1
|
Nakauchi C, Masunaga N, Kagara N, Oshiro C, Shimoda M, Shimazu K. Development of a prediction model for ctDNA detection (Cir-Predict) in breast cancer. Breast Cancer Res Treat 2025; 211:331-339. [PMID: 40055250 PMCID: PMC12006266 DOI: 10.1007/s10549-025-07647-0] [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: 06/24/2024] [Accepted: 02/10/2025] [Indexed: 04/18/2025]
Abstract
PURPOSE The detection of circulating tumor DNA (ctDNA) is a valuable method to predict the risk of recurrence and to detect real-time gene changes. The amount of ctDNA is affected by many factors. Moreover, the detection rate of ctDNA varies from report to report. METHODS The present study evaluated differentially expressed genes using a DNA microarray assay for gene expression in tumors with and without detected ctDNA and constructed a prediction model for the detectability of ctDNA in breast tumor tissues. The model, named Cir-Predict, consisted of 126 probe sets (111 genes) and was constructed in a training set of breast cancer patients (n = 35) and validated in a validation set (n = 13). RESULTS The accuracy, sensitivity, and specificity in training and validation sets were over 90%, and Cir-Predict was significantly associated with ctDNA detection independently of the other conventional clinicopathological parameters in training and validation sets (P < 0.001, P = 0.014, respectively). Cir-Predict (+) was significantly associated with worse recurrence-free survival (P = 0.006). Pathway analysis revealed that nine pathways including tight junction and cell cycle tended to be related to ctDNA detectability. CONCLUSION Cir-Predict not only provides information useful for breast cancer treatment, but also helps the understanding of the mechanism by which ctDNA is detected.
Collapse
Affiliation(s)
- Chiaki Nakauchi
- Department of Breast Surgery, ISEIKAI International General Hospital, 4-14 Minamioogimachi, Kita-ku, Osaka City, Osaka, Japan.
| | - Nanae Masunaga
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Naofumi Kagara
- Department of Breast Surgery, Osaka General Medical Center, 3-1-56, Bandai-Higashi, Sumiyoshi-ku, Osaka City, Osaka, 558-8558, Japan
| | - Chiya Oshiro
- Department of Breast Surgery, Kaizuka City Hospital, 3-10-20 Ichibori, Kaizuka, Osaka, Japan
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-2-E10 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
2
|
Lužaić K, Lachanas K, Vamvakopoulos KO, Sidiropoulos A, Vamvakopoulou D, Nomikos I. Axilla Management in Breast Cancer Surgery: Brief Review and Current Practice Recommendations. Am Surg 2025; 91:834-842. [PMID: 39819186 DOI: 10.1177/00031348251313529] [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] [Indexed: 01/19/2025]
Abstract
The diagnostic and therapeutic approach to the axilla in breast cancer patients has changed significantly over the past 30 years, with the replacement of complete axillary lymph node dissection practices by less invasive approaches. Reference is made to clinical findings that have led to practical treatment recommendations and are paving the way to new levels of de-escalation in breast cancer surgery.
Collapse
Affiliation(s)
- Karla Lužaić
- Department of Emergency Medicine, Institute of Emergency Medicine of Sisak-Moslavina County, Sisak, Croatia
| | - Konstantinos Lachanas
- Department of Public Health and Social Medicine, Koutlimpanio and Triantafylleio General Hospital, Larissa, Greece
| | | | | | | | - Iakovos Nomikos
- Department of Surgery, Rea Maternity Hospital, Athens, Greece
- School of Medicine, European University Cyprus, Nicosia, Cyprus
| |
Collapse
|
3
|
Witkiewicz AK, Wang J, Schultz E, O'Connor TN, O'Connor T, Levine E, Knudsen ES. Using prognostic signatures and machine learning to identify core features associated with response to CDK4/6 inhibitor-based therapy in metastatic breast cancer. Oncogene 2025; 44:1387-1399. [PMID: 40011574 DOI: 10.1038/s41388-025-03308-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 01/06/2025] [Accepted: 02/10/2025] [Indexed: 02/28/2025]
Abstract
CDK4/6 inhibitors in combination with endocrine therapy are widely used to treat HR+/HER2- metastatic breast cancer leading to improved progression-free survival (PFS) compared to single agent endocrine therapy. Over 300 patients receiving standard-of-care CDK4/6 inhibitor combination therapy for metastatic disease were enrolled at a single institution. Clinical, pathological, and gene expression data were employed to define determinants for PFS duration. Visceral disease (HR 1.55, p = 0.0013), prior endocrine therapy (HR 2.34, p < 0.001), and the type of endocrine therapy (HR 2.16, p < 0.001) were highly associated with PFS duration. Multiple pre-defined gene expression signatures were employed to determine association with response to CDK4/6 inhibitor-based therapy. Random survival forest was applied to define key gene expression and clinical features associated with PFS and develop a predictive model. The time to progression predicted by this model was related to the median PFS observed in PALOMA-2/3 and PEARL studies. Interrogating genes identified as highly significant across all studies indicated common enrichment of gene networks associated with cell cycle and estrogen receptor signaling. These findings indicate that there are common features from real-world use of CDK4/6 inhibitors that could be used to infer time to progression and better inform treatment.
Collapse
Affiliation(s)
- Agnieszka K Witkiewicz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA.
- Department of Pathology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA.
| | - Jianxin Wang
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Emily Schultz
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Thomas N O'Connor
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Tracey O'Connor
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Ellis Levine
- Department of Medicine, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA
| | - Erik S Knudsen
- Department of Molecular and Cellular Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Street, Buffalo, NY, 14263, USA.
| |
Collapse
|
4
|
Karthikeyan SK, Chandrashekar DS, Sahai S, Shrestha S, Aneja R, Singh R, Kleer CG, Kumar S, Qin ZS, Nakshatri H, Manne U, Creighton CJ, Varambally S. MammOnc-DB, an integrative breast cancer data analysis platform for target discovery. NPJ Breast Cancer 2025; 11:35. [PMID: 40251157 PMCID: PMC12008238 DOI: 10.1038/s41523-025-00750-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 03/27/2025] [Indexed: 04/20/2025] Open
Abstract
Breast cancer (BCa), a leading malignancy among women, is characterized by morphological and molecular heterogeneity. While early-stage, hormone receptor, and HER2-positive BCa are treatable, triple-negative BCa and metastatic BCa remains largely untreatable. Advances in sequencing and proteomic technologies have improved our understanding of the molecular alterations that occur during BCa initiation and progression and enabled identification of subclass-specific biomarkers and therapeutic targets. Despite the availability of abundant omics data in public repositories, user-friendly tools for multi-omics data analysis and integration are scarce. To address this, we developed a comprehensive BCa data analysis platform called MammOnc-DB ( http://resource.path.uab.edu/MammOnc-Home.html ), comprising data from more than 20,000 BCa samples. MammOnc-DB facilitates hypothesis generation and testing, biomarker discovery, and therapeutic targets identification. The platform also includes pre- and post-treatment data, which can help users identify treatment resistance markers and support combination therapy strategies, offering researchers and clinicians a comprehensive tool for BCa data analysis and visualization.
Collapse
Affiliation(s)
| | | | - Snigdha Sahai
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Sadeep Shrestha
- Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - Ritu Aneja
- School of Health Professions, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - Rajesh Singh
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA, USA
| | - Celina G Kleer
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Sidharth Kumar
- Department of Computer Science, University of Illinois Chicago, Chicago, IL, USA
| | - Zhaohui S Qin
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | | | - Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chad J Creighton
- Department of Medicine and Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Sooryanarayana Varambally
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, USA.
- Department of Biomedical Informatics and Data Science, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
5
|
Bertinelli Salucci C, Bakdi A, Glad IK, Lindqvist BH, Vanem E, De Bin R. Lifetime analysis with monotonic degradation: a boosted first hitting time model based on a homogeneous gamma process. LIFETIME DATA ANALYSIS 2025; 31:300-339. [PMID: 40186714 PMCID: PMC12043765 DOI: 10.1007/s10985-025-09648-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 03/04/2025] [Indexed: 04/07/2025]
Abstract
In the context of time-to-event analysis, First hitting time methods consider the event occurrence as the ending point of some evolving process. The characteristics of the process are of great relevance for the analysis, which makes this class of models interesting and particularly suitable for applications where something about the degradation path is known. In cases where the degradation can only worsen, a monotonic process is the most suitable choice. This paper proposes a boosting algorithm for first hitting time models based on an underlying homogeneous gamma process to account for the monotonicity of the degradation trend. The predictive power and versatility of the algorithm are shown with real data examples from both engineering and biomedical applications, as well as with simulated examples.
Collapse
Affiliation(s)
| | - Azzeddine Bakdi
- Corvus Energy, Tormod Gjestlands veg 51, 3936, Porsgrunn, Norway
| | - Ingrid Kristine Glad
- Department of Mathematics, University of Oslo, Moltke Moes vei 35, 0851, Oslo, Norway
| | - Bo Henry Lindqvist
- Department of Mathematical Sciences, Norwegian University of Science and Technology, Alfred Getz vei 1, 7491, Trondheim, Norway
| | - Erik Vanem
- Department of Mathematics, University of Oslo, Moltke Moes vei 35, 0851, Oslo, Norway
- DNV Group Technology and Research, Veritasveien 1, 1322, Høvik, Norway
| | - Riccardo De Bin
- Department of Mathematics, University of Oslo, Moltke Moes vei 35, 0851, Oslo, Norway
| |
Collapse
|
6
|
Liu Z, Deng J, Xu H, Liu L, Zhang Y, Ba Y, Zhang Z, He F, Xie L. Efficient discovery of robust prognostic biomarkers and signatures in solid tumors. Cancer Lett 2025; 613:217502. [PMID: 39864538 DOI: 10.1016/j.canlet.2025.217502] [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: 11/25/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Recent advancements in multi-omics and big-data technologies have facilitated the discovery of numerous cancer prognostic biomarkers and gene signatures. However, their clinical application remains limited due to poor reproducibility and insufficient independent validation. Despite the availability of high-quality datasets, achieving reliable biomarker identification across multiple cohorts continues to be a significant challenge. To address these issues, we developed a comprehensive platform, SurvivalML, designed to support the discovery and validation of prognostic biomarkers and gene signatures using large-scale and harmonized data from 21 cancer types. Through SurvivalML, we identified DCLRE1B as a novel prognostic biomarker for hepatocellular carcinoma, with experimental confirmation of its role in promoting tumor progression. Additionally, we developed the Chinese glioblastoma prognostic signature (CGPS) and its simplified version, SCGPS, a three-gene model. Both demonstrated superior predictive performance compared to other glioblastoma signatures in our in-house cohort and five independent Chinese datasets. The SCGPS model was further validated in 109 clinical samples using multiplex immunofluorescence, showing strong consistency with the original CGPS model. Overall, SurvivalML provides a robust platform for the identification and validation of prognostic biomarkers and gene signatures, offering a valuable resource for advancing cancer research and clinical application.
