1
|
Wang Y, Zhou S, Quan Y, Liu Y, Zhou B, Chen X, Ma Z, Zhou Y. Label-free spatiotemporal decoding of single-cell fate via acoustic driven 3D tomography. Mater Today Bio 2024; 28:101201. [PMID: 39221213 PMCID: PMC11364901 DOI: 10.1016/j.mtbio.2024.101201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 08/08/2024] [Accepted: 08/11/2024] [Indexed: 09/04/2024] Open
Abstract
Label-free three-dimensional imaging plays a crucial role in unraveling the complexities of cellular functions and interactions in biomedical research. Conventional single-cell optical tomography techniques offer affordability and the convenience of bypassing laborious cell labelling protocols. However, these methods are encumbered by restricted illumination scanning ranges on abaxial plane, resulting in the loss of intricate cellular imaging details. The ability to fully control cellular rotation across all angles has emerged as an optimal solution for capturing comprehensive structural details of cells. Here, we introduce a label-free, cost-effective, and readily fabricated contactless acoustic-induced vibration system, specifically designed to enable multi-degree-of-freedom rotation of cells, ultimately attaining stable in-situ rotation. Furthermore, by integrating this system with advanced deep learning technologies, we perform 3D reconstruction and morphological analysis on diverse cell types, thus validating groups of high-precision cell identification. Notably, long-term observation of cells reveals distinct features associated with drug-induced apoptosis in both cancerous and normal cells populations. This methodology, based on deep learning-enabled cell 3D reconstruction, charts a novel trajectory for groups of real-time cellular visualization, offering promising advancements in the realms of drug screening and post-single-cell analysis, thereby addressing potential clinical requisites.
Collapse
Affiliation(s)
- Yuxin Wang
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Shizheng Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Yue Quan
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Yu Liu
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Bingpu Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Xiuping Chen
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| | - Zhichao Ma
- Institute of Medical Robotics, School of Biomedical Engineering, Shanghai Jiao Tong University, No.800 Dongchuan Road, Shanghai, 200240, China
| | - Yinning Zhou
- Joint Key Laboratory of the Ministry of Education, Institute of Applied Physics and Materials Engineering, University of Macau, Avenida da Universidade, Taipa, Macau, 999078, China
| |
Collapse
|
2
|
Tanwar D, Kaur T, Sudheendranath A, Kumar U, Sharma D. Pd(II) complexes bearing NNS pincer ligands: unveiling potent cytotoxicity against breast and pancreatic cancer. Dalton Trans 2024; 53:9798-9811. [PMID: 38787690 DOI: 10.1039/d4dt00282b] [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: 05/26/2024]
Abstract
The continuously increasing rate of breast cancer is one of the major threats to female health worldwide. Recently, palladium complexes have emerged as impressive candidates with effective biocompatibility and anticancer activities. Hence, in the present study, we report a new series of palladium complexes bearing NNS pincer ligands for cytotoxicity studies. The reaction of thiophenol/4-chlorothiophenol/4-methylthiophenol/4-methoxythiophenol with 2-bromo-N-quinolin-8-yl-acetamide in the presence of sodium hydroxide in ethanol at 80 °C gave [C9H6N-NH-C(O)-CH2-S-Ar] [Ar = C6H5 (L1), C6H4Cl-4 (L2), C6H4Me-4 (L3), and C6H4-OMe-4 (L4)]. The corresponding reaction of L1-L4 with Na2PdCl4 in methanol at room temperature for 3 h resulted in complexes [(L1-H)PdCl] (C1), [(L2-H)PdCl] (C2), [(L3-H)PdCl] (C3), and [(L4-H)PdCl] (C4). All new compounds have been characterized by spectroscopic analyses. The structures of complexes C1, C3, and C4 have also been determined from single-crystal X-ray diffraction data. The cytotoxicities of L1-L4 and C1-C4 have been investigated for breast cancer 4T1 and pancreatic cancer MIA-PaCa-2 cells. The IC50 values for complexes C2 and C3 were observed to be comparable to or higher than those of cisplatin. The stressed morphology and cell death of cancerous cells were successfully observed through cellular morphology analysis and the assessment of cytoskeleton damage.
Collapse
Affiliation(s)
- Deepika Tanwar
- Catalysis and Bioinorganic Research Lab, Department of Chemistry, Deshbandhu College, University of Delhi, New Delhi-110019, India.
