1
|
Wu J, Lai J, Zhao X, Wang Z, Zhang Y, Wang L, Su Y, He Y, Li S, Jiang Y, Han J. DeepCCDS: Interpretable Deep Learning Framework for Predicting Cancer Cell Drug Sensitivity through Characterizing Cancer Driver Signals. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2416958. [PMID: 40397390 DOI: 10.1002/advs.202416958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/18/2025] [Indexed: 05/22/2025]
Abstract
Accurate characterization of cellular states is the foundation for precise prediction of drug sensitivity in cancer cell lines, which in turn is fundamental to realizing precision oncology. However, current deep learning approaches have limitations in characterizing cellular states. They rely solely on isolated genetic markers, overlooking the complex regulatory networks and cellular mechanisms that underlie drug responses. To address this limitation, this work proposes DeepCCDS, a Deep learning framework for Cancer Cell Drug Sensitivity prediction through Characterizing Cancer Driver Signals. DeepCCDS incorporates a prior knowledge network to characterize cancer driver signals, building upon the self-supervised neural network framework. The signals can reflect key mechanisms influencing cancer cell development and drug response, enhancing the model's predictive performance and interpretability. DeepCCDS has demonstrated superior performance in predicting drug sensitivity compared to previous state-of-the-art approaches across multiple datasets. Benefiting from integrating prior knowledge, DeepCCDS exhibits powerful feature representation capabilities and interpretability. Based on these feature representations, we have identified embedding features that could potentially be used for drug screening in new indications. Further, this work demonstrates the applicability of DeepCCDS on solid tumor samples from The Cancer Genome Atlas. This work believes integrating DeepCCDS into clinical decision-making processes can potentially improve the selection of personalized treatment strategies for cancer patients.
Collapse
Affiliation(s)
- Jiashuo Wu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Jiyin Lai
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Xilong Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Ziyi Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yongbao Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Liqiang Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yinchun Su
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yalan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Siyuan Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Ying Jiang
- College of Basic Medical Science, Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Junwei Han
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| |
Collapse
|
2
|
Akhtar M, Nehal N, Gull A, Parveen R, Khan S, Khan S, Ali J. Explicating the transformative role of artificial intelligence in designing targeted nanomedicine. Expert Opin Drug Deliv 2025:1-21. [PMID: 40321117 DOI: 10.1080/17425247.2025.2502022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/01/2025] [Indexed: 05/22/2025]
Abstract
INTRODUCTION Artificial intelligence (AI) has emerged as a transformative force in nanomedicine, revolutionizing drug delivery, diagnostics, and personalized treatment. While nanomedicine offers precise targeted drug delivery and reduced toxic effects, its clinical translation is hindered by biological complexity, unpredictable in vivo behavior, and inefficient trial-and-error approaches. AREAS COVERED This review covers the application of AI and Machine Learning (ML) across the nanomedicine development pipeline, starting from drug and target identification to nanoparticle design, toxicity prediction, and personalized dosing. Different AI/ML models like QSAR, MTK-QSBER, and Alchemite, along with data sources and high-throughput screening methods, have been explored. Real-world applications are critically discussed, including AI-assisted drug repurposing, controlled-release formulations, and cancer-specific delivery systems. EXPERT OPINION AI has emerged as an essential component in designing next-generation nanomedicine. Efficiently handling multidimensional datasets, optimizing formulations, and personalizing treatment regimens, it has sped up the innovation process. However, challenges like data heterogeneity, model transparency, and regulatory gaps remain. Addressing these hurdles through interdisciplinary efforts and emerging innovations like explainable AI and federated learning will pave the way for the clinical translation of AI-driven nanomedicine.
Collapse
Affiliation(s)
- Masheera Akhtar
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, New Delhi, India
| | - Nida Nehal
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, New Delhi, India
| | - Azka Gull
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, New Delhi, India
| | - Rabea Parveen
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, New Delhi, India
| | - Sana Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, New Delhi, India
| | - Saba Khan
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, New Delhi, India
| | - Javed Ali
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, New Delhi, India
| |
Collapse
|
3
|
He Y, Li S, Lan H, Long W, Zhai S, Li M, Wen Z. A Transfer Learning Framework for Predicting and Interpreting Drug Responses via Single-Cell RNA-Seq Data. Int J Mol Sci 2025; 26:4365. [PMID: 40362602 PMCID: PMC12072357 DOI: 10.3390/ijms26094365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 04/29/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Chemotherapy is a fundamental therapy in cancer treatment, yet its effectiveness is often undermined by drug resistance. Understanding the molecular mechanisms underlying drug response remains a major challenge due to tumor heterogeneity, complex cellular interactions, and limited access to clinical samples, which also hinder the performance and interpretability of existing predictive models. Meanwhile, single-cell RNA sequencing (scRNA-seq) has emerged as a powerful tool for uncovering resistance mechanisms, but the systematic collection and utilization of scRNA-seq drug response data remain limited. In this study, we collected scRNA-seq drug response datasets from publicly available web sources and proposed a transfer learning-based framework to align bulk and single cell sequencing data. A shared encoder was designed to project both bulk and single-cell sequencing data into a unified latent space for drug response prediction, while a sparse decoder guided by prior biological knowledge enhanced interpretability by mapping latent features to predefined pathways. The proposed model achieved superior performance across five curated scRNA-seq datasets and yielded biologically meaningful insights through integrated gradient analysis. This work demonstrates the potential of deep learning to advance drug response prediction and underscores the value of scRNA-seq data in supporting related research.
Collapse
Affiliation(s)
- Yujie He
- College of Chemistry, Sichuan University, Chengdu 610064, China; (Y.H.)
| | - Shenghao Li
- College of Chemistry, Sichuan University, Chengdu 610064, China; (Y.H.)
| | - Hao Lan
- College of Chemistry, Sichuan University, Chengdu 610064, China; (Y.H.)
| | - Wulin Long
- College of Chemistry, Sichuan University, Chengdu 610064, China; (Y.H.)
| | - Shengqiu Zhai
- College of Chemistry, Sichuan University, Chengdu 610064, China; (Y.H.)
| | - Menglong Li
- College of Chemistry, Sichuan University, Chengdu 610064, China; (Y.H.)
| | - Zhining Wen
- College of Chemistry, Sichuan University, Chengdu 610064, China; (Y.H.)
- Medical Big Data Center, Sichuan University, Chengdu 610064, China
| |
Collapse
|
4
|
Ye Q, Zeng Y, Jiang L, Kang Y, Pan P, Chen J, Deng Y, Zhao H, He S, Hou T, Hsieh C. A Knowledge-Guided Graph Learning Approach Bridging Phenotype- and Target-Based Drug Discovery. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412402. [PMID: 40047372 PMCID: PMC12021103 DOI: 10.1002/advs.202412402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Revised: 01/24/2025] [Indexed: 04/26/2025]
Abstract
Discovering therapeutic molecules requires the integration of both phenotype-based drug discovery (PDD) and target-based drug discovery (TDD). However, this integration remains challenging due to the inherent heterogeneity, noise, and bias present in biomedical data. In this study, Knowledge-Guided Drug Relational Predictor (KGDRP), a graph representation learning approach is developed that effectively integrates multimodal biomedical data, including network data containing biological system information, gene expression data, and sequence data that incorporates chemical molecular structures, all within a heterogeneous graph (HG) structure. By incorporating biomedical HG (BioHG) into a heterogeneous graph neural network (HGNN)-based architecture, KGDRP exhibits a remarkable 12% improvement compared to previous methods in real-world screening scenarios. Notably, the biology-informed representation, derived from KGDRP, significantly enhance target prioritization by 26% in drug target discovery. Furthermore, zero-shot evaluation on COVID-19 exhibited a notably higher success rate in identifying diverse potential drugs. The utilization of BioHG facilitates a unique KGDRP-based analysis of cell-target-drug interactions, thereby enabling the elucidation of drug mechanisms. Overall, KGDRP provides a robust infrastructure for the seamlessly integration of multimodal data and biomedical networks, effectively accelerating PDD, guiding therapeutic target discovery, and ultimately expediting therapeutic molecule discovery.
Collapse
Affiliation(s)
- Qing Ye
- College of Control Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Yundian Zeng
- College of Control Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Linlong Jiang
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Yu Kang
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Peichen Pan
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Jiming Chen
- College of Control Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Yafeng Deng
- CarbonSilicon AI Technology Co., LtdHangzhouZhejiang310018China
| | - Haitao Zhao
- Center for Intelligent and Biomimetic SystemsShenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdong440305China
| | - Shibo He
- College of Control Science and EngineeringZhejiang UniversityHangzhouZhejiang310027China
| | - Tingjun Hou
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| | - Chang‐Yu Hsieh
- College of Pharmaceutical SciencesZhejiang UniversityHangzhouZhejiang310058China
| |
Collapse
|
5
|
Codicè F, Pancotti C, Rollo C, Moreau Y, Fariselli P, Raimondi D. The specification game: rethinking the evaluation of drug response prediction for precision oncology. J Cheminform 2025; 17:33. [PMID: 40087708 PMCID: PMC11907791 DOI: 10.1186/s13321-025-00972-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 02/13/2025] [Indexed: 03/17/2025] Open
Abstract
Precision oncology plays a pivotal role in contemporary healthcare, aiming to optimize treatments for each patient based on their unique characteristics. This objective has spurred the emergence of various cancer cell line drug response datasets, driven by the need to facilitate pre-clinical studies by exploring the impact of multi-omics data on drug response. Despite the proliferation of machine learning models for Drug Response Prediction (DRP), their validation remains critical to reliably assess their usefulness for drug discovery, precision oncology and their actual ability to generalize over the immense space of cancer cells and chemical compounds. Scientific contribution In this paper we show that the commonly used evaluation strategies for DRP methods can be easily fooled by commonly occurring dataset biases, and they are therefore not able to truly measure the ability of DRP methods to generalize over drugs and cell lines ("specification gaming"). This problem hinders the development of reliable DRP methods and their application to experimental pipelines. Here we propose a new validation protocol composed by three Aggregation Strategies (Global, Fixed-Drug, and Fixed-Cell Line) integrating them with three of the most commonly used train-test evaluation settings, to ensure a truly realistic assessment of the prediction performance. We also scrutinize the challenges associated with using IC50 as a prediction label, showing how its close correlation with the drug concentration ranges worsens the risk of misleading performance assessment, and we indicate an additional reason to replace it with the Area Under the Dose-Response Curve instead.