Collapse
Affiliation(s)
- Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, 100730, Beijing, China; State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 102206, Beijing, China; International Academy of Phronesis Medicine (Guangdong), 510320, Guangdong, China
| | - Jinhai Deng
- Richard Dimbleby Department of Cancer Research, Comprehensive Cancer Centre, Kings College London, London, United Kingdom
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Long Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, 710061, Xi'an, Shanxi, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Zhengyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
| | - Fuchu He
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 102206, Beijing, China; International Academy of Phronesis Medicine (Guangdong), 510320, Guangdong, China; Research Unit of Proteomics Dirven Cancer Precision Medicine, Chinese Academy of Medical Sciences, 102206, Beijing, China.
| | - Linhai Xie
- State Key Laboratory of Medical Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 102206, Beijing, China; International Academy of Phronesis Medicine (Guangdong), 510320, Guangdong, China.
| |
Collapse
|
7
|
Cha J, Lee I. Single-cell network biology enabling cell-type-resolved disease genetics. Genomics Inform 2025; 23:10. [PMID: 40148916 PMCID: PMC11951680 DOI: 10.1186/s44342-025-00042-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Gene network models provide a foundation for graph theory approaches, aiding in the novel discovery of drug targets, disease genes, and genetic mechanisms for various biological functions. Disease genetics must be interpreted within the cellular context of disease-associated cell types, which cannot be achieved with datasets consisting solely of organism-level samples. Single-cell RNA sequencing (scRNA-seq) technology allows computational distinction of cell states which provides a unique opportunity to understand cellular biology that drives disease processes. Importantly, the abundance of cell samples with their transcriptome-wide profile allows the modeling of systemic cell-type-specific gene networks (CGNs), offering insights into gene-cell-disease relationships. In this review, we present reference-based and de novo inference of gene functional interaction networks that we have recently developed using scRNA-seq datasets. We also introduce a compendium of CGNs as a useful resource for cell-type-resolved disease genetics. By leveraging these advances, we envision single-cell network biology as the key approach for mapping the gene-cell-disease axis.
Collapse
Affiliation(s)
- Junha Cha
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| | - Insuk Lee
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Republic of Korea.
| |
Collapse
|
8
|
Tian BX, Yu ZX, Qiu X, Chen LP, Zhuang YL, Chen Q, Gu YH, Hou MJ, Gu YF. Development and validation of a 14-CpG DNA methylation signature and drug targets for prognostic prediction in breast cancer. Front Med (Lausanne) 2025; 12:1548726. [PMID: 40177272 PMCID: PMC11961922 DOI: 10.3389/fmed.2025.1548726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/28/2025] [Indexed: 04/05/2025] Open
Abstract
Background Breast cancer (BC) is the most prevalent cancer among women and a leading cause of cancer-related deaths worldwide. Emerging evidence suggests that DNA methylation, a well-studied epigenetic modification, regulates various cellular processes critical for cancer development and progression and holds promise as a biomarker for cancer diagnosis and prognosis, potentially enhancing the efficacy of precision therapies. Methods We developed a robust prognostic model for BC based on DNA methylation and clinical data from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO). We analyzed the association of the model with clinicopathological features, survival outcomes, and chemotherapy drug sensitivity. Results A set of 216 differentially methylated CpGs was identified by intersecting three datasets (TCGA, GSE22249, and GSE66695). Using univariate Cox proportional hazard and LASSO Cox regression analyses, we constructed a 14-CpG model significantly associated with progression-free interval (PFI), disease-specific survival (DSS), and overall survival (OS) in BC patients. Kaplan-Meier (KM) survival analysis, receiver operating characteristic (ROC) analysis, and nomogram validation confirmed the clinical value of the signature. The Cox analysis showed a significant association between the signature and PFI and DSS in BC patients. KM analysis effectively distinguished high-risk from low-risk patients, while ROC analysis demonstrated high sensitivity and specificity in predicting BC prognosis. A nomogram based on the signature effectively predicted 5- and 10-year PFI and DSS. Additionally, combining our model with clinical risk factors suggested that patients in the I-II & M+ subgroup could benefit from adjuvant chemotherapy regarding PFI, DSS, and OS. Gene Ontology (GO) functional enrichment and KEGG pathway analyses indicated that the top 3,000 differentially expressed genes (DEGs) were enriched in pathways related to DNA replication and repair and cell cycle regulation. Patients in the high-risk group might benefit from drugs targeting DNA replication and repair processes in tumor cells. Conclusion The 14-CpG model serves as a useful biomarker for predicting prognosis in BC patients. When combined with TNM staging, it offers a potential strategy for individualized clinical decision-making, guiding personalized therapeutic regimen selection for clinicians.
Collapse
Affiliation(s)
- Bao-xing Tian
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-xi Yu
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xia Qiu
- Department of Breast Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li-ping Chen
- Department of Breast Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu-lian Zhuang
- Department of Nursing, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Chen
- Department of Nursing, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yan-hua Gu
- Department of Nursing, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng-jie Hou
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai 9th People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-fan Gu
- Department of Breast Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
9
|
Bedi P, Rani S, Gupta B, Bhasin V, Gole P. EpiBrCan-Lite: A lightweight deep learning model for breast cancer subtype classification using epigenomic data. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 260:108553. [PMID: 39667144 DOI: 10.1016/j.cmpb.2024.108553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 11/14/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND AND OBJECTIVES Early breast cancer subtypes classification improves the survival rate as it facilitates prognosis of the patient. In literature this problem was prominently solved by various Machine Learning and Deep Learning techniques. However, these studies have three major shortcomings: huge Trainable Weight Parameters (TWP), suffer from low performance and class imbalance problem. METHODS This paper proposes a lightweight model named EpiBrCan-Lite for classifying breast cancer subtypes using DNA methylation data. This model encompasses three blocks namely Data Encoding, TransGRU, and Classification blocks. In Data Encoding block, the input features are encoded into equal sized chunks and then passed down to TransGRU block which is a modified version of traditional Transformer Encoder (TE). In TransGRU block, MLP module of traditional TE is replaced by GRU module, consisting of two GRU layers to reduce TWP and capture the long-range dependencies of input feature data. Furthermore, output of TransGRU block is passed to Classification block for classifying breast cancer into their subtypes. RESULTS The proposed model is validated using Accuracy, Precision, Recall, F1-score, FPR, and FNR metrics on TCGA breast cancer dataset. This dataset suffers from the class imbalance problem which is mitigated using Synthetic Minority Oversampling Technique (SMOTE). Experimentation results demonstrate that EpiBrCan-Lite model attained 95.85 % accuracy, 95.96 % recall, 95.85 % precision, 95.90 % F1-score, 1.03 % FPR, and 4.12 % FNR despite of utilizing only 1/1500 of TWP than other state-of-the-art models. CONCLUSION EpiBrCan-Lite model is efficiently classifying breast cancer subtypes, and being lightweight, it is suitable to be deployed on low computational powered devices.
Collapse
Affiliation(s)
- Punam Bedi
- Department of Computer Science, University of Delhi, Delhi, India.
| | - Surbhi Rani
- Department of Computer Science, University of Delhi, Delhi, India.
| | - Bhavna Gupta
- Keshav Mahavidyalaya, University of Delhi, New Delhi, India.
| | - Veenu Bhasin
- PGDAV College, University of Delhi, New Delhi, India.
| | - Pushkar Gole
- Department of Computer Science, University of Delhi, Delhi, India.
| |
Collapse
|
10
|
Bal S, Kumari R. The Clinical Significance of the Magee Equations in Breast Cancer Prognostication. Cureus 2025; 17:e81528. [PMID: 40308430 PMCID: PMC12043356 DOI: 10.7759/cureus.81528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
The landscape of breast cancer management has been revolutionized by advancements in molecular diagnostics, yet accessibility and cost remain significant barriers for many patients. Limited-resource settings often face significant challenges in accessing expensive molecular tests, which can impact timely diagnosis and treatment decisions. The Magee equations present a practical, cost-effective solution that can bridge this gap, ensuring that patients, regardless of their financial or geographic limitations, receive appropriate and timely treatment. Originally developed at the University of Pittsburgh Medical Center, these equations offer a reliable alternative for estimating recurrence scores without the prohibitive costs associated with genomic assays.
Collapse
Affiliation(s)
| | - Roopa Kumari
- Pathology, Mount Sinai Morningside, New York, USA
| |
Collapse
|
11
|
Xu J, Jin M, Mu Z, Li Z, Qi R, Han X, Jiang H. Inhibiting melanoma tumor growth: the role of oxidative stress-associated LINC02132 and COPDA1 long non-coding RNAs. Front Immunol 2025; 16:1558292. [PMID: 40092985 PMCID: PMC11906686 DOI: 10.3389/fimmu.2025.1558292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/11/2025] [Indexed: 03/19/2025] Open
Abstract
Background Cutaneous melanoma is a type of malignant tumor that is challenging to predict and is readily stimulated by various factors. Oxidative stress can induce damage and alterations in melanocytes, subsequently triggering immune responses. Given that oxidative stress is a prevalent tumor stimulus, we aimed to enhance melanoma prediction by identifying lncRNA signatures associated with oxidative stress. Methods We screened for oxidative stress-related lncRNAs that could improve melanoma patient prognosis using the TCGA and GTEx databases. Utilizing differentially expressed oxidative stress-related lncRNAs (DE-OSlncRNAs), we constructed a Lasso regression model. The accuracy of the model was validated using univariate and multivariate regression, Kaplan-Meier (K-M) curves, and ROC curves. Subsequently, we conducted immune infiltration analysis, immune checkpoint differential analysis, IC50 pharmaceutical analysis, and gene set enrichment analysis. Investigating the effects of the target gene on melanoma using fluorescence in situ hybridization (FISH), quantitative real-time PCR (qRT-PCR), Edu assay, wound healing assay, transwell assay, flow cytometry, and reactive oxygen species (ROS) detection. Results Thirteen lncRNAs were identified as significant prognostic factors. Four oxidative stress-related lncRNAs (COPDA1, LINC02132, LINC02812, and MIR205HG) were further validated by fluorescence in situ hybridization (FISH), with results consistent with our data analysis. LINC02132 and COPDA1 can influence the proliferation, invasion, migration, and apoptosis of melanoma. Conclusion Our findings suggest that upregulation of the LINC02132 or COPDA1 genes elevates intracellular reactive oxygen species (ROS) levels in melanoma cells, suppresses tumor cell proliferation, migration, and invasion, and promotes apoptosis. These results suggest a novel therapeutic strategy for melanoma treatment.
