS-CoV-2 with 3D structures deposited in the Protein Data Bank (PDB). Five 3D antibody structures associated with the SARS-CoV spike (S) protein are also evaluated for their potential in neutralizing SARS-CoV-2. The interactions of these antibodies with the S protein receptor-binding domain (RBD) are compared with those between angiotensin-converting enzyme 2 and RBD complexes. Due to the orders of magnitude in the discrepancies of experimental binding affinities, we introduce topological data analysis, a variety of network models, and deep learning to analyze the binding strength and therapeutic potential of the 14 antibody-antigen complexes. The current COVID-19 antibody clinical trials, which are not limited to the S protein target, are also reviewed.


Subject(s)
Antibodies, Viral/therapeutic use , COVID-19/therapy , SARS-CoV-2/immunology , Antibodies, Viral/immunology , COVID-19/immunology , COVID-19/virology , Humans , Models, Molecular , SARS-CoV-2/isolation & purification
11.
ArXiv ; 2020 Jun 18.
Article in English | MEDLINE | ID: mdl-32601601

ABSTRACT

Under the global health emergency caused by coronavirus disease 2019 (COVID-19), efficient and specific therapies are urgently needed. Compared with traditional small-molecular drugs, antibody therapies are relatively easy to develop and as specific as vaccines in targeting severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and thus attract much attention in the past few months. This work reviews seven existing antibodies for SARS-CoV-2 spike (S) protein with three-dimensional (3D) structures deposited in the Protein Data Bank. Five antibody structures associated with SARS-CoV are evaluated for their potential in neutralizing SARS-CoV-2. The interactions of these antibodies with the S protein receptor-binding domain (RBD) are compared with those of angiotensin-converting enzyme 2 (ACE2) and RBD complexes. Due to the orders of magnitude in the discrepancies of experimental binding affinities, we introduce topological data analysis (TDA), a variety of network models, and deep learning to analyze the binding strength and therapeutic potential of the aforementioned fourteen antibody-antigen complexes. The current COVID-19 antibody clinical trials, which are not limited to the S protein target, are also reviewed.

12.
J Chem Inf Model ; 60(12): 5682-5698, 2020 12 28.
Article in English | MEDLINE | ID: mdl-32686938

ABSTRACT

Current drug discovery is expensive and time-consuming. It remains a challenging task to create a wide variety of novel compounds that not only have desirable pharmacological properties but also are cheaply available to low-income people. In this work, we develop a generative network complex (GNC) to generate new drug-like molecules based on the multiproperty optimization via the gradient descent in the latent space of an autoencoder. In our GNC, both multiple chemical properties and similarity scores are optimized to generate drug-like molecules with desired chemical properties. To further validate the reliability of the predictions, these molecules are reevaluated and screened by independent 2D fingerprint-based predictors to come up with a few hundreds of new drug candidates. As a demonstration, we apply our GNC to generate a large number of new BACE1 inhibitors, as well as thousands of novel alternative drug candidates for eight existing market drugs, including Ceritinib, Ribociclib, Acalabrutinib, Idelalisib, Dabrafenib, Macimorelin, Enzalutamide, and Panobinostat.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Drug Discovery , Humans , Pharmaceutical Preparations
13.
J Phys Chem Lett ; 11(13): 5373-5382, 2020 Jul 02.
Article in English | MEDLINE | ID: mdl-32543196

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has infected over 7.1 million people and led to over 0.4 million deaths. Currently, there is no specific anti-SARS-CoV-2 medication. New drug discovery typically takes more than 10 years. Drug repositioning becomes one of the most feasible approaches for combating COVID-19. This work curates the largest available experimental data set for SARS-CoV-2 or SARS-CoV 3CL (main) protease inhibitors. On the basis of this data set, we develop validated machine learning models with relatively low root-mean-square error to screen 1553 FDA-approved drugs as well as another 7012 investigational or off-market drugs in DrugBank. We found that many existing drugs might be potentially potent to SARS-CoV-2. The druggability of many potent SARS-CoV-2 3CL protease inhibitors is analyzed. This work offers a foundation for further experimental studies of COVID-19 drug repositioning.