Collapse
Affiliation(s)
- Francesco Codicè
- Department of Medical Sciences, University of Torino, 10123, Torino, Italy.
| | - Corrado Pancotti
- Department of Medical Sciences, University of Torino, 10123, Torino, Italy
| | - Cesare Rollo
- Department of Medical Sciences, University of Torino, 10123, Torino, Italy
| | - Yves Moreau
- ESAT-STADIUS, KU Leuven, Leuven, 3001, Belgium
| | - Piero Fariselli
- Department of Medical Sciences, University of Torino, 10123, Torino, Italy
| | - Daniele Raimondi
- Institut de Génétique Moléculaire de Montpellier, Université de Montpellier, 34293, Montpellier, France
| |
Collapse
|
6
|
Narykov O, Zhu Y, Brettin T, Evrard YA, Partin A, Xia F, Shukla M, Vasanthakumari P, Doroshow JH, Stevens RL. Data imbalance in drug response prediction: multi-objective optimization approach in deep learning setting. Brief Bioinform 2025; 26:bbaf134. [PMID: 40178282 PMCID: PMC11966611 DOI: 10.1093/bib/bbaf134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 04/05/2025] Open
Abstract
Drug response prediction (DRP) methods tackle the complex task of associating the effectiveness of small molecules with the specific genetic makeup of the patient. Anti-cancer DRP is a particularly challenging task requiring costly experiments as underlying pathogenic mechanisms are broad and associated with multiple genomic pathways. The scientific community has exerted significant efforts to generate public drug screening datasets, giving a path to various machine learning models that attempt to reason over complex data space of small compounds and biological characteristics of tumors. However, the data depth is still lacking compared to application domains like computer vision or natural language processing domains, limiting current learning capabilities. To combat this issue and improves the generalizability of the DRP models, we are exploring strategies that explicitly address the imbalance in the DRP datasets. We reframe the problem as a multi-objective optimization across multiple drugs to maximize deep learning model performance. We implement this approach by constructing Multi-Objective Optimization Regularized by Loss Entropy loss function and plugging it into a Deep Learning model. We demonstrate the utility of proposed drug discovery methods and make suggestions for further potential application of the work to achieve desirable outcomes in the healthcare field.
Collapse
Affiliation(s)
- Oleksandr Narykov
- Computing, Environment and Life Sciences, Argonne National Laboratory, 9700 S Cass Ave, Lemont, IL 60439, United States
| | - Yitan Zhu
- Computing, Environment and Life Sciences, Argonne National Laboratory, 9700 S Cass Ave, Lemont, IL 60439, United States
| | - Thomas Brettin
- Computing, Environment and Life Sciences, Argonne National Laboratory, 9700 S Cass Ave, Lemont, IL 60439, United States
| | - Yvonne A Evrard
- Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, 8560 Progress Drive, Frederick, MD 21702, United States
| | - Alexander Partin
- Computing, Environment and Life Sciences, Argonne National Laboratory, 9700 S Cass Ave, Lemont, IL 60439, United States
| | - Fangfang Xia
- Computing, Environment and Life Sciences, Argonne National Laboratory, 9700 S Cass Ave, Lemont, IL 60439, United States
| | - Maulik Shukla
- Computing, Environment and Life Sciences, Argonne National Laboratory, 9700 S Cass Ave, Lemont, IL 60439, United States
| | - Priyanka Vasanthakumari
- Computing, Environment and Life Sciences, Argonne National Laboratory, 9700 S Cass Ave, Lemont, IL 60439, United States
| | - James H Doroshow
- Developmental Therapeutics Branch, National Cancer Institute, 31 Center Dr, Bethesda, MD 20892, United States
| | - Rick L Stevens
- Computing, Environment and Life Sciences, Argonne National Laboratory, 9700 S Cass Ave, Lemont, IL 60439, United States
- Department of Computer Science, The University of Chicago, 5730 S Ellis Ave, Chicago, IL 60637, United States
| |
Collapse
|
7
|
Wang J, Zeng Z, Li Z, Liu G, Zhang S, Luo C, Hu S, Wan S, Zhao L. The clinical application of artificial intelligence in cancer precision treatment. J Transl Med 2025; 23:120. [PMID: 39871340 PMCID: PMC11773911 DOI: 10.1186/s12967-025-06139-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/14/2025] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Artificial intelligence has made significant contributions to oncology through the availability of high-dimensional datasets and advances in computing and deep learning. Cancer precision medicine aims to optimize therapeutic outcomes and reduce side effects for individual cancer patients. However, a comprehensive review describing the impact of artificial intelligence on cancer precision medicine is lacking. OBSERVATIONS By collecting and integrating large volumes of data and applying it to clinical tasks across various algorithms and models, artificial intelligence plays a significant role in cancer precision medicine. Here, we describe the general principles of artificial intelligence, including machine learning and deep learning. We further summarize the latest developments in artificial intelligence applications in cancer precision medicine. In tumor precision treatment, artificial intelligence plays a crucial role in individualizing both conventional and emerging therapies. In specific fields, including target prediction, targeted drug generation, immunotherapy response prediction, neoantigen prediction, and identification of long non-coding RNA, artificial intelligence offers promising perspectives. Finally, we outline the current challenges and ethical issues in the field. CONCLUSIONS Recent clinical studies demonstrate that artificial intelligence is involved in cancer precision medicine and has the potential to benefit cancer healthcare, particularly by optimizing conventional therapies, emerging targeted therapies, and individual immunotherapies. This review aims to provide valuable resources to clinicians and researchers and encourage further investigation in this field.
Collapse
Affiliation(s)
- Jinyu Wang
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Ziyi Zeng
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
- Department of Neonatology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Zehua Li
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Guangyue Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, China
| | - Shunhong Zhang
- Department of Cardiology, Panzhihua Iron and Steel Group General Hospital, Panzhihua, China
| | - Chenchen Luo
- Department of Outpatient Chengbei, the Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
| | - Saidi Hu
- Department of Stomatology, Yaan people's Hospital, Yaan, China
| | - Siran Wan
- Department of Gynaecology and Obstetrics, Yaan people's Hospital, Yaan, China
| | - Linyong Zhao
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy / Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
8
|
Prathaban K, Hande MP. Transforming Healthcare: Artificial Intelligence (AI) Applications in Medical Imaging and Drug Response Prediction. Genome Integr 2025; 15:e20240002. [PMID: 39845982 PMCID: PMC11752870 DOI: 10.14293/genint.15.1.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025] Open
Abstract
Artificial intelligence (AI) offers a broad range of enhancements in medicine. Machine learning and deep learning techniques have shown significant potential in improving diagnosis and treatment outcomes, from assisting clinicians in diagnosing medical images to ascertaining effective drugs for a specific disease. Despite the prospective benefits, adopting AI in clinical settings requires careful consideration, particularly concerning data generalisation and model explainability. This commentary aims to discuss two potential use cases for AI in the field of medicine and the overarching challenges involved in their implementation.
Collapse
Affiliation(s)
- Karthik Prathaban
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - M. Prakash Hande
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
9
|
Xu K, Wang M, Zou X, Liu J, Wei A, Chen J, Tang C. HSTrans: Homogeneous substructures transformer for predicting frequencies of drug-side effects. Neural Netw 2025; 181:106779. [PMID: 39488108 DOI: 10.1016/j.neunet.2024.106779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 08/29/2024] [Accepted: 10/01/2024] [Indexed: 11/04/2024]
Abstract
Identifying the frequencies of drug-side effects is crucial for assessing drug risk-benefit. However, accurately determining these frequencies remains challenging due to the limitations of time and scale in clinical randomized controlled trials. As a result, several computational methods have been proposed to address these issues. Nonetheless, two primary problems still persist. Firstly, most of these methods face challenges in generating accurate predictions for novel drugs, as they heavily depend on the interaction graph between drugs and side effects (SEs) within their modeling framework. Secondly, some previous methods often simply concatenate the features of drugs and SEs, which fails to effectively capture their underlying association. In this work, we present HSTrans, a novel approach that treats drugs and SEs as sets of substructures, leveraging a transformer encoder for unified substructure embedding and incorporating an interaction module for association capture. Specifically, HSTrans extracts drug substructures through a specialized algorithm and identifies effective substructures for each SE by employing an indicator that measures the importance of each substructure and SE. Additionally, HSTrans applies convolutional neural network (CNN) in the interaction module to capture complex relationships between drugs and SEs. Experimental results on datasets from Galeano et al.'s study demonstrate that the proposed method outperforms other state-of-the-art approaches. The demo codes for HSTrans are available at https://github.com/Dtdtxuky/HSTrans/tree/master.
Collapse
Affiliation(s)
- Kaiyi Xu
- School of Computer Science, China University of Geosciences, Wuhan 430074, China
| | - Minhui Wang
- Department of Pharmacy, Lianshui People's Hospital Affiliated to Kangda College of Nanjing Medical University, Huai'an 223300, China
| | - Xin Zou
- School of Computer Science, China University of Geosciences, Wuhan 430074, China
| | - Jingjing Liu
- Department of Cardiac Surgery, Tianjin Chest Hospital, Tianjin 300222, China
| | - Ao Wei
- Department of Cardiology, Tianjin Chest Hospital, Tianjin 300222, China
| | - Jiajia Chen
- Department of Pharmacy, The Affiliated Huai'an Hospital of Xuzhou Medical University and The Second People's Hospital of Huai'an, Huai'an 223002, China.
| | - Chang Tang
- School of Computer Science, China University of Geosciences, Wuhan 430074, China.
| |
Collapse
|
10
|
Hajim WI, Zainudin S, Daud KM, Alheeti K. Golden eagle optimized CONV-LSTM and non-negativity-constrained autoencoder to support spatial and temporal features in cancer drug response prediction. PeerJ Comput Sci 2024; 10:e2520. [PMID: 39896419 PMCID: PMC11784781 DOI: 10.7717/peerj-cs.2520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 10/25/2024] [Indexed: 02/04/2025]
Abstract
Advanced machine learning (ML) and deep learning (DL) methods have recently been utilized in Drug Response Prediction (DRP), and these models use the details from genomic profiles, such as extensive drug screening data and cell line data, to predict the response of drugs. Comparatively, the DL-based prediction approaches provided better learning of such features. However, prior knowledge, like pathway data, is sometimes discarded as irrelevant since the drug response datasets are multidimensional and noisy. Optimized feature learning and extraction processes are suggested to handle this problem. First, the noise and class imbalance problems must be tackled to avoid low identification accuracy, long prediction times, and poor applicability. This article aims to apply the Non-Negativity-Constrained Auto Encoder (NNCAE) network to tackle these issues, enhance the adaptive search for the optimal size of sliding windows, and ensure that deep network architectures are adept at learning the vital hidden features. NNCAE methodology is used after performing the standard pre-processing procedures to handle the noise and class imbalance problem. This class balanced and noise-removed input data features are learned to train the proposed hybrid classifier. The classification model, Golden Eagle Optimization-based Convolutional Long Short-Term Memory neural networks (GEO-Conv-LSTM), is assembled by integrating Convolutional Neural Network CNN and LSTM models, with parameter tuning performed by the GEO algorithm. Evaluations are conducted on two large datasets from the Genomics of Drug Sensitivity in Cancer (GDSC) repository, and the proposed NNCAE-GEO-Conv-LSTM-based approach has achieved 96.99% and 97.79% accuracies, respectively, with reduced processing time and error rate for the DRP problem.