- Department of Chemistry, University of Delhi, New Delhi-110007, India
| | - Tashmeen Kaur
- Institute of Nano Science and Technology, Knowledge City, Mohali, Punjab-140306, India.
| | - Athul Sudheendranath
- Department of Chemistry, Indian Institute of Technology, New Delhi-110016, India
| | - Umesh Kumar
- Catalysis and Bioinorganic Research Lab, Department of Chemistry, Deshbandhu College, University of Delhi, New Delhi-110019, India.
| | - Deepika Sharma
- Institute of Nano Science and Technology, Knowledge City, Mohali, Punjab-140306, India.
| |
Collapse
|
3
|
Zaki M, Elallam O, Jami O, EL Ghoubali D, Jhilal F, Alidrissi N, Ghazal H, Habib N, Abbad F, Benmoussa A, Bakkali F. Advancing Tumor Cell Classification and Segmentation in Ki-67 Images: A Systematic Review of Deep Learning Approaches. LECTURE NOTES IN NETWORKS AND SYSTEMS 2024:94-112. [DOI: 10.1007/978-3-031-52385-4_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
4
|
Lv Z, Cao X, Jin X, Xu S, Deng H. High-accuracy morphological identification of bone marrow cells using deep learning-based Morphogo system. Sci Rep 2023; 13:13364. [PMID: 37591969 PMCID: PMC10435561 DOI: 10.1038/s41598-023-40424-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 08/10/2023] [Indexed: 08/19/2023] Open
Abstract
Accurate identification and classification of bone marrow (BM) nucleated cell morphology are crucial for the diagnosis of hematological diseases. However, the subjective and time-consuming nature of manual identification by pathologists hinders prompt diagnosis and patient treatment. To address this issue, we developed Morphogo, a convolutional neural network-based system for morphological examination. Morphogo was trained using a vast dataset of over 2.8 million BM nucleated cell images. Its performance was evaluated using 508 BM cases that were categorized into five groups based on the degree of morphological abnormalities, comprising a total of 385,207 BM nucleated cells. The results demonstrated Morphogo's ability to identify over 25 different types of BM nucleated cells, achieving a sensitivity of 80.95%, specificity of 99.48%, positive predictive value of 76.49%, negative predictive value of 99.44%, and an overall accuracy of 99.01%. In most groups, Morphogo cell analysis and Pathologists' proofreading showed high intragroup correlation coefficients for granulocytes, erythrocytes, lymphocytes, monocytes, and plasma cells. These findings further validate the practical applicability of the Morphogo system in clinical practice and emphasize its value in assisting pathologists in diagnosing blood disorders.
Collapse
Affiliation(s)
- Zhanwu Lv
- Bone Marrow Chamber, Guangzhou Kingmed Diagnostic Laboratory Group Co., Ltd., Guangzhou, 510330, China.
| | - Xinyi Cao
- Division of Medical Technology Development, Hangzhou Zhiwei Information Technology Co., Ltd., Hangzhou, 310000, China
| | - Xinyi Jin
- Division of Medical Technology Development, Hangzhou Zhiwei Information Technology Co., Ltd., Hangzhou, 310000, China
| | - Shuangqing Xu
- Bone Marrow Chamber, Guangzhou Kingmed Diagnostic Laboratory Group Co., Ltd., Guangzhou, 510330, China
| | - Huangling Deng
- Bone Marrow Chamber, Guangzhou Kingmed Diagnostic Laboratory Group Co., Ltd., Guangzhou, 510330, China
| |
Collapse
|
5
|
Wang Y, Zhang W, Yip H, Qu C, Hu H, Chen X, Lee T, Yang X, Yang B, Kumar P, Lee SY, Casimiro JJ, Zhang J, Wang A, Lam KS. SIC50: Determining drug inhibitory concentrations using a vision transformer and an optimized Sobel operator. PATTERNS (NEW YORK, N.Y.) 2023; 4:100686. [PMID: 36873901 PMCID: PMC9982297 DOI: 10.1016/j.patter.2023.100686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/28/2022] [Accepted: 01/10/2023] [Indexed: 02/05/2023]
Abstract
As a measure of cytotoxic potency, half-maximal inhibitory concentration (IC50) is the concentration at which a drug exerts half of its maximal inhibitory effect against target cells. It can be determined by various methods that require applying additional reagents or lysing the cells. Here, we describe a label-free Sobel-edge-based method, which we name SIC50, for the evaluation of IC50. SIC50 classifies preprocessed phase-contrast images with a state-of-the-art vision transformer and allows for the continuous assessment of IC50 in a faster and more cost-efficient manner. We have validated this method using four drugs and 1,536-well plates and also built a web application. We anticipate that this method will assist in the high-throughput screening of chemical libraries (e.g., small-molecule drugs, small interfering RNA [siRNA], and microRNA and drug discovery).