Collapse
Affiliation(s)
- JingWen Xu
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - MingZhu Jin
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - ZhenZhen Mu
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - ZhengXiu Li
- Department of Dermatology & Key Lab of Dermatology, Ministry of Education and Public Health, National Joint Engineering Research Center for Theranostics of Immunology Skin Diseases, The First Hospital of China Medical University, Shenyang, China
| | - RuiQun Qi
- Department of Dermatology & Key Lab of Dermatology, Ministry of Education and Public Health, National Joint Engineering Research Center for Theranostics of Immunology Skin Diseases, The First Hospital of China Medical University, Shenyang, China
| | - XiuPing Han
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| | - HangHang Jiang
- Department of Dermatology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
12
|
Singhal A, Zhao X, Wall P, So E, Calderini G, Partin A, Koussa N, Vasanthakumari P, Narykov O, Zhu Y, Jones SE, Abbas-Aghababazadeh F, Nair SK, Bélisle-Pipon JC, Jayaram A, Parker BA, Yeung KT, Griffiths JI, Weil R, Nath A, Haibe-Kains B, Ideker T. The Hallmarks of Predictive Oncology. Cancer Discov 2025; 15:271-285. [PMID: 39760657 PMCID: PMC11969157 DOI: 10.1158/2159-8290.cd-24-0760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 08/30/2024] [Accepted: 10/16/2024] [Indexed: 01/07/2025]
Abstract
SIGNIFICANCE As the field of artificial intelligence evolves rapidly, these hallmarks are intended to capture fundamental, complementary concepts necessary for the progress and timely adoption of predictive modeling in precision oncology. Through these hallmarks, we hope to establish standards and guidelines that enable the symbiotic development of artificial intelligence and precision oncology.
Collapse
Affiliation(s)
- Akshat Singhal
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Xiaoyu Zhao
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Patrick Wall
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
| | - Emily So
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Guido Calderini
- Faculty of Health Science, Simon Fraser University, Burnaby, BC, Canada
- École de santé publique, Université de Montréal, Montréal, QC, Canada
| | - Alexander Partin
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - Natasha Koussa
- Cancer Data Science Initiatives, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | - Oleksandr Narykov
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - Yitan Zhu
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - Sara E. Jones
- Cancer Data Science Initiatives, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | | | | | | | | | - Barbara A. Parker
- Moores Cancer Center, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Kay T. Yeung
- Moores Cancer Center, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| | - Jason I. Griffiths
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute, City of Hope National Medical Center, Monrovia, CA, USA
| | - Ryan Weil
- Cancer Data Science Initiatives, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Aritro Nath
- Department of Medical Oncology and Therapeutics Research, Beckman Research Institute, City of Hope National Medical Center, Monrovia, CA, USA
| | - Benjamin Haibe-Kains
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
- Medical Biophysics, University of Toronto, Toronto, Canada
- Vector Institute for Artificial Intelligence, Toronto, Canada
- Department of Biostatistics, Dalla Lana School of Public Health, Toronto, Canada
| | - Trey Ideker
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
- Division of Human Genomics and Precision Medicine, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, USA
- Moores Cancer Center, Department of Medicine, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
13
|
Hortobagyi GN, Lacko A, Sohn J, Cruz F, Ruiz Borrego M, Manikhas A, Hee Park Y, Stroyakovskiy D, Yardley DA, Huang CS, Fasching PA, Crown J, Bardia A, Chia S, Im SA, Martin M, Loi S, Xu B, Hurvitz S, Barrios C, Untch M, Moroose R, Visco F, Parnizari F, Zarate JP, Li Z, Waters S, Chakravartty A, Slamon D. A phase III trial of adjuvant ribociclib plus endocrine therapy versus endocrine therapy alone in patients with HR-positive/HER2-negative early breast cancer: final invasive disease-free survival results from the NATALEE trial. Ann Oncol 2025; 36:149-157. [PMID: 39442617 DOI: 10.1016/j.annonc.2024.10.015] [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: 07/01/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND NATALEE assessed efficacy and tolerability of 3 years of adjuvant ribociclib plus a nonsteroidal aromatase inhibitor (NSAI) compared with an NSAI alone in a broad population of patients with hormone receptor (HR)-positive/human epidermal growth factor 2 (HER2)-negative early breast cancer, including a select group without nodal involvement. This is the final preplanned analysis of invasive disease-free survival (iDFS). PATIENTS AND METHODS Premenopausal/postmenopausal women and men were randomized 1 : 1 to ribociclib (n = 2549; 400 mg/day, 3 weeks on/1 week off for 36 months) plus NSAI (letrozole 2.5 mg/day or anastrozole 1 mg/day for 60 months) or NSAI alone (n = 2552). Men and premenopausal women also received goserelin (3.6 mg once every 28 days). Patients had anatomical stage IIA (N0 with additional risk factors or N1), IIB, or III disease. The primary endpoint was iDFS. Secondary efficacy endpoints were recurrence-free survival (RFS), distant DFS, and overall survival. This final iDFS analysis was planned after ∼500 events. RESULTS At data cut-off (21 July 2023), ribociclib was stopped for 1996 patients (78.3%); 1091 (42.8%) completed 3 years of ribociclib, and ribociclib treatment was ongoing for 528 (20.7%). Median follow-up for iDFS was 33.3 months. Overall, 226 and 283 iDFS events occurred with ribociclib plus NSAI versus NSAI alone, respectively. Ribociclib plus NSAI demonstrated significant iDFS benefit over NSAI alone [hazard ratio 0.749, 95% confidence interval (CI) 0.628-0.892; P = 0.0012]. The 3-year iDFS rates were 90.7% (95% CI 89.3% to 91.8%) versus 87.6% (95% CI 86.1% to 88.9%). A consistent benefit was observed across prespecified subgroups, including stage (II/III) and nodal status (positive/negative). Distant DFS and RFS favored ribociclib plus NSAI. Overall survival data were immature. No new safety signals were observed. CONCLUSIONS With longer follow-up and most patients off ribociclib, NATALEE continues to demonstrate iDFS benefit with ribociclib plus NSAI over NSAI alone in the overall population and across key subgroups. Observed adverse events remained stable.
Collapse
Affiliation(s)
- G N Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA.
| | - A Lacko
- Dolnoslaskie Centrum Onkologii, Wroclaw, Poland
| | - J Sohn
- Severance Hospital, Seoul, Korea
| | - F Cruz
- Instituto Brasileiro de Controle do Câncer, São Paulo, Brazil
| | | | - A Manikhas
- Saint Petersburg City Clinical Oncology Dispensary, Saint Petersburg, Russia
| | | | - D Stroyakovskiy
- Moscow City Oncology Hospital No. 62 of Moscow Healthcare Department, Moscow, Russia
| | - D A Yardley
- Sarah Cannon Research Institute, Nashville, USA
| | - C-S Huang
- National Taiwan University Hospital, National Taiwan University College of Medicine, Taipei City, Taiwan
| | - P A Fasching
- University Hospital Erlangen Comprehensive Cancer Center Erlangen-EMN, Friedrich-Alexander University Erlangen-Nuremberg, Erlangen, Germany
| | - J Crown
- St Vincent's Private Hospital, Dublin, Ireland
| | - A Bardia
- David Geffen School of Medicine at UCLA, Los Angeles, USA
| | - S Chia
- BC Cancer - Vancouver, Vancouver, Canada
| | - S-A Im
- Cancer Research Institute, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - M Martin
- Instituto de Investigación Sanitaria Gregorio Marañón, Centro de Investigación Biomédica en Red de Cáncer, Grupo Español de Investigación en Cáncer de Mama, Universidad Complutense, Madrid, Spain
| | - S Loi
- Peter MacCallum Cancer Centre, Melbourne, Australia
| | - B Xu
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, and Peking Union Medical College, Beijing, China
| | - S Hurvitz
- University of Washington, Fred Hutchinson Cancer Center, Seattle, USA
| | - C Barrios
- Latin American Cooperative Oncology Group, Porto Alegre, Brazil
| | - M Untch
- Interdisciplinary Breast Cancer Center, Helios Klinikum Berlin-Buch, Berlin, Germany
| | - R Moroose
- Orlando Health Cancer Institute, Orlando
| | - F Visco
- National Breast Cancer Coalition, Washington, USA
| | - F Parnizari
- TRIO - Translational Research in Oncology, Montevideo, Uruguay
| | - J P Zarate
- Novartis Pharmaceuticals Corporation, East Hanover, USA
| | - Z Li
- Novartis Pharmaceuticals Corporation, East Hanover, USA
| | - S Waters
- Novartis Ireland, Dublin, Ireland
| | | | - D Slamon
- David Geffen School of Medicine at UCLA, Los Angeles, USA
| |
Collapse
|
14
|
Rios-Hoyo A, Xiong K, Dai J, Yau C, Marczyk M, García-Milian R, Wolf DM, Huppert LA, Nanda R, Hirst GL, Cobain EF, van ‘t Veer LJ, Esserman LJ, Pusztai L. Hormone Receptor-Positive HER2-Negative/MammaPrint High-2 Breast Cancers Closely Resemble Triple-Negative Breast Cancers. Clin Cancer Res 2025; 31:403-413. [PMID: 39561272 PMCID: PMC11747811 DOI: 10.1158/1078-0432.ccr-24-1553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 09/16/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024]
Abstract
PURPOSE The MammaPrint (MP) prognostic assay categorizes breast cancers into high- and low-risk subgroups, and the high-risk group can be further subdivided into high-1 (MP-H1), and very high-risk high-2 (MP-H2). The aim of this analysis was to assess clinical and molecular differences between the hormone receptor-positive (HR+)/HER2-negative MP-H1, -H2, and triple-negative (TN) MP-H1 and -H2 cancers. EXPERIMENTAL DESIGN Pretreatment gene expression data from 742 HER2-negative breast cancers enrolled in the I-SPY2 neoadjuvant trial were used. Prognostic risk categories were assigned using the MP assay. Transcriptional similarities across the four receptor and prognostic groups were assessed using principal component analyses and by identifying differentially expressed genes. We also examined pathologic complete response rates and event-free survivals by risk group. RESULTS Principal component analysis showed that HR+/MP-H2 tumors clustered with TN/MP-H2 cancers. Only 125 genes showed differential expression between the HR+/MP-H2 and TN/MP-H2 cancers, whereas 1,465 genes were differentially expressed between HR+/MP-H2 and -H1. Gene set analysis revealed similarly high expression of cell cycle, DNA repair, and immune infiltration-related pathways in HR+/MP-H2 and TN/MP-H2 cancers. HR+/MP-H2 cancers also showed low estrogen receptor-related gene expression. Pathologic complete response rates were similarly high in TN/MP-H2 and HR+/MP-H2 cancers (42% vs. 30.5%; P = 0.11), and MP-H2 cancers with residual cancer had similarly poor event-free survival regardless of estrogen receptor status. CONCLUSIONS In conclusion, HR+/MP-H2 cancers closely resemble TN breast cancers in transcriptional and clinical features and benefit from similar treatment strategies.
