ArticlePublisher preview available

Prediction of drug efficacy from transcriptional profiles with deep learning

Authors:
To read the full-text of this research, you can request a copy directly from the authors.

Abstract and Figures

Drug discovery focused on target proteins has been a successful strategy, but many diseases and biological processes lack obvious targets to enable such approaches. Here, to overcome this challenge, we describe a deep learning–based efficacy prediction system (DLEPS) that identifies drug candidates using a change in the gene expression profile in the diseased state as input. DLEPS was trained using chemically induced changes in transcriptional profiles from the L1000 project. We found that the changes in transcriptional profiles for previously unexamined molecules were predicted with a Pearson correlation coefficient of 0.74. We examined three disorders and experimentally tested the top drug candidates in mouse disease models. Validation showed that perillen, chikusetsusaponin IV and trametinib confer disease-relevant impacts against obesity, hyperuricemia and nonalcoholic steatohepatitis, respectively. DLEPS can generate insights into pathogenic mechanisms, and we demonstrate that the MEK–ERK signaling pathway is a target for developing agents against nonalcoholic steatohepatitis. Our findings suggest that DLEPS is an effective tool for drug repurposing and discovery. Drug discovery based on transcriptional profiling does not require knowledge of protein targets.
Statistical and structural analysis of DLEPS’ performance a, The distribution of maximum Tanimoto Similarity based on CDK fingerprint (CDK TS) of each test molecule among comparison with all training molecules. b, c, The distribution of Pearson correlation coefficient r of predicted versus empirical changes of transcriptional profiles (CTPs) of test molecules with CDK TS < 0.4 (b) (mean r = 0.60, peak r = 0.8) and with CDK TS > 0.4 (c) (mean r = 0.79, peak r = 0.93). d, A few well-predicted test molecules (r > 0.74) and their most similar molecules in the training set, indicating DLEPS is capable of predicting CTPs of structurally novel molecules. The Maximum Common Sub-Structures (MCSS) are highlighted in cyan. e, The distribution of Pearson correlation coefficient r of predicted versus empirical CTPs among selected molecule pairs. One molecule in these pairs is from well-predicted test set (r > 0.74, n = 2033 out of 3000) and the other one in the pair is a structurally similar molecule from the training set, with CDK TS > 0.35. The mean Pearson r equals to 0.50. f, As comparison, Pearson r for randomly permutated pairs equals to 0.07. g−i, Similarity versus correlation analysis of molecule pairs. g, Principal component analysis (PCA) of CTPs of test molecule BRD-K70918941 and its most similar molecules in training set. MCSS were highlighted in cyan for each molecule. DLEPS predicted CTP was highlighted in red. The heatmap of CDK Tanimoto similarity (h) and correlation coefficient matrix (i) of sampled pairs. j, Scatter plot of CDK TS versus correlation coefficient of CTPs, indicating that high CDK TS not necessarily yield high correlation and vice versa. k, The exemplar fragments tend to disrupt (upper) and retain (bottom) the CTPs, analyzed from the well (r > 0.80) / poorly (-0.3 < r < 0.3) correlated pair groups in e).
