1
|
Singh A, Georgy JT, Dhananjayan S, Sigamani E, John AO, Joel A, Chandramohan J, Abarna R, Rebekah G, Backianathan S, Abraham DT, Paul MJ, Chacko RT, Manipadam MT, Pai R. Comparative analysis of mutational patterns in triple negative breast cancer before and after neoadjuvant chemotherapy in patients with residual disease. Gene 2024; 895:147980. [PMID: 37951371 DOI: 10.1016/j.gene.2023.147980] [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: 06/07/2023] [Revised: 09/13/2023] [Accepted: 11/08/2023] [Indexed: 11/14/2023]
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease with poor survival compared to other subtypes. Patients with residual disease after neoadjuvant chemotherapy (NAC) face an increased risk of relapse and death. We aimed to characterize the mutational landscape of this subset to offer insights into relapse pathogenesis and potential therapeutic targets. We retrospectively analyzed archived paired (pre- and post-NAC) tumor samples from 25 patients with TNBC with residual disease using a targeted 72-gene next-generation sequencing panel. Our findings revealed a stable mutational burden in both pre- and post-NAC samples, with a median count of 12 variants (IQR 7-17.25) per sample. TP53, PMS2, PTEN, ERBB2, and NOTCH1 variants were observed in pre-NAC samples predominantly. Notably, post-NAC samples exhibited a significant increase in AR gene mutations, suggesting potential prognostic and predictive implications. No difference in mutational burden was found between patients who did and did not receive platinum (p = 0.94), or between those with and without recurrence (p = 0.49). We employed K-means clustering to categorize the patients based on their variant profiles, aiding in the prediction of possible patterns associated with recurrence. Our study was limited by its small sample size and retrospective design, suggesting the need for further validation in larger prospective cohorts.
Collapse
Affiliation(s)
- Ashish Singh
- Department of Medical Oncology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Josh Thomas Georgy
- Department of Medical Oncology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Sakthi Dhananjayan
- Department of Pathology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Elanthenral Sigamani
- Department of Pathology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Ajoy Oommen John
- Department of Medical Oncology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Anjana Joel
- Department of Medical Oncology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Jagan Chandramohan
- Department of Pathology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Rajadurai Abarna
- Department of Pathology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Grace Rebekah
- Department of Biostatistics, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Selvamani Backianathan
- Department of Radiotherapy, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | - Deepak Thomas Abraham
- Department of Endocrine Surgery, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | | | - Raju Titus Chacko
- Department of Medical Oncology, Christian Medical College, Vellore, Tamil Nadu 632004, India
| | | | - Rekha Pai
- Department of Pathology, Christian Medical College, Vellore, Tamil Nadu 632004, India.
| |
Collapse
|
2
|
Zhang Z, Wei X. Artificial intelligence-assisted selection and efficacy prediction of antineoplastic strategies for precision cancer therapy. Semin Cancer Biol 2023; 90:57-72. [PMID: 36796530 DOI: 10.1016/j.semcancer.2023.02.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 01/12/2023] [Accepted: 02/13/2023] [Indexed: 02/16/2023]
Abstract
The rapid development of artificial intelligence (AI) technologies in the context of the vast amount of collectable data obtained from high-throughput sequencing has led to an unprecedented understanding of cancer and accelerated the advent of a new era of clinical oncology with a tone of precision treatment and personalized medicine. However, the gains achieved by a variety of AI models in clinical oncology practice are far from what one would expect, and in particular, there are still many uncertainties in the selection of clinical treatment options that pose significant challenges to the application of AI in clinical oncology. In this review, we summarize emerging approaches, relevant datasets and open-source software of AI and show how to integrate them to address problems from clinical oncology and cancer research. We focus on the principles and procedures for identifying different antitumor strategies with the assistance of AI, including targeted cancer therapy, conventional cancer therapy, and cancer immunotherapy. In addition, we also highlight the current challenges and directions of AI in clinical oncology translation. Overall, we hope this article will provide researchers and clinicians with a deeper understanding of the role and implications of AI in precision cancer therapy, and help AI move more quickly into accepted cancer guidelines.
