1
|
Abbas Z, Kim S, Lee N, Kazmi SAW, Lee SW. A robust ensemble framework for anticancer peptide classification using multi-model voting approach. Comput Biol Med 2025; 188:109750. [PMID: 40032410 DOI: 10.1016/j.compbiomed.2025.109750] [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: 10/30/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 03/05/2025]
Abstract
Anticancer peptides (ACPs) hold great potential for cancer therapeutics, yet accurately identifying them remains a challenging task due to the complexity of peptide sequences and their interactions with biological systems. In this study, we propose a novel machine learning-based framework for ACP classification, integrating multiple feature sets, including sequence composition, physicochemical properties, and embedding features derived from pre-trained language models. We evaluate the performance of various classifiers on benchmark datasets and compare our model against state-of-the-art methods. The results demonstrate that our model outperforms existing methods such as UniDL4BioPep, ACPred-Fuse, and iACP with an accuracy of 75.58%, an AUC of 0.8272, and an MCC of 0.5119. Our approach provides a more balanced sensitivity of 0.7384 and specificity of 0.773, ensuring robust identification of both ACPs and non-ACPs. These findings suggest that incorporating diverse feature sets can significantly enhance ACP classification, potentially facilitating the discovery of novel anticancer peptides for therapeutic applications.
Collapse
Affiliation(s)
- Zeeshan Abbas
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea; Department of Artificial Intelligence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Sunyeup Kim
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | - Nangkyeong Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea
| | | | - Seung Won Lee
- Department of Precision Medicine, Sungkyunkwan University School of Medicine, Suwon, Republic of Korea; Department of Artificial Intelligence, Sungkyunkwan University, Suwon 16419, Republic of Korea; Department of Metabiohealth, Sungkyunkwan University, Suwon 16419, Republic of Korea; Personalized Cancer Immunotherapy Research Center, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea.
| |
Collapse
|
2
|
Geng A, Luo Z, Li A, Zhang Z, Zou Q, Wei L, Cui F. ACP-CLB: An Anticancer Peptide Prediction Model Based on Multichannel Discriminative Processing and Integration of Large Pretrained Protein Language Models. J Chem Inf Model 2025; 65:2336-2349. [PMID: 39969847 DOI: 10.1021/acs.jcim.4c02072] [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: 02/20/2025]
Abstract
MOTIVATION Cancer affects millions globally, and as research advances, our understanding and treatment of cancer evolve. Compared to conventional treatments with significant side effects, anticancer peptides (ACPs) have gained considerable attention. Validating ACPs through wet-lab experiments is time-consuming and costly. However, numerous artificial intelligence methods are now used for ACP identification and classification. These methods typically apply a uniform strategy to all feature types, overlooking the potential benefits of more specialized processing for different feature types. INNOVATION In this paper, we propose a framework based on multichannel discriminative processing, where different neural networks are applied to process various feature types, optimizing their respective feature vectors. Additionally, we leverage Large Pretrained Protein Language Models to capture deeper sequence features, further enhancing the model's performance. Contributions: To better validate the overall performance and generalization ability of the model, we compared it with state-of-the-art models using four different data sets (AntiCp2Main, AntiCp2 Alternate, ACP740, cACP-DeepGram). The results show significant improvements across most metrics. Additionally, our proposed framework better assists researchers in distinguishing and identifying ACPs and further validates the need for distinct processing methods for different feature types.
Collapse
Affiliation(s)
- Aoyun Geng
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Zhenjie Luo
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Aohan Li
- Graduate School of Informatics and Engineering, The University of Electro-Communications, Tokyo 182-8585, Japan
| | - Zilong Zhang
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu 610054, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China
| | - Leyi Wei
- Centre for Artificial Intelligence driven Drug Discovery, Faculty of Applied Science, Macao Polytechnic University, Macao SAR 999078, China
- School of Informatics, Xiamen University, Xiamen 361000, China
| | - Feifei Cui
- School of Computer Science and Technology, Hainan University, Haikou 570228, China
| |
Collapse
|
3
|
Hou D, Zhou H, Tang Y, Liu Z, Su L, Guo J, Pathak JL, Wu L. Dynamic Visualization of Computer-Aided Peptide Design for Cancer Therapeutics. Drug Des Devel Ther 2025; 19:1043-1065. [PMID: 39974609 PMCID: PMC11837852 DOI: 10.2147/dddt.s497126] [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: 09/21/2024] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
Purpose Cancer stands as a significant global public health concern, with traditional therapies potentially yielding severe side effects. Peptide-based cancer therapy is increasingly employed for diseases like cancer due to its advantages of excellent targeting, biocompatibility, and convenient synthesis. With advancements in computer technology and bioinformatics, rational design strategies based on computer technology have been employed to develop more cost-effective and potent anticancer peptides (ACPs). This study aims to explore the current status, hotspots, and future trends in the field of computer-aided design of peptides for cancer treatment through a bibliometric analysis. Methods A total of 1547 relevant publications published from 2006 to 2024 were collected from the Web of Science Core Collection. Bibliometric analysis was conducted using tools like CiteSpace, VOSviewer, Bibliometrix, Origin, and an online bibliometric platform. Results The research in this field has shown a steady growth trend, with the United States and China making the most significant contributions. Currently, ACP research mainly focuses on cell-penetrating peptides related to drug delivery, which are expected to become future research hotspots. Beyond that, peptide vaccines associated with immunotherapy are also worthy of attention. In addition, molecular dynamics simulation and molecular docking are currently popular research methods. At the same time, deep learning is the emerging keyword, indicating its potential for a more significant impact on future peptide design. Conclusion Deep learning technology represents emerging research hotspots with immense potential and promising prospects. As cutting-edge research directions, cellularly penetrating peptides and polypeptide immunotherapy are expected to achieve breakthroughs in cancer treatment. This study provides valuable insights into the computer-aided design of peptides in cancer therapy, contributing significantly to advancing the in-depth research and applications in this area.
Collapse
Affiliation(s)
- Dan Hou
- Department of Basic Oral Medicine, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, Guangdong, 510182, People’s Republic of China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, People’s Republic of China
- Department of Oral and Maxillofacial Surgery/Oral Pathology, Amsterdam UMC/VUmc and Academic Centre for Dentistry Amsterdam (ACTA), Vrije Universiteit Amsterdam, Amsterdam Movement Science, Amsterdam, 1081 hZ, the Netherlands
| | - Haobin Zhou
- Department of Basic Oral Medicine, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, Guangdong, 510182, People’s Republic of China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, People’s Republic of China
| | - Yuting Tang
- Department of Basic Oral Medicine, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, Guangdong, 510182, People’s Republic of China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, People’s Republic of China
| | - Ziyuan Liu
- Department of Basic Oral Medicine, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, Guangdong, 510182, People’s Republic of China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, People’s Republic of China
| | - Lin Su
- Department of Basic Oral Medicine, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, Guangdong, 510182, People’s Republic of China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, People’s Republic of China
| | - Junkai Guo
- Department of Basic Oral Medicine, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, Guangdong, 510182, People’s Republic of China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, People’s Republic of China
| | - Janak Lal Pathak
- Department of Basic Oral Medicine, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, Guangdong, 510182, People’s Republic of China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, People’s Republic of China
| | - Lihong Wu
- Department of Basic Oral Medicine, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Medical University, Guangzhou, Guangdong, 510182, People’s Republic of China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou, Guangdong, 510182, People’s Republic of China
| |
Collapse
|
4
|
Shahid, Hayat M, Alghamdi W, Akbar S, Raza A, Kadir RA, Sarker MR. pACP-HybDeep: predicting anticancer peptides using binary tree growth based transformer and structural feature encoding with deep-hybrid learning. Sci Rep 2025; 15:565. [PMID: 39747941 PMCID: PMC11695694 DOI: 10.1038/s41598-024-84146-0] [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: 08/12/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
Worldwide, Cancer remains a significant health concern due to its high mortality rates. Despite numerous traditional therapies and wet-laboratory methods for treating cancer-affected cells, these approaches often face limitations, including high costs and substantial side effects. Recently the high selectivity of peptides has garnered significant attention from scientists due to their reliable targeted actions and minimal adverse effects. Furthermore, keeping the significant outcomes of the existing computational models, we propose a highly reliable and effective model namely, pACP-HybDeep for the accurate prediction of anticancer peptides. In this model, training peptides are numerically encoded using an attention-based ProtBERT-BFD encoder to extract semantic features along with CTDT-based structural information. Furthermore, a k-nearest neighbor-based binary tree growth (BTG) algorithm is employed to select an optimal feature set from the multi-perspective vector. The selected feature vector is subsequently trained using a CNN + RNN-based deep learning model. Our proposed pACP-HybDeep model demonstrated a high training accuracy of 95.33%, and an AUC of 0.97. To validate the generalization capabilities of the model, our pACP-HybDeep model achieved accuracies of 94.92%, 92.26%, and 91.16% on independent datasets Ind-S1, Ind-S2, and Ind-S3, respectively. The demonstrated efficacy, and reliability of the pACP-HybDeep model using test datasets establish it as a valuable tool for researchers in academia and pharmaceutical drug design.
Collapse
Affiliation(s)
- Shahid
- Department of Computer Science, Abdul Wali Khan University Mardan, Mardan, 23200, KP, Pakistan
| | - Maqsood Hayat
- Department of Computer Science, Abdul Wali Khan University Mardan, Mardan, 23200, KP, Pakistan.
| | - Wajdi Alghamdi
- Department of Information Technology, Faculty of Computing and Information Technology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shahid Akbar
- Department of Computer Science, Abdul Wali Khan University Mardan, Mardan, 23200, KP, Pakistan.
- Institute of Fundamental and Frontier Sciences, University of Electronic Science and Technology of China, Chengdu, 610054, China.
| | - Ali Raza
- Department of Computer Science, MY University, Islamabad, 45750, Pakistan
| | - Rabiah Abdul Kadir
- Institute of Visual Informatics, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia.
| | - Mahidur R Sarker
- Institute of Visual Informatics, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
- Universidad de Dise˜no, Innovaci´on y Tecnología, UDIT, Av. Alfonso XIII, 97, 28016, Madrid, Spain
| |
Collapse
|
5
|
Lin Z, Assaraf YG, Kwok HF. Peptides for microbe-induced cancers: latest therapeutic strategies and their advanced technologies. Cancer Metastasis Rev 2024; 43:1315-1336. [PMID: 39008152 DOI: 10.1007/s10555-024-10197-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 06/14/2024] [Indexed: 07/16/2024]
Abstract
Cancer is a significant global health concern associated with multiple distinct factors, including microbial and viral infections. Numerous studies have elucidated the role of microorganisms, such as Helicobacter pylori (H. pylori), as well as viruses for example human papillomavirus (HPV), hepatitis B virus (HBV), and hepatitis C virus (HCV), in the development of human malignancies. Substantial attention has been focused on the treatment of these microorganism- and virus-associated cancers, with promising outcomes observed in studies employing peptide-based therapies. The current paper provides an overview of microbe- and virus-induced cancers and their underlying molecular mechanisms. We discuss an assortment of peptide-based therapies which are currently being developed, including tumor-targeting peptides and microbial/viral peptide-based vaccines. We describe the major technological advancements that have been made in the design, screening, and delivery of peptides as anticancer agents. The primary focus of the current review is to provide insight into the latest research and development in this field and to provide a realistic glimpse into the future of peptide-based therapies for microbe- and virus-induced neoplasms.
Collapse
Affiliation(s)
- Ziqi Lin
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR
| | - Yehuda G Assaraf
- The Fred Wyszkowski Cancer Research Lab, Faculty of Biology, Technion-Israel Instituteof Technology, Haifa, 3200003, Israel
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida da Universidade, Taipa, Macau SAR.