… 
Transcriptional analysis of perillen treated mice, extra DLEPS analysis and pharmacokinetic analysis of perillen a, Blood uric acid levels (BUA) of control, HUA model mice and HUA model mice treated with 4 molecules from negative set (Marbofloxacin, Captopril, Parecoxib and Mupirocin at 20 mg/kg, n = 6). b, Kidney index of normal, HUA model mice and perillen treated HUA model mice at 2.5, 5 and 10 mg/kg and topiroxostat treated HUA model mice (n = 6). Body weight (c) Food intake (d) and water intake (e) of mice with treatment of perillen for 7 days (c−e, n = 6). f, Principal component analysis of normal, HUA model, and HUA model perillen-treated mice (n = 3). g, Scatter plot of gene expression in HUA model versus non-induced control mice. The color gradient represents dot intensity. h, Scatter plot of gene expression in perillen treated mice versus that of HUA model mice. i, Scatter plot of slopes in h) versus that in g) (r = -0.23, P < 3e-232). j, GO analysis of upregulated genes in model mice (n = 3). k, GO analysis of downregulated genes in perillen treated model mice (n = 3). l−n, Extra analysis of anti-inflammation and fibrosis score using a NASH phase IV gene signatures (l) and hepatic steatosis gene signatures (m). Big red dot highlights perillen, indicating prediction of perillen is robust to various inflammation/fibrosis gene signatures. n, Scatter plot of the inflammation/fibrosis score in Fig. 4b versus the NASH phase IV score (r = 0.51, P < 2e-238), indicating a well correlation of these two scores. o, Chromatograms of perillen. p, The serum concentration-time curves of perillen for 4 various conditions. * P < 0.05, ** P < 0.01, **** P < 0.0001 compared with model group. ## P < 0.01, #### P < 0.0001 compared with normal group (Normal). All P values were determined by two-tailed paired t-test. All data are presented as the mean ± sem. Source data
… 
Histological, serum and TUNEL analysis of molecules treated MCD model mice 8 week-old mice were housed at 22°C, received MCD diets for two weeks, and then treated with positively predicted compounds: Normilin (6 mg/kg), Lupenone (2 or 6 mg/kg), Telmisartan (10 mg/kg), Bendroflumethiazide (1.5 mg/kg), GI02002 (10 mg/kg), Ravoxertinib (1 mg/kg) in a), and with negatively predicted compounds: Butoconazole (10 mg/Kg), Benfotiamine (10 mg/kg), Menatetrenone (2.5 mg/kg), Phenacetin (70 mg/kg), GI02002 (10 mg/kg, positive control) or vehicle (0.5%CMC-Na containing 3%DMSO) in b−d) by i.p. injection for 14 days. a, H&E (hematoxylin and eosin) staining of liver (3 mice replicates). b, Serum ALT and AST level (n = 6 in MCD, Butoconazole and Phenacetin group, and n = 7 in other groups, The P values of ALT in each group compared with model group were 0.8374, 0.4412, 0.5640, 0.1975 and 0.0002 respectively. The P values of AST in each group compared with model group were 0.6609, 0.5452, 0.1093, 0.8002 and 0.0002, respectively). c, Serum CHO and TG level (n = 6 in MCD, Butoconazole and Phenacetin group, and n = 7 in other groups, The P values of CHO in each group compared with model group were 0.1014, 0.1176, 0.0958, 0.0909 and 0.0177, respectively. The P values of TG in each group compared with model group were 0.8872, 0.5317, 0.4414, 0.2618 and 0.9238, respectively). d, H&E staining of liver (upper row, 3 mice replicates). Scale bar indicates 50 μm. Oil-Red staining of liver (bottom row, 3 mice replicates). Scale bar indicates 100 μm. e, Representative images of TUNEL staining (3 mice replicates, The P values model group compared with normal group were < 0.0001 and the P values Trametinib group compared with model group were < 0.0001, respectively). Scale bar indicates 200 μm. All P values were determined by two-tailed paired t-test.* P < 0.05, *** P < 0.001, **** P < 0.0001 compared with model group (MCD). #### P < 0.0001 compared with normal group (Normal). All data are presented as the mean ± sem. Source data
… 
This content is subject to copyright. Terms and conditions apply.
Articles
https://doi.org/10.1038/s41587-021-00946-z
1Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, China. 2Department of Pharmacology, School of Basic
Medical Sciences, Health Science Center, Peking University, Beijing, China. 3Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals
Tech. Co. Ltd., Beijing, China. 4Department of Anatomy, Histology and Embryology, Neuroscience Research Institute, Health Science Center, Peking
University, Beijing, China. 5State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Health Science Center, Peking
University, Beijing, China. 6These authors contributed equally: Jie Zhu, Jingxiang Wang, Xin Wang, Mingjing Gao, Bingbing Guo. 7These authors jointly
supervised this work: Hong Zhu, Ning Zhang, Ruimao Zheng, Zhengwei Xie. e-mail: rainbow_zhou@126.com; zhangning@bjmu.edu.cn;
rmzheng@pku.edu.cn; xiezhengwei@hsc.pku.edu.cn
Recent developments in the application of deep learning to
diverse areas (for example, natural language processing, com-
puter vision and so on) suggest the potential of advanced
algorithms for the assessment of chemicals in applications such as
molecular encoding, chemical synthesis route planning and inhibi-
tor target prediction15. Combined with resources developed in
computational chemistry, these deep learning tools are changing
the landscape of chemical and pharmaceutical research and devel-
opment (for example, enabling rapid sampling of a vast chemical
space and allowing researchers to make accurate predictions about
structure–function relationships).