Collapse
Affiliation(s)
- Alejandro Rios-Hoyo
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
- To be considered as first authors
| | - Kaitlyn Xiong
- Yale School of Medicine, New Haven, Connecticut, USA
- To be considered as first authors
| | - Jiawei Dai
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christina Yau
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Michal Marczyk
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
- Department of Data Science and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Rolando García-Milian
- Bioinformatics Support Program, Research and Education Services, Cushing/Whitney Medical Library, Yale University, New Haven, CT, United States of America
| | - Denise M. Wolf
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Laura A. Huppert
- University of California San Francisco Comprehensive Cancer Center, San Francisco, CA, USA
| | - Rita Nanda
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine & Biological Sciences, Chicago, IL, USA
| | - Gillian L. Hirst
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | | | - Laura J. van ‘t Veer
- Department of Laboratory Medicine, University of California, San Francisco, CA, USA
| | - Laura J. Esserman
- Department of Surgery, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lajos Pusztai
- Yale Cancer Center, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
15
|
Bernal Astrain G, Strakhova R, Jo CH, Teszner E, Killoran RC, Smith MJ. The small GTPase MRAS is a broken switch. Nat Commun 2025; 16:647. [PMID: 39809765 PMCID: PMC11733253 DOI: 10.1038/s41467-025-55967-y] [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: 08/28/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
Intense research on founding members of the RAS superfamily has defined our understanding of these critical signalling proteins, leading to the premise that small GTPases function as molecular switches dependent on differential nucleotide loading. The closest homologs of H/K/NRAS are the three-member RRAS family, and interest in the MRAS GTPase as a regulator of MAPK activity has recently intensified. We show here that MRAS does not function as a classical switch and is unable to exchange GDP-to-GTP in solution or when tethered to a lipid bilayer. The exchange defect is unaffected by inclusion of the GEF SOS1 and is conserved in a distal ortholog from nematodes. Synthetic activating mutations widely used to study the function of MRAS in a presumed GTP-loaded state do not increase exchange, but instead drive effector binding due to sampling of an activated conformation in the GDP-loaded state. This includes nucleation of the SHOC2-PP1Cα holophosphatase complex. Acquisition of NMR spectra from isotopically labeled MRAS in live cells validated the GTPase remains fully GDP-loaded, even a supposed activated mutant. These data show that RAS GTPases, including those most similar to KRAS, have disparate biochemical activities and challenge current dogma on MRAS, suggesting previous data may need reinterpretation.
Collapse
Affiliation(s)
- Gabriela Bernal Astrain
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Programmes de biologie moléculaire, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Regina Strakhova
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Programmes de biologie moléculaire, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Chang Hwa Jo
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Emma Teszner
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
- Programmes de biologie moléculaire, Université de Montréal, Montreal, QC, H3C 3J7, Canada
| | - Ryan C Killoran
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada
| | - Matthew J Smith
- Institute for Research in Immunology and Cancer (IRIC), Université de Montréal, Montréal, QC, H3T 1J4, Canada.
- Programmes de biologie moléculaire, Université de Montréal, Montreal, QC, H3C 3J7, Canada.
- Department of Pathology and Cell Biology, Faculty of Medicine, Université de Montréal, Montréal, QC, H3T 1J4, Canada.
| |
Collapse
|
16
|
Jiang G, Wang Y. Development of endosome-related gene signature for the prediction of prognosis and therapeutic response in breast cancer. Medicine (Baltimore) 2025; 104:e41230. [PMID: 39792732 PMCID: PMC11730407 DOI: 10.1097/md.0000000000041230] [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: 05/11/2024] [Accepted: 12/18/2024] [Indexed: 01/12/2025] Open
Abstract
Endosomes play a pivotal role in cellular biology, orchestrating processes such as endocytosis, molecular trafficking, signal transduction, and recycling of cellular materials. This study aims to construct an endosome-related gene (ERG)-derived risk signature for breast cancer prognosis. Transcriptomic and clinical data were retrieved from The Cancer Genome Atlas and the University of California Santa Cruz databases to build and validate the model. A Lasso Cox regression model was employed for risk signature construction. The immune landscape was assessed using CIBERSORT and ESTIMATE algorithms, while drug sensitivity was evaluated via the pRRophetic algorithm. Gene set enrichment analysis and gene set variation analysis were applied to evaluate gene expression patterns. A nomogram was constructed and validated for predicting breast cancer outcomes. The expression of ERGs in breast cancer cells and tissues was further validated. Sixty-one ERGs associated with breast cancer prognosis were identified, with 23 selected for constructing the risk signature. This signature stratified breast cancer patients into high- and low-risk groups, where the low-risk group exhibited significantly better prognosis. Notably, younger patients tended to have lower risk scores compared to older ones. The low-risk group exhibited enhanced sensitivity to the majority of the drugs tested, accompanied by increased infiltration of T cells and M1 macrophages. Additionally, cell cycle pathways were suppressed in the low-risk group, whereas antigen binding functions were significantly activated. Ultimately, risk score and age were identified as independent prognostic factors for breast cancer, and these factors were incorporated into a nomogram that demonstrated excellent performance in prognosis assessment. Finally, external cohort validated the dysregulation of the risk score-associated ERGs in breast cancer cells and tissues. This study successfully established an ERG-derived breast cancer risk signature and nomogram, elucidating their potential value in prognosis prediction and evaluation of therapeutic response.
Collapse
Affiliation(s)
- Guowei Jiang
- Department of Breast, Haining Maternity and Child Health Care Hospital, Haining, Zhejieng, China
| | - Ye Wang
- Department of Breast, Haining Maternity and Child Health Care Hospital, Haining, Zhejieng, China
| |
Collapse
|
17
|
Kanu GA, Mouselly A, Mohamed AA. Foundations and applications of computational genomics. DEEP LEARNING IN GENETICS AND GENOMICS 2025:59-75. [DOI: 10.1016/b978-0-443-27574-6.00007-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
18
|
Fisher EB. Spinoza, Liberation From Causation, and Community Health Promotion. Am J Health Promot 2025; 39:172-175. [PMID: 39358208 PMCID: PMC11568658 DOI: 10.1177/08901171241286876] [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: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 10/04/2024]
Abstract
What sense does it make to say that a new program implemented in a community with roots as old as evolution caused an observed health benefit? Evaluation of community approaches has often sought to isolate the causal roles of interventions. Central to this is the assumption that there are causes to be proven and isolated. Benedict Spinoza (1632-1677) dismissed the concept of cause, arguing that all things, "substances," are not caused but simply are. Actions of things in nature can influence each other, e.g., erosion of a mountain, but their substance, the mountains simply are. For Spinoza, satisfaction in life comes from realizing and acting in accord with our substance, but this requires communities that support such realization and action. Thus, communities and the vast influences they contain are central to human welfare. Interventions within them do not cause benefits but join with the history, culture, and numerous other features of the community in becoming part of how the community influences its members. Implications include a) expanding the social ecological model fully to embrace multiple influences - including innovative programs - and interactions among them, and c) varied research methods to identify practical lessons about how communities may adopt and incorporate innovations to engender change, rather than a catalogue of interventions that are supposed to change them.
Collapse
Affiliation(s)
- Edwin B. Fisher
- Peers for Progress and Department of Health Behavior, Gillings School of Global Public Health, University of North Carolina Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
19
|
Hu H, Li X, Xu Z, Tao Y, Zhao L, You H, Xu G, Zhang T, Zhang Y, Fan H, Wang X, Chen W, Lin CG, Zheng H. OPG promotes lung metastasis by reducing CXCL10 production of monocyte-derived macrophages and decreasing NK cell recruitment. EBioMedicine 2025; 111:105503. [PMID: 39674088 PMCID: PMC11700254 DOI: 10.1016/j.ebiom.2024.105503] [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: 08/06/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 12/16/2024] Open
Abstract
BACKGROUND Lung metastasis is a critical and often fatal progression in cancer patients, with monocyte-derived macrophages (Mo-macs) playing multifaceted roles in this process. Despite the recognized importance of Mac-macs, most studies focus on these cells themselves, while the precise mechanisms through which tumor cells manipulate Mo-macs to promote metastasis remain poorly understood. METHODS We developed an in vivo CRISPR screening system to identify genes involved in macrophage-dependent metastasis by depleting Mo-macs. Osteoprotegerin (OPG) was identified as the factor significantly enhances lung metastasis. We validated its function in lung metastasis by modulating the expression of OPG in an array of cell lines and performed spontaneous and experimental lung metastasis assays. Genetically engineered mice were utilized to confirm the role of RANKL-RANK signaling in OPG-mediated metastasis. Additionally, we employed different neutralizing antibodies to elucidate the roles of Mo-macs and NK cells and inhibitor to clarify the role of CXCL10 signaling. FINDINGS Employing in vivo screening techniques, we elucidate the role of OPG, a protein secreted by cancer cells, in driving lung metastasis, contingent upon regulating Mo-mac activity. OPG blocks the signaling cascade between receptor activator of nuclear factor kappa-B ligand (RANKL) and its receptor RANK on Mo-macs, thereby hindering Mo-macs from secreting CXCL10, a chemokine crucial for recruiting natural killer (NK) cells that help control lung metastasis. Moreover, we observe an enrichment of OPG amplifications in metastatic cancer patients, and elevated levels of OPG expression in lung metastatic sites compared to paired primary breast cancer samples. INTERPRETATION Our work revealed that OPG works as a lung metastasis promoting factor by blocking the RANKL-RANK-CXCL10 axis to drive the paucity of NK cells, which could be a therapeutic target for lung metastatic cancer patients. FUNDING The full list of funding supporting this study can be found in the Acknowledgements section.