- MoE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR.
| |
Collapse
|
6
|
Wang X, Zhang Z, Liu C. iACP-DFSRA: Identification of Anticancer Peptides Based on a Dual-channel Fusion Strategy of ResCNN and Attention. J Mol Biol 2024; 436:168810. [PMID: 39362624 DOI: 10.1016/j.jmb.2024.168810] [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: 07/09/2024] [Revised: 09/10/2024] [Accepted: 09/27/2024] [Indexed: 10/05/2024]
Abstract
Anticancer peptides (ACPs) have been widely applied in the treatment of cancer owing to good safety, rational side effects, and high selectivity. However, the number of ACPs that have been experimentally validated is limited as identification of ACPs is extremely expensive. Hence, accurate and cost-effective identification methods for ACPs are urgently needed. In this work, we proposed a deep learning-based model, named iACP-DFSRA, for ACPs identification. Specifically, we adopted two kinds of sequence embedding technologies, ProtBert_BFD pre-training language model and handcrafted features to encode protein sequences. Then, the LightGBM was used for feature selection, and the selected features were input into ResCNN and Attention mechanism, respectively, to extract local and global features. Finally, the concatenate features were deeply fused by using the Attention mechanism to allow key features to be paid more attention to by the model and make predictions by fully connected layer. The results of 10-fold cross-validation demonstrated that the iACP-DFSRA model delivered improved results in most metrics with Sp of 94.15%, Sn of 95.32%, Acc of 94.74% and MCC of 89.48% compared to the latest AACFlow model. Indeed, the iACP-DFSRA model is the only model with Acc > 90% and MCC > 80% on this independent test dataset. Furthermore, we have further demonstrated the superiority of our model on additional datasets. In addition, t-SNE and SHAP interpretation analysis demonstrated that it is crucial to use two channels for feature extraction and use the Attention mechanism for deep fusion, which helps the iACP-DFSRA to predict ACPs more effectively.
Collapse
Affiliation(s)
- Xin Wang
- School of Science, Dalian Maritime University, Dalian 116026, China.
| | - Zimeng Zhang
- School of Science, Dalian Maritime University, Dalian 116026, China
| | - Chang Liu
- School of Science, Dalian Maritime University, Dalian 116026, China
| |
Collapse
|
7
|
Wang X, Wang S. ACP-PDAFF: Pretrained model and dual-channel attentional feature fusion for anticancer peptides prediction. Comput Biol Chem 2024; 112:108141. [PMID: 38996756 DOI: 10.1016/j.compbiolchem.2024.108141] [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: 01/01/2024] [Revised: 05/26/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024]
Abstract
Anticancer peptides(ACPs) have attracted significant interest as a novel method of treating cancer due to their ability to selectively kill cancer cells without damaging normal cells. Many artificial intelligence-based methods have demonstrated impressive performance in predicting ACPs. Nevertheless, the limitations of existing methods in feature engineering include handcrafted features driven by prior knowledge, insufficient feature extraction, and inefficient feature fusion. In this study, we propose a model based on a pretrained model, and dual-channel attentional feature fusion(DAFF), called ACP-PDAFF. Firstly, to reduce the heavy dependence on expert knowledge-based handcrafted features, binary profile features (BPF) and physicochemical properties features(PCPF) are used as inputs to the transformer model. Secondly, aimed at learning more diverse feature informations of ACPs, a pretrained model ProtBert is utilized. Thirdly, for better fusion of different feature channels, DAFF is employed. Finally, to evaluate the performance of the model, we compare it with other methods on five benchmark datasets, including ACP-Mixed-80 dataset, Main and Alternate datasets of AntiCP 2.0, LEE and Independet dataset, and ACPred-Fuse dataset. And the accuracies obtained by ACP-PDAFF are 0.86, 0.80, 0.94, 0.97 and 0.95 on five datasets, respectively, higher than existing methods by 1% to 12%. Therefore, by learning rich feature informations and effectively fusing different feature channels, ACD-PDAFF achieves outstanding performance. Our code and the datasets are available at https://github.com/wongsing/ACP-PDAFF.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Computer Science and Engineering, School of Information Science and Engineering, Yunnan University, Kunming, 650504, Yunnan, China
| | - Shunfang Wang
- Department of Computer Science and Engineering, School of Information Science and Engineering, Yunnan University, Kunming, 650504, Yunnan, China.
| |
Collapse
|
8
|
Isaac KS, Combe M, Potter G, Sokolenko S. Machine learning tools for peptide bioactivity evaluation - Implications for cell culture media optimization and the broader cultivated meat industry. Curr Res Food Sci 2024; 9:100842. [PMID: 39435450 PMCID: PMC11491887 DOI: 10.1016/j.crfs.2024.100842] [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: 05/31/2024] [Accepted: 09/07/2024] [Indexed: 10/23/2024] Open
Abstract
Although bioactive peptides have traditionally been studied for their health-promoting qualities in the context of nutrition and medicine, the past twenty years have seen a steady increase in their application to cell culture media optimization. Complex natural sources of bioactive peptides, such as hydrolysates, offer a sustainable and cost-effective means of promoting cellular growth, making them an essential component of scaling-up cultivated meat production. However, the sheer diversity of hydrolysates makes product selection difficult, highlighting the need for functional characterization. Traditional wet-lab techniques for isolating and estimating peptide bioactivity cannot keep pace with peptide identification using high-throughput tools such as mass spectrometry, requiring the development and use of machine learning-based classifiers. This review provides a comprehensive list of available software tools to evaluate peptide bioactivity, classified and compared based on the algorithm, training set, functionality, and limitations of the underlying models. We curated independent test sets to compare the predictive performance of different models based on specific bioactivity classification relevant to promoting cell culture growth: antioxidant and anti-inflammatory. A comprehensive screening of all bioactivity classifiers revealed that while there are approximately fifty tools to elucidate antimicrobial activity and sixteen that predict anti-inflammatory activity, fewer tools are available for other functionalities related to cell growth - five that predict antioxidant activity and two for growth factor and/or cell signaling prediction. A thorough evaluation of the available tools revealed significant issues with sensitivity, specificity, and overall accuracy. Despite the overall interest in estimating peptide bioactivity, our work highlights key gaps in the broader adoption of existing software for the specific application of cell culture media optimization in the context of cultivated meat and beyond.
Collapse
Affiliation(s)
- Kathy Sharon Isaac
- Process Engineering and Applied Science, Dalhousie University, 5273 DaCosta Row, PO Box 15000, Halifax, B3H 4R2, NS, Canada
| | - Michelle Combe
- Process Engineering and Applied Science, Dalhousie University, 5273 DaCosta Row, PO Box 15000, Halifax, B3H 4R2, NS, Canada
| | | | - Stanislav Sokolenko
- Process Engineering and Applied Science, Dalhousie University, 5273 DaCosta Row, PO Box 15000, Halifax, B3H 4R2, NS, Canada
| |
Collapse
|
9
|
Garai S, Thomas J, Dey P, Das D. LGBM-ACp: an ensemble model for anticancer peptide prediction and in silico screening with potential drug targets. Mol Divers 2024; 28:1965-1981. [PMID: 36637711 DOI: 10.1007/s11030-023-10602-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/06/2023] [Indexed: 01/14/2023]
Abstract
Conventional cancer therapies are highly expensive and have serious complications. An alternative approach now emphasizes on the development of small, biologically active peptides without acute toxicity. Experimental screening to find curative anticancer peptides (ACP) often gives rise to multiple obstacles and is time dependent. Consequently, developing an effective computational technique to identify promising ACP candidates prior to preclinical research is in high demand. This study proposed a machine-learning framework that used the light gradient-boosting machine as a classifier and two compositional and two binary profile features as input. The ensemble model displayed an accuracy, MCC, and AUROC of 97.52%, 0.91, and 0.98, respectively, which outclassed most of the existing sequence-based computational tools. A distinct dataset of non-mutagenic, non-toxic, and non-inhibitory Cytochrome P-450 peptides was used to validate the hybrid model. The most relevant ACP in the alternative dataset was compared with two standard ACPs, beta defensin 2, and cecropin-A. Molecular docking of the predicted peptide revealed that it has a strong binding affinity with twenty-five anticancer drug targets, most notably phosphoenolpyruvate carboxykinase (- 7.2 kcal/mol). Additionally, molecular dynamics simulation and principal component analysis supported the stability of the peptide-receptor complex. Overall, the present findings will take a step forward in rational drug design through rapid identification and screening of therapeutic peptides.
Collapse
Affiliation(s)
- Swarnava Garai
- Department of Bioengineering, NIT Agartala, Tripura, 799046, India
| | - Juanit Thomas
- Department of Bioengineering, NIT Agartala, Tripura, 799046, India
| | - Palash Dey
- Civil Engineering Department, The ICFAI University, Tripura, 799210, India
| | - Deeplina Das
- Department of Bioengineering, NIT Agartala, Tripura, 799046, India.
| |
Collapse
|
10
|
Ullah F, Salam A, Nadeem M, Amin F, AlSalman H, Abrar M, Alfakih T. Extended dipeptide composition framework for accurate identification of anticancer peptides. Sci Rep 2024; 14:17381. [PMID: 39075193 PMCID: PMC11286958 DOI: 10.1038/s41598-024-68475-8] [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: 06/19/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024] Open
Abstract
The identification of anticancer peptides (ACPs) is crucial, especially in the development of peptide-based cancer therapy. The classical models such as Split Amino Acid Composition (SAAC) and Pseudo Amino Acid Composition (PseAAC) lack the incorporation of feature representation. These advancements improve the predictive accuracy and efficiency of ACP identification. Thus, the effort of this research is to propose and develop an advanced framework based on feature extraction. Thus, to achieve this objective herein we propose an Extended Dipeptide Composition (EDPC) framework. The proposed EDPC framework extends the dipeptide composition by considering the local sequence environment information and reforming the CD-HIT framework to remove noise and redundancy. To measure the accuracy, we have performed several experiments. These experiments were employed using four famous machine learning (ML) algorithms named; Support Vector Machine (SVM), Decision Tree (DT), Random Forest (RF), and K Nearest Neighbor (KNN). For comparisons, we have used accuracy, specificity, sensitivity, precision, recall, and F1-Score as evaluation criteria. The reliability of the proposed framework is further evaluated using statistical significance tests. As a result, the proposed EDPC framework exhibited enhanced performance than SAAC and PseAAC, where the SVM model delivered the highest accuracy of 96. 6% and significant enhancements in specificity, sensitivity, precision, and F1-score over multiple datasets. Due to the incorporation of enhanced feature representation and the incorporation of local and global sequence profiles proposed EDPC achieves higher classification performance. The proposed frameworks can deal with noise and also duplicating features. These are accompanied by a wide range of feature representations. Finally, our proposed framework can be used for clinical applications where ACP identification is essential. Future works will include extending to a larger variety of datasets, incorporating tertiary structural information, and using deep learning techniques to improve the proposed EDPC.
Collapse
Affiliation(s)
- Faizan Ullah
- Department of Computer Science, Bacha Khan University, Charsadda, 24420, Pakistan
| | - Abdu Salam
- Department of Computer Science, Abdul Wali Khan University, Mardan, 23200, Pakistan
| | - Muhammad Nadeem
- Department of Computer Science and Software Engineering, International Islamic University, Islamabad, 44000, Pakistan
| | - Farhan Amin
- School of Computer Science and Engineering, Yeungnam University, Gyeongsan, 38541, Korea.
| | - Hussain AlSalman
- Department of Computer Science, College of Computer and Information Sciences, King Saud University, 11543, Riyadh, Saudi Arabia.
| | - Mohammad Abrar
- Faculty of Computer Studies, Arab Open University, Muscat, Oman
| | - Taha Alfakih
- Department of Information Systems, College of Computer and Information Sciences, King Saud University, 11543, Riyadh, Saudi Arabia
| |
Collapse
|
11
|
Kao HJ, Weng TH, Chen CH, Chen YC, Chi YH, Huang KY, Weng SL. Integrating In Silico and In Vitro Approaches to Identify Natural Peptides with Selective Cytotoxicity against Cancer Cells. Int J Mol Sci 2024; 25:6848. [PMID: 38999958 PMCID: PMC11240926 DOI: 10.3390/ijms25136848] [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: 05/15/2024] [Revised: 06/14/2024] [Accepted: 06/18/2024] [Indexed: 07/14/2024] Open
Abstract
Anticancer peptides (ACPs) are bioactive compounds known for their selective cytotoxicity against tumor cells via various mechanisms. Recent studies have demonstrated that in silico machine learning methods are effective in predicting peptides with anticancer activity. In this study, we collected and analyzed over a thousand experimentally verified ACPs, specifically targeting peptides derived from natural sources. We developed a precise prediction model based on their sequence and structural features, and the model's evaluation results suggest its strong predictive ability for anticancer activity. To enhance reliability, we integrated the results of this model with those from other available methods. In total, we identified 176 potential ACPs, some of which were synthesized and further evaluated using the MTT colorimetric assay. All of these putative ACPs exhibited significant anticancer effects and selective cytotoxicity against specific tumor cells. In summary, we present a strategy for identifying and characterizing natural peptides with selective cytotoxicity against cancer cells, which could serve as novel therapeutic agents. Our prediction model can effectively screen new molecules for potential anticancer activity, and the results from in vitro experiments provide compelling evidence of the candidates' anticancer effects and selective cytotoxicity.