Drug development based on target proteins has been a suc-
cessful approach in the past decades, but these methods cannot
address diseases that lack well-defined protein targets. One strategy
for developing drugs to treat these diseases would be to generate a
model capable of predicting efficacy independent of specific targets.
A recent study showed how a new antibiotic candidate for treating
Escherichia coli infections was found using a customized deep learn-
ing model6. However, this kind of model is built on a case-by-case
basis and relies on phenotypic data specific to a single disease state;
that is, it lacks the ability to generalize to other diseases.
Given that most diseases are associated with characteris-
tic changes in gene expression profiles, such changes are used as
indicators reflecting the underlying mechanisms of diseases, an
assumption embodied in the Connectivity Map (CMap) concept710.
However, CMap is applicable only to the molecules whose tran-
scriptional profiles have already been experimentally assessed. We
envisioned that a model capable of predicting chemically inducible
changes in transcriptional profiles (CTPs) for an unlimited number
of small molecules would make it much easier to find potent agents
to develop as treatments for most diseases. First, we constructed a
neural network using simplified molecular-input line-entry system
(SMILES) chemical encoding as input to fit CTPs that were mea-
sured in the L1000 project11 (Fig. 1a). Second, using gene signatures
specific to pathological contexts, we employed gene set enrichment
analysis (GSEA)12 to evaluate the potential efficacy of compounds
against these diseases. We refer to this approach and model as
DLEPS.
Results
The architecture and training of DLEPS. To build a general-
purpose model that is suitable for use with many diseases, especially
for disorders without well-defined targets, we developed DLEPS
comprising two stages. First, we trained a deep neural network to
predict CTPs based on data from cell culture screening with diverse
compounds (Fig. 1a). The SMILES encoding of small molecules was
initially parsed to a grammar tree13, which was then encoded to a
point randomly in a high-dimensional sphere (Fig. 1a, middle). The
latent vector was further passed to a deep dense network to predict
the CTPs (Fig. 1a, right).
Second, we selected upregulated and downregulated gene sig-
natures that should reflect pathological changes in gene expression
levels; here, we employed GSEA, which has been adopted in CMap,
to compute an enrichment score as the efficacy score7,9. According
to this score, we finally selected several top-ranked candidate small
molecules to be assayed with cell cultures or directly in animal models
Prediction of drug efficacy from transcriptional
profiles with deep learning
Jie Zhu 1,2,6, Jingxiang Wang3,6, Xin Wang2,6, Mingjing Gao3,6, Bingbing Guo4,6, Miaomiao Gao1,
Jiarui Liu4, Yanqiu Yu1, Liang Wang2, Weikaixin Kong 5, Yongpan An2, Zurui Liu3, Xinpei Sun 1,
Zhuo Huang 5, Hong Zhou2,7 ✉ , Ning Zhang1,7 ✉ , Ruimao Zheng4,7 ✉ and Zhengwei Xie 1,3,7 ✉
Drug discovery focused on target proteins has been a successful strategy, but many diseases and biological processes lack obvi-
ous targets to enable such approaches. Here, to overcome this challenge, we describe a deep learning–based efficacy prediction
system (DLEPS) that identifies drug candidates using a change in the gene expression profile in the diseased state as input.