Collapse
Affiliation(s)
- Zhe Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, PR China; State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610041, PR China.
| |
Collapse
|
3
|
WeDIV – An improved k-means clustering algorithm with a weighted distance and a novel internal validation index. EGYPTIAN INFORMATICS JOURNAL 2022. [DOI: 10.1016/j.eij.2022.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
4
|
Goettsch KA, Zhang L, Singh AB, Dhawan P, Bastola DK. Reliable epithelial-mesenchymal transition biomarkers for colorectal cancer detection. Biomark Med 2022; 16:889-901. [PMID: 35892269 PMCID: PMC9442548 DOI: 10.2217/bmm-2022-0071] [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: 12/24/2022] Open
Abstract
Aims: To combat increases in colorectal cancer (CRC) incidence and mortality, biomarkers among differentially expressed genes (DEGs) have been identified to objectively detect cancer. However, DEGs are numerous, and additional parameters may identify more reliable biomarkers. Here, CRC DEGs were filtered into a prioritized list of biomarkers. Materials & methods: Two independent datasets (COAD-READ [n = 698] and GSE50760 [n = 36]) were input alternatively to the recently published data-driven reference method. Results were filtered based on epithelial-mesenchymal transition enrichment (χ-square statistic: 919.05; p = 2.2e-16) to produce 37 potential CRC biomarkers. Results: All 37 genes reliably classified CRC samples and ETV4, CLDN1 and CA2 together were top-ranked by DDR (accuracy: 89%; F1 score: 0.89). Conclusion: Biological and statistical information were combined to produce a better set of CRC detection biomarkers.
Collapse
Affiliation(s)
- Kaitlin A Goettsch
- School of Interdisciplinary Informatics, College of Information Science & Technology, University of Nebraska at Omaha, 1110 S. 67th Street, Omaha, NE 68182, USA
| | - Ling Zhang
- School of Interdisciplinary Informatics, College of Information Science & Technology, University of Nebraska at Omaha, 1110 S. 67th Street, Omaha, NE 68182, USA
| | - Amar B Singh
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, 42nd & Emile Streets, Omaha, NE 68198, USA.,Veterans Affairs Nebraska - Western Iowa Health Care System, Research Service, Omaha, NE 68105, USA
| | - Punita Dhawan
- Department of Biochemistry & Molecular Biology, University of Nebraska Medical Center, 42nd & Emile Streets, Omaha, NE 68198, USA
| | - Dhundy K Bastola
- School of Interdisciplinary Informatics, College of Information Science & Technology, University of Nebraska at Omaha, 1110 S. 67th Street, Omaha, NE 68182, USA
| |
Collapse
|
5
|
Framework for classification of cancer gene expression data using Bayesian hyper-parameter optimization. Med Biol Eng Comput 2021; 59:2353-2371. [PMID: 34609687 DOI: 10.1007/s11517-021-02442-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 09/13/2021] [Indexed: 10/20/2022]
Abstract
Computational classification of cancers is an important research problem. Gene expression data has 1000s of features, very few samples, and a class imbalance problem. In this paper, we have proposed a framework for the classification of cancer gene expression profiles. The framework consists of a pipeline of methods for data pre-processing, feature selection, and classification. Data pre-processing is done by standard scaling and normalization of the features. The feature selection is performed in two steps. First, recursive feature elimination (RFE) is used; then, a genetic algorithm is applied only in case RFE results in a feature subset of size more than a specific threshold. Next, is a meta-pool of diverse, individual as well as ensemble classifiers. Hyper-parameters of each member in the meta-pool are optimized using Bayesian Optimization. An algorithm is developed to select the best classifier from the meta-pool based on classification accuracy and computation time taken. We evaluated the framework on 6 publicly available microarray datasets and the PAN-Cancer RNA Sequencing dataset. We found that the classifier selected by the proposed framework produced significant improvement in classification accuracy and computation time required to predict labels for test datasets. A detailed comparison with the state-of-the-art methods shows that the proposed framework outperforms all of them.