Collapse
Affiliation(s)
- Hui-Ju Kao
- Department of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Department of Medical Research, Hsinchu Municipal MacKay Children's Hospital, Hsinchu City 300, Taiwan
| | - Tzu-Han Weng
- Department of Dermatology, MacKay Memorial Hospital, Taipei City 104, Taiwan
| | - Chia-Hung Chen
- Department of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Department of Medical Research, Hsinchu Municipal MacKay Children's Hospital, Hsinchu City 300, Taiwan
| | - Yu-Chi Chen
- Department of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Department of Medical Research, Hsinchu Municipal MacKay Children's Hospital, Hsinchu City 300, Taiwan
| | - Yu-Hsiang Chi
- National Center for High-Performance Computing, Hsinchu City 300, Taiwan
| | - Kai-Yao Huang
- Department of Medical Research, Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Department of Medical Research, Hsinchu Municipal MacKay Children's Hospital, Hsinchu City 300, Taiwan
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
- Institute of Biomedical Sciences, MacKay Medical College, New Taipei City 252, Taiwan
| | - Shun-Long Weng
- Department of Medicine, MacKay Medical College, New Taipei City 252, Taiwan
- Department of Obstetrics and Gynecology, Hsinchu MacKay Memorial Hospital, Hsinchu City 300, Taiwan
- Department of Obstetrics and Gynecology, Hsinchu Municipal MacKay Children's Hospital, Hsinchu City 300, Taiwan
| |
Collapse
|
12
|
Ghafoor H, Asim MN, Ibrahim MA, Ahmed S, Dengel A. CAPTURE: Comprehensive anti-cancer peptide predictor with a unique amino acid sequence encoder. Comput Biol Med 2024; 176:108538. [PMID: 38759585 DOI: 10.1016/j.compbiomed.2024.108538] [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: 01/08/2024] [Revised: 04/26/2024] [Accepted: 04/28/2024] [Indexed: 05/19/2024]
Abstract
Anticancer peptides (ACPs) key properties including bioactivity, high efficacy, low toxicity, and lack of drug resistance make them ideal candidates for cancer therapies. To deeply explore the potential of ACPs and accelerate development of cancer therapies, although 53 Artificial Intelligence supported computational predictors have been developed for ACPs and non ACPs classification but only one predictor has been developed for ACPs functional types annotations. Moreover, these predictors extract amino acids distribution patterns to transform peptides sequences into statistical vectors that are further fed to classifiers for discriminating peptides sequences and annotating peptides functional classes. Overall, these predictors remain fail in extracting diverse types of amino acids distribution patterns from peptide sequences. The paper in hand presents a unique CARE encoder that transforms peptides sequences into statistical vectors by extracting 4 different types of distribution patterns including correlation, distribution, composition, and transition. Across public benchmark dataset, proposed encoder potential is explored under two different evaluation settings namely; intrinsic and extrinsic. Extrinsic evaluation indicates that 12 different machine learning classifiers achieve superior performance with the proposed encoder as compared to 55 existing encoders. Furthermore, an intrinsic evaluation reveals that, unlike existing encoders, the proposed encoder generates more discriminative clusters for ACPs and non-ACPs classes. Across 8 public benchmark ACPs and non-ACPs classification datasets, proposed encoder and Adaboost classifier based CAPTURE predictor outperforms existing predictors with an average accuracy, recall and MCC score of 1%, 4%, and 2% respectively. In generalizeability evaluation case study, across 7 benchmark anti-microbial peptides classification datasets, CAPTURE surpasses existing predictors by an average AU-ROC of 2%. CAPTURE predictive pipeline along with label powerset method outperforms state-of-the-art ACPs functional types predictor by 5%, 5%, 5%, 6%, and 3% in terms of average accuracy, subset accuracy, precision, recall, and F1 respectively. CAPTURE web application is available at https://sds_genetic_analysis.opendfki.de/CAPTURE.
Collapse
Affiliation(s)
- Hina Ghafoor
- Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, 67663, Germany; German Research Center for Artificial Intelligence GmbH, Kaiserslautern, 67663, Germany
| | - Muhammad Nabeel Asim
- German Research Center for Artificial Intelligence GmbH, Kaiserslautern, 67663, Germany.
| | - Muhammad Ali Ibrahim
- Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, 67663, Germany; German Research Center for Artificial Intelligence GmbH, Kaiserslautern, 67663, Germany
| | - Sheraz Ahmed
- German Research Center for Artificial Intelligence GmbH, Kaiserslautern, 67663, Germany
| | - Andreas Dengel
- Department of Computer Science, Rhineland-Palatinate Technical University of Kaiserslautern-Landau, Kaiserslautern, 67663, Germany; German Research Center for Artificial Intelligence GmbH, Kaiserslautern, 67663, Germany
| |
Collapse
|
13
|
Lv Y, Feng G, Yang L, Wu X, Wang C, Ye A, wang S, Xu C, Shi H. Differential whole-genome doubling based signatures for improvement on clinical outcomes and drug response in patients with breast cancer. Heliyon 2024; 10:e28586. [PMID: 38576569 PMCID: PMC10990872 DOI: 10.1016/j.heliyon.2024.e28586] [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: 08/12/2023] [Revised: 03/20/2024] [Accepted: 03/20/2024] [Indexed: 04/06/2024] Open
Abstract
Whole genome doublings (WGD), a hallmark of human cancer, is pervasive in breast cancer patients. However, the molecular mechanism of the complete impact of WGD on survival and treatment response in breast cancer remains unclear. To address this, we performed a comprehensive and systematic analysis of WGD, aiming to identify distinct genetic alterations linked to WGD and highlight its improvement on clinical outcomes and treatment response for breast cancer. A linear regression model along with weighted gene co-expression network analysis (WGCNA) was applied on The Cancer Genome Atlas (TCGA) dataset to identify critical genes related to WGD. Further Cox regression models with random selection were used to optimize the most useful prognostic markers in the TCGA dataset. The clinical implication of the risk model was further assessed through prognostic impact evaluation, tumor stratification, functional analysis, genomic feature difference analysis, drug response analysis, and multiple independent datasets for validation. Our findings revealed a high aneuploidy burden, chromosomal instability (CIN), copy number variation (CNV), and mutation burden in breast tumors exhibiting WGD events. Moreover, 247 key genes associated with WGD were identified from the distinct genomic patterns in the TCGA dataset. A risk model consisting of 22 genes was optimized from the key genes. High-risk breast cancer patients were more prone to WGD and exhibited greater genomic diversity compared to low-risk patients. Some oncogenic signaling pathways were enriched in the high-risk group, while primary immune deficiency pathways were enriched in the low-risk group. We also identified a risk gene, ANLN (anillin), which displayed a strong positive correlation with two crucial WGD genes, KIF18A and CCNE2. Tumors with high expression of ANLN were more prone to WGD events and displayed worse clinical survival outcomes. Furthermore, the expression levels of these risk genes were significantly associated with the sensitivities of BRCA cell lines to multiple drugs, providing valuable insights for targeted therapies. These findings will be helpful for further improvement on clinical outcomes and contribution to drug development in breast cancer.
Collapse
Affiliation(s)
| | | | - Lei Yang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xiaoliang Wu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Chengyi Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Aokun Ye
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Shuyuan wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Chaohan Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Hongbo Shi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| |
Collapse
|
14
|
Yang X, Jin J, Wang R, Li Z, Wang Y, Wei L. CACPP: A Contrastive Learning-Based Siamese Network to Identify Anticancer Peptides Based on Sequence Only. J Chem Inf Model 2024; 64:2807-2816. [PMID: 37252890 DOI: 10.1021/acs.jcim.3c00297] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Anticancer peptides (ACPs) recently have been receiving increasing attention in cancer therapy due to their low consumption, few adverse side effects, and easy accessibility. However, it remains a great challenge to identify anticancer peptides via experimental approaches, requiring expensive and time-consuming experimental studies. In addition, traditional machine-learning-based methods are proposed for ACP prediction mainly depending on hand-crafted feature engineering, which normally achieves low prediction performance. In this study, we propose CACPP (Contrastive ACP Predictor), a deep learning framework based on the convolutional neural network (CNN) and contrastive learning for accurately predicting anticancer peptides. In particular, we introduce the TextCNN model to extract the high-latent features based on the peptide sequences only and exploit the contrastive learning module to learn more distinguishable feature representations to make better predictions. Comparative results on the benchmark data sets indicate that CACPP outperforms all the state-of-the-art methods in the prediction of anticancer peptides. Moreover, to intuitively show that our model has good classification ability, we visualize the dimension reduction of the features from our model and explore the relationship between ACP sequences and anticancer functions. Furthermore, we also discuss the influence of data set construction on model prediction and explore our model performance on the data sets with verified negative samples.
Collapse
Affiliation(s)
- Xuetong Yang
- School of Software, Shandong University, Jinan 250101, China
- Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan 250101, China
| | - Junru Jin
- School of Software, Shandong University, Jinan 250101, China
- Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan 250101, China
| | - Ruheng Wang
- School of Software, Shandong University, Jinan 250101, China
- Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan 250101, China
| | - Zhongshen Li
- School of Software, Shandong University, Jinan 250101, China
- Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan 250101, China
| | - Yu Wang
- School of Software, Shandong University, Jinan 250101, China
- Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan 250101, China
| | - Leyi Wei
- School of Software, Shandong University, Jinan 250101, China
- Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan 250101, China
| |
Collapse
|
15
|
Mitchell SM, Heise RM, Murray ME, Lambo DJ, Daso RE, Banerjee IA. An investigation of binding interactions of tumor-targeted peptide conjugated polyphenols with the kinase domain of ephrin B4 and B2 receptors. Mol Divers 2024; 28:817-849. [PMID: 36847923 PMCID: PMC9969393 DOI: 10.1007/s11030-023-10621-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 02/02/2023] [Indexed: 03/01/2023]
Abstract
Recent studies have shown that Ephrin receptors may be upregulated in several types of cancers including breast, ovarian and endometrial cancers, making them a target for drug design. In this work, we have utilized a target-hopping approach to design new natural product-peptide conjugates and examined their interactions with the kinase-binding domain of EphB4 and EphB2 receptors. The peptide sequences were generated through point mutations of the known EphB4 antagonist peptide TNYLFSPNGPIA. Their anticancer properties and secondary structures were analyzed computationally. Conjugates of most optimum of peptides were then designed by binding the N-terminal of the peptides with the free carboxyl group of the polyphenols sinapate, gallate and coumarate, which are known for their inherent anticancer properties. To investigate if these conjugates have a potential to bind to the kinase domain, we carried out docking studies and MMGBSA free energy calculations of the trajectories based on the molecular dynamics simulations, with both the apo and the ATP bound kinase domains of both receptors. In most cases binding interactions occurred within the catalytic loop region, while in some cases the conjugates were found to spread out across the N-lobe and the DFG motif region. The conjugates were further tested for prediction of pharmacokinetic properties using ADME studies. Our results indicated that the conjugates were lipophilic and MDCK permeable with no CYP interactions. These findings provide an insight into the molecular interactions of these peptides and conjugates with the kinase domain of the EphB4 and EphB2 receptor. As a proof of concept, we synthesized and carried out SPR analysis with two of the conjugates (gallate-TNYLFSPNGPIA and sinapate-TNYLFSPNGPIA). Results indicated that the conjugates showed higher binding with the EphB4 receptor and minimal binding to EphB2 receptor. Sinapate-TNYLFSPNGPIA showed inhibitory activity against EphB4. These studies reveal that some of the conjugates may be developed for further investigation into in vitro and in vivo studies and potential development as therapeutics.