DLEPS was trained using chemically induced changes in transcriptional profiles from the L1000 project. We found that the
changes in transcriptional profiles for previously unexamined molecules were predicted with a Pearson correlation coefficient
of 0.74. We examined three disorders and experimentally tested the top drug candidates in mouse disease models. Validation
showed that perillen, chikusetsusaponin IV and trametinib confer disease-relevant impacts against obesity, hyperuricemia and
nonalcoholic steatohepatitis, respectively. DLEPS can generate insights into pathogenic mechanisms, and we demonstrate that
the MEK–ERK signaling pathway is a target for developing agents against nonalcoholic steatohepatitis. Our findings suggest
that DLEPS is an effective tool for drug repurposing and discovery.
NATURE BIOTECHNOLOGY | VOL 39 | NOVEMBER 2021 | 1444–1452 | www.nature.com/naturebiotechnology
1444
Content courtesy of Springer Nature, terms of use apply. Rights reserved
... This challenge has spurred the development of deep learning models capable of predicting transcriptional profiles for novel chemicals using publicly available data. DLEPS is a deep neural network designed to predict gene expression responses to new chemicals without cell-type specificity 13 . Furthermore, DeepCE 14 and CIGER 15 utilize one-hot encoding to distinguish between cell types, learning from diverse perturbational profiles. ...
... (3) Even when a target is identified, the resulting drug may struggle to reach its target within the cell due to poor cell permeability, thereby hindering the achievement of the desired therapeutic effect 39,40 . These challenges have spurred the emergence of phenotype-based approaches, which directly analyze overall cellular response to drugs, offering a more holistic understanding of disease mechanisms, and the potential for discoveries of novel drug mechanisms and therapeutic opportunities 13,41 . ...
Article
Full-text available
Artificial intelligence transforms drug discovery, with phenotype-based approaches emerging as a promising alternative to target-based methods, overcoming limitations like lack of well-defined targets. While chemical-induced transcriptional profiles offer a comprehensive view of drug mechanisms, inherent noise often obscures the true signal, hindering their potential for meaningful insights. Here, we highlight the development of TranSiGen, a deep generative model employing self-supervised representation learning. TranSiGen analyzes basal cell gene expression and molecular structures to reconstruct chemical-induced transcriptional profiles with high accuracy. By capturing both cellular and compound information, TranSiGen-derived representations demonstrate efficacy in diverse downstream tasks like ligand-based virtual screening, drug response prediction, and phenotype-based drug repurposing. Notably, in vitro validation of TranSiGen’s application in pancreatic cancer drug discovery highlights its potential for identifying effective compounds. We envisage that integrating TranSiGen into the drug discovery and mechanism research holds significant promise for advancing biomedicine.
Article
Drug response is conventionally measured at the cell level, often quantified by metrics like IC50. However, to gain a deeper understanding of drug response, cellular outcomes need to be understood in terms of pathway perturbation. This perspective leads us to recognize a challenge posed by the gap between two widely used large-scale databases, LINCS L1000 and GDSC, measuring drug response at different levels—L1000 captures information at the gene expression level, while GDSC operates at the cell line level. Our study aims to bridge this gap by integrating the two databases through transfer learning, focusing on condition-specific perturbations in gene interactions from L1000 to interpret drug response integrating both gene and cell levels in GDSC. This transfer learning strategy involves pretraining on the transcriptomic-level L1000 dataset, with parameter-frozen fine-tuning to cell line-level drug response. Our novel condition-specific gene–gene attention (CSG2A) mechanism dynamically learns gene interactions specific to input conditions, guided by both data and biological network priors. The CSG2A network, equipped with transfer learning strategy, achieves state-of-the-art performance in cell line-level drug response prediction. In two case studies, well-known mechanisms of drugs are well represented in both the learned gene–gene attention and the predicted transcriptomic profiles. This alignment supports the modeling power in terms of interpretability and biological relevance. Furthermore, our model’s unique capacity to capture drug response in terms of both pathway perturbation and cell viability extends predictions to the patient level using TCGA data, demonstrating its expressive power obtained from both gene and cell levels. Availability and implementation The source code for the CSG2A network is available at https://github.com/eugenebang/CSG2A.