Collapse
Affiliation(s)
- Wesam Ibrahim Hajim
- Department of Applied Geology, College of Sciences, University of Tikrit, Tikrit, Salah ad Din, Iraq
- Center for Artificial Intelligence Technology, Faculty of Information Science and Technology, Universiti Kebangsaan Malaysia, Selangor, Malaysia
| | - Suhaila Zainudin
- Center for Artificial Intelligence Technology, Faculty of Information Science and Technology, Universiti Kebangsaan Malaysia, Selangor, Malaysia
| | - Kauthar Mohd Daud
- Center for Artificial Intelligence Technology, Faculty of Information Science and Technology, Universiti Kebangsaan Malaysia, Selangor, Malaysia
| | - Khattab Alheeti
- Department of Computer Networking Systems College of Computer Sciences and Information Technology, University of Anbar, Ramadi, Al Anbar, Iraq
| |
Collapse
|
11
|
Cai Z, Poulos RC, Aref A, Robinson PJ, Reddel RR, Zhong Q. DeePathNet: A Transformer-Based Deep Learning Model Integrating Multiomic Data with Cancer Pathways. CANCER RESEARCH COMMUNICATIONS 2024; 4:3151-3164. [PMID: 39530738 PMCID: PMC11652962 DOI: 10.1158/2767-9764.crc-24-0285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Revised: 10/10/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
SIGNIFICANCE DeePathNet integrates cancer-specific biological pathways using transformer-based deep learning for enhanced cancer analysis. It outperforms existing models in predicting drug responses, cancer types, and subtypes. By enabling pathway-level biomarker discovery, DeePathNet represents a significant advancement in cancer research and could lead to more effective treatments.
Collapse
Affiliation(s)
- Zhaoxiang Cai
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Rebecca C. Poulos
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Adel Aref
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Phillip J. Robinson
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Roger R. Reddel
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| | - Qing Zhong
- ProCan, Children’s Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, Australia
| |
Collapse
|
12
|
Jin I, Lee S, Schmuhalek M, Nam H. DD-PRiSM: a deep learning framework for decomposition and prediction of synergistic drug combinations. Brief Bioinform 2024; 26:bbae717. [PMID: 39800875 PMCID: PMC11725392 DOI: 10.1093/bib/bbae717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/16/2025] Open
Abstract
Combination therapies have emerged as a promising approach for treating complex diseases, particularly cancer. However, predicting the efficacy and safety profiles of these therapies remains a significant challenge, primarily because of the complex interactions among drugs and their wide-ranging effects. To address this issue, we introduce DD-PRiSM (Decomposition of Drug-Pair Response into Synergy and Monotherapy effect), a deep-learning pipeline that predicts the effects of combination therapy. DD-PRiSM consists of two predictive models. The first is the Monotherapy model, which predicts parameters of the drug response curve based on drug structure and cell line gene expression. This reconstructed curve is then used to predict cell viability at the given drug dosage. The second is the Combination therapy model, which predicts the efficacy of drug combinations by analyzing individual drug effects and their synergistic interactions with a specific dosage level of individual drugs. The efficacy of DD-PRiSM is demonstrated through its performance metrics, achieving a root mean square error of 0.0854, a Pearson correlation coefficient of 0.9063, and an R2 of 0.8209 for unseen pairs. Furthermore, DD-PRiSM distinguishes itself by its capability to decompose combination therapy efficacy, successfully identifying synergistic drug pairs. We demonstrated synergistic responses vary across cancer types and identified hub drugs that trigger synergistic effects. Finally, we suggested a promising drug pair through our case study.
Collapse
Affiliation(s)
- Iljung Jin
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST),Buk-gu, Gwangju 61005, Republic of Korea
| | - Songyeon Lee
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST),Buk-gu, Gwangju 61005, Republic of Korea
| | - Martin Schmuhalek
- AI Graduate School, Gwangju Institute of Science and Technology (GIST), Buk-gu, Gwangju 61005, Republic of Korea
| | - Hojung Nam
- School of Electrical Engineering and Computer Science, Gwangju Institute of Science and Technology (GIST),Buk-gu, Gwangju 61005, Republic of Korea
- AI Graduate School, Gwangju Institute of Science and Technology (GIST), Buk-gu, Gwangju 61005, Republic of Korea
- Center for AI-Applied High Efficiency Drug Discovery (AHEDD), Gwangju Institute of Science and Technology (GIST), Buk-gu, Gwangju 61005, Republic of Korea
| |
Collapse
|
13
|
Xia X, Zhu C, Zhong F, Liu L. TransCDR: a deep learning model for enhancing the generalizability of drug activity prediction through transfer learning and multimodal data fusion. BMC Biol 2024; 22:227. [PMID: 39385185 PMCID: PMC11462810 DOI: 10.1186/s12915-024-02023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/30/2024] [Indexed: 10/11/2024] Open
Abstract
BACKGROUND Accurate and robust drug response prediction is of utmost importance in precision medicine. Although many models have been developed to utilize the representations of drugs and cancer cell lines for predicting cancer drug responses (CDR), their performances can be improved by addressing issues such as insufficient data modality, suboptimal fusion algorithms, and poor generalizability for novel drugs or cell lines. RESULTS We introduce TransCDR, which uses transfer learning to learn drug representations and fuses multi-modality features of drugs and cell lines by a self-attention mechanism, to predict the IC50 values or sensitive states of drugs on cell lines. We are the first to systematically evaluate the generalization of the CDR prediction model to novel (i.e., never-before-seen) compound scaffolds and cell line clusters. TransCDR shows better generalizability than 8 state-of-the-art models. TransCDR outperforms its 5 variants that train drug encoders (i.e., RNN and AttentiveFP) from scratch under various scenarios. The most critical contributors among multiple drug notations and omics profiles are Extended Connectivity Fingerprint and genetic mutation. Additionally, the attention-based fusion module further enhances the predictive performance of TransCDR. TransCDR, trained on the GDSC dataset, demonstrates strong predictive performance on the external testing set CCLE. It is also utilized to predict missing CDRs on GDSC. Moreover, we investigate the biological mechanisms underlying drug response by classifying 7675 patients from TCGA into drug-sensitive or drug-resistant groups, followed by a Gene Set Enrichment Analysis. CONCLUSIONS TransCDR emerges as a potent tool with significant potential in drug response prediction.
Collapse
Affiliation(s)
- Xiaoqiong Xia
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Chaoyu Zhu
- Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China
| | - Fan Zhong
- Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China.
| | - Lei Liu
- Intelligent Medicine Institute, Fudan University, Shanghai, 200032, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai, 200120, China.
| |
Collapse
|
14
|
Hu X, Zhang P, Zhang J, Deng L. DeepFusionCDR: Employing Multi-Omics Integration and Molecule-Specific Transformers for Enhanced Prediction of Cancer Drug Responses. IEEE J Biomed Health Inform 2024; 28:6248-6258. [PMID: 38935469 DOI: 10.1109/jbhi.2024.3417014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Deep learning approaches have demonstrated remarkable potential in predicting cancer drug responses (CDRs), using cell line and drug features. However, existing methods predominantly rely on single-omics data of cell lines, potentially overlooking the complex biological mechanisms governing cell line responses. This paper introduces DeepFusionCDR, a novel approach employing unsupervised contrastive learning to amalgamate multi-omics features, including mutation, transcriptome, methylome, and copy number variation data, from cell lines. Furthermore, we incorporate molecular SMILES-specific transformers to derive drug features from their chemical structures. The unified multi-omics and drug signatures are combined, and a multi-layer perceptron (MLP) is applied to predict IC50 values for cell line-drug pairs. Moreover, this MLP can discern whether a cell line is resistant or sensitive to a particular drug. We assessed DeepFusionCDR's performance on the GDSC dataset and juxtaposed it against cutting-edge methods, demonstrating its superior performance in regression and classification tasks. We also conducted ablation studies and case analyses to exhibit the effectiveness and versatility of our proposed approach. Our results underscore the potential of DeepFusionCDR to enhance CDR predictions by harnessing the power of multi-omics fusion and molecular-specific transformers. The prediction of DeepFusionCDR on TCGA patient data and case study highlight the practical application scenarios of DeepFusionCDR in real-world environments.
Collapse
|
15
|
Saranya KR, Vimina ER. DRN-CDR: A cancer drug response prediction model using multi-omics and drug features. Comput Biol Chem 2024; 112:108175. [PMID: 39191166 DOI: 10.1016/j.compbiolchem.2024.108175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 08/09/2024] [Accepted: 08/14/2024] [Indexed: 08/29/2024]
Abstract
Cancer drug response (CDR) prediction is an important area of research that aims to personalize cancer therapy, optimizing treatment plans for maximum effectiveness while minimizing potential negative effects. Despite the advancements in Deep learning techniques, the effective integration of multi-omics data for drug response prediction remains challenging. In this paper, a regression method using Deep ResNet for CDR (DRN-CDR) prediction is proposed. We aim to explore the potential of considering sole cancer genes in drug response prediction. Here the multi-omics data such as gene expressions, mutation data, and methylation data along with the molecular structural information of drugs were integrated to predict the IC50 values of drugs. Drug features are extracted by employing a Uniform Graph Convolution Network, while Cell line features are extracted using a combination of Convolutional Neural Network and Fully Connected Networks. These features are then concatenated and fed into a deep ResNet for the prediction of IC50 values between Drug - Cell line pairs. The proposed method yielded higher Pearson's correlation coefficient (rp) of 0.7938 with lowest Root Mean Squared Error (RMSE) value of 0.92 when compared with similar methods of tCNNS, MOLI, DeepCDR, TGSA, NIHGCN, DeepTTA, GraTransDRP and TSGCNN. Further, when the model is extended to a classification problem to categorize drugs as sensitive or resistant, we achieved AUC and AUPR measures of 0.7623 and 0.7691, respectively. The drugs such as Tivozanib, SNX-2112, CGP-60474, PHA-665752, Foretinib etc., exhibited low median IC50 values and were found to be effective anti-cancer drugs. The case studies with different TCGA cancer types also revealed the effectiveness of SNX-2112, CGP-60474, Foretinib, Cisplatin, Vinblastine etc. This consistent pattern strongly suggests the effectiveness of the model in predicting CDR.