Collapse
Affiliation(s)
- Saige M Mitchell
- Department of Chemistry, Fordham University, 441 E. Fordham Rd, Bronx, NY, 10458, USA
| | - Ryan M Heise
- Department of Chemistry, Fordham University, 441 E. Fordham Rd, Bronx, NY, 10458, USA
| | - Molly E Murray
- Department of Chemistry, Fordham University, 441 E. Fordham Rd, Bronx, NY, 10458, USA
| | - Dominic J Lambo
- Department of Chemistry, Fordham University, 441 E. Fordham Rd, Bronx, NY, 10458, USA
| | - Rachel E Daso
- Department of Chemistry, Fordham University, 441 E. Fordham Rd, Bronx, NY, 10458, USA
| | - Ipsita A Banerjee
- Department of Chemistry, Fordham University, 441 E. Fordham Rd, Bronx, NY, 10458, USA.
| |
Collapse
|
16
|
Li C, Jin K. Chemical Strategies towards the Development of Effective Anticancer Peptides. Curr Med Chem 2024; 31:1839-1873. [PMID: 37170992 DOI: 10.2174/0929867330666230426111157] [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: 11/01/2022] [Revised: 01/28/2023] [Accepted: 02/24/2023] [Indexed: 05/13/2023]
Abstract
Cancer is increasingly recognized as one of the primary causes of death and has become a multifaceted global health issue. Modern medical science has made significant advancements in the diagnosis and therapy of cancer over the past decade. The detrimental side effects, lack of efficacy, and multidrug resistance of conventional cancer therapies have created an urgent need for novel anticancer therapeutics or treatments with low cytotoxicity and drug resistance. The pharmaceutical groups have recognized the crucial role that peptide therapeutic agents can play in addressing unsatisfied healthcare demands and how these become great supplements or even preferable alternatives to biological therapies and small molecules. Anticancer peptides, as a vibrant therapeutic strategy against various cancer cells, have demonstrated incredible anticancer potential due to high specificity and selectivity, low toxicity, and the ability to target the surface of traditional "undruggable" proteins. This review will provide the research progression of anticancer peptides, mainly focusing on the discovery and modifications along with the optimization and application of these peptides in clinical practice.
Collapse
Affiliation(s)
- Cuicui Li
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Kang Jin
- Key Laboratory of Chemical Biology (Ministry of Education), Department of Medicinal Chemistry, School of Pharmacy, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| |
Collapse
|
17
|
La Paglia L, Vazzana M, Mauro M, Urso A, Arizza V, Vizzini A. Bioactive Molecules from the Innate Immunity of Ascidians and Innovative Methods of Drug Discovery: A Computational Approach Based on Artificial Intelligence. Mar Drugs 2023; 22:6. [PMID: 38276644 PMCID: PMC10817596 DOI: 10.3390/md22010006] [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: 11/20/2023] [Revised: 12/12/2023] [Accepted: 12/17/2023] [Indexed: 01/27/2024] Open
Abstract
The study of bioactive molecules of marine origin has created an important bridge between biological knowledge and its applications in biotechnology and biomedicine. Current studies in different research fields, such as biomedicine, aim to discover marine molecules characterized by biological activities that can be used to produce potential drugs for human use. In recent decades, increasing attention has been paid to a particular group of marine invertebrates, the Ascidians, as they are a source of bioactive products. We describe omics data and computational methods relevant to identifying the mechanisms and processes of innate immunity underlying the biosynthesis of bioactive molecules, focusing on innovative computational approaches based on Artificial Intelligence. Since there is increasing attention on finding new solutions for a sustainable supply of bioactive compounds, we propose that a possible improvement in the biodiscovery pipeline might also come from the study and utilization of marine invertebrates' innate immunity.
Collapse
Affiliation(s)
- Laura La Paglia
- Istituto di Calcolo e Reti ad Alte Prestazioni–Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy; (L.L.P.); (A.U.)
| | - Mirella Vazzana
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche–Università di Palermo, Via Archirafi 18, 90100 Palermo, Italy; (M.V.); (M.M.); (V.A.)
| | - Manuela Mauro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche–Università di Palermo, Via Archirafi 18, 90100 Palermo, Italy; (M.V.); (M.M.); (V.A.)
| | - Alfonso Urso
- Istituto di Calcolo e Reti ad Alte Prestazioni–Consiglio Nazionale delle Ricerche, Via Ugo La Malfa 153, 90146 Palermo, Italy; (L.L.P.); (A.U.)
| | - Vincenzo Arizza
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche–Università di Palermo, Via Archirafi 18, 90100 Palermo, Italy; (M.V.); (M.M.); (V.A.)
| | - Aiti Vizzini
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche–Università di Palermo, Via Archirafi 18, 90100 Palermo, Italy; (M.V.); (M.M.); (V.A.)
| |
Collapse
|
18
|
Sun M, Hu H, Pang W, Zhou Y. ACP-BC: A Model for Accurate Identification of Anticancer Peptides Based on Fusion Features of Bidirectional Long Short-Term Memory and Chemically Derived Information. Int J Mol Sci 2023; 24:15447. [PMID: 37895128 PMCID: PMC10607064 DOI: 10.3390/ijms242015447] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/10/2023] [Accepted: 10/20/2023] [Indexed: 10/29/2023] Open
Abstract
Anticancer peptides (ACPs) have been proven to possess potent anticancer activities. Although computational methods have emerged for rapid ACPs identification, their accuracy still needs improvement. In this study, we propose a model called ACP-BC, a three-channel end-to-end model that utilizes various combinations of data augmentation techniques. In the first channel, features are extracted from the raw sequence using a bidirectional long short-term memory network. In the second channel, the entire sequence is converted into a chemical molecular formula, which is further simplified using Simplified Molecular Input Line Entry System notation to obtain deep abstract features through a bidirectional encoder representation transformer (BERT). In the third channel, we manually selected four effective features according to dipeptide composition, binary profile feature, k-mer sparse matrix, and pseudo amino acid composition. Notably, the application of chemical BERT in predicting ACPs is novel and successfully integrated into our model. To validate the performance of our model, we selected two benchmark datasets, ACPs740 and ACPs240. ACP-BC achieved prediction accuracy with 87% and 90% on these two datasets, respectively, representing improvements of 1.3% and 7% compared to existing state-of-the-art methods on these datasets. Therefore, systematic comparative experiments have shown that the ACP-BC can effectively identify anticancer peptides.
Collapse
Affiliation(s)
- Mingwei Sun
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (M.S.); (H.H.)
| | - Haoyuan Hu
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (M.S.); (H.H.)
| | - Wei Pang
- School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh EH14 4AS, UK;
| | - You Zhou
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun 130012, China; (M.S.); (H.H.)
- College of Software, Jilin University, Changchun 130012, China
| |
Collapse
|
19
|
Barman P, Joshi S, Sharma S, Preet S, Sharma S, Saini A. Strategic Approaches to Improvise Peptide Drugs as Next Generation Therapeutics. Int J Pept Res Ther 2023; 29:61. [PMID: 37251528 PMCID: PMC10206374 DOI: 10.1007/s10989-023-10524-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2023] [Indexed: 05/31/2023]
Abstract
In recent years, the occurrence of a wide variety of drug-resistant diseases has led to an increase in interest in alternate therapies. Peptide-based drugs as an alternate therapy hold researchers' attention in various therapeutic fields such as neurology, dermatology, oncology, metabolic diseases, etc. Previously, they had been overlooked by pharmaceutical companies due to certain limitations such as proteolytic degradation, poor membrane permeability, low oral bioavailability, shorter half-life, and poor target specificity. Over the last two decades, these limitations have been countered by introducing various modification strategies such as backbone and side-chain modifications, amino acid substitution, etc. which improve their functionality. This has led to a substantial interest of researchers and pharmaceutical companies, moving the next generation of these therapeutics from fundamental research to the market. Various chemical and computational approaches are aiding the production of more stable and long-lasting peptides guiding the formulation of novel and advanced therapeutic agents. However, there is not a single article that talks about various peptide design approaches i.e., in-silico and in-vitro along with their applications and strategies to improve their efficacy. In this review, we try to bring different aspects of peptide-based therapeutics under one article with a clear focus to cover the missing links in the literature. This review draws emphasis on various in-silico approaches and modification-based peptide design strategies. It also highlights the recent progress made in peptide delivery methods important for their enhanced clinical efficacy. The article would provide a bird's-eye view to researchers aiming to develop peptides with therapeutic applications. Graphical Abstract
Collapse
Affiliation(s)
- Panchali Barman
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Sector 14, Chandigarh, 160014 India
| | - Shubhi Joshi
- Energy Research Centre, Panjab University, Sector 14, Chandigarh, 160014 India
| | - Sheetal Sharma
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| | - Simran Preet
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| | - Shweta Sharma
- Institute of Forensic Science and Criminology (UIEAST), Panjab University, Sector 14, Chandigarh, 160014 India
| | - Avneet Saini
- Department of Biophysics, Panjab University, Sector 25, Chandigarh, U.T 160014 India
| |
Collapse
|
20
|
Ghaly G, Tallima H, Dabbish E, Badr ElDin N, Abd El-Rahman MK, Ibrahim MAA, Shoeib T. Anti-Cancer Peptides: Status and Future Prospects. Molecules 2023; 28:molecules28031148. [PMID: 36770815 PMCID: PMC9920184 DOI: 10.3390/molecules28031148] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/26/2022] [Accepted: 01/19/2023] [Indexed: 01/26/2023] Open
Abstract
The dramatic rise in cancer incidence, alongside treatment deficiencies, has elevated cancer to the second-leading cause of death globally. The increasing morbidity and mortality of this disease can be traced back to a number of causes, including treatment-related side effects, drug resistance, inadequate curative treatment and tumor relapse. Recently, anti-cancer bioactive peptides (ACPs) have emerged as a potential therapeutic choice within the pharmaceutical arsenal due to their high penetration, specificity and fewer side effects. In this contribution, we present a general overview of the literature concerning the conformational structures, modes of action and membrane interaction mechanisms of ACPs, as well as provide recent examples of their successful employment as targeting ligands in cancer treatment. The use of ACPs as a diagnostic tool is summarized, and their advantages in these applications are highlighted. This review expounds on the main approaches for peptide synthesis along with their reconstruction and modification needed to enhance their therapeutic effect. Computational approaches that could predict therapeutic efficacy and suggest ACP candidates for experimental studies are discussed. Future research prospects in this rapidly expanding area are also offered.
Collapse
Affiliation(s)
- Gehane Ghaly
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt
| | - Hatem Tallima
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt
| | - Eslam Dabbish
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt
| | - Norhan Badr ElDin
- Analytical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr-El Aini Street, Cairo 11562, Egypt
| | - Mohamed K. Abd El-Rahman
- Analytical Chemistry Department, Faculty of Pharmacy, Cairo University, Kasr-El Aini Street, Cairo 11562, Egypt
- Department of Chemistry and Chemical Biology, Harvard University, 12 Oxford Street, Cambridge, MA 02138, USA
| | - Mahmoud A. A. Ibrahim
- Computational Chemistry Laboratory, Chemistry Department, Faculty of Science, Minia University, Minia 61519, Egypt
- School of Health Sciences, University of Kwa-Zulu-Natal, Westville, Durban 4000, South Africa
| | - Tamer Shoeib
- Department of Chemistry, The American University in Cairo, New Cairo 11835, Egypt
- Correspondence:
| |
Collapse
|
21
|
Yuan Q, Chen K, Yu Y, Le NQK, Chua MCH. Prediction of anticancer peptides based on an ensemble model of deep learning and machine learning using ordinal positional encoding. Brief Bioinform 2023; 24:6987656. [PMID: 36642410 DOI: 10.1093/bib/bbac630] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 12/01/2022] [Accepted: 12/28/2022] [Indexed: 01/17/2023] Open
Abstract
Anticancer peptides (ACPs) are the types of peptides that have been demonstrated to have anticancer activities. Using ACPs to prevent cancer could be a viable alternative to conventional cancer treatments because they are safer and display higher selectivity. Due to ACP identification being highly lab-limited, expensive and lengthy, a computational method is proposed to predict ACPs from sequence information in this study. The process includes the input of the peptide sequences, feature extraction in terms of ordinal encoding with positional information and handcrafted features, and finally feature selection. The whole model comprises of two modules, including deep learning and machine learning algorithms. The deep learning module contained two channels: bidirectional long short-term memory (BiLSTM) and convolutional neural network (CNN). Light Gradient Boosting Machine (LightGBM) was used in the machine learning module. Finally, this study voted the three models' classification results for the three paths resulting in the model ensemble layer. This study provides insights into ACP prediction utilizing a novel method and presented a promising performance. It used a benchmark dataset for further exploration and improvement compared with previous studies. Our final model has an accuracy of 0.7895, sensitivity of 0.8153 and specificity of 0.7676, and it was increased by at least 2% compared with the state-of-the-art studies in all metrics. Hence, this paper presents a novel method that can potentially predict ACPs more effectively and efficiently. The work and source codes are made available to the community of researchers and developers at https://github.com/khanhlee/acp-ope/.