Collapse
Affiliation(s)
- Yongheng Wang
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
| | - Weidi Zhang
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Hoyin Yip
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA 95817, USA
| | | | - Hongru Hu
- Integrative Genetics and Genomics, University of California, Davis, Davis, CA 95616, USA
| | - Xiaotie Chen
- Department of Mathematics, University of California, Davis, Davis, CA 95616, USA
| | - Teresa Lee
- Department of Biomedical Engineering, Yale University, New Haven, CT 06511, USA
| | - Xi Yang
- Intel, Santa Clara, CA 95054, USA
| | - Bingjun Yang
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Priyadarsini Kumar
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children Northern California, UC Davis School of Medicine, Sacramento, CA 96817, USA
| | - Su Yeon Lee
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Javier J. Casimiro
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA 95817, USA
| | - Jiawei Zhang
- Department of Computer Science, IFM Lab, University of California, Davis, Davis, CA 95616, USA
| | - Aijun Wang
- Department of Biomedical Engineering, University of California, Davis, Davis, CA 95616, USA
- Center for Surgical Bioengineering, Department of Surgery, University of California, Davis, School of Medicine, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospital for Children Northern California, UC Davis School of Medicine, Sacramento, CA 96817, USA
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, UC Davis NCI-designated Comprehensive Cancer Center, University of California, Davis, Sacramento, CA 95817, USA
- Division of Hematology and Oncology, Department of Internal Medicine, School of Medicine, University of California, Davis, Sacramento, CA 95817, USA
| |
Collapse
|
6
|
Tanwar D, Kaur T, Kumar R, Ahluwalia D, Sharma D, Kumar U. Nickel Complexes Bearing ONS Chelating Ligands: A Promising Contender for In Vitro Cytotoxicity Effects on Human Pancreatic Cancer MIA-PaCa-2 Cells. ACS APPLIED BIO MATERIALS 2023; 6:134-145. [PMID: 36599051 DOI: 10.1021/acsabm.2c00787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The highly chronic human pancreatic cancer cell is one of the major reasons for cancerous death. Nickel complexes are recently gaining interest in anticancer activities on different types of cancer cells. Hence, in this study, we synthesized and characterized a series of ONS donor ligands [2-HO-C6H4-CH═N-(C6H4)-SH] (L1), [2-OH-3-OMe-C6H3-CH═N-(C6H4)-SH] (L2), [2-OH-3,5-(C(Me)3)2-C6H2-CH═N-(C6H4)-SH] (L3), [2-OH-C6H4-CH═N-(C6H4)-SMe] (L4), [2-OH-3-OMe-C6H3-CH═N-(C6H4)-SMe] (L5), [2-OH-3,5-(C(Me)3)2-C6H2-CH═N-(C6H4)-SMe] (L6) and their Ni(II) metal complexes [(MeOH)Ni(L1-L1-4H)] (1), [(MeOH)Ni(L2-L2-4H)] (2), [(MeOH)Ni(L3-L3-4H)] (3), [(L4-H)2Ni] (4), [(L5-H)2Ni] (5), and [(L6-H)2Ni] (6). The single-crystal X-ray diffraction data of complexes 1 and 4 were collected to elucidate the geometry around the metal center. The anticancer activity of complexes 1-6 was investigated on human pancreatic cancer cell line MIA-PaCa-2, which revealed that complexes 4 and 6 were the most significantly effective in decreasing the cell viability of cancer cells at the lowest dose. The structure parameters obtained from single-crystal X-ray diffraction data are found to be in good agreement with the data from density functional theory and Hirshfeld surface analysis for complex 1.