Collapse
Affiliation(s)
- Haitian Hu
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Xuan Li
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Zhanao Xu
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuwei Tao
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Luyang Zhao
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Huiwen You
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Guoyuan Xu
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Tengjiang Zhang
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Yuan Zhang
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Huijuan Fan
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Xuxiang Wang
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Wenjing Chen
- SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China
| | - Christopher G Lin
- Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China
| | - Hanqiu Zheng
- State Key Laboratory of Molecular Oncology and Center for Cancer Biology, School of Basic Medical Sciences, Tsinghua University, Beijing, 100084, China; SXMU-Tsinghua Collaborative Innovation Center for Frontier Medicine, Shanxi Medical University, Taiyuan, Shanxi Province, 030001, China.
| |
Collapse
|
20
|
Xu Y, Qi Y, Lu Z, Tan Y, Chen D, Luo H. Navigating precision: the crucial role of next-generation sequencing recurrence risk assessment in tailoring adjuvant therapy for hormone receptor-positive, human epidermal growth factor Receptor2-negative early breast cancer. Cancer Biol Ther 2024; 25:2405060. [PMID: 39304993 PMCID: PMC11418226 DOI: 10.1080/15384047.2024.2405060] [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: 07/22/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 09/25/2024] Open
Abstract
Hormone receptor-positive (HR+)/human epidermal growth factor receptor 2-negative (HER2-) breast cancer is the most common subtype, representing over two-thirds of new diagnoses. Adjuvant therapy, which encompasses various medications and treatment durations, is the standard approach for managing early stage HR+ HER2- breast cancer. Optimizing treatment is essential to minimize unnecessary side effects while addressing the biological variability inherent in HR+/HER2- breast cancers. Incorporating biological biomarkers into treatment decisions, alongside traditional clinical factors, is vital. Gene expression assays can identify patients unlikely to benefit from adjuvant chemotherapy, thereby refining treatment strategies and improving risk assessment. This paper reviews evidence for several genomic tests, including Oncotype DX, MammaPrint, Breast Cancer Index, RucurIndex, and EndoPredict, which assist in tailoring adjuvant therapy. Additionally, we explore the role of liquid biopsies in personalizing treatment, emphasizing the importance of considering late relapse risks and potential benefits of extended systemic therapy for HR+/HER2- breast cancer patients.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Female
- Chemotherapy, Adjuvant/methods
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Risk Assessment/methods
- Neoplasm Recurrence, Local/genetics
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- High-Throughput Nucleotide Sequencing/methods
- Precision Medicine/methods
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
Collapse
Affiliation(s)
- Ying Xu
- Department of Obestetrics and Gynecology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China
| | - Yingxue Qi
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd. Nanjing Simcere Medical Laboratory Science Co. Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Zhongyu Lu
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd. Nanjing Simcere Medical Laboratory Science Co. Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Yuan Tan
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd. Nanjing Simcere Medical Laboratory Science Co. Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
| | - Dongsheng Chen
- The Medical Department, Jiangsu Simcere Diagnostics Co. Ltd. Nanjing Simcere Medical Laboratory Science Co. Ltd., The State Key Laboratory of Neurology and Oncology Drug Development, Nanjing, China
- Cancer Center, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
- Center of Translational Medicine, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Haijun Luo
- Department of Pathology, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
| |
Collapse
|
21
|
Mo M, Hou C, Yuan H, Zhao R, Chen M, Jiang Y, Xu K, Zhang T, Chen X, Suo C. Shared genetic factors and the interactions with fresh fruit intake contributes to four types squamous cell carcinomas. PLoS One 2024; 19:e0316087. [PMID: 39739889 DOI: 10.1371/journal.pone.0316087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 12/05/2024] [Indexed: 01/02/2025] Open
Abstract
Studies have reported risk factors for a single-squamous cell carcinoma(Single-SCCs). However, the shared common germline genetic factors and environmental factors have not been well elucidated with respect to augmented risk of pan-squamous cell carcinoma(Pan-SCCs). By integrating a large-scale genotype data of 1,928 Pan-SCCs cases and 7,712 age- and sex-matched controls in the UK Biobank cohort, as well as multiple transcriptome and protein databases, we conducted a multi-omics analysis. Genome-wide association analysis (GWAS) was used to identify genetic susceptibility loci of SCCs. High resolution human leucocyte antigen (HLA) alleles and corresponding amino acid sequences were imputed using SNP2HLA and tested for association with SCCs. Credible risk variants (CRVs) were combined risk SNPs reported in GWAS Catalog and our study, followed by comprehensive bioinformatics analyses. We identified six novel index SNPs in the progression of SCCs, which were also strongly interacted with fresh fruit intake. Moreover, our study systematically characterize the HLA variants and their relationship to SCCs susceptibility. We identified HLA-A*01 and six HLA-A amino acid position were associated independently with SCCs. Credible risk variants were annotated to 469 target genes, further GO and KEGG Pathway Enrichment Analysis showed that SCCs genes were primarily involved in immune-related pathways, espechially regulated by HLA region. The transcriptome analysis showed that there were 270 differentially expressed genes(DEGs), with the upregulated genes were enriched in the regulation of stem cell differentiation, proliferation, development, and maintenance. The PPI Network and Modular Analysis uncovered the Keratin(KRT) genes may serve as a potential marker in SCCs. Our results illustrate the molecular basis of both well-studied and new susceptibility loci of SCCs, providing not only novel insights into the genetic commonality among SCCs but also a set of plausible gene targets for post-GWAS functional experiments.
Collapse
Affiliation(s)
- Mengqing Mo
- Department of Epidemiology & Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
- Department of Outpatient Office, Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Can Hou
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huangbo Yuan
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, Human Phenome Institute, Fudan University, Shanghai, China
| | - Renjia Zhao
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, Human Phenome Institute, Fudan University, Shanghai, China
| | - Mingyang Chen
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, Human Phenome Institute, Fudan University, Shanghai, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Kelin Xu
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, China
| | - Tiejun Zhang
- Department of Epidemiology & Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Zhangjiang Fudan International Innovation Center, Human Phenome Institute, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
- Yiwu Research Institute of Fudan University, Yiwu, Zhejiang, China
| | - Chen Suo
- Department of Epidemiology & Ministry of Education Key Laboratory of Public Health Safety, School of Public Health, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
- Shanghai Institute of Infectious Disease and Biosecurity, Shanghai, China
| |
Collapse
|
22
|
Zhang D, Wang J, Cai S, Surtihadi J. Skewness-Corrected Confidence Intervals for Predictive Values in Enrichment Studies. Stat Med 2024; 43:5862-5871. [PMID: 39567245 DOI: 10.1002/sim.10283] [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: 08/11/2023] [Revised: 08/22/2024] [Accepted: 10/30/2024] [Indexed: 11/22/2024]
Abstract
The positive predictive value (PPV) and negative predictive value (NPV) can be expressed as functions of disease prevalence (ρ $$ \rho $$ ) and the ratios of two binomial proportions (ϕ $$ \phi $$ ), whereϕ ppv = 1 - specificity sensitivity $$ {\phi}_{ppv}=\frac{1- specificity}{sensitivity} $$ andϕ npv = 1 - sensitivity specificity $$ {\phi}_{npv}=\frac{1- sensitivity}{specificity} $$ . In prospective studies, where the proportion of subjects with the disease in the study cohort is an unbiased estimate of the disease prevalence, the confidence intervals (CIs) of PPV and NPV can be estimated using established methods for single proportion. However, in enrichment studies, such as case-control studies, where the proportion of diseased subjects significantly differs from disease prevalence, estimating CIs for PPV and NPV remains a challenge in terms of skewness and overall coverage, especially under extreme conditions (e.g.,NPV = 1 $$ \mathrm{NPV}=1 $$ ). In this article, we extend the method adopted by Li, where CIs for PPV and NPV were derived from those ofϕ $$ \phi $$ . We explored additional CI methods forϕ $$ \phi $$ , including those by Gart & Nam (GN), MoverJ, and Walter and convert their corresponding CIs for PPV and NPV. Through simulations, we compared these methods with established CI methods, Fieller, Pepe, and Delta in terms of skewness and overall coverage. While no method proves universally optimal, GN and MoverJ methods generally emerge as recommended choices.
Collapse
Affiliation(s)
- Dadong Zhang
- Biostatistics, Illumina Inc., San Diego, California, USA
| | - Jingye Wang
- Biostatistics, Illumina Inc., San Diego, California, USA
| | - Suqin Cai
- Biostatistics, Illumina Inc., San Diego, California, USA
| | | |
Collapse
|
23
|
Kavgaci G, Sahin TK, Muderrisoglu T, Ileri S, Guven DC, Aksoy S. Post-operative serum CEA predicts prognosis in HR-positive/HER2-negative early breast cancer. Expert Rev Anticancer Ther 2024; 24:1319-1326. [PMID: 39673491 DOI: 10.1080/14737140.2024.2443009] [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: 09/10/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND The prognostic role of preoperative carcinoembryonic antigen (CEA) in breast cancer is recognized, but the impact of postoperative CEA levels on survival in early breast cancer is uncertain. RESEARCH DESIGN AND METHODS We conducted a retrospective study of 921 non-metastatic breast cancer patients treated at anonymized. Patients were categorized as normal (CEA ≤3 µg/L) or elevated (CEA >3 µg/L). RESULTS Elevated postoperative CEA levels were associated with shorter disease-free survival (DFS) (median, 174.6 vs. 239.8 months; hazard ratio (HR): 1.80; 95% confidence interval (CI): 1.27-2.56; p < 0.001) and overall survival (OS) (median, 174.6 vs. 261.1 months; HR:2.34; 95% CI: 1.59-3.45; p < 0.001). Elevated CEA was associated with shorter DFS (median, 174.6 months vs. not reached (NR); HR:2.30; 95% CI: 1.03-5.19; p = 0.043) and OS (NR vs. NR; HR: 2.81; 95% CI: 1.06-7.48; p = 0.039) in stage 1, shorter DFS (median, 239. 8 vs. 141.1 months; HR: 1.95; 95% CI: 1.28-2.98; p = 0.002) and OS (median, 169 vs. 261.1 months; HR: 2.56; 95% CI: 1.6-4.12; p < 0.001) in stage 2 and shorter OS (median, 65 vs. 183.1 months; HR: 3.25; 95% CI: 1.19-8.83; p = 0.021) in stage 3. CONCLUSIONS Elevated postoperative CEA indicates worse DFS and OS in patients with HR-positive/HER2-negative early breast cancer.
Collapse
Affiliation(s)
- Gozde Kavgaci
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkiye
| | - Taha Koray Sahin
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkiye
| | - Tugcenur Muderrisoglu
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkiye
| | - Serez Ileri
- Department of Internal Medicine, Hacettepe University Faculty of Medicine, Ankara, Turkiye
| | - Deniz Can Guven
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkiye
| | - Sercan Aksoy
- Department of Medical Oncology, Hacettepe University Cancer Institute, Ankara, Turkiye
| |
Collapse
|
24
|
Palmal S, Saha S, Arya N, Tripathy S. CAGCL: Predicting Short- and Long-Term Breast Cancer Survival With Cross-Modal Attention and Graph Contrastive Learning. IEEE J Biomed Health Inform 2024; 28:7382-7391. [PMID: 39236140 DOI: 10.1109/jbhi.2024.3449756] [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: 09/07/2024]
Abstract
In breast cancer treatment, accurately predicting how long a patient will survive is crucial for decision-making. This information guides treatment choices and supports patients' psychological recovery. To address this challenge, we introduce a novel predictive model to forecast breast cancer prognosis by leveraging diverse data sources, including clinical records, copy number variation, gene expressions, DNA methylation, microRNA (miRSeq) sequencing, and whole slide image data from the TCGA Database. The methodology incorporates graph contrastive learning with cross-modality attention (CAGCL), considering all possible combinations of the six distinct data modalities. Feature embeddings are enhanced through graph contrastive learning, which identifies subtle differences and similarities among samples. Further, to learn the complementary nature of information across multiple data modalities, a cross-attention framework is proposed and applied to the graph contrastive learning-based extracted features from various data sources for breast cancer survival prediction. It performs a binary classification to anticipate the likelihood of short- and long-term breast cancer survivors, delineated by a five-year threshold. The proposed model (CAGCL) showcases superior performance compared to baseline models and other state-of-the-art models. The model attains an accuracy of 0.932, a sensitivity of 0.954, a precision of 0.958, an F1 score of 0.956, and an AUC of 0.948, underscoring its effectiveness in predicting breast cancer survival.