Collapse
Affiliation(s)
- Qitong Yuan
- Institute of Systems Science, National University of Singapore, 25 Heng Mui Keng Terrace, 119615, Singapore, Singapore
| | - Keyi Chen
- Institute of Systems Science, National University of Singapore, 25 Heng Mui Keng Terrace, 119615, Singapore, Singapore
| | - Yimin Yu
- Institute of Systems Science, National University of Singapore, 25 Heng Mui Keng Terrace, 119615, Singapore, Singapore
| | - Nguyen Quoc Khanh Le
- Professional Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, 250 Wuxing St, 106, Taipei, Taiwan.,Research Center for Artificial Intelligence in Medicine, Taipei Medical University, 250 Wuxing St, 106, Taipei, Taiwan.,Translational Imaging Research Center, Taipei Medical University Hospital, 252 Wuxing St, 110, Taipei, Taiwan
| | - Matthew Chin Heng Chua
- Institute of Systems Science, National University of Singapore, 25 Heng Mui Keng Terrace, 119615, Singapore, Singapore
| |
Collapse
|
22
|
Kordi M, Borzouyi Z, Chitsaz S, Asmaei MH, Salami R, Tabarzad M. Antimicrobial peptides with anticancer activity: Today status, trends and their computational design. Arch Biochem Biophys 2023; 733:109484. [PMID: 36473507 DOI: 10.1016/j.abb.2022.109484] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/29/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022]
Abstract
Some antimicrobial peptides have been shown to be able to inhibit the proliferation of cancer cell lines. Various strategies for treating cancers with active peptides have been pursued. According to the reports, anticancer peptides are important therapeutic peptides, which can act through two distinct pathways: they either just create pores in the cell membrane, or they have a vital intracellular target. In this review, publications up to Sep. 2021 had extracted form Scopus and PubMed using "antimicrobial peptide" and "anticancer peptide" as keywords. In second step, "computational design" related publications extracted. Among publications, those have similar scopes were classified and selected based on mechanisms of action and application. In this review, the most recent advances in the field of antimicrobial peptides with anti-cancer activities have been summarized. Freely available webservers such as AntiCP, ACPP, iACP, iACP-GAEnsC, ACPred are discussed here. In conclusion, despite some limitations of ACPs such as production cost and challenges, short half-life and toxicity on normal cells, the beneficial properties of AMPs make some of them good therapeutic agents for cancer therapy. Towards designing novel ACPs, the computational methods have substantial position and have been used progressively, today.
Collapse
Affiliation(s)
- Masoumeh Kordi
- Department of Plant Science and Biotechnology, School of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Zeynab Borzouyi
- Department of Agriculture, School of Agriculture and Plant Breeding, Islamic Azad University, Sabzevar, Iran
| | - Saideh Chitsaz
- Department of Microbiology, Islamic Azad University, Karaj, Iran
| | | | - Robab Salami
- Department of Plant Science and Biotechnology, School of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Maryam Tabarzad
- Protein Technology Research Center, Shahid Beheshti University of Medical Science, Iran.
| |
Collapse
|
23
|
Liang Y, Ma X. iACP-GE: accurate identification of anticancer peptides by using gradient boosting decision tree and extra tree. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2023; 34:1-19. [PMID: 36562289 DOI: 10.1080/1062936x.2022.2160011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 12/12/2022] [Indexed: 06/17/2023]
Abstract
Cancer is one of the main diseases threatening human life, accounting for millions of deaths around the world each year. Traditional physical and chemical methods for cancer treatment are extremely time-consuming, lab-intensive, expensive, inefficient and difficult to be applied in a high-throughput way. Hence, it is an urgent task to develop automated computational methods to enable fast and accurate identification of anticancer peptides (ACPs). In this paper, we develop a novel model named iACP-GE to identify ACPs. Multi-features are extracted by using binary encoding, enhanced grouped amino acid composition and BLOSUM62 encoding based on the N5C5 sequence, as well as detrended forward moving-average auto-cross correlation analysis based on physicochemical properties of 20 natural amino acids. Thus, 835 features are obtained for each sample, in order to avoid information redundancy, gradient boosting decision tree was adopted as the feature selection strategy. Then, the optimal feature subset is input to the extra tree classifier. The accuracies of ACP740 and ACP240 datasets with the 5-fold cross-validation were 90.54% and 91.25%, respectively. Experimental results indicate that iACP-GE significantly outperforms several existing models on ACP740 and ACP240 datasets and can be used as an effective tool for the identification of ACPs. The datasets and source codes for iACP-GE are available at https://github.com/yunyunliang88/iACP-GE.
Collapse
Affiliation(s)
- Y Liang
- School of Science, Xi'an Polytechnic University, Xi'an, P. R. China
| | - X Ma
- School of Science, Xi'an Polytechnic University, Xi'an, P. R. China
| |
Collapse
|
24
|
Wu X, Zeng W, Lin F. GCNCPR-ACPs: a novel graph convolution network method for ACPs prediction. BMC Bioinformatics 2022; 23:560. [PMID: 36564705 PMCID: PMC9789540 DOI: 10.1186/s12859-022-04771-2] [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: 05/27/2022] [Accepted: 05/31/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Anticancer peptide (ACP) inhibits and kills tumor cells. Research on ACP is of great significance for the development of new drugs, and the prediction of ACPs and non-ACPs is the new hotspot. RESULTS We propose a new machine learning-based method named GCNCPR-ACPs (a Graph Convolutional Neural Network Method based on collapse pooling and residual network to predict the ACPs), which automatically and accurately predicts ACPs using residual graph convolution networks, differentiable graph pooling, and features extracted using peptide sequence information extraction. The GCNCPR-ACPs method can effectively capture different levels of node attributes for amino acid node representation learning, GCNCPR-ACPs uses node2vec and one-hot embedding methods to extract initial amino acid features for ACP prediction. CONCLUSIONS Experimental results of ten-fold cross-validation and independent validation based on different metrics showed that GCNCPR-ACPs significantly outperformed state-of-the-art methods. Specifically, the evaluation indicators of Matthews Correlation Coefficient (MCC) and AUC of our predicator were 69.5% and 90%, respectively, which were 4.3% and 2% higher than those of the other predictors, respectively, in ten-fold cross-validation. And in the independent test, the scores of MCC and SP were 69.6% and 93.9%, respectively, which were 37.6% and 5.5% higher than those of the other predictors, respectively. The overall results showed that the GCNCPR-ACPs method proposed in the current paper can effectively predict ACPs.
Collapse
Affiliation(s)
- Xiujin Wu
- grid.12955.3a0000 0001 2264 7233School of Informatics, Xiamen University, Xiamen, Fujian China
| | - Wenhua Zeng
- grid.12955.3a0000 0001 2264 7233School of Informatics, Xiamen University, Xiamen, Fujian China
| | - Fan Lin
- grid.12955.3a0000 0001 2264 7233School of Informatics, Xiamen University, Xiamen, Fujian China ,grid.2515.30000 0004 0378 8438Computational Health Informatics Program, Boston Children’s Hospital, Boston, MA USA
| |
Collapse
|
25
|
ACPred-BMF: bidirectional LSTM with multiple feature representations for explainable anticancer peptide prediction. Sci Rep 2022; 12:21915. [PMID: 36535969 PMCID: PMC9763336 DOI: 10.1038/s41598-022-24404-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 11/15/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer has become a major factor threatening human life and health. Under the circumstance that traditional treatment methods such as chemotherapy and radiotherapy are not highly specific and often cause severe side effects and toxicity, new treatment methods are urgently needed. Anticancer peptide drugs have low toxicity, stronger efficacy and specificity, and have emerged as a new type of cancer treatment drugs. However, experimental identification of anticancer peptides is time-consuming and expensive, and difficult to perform in a high-throughput manner. Computational identification of anticancer peptides can make up for the shortcomings of experimental identification. In this study, a deep learning-based predictor named ACPred-BMF is proposed for the prediction of anticancer peptides. This method uses the quantitative and qualitative properties of amino acids, binary profile feature to numerical representation for the peptide sequences. The Bidirectional LSTM network architecture is used in the model, and the attention mechanism is also considered. To alleviate the black-box problem of deep learning model prediction, we visualized the automatically extracted features and used the Shapley additive explanations algorithm to determine the importance of features to further understand the anticancer peptide mechanism. The results show that our method is one of the state-of-the-art anticancer peptide predictors. A web server as the implementation of ACPred-BMF that can be accessed via: http://mialab.ruc.edu.cn/ACPredBMFServer/ .
Collapse
|
26
|
Function Prediction of Peptide Toxins with Sequence-Based Multi-Tasking PU Learning Method. Toxins (Basel) 2022; 14:toxins14110811. [PMID: 36422985 PMCID: PMC9696491 DOI: 10.3390/toxins14110811] [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: 08/27/2022] [Revised: 10/28/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022] Open
Abstract
Peptide toxins generally have extreme pharmacological activities and provide a rich source for the discovery of drug leads. However, determining the optimal activity of a new peptide can be a long and expensive process. In this study, peptide toxins were retrieved from Uniprot; three positive-unlabeled (PU) learning schemes, adaptive basis classifier, two-step method, and PU bagging were adopted to develop models for predicting the biological function of new peptide toxins. All three schemes were embedded with 14 machine learning classifiers. The prediction results of the adaptive base classifier and the two-step method were highly consistent. The models with top comprehensive performances were further optimized by feature selection and hyperparameter tuning, and the models were validated by making predictions for 61 three-finger toxins or the external HemoPI dataset. Biological functions that can be identified by these models include cardiotoxicity, vasoactivity, lipid binding, hemolysis, neurotoxicity, postsynaptic neurotoxicity, hypotension, and cytolysis, with relatively weak predictions for hemostasis and presynaptic neurotoxicity. These models are discovery-prediction tools for active peptide toxins and are expected to accelerate the development of peptide toxins as drugs.
Collapse
|
27
|
Bahadory S, Sadraei J, Zibaei M, Pirestani M, Dalimi A. In vitro anti-gastrointestinal cancer activity of Toxocara canis-derived peptide: Analyzing the expression level of factors related to cell proliferation and tumor growth. Front Pharmacol 2022; 13:878724. [PMID: 36204226 PMCID: PMC9530354 DOI: 10.3389/fphar.2022.878724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Recently, a hypothesis about the negative relationship between cancers and parasites has been proposed and investigated; some parasitic worms and their products can affect the cancer cell proliferation. Due to the potential anti-cancer effect of helminthic parasites, in the present study, the excretory–secretory protein of Toxocara canis (T. canis) parasite was used to evaluate the possible anti-cancer properties and their effect on gastrointestinal and liver cancer cell proliferation-related genes in laboratory conditions. Methods and materials: The selected synthesized peptide fraction from the T. canis excretory–secretory Troponin protein peptide (ES TPP) was exposed at 32, 64, 128, and 256 μg/ml concentrations to three gastrointestinal cancer cell lines AGS, HT-29, and Caco 2, as well as HDF cells as normal cell lines. We used the MTT assay to evaluate cellular changes and cell viability (CV). Variations in gene (Bcl-2, APAF1, ZEB1, VEGF, cyclin-D1, and caspase-3) expression were analyzed by real-time RT-PCR. Results: After 24 h of exposure to pept1ides and cell lines, a decrease in CV was observed at a concentration of 64 μg/ml and compared to the control group. Then, after 48 h, a significant decrease in the CV of Caco 2 cells was observed at a concentration of 32 μg/ml; in the other cancer cell lines, concentrations above 32 μg/ml were effective. The peptide was able to significantly alter the expression of the studied genes at a concentration of 100 μg/ml. Conclusion: Although the studied peptide at high concentrations could have a statistically significant effect on cancer cells, it is still far from the standard drug and can be optimized and promising in future studies.
Collapse
Affiliation(s)
- Saeed Bahadory
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Javid Sadraei
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- *Correspondence: Javid Sadraei,
| | - Mohammad Zibaei
- Department of Parasitology and Mycology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
- Evidence-Based Phytotherapy and Complementary Medicine Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Majid Pirestani
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolhossein Dalimi
- Department of Parasitology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
28
|
Liu J, Li M, Chen X. AntiMF: A deep learning framework for predicting anticancer peptides based on multi-view feature extraction. Methods 2022; 207:38-43. [PMID: 36100141 DOI: 10.1016/j.ymeth.2022.07.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/20/2022] [Accepted: 07/26/2022] [Indexed: 01/10/2023] Open
Abstract
In recent years, anticancer peptides have emerged as a new viable option in cancer therapy, with the ability to overcome the considerable side effects and poor outcomes of standard cancer therapies. Accurate anticancer peptide identification can facilitate its finding and speed up its application in treating cancer. However, many recent approaches are based on machine learning, which not only restricts the representation ability of the models but also requires a complex hand-crafted feature extraction process. Here, we propose AntiMF, a deep learning model that utilizes multi-view mechanism based on different feature extraction models. Comparative results show that our model has a better performance than the state-of-the-art methods in the prediction of anticancer peptides. By using an ensemble learning framework to extract representation, AntiMF can capture the different dimensional information, which can make model representation more complete. Moreover, we visualize what AntiMF learns on one of its ensemble models to intuitively show the effectivity of our model, indicating that AntiMF has the great potential ability to be an effective and useful model to identify anticancer peptides accurately.