Collapse
Affiliation(s)
- Deepika Tanwar
- Catalysis and Bioinorganic Research Lab, Department of Chemistry, Deshbandhu College, University of Delhi, New Delhi110019, India.,Department of Chemistry, University of Delhi, New Delhi110007, India
| | - Tashmeen Kaur
- Institute of Nano Science and Technology, Knowledge City, Mohali, Punjab140306, India
| | - Robin Kumar
- Catalysis and Bioinorganic Research Lab, Department of Chemistry, Deshbandhu College, University of Delhi, New Delhi110019, India
| | - Deepali Ahluwalia
- Department of Applied Chemistry, Delhi Technological University, New Delhi110042, India
| | - Deepika Sharma
- Institute of Nano Science and Technology, Knowledge City, Mohali, Punjab140306, India
| | - Umesh Kumar
- Catalysis and Bioinorganic Research Lab, Department of Chemistry, Deshbandhu College, University of Delhi, New Delhi110019, India
| |
Collapse
|
7
|
Zhao Y, Zhang J, Hu D, Qu H, Tian Y, Cui X. Application of Deep Learning in Histopathology Images of Breast Cancer: A Review. MICROMACHINES 2022; 13:2197. [PMID: 36557496 PMCID: PMC9781697 DOI: 10.3390/mi13122197] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/04/2022] [Accepted: 12/09/2022] [Indexed: 06/17/2023]
Abstract
With the development of artificial intelligence technology and computer hardware functions, deep learning algorithms have become a powerful auxiliary tool for medical image analysis. This study was an attempt to use statistical methods to analyze studies related to the detection, segmentation, and classification of breast cancer in pathological images. After an analysis of 107 articles on the application of deep learning to pathological images of breast cancer, this study is divided into three directions based on the types of results they report: detection, segmentation, and classification. We introduced and analyzed models that performed well in these three directions and summarized the related work from recent years. Based on the results obtained, the significant ability of deep learning in the application of breast cancer pathological images can be recognized. Furthermore, in the classification and detection of pathological images of breast cancer, the accuracy of deep learning algorithms has surpassed that of pathologists in certain circumstances. Our study provides a comprehensive review of the development of breast cancer pathological imaging-related research and provides reliable recommendations for the structure of deep learning network models in different application scenarios.
Collapse
Affiliation(s)
- Yue Zhao
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Shenyang 110169, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry, Northeastern University, Shenyang 110169, China
| | - Jie Zhang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
| | - Dayu Hu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
| | - Hui Qu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
| | - Ye Tian
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
| | - Xiaoyu Cui
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang 110169, China
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Shenyang 110169, China
- Key Laboratory of Data Analytics and Optimization for Smart Industry, Northeastern University, Shenyang 110169, China
| |
Collapse
|
8
|
Qin Q, Wang T, Xu Z, Liu S, Zhang H, Du Z, Wang J, Wang Y, Wang Z, Yuan S, Wu J, He W, Wang C, Yan X, Wang Y, Jiang X. Ectoderm-derived frontal bone mesenchymal stem cells promote traumatic brain injury recovery by alleviating neuroinflammation and glutamate excitotoxicity partially via FGF1. Stem Cell Res Ther 2022; 13:341. [PMID: 35883153 PMCID: PMC9327213 DOI: 10.1186/s13287-022-03032-6] [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: 12/22/2021] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Background Traumatic brain injury (TBI) leads to cell and tissue impairment, as well as functional deficits. Stem cells promote structural and functional recovery and thus are considered as a promising therapy for various nerve injuries. Here, we aimed to investigate the role of ectoderm-derived frontal bone mesenchymal stem cells (FbMSCs) in promoting cerebral repair and functional recovery in a murine TBI model. Methods A murine TBI model was established by injuring C57BL/6 N mice with moderate-controlled cortical impact to evaluate the extent of brain damage and behavioral deficits. Ectoderm-derived FbMSCs were isolated from the frontal bone and their characteristics were assessed using multiple differentiation assays, flow cytometry and microarray analysis. Brain repairment and functional recovery were analyzed at different days post-injury with or without FbMSC application. Behavioral tests were performed to assess learning and memory improvements. RNA sequencing analysis, immunofluorescence staining, and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) were used to examine inflammation reaction and neural regeneration. In vitro co-culture analysis and quantification of glutamate transportation were carried out to explore the possible mechanism of neurogenesis and functional recovery promoted by FbMSCs. Results Ectoderm-derived FbMSCs showed fibroblast like morphology and osteogenic differentiation capacity. FbMSCs were CD105, CD29 positive and CD45, CD31 negative. Different from mesoderm-derived MSCs, FbMSCs expressed the ectoderm-specific transcription factor Tfap2β. TBI mice showed impaired learning and memory deficits. Microglia and astrocyte activation, as well as neural damage, were significantly increased post-injury. FbMSC application ameliorated the behavioral deficits of TBI mice and promoted neural regeneration. RNA sequencing analysis showed that signal pathways related to inflammation decreased, whereas those related to neural activation increased. Immunofluorescence staining and qRT-PCR data revealed that microglial activation and astrocyte polarization to the A1 phenotype were suppressed by FbMSC application. In addition, FGF1 secreted from FbMSCs enhanced glutamate transportation by astrocytes and alleviated the cytotoxic effect of excessive glutamate on neurons. Conclusions Ectoderm-derived FbMSC application significantly alleviated neuroinflammation, brain injury, and excitatory toxicity to neurons, improved cognition and behavioral deficits in TBI mice. Therefore, ectoderm-derived FbMSCs could be ideal therapeutic candidates for TBI which mostly affect cells from the same embryonic origins as FbMSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03032-6.