Article
Full-text available
Traditional single nucleic acid assays can only detect one target while multiple nucleic acid assays can detect multiple targets simultaneously, providing comprehensive and accurate information. Fluorescent microspheres in multiplexed nucleic acid detection offer high sensitivity, specificity, multiplexing, flexibility, and scalability advantages, enabling precise, real-time results and supporting clinical diagnosis and research. However, multiplexed assays face challenges like complexity, costs, and sample handling issues. The review explores the recent advancements and applications of fluorescent microspheres in multiple nucleic acid detection. It discusses the versatility of fluorescent microspheres in various fields, such as disease diagnosis, drug screening, and personalized medicine. The review highlights the possibility of adjusting the performance of fluorescent microspheres by modifying concentrations and carrier forms, allowing for tailored applications. It emphasizes the potential of fluorescent microsphere technology in revolutionizing nucleic acid detection and advancing health, disease treatment, and medical research.
Article
Full-text available
Upon a diagnosis, the clinical team faces two main questions: what treatment, and at what dose? Clinical trials' results provide the basis for guidance and support for official protocols that clinicians use to base their decisions. However, individuals do not consistently demonstrate the reported response from relevant clinical trials. The decision complexity increases with combination treatments where drugs administered together can interact with each other, which is often the case. Additionally, the individual's response to the treatment varies with the changes in their condition. In practice, the drug and the dose selection depend significantly on the medical protocol and the medical team's experience. As such, the results are inherently varied and often suboptimal. Big data and Artificial Intelligence (AI) approaches have emerged as excellent decision-making tools, but multiple challenges limit their application. AI is a rapidly evolving and dynamic field with the potential to revolutionize various aspects of human life. AI has become increasingly crucial in drug discovery and development. AI enhances decision-making across different disciplines, such as medicinal chemistry, molecular and cell biology, pharmacology, pathology, and clinical practice. In addition to these, AI contributes to patient population selection and stratification. The need for AI in healthcare is evident as it aids in enhancing data accuracy and ensuring the quality care necessary for effective patient treatment. AI is pivotal in improving success rates in clinical practice. The increasing significance of AI in drug discovery, development, and clinical trials is underscored by many scientific publications. Despite the numerous advantages of AI, such as enhancing and advancing Precision Medicine (PM) and remote patient monitoring, unlocking its full potential in healthcare requires addressing fundamental concerns. These concerns include data quality, the lack of well-annotated large datasets, data privacy and safety issues, biases in AI algorithms, legal and ethical challenges, and obstacles related to cost and implementation. Nevertheless, integrating AI in clinical medicine will improve diagnostic accuracy and treatment outcomes, contribute to more efficient healthcare delivery, reduce costs, and facilitate better patient experiences, making healthcare more sustainable. This article reviews AI applications in drug development and clinical practice, making healthcare more sustainable, and highlights concerns and limitations in applying AI.
Article
Full-text available
In clinical practice, colon cancer is a prevalent malignant tumor of the digestive system, characterized by a complex and progressive process involving multiple genes and molecular pathways. Historically, research efforts have primarily focused on investigating individual genes; however, our current study aims to explore the collective impact of multiple genes on colon cancer and to identify potential therapeutic targets associated with these genes. For this research, we acquired the gene expression profiles and RNA sequencing data of colon cancer from TCGA. Subsequently, we conducted differential gene expression analysis using R, followed by GO and KEGG pathway enrichment analyses. To construct a protein–protein interaction (PPI) network, we selected survival-related genes using the log-rank test and single-factor Cox regression analysis. Additionally, we performed LASSO regression analysis, immune infiltration analysis, mutation analysis, and cMAP analysis, as well as an investigation into ferroptosis. Our differential expression and survival analyses identified 47 hub genes, and subsequent LASSO regression analysis refined the focus to 23 key genes. These genes are closely linked to cancer metastasis, proliferation, apoptosis, cell cycle regulation, signal transduction, cancer microenvironment, immunotherapy, and neurodevelopment. Overall, the hub genes discovered in our study are pivotal in colon cancer and are anticipated to serve as important biological markers for the diagnosis and treatment of the disease.