Collapse
Affiliation(s)
- Jingjing Liu
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Minghao Li
- Beidahuang Industry Group General Hospital, Harbin 150001, China
| | - Xin Chen
- Eye Hospital, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China; Department of Neurosurgical Laboratory, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
29
|
Zakharova E, Orsi M, Capecchi A, Reymond J. Machine Learning Guided Discovery of Non-Hemolytic Membrane Disruptive Anticancer Peptides. ChemMedChem 2022; 17:e202200291. [PMID: 35880810 PMCID: PMC9541320 DOI: 10.1002/cmdc.202200291] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/29/2022] [Indexed: 12/05/2022]
Abstract
Most antimicrobial peptides (AMPs) and anticancer peptides (ACPs) fold into membrane disruptive cationic amphiphilic α-helices, many of which are however also unpredictably hemolytic and toxic. Here we exploited the ability of recurrent neural networks (RNN) to distinguish active from inactive and non-hemolytic from hemolytic AMPs and ACPs to discover new non-hemolytic ACPs. Our discovery pipeline involved: 1) sequence generation using either a generative RNN or a genetic algorithm, 2) RNN classification for activity and hemolysis, 3) selection for sequence novelty, helicity and amphiphilicity, and 4) synthesis and testing. Experimental evaluation of thirty-three peptides resulted in eleven active ACPs, four of which were non-hemolytic, with properties resembling those of the natural ACP lasioglossin III. These experiments show the first example of direct machine learning guided discovery of non-hemolytic ACPs.
Collapse
Affiliation(s)
- Elena Zakharova
- Department of ChemistryBiochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| | - Markus Orsi
- Department of ChemistryBiochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| | - Alice Capecchi
- Department of ChemistryBiochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| | - Jean‐Louis Reymond
- Department of ChemistryBiochemistry and Pharmaceutical SciencesUniversity of BernFreiestrasse 33012BernSwitzerland
| |
Collapse
|
30
|
Zhu L, Ye C, Hu X, Yang S, Zhu C. ACP-check: An anticancer peptide prediction model based on bidirectional long short-term memory and multi-features fusion strategy. Comput Biol Med 2022; 148:105868. [PMID: 35868046 DOI: 10.1016/j.compbiomed.2022.105868] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/14/2022] [Accepted: 07/09/2022] [Indexed: 11/16/2022]
Abstract
The anticancer peptide is an emerging anticancer drug that has become an effective alternative to chemotherapy and targeted therapy due to fewer side effects and resistance. The traditional biological experimental method for identifying anticancer peptides is a time-consuming and complicated process that hinders large-scale, rapid, and effective identification. In this paper, we propose a model based on a bidirectional long short-term memory network and multi-features fusion, called ACP-check, which employs a bidirectional long short-term memory network to extract time-dependent information features from peptide sequences, and combines them with amino acid sequence features including binary profile feature, dipeptide composition, the composition of k-spaced amino acid group pairs, amino acid composition, and sequence-order-coupling number. To verify the performance of the model, six benchmark datasets are selected, including ACPred-Fuse, ACPred-FL, ACP240, ACP740, main and alternate datasets of AntiCP2.0. In terms of Matthews correlation coefficients, ACP-check obtains 0.37, 0.82, 0.80, 0.75, 0.56, and 0.86 on six datasets respectively, which is an improvement by 2%-86% than existing state-of-the-art anticancer peptides prediction methods. Furthermore, ACP-check achieves prediction accuracy with 0.91, 0.91, 0.90, 0.87, 0.78, and 0.93 respectively, which increases range from 1%-49%. Overall, the comparison experiment shows that ACP-check can accurately identify anticancer peptides by sequence-level information. The code and data are available at http://www.cczubio.top/ACP-check/.
Collapse
Affiliation(s)
- Lun Zhu
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, 213164, China
| | - Chenyang Ye
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, 213164, China
| | - Xuemei Hu
- Key Laboratory of Symbol Computation and Knowledge Engineering of Ministry of Education, College of Computer Science and Technology, Jilin University, Changchun, 130012, China
| | - Sen Yang
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, 213164, China; Changzhou No.2 People's Hospital, the Affiliated Hospital of Nanjing Medical University, Changzhou, 213164, China.
| | - Chenyang Zhu
- School of Computer Science and Artificial Intelligence Aliyun School of Big Data School of Software, Changzhou University, Changzhou, 213164, China
| |
Collapse
|
31
|
Akbar S, Hayat M, Tahir M, Khan S, Alarfaj FK. cACP-DeepGram: Classification of anticancer peptides via deep neural network and skip-gram-based word embedding model. Artif Intell Med 2022; 131:102349. [DOI: 10.1016/j.artmed.2022.102349] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/24/2022] [Accepted: 07/04/2022] [Indexed: 12/28/2022]
|
32
|
Zou H, Yang F, Yin Z. Integrating multiple sequence features for identifying anticancer peptides. Comput Biol Chem 2022; 99:107711. [DOI: 10.1016/j.compbiolchem.2022.107711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 05/16/2022] [Accepted: 05/29/2022] [Indexed: 11/03/2022]
|
33
|
Li Y, Li X, Liu Y, Yao Y, Huang G. MPMABP: A CNN and Bi-LSTM-Based Method for Predicting Multi-Activities of Bioactive Peptides. Pharmaceuticals (Basel) 2022; 15:707. [PMID: 35745625 PMCID: PMC9231127 DOI: 10.3390/ph15060707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/23/2022] [Accepted: 05/30/2022] [Indexed: 12/30/2022] Open
Abstract
Bioactive peptides are typically small functional peptides with 2-20 amino acid residues and play versatile roles in metabolic and biological processes. Bioactive peptides are multi-functional, so it is vastly challenging to accurately detect all their functions simultaneously. We proposed a convolution neural network (CNN) and bi-directional long short-term memory (Bi-LSTM)-based deep learning method (called MPMABP) for recognizing multi-activities of bioactive peptides. The MPMABP stacked five CNNs at different scales, and used the residual network to preserve the information from loss. The empirical results showed that the MPMABP is superior to the state-of-the-art methods. Analysis on the distribution of amino acids indicated that the lysine preferred to appear in the anti-cancer peptide, the leucine in the anti-diabetic peptide, and the proline in the anti-hypertensive peptide. The method and analysis are beneficial to recognize multi-activities of bioactive peptides.
Collapse
Affiliation(s)
- You Li
- School of Electrical Engineering, Shaoyang University, Shaoyang 422000, China; (Y.L.); (X.L.)
| | - Xueyong Li
- School of Electrical Engineering, Shaoyang University, Shaoyang 422000, China; (Y.L.); (X.L.)
| | - Yuewu Liu
- College of Information and Intelligence, Hunan Agricultural University, Changsha 410128, China;
| | - Yuhua Yao
- School of Mathematics and Statistics, Hainan Normal University, Haikou 571158, China;
| | - Guohua Huang
- School of Electrical Engineering, Shaoyang University, Shaoyang 422000, China; (Y.L.); (X.L.)
| |
Collapse
|
34
|
Feng G, Yao H, Li C, Liu R, Huang R, Fan X, Ge R, Miao Q. ME-ACP: Multi-view neural networks with ensemble model for identification of anticancer peptides. Comput Biol Med 2022; 145:105459. [DOI: 10.1016/j.compbiomed.2022.105459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/22/2022] [Accepted: 03/24/2022] [Indexed: 12/26/2022]
|
35
|
To Assist Oncologists: An Efficient Machine Learning-Based Approach for Anti-Cancer Peptides Classification. SENSORS 2022; 22:s22114005. [PMID: 35684624 PMCID: PMC9185351 DOI: 10.3390/s22114005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/19/2022] [Accepted: 05/20/2022] [Indexed: 12/10/2022]
Abstract
In the modern technological era, Anti-cancer peptides (ACPs) have been considered a promising cancer treatment. It’s critical to find new ACPs to ensure a better knowledge of their functioning processes and vaccine development. Thus, timely and efficient ACPs using a computational technique are highly needed because of the enormous peptide sequences generated in the post-genomic era. Recently, numerous adaptive statistical algorithms have been developed for separating ACPs and NACPs. Despite great advancements, existing approaches still have insufficient feature descriptors and learning methods, limiting predictive performance. To address this, a trustworthy framework is developed for the precise identification of ACPs. Particularly, the presented approach incorporates four hypothetical feature encoding mechanisms namely: amino acid, dipeptide, tripeptide, and an improved version of pseudo amino acid composition are applied to indicate the motif of the target class. Moreover, principal component analysis (PCA) is employed for feature pruning, while selecting optimal, deep, and highly variated features. Due to the diverse nature of learning, experiments are performed over numerous algorithms to select the optimum operating method. After investigating the empirical outcomes, the support vector machine with hybrid feature space shows better performance. The proposed framework achieved an accuracy of 97.09% and 98.25% over the benchmark and independent datasets, respectively. The comparative analysis demonstrates that our proposed model outperforms as compared to the existing methods and is beneficial in drug development, and oncology.
Collapse
|
36
|
Multi-channel CNN based anticancer peptides identification. Anal Biochem 2022; 650:114707. [PMID: 35568159 DOI: 10.1016/j.ab.2022.114707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 01/27/2022] [Accepted: 04/27/2022] [Indexed: 11/20/2022]
Abstract
Cancer is one of the most dangerous diseases in the world that often leads to misery and death. Current treatments include different kinds of anticancer therapy which exhibit different types of side effects. Because of certain physicochemical properties, anticancer peptides (ACPs) have opened a new path of treatments for this deadly disease. That is why a well-performed methodology for identifying novel anticancer peptides has great importance in the fight against cancer. In addition to the laboratory techniques, various machine learning and deep learning methodologies have developed in recent years for this task. Although these models have shown reasonable predictive ability, there's still room for improvement in terms of performance and exploring new types of algorithms. In this work, we have proposed a novel multi-channel convolutional neural network (CNN) for identifying anticancer peptides from protein sequences. We have collected data from the existing state-of-the-art methodologies and applied binary encoding for data preprocessing. We have also employed k-fold cross-validation to train our models on benchmark datasets and compared our models' performance on the independent datasets. The comparison has indicated our models' superiority on various evaluation metrics. We think our work can be a valuable asset in finding novel anticancer peptides. We have provided a user-friendly web server for academic purposes and it is publicly available at: \texttt{http://103.99.176.239/iacp-cnn/}.
Collapse
|
37
|
Development of Anticancer Peptides Using Artificial Intelligence and Combinational Therapy for Cancer Therapeutics. Pharmaceutics 2022; 14:pharmaceutics14050997. [PMID: 35631583 PMCID: PMC9147327 DOI: 10.3390/pharmaceutics14050997] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 01/27/2023] Open
Abstract
Cancer is a group of diseases causing abnormal cell growth, altering the genome, and invading or spreading to other parts of the body. Among therapeutic peptide drugs, anticancer peptides (ACPs) have been considered to target and kill cancer cells because cancer cells have unique characteristics such as a high negative charge and abundance of microvilli in the cell membrane when compared to a normal cell. ACPs have several advantages, such as high specificity, cost-effectiveness, low immunogenicity, minimal toxicity, and high tolerance under normal physiological conditions. However, the development and identification of ACPs are time-consuming and expensive in traditional wet-lab-based approaches. Thus, the application of artificial intelligence on the approaches can save time and reduce the cost to identify candidate ACPs. Recently, machine learning (ML), deep learning (DL), and hybrid learning (ML combined DL) have emerged into the development of ACPs without experimental analysis, owing to advances in computer power and big data from the power system. Additionally, we suggest that combination therapy with classical approaches and ACPs might be one of the impactful approaches to increase the efficiency of cancer therapy.