Collapse
|
6
|
Zhang S, Li J, Gao H, Tong Y, Li P, Wang Y, Du L, Wang C. lncRNA Profiles Enable Prognosis Prediction and Subtyping for Esophageal Squamous Cell Carcinoma. Front Cell Dev Biol 2021; 9:656554. [PMID: 34127945 PMCID: PMC8196240 DOI: 10.3389/fcell.2021.656554] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 04/08/2021] [Indexed: 12/24/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) have emerged as useful prognostic markers in many tumors. In this study, we investigated the potential application of lncRNA markers for the prognostic prediction of esophageal squamous cell carcinoma (ESCC). We identified ESCC-associated lncRNAs by comparing ESCC tissues with normal tissues. Subsequently, Kaplan–Meier (KM) method in combination with the univariate Cox proportional hazards regression (UniCox) method was used to screen prognostic lncRNAs. By combining the differential and prognostic lncRNAs, we developed a prognostic model using cox stepwise regression analysis. The obtained prognostic prediction model could effectively predict the 3- and 5-year prognosis and survival of ESCC patients by time-dependent receiver operating characteristic (ROC) curves (area under curve = 0.87 and 0.89, respectively). Besides, a lncRNA-based classification of ESCC was generated using k-mean clustering method and we obtained two clusters of ESCC patients with association with race and Barrett’s esophagus (BE) (both P < 0.001). Finally, we found that lncRNA AC007128.1 was upregulated in both ESCC cells and tissues and associated with poor prognosis of ESCC patients. Furthermore, AC007128.1 could promote epithelial-mesenchymal transition (EMT) of ESCC cells by increasing the activation of MAPK/ERK and MAPK/p38 signaling pathways. Collectively, our findings indicated the potentials of lncRNA markers in the prognosis, molecular subtyping, and EMT of ESCC.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Juan Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huiru Gao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yao Tong
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Peilong Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yunshan Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| | - Chuanxin Wang
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Engineering & Technology Research Center for Tumor Marker Detection, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Clinical Laboratory, Jinan, China
| |
Collapse
|
7
|
Zhu X, Tian X, Ji L, Zhang X, Cao Y, Shen C, Hu Y, Wong JWH, Fang JY, Hong J, Chen H. A tumor microenvironment-specific gene expression signature predicts chemotherapy resistance in colorectal cancer patients. NPJ Precis Oncol 2021; 5:7. [PMID: 33580207 PMCID: PMC7881244 DOI: 10.1038/s41698-021-00142-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 10/21/2020] [Indexed: 12/14/2022] Open
Abstract
Studies have shown that tumor microenvironment (TME) might affect drug sensitivity and the classification of colorectal cancer (CRC). Using TME-specific gene signature to identify CRC subtypes with distinctive clinical relevance has not yet been tested. A total of 18 "bulk" RNA-seq datasets (total n = 2269) and four single-cell RNA-seq datasets were included in this study. We constructed a "Signature associated with FOLFIRI resistant and Microenvironment" (SFM) that could discriminate both TME and drug sensitivity. Further, SFM subtypes were identified using K-means clustering and verified in three independent cohorts. Nearest template prediction algorithm was used to predict drug response. TME estimation was performed by CIBERSORT and microenvironment cell populations-counter (MCP-counter) methods. We identified six SFM subtypes based on SFM signature that discriminated both TME and drug sensitivity. The SFM subtypes were associated with distinct clinicopathological, molecular and phenotypic characteristics, specific enrichments of gene signatures, signaling pathways, prognosis, gut microbiome patterns, and tumor lymphocytes infiltration. Among them, SFM-C and -F were immune suppressive. SFM-F had higher stromal fraction with epithelial-to-mesenchymal transition phenotype, while SFM-C was characterized as microsatellite instability phenotype which was responsive to immunotherapy. SFM-D, -E, and -F were sensitive to FOLFIRI and FOLFOX, while SFM-A, -B, and -C were responsive to EGFR inhibitors. Finally, SFM subtypes had strong prognostic value in which SFM-E and -F had worse survival than other subtypes. SFM subtypes enable the stratification of CRC with potential chemotherapy response thereby providing more precise therapeutic options for these patients.
