scholarly journals Predicting effective drug combinations using gradient tree boosting based on features extracted from drug-protein heterogeneous network

2019 ◽  
Vol 20 (1) ◽  
Author(s):  
Hui Liu ◽  
Wenhao Zhang ◽  
Lixia Nie ◽  
Xiancheng Ding ◽  
Judong Luo ◽  
...  

Abstract Background Although targeted drugs have contributed to impressive advances in the treatment of cancer patients, their clinical benefits on tumor therapies are greatly limited due to intrinsic and acquired resistance of cancer cells against such drugs. Drug combinations synergistically interfere with protein networks to inhibit the activity level of carcinogenic genes more effectively, and therefore play an increasingly important role in the treatment of complex disease. Results In this paper, we combined the drug similarity network, protein similarity network and known drug-protein associations into a drug-protein heterogenous network. Next, we ran random walk with restart (RWR) on the heterogenous network using the combinatorial drug targets as the initial probability, and obtained the converged probability distribution as the feature vector of each drug combination. Taking these feature vectors as input, we trained a gradient tree boosting (GTB) classifier to predict new drug combinations. We conducted performance evaluation on the widely used drug combination data set derived from the DCDB database. The experimental results show that our method outperforms seven typical classifiers and traditional boosting algorithms. Conclusions The heterogeneous network-derived features introduced in our method are more informative and enriching compared to the primary ontology features, which results in better performance. In addition, from the perspective of network pharmacology, our method effectively exploits the topological attributes and interactions of drug targets in the overall biological network, which proves to be a systematic and reliable approach for drug discovery.

2016 ◽  
Vol 2016 ◽  
pp. 1-11 ◽  
Author(s):  
Xiangyi Li ◽  
Guangrong Qin ◽  
Qingmin Yang ◽  
Lanming Chen ◽  
Lu Xie

Drug combination is a powerful and promising approach for complex disease therapy such as cancer and cardiovascular disease. However, the number of synergistic drug combinations approved by the Food and Drug Administration is very small. To bridge the gap between urgent need and low yield, researchers have constructed various models to identify synergistic drug combinations. Among these models, biomolecular network-based model is outstanding because of its ability to reflect and illustrate the relationships among drugs, disease-related genes, therapeutic targets, and disease-specific signaling pathways as a system. In this review, we analyzed and classified models for synergistic drug combination prediction in recent decade according to their respective algorithms. Besides, we collected useful resources including databases and analysis tools for synergistic drug combination prediction. It should provide a quick resource for computational biologists who work with network medicine or synergistic drug combination designing.


2021 ◽  
Author(s):  
Heming Zhang ◽  
Yixin Chen ◽  
Philip R Payne ◽  
Fuhai Li

Complex signaling pathways/networks are believed to be responsible for drug resistance in cancer therapy. Drug combinations inhibiting multiple signaling targets within cancer-related signaling networks have the potential to reduce drug resistance. Deep learning models have been reported to predict drug combinations. However, these models are hard to be interpreted in terms of mechanism of synergy (MoS), and thus cannot well support the human-AI based clinical decision making. Herein, we proposed a novel computational model, DeepSignalingFlow, which seeks to address the preceding two challenges. Specifically, a graph convolutional network (GCN) was developed based on a core cancer signaling network consisting of 1584 genes, with gene expression and copy number data derived from 46 core cancer signaling pathways. The novel up-stream signaling-flow (from up-stream signaling to drug targets), and the down-stream signaling-flow (from drug targets to down-stream signaling), were designed using trainable weights of network edges. The numerical features (accumulated information due to the signaling-flows of the signaling network) of drug nodes that link to drug targets were then used to predict the synergy scores of such drug combinations. The model was evaluated using the NCI ALMANAC drug combination screening data. The evaluation results showed that the proposed DeepSignalingFlow model can not only predict drug combination synergy score, but also interpret potentially interpretable MoS of drug combinations.


PeerJ ◽  
2017 ◽  
Vol 5 ◽  
pp. e3030 ◽  
Author(s):  
Frank Christian Kischkel ◽  
Julia Eich ◽  
Carina I. Meyer ◽  
Paula Weidemüller ◽  
Jens Krapfl ◽  
...  