Collapse
|
38
|
Wu X, Zeng W, Lin F, Xu P, Li X. Anticancer Peptide Prediction via Multi-Kernel CNN and Attention Model. Front Genet 2022; 13:887894. [PMID: 35571059 PMCID: PMC9092594 DOI: 10.3389/fgene.2022.887894] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 03/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Modern lifestyles mean that people are more likely to suffer from some form of cancer. As anticancer peptides can effectively kill cancer cells and play an important role in fighting cancer, they have been a subject of increasing research interest. Methods: This study presents a useful tool to identify the anticancer peptides based on a multi-kernel CNN and attention model, called ACP-MCAM. This model can automatically learn adaptive embedding and the context sequence features of ACP. In addition, to obtain better interpretability and integrity, we visualized the model. Results: Benchmarking comparison shows that ACP-MCAM significantly outperforms several state-of-the-art models. Different encoding schemes have different impacts on the performance of the model. We also studied tmethod parameter optimization. Conclusion: The ACP-MCAM can integrate multi-kernel CNN and self-attention mechanism, which outperforms the previous model in identifying anticancer peptides. It is expected that the work will provide new research ideas for anticancer peptide prediction in the future. In addition, this work will promote the development of the interdisciplinary field of artificial intelligence and biomedicine.
Collapse
Affiliation(s)
- Xiujin Wu
- School of Informatics, Xiamen University, Xiamen, China
| | - Wenhua Zeng
- School of Informatics, Xiamen University, Xiamen, China
| | - Fan Lin
- School of Informatics, Xiamen University, Xiamen, China
- Boston Children’s Hospital, Boston, MA, United States
| | - Peng Xu
- Chongqing Michong Technology Co., Ltd., Chongqing, China
| | | |
Collapse
|
39
|
Ramazi S, Mohammadi N, Allahverdi A, Khalili E, Abdolmaleki P. A review on antimicrobial peptides databases and the computational tools. Database (Oxford) 2022; 2022:baac011. [PMID: 35305010 PMCID: PMC9216472 DOI: 10.1093/database/baac011] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/04/2022] [Accepted: 02/28/2022] [Indexed: 12/29/2022]
Abstract
Antimicrobial Peptides (AMPs) have been considered as potential alternatives for infection therapeutics since antibiotic resistance has been raised as a global problem. The AMPs are a group of natural peptides that play a crucial role in the immune system in various organisms AMPs have features such as a short length and efficiency against microbes. Importantly, they have represented low toxicity in mammals which makes them potential candidates for peptide-based drugs. Nevertheless, the discovery of AMPs is accompanied by several issues which are associated with labour-intensive and time-consuming wet-lab experiments. During the last decades, numerous studies have been conducted on the investigation of AMPs, either natural or synthetic type, and relevant data are recently available in many databases. Through the advancement of computational methods, a great number of AMP data are obtained from publicly accessible databanks, which are valuable resources for mining patterns to design new models for AMP prediction. However, due to the current flaws in assessing computational methods, more interrogations are warranted for accurate evaluation/analysis. Considering the diversity of AMPs and newly reported ones, an improvement in Machine Learning algorithms are crucial. In this review, we aim to provide valuable information about different types of AMPs, their mechanism of action and a landscape of current databases and computational tools as resources to collect AMPs and beneficial tools for the prediction and design of a computational model for new active AMPs.
Collapse
Affiliation(s)
- Shahin Ramazi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, Tehran 14115-111, Iran
| | - Neda Mohammadi
- Department of Medical Biotechnology, Faculty of Allied Medical Sciences, Iran University of Medical Sciences, Hemmat Highway, Tehran 1449614535, Iran
- Institute of Pharmacology and Toxicology, University of Bonn, Biomedical Center, Venusberg Campus 1, Bonn 53127, Germany
| | - Abdollah Allahverdi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, Tehran 14115-111, Iran
| | - Elham Khalili
- Department of Plant Biology, Faculty of Biological Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, Tehran 14115-111, Iran
| | - Parviz Abdolmaleki
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Jalal Ale Ahmad Highway, Tehran 14115-111, Iran
| |
Collapse
|
40
|
Zou H, Yang F, Yin Z. iTTCA-MFF: identifying tumor T cell antigens based on multiple feature fusion. Immunogenetics 2022; 74:447-454. [PMID: 35246701 DOI: 10.1007/s00251-022-01258-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/26/2022] [Indexed: 11/05/2022]
Abstract
Cancer is a terrible disease, recent studies reported that tumor T cell antigens (TTCAs) may play a promising role in cancer treatment. Since experimental methods are still expensive and time-consuming, it is highly desirable to develop automatic computational methods to identify tumor T cell antigens from the huge amount of natural and synthetic peptides. Hence, in this study, a novel computational model called iTTCA-MFF was proposed to identify TTCAs. In order to describe the sequence effectively, the physicochemical (PC) properties of amino acid and residue pairwise energy content matrix (RECM) were firstly employed to encode peptide sequences. Then, two different approaches including covariance and Pearson's correlation coefficient (PCC) were used to collect discriminative information from PC and RECM matrixes. Next, an effective feature selection approach called the least absolute shrinkage and selection operator (LAASO) was adopted to select the optimal features. These selected optimal features were fed into support vector machine (SVM) for identifying TTCAs. We performed experiments on two different datasets, experimental results indicated that the proposed method is promising and may play a complementary role to the existing methods for identifying TTCAs. The datasets and codes can be available at https://figshare.com/articles/online_resource/iTTCA-MFF/17636120 .
Collapse
Affiliation(s)
- Hongliang Zou
- School of Communications and Electronics, Jiangxi Science and Technology Normal University, Nanchang, 330003, China.
| | - Fan Yang
- School of Communications and Electronics, Jiangxi Science and Technology Normal University, Nanchang, 330003, China
| | - Zhijian Yin
- School of Communications and Electronics, Jiangxi Science and Technology Normal University, Nanchang, 330003, China
| |
Collapse
|
41
|
Qi L, Gao X, Pan D, Sun Y, Cai Z, Xiong Y, Dang Y. Research progress in the screening and evaluation of umami peptides. Compr Rev Food Sci Food Saf 2022; 21:1462-1490. [PMID: 35201672 DOI: 10.1111/1541-4337.12916] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 12/22/2021] [Accepted: 01/03/2022] [Indexed: 12/22/2022]
Abstract
Umami is an important element affecting food taste, and the development of umami peptides is a topic of interest in food-flavoring research. The existing technology used for traditional screening of umami peptides is time-consuming and labor-intensive, making it difficult to meet the requirements of high-throughput screening, which limits the rapid development of umami peptides. The difficulty in performing a standard measurement of umami intensity is another problem that restricts the development of umami peptides. The existing methods are not sensitive and specific, making it difficult to achieve a standard evaluation of umami taste. This review summarizes the umami receptors and umami peptides, focusing on the problems restricting the development of umami peptides, high-throughput screening, and establishment of evaluation standards. The rapid screening of umami peptides was realized based on molecular docking technology and a machine learning method, and the standard evaluation of umami could be realized with a bionic taste sensor. The progress of rapid screening and evaluation methods significantly promotes the study of umami peptides and increases its application in the seasoning industry.
Collapse
Affiliation(s)
- Lulu Qi
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of AgroProducts, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Xinchang Gao
- Department of Chemistry, Tsinghua University, Beijing, China
| | - Daodong Pan
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of AgroProducts, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China.,National R&D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, Jiangxi, China
| | - Yangying Sun
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of AgroProducts, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Zhendong Cai
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of AgroProducts, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Yongzhao Xiong
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of AgroProducts, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| | - Yali Dang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of AgroProducts, College of Food and Pharmaceutical Sciences, Ningbo University, Ningbo, Zhejiang, China
| |
Collapse
|
42
|
Molecular dynamics simulations, docking and MMGBSA studies of newly designed peptide-conjugated glucosyloxy stilbene derivatives with tumor cell receptors. Mol Divers 2022; 26:2717-2743. [PMID: 35037187 DOI: 10.1007/s11030-021-10354-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 11/17/2021] [Indexed: 10/19/2022]
Abstract
In this work, for the first time, we designed derivatives of beta-D-glucosyloxy-3-hydroxy-trans-stiblene-2-carboxylic acid (GHS), by conjugating GHS with tumor targeting peptides RPARPAR and GGKRPAR to target over-expressed receptors in tumor cells. The sequences RPARPAR and GGKRPAR are known to target the neuropilin1 (NRP1) receptor due to the C-terminal Arg domain; however, their effectiveness has never been examined with other commonly over-expressed receptors in tumor cells, particularly of chronic lymphocytic leukemia that include integrin α1β1 and CD22. By conjugating these peptides with GHS, which is known for its inherent anti-cancer properties, the goal is to further enhance tumor cell targeting by developing compounds that can target multiple receptors. The physicochemical properties of the conjugates and individual peptides were analyzed using Turbomole and COSMOthermX20 in order to determine their hydrogen bond accepting and donating capabilities. The web server POCASA was used in order to determine the surface cavities and binding pockets of the three receptors. To explore the binding affinities, we conducted molecular docking studies with the peptides and the conjugates with each of the receptors. After molecular docking, the complexes were analyzed using Protein-Ligand Interaction Profiler to determine the types of interactions involved. Molecular dynamics simulation studies were conducted to explore the stability of the receptor-ligand complexes. Our results indicated that in most cases the conjugates showed higher binding and stability with the receptors. Additionally, highly stable complexes of conjugates were obtained with CD22, NRP1 and in most cases with the integrin α1β1 receptor as well. The binding energies were calculated for each of the receptor ligand complexes through trajectory analysis using MMGBSA studies. SwissADME studies revealed that the compounds showed low GI absorption and were not found to be CYP inhibitors and had bioavailability score that would allow them to be considered as potential drug candidates. Overall, our results for the first time show that the designed conjugates can target multiple over-expressed receptors in tumor cells and may be potentially developed as future therapeutics for targeting tumor cells.
Collapse
|
43
|
In Silico Studies of Tumor Targeted Peptide-Conjugated Natural Products for Targeting Over-Expressed Receptors in Breast Cancer Cells Using Molecular Docking, Molecular Dynamics and MMGBSA Calculations. APPLIED SCIENCES-BASEL 2022. [DOI: 10.3390/app12010515] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In this work, in silico studies were carried out for the design of diterpene and polyphenol-peptide conjugates to potentially target over-expressed breast tumor cell receptors. Four point mutations were induced into the known tumor-targeting peptide sequence YHWYGYTPQN at positions 1, 2, 8 and 10, resulting in four mutated peptides. Each peptide was separately conjugated with either chlorogenate, carnosate, gallate, or rosmarinate given their known anti-tumor activities, creating dual targeting compounds. Molecular docking studies were conducted with the epidermal growth factor receptor (EGFR), to which the original peptide sequence is known to bind, as well as the estrogen receptor (ERα) and peroxisome proliferator-activated receptor (PPARα) using both Autodock Vina and FireDock. Based on docking results, peptide conjugates and peptides were selected and subjected to molecular dynamics simulations. MMGBSA calculations were used to further probe the binding energies. ADME studies revealed that the compounds were not CYP substrates, though most were Pgp substrates. Additionally, most of the peptides and conjugates showed MDCK permeability. Our results indicated that several of the peptide conjugates enhanced binding interactions with the receptors and resulted in stable receptor-ligand complexes; Furthermore, they may successfully target ERα and PPARα in addition to EGFR and may be further explored for synthesis and biological studies for therapeutic applications.
Collapse
|
44
|
Arif M, Ahmed S, Ge F, Kabir M, Khan YD, Yu DJ, Thafar M. StackACPred: Prediction of anticancer peptides by integrating optimized multiple feature descriptors with stacked ensemble approach. CHEMOMETRICS AND INTELLIGENT LABORATORY SYSTEMS 2022; 220:104458. [DOI: 10.1016/j.chemolab.2021.104458] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
|
45
|
Wang H, Zhao J, Zhao H, Li H, Wang J. CL-ACP: a parallel combination of CNN and LSTM anticancer peptide recognition model. BMC Bioinformatics 2021; 22:512. [PMID: 34670488 PMCID: PMC8527680 DOI: 10.1186/s12859-021-04433-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/05/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Anticancer peptides are defence substances with innate immune functions that can selectively act on cancer cells without harming normal cells and many studies have been conducted to identify anticancer peptides. In this paper, we introduce the anticancer peptide secondary structures as additional features and propose an effective computational model, CL-ACP, that uses a combined network and attention mechanism to predict anticancer peptides. RESULTS The CL-ACP model uses secondary structures and original sequences of anticancer peptides to construct the feature space. The long short-term memory and convolutional neural network are used to extract the contextual dependence and local correlations of the feature space. Furthermore, a multi-head self-attention mechanism is used to strengthen the anticancer peptide sequences. Finally, three categories of feature information are classified by cascading. CL-ACP was validated using two types of datasets, anticancer peptide datasets and antimicrobial peptide datasets, on which it achieved good results compared to previous methods. CL-ACP achieved the highest AUC values of 0.935 and 0.972 on the anticancer peptide and antimicrobial peptide datasets, respectively. CONCLUSIONS CL-ACP can effectively recognize antimicrobial peptides, especially anticancer peptides, and the parallel combined neural network structure of CL-ACP does not require complex feature design and high time cost. It is suitable for application as a useful tool in antimicrobial peptide design.