Article
In this review, we review some existing/published computational frameworks for drug repositioning, organized on the basis of the type of biomedical input data analyzed and the computational algorithms involved. We also outline some exciting new directions that drug repurposing research may take, as pioneered by the generative AI revolution.
Article
Full-text available
The prediction of drug properties plays a vital role in drug research. However, the drug property prediction accuracy of traditional methods is limited due to their inability to fully capture molecular structure and function information. As a result, the use of graph neural networks has attracted significant attention as an effective drug property prediction approach. Nevertheless, traditional graph neural networks still exhibit certain drawbacks in this regard, including their disregard of the interaction information between nodes and edges, the loss of local information during global pooling operations, and the absence of feature fusion mechanisms. This study proposes an enhanced graph neural network (GNN) model that incorporates an attention mechanism, multiscale pooling, an adaptive weight generator, and an activation function to predict drug properties. A comparative analysis with the conventional graph neural network model reveals significant improvements in terms of predicting the side effects of drugs on the heart and liver, with increases of 1%, 7%, and 13%. Furthermore, the enhanced graph neural network model exhibits good performance across the remaining two datasets. Empirical findings underscore the efficacy of the model in drug property prediction tasks, and it is characterized by enhanced predictive precision and robust performance outcomes.
Article
Full-text available
Hyperuricemia (HUA) is a risk factor for chronic kidney disease (CKD). Serum uric acid (SUA) levels in CKD stage 3–4 patients closely correlate with hyperuricemic nephropathy (HN) morbidity. New uric acid (UA)-lowering strategies are required to prevent CKD. The multiple-purpose connectivity map (CMAP) was used to discover potential molecules against HUA and renal fibrosis. We used HUA and unilateral ureteral occlusion (UUO) model mice to verify renoprotective effects of molecules and explore related mechanisms. In vitro experiments were performed in HepG2 and NRK-52E cells induced by UA. Esculetin was the top scoring compound and lowered serum uric acid (SUA) levels with dual functions on UA excretion. Esculetin exerted these effects by inhibiting expression and activity of xanthine oxidase (XO) in liver, and modulating UA transporters in kidney. The mechanism by which esculetin suppressed XO was related to inhibiting the nuclear translocation of hexokinase 2 (HK2). Esculetin was anti-fibrotic in HUA and UUO mice through inhibiting TGF-β1-activated profibrotic signals. The renoprotection effects of esculetin in HUA mice were associated with lower SUA, alleviation of oxidative stress, and inhibition of fibrosis. Esculetin is a candidate urate-lowering drug with renoprotective activity and the ability to inhibit XO, promote excretion of UA, protect oxidative stress injury, and reduce renal fibrosis.
Article
Full-text available
It has been demonstrated that Chikusetsusaponin IVa (CsIVa) possesses abundant biological activities. Herein, using LPS to establish acute inflammation model of mouse liver and cell line inflammation model, we investigated whether miR-155/GSK-3β regulated NF-κB signaling pathway, and CsIVa exerted anti-inflammatory effects by regulating miR-155/GSK-3β signaling pathway. Our results showed that LPS induced high expression of miR-155 and miR-155 promoted macrophage activation through GSK-3β. In addition, CsIVa inhibited inflammatory responses in LPS-induced mouse liver and RAW264.7 cells. Furthermore, we demonstrated that CsIVa improved the inflammatory response in LPS-induced RAW264.7 cells by inhibiting miR-155, increasing GSK-3β expression, and inhibiting NF-κB signaling pathway. In conclusion, our study reveals that CsIVa suppresses LPS-triggered immune response by miR-155/GSK-3β-NF-κB signaling pathway.