Collapse
|
25
|
Derakhshandeh R, Zhu Y, Li J, Hester D, Younis R, Koka R, Jones LP, Sun W, Goloubeva O, Tkaczuk K, Bates J, Reader J, Webb TJ. Identification of Functional Immune Biomarkers in Breast Cancer Patients. Int J Mol Sci 2024; 25:12309. [PMID: 39596374 PMCID: PMC11595306 DOI: 10.3390/ijms252212309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer immunotherapy has emerged as an effective, personalized treatment for certain patients, particularly for those with hematological malignancies. However, its efficacy in breast cancer has been marginal-perhaps due to cold, immune-excluded, or immune-desert tumors. Natural killer T (NKT) cells play a critical role in cancer immune surveillance and are reduced in cancer patients. Thus, we hypothesized that NKT cells could serve as a surrogate marker for immune function. In order to assess which breast cancer patients would likely benefit from immune cell-based therapies, we have developed a quantitative method to rapidly assess NKT function using stimulation with artificial antigen presenting cells followed by quantitative real-time PCR for IFN-γ. We observed a significant reduction in the percentage of circulating NKT cells in breast cancer patients, compared to healthy donors; however, the majority of patients had functional NKT cells. When we compared BC patients with highly functional NKT cells, as indicated by high IFN-γ induction, to those with little to no induction, following stimulation of NKT cells, there was no significant difference in NKT cell number between the groups, suggesting functional loss has more impact than physical loss of this subpopulation of T cells. In addition, we assessed the percentage of tumor-infiltrating lymphocytes and PD-L1 expression within the tumor microenvironment in the low and high responders. Further characterization of immune gene signatures in these groups identified a concomitant decrease in the induction of TNFα, LAG3, and LIGHT in the low responders. We next investigated the mechanisms by which breast cancers suppress NKT-mediated anti-tumor immune responses. We found that breast cancers secrete immunosuppressive lipids, and treatment with commonly prescribed medications that modulate lipid metabolism, can reduce tumor growth and restore NKT cell responses.
Collapse
Affiliation(s)
- Roshanak Derakhshandeh
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Yuyi Zhu
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Junxin Li
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Danubia Hester
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Rania Younis
- Department of Oncology and Diagnostic Sciences, University of Maryland School of Dentistry, Baltimore, MD 21201, USA;
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
| | - Rima Koka
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Laundette P. Jones
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Wenji Sun
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Olga Goloubeva
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
- Department of Epidemiology and Public Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Katherine Tkaczuk
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
| | - Joshua Bates
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
| | - Jocelyn Reader
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
- Department of Pharmaceutical Sciences, University of Maryland Eastern Shore, Princess Anne, MD 21853, USA
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Tonya J. Webb
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (R.D.); (Y.Z.); (J.L.); (D.H.); (W.S.); (J.B.)
- Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland, Baltimore, Baltimore, MD 21201, USA; (R.K.); (L.P.J.); (O.G.); (K.T.); (J.R.)
| |
Collapse
|
26
|
Generali D, Rocca A, Strina C, Milani M, Fiorino E, Cervoni V, Azzini C, Saracino A, Ciliberto I, Ziglioli N, Alberio M, Giudici F, Dester M, Ciani O, Fornaro G, Aguggini S, Dreezen C, Pronin D, Ende S. Assessing the long-term prognostic ability of the 70 gene expression signature MammaPrint in an Italian single-center prospective cohort study of early-stage intermediate-risk breast cancer patients. Heliyon 2024; 10:e39485. [PMID: 39553665 PMCID: PMC11564935 DOI: 10.1016/j.heliyon.2024.e39485] [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: 09/19/2024] [Revised: 10/09/2024] [Accepted: 10/15/2024] [Indexed: 11/19/2024] Open
Abstract
Purpose The aim of this study was to assess the prognostic performance of the 70-gene signature, MammaPrint, in an Italian single-center prospective cohort of early-stage intermediate-risk breast cancer (BC) patients. Methods A total of 195 eligible early BC cases were tested for genomic risk between 2006 and 2013. In this retrospective analysis, the association of genomic risk with distant metastasis-free survival (DMFS) and overall survival (OS) were assessed using Cox regression models, adjusting for clinical and pathological tumor characteristics. Results MammaPrint identified 118 (60.5 %) patients with genomically Low Risk tumors and 77 (39.5 %) patients with genomically High Risk tumors. Age, menopausal status, tumor size, receptor status, and nodal status were comparable between MammaPrint Risk categories. The median follow-up was 8.4 years for DMFS and 9.3 years for OS; 8-year follow-up was reported for both endpoints. The 8-year DMFS was 90.4 % (95 % CI 84.9-95.9) in patients with MammaPrint Low Risk tumors compared to 60.8 % (95 % CI 49.8-71.8) for patients with High Risk tumors. Patients with MammaPrint Low Risk tumors exhibited significantly superior 8-year OS (97.3 %; 95 % CI 94.4-100) compared with MammaPrint High Risk tumors (89.5 %; 95 % CI 82.6-96.4; p = 0.028). Multivariate analyses identified MammaPrint as significantly associated with 8-year DMFS and MammaPrint together with Progesterone Receptor positivity with 8-year OS. Conclusion The prognostic performance of MammaPrint was demonstrated in early-stage clinically intermediate to high-risk BC patients. Moreover, patients with MammaPrint Low Risk tumors had good outcome regardless of treatment regimen, thus supporting personalized treatment choices.
Collapse
Affiliation(s)
- Daniele Generali
- Multidisciplinary Unit of Breast Cancer, ASST of Cremona, Cremona, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Italy
| | - Andrea Rocca
- Department of Medical, Surgical and Health Sciences, University of Trieste, Italy
| | - Carla Strina
- Multidisciplinary Unit of Breast Cancer, ASST of Cremona, Cremona, Italy
| | - Manuela Milani
- Multidisciplinary Unit of Breast Cancer, ASST of Cremona, Cremona, Italy
| | - Enrico Fiorino
- Department of Medical, Surgical and Health Sciences, University of Trieste, Italy
| | - Valeria Cervoni
- Multidisciplinary Unit of Breast Cancer, ASST of Cremona, Cremona, Italy
| | - Carlo Azzini
- Multidisciplinary Unit of Breast Cancer, ASST of Cremona, Cremona, Italy
| | - Antonella Saracino
- Multidisciplinary Unit of Breast Cancer, ASST of Cremona, Cremona, Italy
| | - Ingnazio Ciliberto
- Multidisciplinary Unit of Breast Cancer, ASST of Cremona, Cremona, Italy
| | - Nicoletta Ziglioli
- Multidisciplinary Unit of Breast Cancer, ASST of Cremona, Cremona, Italy
| | - Marzia Alberio
- Multidisciplinary Unit of Breast Cancer, ASST of Cremona, Cremona, Italy
| | - Fabiola Giudici
- Cancer Epidemiology Unit, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Martina Dester
- Multidisciplinary Unit of Breast Cancer, ASST of Cremona, Cremona, Italy
| | - Oriana Ciani
- Centre for Research on Health and Social Care Management, SDA Bocconi, Milan, Italy
| | - Giulia Fornaro
- Centre for Research on Health and Social Care Management, SDA Bocconi, Milan, Italy
| | - Sergio Aguggini
- Multidisciplinary Unit of Breast Cancer, ASST of Cremona, Cremona, Italy
| | | | | | | |
Collapse
|
27
|
Guo R, Wang R, Zhang W, Li Y, Wang Y, Wang H, Li X, Song J. Mechanisms of Action of HSP110 and Its Cognate Family Members in Carcinogenesis. Onco Targets Ther 2024; 17:977-989. [PMID: 39553399 PMCID: PMC11568853 DOI: 10.2147/ott.s496403] [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: 09/15/2024] [Accepted: 10/27/2024] [Indexed: 11/19/2024] Open
Abstract
Tumors, as chronic malignant diseases that account for about 20% of all deaths worldwide, are the number one threat to human health. Until now there is no reliable treatment for most types of tumors. Tumorigenesis and cellular carcinogenesis remain difficult challenges due to their complex etiology and unknown mechanisms. As stress process regulating molecules and protein folding promoters, heat shock proteins (HSPs) play an important role in cancer development. Most studies have shown that HSPs are one of the major anticancer drug targets. HSPs are not only modulators of the cellular stress response, but are also closely associated with tumor initiation, progression, and drug resistance, so understanding the mechanism of the HSP family involved in cellular carcinogenesis is an important part of understanding tumorigenesis and enabling anticancer drug development. In this review, we discuss the functions and mechanisms of key members of the HSP family (HSP70, HSP90, and HSP110) in participating in the process of tumorigenesis and cell carcinogenesis, and look forward to the prospect of key members of the HSP family in targeted cancer therapy.