Collapse
Affiliation(s)
- Huiqing Wang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030024, China
| | - Jian Zhao
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030024, China.
| | - Hong Zhao
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030024, China
| | - Haolin Li
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030024, China
| | - Juan Wang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, 030024, China
| |
Collapse
|
46
|
Lv Z, Cui F, Zou Q, Zhang L, Xu L. Anticancer peptides prediction with deep representation learning features. Brief Bioinform 2021; 22:bbab008. [PMID: 33529337 DOI: 10.1093/bib/bbab008] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/20/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022] Open
Abstract
Anticancer peptides constitute one of the most promising therapeutic agents for combating common human cancers. Using wet experiments to verify whether a peptide displays anticancer characteristics is time-consuming and costly. Hence, in this study, we proposed a computational method named identify anticancer peptides via deep representation learning features (iACP-DRLF) using light gradient boosting machine algorithm and deep representation learning features. Two kinds of sequence embedding technologies were used, namely soft symmetric alignment embedding and unified representation (UniRep) embedding, both of which involved deep neural network models based on long short-term memory networks and their derived networks. The results showed that the use of deep representation learning features greatly improved the capability of the models to discriminate anticancer peptides from other peptides. Also, UMAP (uniform manifold approximation and projection for dimension reduction) and SHAP (shapley additive explanations) analysis proved that UniRep have an advantage over other features for anticancer peptide identification. The python script and pretrained models could be downloaded from https://github.com/zhibinlv/iACP-DRLF or from http://public.aibiochem.net/iACP-DRLF/.
Collapse
Affiliation(s)
- Zhibin Lv
- University of Electronic Science and Technology of China
| | - Feifei Cui
- University of Electronic Science and Technology of China
| | - Quan Zou
- Institute of Fundamental and Frontier Sciences at University of Electronic Science and Technology of China
| | - Lichao Zhang
- School of Intelligent Manufacturing and Equipment, Shenzhen Institute of Information Technology, China
| | - Lei Xu
- School of Electronic and Communication Engineering, Shenzhen Polytechnic, China
| |
Collapse
|
47
|
Cao R, Wang M, Bin Y, Zheng C. DLFF-ACP: prediction of ACPs based on deep learning and multi-view features fusion. PeerJ 2021; 9:e11906. [PMID: 34414035 PMCID: PMC8344685 DOI: 10.7717/peerj.11906] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 07/14/2021] [Indexed: 01/10/2023] Open
Abstract
An emerging type of therapeutic agent, anticancer peptides (ACPs), has attracted attention because of its lower risk of toxic side effects. However process of identifying ACPs using experimental methods is both time-consuming and laborious. In this study, we developed a new and efficient algorithm that predicts ACPs by fusing multi-view features based on dual-channel deep neural network ensemble model. In the model, one channel used the convolutional neural network CNN to automatically extract the potential spatial features of a sequence. Another channel was used to process and extract more effective features from handcrafted features. Additionally, an effective feature fusion method was explored for the mutual fusion of different features. Finally, we adopted the neural network to predict ACPs based on the fusion features. The performance comparisons across the single and fusion features showed that the fusion of multi-view features could effectively improve the model's predictive ability. Among these, the fusion of the features extracted by the CNN and composition of k-spaced amino acid group pairs achieved the best performance. To further validate the performance of our model, we compared it with other existing methods using two independent test sets. The results showed that our model's area under curve was 0.90, which was higher than that of the other existing methods on the first test set and higher than most of the other existing methods on the second test set. The source code and datasets are available at https://github.com/wame-ng/DLFF-ACP.
Collapse
Affiliation(s)
- Ruifen Cao
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, School of Computer Science and Technology, Anhui University, Hefei, Anhui, China
- Engineering Research Center of Big Data Application in Private Health Medicine, Fujian Province University, Putian, Fujian, China
| | - Meng Wang
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, School of Computer Science and Technology, Anhui University, Hefei, Anhui, China
| | - Yannan Bin
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, School of Computer Science and Technology, Anhui University, Hefei, Anhui, China
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, China
| | - Chunhou Zheng
- Key Laboratory of Intelligent Computing and Signal Processing of Ministry of Education, School of Computer Science and Technology, Anhui University, Hefei, Anhui, China
- Engineering Research Center of Big Data Application in Private Health Medicine, Fujian Province University, Putian, Fujian, China
| |
Collapse
|
48
|
Chen J, Cheong HH, Siu SWI. xDeep-AcPEP: Deep Learning Method for Anticancer Peptide Activity Prediction Based on Convolutional Neural Network and Multitask Learning. J Chem Inf Model 2021; 61:3789-3803. [PMID: 34327990 DOI: 10.1021/acs.jcim.1c00181] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Cancer is one of the leading causes of death worldwide. Conventional cancer treatment relies on radiotherapy and chemotherapy, but both methods bring severe side effects to patients, as these therapies not only attack cancer cells but also damage normal cells. Anticancer peptides (ACPs) are a promising alternative as therapeutic agents that are efficient and selective against tumor cells. Here, we propose a deep learning method based on convolutional neural networks to predict biological activity (EC50, LC50, IC50, and LD50) against six tumor cells, including breast, colon, cervix, lung, skin, and prostate. We show that models derived with multitask learning achieve better performance than conventional single-task models. In repeated 5-fold cross validation using the CancerPPD data set, the best models with the applicability domain defined obtain an average mean squared error of 0.1758, Pearson's correlation coefficient of 0.8086, and Kendall's correlation coefficient of 0.6156. As a step toward model interpretability, we infer the contribution of each residue in the sequence to the predicted activity by means of feature importance weights derived from the convolutional layers of the model. The present method, referred to as xDeep-AcPEP, will help to identify effective ACPs in rational peptide design for therapeutic purposes. The data, script files for reproducing the experiments, and the final prediction models can be downloaded from http://github.com/chen709847237/xDeep-AcPEP. The web server to directly access this prediction method is at https://app.cbbio.online/acpep/home.
Collapse
Affiliation(s)
- Jiarui Chen
- Department of Computer and Information Science, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Hong Hin Cheong
- Department of Computer and Information Science, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China
| | - Shirley W I Siu
- Department of Computer and Information Science, University of Macau, Avenida da Universidade, Taipa, Macau 999078, China.,School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800 USM, Penang, Malaysia
| |
Collapse
|
49
|
He W, Wang Y, Cui L, Su R, Wei L. Learning embedding features based on multi-sense-scaled attention architecture to improve the predictive performance of anticancer peptides. Bioinformatics 2021; 37:4684-4693. [PMID: 34323948 DOI: 10.1093/bioinformatics/btab560] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 07/03/2021] [Accepted: 07/28/2021] [Indexed: 01/10/2023] Open
Abstract
MOTIVATION Anticancer peptides (ACPs) have recently emerged as effective anticancer drugs in cancer therapy. Machine-learning-based predictors have been developed to identify ACPs and achieve satisfactory performance. However, existing methods suffer from experience-based feature engineering, which not only restricts the representation ability of the models to a certain extent but also lacks adaptivity for different data, limiting the further improvement of the predictive performance and impacting the robustness of the predictive models. To alleviate the above problems, we propose a novel deep-learning-based predictor named ACPred-LAF, in which we propose a novel multi-sense and multi-scaled embedding algorithm to automatically learn and extract context sequential characteristics of ACPs. RESULTS Through the feature comparative analysis, we demonstrate that our learnable and self-adaptive embedding features are better than hand-crafted features in capturing discriminative information, which can effectively benefit the performance improvement for ACP prediction. In addition, benchmarking comparison results demonstrate that our ACPred-LAF outperforms the state-of-the-art methods both on existing benchmark datasets and our newly constructed dataset. Furthermore, we also prove and validate the robustness of the model via the data interference experiment. To avoid potential evaluation bias, here we construct a new ACP benchmark dataset named ACP-Mixed by integrating existing datasets. We expect our newly constructed dataset to be a golden standard benchmark dataset in this field. To facilitate the use of our model, we develop a web server as the implementation of ACPred-LAF. AVAILABILITY Our proposed ACPred-LAF, newly constructed benchmark dataset ACP-Mixed are open source collaborative initiatives available in the GitHub repository (https://github.com/TearsWaiting/ACPred-LAF). Besides, a webserver as the implementation of ACPred-LAF that can be accessed via: http://server.malab.cn/ACPred-LAF. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Wenjia He
- School of Software, Shandong University, Jinan, China.,Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China
| | - Yu Wang
- School of Software, Shandong University, Jinan, China.,Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China
| | - Lizhen Cui
- School of Software, Shandong University, Jinan, China.,Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China
| | - Ran Su
- College of Intelligence and Computing, Tianjin University, Tianjin, China
| | - Leyi Wei
- School of Software, Shandong University, Jinan, China.,Joint SDU-NTU Centre for Artificial Intelligence Research (C-FAIR), Shandong University, Jinan, China
| |
Collapse
|
50
|
Liang X, Li F, Chen J, Li J, Wu H, Li S, Song J, Liu Q. Large-scale comparative review and assessment of computational methods for anti-cancer peptide identification. Brief Bioinform 2021; 22:bbaa312. [PMID: 33316035 PMCID: PMC8294543 DOI: 10.1093/bib/bbaa312] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/30/2020] [Accepted: 08/25/2020] [Indexed: 12/13/2022] Open
Abstract
Anti-cancer peptides (ACPs) are known as potential therapeutics for cancer. Due to their unique ability to target cancer cells without affecting healthy cells directly, they have been extensively studied. Many peptide-based drugs are currently evaluated in the preclinical and clinical trials. Accurate identification of ACPs has received considerable attention in recent years; as such, a number of machine learning-based methods for in silico identification of ACPs have been developed. These methods promote the research on the mechanism of ACPs therapeutics against cancer to some extent. There is a vast difference in these methods in terms of their training/testing datasets, machine learning algorithms, feature encoding schemes, feature selection methods and evaluation strategies used. Therefore, it is desirable to summarize the advantages and disadvantages of the existing methods, provide useful insights and suggestions for the development and improvement of novel computational tools to characterize and identify ACPs. With this in mind, we firstly comprehensively investigate 16 state-of-the-art predictors for ACPs in terms of their core algorithms, feature encoding schemes, performance evaluation metrics and webserver/software usability. Then, comprehensive performance assessment is conducted to evaluate the robustness and scalability of the existing predictors using a well-prepared benchmark dataset. We provide potential strategies for the model performance improvement. Moreover, we propose a novel ensemble learning framework, termed ACPredStackL, for the accurate identification of ACPs. ACPredStackL is developed based on the stacking ensemble strategy combined with SVM, Naïve Bayesian, lightGBM and KNN. Empirical benchmarking experiments against the state-of-the-art methods demonstrate that ACPredStackL achieves a comparative performance for predicting ACPs. The webserver and source code of ACPredStackL is freely available at http://bigdata.biocie.cn/ACPredStackL/ and https://github.com/liangxiaoq/ACPredStackL, respectively.
Collapse
Affiliation(s)
- Xiao Liang
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China
- Shaanxi Key Laboratory of Agricultural Information Perception and Intelligent Service, Yangling, Shaanxi 712100, China
| | - Fuyi Li
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
- Monash Centre for Data Science, Monash University, Melbourne, VIC 3800, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Jinxiang Chen
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China
| | - Junlong Li
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China
| | - Hao Wu
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China
| | - Shuqin Li
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China
- Shaanxi Key Laboratory of Agricultural Information Perception and Intelligent Service, Yangling, Shaanxi 712100, China
| | - Jiangning Song
- Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
- Monash Centre for Data Science, Monash University, Melbourne, VIC 3800, Australia
- ARC Centre of Excellence in Advanced Molecular Imaging, Monash University, Melbourne, VIC 3800, Australia
| | - Quanzhong Liu
- College of Information Engineering, Northwest A&F University, Yangling, 712100, China
- Shaanxi Key Laboratory of Agricultural Information Perception and Intelligent Service, Yangling, Shaanxi 712100, China
| |
Collapse
|