GET THE APP

Deep Cancer Map: A Flexible Profound Learning Stage for Target and Cell-Based Anticancer Medication Disclosure
..

Research and Reports in Medical Sciences

ISSN: 2952-8127

Open Access

Mini Review - (2023) Volume 7, Issue 3

Deep Cancer Map: A Flexible Profound Learning Stage for Target and Cell-Based Anticancer Medication Disclosure

Ling Wang*
*Correspondence: Ling Wang, Department of Biology and Biological Engineering, South China University of Technology, Guangzhou, China, Email:
Department of Biology and Biological Engineering, South China University of Technology, Guangzhou, China

Received: 01-May-2023, Manuscript No. rrms-23-102571; Editor assigned: 03-May-2023, Pre QC No. P-102571; Reviewed: 16-May-2023, QC No. Q-102571; Revised: 22-May-2023, Manuscript No. R-102571; Published: 30-May-2023 , DOI: 10.37421/2952-8127.2023.7.111
Citation: Wang, Ling. “Deep Cancer Map: A Flexible Profound Learning Stage for Target and Cell-Based Anticancer Medication Disclosure.” Res Rep Med Sci 7 (2023): 111.
Copyright: © 2023 Wang L. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Abstract

Finding new anticancer medications has been generally concerned and stays an open test. Both phenotypic-based experimental screening and target-based experimental screening are common approaches to the discovery of anticancer drugs. Both of these approaches are time-consuming, labor-intensive, and expensive. In this review, we gathered 485,900 mixtures including in 3,919,974 bioactivity records against 426 anticancer targets and 346 disease cell lines from scholastic writing, as well as 60 growth cell lines from NCI-60 board. The FP-GNN deep learning method was then used to create a total of 832 classification models, including 426 target- and 406 cell-based predictive models, to predict the inhibitory activity of compounds against targets and tumor cell lines. The FP-GNN models outperform conventional machine learning and deep learning in terms of overall predictive performance, achieving the highest AUC values of 0.91, 0.88, and 0.91 for the test sets of targets, academia-sourced cancer cell lines, and NCI-60 cancer cell lines, respectively. Based on these high-quality models, the user-friendly webserver Deep Cancer Map and its local version made it possible for users to perform anticancer drug discovery tasks like large-scale virtual screening, profiling prediction of anticancer agents, target fishing, and drug repositioning. We guess this stage to speed up the disclosure of anticancer medications in the field.

Keywords

Deep cancer map • Profound learning • Target-based • Cell-based • Anticancer medication

Introduction

Cancer is a serious issue affecting human health that affects people all over the world. Chemotherapeutics, perhaps of the most generally involved methodology in disease treatment, mean to obstruct the quickly and confused isolating cancer cells out of ordinary components of development concealment control. However, numerous anticancer medications have unfavorable therapeutic effects and a variety of typical flaws. For a certain something, the low explicitness of existing chemotherapeutic medications for disease cells prompts killing ordinary cells, particularly those in the quickly multiplying stage, bringing about serious unfriendly impacts. Another factor that hinders the clinical efficacy and applications of conventional chemotherapeutics is the tumor cells' resistance to them. For instance, one normal system for disease cells to get obstruction is through the dynamic efflux capability of ATP-restricting tape carrier proteins, which can make a few medications be insufficient, for example, Vincristine and Adriamycin. Likewise, the ATP-autonomous habits, like chromosomal qualities transformation and changed drug pervasion, have additionally been accounted for as of late. Likewise, there is a dire need to distinguish novel anticancer medications to conquer these inadequacies [1,2].

Literature Review

The explosive growth of biological data has had a promising impact on drug discovery since the 1990s, when high-throughput screening and combinatorial chemistry made significant progress. For instance, CHEMBL is a massive, open-access, manually curated database that collects bioactivity and medicinal chemistry data. It contains over 5.4 million bioactivity data. The database, which will be released on January 26, 2023, currently contains more than 20 million compound-target pairs derived from over 2.3 million compounds tested against over 15,000 targets [3]. Drug Bank and Scifinder are two similar databases for medicinal chemistry that collect drug chemical structures, pharmacological actions, acting protein targets, and physiological pathways. The computationalbased approaches to drug development began to demonstrate potential research value and application directions on the basis of the rapidly expanding and aggregated data.

Discussion

Target-based drug discovery and phenotypic-based drug discovery are the two most common approaches to anticancer drug discovery in conventional research channels. In recent decades, TDD has become the standard method in the pharmaceutical industry due to its advantages of high screening capacity, relatively low cost, and ease of achieving efficient structure-activity relationship for subsequent lead optimization. Nonetheless, a significant deficiency of TDD is that ID and approval of druggable anticancer targets is very troublesome, as it requires evidence that medication competitors following up on potential objective have distinct clinical helpful impacts in malignant growth therapy, without unsatisfactory secondary effects, which is almost unimaginable for execution in the beginning phase of TDD.