Background To find the best individual chemotherapy for cancer patients, the efficacy of different chemotherapeutic drugs can be predicted by pretesting tumor samples in vitro via the chemotherapy-resistance (CTR)-Test®. Although drug combinations are widely used among cancer therapy, so far only single drugs are tested by this and other tests. However, several first line chemotherapies are combining two or more chemotherapeutics, leading to the necessity of drug combination testing methods. Methods We established a system to measure and predict the efficacy of chemotherapeutic drug combinations with the help of the Loewe additivity concept in combination with the CTR-test. A combination is measured by using half of the monotherapy’s concentration of both drugs simultaneously. With this method, the efficacy of a combination can also be calculated based on single drug measurements. Results The established system was tested on a data set of ovarian carcinoma samples using the combination carboplatin and paclitaxel and confirmed by using other tumor species and chemotherapeutics. Comparing the measured and the calculated values of the combination testings revealed a high correlation. Additionally, in 70% of the cases the measured and the calculated values lead to the same chemotherapeutic resistance category of the tumor. Conclusion Our data suggest that the best drug combination consists of the most efficient single drugs and the worst drug combination of the least efficient single drugs. Our results showed that single measurements are sufficient to predict combinations in specific cases but there are exceptions in which it is necessary to measure combinations, which is possible with the presented system.


2020 ◽  
Vol 20 (14) ◽  
pp. 1114-1131 ◽  
Author(s):  
Kanisha Shah ◽  
Rakesh M. Rawal

Cancer is a complex disease that has the ability to develop resistance to traditional therapies. The current chemotherapeutic treatment has become increasingly sophisticated, yet it is not 100% effective against disseminated tumours. Anticancer drugs resistance is an intricate process that ascends from modifications in the drug targets suggesting the need for better targeted therapies in the therapeutic arsenal. Advances in the modern techniques such as DNA microarray, proteomics along with the development of newer targeted drug therapies might provide better strategies to overcome drug resistance. This drug resistance in tumours can be attributed to an individual’s genetic differences, especially in tumoral somatic cells but acquired drug resistance is due to different mechanisms, such as cell death inhibition (apoptosis suppression) altered expression of drug transporters, alteration in drug metabolism epigenetic and drug targets, enhancing DNA repair and gene amplification. This review also focusses on the epigenetic modifications and microRNAs, which induce drug resistance and contributes to the formation of tumour progenitor cells that are not destroyed by conventional cancer therapies. Lastly, this review highlights different means to prevent the formation of drug resistant tumours and provides future directions for better treatment of these resistant tumours.


Bone Research ◽  
2021 ◽  
Vol 9 (1) ◽  
Author(s):  
Xiaowei Zhu ◽  
Weiyang Bai ◽  
Houfeng Zheng

AbstractOsteoporosis is a common skeletal disease, affecting ~200 million people around the world. As a complex disease, osteoporosis is influenced by many factors, including diet (e.g. calcium and protein intake), physical activity, endocrine status, coexisting diseases and genetic factors. In this review, we first summarize the discovery from genome-wide association studies (GWASs) in the bone field in the last 12 years. To date, GWASs and meta-analyses have discovered hundreds of loci that are associated with bone mineral density (BMD), osteoporosis, and osteoporotic fractures. However, the GWAS approach has sometimes been criticized because of the small effect size of the discovered variants and the mystery of missing heritability, these two questions could be partially explained by the newly raised conceptual models, such as omnigenic model and natural selection. Finally, we introduce the clinical use of GWAS findings in the bone field, such as the identification of causal clinical risk factors, the development of drug targets and disease prediction. Despite the fruitful GWAS discoveries in the bone field, most of these GWAS participants were of European descent, and more genetic studies should be carried out in other ethnic populations to benefit disease prediction in the corresponding population.