Collapse
Affiliation(s)
- K R Saranya
- Department of Computer Science and IT, School of Computing, Amrita Vishwa Vidyapeetham, Kochi Campus, India
| | - E R Vimina
- Department of Computer Science and IT, School of Computing, Amrita Vishwa Vidyapeetham, Kochi Campus, India.
| |
Collapse
|
16
|
Pak M, Bang D, Sung I, Kim S, Lee S. DGDRP: drug-specific gene selection for drug response prediction via re-ranking through propagating and learning biological network. Front Genet 2024; 15:1441558. [PMID: 39371421 PMCID: PMC11450864 DOI: 10.3389/fgene.2024.1441558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Introduction: Drug response prediction, especially in terms of cell viability prediction, is a well-studied research problem with significant implications for personalized medicine. It enables the identification of the most effective drugs based on individual genetic profiles, aids in selecting potential drug candidates, and helps identify biomarkers that predict drug efficacy and toxicity.A deeper investigation on drug response prediction reveals that drugs exert their effects by targeting specific proteins, which in turn perturb related genes in cascading ways. This perturbation affects cellular pathways and regulatory networks, ultimately influencing the cellular response to the drug. Identifying which genes are perturbed and how they interact can provide critical insights into the mechanisms of drug action. Hence, the problem of predicting drug response can be framed as a dual problem involving both the prediction of drug efficacy and the selection of drug-specific genes. Identifying these drug-specific genes (biomarkers) is crucial because they serve as indicators of how the drug will affect the biological system, thereby facilitating both drug response prediction and biomarker discovery.Methods: In this study, we propose DGDRP (Drug-specific Gene selection for Drug Response Prediction), a graph neural network (GNN)-based model that uses a novel rank-and-re-rank process for drug-specific gene selection. DGDRP first ranks genes using a pathway knowledge-enhanced network propagation algorithm based on drug target information, ensuring biological relevance. It then re-ranks genes based on the similarity between gene and drug target embeddings learned from the GNN, incorporating semantic relationships. Thus, our model adaptively learns to select drug mechanism-associated genes that contribute to drug response prediction. This integrated approach not only improves drug response predictions compared to other gene selection methods but also allows for effective biomarker discovery.Discussion: As a result, our approach demonstrates improved drug response predictions compared to other gene selection methods and demonstrates comparability with state-of-the-art deep learning models. Case studies further support our method by showing alignment of selected gene sets with the mechanisms of action of input drugs.Conclusion: Overall, DGDRP represents a deep learning based re-ranking strategy, offering a robust gene selection framework for more accurate drug response prediction. The source code for DGDRP can be found at: https://github.com/minwoopak/heteronet.
Collapse
Affiliation(s)
- Minwoo Pak
- Department of Computer Science and Engineering, Seoul National University, Seoul, Republic of Korea
| | - Dongmin Bang
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
- Aigendrug Co., Ltd., Seoul, Republic of Korea
| | - Inyoung Sung
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
| | - Sun Kim
- Department of Computer Science and Engineering, Seoul National University, Seoul, Republic of Korea
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, Seoul, Republic of Korea
| | - Sunho Lee
- Aigendrug Co., Ltd., Seoul, Republic of Korea
| |
Collapse
|
17
|
Ren Y, Wu C, Zhou H, Hu X, Miao Z. Dual-extraction modeling: A multi-modal deep-learning architecture for phenotypic prediction and functional gene mining of complex traits. PLANT COMMUNICATIONS 2024; 5:101002. [PMID: 38872306 DOI: 10.1016/j.xplc.2024.101002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 05/27/2024] [Accepted: 06/11/2024] [Indexed: 06/15/2024]
Abstract
Despite considerable advances in extracting crucial insights from bio-omics data to unravel the intricate mechanisms underlying complex traits, the absence of a universal multi-modal computational tool with robust interpretability for accurate phenotype prediction and identification of trait-associated genes remains a challenge. This study introduces the dual-extraction modeling (DEM) approach, a multi-modal deep-learning architecture designed to extract representative features from heterogeneous omics datasets, enabling the prediction of complex trait phenotypes. Through comprehensive benchmarking experiments, we demonstrate the efficacy of DEM in classification and regression prediction of complex traits. DEM consistently exhibits superior accuracy, robustness, generalizability, and flexibility. Notably, we establish its effectiveness in predicting pleiotropic genes that influence both flowering time and rosette leaf number, underscoring its commendable interpretability. In addition, we have developed user-friendly software to facilitate seamless utilization of DEM's functions. In summary, this study presents a state-of-the-art approach with the ability to effectively predict qualitative and quantitative traits and identify functional genes, confirming its potential as a valuable tool for exploring the genetic basis of complex traits.
Collapse
Affiliation(s)
- Yanlin Ren
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Chenhua Wu
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - He Zhou
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Xiaona Hu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shaanxi 712100, China.
| | - Zhenyan Miao
- State Key Laboratory for Crop Stress Resistance and High-Efficiency Production, Center of Bioinformatics, College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China; Key Laboratory of Biology and Genetics Improvement of Maize in Arid Area of Northwest Region, Ministry of Agriculture, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|
18
|
Huang Z, Fan Z, Shen S, Wu M, Deng L. MolMVC: Enhancing molecular representations for drug-related tasks through multi-view contrastive learning. Bioinformatics 2024; 40:ii190-ii197. [PMID: 39230706 PMCID: PMC11373324 DOI: 10.1093/bioinformatics/btae386] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/05/2024] Open
Abstract
MOTIVATION Effective molecular representation is critical in drug development. The complex nature of molecules demands comprehensive multi-view representations, considering 1D, 2D, and 3D aspects, to capture diverse perspectives. Obtaining representations that encompass these varied structures is crucial for a holistic understanding of molecules in drug-related contexts. RESULTS In this study, we introduce an innovative multi-view contrastive learning framework for molecular representation, denoted as MolMVC. Initially, we use a Transformer encoder to capture 1D sequence information and a Graph Transformer to encode the intricate 2D and 3D structural details of molecules. Our approach incorporates a novel attention-guided augmentation scheme, leveraging prior knowledge to create positive samples tailored to different molecular data views. To align multi-view molecular positive samples effectively in latent space, we introduce an adaptive multi-view contrastive loss (AMCLoss). In particular, we calculate AMCLoss at various levels within the model to effectively capture the hierarchical nature of the molecular information. Eventually, we pre-train the encoders via minimizing AMCLoss to obtain the molecular representation, which can be used for various down-stream tasks. In our experiments, we evaluate the performance of our MolMVC on multiple tasks, including molecular property prediction (MPP), drug-target binding affinity (DTA) prediction and cancer drug response (CDR) prediction. The results demonstrate that the molecular representation learned by our MolMVC can enhance the predictive accuracy on these tasks and also reduce the computational costs. Furthermore, we showcase MolMVC's efficacy in drug repositioning across a spectrum of drug-related applications. AVAILABILITY AND IMPLEMENTATION The code and pre-trained model are publicly available at https://github.com/Hhhzj-7/MolMVC.
Collapse
Affiliation(s)
- Zhijian Huang
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Ziyu Fan
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Siyuan Shen
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Min Wu
- Institute for Infocomm Research, Agency for Science, Technology and Research (A*STAR), Singapore 138632, Singapore
| | - Lei Deng
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
19
|
Mohammadzadeh-Vardin T, Ghareyazi A, Gharizadeh A, Abbasi K, Rabiee HR. DeepDRA: Drug repurposing using multi-omics data integration with autoencoders. PLoS One 2024; 19:e0307649. [PMID: 39058696 PMCID: PMC11280260 DOI: 10.1371/journal.pone.0307649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Cancer treatment has become one of the biggest challenges in the world today. Different treatments are used against cancer; drug-based treatments have shown better results. On the other hand, designing new drugs for cancer is costly and time-consuming. Some computational methods, such as machine learning and deep learning, have been suggested to solve these challenges using drug repurposing. Despite the promise of classical machine-learning methods in repurposing cancer drugs and predicting responses, deep-learning methods performed better. This study aims to develop a deep-learning model that predicts cancer drug response based on multi-omics data, drug descriptors, and drug fingerprints and facilitates the repurposing of drugs based on those responses. To reduce multi-omics data's dimensionality, we use autoencoders. As a multi-task learning model, autoencoders are connected to MLPs. We extensively tested our model using three primary datasets: GDSC, CTRP, and CCLE to determine its efficacy. In multiple experiments, our model consistently outperforms existing state-of-the-art methods. Compared to state-of-the-art models, our model achieves an impressive AUPRC of 0.99. Furthermore, in a cross-dataset evaluation, where the model is trained on GDSC and tested on CCLE, it surpasses the performance of three previous works, achieving an AUPRC of 0.72. In conclusion, we presented a deep learning model that outperforms the current state-of-the-art regarding generalization. Using this model, we could assess drug responses and explore drug repurposing, leading to the discovery of novel cancer drugs. Our study highlights the potential for advanced deep learning to advance cancer therapeutic precision.
Collapse
Affiliation(s)
- Taha Mohammadzadeh-Vardin
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| | - Amin Ghareyazi
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| | - Ali Gharizadeh
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| | - Karim Abbasi
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
- Faculty of Mathematics and Computer Science, Kharazmi University, Tehran, Iran
| | - Hamid R. Rabiee
- Department of Computer Engineering, Bioinformatics and Computational Biology Lab, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
20
|
Bang D, Koo B, Kim S. Transfer learning of condition-specific perturbation in gene interactions improves drug response prediction. Bioinformatics 2024; 40:i130-i139. [PMID: 38940127 PMCID: PMC11256952 DOI: 10.1093/bioinformatics/btae249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024] Open
Abstract
SUMMARY 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.