Subject(s)
Antiviral Agents/metabolism , Coronavirus Infections/drug therapy , Cysteine Proteinase Inhibitors/metabolism , Drug Repositioning , Pneumonia, Viral/drug therapy , Betacoronavirus/enzymology , COVID-19 , Coronavirus 3C Proteases , Coronavirus Infections/enzymology , Cysteine Endopeptidases/metabolism , Databases, Protein/statistics & numerical data , Humans , Machine Learning , Pandemics , Pneumonia, Viral/enzymology , Protein Binding , SARS-CoV-2 , Viral Nonstructural Proteins/antagonists & inhibitors , Viral Nonstructural Proteins/metabolism , COVID-19 Drug Treatment
14.
Int J Numer Method Biomed Eng ; 36(9): e3376, 2020 09.
Article in English | MEDLINE | ID: mdl-32515170

ABSTRACT

Persistent homology is constrained to purely topological persistence, while multiscale graphs account only for geometric information. This work introduces persistent spectral theory to create a unified low-dimensional multiscale paradigm for revealing topological persistence and extracting geometric shapes from high-dimensional datasets. For a point-cloud dataset, a filtration procedure is used to generate a sequence of chain complexes and associated families of simplicial complexes and chains, from which we construct persistent combinatorial Laplacian matrices. We show that a full set of topological persistence can be completely recovered from the harmonic persistent spectra, that is, the spectra that have zero eigenvalues, of the persistent combinatorial Laplacian matrices. However, non-harmonic spectra of the Laplacian matrices induced by the filtration offer another powerful tool for data analysis, modeling, and prediction. In this work, fullerene stability is predicted by using both harmonic spectra and non-harmonic persistent spectra, while the latter spectra are successfully devised to analyze the structure of fullerenes and model protein flexibility, which cannot be straightforwardly extracted from the current persistent homology. The proposed method is found to provide excellent predictions of the protein B-factors for which current popular biophysical models break down.


Subject(s)
Data Analysis , Proteins/chemistry
15.
bioRxiv ; 2020 Feb 04.
Article in English | MEDLINE | ID: mdl-32511308

ABSTRACT

Wuhan coronavirus, called 2019-nCoV, is a newly emerged virus that infected more than 9692 people and leads to more than 213 fatalities by January 30, 2020. Currently, there is no effective treatment for this epidemic. However, the viral protease of a coronavirus is well-known to be essential for its replication and thus is an effective drug target. Fortunately, the sequence identity of the 2019-nCoV protease and that of severe-acute respiratory syndrome virus (SARS-CoV) is as high as 96.1%. We show that the protease inhibitor binding sites of 2019-nCoV and SARS-CoV are almost identical, which means all potential anti-SARS-CoV chemotherapies are also potential 2019-nCoV drugs. Here, we report a family of potential 2019-nCoV drugs generated by a machine intelligence-based generative network complex (GNC). The potential effectiveness of treating 2019-nCoV by using some existing HIV drugs is also analyzed.

16.
bioRxiv ; 2020 Feb 13.
Article in English | MEDLINE | ID: mdl-32511344

ABSTRACT

The World Health Organization (WHO) has declared the 2019 novel coronavirus (2019-nCoV) infection outbreak a global health emergency. Currently, there is no effective anti-2019-nCoV medication. The sequence identity of the 3CL proteases of 2019-nCoV and SARS is 96%, which provides a sound foundation for structural-based drug repositioning (SBDR). Based on a SARS 3CL protease X-ray crystal structure, we construct a 3D homology structure of 2019-nCoV 3CL protease. Based on this structure and existing experimental datasets for SARS 3CL protease inhibitors, we develop an SBDR model based on machine learning and mathematics to screen 1465 drugs in the DrugBank that have been approved by the U.S. Food and Drug Administration (FDA). We found that many FDA approved drugs are potentially highly potent to 2019-nCoV.

17.
Phys Chem Chem Phys ; 22(16): 8373-8390, 2020 Apr 29.
Article in English | MEDLINE | ID: mdl-32266895

ABSTRACT

Recently, molecular fingerprints extracted from three-dimensional (3D) structures using advanced mathematics, such as algebraic topology, differential geometry, and graph theory have been paired with efficient machine learning, especially deep learning algorithms to outperform other methods in drug discovery applications and competitions. This raises the question of whether classical 2D fingerprints are still valuable in computer-aided drug discovery. This work considers 23 datasets associated with four typical problems, namely protein-ligand binding, toxicity, solubility and partition coefficient to assess the performance of eight 2D fingerprints. Advanced machine learning algorithms including random forest, gradient boosted decision tree, single-task deep neural network and multitask deep neural network are employed to construct efficient 2D-fingerprint based models. Additionally, appropriate consensus models are built to further enhance the performance of 2D-fingerprint-based methods. It is demonstrated that 2D-fingerprint-based models perform as well as the state-of-the-art 3D structure-based models for the predictions of toxicity, solubility, partition coefficient and protein-ligand binding affinity based on only ligand information. However, 3D structure-based models outperform 2D fingerprint-based methods in complex-based protein-ligand binding affinity predictions.