Article
Full-text available
Objectives Although NaV1.7 sodium channel is a promising drug target for pain, traditional screening strategies for discovery of NaV1.7 inhibitors are very painstaking and time-consuming. Herein, we aimed to build machine learning models for screening and design of potent and effective NaV1.7 sodium channel inhibitors. Materials and Methods We customized the imbalanced data set from ChEMBL and BindingDB to train and filter the best classification model. Then the whole-cell voltage-clamp was employed to validate the inhibitors. We assembled a molecular group optimization method by combining Grammar Variational Autoencoder, classification model and simulated annealing. Results and Conclusion We found the RF-CDK model (Random forest + CDK figureprint) performs best in the imbalanced data set. Of the three compounds that may have inhibitory effects, nortriptyline has been experimentally verified. In the molecule optimization process, 40 molecules located in the applicability domain of RF-CDK were used as start point, among which 34 molecules evolved to molecules with greater molecular scores (MS). The molecule with highest MS was derived from CHEMBL2325245. The model and method we developed for NaV1.7 inhibitors are also applicable to other targets.
Article
Full-text available
We have developed a deep generative model, generative tensorial reinforcement learning (GENTRL), for de novo small-molecule design. GENTRL optimizes synthetic feasibility, novelty, and biological activity. We used GENTRL to discover potent inhibitors of discoidin domain receptor 1 (DDR1), a kinase target implicated in fibrosis and other diseases, in 21 days. Four compounds were active in biochemical assays, and two were validated in cell-based assays. One lead candidate was tested and demonstrated favorable pharmacokinetics in mice. A machine learning model allows the identification of new small-molecule kinase inhibitors in days.
Article
Full-text available
Rationale: Osteoporosis is a severe bone disorder that is a threat to our aging population. Excessive osteoclast formation and bone resorption lead to changes in trabecular bone volume and architecture, leaving the bones vulnerable to fracture. Therapeutic approaches of inhibiting osteoclastogenesis and bone resorption have been proven to be an efficient approach to prevent osteoporosis. In our study, we have demonstrated for the first time that Loureirin B (LrB) inhibits ovariectomized osteoporosis and explored its underlying mechanisms of action in vitro. Methods: We examined the effects of LrB on RANKL-induced osteoclast differentiation and bone resorption, and its impacts on RANKL-induced NFATc1 activation, calcium oscillations and reactive oxygen species (ROS) production in osteoclasts in vitro. We assessed the in vivo efficacy of LrB using an ovariectomy (OVX)-induced osteoporosis model, which was analyzed using micro-computed tomography (micro-CT) and bone histomorphometry. Results: We found that LrB represses osteoclastogenesis, bone resorption, F-actin belts formation, osteoclast specific gene expressions, ROS activity and calcium oscillations through preventing NFATc1 translocation and expression as well as affecting MAPK-NFAT signaling pathways in vitro. Our in vivo study indicated that LrB prevents OVX-induced osteoporosis and preserves bone volume by repressing osteoclast activity and function. Conclusions: Our findings confirm that LrB can attenuate osteoclast formation and OVX-induced osteoporosis. This novel and exciting discovery could pave the way for the development of LrB as a potential therapeutic treatment for osteoporosis.
Article
Due to the rapid emergence of antibiotic-resistant bacteria, there is a growing need to discover new antibiotics. To address this challenge, we trained a deep neural network capable of predicting molecules with antibacterial activity. We performed predictions on multiple chemical libraries and discovered a molecule from the Drug Repurposing Hub-halicin-that is structurally divergent from conventional antibiotics and displays bactericidal activity against a wide phylogenetic spectrum of pathogens including Mycobacterium tuberculosis and carbapenem-resistant Enterobacteriaceae. Halicin also effectively treated Clostridioides difficile and pan-resistant Acinetobacter baumannii infections in murine models. Additionally, from a discrete set of 23 empirically tested predictions from >107 million molecules curated from the ZINC15 database, our model identified eight antibacterial compounds that are structurally distant from known antibiotics. This work highlights the utility of deep learning approaches to expand our antibiotic arsenal through the discovery of structurally distinct antibacterial molecules.