Collapse
Affiliation(s)
- Qiaozhen Qin
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China.,Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, People's Republic of China
| | - Ting Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhenhua Xu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Shuirong Liu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Heyang Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhangzhen Du
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Jianing Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Yadi Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhenning Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Shanshan Yuan
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Jiamei Wu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Wenyan He
- China National Clinical Research Center for Neurological Diseases, Jing-Jin Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Changzhen Wang
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Xinlong Yan
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, People's Republic of China.
| | - Yan Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China. .,Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| | - Xiaoxia Jiang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China. .,Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| |
Collapse
|
9
|
Numerical learning of deep features from drug-exposed cell images to calculate IC50 without staining. Sci Rep 2022; 12:6610. [PMID: 35459284 PMCID: PMC9033873 DOI: 10.1038/s41598-022-10643-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 04/07/2022] [Indexed: 11/25/2022] Open
Abstract
To facilitate rapid determination of cellular viability caused by the inhibitory effect of drugs, numerical deep learning algorithms was used for unlabeled cell culture images captured by a light microscope as input. In this study, A549, HEK293, and NCI-H1975 cells were cultured, each of which have different molecular shapes and levels of drug responsiveness to doxorubicin (DOX). The microscopic images of these cells following exposure to various concentrations of DOX were trained with the measured value of cell viability using a colorimetric cell proliferation assay. Convolutional neural network (CNN) models for the study cells were constructed using augmented image data; the predicted cell viability using CNN models was compared to the cell viability measured by colorimetric assay. The linear relationship coefficient (r2) between measured and predicted cell viability was determined as 0.94–0.95 for the three cell types. In addition, the measured and predicted IC50 values were not statistically different. When drug responsiveness was estimated using allogenic models that were trained with a different cell type, the correlation coefficient decreased to 0.004085–0.8643. Our models could be applied to label-free cells to conduct rapid and large-scale tests while minimizing cost and labor, such as high-throughput screening for drug responsiveness.
Collapse
|
10
|
Varon E, Blumrosen G, Shefi O. A predictive model for personalization of nanotechnology-based phototherapy in cancer treatment. Front Oncol 2022; 12:1037419. [PMID: 36911792 PMCID: PMC9999042 DOI: 10.3389/fonc.2022.1037419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 11/21/2022] [Indexed: 01/06/2023] Open
Abstract
A major challenge in radiation oncology is the prediction and optimization of clinical responses in a personalized manner. Recently, nanotechnology-based cancer treatments are being combined with photodynamic therapy (PDT) and photothermal therapy (PTT). Predictive models based on machine learning techniques can be used to optimize the clinical setup configuration, including such parameters as laser radiation intensity, treatment duration, and nanoparticle features. In this article we demonstrate a methodology that can be used to identify the optimal treatment parameters for PDT and PTT by collecting data from in vitro cytotoxicity assay of PDT/PTT-induced cell death using a single nanocomplex. We construct three machine learning prediction models, employing regression, interpolation, and low- degree analytical function fitting, to predict the laser radiation intensity and duration settings that maximize the treatment efficiency. To examine the accuracy of these prediction models, we construct a dedicated dataset for PDT, PTT, and a combined treatment; this dataset is based on cell death measurements after light radiation treatment and is divided into training and test sets. The preliminary results show that the performance of all three models is sufficient, with death rate errors of 0.09, 0.15, and 0.12 for the regression, interpolation, and analytical function fitting approaches, respectively. Nevertheless, due to its simple form, the analytical function method has an advantage in clinical application and can be used for further analysis of the sensitivity of performance to the treatment parameters. Overall, the results of this study form a baseline for a future personalized prediction model based on machine learning in the domain of combined nanotechnology- and phototherapy-based cancer treatment.
Collapse
Affiliation(s)
- Eli Varon
- Faculty of Engineering, Bar-Ilan University, Ramat Gan, Israel.,Bar-Ilan Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel
| | - Gaddi Blumrosen
- Department of Digital Medical Technologies, Holon Institute of Technology, Holon, Israel.,Department of Computer Science, Holon Institute of Technology, Holon, Israel
| | - Orit Shefi
- Faculty of Engineering, Bar-Ilan University, Ramat Gan, Israel.,Bar-Ilan Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan, Israel.,Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|