Collapse
Affiliation(s)
- Dongmin Bang
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
- AIGENDRUG Co., Ltd., Seoul, 08758, Republic of Korea
| | - Bonil Koo
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
- AIGENDRUG Co., Ltd., Seoul, 08758, Republic of Korea
| | - Sun Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, 08826, Republic of Korea
- AIGENDRUG Co., Ltd., Seoul, 08758, Republic of Korea
- Department of Computer Science and Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- Interdisciplinary Program in Artificial Intelligence, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
21
|
Abbasi M, Carvalho FG, Ribeiro B, Arrais JP. Predicting drug activity against cancer through genomic profiles and SMILES. Artif Intell Med 2024; 150:102820. [PMID: 38553160 DOI: 10.1016/j.artmed.2024.102820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/09/2024] [Accepted: 02/21/2024] [Indexed: 04/02/2024]
Abstract
Due to the constant increase in cancer rates, the disease has become a leading cause of death worldwide, enhancing the need for its detection and treatment. In the era of personalized medicine, the main goal is to incorporate individual variability in order to choose more precisely which therapy and prevention strategies suit each person. However, predicting the sensitivity of tumors to anticancer treatments remains a challenge. In this work, we propose two deep neural network models to predict the impact of anticancer drugs in tumors through the half-maximal inhibitory concentration (IC50). These models join biological and chemical data to apprehend relevant features of the genetic profile and the drug compounds, respectively. In order to predict the drug response in cancer cell lines, this study employed different DL methods, resorting to Recurrent Neural Networks (RNNs) and Convolutional Neural Networks (CNNs). In the first stage, two autoencoders were pre-trained with high-dimensional gene expression and mutation data of tumors. Afterward, this genetic background is transferred to the prediction models that return the IC50 value that portrays the potency of a substance in inhibiting a cancer cell line. When comparing RSEM Expected counts and TPM as methods for displaying gene expression data, RSEM has been shown to perform better in deep models and CNNs model can obtain better insight in these types of data. Moreover, the obtained results reflect the effectiveness of the extracted deep representations in the prediction of the IC50 value that portrays the potency of a substance in inhibiting a tumor, achieving a performance of a mean squared error of 1.06 and surpassing previous state-of-the-art models.
Collapse
Affiliation(s)
- Maryam Abbasi
- Centre for Informatics and Systems of the University of Coimbra, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal; Polytechnic Institute of Coimbra, Applied Research Institute, Coimbra, Portugal; Research Centre for Natural Resources Environment and Society (CERNAS), Polytechnic Institute of Coimbra, Coimbra, Portugal.
| | - Filipa G Carvalho
- Centre for Informatics and Systems of the University of Coimbra, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal
| | - Bernardete Ribeiro
- Centre for Informatics and Systems of the University of Coimbra, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal
| | - Joel P Arrais
- Centre for Informatics and Systems of the University of Coimbra, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
22
|
Nguyen T, Campbell A, Kumar A, Amponsah E, Fiterau M, Shahriyari L. Optimal fusion of genotype and drug embeddings in predicting cancer drug response. Brief Bioinform 2024; 25:bbae227. [PMID: 38754407 PMCID: PMC11097979 DOI: 10.1093/bib/bbae227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 04/14/2024] [Accepted: 04/25/2024] [Indexed: 05/18/2024] Open
Abstract
Predicting cancer drug response using both genomics and drug features has shown some success compared to using genomics features alone. However, there has been limited research done on how best to combine or fuse the two types of features. Using a visible neural network with two deep learning branches for genes and drug features as the base architecture, we experimented with different fusion functions and fusion points. Our experiments show that injecting multiplicative relationships between gene and drug latent features into the original concatenation-based architecture DrugCell significantly improved the overall predictive performance and outperformed other baseline models. We also show that different fusion methods respond differently to different fusion points, indicating that the relationship between drug features and different hierarchical biological level of gene features is optimally captured using different methods. Considering both predictive performance and runtime speed, tensor product partial is the best-performing fusion function to combine late-stage representations of drug and gene features to predict cancer drug response.
Collapse
Affiliation(s)
- Trang Nguyen
- Department of Computer Science, University of Massachusetts Amherst, Amherst 01002, MA, United States
| | - Anthony Campbell
- Department of Computer Science, University of Massachusetts Amherst, Amherst 01002, MA, United States
| | - Ankit Kumar
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst 01002, MA, United States
| | - Edwin Amponsah
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst 01002, MA, United States
| | - Madalina Fiterau
- Department of Computer Science, University of Massachusetts Amherst, Amherst 01002, MA, United States
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst 01002, MA, United States
| |
Collapse
|
23
|
Lao C, Zheng P, Chen H, Liu Q, An F, Li Z. DeepAEG: a model for predicting cancer drug response based on data enhancement and edge-collaborative update strategies. BMC Bioinformatics 2024; 25:105. [PMID: 38461284 PMCID: PMC10925015 DOI: 10.1186/s12859-024-05723-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Accepted: 02/27/2024] [Indexed: 03/11/2024] Open
Abstract
MOTIVATION The prediction of cancer drug response is a challenging subject in modern personalized cancer therapy due to the uncertainty of drug efficacy and the heterogeneity of patients. It has been shown that the characteristics of the drug itself and the genomic characteristics of the patient can greatly influence the results of cancer drug response. Therefore, accurate, efficient, and comprehensive methods for drug feature extraction and genomics integration are crucial to improve the prediction accuracy. RESULTS Accurate prediction of cancer drug response is vital for guiding the design of anticancer drugs. In this study, we propose an end-to-end deep learning model named DeepAEG which is based on a complete-graph update mode to predict IC50. Specifically, we integrate an edge update mechanism on the basis of a hybrid graph convolutional network to comprehensively learn the potential high-dimensional representation of topological structures in drugs, including atomic characteristics and chemical bond information. Additionally, we present a novel approach for enhancing simplified molecular input line entry specification data by employing sequence recombination to eliminate the defect of single sequence representation of drug molecules. Our extensive experiments show that DeepAEG outperforms other existing methods across multiple evaluation parameters in multiple test sets. Furthermore, we identify several potential anticancer agents, including bortezomib, which has proven to be an effective clinical treatment option. Our results highlight the potential value of DeepAEG in guiding the design of specific cancer treatment regimens.
Collapse
Affiliation(s)
- Chuanqi Lao
- Research Center for Graph Computing, Zhejiang Lab, Yuhang, Hangzhou, 311121, Zhejiang, China
| | - Pengfei Zheng
- Research Center for Graph Computing, Zhejiang Lab, Yuhang, Hangzhou, 311121, Zhejiang, China
| | - Hongyang Chen
- Research Center for Graph Computing, Zhejiang Lab, Yuhang, Hangzhou, 311121, Zhejiang, China.
| | - Qiao Liu
- Department of Statistics, Stanford University, Stanford, Palo Alto, CA, 94305, USA
| | - Feng An
- Research Center for Graph Computing, Zhejiang Lab, Yuhang, Hangzhou, 311121, Zhejiang, China
| | - Zhao Li
- Research Center for Graph Computing, Zhejiang Lab, Yuhang, Hangzhou, 311121, Zhejiang, China
| |
Collapse
|
24
|
Lin CX, Guan Y, Li HD. Artificial intelligence approaches for molecular representation in drug response prediction. Curr Opin Struct Biol 2024; 84:102747. [PMID: 38091924 DOI: 10.1016/j.sbi.2023.102747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/26/2023] [Accepted: 11/26/2023] [Indexed: 02/09/2024]
Abstract
Drug response prediction is essential for drug development and disease treatment. One key question in predicting drug response is the representation of molecules, which has been greatly advanced by artificial intelligence (AI) techniques in recent years. In this review, we first describe different types of representation methods, pinpointing their key principles and discussing their limitations. Thereafter we discuss potential ways how these methods could be further developed. We expect that this review will provide useful guidance for researchers in the community.
Collapse
Affiliation(s)
- Cui-Xiang Lin
- School of Mathematics and Computational Science, Xiangtan University, Xiangtan, 411105, Hunan Province, PR China
| | - Yuanfang Guan
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Hong-Dong Li
- School of Computer Science and Engineering, Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, Hunan 410083, PR China.
| |
Collapse
|
25
|
Jiang L, Qu S, Yu Z, Wang J, Liu X. MOASL: Predicting drug mechanism of actions through similarity learning with transcriptomic signature. Comput Biol Med 2024; 169:107853. [PMID: 38104518 DOI: 10.1016/j.compbiomed.2023.107853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 11/02/2023] [Accepted: 12/11/2023] [Indexed: 12/19/2023]
Abstract
Understanding the mechanisms of actions (MOAs) of compounds is crucial in drug discovery. A common step in drug MOAs annotation is to query the dysregulated gene signatures induced by drugs in a reference library of pre-defined signatures. However, traditional similarity-based computational strategies face challenges when dealing with high-dimensional and noisy transcriptional signature data. To address this issue, we introduce MOASL (MOAs prediction via Similarity Learning), a novel approach that contrastive to learn similarity embeddings among signatures with shared MOAs automatically. We evaluated the accuracy of signature matching on various transcriptional activity score (TAS) datasets and individual cell lines by using MOASL. The results show MOASL achieved higher performance over several statistical and machine learning methods. Furthermore, we provided the rationale of our model by visualizing the signature annotation procedure. Using MOASL, the MOAs label of query signature could be conveniently defined by calculating the similarity between the query embedding and the reference embeddings. Finally, we applied MOASL to repurpose thousands of compounds as glucocorticoid receptor (GR) agonists, accurately identifying 8 out of the top 10 compounds. MOASL is conveniently accessible on GitHub at https://github.com/jianglikun/MOASL, empowering researchers and practitioners in the field of drug discovery to predict the MOAs of drug.
Collapse
Affiliation(s)
- Likun Jiang
- Department of Computer Science, Xiamen University, Xiamen 361005, PR China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361005, PR China
| | - Susu Qu
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, PR China; Chinese Institute for Brain Research, Beijing 102206, PR China
| | - Zhengqiu Yu
- National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361005, PR China; School of Medicine, Xiamen University, Xiamen 361005, PR China
| | - Jianmin Wang
- The Interdisciplinary Graduate Program in Integrative Biotechnology and Translational Medicine, Yonsei University, Incheon 21983, South Korea
| | - Xiangrong Liu
- Department of Computer Science, Xiamen University, Xiamen 361005, PR China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen 361005, PR China.
| |
Collapse
|
26
|
Vasanthakumari P, Zhu Y, Brettin T, Partin A, Shukla M, Xia F, Narykov O, Weil MR, Stevens RL. A Comprehensive Investigation of Active Learning Strategies for Conducting Anti-Cancer Drug Screening. Cancers (Basel) 2024; 16:530. [PMID: 38339281 PMCID: PMC10854925 DOI: 10.3390/cancers16030530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/12/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
It is well-known that cancers of the same histology type can respond differently to a treatment. Thus, computational drug response prediction is of paramount importance for both preclinical drug screening studies and clinical treatment design. To build drug response prediction models, treatment response data need to be generated through screening experiments and used as input to train the prediction models. In this study, we investigate various active learning strategies of selecting experiments to generate response data for the purposes of (1) improving the performance of drug response prediction models built on the data and (2) identifying effective treatments. Here, we focus on constructing drug-specific response prediction models for cancer cell lines. Various approaches have been designed and applied to select cell lines for screening, including a random, greedy, uncertainty, diversity, combination of greedy and uncertainty, sampling-based hybrid, and iteration-based hybrid approach. All of these approaches are evaluated and compared using two criteria: (1) the number of identified hits that are selected experiments validated to be responsive, and (2) the performance of the response prediction model trained on the data of selected experiments. The analysis was conducted for 57 drugs and the results show a significant improvement on identifying hits using active learning approaches compared with the random and greedy sampling method. Active learning approaches also show an improvement on response prediction performance for some of the drugs and analysis runs compared with the greedy sampling method.