Conference Paper
Deep generative models have been wildly successful at learning coherent latent representations for continuous data such as natural images, artwork, and audio. However, generative modeling of discrete data such as arithmetic expressions and molecular structures still poses significant challenges. Crucially, state-of-the-art methods often produce outputs that are not valid. We make the key observation that frequently, discrete data can be represented as a parse tree from a context-free grammar. We propose a variational autoencoder which directly encodes from and decodes to these parse trees, ensuring the generated outputs are always syntactically valid. Surprisingly, we show that not only does our model more often generate valid outputs, it also learns a more coherent latent space in which nearby points decode to similar discrete outputs. We demonstrate the effectiveness of our learned models by showing their improved performance in Bayesian optimization for symbolic regression and molecule generation.
Article
Purpose: Extracellular signal-regulated kinase 1 and 2 (ERK1/2) signaling can be dysregulated in cancer. GDC-0994 is an oral inhibitor of ERK1/2. A first-in-human, Phase I dose escalation study of GDC-0994 was conducted in patients with locally advanced or metastatic solid tumors. Experimental design: GDC-0994 was administered once daily on a 21-day on/7-day off schedule to evaluate safety, pharmacokinetics, and preliminary signs of efficacy. Patients with pancreatic adenocarcinoma and BRAF-mutant colorectal cancer (CRC) were enrolled in the expansion stage. Results: Forty-seven patients were enrolled in 6 successive cohorts (50-800 mg). A single DLT of Grade 3 rash occurred at 600 mg. The most common drug-related adverse events (AE) were diarrhea, rash, nausea, fatigue, and vomiting. PK data showed dose-proportional increases in exposure, with a mean half-life of 23 hours, supportive of once daily dosing. In evaluable paired biopsies, MAPK pathway inhibition ranged from 19-51%. Partial metabolic responses by FDG-PET were observed in 11/20 patients across dose levels in multiple tumor types. Overall, 15/45 (33%) patients had a best overall response of stable disease and 2 patients with BRAF-mutant CRC had a confirmed partial response. Conclusions: GDC-0994 had an acceptable safety profile and pharmacodynamic effects were observed by FDG-PET and in serial tumor biopsies. Single agent activity was observed in two patients with BRAF-mutant CRC.
Article
Isoginkgetin is a biflavonoid compound isolated from the leaf extracts of Ginkgo biloba. In this study, an liquid chromatography‐tandem mass spectrometry (LC/MS/MS) with liquid‐liquid extraction was developed and validated for the analysis of isoginkgetin in rat plasma. In the process of chromatographic separation, selected reaction monitoring transitions for isoginkgetin and IS were m/z 566.8→134.7 and m/z 430.8→269.3, respectively. The validation parameters including selectivity, linearity, LLOQ, accuracy, precision, matrix effect, stability and recovery were satisfactory. The intra‐ and inter‐batch precision (RSD%) were below 12.1% in plasma, while the accuracy (RE%) was within ±14.3%. This method was employed to the pharmacokinetic study on rats after the intravenous administration of isoginkgetin.
Article
Caloric excess and sedentary lifestyle have led to a global epidemic of obesity and metabolic syndrome. The hepatic consequence of metabolic syndrome and obesity, nonalcoholic fatty liver disease (NAFLD), is estimated to affect up to one-third of the adult population in many developed and developing countries. This spectrum of liver disease ranges from simple steatosis to nonalcoholic steatohepatitis (NASH) and cirrhosis. Owing to the high prevalence of NAFLD, especially in industrialized countries but also worldwide, and the consequent burden of progressive liver disease, there is mounting epidemiological evidence that NAFLD has rapidly become a leading aetiology underlying many cases of hepatocellular carcinoma (HCC). In this Review, we discuss NAFLD-associated HCC, including its epidemiology, the key features of the hepatic NAFLD microenvironment (for instance, adaptive and innate immune responses) that promote hepatocarcinogenesis and the management of HCC in patients with obesity and associated metabolic comorbidities. The challenges and future directions of research will also be discussed, including clinically relevant biomarkers for early detection, treatment stratification and monitoring as well as approaches to therapies for both prevention and treatment in those at risk or presenting with NAFLD-associated HCC.