Author(s):  
Guizhou Hu ◽  
Martin M. Root

Background No methodology is currently available to allow the combining of individual risk factor information derived from different longitudinal studies for a chronic disease in a multivariate fashion. This paper introduces such a methodology, named Synthesis Analysis, which is essentially a multivariate meta-analytic technique. Design The construction and validation of statistical models using available data sets. Methods and results Two analyses are presented. (1) With the same data, Synthesis Analysis produced a similar prediction model to the conventional regression approach when using the same risk variables. Synthesis Analysis produced better prediction models when additional risk variables were added. (2) A four-variable empirical logistic model for death from coronary heart disease was developed with data from the Framingham Heart Study. A synthesized prediction model with five new variables added to this empirical model was developed using Synthesis Analysis and literature information. This model was then compared with the four-variable empirical model using the first National Health and Nutrition Examination Survey (NHANES I) Epidemiologic Follow-up Study data set. The synthesized model had significantly improved predictive power ( x2 = 43.8, P < 0.00001). Conclusions Synthesis Analysis provides a new means of developing complex disease predictive models from the medical literature.


2018 ◽  
Vol 62 (4) ◽  
Author(s):  
Suvitha Subramaniam ◽  
Christoph D. Schmid ◽  
Xue Li Guan ◽  
Pascal Mäser

ABSTRACT Combinatorial chemotherapy is necessary for the treatment of malaria. However, finding a suitable partner drug for a new candidate is challenging. Here we develop an algorithm that identifies all of the gene pairs of Plasmodium falciparum that possess orthologues in yeast that have a synthetic lethal interaction but are absent in humans. This suggests new options for drug combinations, particularly for inhibitors of targets such as P. falciparum calcineurin, cation ATPase 4, or phosphatidylinositol 4-kinase.


2016 ◽  
Vol 12 (2) ◽  
pp. 520-531 ◽  
Author(s):  
Xiao-Ying Yan ◽  
Shao-Wu Zhang ◽  
Song-Yao Zhang

By implementing label propagation on drug/target similarity network with mutual interaction information derived from drug–target heterogeneous network, LPMIHN algorithm identifies potential drug–target interactions.


2016 ◽  
Vol 40 (7) ◽  
pp. 867-881 ◽  
Author(s):  
Dingguo Yu ◽  
Nan Chen ◽  
Xu Ran

Purpose With the development and application of mobile internet access, social media represented by Weibo, WeChat, etc. has become the main channel for information release and sharing. High-impact users in social networks are key factors stimulating the large-scale propagation of information within social networks. User influence is usually related to the user’s attention rate, activity level, and message content. The paper aims to discuss these issues. Design/methodology/approach In this paper, the authors focused on Sina Weibo users, centered on users’ behavior and interactive information, and formulated a weighted interactive information network model, then present a novel computational model for Weibo user influence, which combined multiple indexes such as the user’s attention rate, activity level, and message content influence, etc., the model incorporated the time dimension, through the calculation of users’ attribute influence and interactive influence, to comprehensively measure the user influence of Sina Weibo users. Findings Compared with other models, the model reflected the dynamics and timeliness of the user influence in a more accurate way. Extensive experiments are conducted on the real-world data set, and the results validate the performance of the approach, and demonstrate the effectiveness of the dynamics and timeliness. Due to the similarity in platform architecture and user behavior between Sina Weibo and Twitter, the calculation model is also applicable to Twitter. Originality/value This paper presents a novel computational model for Weibo user influence, which combined multiple indexes such as the user’s attention rate, activity level, and message content influence, etc.


2022 ◽  
Author(s):  
Nurcan Tuncbag ◽  
Seyma Unsal Beyge

Abstract Heterogeneity across tumors is the main obstacle in developing treatment strategies. Drug molecules not only perturb their immediate protein targets but also modulate multiple signaling pathways. In this study, we explored the networks modulated by several drug molecules across multiple cancer cell lines by integrating the drug targets with transcriptomic and phosphoproteomic data. As a result, we obtained 236 reconstructed networks covering five cell lines and 70 drugs. A rigorous topological and pathway analysis showed that chemically and functionally different drugs may modulate overlapping networks. Additionally, we revealed a set of tumor-specific hidden pathways with the help of drug network models that are not detectable from the initial data. The difference in the target selectivity of the drugs leads to disjoint networks despite sharing the exact mechanism of action, e.g., HDAC inhibitors. We also used the reconstructed network models to study potential drug combinations based on the topological separation, found literature evidence for a set of drug pairs. Overall, the network-level exploration of the drug perturbations may potentially help optimize treatment strategies and suggest new drug combinations.


Sign in / Sign up

Export Citation Format

Share Document