This lethal slack in target approval has been as of late recognized as one of the potential explanations behind the low probability of clinical interpretation. PDD is a unique yet huge technique that has restored interest as of late, permitting direct perception of atoms balancing the way of behaving of organic frameworks. PDD is eluded to an option in contrast to TDD because of its higher clinicaltranslatability. For instance, the cancer cell-based screening model, which is a typical biological system, has been used to discover numerous anticancer drugs with success. Be that as it may, the principal trouble of PDD is the way to fish the likely focuses for a given medication competitor rapidly. In spite of the fact that there are numerous exploratory and computational techniques for target fishing, obviously, finding possible focuses for a given medication from numerous proteins in the human body is difficult. PDD and TDD can clearly work together to discover new anticancer drugs.

Machine learning and deep learning algorithms are used to develop predictive and/or generative models for the discovery of new anticancer agents based on a variety of molecular representations, such as molecular descriptors, fingerprints, and graphs, as the amount of target- and phenotypic-based screening data increases in the field of anticancer drug discovery. For instance, in the parts of using objective based screening information, Chen et al. utilizing the support vector machine method, a nanomolar dual inhibitor of FGFR4 and EGFR was discovered. Yang and co. In addition, ML and DL strategies have been utilized for the inhibitory movement expectation of different kinases inhibitors and profiling forecast of kinase inhibitors in the field of anticancer medication revelation [4].

The most common phenotypical-based screening method for determining a compound's inhibitory activity against tumor cell lines has been cell-based screening, which has made it much easier to find new anticancer drugs. Luo et al. developed naive Bayesian based predictive models for predicting anticancer molecules' cellular inhibitory activity, and they later discovered novel and diverse anticancer agents with significant inhibitory activity against a variety of tumor cell lines. He et al. recently reported ML models with interpretability for the precise prediction of active molecules that would be effective against breast cancer cells. In addition, it has been reported that DDR1, AKT, and CDK9 kinase inhibitors have been discovered using DL-based molecular generative models for the treatment of cancer. These models have all been used to find new active molecules that fight cancer in real-world situations, proving that ML and DL methods are important for speeding up drug discovery for cancer [5,6].

Conclusion

It has been a formidable challenge to design and discover new cancer drugs. Deep Cancer Map, the first comprehensive platform based on high-quality deep learning models for anticancer drug discovery, was introduced in this study as a versatile and user-friendly webserver. There are currently 832 robust and accurate predictive models in Deep Cancer Map that have been trained using carefully selected modeling datasets from target- and cell-based pharmacological screening data.

Acknowledgement

None.

Conflict of Interest

None.

References

  1. Miller, Kimberly D., Rebecca L. Siegel, Chun Chieh Lin and Angela B. Mariotto, et al. "Cancer treatment and survivorship statistics, 2016." Cancer J Clin 66 (2016): 271-289.
  2. Google Scholar, Crossref, Indexed at

  3. Chen, Wanqing, Rongshou Zheng, Peter D. Baade and Siwei Zhang, et al. "Cancer statistics in China, 2015." Cancer J Clin 66 (2016): 115-132.
  4. Google Scholar, Crossref, Indexed at

  5. Chae, Young Kwang, Alan P. Pan, Andrew A. Davis and Sandip P. Patel, et al. "Path toward precision oncology: Review of targeted therapy studies and tools to aid in defining “actionability” of a molecular lesion and patient management support." Mol Cancer Ther 16 (2017): 2645-2655.
  6. Google Scholar, Crossref, Indexed at

  7. Patel, Sandip Kumar, Bhawana George and Vineeta Rai. "Artificial intelligence to decode cancer mechanism: Beyond patient stratification for precision oncology." Front Pharmacol 11 (2020): 1177.
  8. Google Scholar, Crossref, Indexed at

  9. Huang, Shigao, Jie Yang, Simon Fong and Qi Zhao. "Artificial intelligence in cancer diagnosis and prognosis: Opportunities and challenges." Cancer Lett 471 (2020): 61-71.
  10. Google Scholar, Crossref, Indexed at

  11. Leslie P. L., Franklin D. A., Liu Y. and Zhang Y. “p53 regulates the expression of LRP1 and apoptosis through a stress intensity-dependent microRNA feedback loop.” Cell Rep 6 (2018): 1484–1495.
  12. Google Scholar, Crossref, Indexed at

arrow_upward arrow_upward