Collapse
Affiliation(s)
- Rongqi Guo
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, 224000, People’s Republic of China
- Medical School of Nantong University, Nantong, 226007, People’s Republic of China
| | - Rui Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, 224000, People’s Republic of China
- Medical School of Nantong University, Nantong, 226007, People’s Republic of China
| | - Weisong Zhang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, 224000, People’s Republic of China
- Medical School of Nantong University, Nantong, 226007, People’s Republic of China
| | - Yangyang Li
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, 224000, People’s Republic of China
- Medical School of Nantong University, Nantong, 226007, People’s Republic of China
| | - Yihao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, 224000, People’s Republic of China
- Medical School of Nantong University, Nantong, 226007, People’s Republic of China
| | - Hao Wang
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, 224000, People’s Republic of China
- Medical School of Nantong University, Nantong, 226007, People’s Republic of China
| | - Xia Li
- Department of General Medicine, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, 224000, People’s Republic of China
| | - Jianxiang Song
- Department of Thoracic Surgery, Affiliated Hospital 6 of Nantong University, Yancheng Third People’s Hospital, Yancheng, 224000, People’s Republic of China
- Medical School of Nantong University, Nantong, 226007, People’s Republic of China
| |
Collapse
|
28
|
Mehta K, Sharma A, Mehta A, Tayal J. Enhancing negative control selection: A comparative analysis of random and targeted sampling techniques for obtaining High-Quality RNA from normal breast tissue. Biol Methods Protoc 2024; 9:bpae083. [PMID: 39659669 PMCID: PMC11631398 DOI: 10.1093/biomethods/bpae083] [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: 08/14/2024] [Revised: 10/21/2024] [Accepted: 11/01/2024] [Indexed: 12/12/2024] Open
Abstract
Molecular profiling is a crucial aspect of cancer therapy selection, underscoring the necessity for representative sampling of both tumor and normal tissues. While much attention has been given to representative tumor sampling, there has been a notable lack of exploration into the issue of poor RNA quality in normal breast tissue processing. Normal breast tissue from the same patient is often used as a negative control for most "-omics" experiments. RNA extracted from normal breast tissues frequently contains nucleic acids from surrounding adipocytes, endothelial cells, and immune cells, leading to a low representation of ductal elements and skewed results. Therefore, ensuring a complete representation of breast glandular tissue is imperative. The study aimed to investigate the variations in RNA enrichment between a random sampling technique and a targeted sampling approach when visually selecting normal breast tissue sections as negative controls for "-omics" experiments. Fifteen female breast cancer subjects who underwent Modified Radical Mastectomy were selected for the study. Normal Breast tissue was visually examined, and samples were collected from random fat pockets (random sampling) and fibromuscular grey-white streak areas (targeted sampling). RNA was isolated, followed by spectrophotometric analysis, agarose gel electrophoresis and Agilent Tape station analysis. Histopathological assessments and a gene expression study for housekeeping genes were performed on both subsets. Tissues collected through targeted sampling exhibited significantly higher RNA quality than those obtained via random sampling. Histopathological analysis revealed cellular areas abundant in terminal ductular units within the targeted samples, and a final validation qPCR showed that the targeted samples were the most representative of normal breast glandular tissue. The comparative analysis of the two sampling methods clearly indicates that the targeted approach, with its superior accuracy and reliability, is the more practical choice for obtaining representative normal breast glandular tissue for "-omics" experiments.
Collapse
Affiliation(s)
- Komal Mehta
- Biorepository, Department of Research, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi 110085, India
| | - Archana Sharma
- Biorepository, Department of Research, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi 110085, India
| | - Anurag Mehta
- Histopathology, Department of Laboratory Services, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi 110085, India
| | - Juhi Tayal
- Biorepository, Department of Research, Rajiv Gandhi Cancer Institute and Research Centre, New Delhi 110085, India
| |
Collapse
|
29
|
van 't Veer LJ, Meershoek-Klein Kranenbarg E, Duijm-de Carpentier M, Van de Velde CJH, Kleijn M, Dreezen C, Menicucci AR, Audeh W, Liefers GJ. Selection of Patients With Early-Stage Breast Cancer for Extended Endocrine Therapy: A Secondary Analysis of the IDEAL Randomized Clinical Trial. JAMA Netw Open 2024; 7:e2447530. [PMID: 39602119 DOI: 10.1001/jamanetworkopen.2024.47530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
Importance There is a need for biomarkers that predict late recurrence risk and extended endocrine therapy (EET) benefit among patients with early-stage breast cancer (EBC). MammaPrint, a 70-gene expression risk-of-recurrence assay, has been found to project significant EET benefit in patients with assay-classified low-risk tumors. Objective To determine the test's utility in identifying which patients with EBC in the IDEAL (Investigation on the Duration of Extended Adjuvant Letrozole) trial could benefit from 5-year vs 2.5-year letrozole treatment. Design, Setting, and Participants This secondary analysis of the IDEAL randomized clinical trial evaluated postmenopausal women with hormone receptor-positive EBC who were assigned to either 2.5 or 5 years of EET, with 10 years of follow-up after randomization. A 70-gene assay was used to classify tumors as high, low, or ultralow risk. Adverse event (AE) frequency and treatment compliance were evaluated. Statistical analyses were performed from April 2022 to September 2024. Interventions After 5 years of endocrine therapy, patients were randomized to 2.5 or 5 years of EET with letrozole. Main Outcomes and Measures Primary end point was distant recurrence (DR). Cox proportional hazard regression models and likelihood ratios tested the interaction between treatment and gene expression assay. Results Among 515 women included (mean [SD] age at randomization, 59.9 [9.5] years), 265 were in the 2.5-year treatment arm and 250 in the 5-year treatment arm. Of these patients, 223 (43.3%) patients with 70-gene assay-classified low-risk tumors had a significant absolute benefit of 10.1% for DR (hazard ratio, 0.32; 95% CI, 0.12-0.87; P = .03). Treatment interaction was not significant for DR. Of patients with either 70-gene assay-classified high-risk tumors (259 [50.3%]) or ultralow risk tumors (33 [6.4%]), 5 years vs 2.5 years of EET was not associated with improved benefit for DR. As expected, rates of AEs and treatment discontinuation were comparable among the different 70-gene assay risk groups in each treatment arm. Conclusions and Relevance This secondary analysis of the IDEAL trial found that the 70-gene assay identified patients with low-risk tumors who could benefit from 5-year vs 2.5-year EET. These findings suggest that this gene expression assay could go beyond guiding neoadjuvant and adjuvant chemotherapy decisions to informing the optimal duration of adjuvant endocrine therapy. Trial Registration EU Clinical Trials Register Eudra CT: 2006-003958-16.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Gerrit-Jan Liefers
- Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
30
|
Liao Y, Fu J, Lu X, Qian Z, Yu Y, Zhu L, Pan J, Li P, Zhu Q, Li X, Sun W, Wang X, Cao W. High chromosomal instability is associated with higher 10-year risks of recurrence for hormone receptor-positive, human epidermal growth factor receptor 2-negative breast cancer patients: clinical evidence from a large-scale, multiple-site, retrospective study. J Pathol Clin Res 2024; 10:e70011. [PMID: 39545625 PMCID: PMC11565440 DOI: 10.1002/2056-4538.70011] [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: 06/17/2024] [Revised: 10/06/2024] [Accepted: 10/24/2024] [Indexed: 11/17/2024]
Abstract
Long-term survival varies among hormone receptor-positive (HR+) and human epidermal growth factor receptor 2-negative (HER2-) breast cancer patients and is seriously impaired by metastasis. Chromosomal instability (CIN) was one of the key drivers of breast cancer metastasis. Here we evaluate CIN and 10-year invasive disease-free survival (iDFS) and overall survival (OS) in HR+/HER2-- breast cancer. In this large-scale, multiple-site, retrospective study, 354 HR+/HER2- breast cancer patients were recruited. Of these, 204 patients were used for internal training, 70 for external validation, and 80 for cross-validation. All medical records were carefully reviewed to obtain the disease recurrence information. Formalin-fixed paraffin-embedded tissue samples were collected, followed by low-pass whole-genome sequencing with a median genome coverage of 1.86X using minimal 1 ng DNA input. CIN was then assessed using a customized bioinformatics workflow. Three or more instances of CIN per sample was defined as high CIN and the frequency was 42.2% (86/204) in the internal cohort. High CIN correlated significantly with increased lymph node metastasis, vascular invasion, progesterone receptor negative status, HER2 low, worse pathological type, and performed as an independent prognostic factor for HR+/- breast cancer. Patients with high CIN had shorter iDFS and OS than those with low CIN [10-year iDFS 11.1% versus 82.2%, hazard ratio (HR) = 11.12, p < 0.01; 10-year OS 45.7% versus 94.3%, HR = 14.17, p < 0.01]. These findings were validated in two external cohorts with 70 breast cancer patients. Moreover, high CIN could predict the prognosis more accurately than Adjuvant! Online score (10-year iDFS 11.1% versus 48.6%, HR = 2.71, p < 0.01). Cross-validation analysis found that high consistency (83.8%) was observed between CIN and MammaPrint score, while only 45% between CIN and Adjuvant! Online score. In conclusion, high CIN is an independent prognostic indicator for HR+/HER2- breast cancer with shorter iDFS and OS and holds promise for predicting recurrence and metastasis.
Collapse
MESH Headings
- Humans
- Breast Neoplasms/pathology
- Breast Neoplasms/genetics
- Breast Neoplasms/mortality
- Female
- Retrospective Studies
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/analysis
- Middle Aged
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/genetics
- Chromosomal Instability
- Adult
- Aged
- Receptors, Progesterone/metabolism
- Receptors, Progesterone/analysis
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/analysis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/analysis
- Disease-Free Survival
- Risk Factors
Collapse
Affiliation(s)
- Yu‐Yang Liao
- Department of Breast Medical OncologyZhejiang Cancer HospitalHangzhouPR China
- Postgraduate Training Base Alliance of Wenzhou Medical UniversityZhejiang Cancer HospitalHangzhouPR China
| | - Jianfei Fu
- Department of Medical Oncology, Affiliated Jinhua HospitalZhejiang University School of Medicine (Jinhua Municipal Central Hospital)JinhuaPR China
| | - Xiang Lu
- Department of Breast DiseaseAffiliated Hospital of Jiaxing University (First Hospital of Jiaxing)JiaxingPR China
| | | | - Yang Yu
- Department of Breast SurgeryZhejiang Cancer HospitalHangzhouPR China
| | - Liang Zhu
- Department of PathologyZhejiang Cancer HospitalHangzhouPR China
| | - Jia‐Ni Pan
- Department of Breast Medical OncologyZhejiang Cancer HospitalHangzhouPR China
| | - Pu‐Chun Li
- Department of Breast Medical OncologyZhejiang Cancer HospitalHangzhouPR China
- Postgraduate Training Base Alliance of Wenzhou Medical UniversityZhejiang Cancer HospitalHangzhouPR China
| | - Qiao‐Yan Zhu
- Department of Breast Medical OncologyZhejiang Cancer HospitalHangzhouPR China
- The Second Clinical Medical College of Zhejiang Chinese Medical UniversityHangzhouPR China
| | - Xiaolin Li
- Department of Breast Medical OncologyZhejiang Cancer HospitalHangzhouPR China
| | - Wenyong Sun
- Department of PathologyZhejiang Cancer HospitalHangzhouPR China
| | - Xiao‐Jia Wang
- Department of Breast Medical OncologyZhejiang Cancer HospitalHangzhouPR China
| | - Wen‐Ming Cao
- Department of Breast Medical OncologyZhejiang Cancer HospitalHangzhouPR China
| |
Collapse
|
31
|
Ooko E, Ali NT, Efferth T. Identification of Cuproptosis-Associated Prognostic Gene Expression Signatures from 20 Tumor Types. BIOLOGY 2024; 13:793. [PMID: 39452102 PMCID: PMC11505359 DOI: 10.3390/biology13100793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024]
Abstract
We investigated the mRNA expression of 124 cuproptosis-associated genes in 7489 biopsies from 20 different tumor types of The Cancer Genome Atlas (TCGA). The KM plotter algorithm has been used to calculate Kaplan-Meier statistics and false discovery rate (FDR) corrections. Interaction networks have been generated using Ingenuity Pathway Analysis (IPA). High mRNA expression of 63 out of 124 genes significantly correlated with shorter survival times of cancer patients across all 20 tumor types. IPA analyses revealed that their gene products were interconnected in canonical pathways (e.g., cancer, cell death, cell cycle, cell signaling). Four tumor entities showed a higher accumulation of genes than the other cancer types, i.e., renal clear cell carcinoma (n = 21), renal papillary carcinoma (n = 13), kidney hepatocellular carcinoma (n = 13), and lung adenocarcinoma (n = 9). These gene clusters may serve as prognostic signatures for patient survival. These signatures were also of prognostic value for tumors with high mutational rates and neoantigen loads. Cuproptosis is of prognostic significance for the survival of cancer patients. The identification of specific gene signatures deserves further exploration for their clinical utility in routine diagnostics.