Collapse
Affiliation(s)
- Priyanka Vasanthakumari
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (A.P.); (M.S.); (F.X.); (O.N.)
| | - Yitan Zhu
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (A.P.); (M.S.); (F.X.); (O.N.)
| | - Thomas Brettin
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (T.B.); (R.L.S.)
| | - Alexander Partin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (A.P.); (M.S.); (F.X.); (O.N.)
| | - Maulik Shukla
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (A.P.); (M.S.); (F.X.); (O.N.)
| | - Fangfang Xia
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (A.P.); (M.S.); (F.X.); (O.N.)
| | - Oleksandr Narykov
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (A.P.); (M.S.); (F.X.); (O.N.)
| | - Michael Ryan Weil
- Cancer Research Technology Program, Cancer Data Science Initiatives, Frederick National Laboratory for Cancer Research, Frederick, MD 21701, USA;
| | - Rick L. Stevens
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (T.B.); (R.L.S.)
- Department of Computer Science, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
27
|
Guo Y, Hu H, Chen W, Yin H, Wu J, Hsieh CY, He Q, Cao J. SynergyX: a multi-modality mutual attention network for interpretable drug synergy prediction. Brief Bioinform 2024; 25:bbae015. [PMID: 38340091 PMCID: PMC10858681 DOI: 10.1093/bib/bbae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 12/18/2023] [Indexed: 02/12/2024] Open
Abstract
Discovering effective anti-tumor drug combinations is crucial for advancing cancer therapy. Taking full account of intricate biological interactions is highly important in accurately predicting drug synergy. However, the extremely limited prior knowledge poses great challenges in developing current computational methods. To address this, we introduce SynergyX, a multi-modality mutual attention network to improve anti-tumor drug synergy prediction. It dynamically captures cross-modal interactions, allowing for the modeling of complex biological networks and drug interactions. A convolution-augmented attention structure is adopted to integrate multi-omic data in this framework effectively. Compared with other state-of-the-art models, SynergyX demonstrates superior predictive accuracy in both the General Test and Blind Test and cross-dataset validation. By exhaustively screening combinations of approved drugs, SynergyX reveals its ability to identify promising drug combination candidates for potential lung cancer treatment. Another notable advantage lies in its multidimensional interpretability. Taking Sorafenib and Vorinostat as an example, SynergyX serves as a powerful tool for uncovering drug-gene interactions and deciphering cell selectivity mechanisms. In summary, SynergyX provides an illuminating and interpretable framework, poised to catalyze the expedition of drug synergy discovery and deepen our comprehension of rational combination therapy.
Collapse
Affiliation(s)
- Yue Guo
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
| | - Haitao Hu
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Polytechnic Institute, Zhejiang University, 269 Shixiang Road,310000, Hangzhou, Zhejiang, China
| | - Wenbo Chen
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Polytechnic Institute, Zhejiang University, 269 Shixiang Road,310000, Hangzhou, Zhejiang, China
| | - Hao Yin
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Polytechnic Institute, Zhejiang University, 269 Shixiang Road,310000, Hangzhou, Zhejiang, China
| | - Jian Wu
- Second Affiliated Hospital School of Medicine, School of Public Health, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
| | - Chang-Yu Hsieh
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, 291 Fucheng Road, 310018, Hangzhou, Zhejiang, China
- College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
| | - Qiaojun He
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, 310020, Hangzhou, Zhejiang, China
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, 291 Fucheng Road, 310018, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
| | - Ji Cao
- Institute of Pharmacology and Toxicology, Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Engineering Research Center of Innovative Anticancer Drugs, Ministry of Education, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
- Center for Medical Research and Innovation in Digestive System Tumors, Ministry of Education, 310020, Hangzhou, Zhejiang, China
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, 291 Fucheng Road, 310018, Hangzhou, Zhejiang, China
- Cancer Center, Zhejiang University, 866 Yuhangtang Road, 310058, Hangzhou, Zhejiang, China
| |
Collapse
|
28
|
Mondello A, Dal Bo M, Toffoli G, Polano M. Machine learning in onco-pharmacogenomics: a path to precision medicine with many challenges. Front Pharmacol 2024; 14:1260276. [PMID: 38264526 PMCID: PMC10803549 DOI: 10.3389/fphar.2023.1260276] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
Over the past two decades, Next-Generation Sequencing (NGS) has revolutionized the approach to cancer research. Applications of NGS include the identification of tumor specific alterations that can influence tumor pathobiology and also impact diagnosis, prognosis and therapeutic options. Pharmacogenomics (PGx) studies the role of inheritance of individual genetic patterns in drug response and has taken advantage of NGS technology as it provides access to high-throughput data that can, however, be difficult to manage. Machine learning (ML) has recently been used in the life sciences to discover hidden patterns from complex NGS data and to solve various PGx problems. In this review, we provide a comprehensive overview of the NGS approaches that can be employed and the different PGx studies implicating the use of NGS data. We also provide an excursus of the ML algorithms that can exert a role as fundamental strategies in the PGx field to improve personalized medicine in cancer.
Collapse
Affiliation(s)
| | | | | | - Maurizio Polano
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Aviano, Italy
| |
Collapse
|
29
|
Narykov O, Zhu Y, Brettin T, Evrard YA, Partin A, Shukla M, Xia F, Clyde A, Vasanthakumari P, Doroshow JH, Stevens RL. Integration of Computational Docking into Anti-Cancer Drug Response Prediction Models. Cancers (Basel) 2023; 16:50. [PMID: 38201477 PMCID: PMC10777918 DOI: 10.3390/cancers16010050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/01/2023] [Accepted: 12/07/2023] [Indexed: 01/12/2024] Open
Abstract
Cancer is a heterogeneous disease in that tumors of the same histology type can respond differently to a treatment. Anti-cancer drug response prediction is of paramount importance for both drug development and patient treatment design. Although various computational methods and data have been used to develop drug response prediction models, it remains a challenging problem due to the complexities of cancer mechanisms and cancer-drug interactions. To better characterize the interaction between cancer and drugs, we investigate the feasibility of integrating computationally derived features of molecular mechanisms of action into prediction models. Specifically, we add docking scores of drug molecules and target proteins in combination with cancer gene expressions and molecular drug descriptors for building response models. The results demonstrate a marginal improvement in drug response prediction performance when adding docking scores as additional features, through tests on large drug screening data. We discuss the limitations of the current approach and provide the research community with a baseline dataset of the large-scale computational docking for anti-cancer drugs.
Collapse
Affiliation(s)
- Oleksandr Narykov
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Yitan Zhu
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Thomas Brettin
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Yvonne A. Evrard
- Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA;
| | - Alexander Partin
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Maulik Shukla
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Fangfang Xia
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - Austin Clyde
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
- Department of Computer Science, The University of Chicago, Chicago, IL 60637, USA
| | - Priyanka Vasanthakumari
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
| | - James H. Doroshow
- Developmental Therapeutics Branch, National Cancer Institute, Bethesda, MD 20892, USA;
| | - Rick L. Stevens
- Computing, Environment and Life Sciences, Argonne National Laboratory, Lemont, IL 60439, USA; (Y.Z.); (T.B.); (A.P.); (M.S.); (F.X.); (P.V.); (R.L.S.)
- Department of Computer Science, The University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
30
|
Zhang Y, Liu C, Liu M, Liu T, Lin H, Huang CB, Ning L. Attention is all you need: utilizing attention in AI-enabled drug discovery. Brief Bioinform 2023; 25:bbad467. [PMID: 38189543 PMCID: PMC10772984 DOI: 10.1093/bib/bbad467] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Revised: 11/03/2023] [Accepted: 11/25/2023] [Indexed: 01/09/2024] Open
Abstract
Recently, attention mechanism and derived models have gained significant traction in drug development due to their outstanding performance and interpretability in handling complex data structures. This review offers an in-depth exploration of the principles underlying attention-based models and their advantages in drug discovery. We further elaborate on their applications in various aspects of drug development, from molecular screening and target binding to property prediction and molecule generation. Finally, we discuss the current challenges faced in the application of attention mechanisms and Artificial Intelligence technologies, including data quality, model interpretability and computational resource constraints, along with future directions for research. Given the accelerating pace of technological advancement, we believe that attention-based models will have an increasingly prominent role in future drug discovery. We anticipate that these models will usher in revolutionary breakthroughs in the pharmaceutical domain, significantly accelerating the pace of drug development.
Collapse
Affiliation(s)
- Yang Zhang
- Innovative Institute of Chinese Medicine and Pharmacy, Academy for Interdiscipline, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Caiqi Liu
- Department of Gastrointestinal Medical Oncology, Harbin Medical University Cancer Hospital, No.150 Haping Road, Nangang District, Harbin, Heilongjiang 150081, China
- Key Laboratory of Molecular Oncology of Heilongjiang Province, No.150 Haping Road, Nangang District, Harbin, Heilongjiang 150081, China
| | - Mujiexin Liu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tianyuan Liu
- Graduate School of Science and Technology, University of Tsukuba, Tsukuba, Japan
| | - Hao Lin
- School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Cheng-Bing Huang
- School of Computer Science and Technology, Aba Teachers University, Aba, China
| | - Lin Ning
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou, Zhejiang, China
- School of Healthcare Technology, Chengdu Neusoft University, Chengdu 611844, China
| |
Collapse
|
31
|
Zhao H, Ni P, Zhao Q, Liang X, Ai D, Erhardt S, Wang J, Li Y, Wang J. Identifying the serious clinical outcomes of adverse reactions to drugs by a multi-task deep learning framework. Commun Biol 2023; 6:870. [PMID: 37620651 PMCID: PMC10449791 DOI: 10.1038/s42003-023-05243-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 08/14/2023] [Indexed: 08/26/2023] Open
Abstract
Adverse Drug Reactions (ADRs) have a direct impact on human health. As continuous pharmacovigilance and drug monitoring prove to be costly and time-consuming, computational methods have emerged as promising alternatives. However, most existing computational methods primarily focus on predicting whether or not the drug is associated with an adverse reaction and do not consider the core issue of drug benefit-risk assessment-whether the treatment outcome is serious when adverse drug reactions occur. To this end, we categorize serious clinical outcomes caused by adverse reactions to drugs into seven distinct classes and present a deep learning framework, so-called GCAP, for predicting the seriousness of clinical outcomes of adverse reactions to drugs. GCAP has two tasks: one is to predict whether adverse reactions to drugs cause serious clinical outcomes, and the other is to infer the corresponding classes of serious clinical outcomes. Experimental results demonstrate that our method is a powerful and robust framework with high extendibility. GCAP can serve as a useful tool to successfully address the challenge of predicting the seriousness of clinical outcomes stemming from adverse reactions to drugs.