Collapse
Affiliation(s)
- Xiaoqiang Zhu
- State Key Laboratory for Oncogenes and Related Genes, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Xianglong Tian
- State Key Laboratory for Oncogenes and Related Genes, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Linhua Ji
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xinyu Zhang
- State Key Laboratory for Oncogenes and Related Genes, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yingying Cao
- State Key Laboratory for Oncogenes and Related Genes, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chaoqin Shen
- State Key Laboratory for Oncogenes and Related Genes, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ye Hu
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Women's Cancer Program at the Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jason W H Wong
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Jing-Yuan Fang
- State Key Laboratory for Oncogenes and Related Genes, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Jie Hong
- State Key Laboratory for Oncogenes and Related Genes, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Haoyan Chen
- State Key Laboratory for Oncogenes and Related Genes, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
8
|
Bertsimas D, Wiberg H. Machine Learning in Oncology: Methods, Applications, and Challenges. JCO Clin Cancer Inform 2020; 4:885-894. [PMID: 33058693 PMCID: PMC7608565 DOI: 10.1200/cci.20.00072] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2020] [Indexed: 01/16/2023] Open
Affiliation(s)
- Dimitris Bertsimas
- Sloan School of Management, Massachusetts Institute of Technology, Cambridge, MA
- Operations Research Center, Massachusetts Institute of Technology, Cambridge, MA
| | - Holly Wiberg
- Operations Research Center, Massachusetts Institute of Technology, Cambridge, MA
| |
Collapse
|
9
|
Dimension Reduction and Clustering Models for Single-Cell RNA Sequencing Data: A Comparative Study. Int J Mol Sci 2020; 21:ijms21062181. [PMID: 32235704 PMCID: PMC7139673 DOI: 10.3390/ijms21062181] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/09/2020] [Accepted: 03/20/2020] [Indexed: 12/30/2022] Open
Abstract
With recent advances in single-cell RNA sequencing, enormous transcriptome datasets have been generated. These datasets have furthered our understanding of cellular heterogeneity and its underlying mechanisms in homogeneous populations. Single-cell RNA sequencing (scRNA-seq) data clustering can group cells belonging to the same cell type based on patterns embedded in gene expression. However, scRNA-seq data are high-dimensional, noisy, and sparse, owing to the limitation of existing scRNA-seq technologies. Traditional clustering methods are not effective and efficient for high-dimensional and sparse matrix computations. Therefore, several dimension reduction methods have been introduced. To validate a reliable and standard research routine, we conducted a comprehensive review and evaluation of four classical dimension reduction methods and five clustering models. Four experiments were progressively performed on two large scRNA-seq datasets using 20 models. Results showed that the feature selection method contributed positively to high-dimensional and sparse scRNA-seq data. Moreover, feature-extraction methods were able to promote clustering performance, although this was not eternally immutable. Independent component analysis (ICA) performed well in those small compressed feature spaces, whereas principal component analysis was steadier than all the other feature-extraction methods. In addition, ICA was not ideal for fuzzy C-means clustering in scRNA-seq data analysis. K-means clustering was combined with feature-extraction methods to achieve good results.
Collapse
|
10
|
Mutation Clusters from Cancer Exome. Genes (Basel) 2017; 8:genes8080201. [PMID: 28809811 PMCID: PMC5575665 DOI: 10.3390/genes8080201] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 07/26/2017] [Accepted: 08/07/2017] [Indexed: 11/17/2022] Open
Abstract
We apply our statistically deterministic machine learning/clustering algorithm *K-means (recently developed in https://ssrn.com/abstract=2908286) to 10,656 published exome samples for 32 cancer types. A majority of cancer types exhibit a mutation clustering structure. Our results are in-sample stable. They are also out-of-sample stable when applied to 1389 published genome samples across 14 cancer types. In contrast, we find in- and out-of-sample instabilities in cancer signatures extracted from exome samples via nonnegative matrix factorization (NMF), a computationally-costly and non-deterministic method. Extracting stable mutation structures from exome data could have important implications for speed and cost, which are critical for early-stage cancer diagnostics, such as novel blood-test methods currently in development.
Collapse
|