Subject(s)
Chemistry Techniques, Analytical/standards , Drug Discovery/methods , Peptide Mapping , Algorithms , Datasets as Topic
18.
Phys Chem Chem Phys ; 22(8): 4343-4367, 2020 Feb 26.
Article in English | MEDLINE | ID: mdl-32067019

ABSTRACT

Recently, machine learning (ML) has established itself in various worldwide benchmarking competitions in computational biology, including Critical Assessment of Structure Prediction (CASP) and Drug Design Data Resource (D3R) Grand Challenges. However, the intricate structural complexity and high ML dimensionality of biomolecular datasets obstruct the efficient application of ML algorithms in the field. In addition to data and algorithm, an efficient ML machinery for biomolecular predictions must include structural representation as an indispensable component. Mathematical representations that simplify the biomolecular structural complexity and reduce ML dimensionality have emerged as a prime winner in D3R Grand Challenges. This review is devoted to the recent advances in developing low-dimensional and scalable mathematical representations of biomolecules in our laboratory. We discuss three classes of mathematical approaches, including algebraic topology, differential geometry, and graph theory. We elucidate how the physical and biological challenges have guided the evolution and development of these mathematical apparatuses for massive and diverse biomolecular data. We focus the performance analysis on protein-ligand binding predictions in this review although these methods have had tremendous success in many other applications, such as protein classification, virtual screening, and the predictions of solubility, solvation free energies, toxicity, partition coefficients, protein folding stability changes upon mutation, etc.


Subject(s)
Computational Biology , Models, Biological , Algorithms , Molecular Sequence Data
19.
J Chem Inf Model ; 60(3): 1235-1244, 2020 03 23.
Article in English | MEDLINE | ID: mdl-31977216

ABSTRACT

Machine learning approaches have had tremendous success in various disciplines. However, such success highly depends on the size and quality of datasets. Scientific datasets are often small and difficult to collect. Currently, improving machine learning performance for small scientific datasets remains a major challenge in many academic fields, such as bioinformatics or medical science. Gradient boosting decision tree (GBDT) is typically optimal for small datasets, while deep learning often performs better for large datasets. This work reports a boosting tree-assisted multitask deep learning (BTAMDL) architecture that integrates GBDT and multitask deep learning (MDL) to achieve near-optimal predictions for small datasets when there exists a large dataset that is well correlated to the small datasets. Two BTAMDL models are constructed, one utilizing purely MDL output as GBDT input while the other admitting additional features in GBDT input. The proposed BTAMDL models are validated on four categories of datasets, including toxicity, partition coefficient, solubility, and solvation. It is found that the proposed BTAMDL models outperform the current state-of-the-art methods in various applications involving small datasets.


Subject(s)
Deep Learning , Computational Biology , Machine Learning , Solubility
20.
J Comput Aided Mol Des ; 34(2): 131-147, 2020 02.
Article in English | MEDLINE | ID: mdl-31734815

ABSTRACT

We present the performances of our mathematical deep learning (MathDL) models for D3R Grand Challenge 4 (GC4). This challenge involves pose prediction, affinity ranking, and free energy estimation for beta secretase 1 (BACE) as well as affinity ranking and free energy estimation for Cathepsin S (CatS). We have developed advanced mathematics, namely differential geometry, algebraic graph, and/or algebraic topology, to accurately and efficiently encode high dimensional physical/chemical interactions into scalable low-dimensional rotational and translational invariant representations. These representations are integrated with deep learning models, such as generative adversarial networks (GAN) and convolutional neural networks (CNN) for pose prediction and energy evaluation, respectively. Overall, our MathDL models achieved the top place in pose prediction for BACE ligands in Stage 1a. Moreover, our submissions obtained the highest Spearman correlation coefficient on the affinity ranking of 460 CatS compounds, and the smallest centered root mean square error on the free energy set of 39 CatS molecules. It is worthy to mention that our method on docking pose predictions has significantly improved from our previous ones.


Subject(s)
Deep Learning , Drug Design , Amyloid Precursor Protein Secretases/chemistry , Amyloid Precursor Protein Secretases/metabolism , Aspartic Acid Endopeptidases/chemistry , Aspartic Acid Endopeptidases/metabolism , Binding Sites , Cathepsins/chemistry , Cathepsins/metabolism , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Thermodynamics
SELECTION OF CITATIONS
SEARCH DETAIL