Collapse
Affiliation(s)
- Haochen Zhao
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
- Xiangjiang Laboratory, Changsha, 410205, China
| | - Peng Ni
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
| | - Qichang Zhao
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
| | - Xiao Liang
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China
| | - Di Ai
- Department of Pathology and Laboratory Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Shannon Erhardt
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jun Wang
- Department of Pediatrics, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Yaohang Li
- Department of Computer Science, Old Dominion University, Norfolk, VA, 23529-0001, USA
| | - Jianxin Wang
- School of Computer Science and Engineering, Central South University, Changsha, 410083, China.
- Hunan Provincial Key Lab on Bioinformatics, Central South University, Changsha, 410083, China.
- Xiangjiang Laboratory, Changsha, 410205, China.
| |
Collapse
|
32
|
Xu X, Qi Z, Han X, Xu A, Geng Z, He X, Ren Y, Duo Z. Predicting anticancer drug sensitivity on distributed data sources using federated deep learning. Heliyon 2023; 9:e18615. [PMID: 37593639 PMCID: PMC10427996 DOI: 10.1016/j.heliyon.2023.e18615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/12/2023] [Accepted: 07/24/2023] [Indexed: 08/19/2023] Open
Abstract
Drug sensitivity prediction plays a crucial role in precision cancer therapy. Collaboration among medical institutions can lead to better performance in drug sensitivity prediction. However, patient privacy and data protection regulation remain a severe impediment to centralized prediction studies. For the first time, we proposed a federated drug sensitivity prediction model with high generalization, combining distributed data sources while protecting private data. Cell lines are first classified into three categories using the waterfall method. Focal loss for solving class imbalance is then embedded into the horizontal federated deep learning framework, i.e., HFDL-fl is presented. Applying HFDL-fl to homogeneous and heterogeneous data, we obtained HFDL-Cross and HFDL-Within. Our comprehensive experiments demonstrated that (i) collaboration by HFDL-fl outperforms private model on local data, (ii) focal loss function can effectively improve model performance to classify cell lines in sensitive and resistant categories, and (iii) HFDL-fl is not significantly affected by data heterogeneity. To summarize, HFDL-fl provides a valuable solution to break down the barriers between medical institutions for privacy-preserving drug sensitivity prediction and therefore facilitates the development of cancer precision medicine and other privacy-related biomedical research.
Collapse
Affiliation(s)
- Xiaolu Xu
- School of Computer and Artificial Intelligence, Liaoning Normal University, Dalian 116029, China
| | - Zitong Qi
- Department of Statistics, University of Washington, Seattle, WA 98195, USA
| | - Xiumei Han
- College of Artificial Intelligence, Dalian Maritime University, Dalian 116026, China
| | - Aiguo Xu
- Department of Oncology, The Second People's Hospital of Lianyungang, Lianyungang 222023, China
| | - Zhaohong Geng
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian 116023, China
| | - Xinyu He
- School of Computer and Artificial Intelligence, Liaoning Normal University, Dalian 116029, China
| | - Yonggong Ren
- School of Computer and Artificial Intelligence, Liaoning Normal University, Dalian 116029, China
| | - Zhaojun Duo
- School of Computer and Artificial Intelligence, Liaoning Normal University, Dalian 116029, China
| |
Collapse
|
33
|
Zhan Y, Guo J, Philip Chen CL, Meng XB. iBT-Net: an incremental broad transformer network for cancer drug response prediction. Brief Bioinform 2023:bbad256. [PMID: 37429577 DOI: 10.1093/bib/bbad256] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/30/2023] [Accepted: 06/15/2023] [Indexed: 07/12/2023] Open
Abstract
In modern precision medicine, it is an important research topic to predict cancer drug response. Due to incomplete chemical structures and complex gene features, however, it is an ongoing work to design efficient data-driven methods for predicting drug response. Moreover, since the clinical data cannot be easily obtained all at once, the data-driven methods may require relearning when new data are available, resulting in increased time consumption and cost. To address these issues, an incremental broad Transformer network (iBT-Net) is proposed for cancer drug response prediction. Different from the gene expression features learning from cancer cell lines, structural features are further extracted from drugs by Transformer. Broad learning system is then designed to integrate the learned gene features and structural features of drugs to predict the response. With the capability of incremental learning, the proposed method can further use new data to improve its prediction performance without retraining totally. Experiments and comparison studies demonstrate the effectiveness and superiority of iBT-Net under different experimental configurations and continuous data learning.
Collapse
Affiliation(s)
- Yongkang Zhan
- School of Computer Science & Engineering,South China University of Technology, 510006, China
| | - Jifeng Guo
- School of Computer Science & Engineering,South China University of Technology, 510006, China
| | - C L Philip Chen
- School of Computer Science & Engineering,South China University of Technology, 510006, China
- Brain and Affective Cognitive Research Center, Pazhou Lab, 510335, China
| | - Xian-Bing Meng
- School of Electromechanical Engineering, Guangdong University of Technology, 510006, China
| |
Collapse
|
34
|
Huang Z, Zhang P, Deng L. DeepCoVDR: deep transfer learning with graph transformer and cross-attention for predicting COVID-19 drug response. Bioinformatics 2023; 39:i475-i483. [PMID: 37387168 DOI: 10.1093/bioinformatics/btad244] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/01/2023] Open
Abstract
MOTIVATION The coronavirus disease 2019 (COVID-19) remains a global public health emergency. Although people, especially those with underlying health conditions, could benefit from several approved COVID-19 therapeutics, the development of effective antiviral COVID-19 drugs is still a very urgent problem. Accurate and robust drug response prediction to a new chemical compound is critical for discovering safe and effective COVID-19 therapeutics. RESULTS In this study, we propose DeepCoVDR, a novel COVID-19 drug response prediction method based on deep transfer learning with graph transformer and cross-attention. First, we adopt a graph transformer and feed-forward neural network to mine the drug and cell line information. Then, we use a cross-attention module that calculates the interaction between the drug and cell line. After that, DeepCoVDR combines drug and cell line representation and their interaction features to predict drug response. To solve the problem of SARS-CoV-2 data scarcity, we apply transfer learning and use the SARS-CoV-2 dataset to fine-tune the model pretrained on the cancer dataset. The experiments of regression and classification show that DeepCoVDR outperforms baseline methods. We also evaluate DeepCoVDR on the cancer dataset, and the results indicate that our approach has high performance compared with other state-of-the-art methods. Moreover, we use DeepCoVDR to predict COVID-19 drugs from FDA-approved drugs and demonstrate the effectiveness of DeepCoVDR in identifying novel COVID-19 drugs. AVAILABILITY AND IMPLEMENTATION https://github.com/Hhhzj-7/DeepCoVDR.
Collapse
Affiliation(s)
- Zhijian Huang
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Pan Zhang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha 410083, China
| | - Lei Deng
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
35
|
Zhang Z, Wei X. Artificial intelligence-assisted selection and efficacy prediction of antineoplastic strategies for precision cancer therapy. Semin Cancer Biol 2023; 90:57-72. [PMID: 36796530 DOI: 10.1016/j.semcancer.2023.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/12/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
The rapid development of artificial intelligence (AI) technologies in the context of the vast amount of collectable data obtained from high-throughput sequencing has led to an unprecedented understanding of cancer and accelerated the advent of a new era of clinical oncology with a tone of precision treatment and personalized medicine. However, the gains achieved by a variety of AI models in clinical oncology practice are far from what one would expect, and in particular, there are still many uncertainties in the selection of clinical treatment options that pose significant challenges to the application of AI in clinical oncology. In this review, we summarize emerging approaches, relevant datasets and open-source software of AI and show how to integrate them to address problems from clinical oncology and cancer research. We focus on the principles and procedures for identifying different antitumor strategies with the assistance of AI, including targeted cancer therapy, conventional cancer therapy, and cancer immunotherapy. In addition, we also highlight the current challenges and directions of AI in clinical oncology translation. Overall, we hope this article will provide researchers and clinicians with a deeper understanding of the role and implications of AI in precision cancer therapy, and help AI move more quickly into accepted cancer guidelines.
Collapse
Affiliation(s)
- Zhe Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
36
|
Partin A, Brettin T, Zhu Y, Dolezal JM, Kochanny S, Pearson AT, Shukla M, Evrard YA, Doroshow JH, Stevens RL. Data augmentation and multimodal learning for predicting drug response in patient-derived xenografts from gene expressions and histology images. Front Med (Lausanne) 2023; 10:1058919. [PMID: 36960342 PMCID: PMC10027779 DOI: 10.3389/fmed.2023.1058919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 02/10/2023] [Indexed: 03/09/2023] Open
Abstract
Patient-derived xenografts (PDXs) are an appealing platform for preclinical drug studies. A primary challenge in modeling drug response prediction (DRP) with PDXs and neural networks (NNs) is the limited number of drug response samples. We investigate multimodal neural network (MM-Net) and data augmentation for DRP in PDXs. The MM-Net learns to predict response using drug descriptors, gene expressions (GE), and histology whole-slide images (WSIs). We explore whether combining WSIs with GE improves predictions as compared with models that use GE alone. We propose two data augmentation methods which allow us training multimodal and unimodal NNs without changing architectures with a single larger dataset: 1) combine single-drug and drug-pair treatments by homogenizing drug representations, and 2) augment drug-pairs which doubles the sample size of all drug-pair samples. Unimodal NNs which use GE are compared to assess the contribution of data augmentation. The NN that uses the original and the augmented drug-pair treatments as well as single-drug treatments outperforms NNs that ignore either the augmented drug-pairs or the single-drug treatments. In assessing the multimodal learning based on the MCC metric, MM-Net outperforms all the baselines. Our results show that data augmentation and integration of histology images with GE can improve prediction performance of drug response in PDXs.
Collapse
Affiliation(s)
- Alexander Partin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Thomas Brettin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Yitan Zhu
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - James M. Dolezal
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, United States
| | - Sara Kochanny
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, United States
| | - Alexander T. Pearson
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medical Center, Chicago, IL, United States
| | - Maulik Shukla
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Yvonne A. Evrard
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD, United States
| | - James H. Doroshow
- Division of Cancer Therapeutics and Diagnosis, National Cancer Institute, Bethesda, MD, United States
| | - Rick L. Stevens
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
- Department of Computer Science, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
37
|
Yang X, Niu Z, Liu Y, Song B, Lu W, Zeng L, Zeng X. Modality-DTA: Multimodality Fusion Strategy for Drug-Target Affinity Prediction. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:1200-1210. [PMID: 36083952 DOI: 10.1109/tcbb.2022.3205282] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Prediction of the drug-target affinity (DTA) plays an important role in drug discovery. Existing deep learning methods for DTA prediction typically leverage a single modality, namely simplified molecular input line entry specification (SMILES) or amino acid sequence to learn representations. SMILES or amino acid sequences can be encoded into different modalities. Multimodality data provide different kinds of information, with complementary roles for DTA prediction. We propose Modality-DTA, a novel deep learning method for DTA prediction that leverages the multimodality of drugs and targets. A group of backward propagation neural networks is applied to ensure the completeness of the reconstruction process from the latent feature representation to original multimodality data. The tag between the drug and target is used to reduce the noise information in the latent representation from multimodality data. Experiments on three benchmark datasets show that our Modality-DTA outperforms existing methods in all metrics. Modality-DTA reduces the mean square error by 15.7% and improves the area under the precisionrecall curve by 12.74% in the Davis dataset. We further find that the drug modality Morgan fingerprint and the target modality generated by one-hot-encoding play the most significant roles. To the best of our knowledge, Modality-DTA is the first method to explore multimodality for DTA prediction.
Collapse
|
38
|
Moon S, Kim HJ, Lee Y, Lee YJ, Jung S, Lee JS, Hahn SH, Kim K, Roh JY, Nam S. Oncogenic signaling pathways and hallmarks of cancer in Korean patients with acral melanoma. Comput Biol Med 2023; 154:106602. [PMID: 36716688 DOI: 10.1016/j.compbiomed.2023.106602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 12/23/2022] [Accepted: 01/22/2023] [Indexed: 01/25/2023]
Abstract
Acral melanoma (AM), a rare subtype of cutaneous melanoma, shows higher incidence in Asians, including Koreans, than in Caucasians. However, the genetic modification associated with AM in Koreans is not well known and has not been comprehensively investigated in terms of oncogenic signaling, and hallmarks of cancer. We performed whole-exome and RNA sequencing for Korean patients with AM and acquired the genetic alterations and gene expression profiles. KIT alterations (previously known to be recurrent alterations in AM) and CDK4/CCND1 copy number amplifications were identified in the patients. Genetic and transcriptomic alterations in patients with AM were functionally converge to the hallmarks of cancer and oncogenic pathways, including 'proliferative signal persistence', 'apoptotic resistance', and 'activation of invasion and metastasis', despite the heterogeneous somatic mutation profiles of Korean patients with AM. This study may provide a molecular understanding for therapeutic strategy for AM.
Collapse
Affiliation(s)
- SeongRyeol Moon
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, 21999, South Korea
| | - Hee Joo Kim
- Department of Dermatology, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, South Korea
| | - Yeeun Lee
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, 21999, South Korea
| | - Yu Joo Lee
- Department of Genome Medicine and Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, South Korea
| | - Sungwon Jung
- Department of Genome Medicine and Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, South Korea
| | - Jin Sook Lee
- Department of Genome Medicine and Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, South Korea; Department of Pediatrics, Seoul National University Hospital Child Cancer and Rare Disease Administration, Seoul National University Children's Hospital, Seoul, 03080, South Korea
| | - Si Houn Hahn
- Division of Genetic Medicine, Department of Pediatrics, University of Washington School of Medicine, Seattle Children's Hospital, Seattle, WA, 98105, USA
| | | | - Joo Young Roh
- Department of Dermatology, Ewha Womans University College of Medicine, Seoul Hospital, Seoul, 07804, South Korea.
| | - Seungyoon Nam
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, 21999, South Korea; Department of Genome Medicine and Science, Gachon Institute of Genome Medicine and Science, Gachon University Gil Medical Center, Gachon University College of Medicine, Incheon, 21565, South Korea; AI Convergence Center for Medical Science, Gachon University College of Medicine, Incheon, 21565, South Korea.
| |
Collapse
|
39
|
Partin A, Brettin TS, Zhu Y, Narykov O, Clyde A, Overbeek J, Stevens RL. Deep learning methods for drug response prediction in cancer: Predominant and emerging trends. Front Med (Lausanne) 2023; 10:1086097. [PMID: 36873878 PMCID: PMC9975164 DOI: 10.3389/fmed.2023.1086097] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Cancer claims millions of lives yearly worldwide. While many therapies have been made available in recent years, by in large cancer remains unsolved. Exploiting computational predictive models to study and treat cancer holds great promise in improving drug development and personalized design of treatment plans, ultimately suppressing tumors, alleviating suffering, and prolonging lives of patients. A wave of recent papers demonstrates promising results in predicting cancer response to drug treatments while utilizing deep learning methods. These papers investigate diverse data representations, neural network architectures, learning methodologies, and evaluations schemes. However, deciphering promising predominant and emerging trends is difficult due to the variety of explored methods and lack of standardized framework for comparing drug response prediction models. To obtain a comprehensive landscape of deep learning methods, we conducted an extensive search and analysis of deep learning models that predict the response to single drug treatments. A total of 61 deep learning-based models have been curated, and summary plots were generated. Based on the analysis, observable patterns and prevalence of methods have been revealed. This review allows to better understand the current state of the field and identify major challenges and promising solution paths.
Collapse
Affiliation(s)
- Alexander Partin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Thomas S. Brettin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Yitan Zhu
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Oleksandr Narykov
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Austin Clyde
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Jamie Overbeek
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
| | - Rick L. Stevens
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, United States
- Department of Computer Science, The University of Chicago, Chicago, IL, United States
| |
Collapse
|
40
|
Wang H, Dai C, Wen Y, Wang X, Liu W, He S, Bo X, Peng S. GADRP: graph convolutional networks and autoencoders for cancer drug response prediction. Brief Bioinform 2023; 24:6865039. [PMID: 36460622 DOI: 10.1093/bib/bbac501] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/19/2022] [Accepted: 10/22/2022] [Indexed: 12/04/2022] Open
Abstract
Drug response prediction in cancer cell lines is of great significance in personalized medicine. In this study, we propose GADRP, a cancer drug response prediction model based on graph convolutional networks (GCNs) and autoencoders (AEs). We first use a stacked deep AE to extract low-dimensional representations from cell line features, and then construct a sparse drug cell line pair (DCP) network incorporating drug, cell line, and DCP similarity information. Later, initial residual and layer attention-based GCN (ILGCN) that can alleviate over-smoothing problem is utilized to learn DCP features. And finally, fully connected network is employed to make prediction. Benchmarking results demonstrate that GADRP can significantly improve prediction performance on all metrics compared with baselines on five datasets. Particularly, experiments of predictions of unknown DCP responses, drug-cancer tissue associations, and drug-pathway associations illustrate the predictive power of GADRP. All results highlight the effectiveness of GADRP in predicting drug responses, and its potential value in guiding anti-cancer drug selection.
Collapse
Affiliation(s)
- Hong Wang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Chong Dai
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.,Department of Bioinformatics, Beijing Institute of Health Service and Transfusion Medicine, Beijing 100850, China
| | - Yuqi Wen
- Department of Bioinformatics, Beijing Institute of Health Service and Transfusion Medicine, Beijing 100850, China
| | - Xiaoqi Wang
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Wenjuan Liu
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China
| | - Song He
- Department of Bioinformatics, Beijing Institute of Health Service and Transfusion Medicine, Beijing 100850, China
| | - Xiaochen Bo
- Department of Bioinformatics, Beijing Institute of Health Service and Transfusion Medicine, Beijing 100850, China
| | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha 410082, China.,The State Key Laboratory of Chemo/Biosensing and Chemometrics, Hunan University, Changsha 410082, China
| |
Collapse
|
41
|
Chen J, Hao L, Qian X, Lin L, Pan Y, Han X. Machine learning models based on immunological genes to predict the response to neoadjuvant therapy in breast cancer patients. Front Immunol 2022; 13:948601. [PMID: 35935976 PMCID: PMC9352856 DOI: 10.3389/fimmu.2022.948601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/29/2022] [Indexed: 12/13/2022] Open
Abstract
Breast cancer (BC) is the most common malignancy worldwide and neoadjuvant therapy (NAT) plays an important role in the treatment of patients with early BC. However, only a subset of BC patients can achieve pathological complete response (pCR) and benefit from NAT. It is therefore necessary to predict the responses to NAT. Although many models to predict the response to NAT based on gene expression determined by the microarray platform have been proposed, their applications in clinical practice are limited due to the data normalization methods during model building and the disadvantages of the microarray platform compared with the RNA-seq platform. In this study, we first reconfirmed the correlation between immune profiles and pCR in an RNA-seq dataset. Then, we employed multiple machine learning algorithms and a model stacking strategy to build an immunological gene based model (Ipredictor model) and an immunological gene and receptor status based model ICpredictor model) in the RNA-seq dataset. The areas under the receiver operator characteristic curves for the Ipredictor model and ICpredictor models were 0.745 and 0.769 in an independent external test set based on the RNA-seq platform, and were 0.716 and 0.752 in another independent external test set based on the microarray platform. Furthermore, we found that the predictive score of the Ipredictor model was correlated with immune microenvironment and genomic aberration markers. These results demonstrated that the models can accurately predict the response to NAT for BC patients and will contribute to individualized therapy.
Collapse
Affiliation(s)
- Jian Chen
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Center for Cancer Bioimmunotherapy of Anhui Province, Hefei, China
| | - Li Hao
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Center for Cancer Bioimmunotherapy of Anhui Province, Hefei, China
| | - Xiaojun Qian
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Center for Cancer Bioimmunotherapy of Anhui Province, Hefei, China
| | - Lin Lin
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Center for Cancer Bioimmunotherapy of Anhui Province, Hefei, China
| | - Yueyin Pan
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Center for Cancer Bioimmunotherapy of Anhui Province, Hefei, China
| | - Xinghua Han
- Department of Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Center for Cancer Bioimmunotherapy of Anhui Province, Hefei, China
| |
Collapse
|