Collapse
Affiliation(s)
- Ednah Ooko
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA;
- Department of Biological Sciences, School of Natural and Applied Sciences, Masinde Muliro University of Science and Technology, Kakamega 190-50100, Kenya
| | - Nadeen T. Ali
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany;
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Staudinger Weg 5, 55128 Mainz, Germany;
| |
Collapse
|
32
|
Torland LA, Lai X, Kumar S, Riis MH, Geisler J, Lüders T, Tekpli X, Kristensen V, Sahlberg K, Tahiri A. Benign breast tumors may arise on different immunological backgrounds. Mol Oncol 2024; 18:2495-2509. [PMID: 38757377 PMCID: PMC11459044 DOI: 10.1002/1878-0261.13655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 12/21/2023] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
Benign breast tumors are a nonthreatening condition defined as abnormal cell growth within the breast without the ability to invade nearby tissue. However, benign lesions hold valuable biological information that can lead us toward better understanding of tumor biology. In this study, we have used two pathway analysis algorithms, Pathifier and gene set variation analysis (GSVA), to identify biological differences between normal breast tissue, benign tumors and malignant tumors in our clinical dataset. Our results revealed that one-third of all pathways that were significantly different between benign and malignant tumors were immune-related pathways, and 227 of them were validated by both methods and in the METABRIC dataset. Furthermore, five of these pathways (all including genes involved in cytokine and interferon signaling) were related to overall survival in cancer patients in both datasets. The cellular moieties that contribute to immune differences in malignant and benign tumors were analyzed using the deconvolution tool, CIBERSORT. The results showed that levels of some immune cells were specifically higher in benign than in malignant tumors, and this was especially the case for resting dendritic cells and follicular T-helper cells. Understanding the distinct immune profiles of benign and malignant breast tumors may aid in developing noninvasive diagnostic methods to differentiate between them in the future.
Collapse
Affiliation(s)
- Lilly Anne Torland
- Department of Clinical Molecular Biology (EpiGen)Akershus University HospitalLørenskogNorway
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
- Department of Research and InnovationVestre Viken HF, Drammen HospitalNorway
| | - Xiaoran Lai
- Oslo Centre for Biostatistics and Epidemiology, Faculty of MedicineUniversity of OsloNorway
| | - Surendra Kumar
- Department of Ocean SciencesMemorial University of NewfoundlandSt. John'sCanada
| | - Margit H. Riis
- Department of Breast and Endocrine Surgery, Clinic of CancerOslo University HospitalNorway
| | - Jürgen Geisler
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
- Department of OncologyAkershus University HospitalLørenskogNorway
| | - Torben Lüders
- Department of Clinical Molecular Biology (EpiGen)Akershus University HospitalLørenskogNorway
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
| | - Xavier Tekpli
- Department of Medical GeneticsOslo University HospitalOsloNorway
| | - Vessela Kristensen
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
- Department of Medical GeneticsOslo University HospitalOsloNorway
| | - Kristine Sahlberg
- Institute of Clinical Medicine, Faculty of MedicineUniversity of OsloNorway
- Department of Research and InnovationVestre Viken HF, Drammen HospitalNorway
| | - Andliena Tahiri
- Department of Clinical Molecular Biology (EpiGen)Akershus University HospitalLørenskogNorway
- Department of Research and InnovationVestre Viken HF, Drammen HospitalNorway
| |
Collapse
|
33
|
Mathur A, Arya N, Pasupa K, Saha S, Roy Dey S, Saha S. Breast cancer prognosis through the use of multi-modal classifiers: current state of the art and the way forward. Brief Funct Genomics 2024; 23:561-569. [PMID: 38688724 DOI: 10.1093/bfgp/elae015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 03/01/2024] [Accepted: 04/09/2024] [Indexed: 05/02/2024] Open
Abstract
We present a survey of the current state-of-the-art in breast cancer detection and prognosis. We analyze the evolution of Artificial Intelligence-based approaches from using just uni-modal information to multi-modality for detection and how such paradigm shift facilitates the efficacy of detection, consistent with clinical observations. We conclude that interpretable AI-based predictions and ability to handle class imbalance should be considered priority.
Collapse
Affiliation(s)
- Archana Mathur
- Department of Information Science and Engineering, Nitte Meenakshi Institute of Technology, Yelahanka, 560064, Karnataka, India
| | - Nikhilanand Arya
- School of Computer Engineering, Kalinga Institute of Industrial Technology, Deemed to be University, Bhubaneshwar, 751024, Odisha, India
| | - Kitsuchart Pasupa
- School of Information Technology, King Mongkut's Institute of Technology Ladkrabang, 1 Soi Chalongkrung 1, 10520, Bangkok, Thailand
| | - Sriparna Saha
- Computer Science and Engineering, Indian Institute of Technology Patna, Bihta, 801106, Bihar, India
| | - Sudeepa Roy Dey
- Department of Computer Science and Engineering, PES University, Hosur Road, 560100, Karnataka, India
| | - Snehanshu Saha
- CSIS and APPCAIR, BITS Pilani K.K Birla Goa Campus, Goa, 403726, Goa, India
- Div of AI Research, HappyMonk AI, Bangalore, 560078, Karnataka, India
| |
Collapse
|
34
|
Varambally S, Karthikeyan SK, Chandrashekar D, Sahai S, Shrestha S, Aneja R, Singh R, Kleer C, Kumar S, Qin Z, Nakshatri H, Manne U, Creighton C. MammOnc-DB, an integrative breast cancer data analysis platform for target discovery. RESEARCH SQUARE 2024:rs.3.rs-4926362. [PMID: 39399665 PMCID: PMC11469468 DOI: 10.21203/rs.3.rs-4926362/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Breast cancer (BCa) is one of the most common malignancies among women worldwide. It is a complex disease that is characterized by morphological and molecular heterogeneity. In the early stages of the disease, most BCa cases are treatable, particularly hormone receptor-positive and HER2-positive tumors. Unfortunately, triple-negative BCa and metastases to distant organs are largely untreatable with current medical interventions. Recent advances in sequencing and proteomic technologies have improved our understanding of the molecular changes that occur during breast cancer initiation and progression. In this era of precision medicine, researchers and clinicians aim to identify subclass-specific BCa biomarkers and develop new targets and drugs to guide treatment. Although vast amounts of omics data including single cell sequencing data, can be accessed through public repositories, there is a lack of user-friendly platforms that integrate information from multiple studies. Thus, to meet the need for a simple yet effective and integrative BCa tool for multi-omics data analysis and visualization, we developed a comprehensive BCa data analysis platform called MammOnc-DB (http://resource.path.uab.edu/MammOnc-Home.html), comprising data from more than 20,000 BCa samples. MammOnc-DB was developed to provide a unique resource for hypothesis generation and testing, as well as for the discovery of biomarkers and therapeutic targets. The platform also provides pre- and post-treatment data, which can help users identify treatment resistance markers and patient groups that may benefit from combination therapy.
Collapse
|
35
|
Sun A, Franzmann EJ, Chen Z, Cai X. Deep contrastive learning for predicting cancer prognosis using gene expression values. Brief Bioinform 2024; 25:bbae544. [PMID: 39471411 PMCID: PMC11521346 DOI: 10.1093/bib/bbae544] [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: 06/10/2024] [Revised: 09/09/2024] [Accepted: 10/18/2024] [Indexed: 11/01/2024] Open
Abstract
Recent advancements in image classification have demonstrated that contrastive learning (CL) can aid in further learning tasks by acquiring good feature representation from a limited number of data samples. In this paper, we applied CL to tumor transcriptomes and clinical data to learn feature representations in a low-dimensional space. We then utilized these learned features to train a classifier to categorize tumors into a high- or low-risk group of recurrence. Using data from The Cancer Genome Atlas (TCGA), we demonstrated that CL can significantly improve classification accuracy. Specifically, our CL-based classifiers achieved an area under the receiver operating characteristic curve (AUC) greater than 0.8 for 14 types of cancer, and an AUC greater than 0.9 for 3 types of cancer. We also developed CL-based Cox (CLCox) models for predicting cancer prognosis. Our CLCox models trained with the TCGA data outperformed existing methods significantly in predicting the prognosis of 19 types of cancer under consideration. The performance of CLCox models and CL-based classifiers trained with TCGA lung and prostate cancer data were validated using the data from two independent cohorts. We also show that the CLCox model trained with the whole transcriptome significantly outperforms the Cox model trained with the 16 genes of Oncotype DX that is in clinical use for breast cancer patients. The trained models and the Python codes are publicly accessible and provide a valuable resource that will potentially find clinical applications for many types of cancer.
Collapse
Affiliation(s)
- Anchen Sun
- Department of Electrical and Computer Engineering, University of Miami, Miami, FL 33146, United States
| | - Elizabeth J Franzmann
- Department of Otolaryngology, University of Miami, Miami, FL 33146, United States
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33146, United States
| | - Zhibin Chen
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33146, United States
- Department of Microbiology and Immunology, University of Miami, Miami, FL 33146, United States
| | - Xiaodong Cai
- Department of Electrical and Computer Engineering, University of Miami, Miami, FL 33146, United States
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33146, United States
| |
Collapse
|
36
|
Qi YJ, Su GH, You C, Zhang X, Xiao Y, Jiang YZ, Shao ZM. Radiomics in breast cancer: Current advances and future directions. Cell Rep Med 2024; 5:101719. [PMID: 39293402 PMCID: PMC11528234 DOI: 10.1016/j.xcrm.2024.101719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/10/2024] [Accepted: 08/14/2024] [Indexed: 09/20/2024]
Abstract
Breast cancer is a common disease that causes great health concerns to women worldwide. During the diagnosis and treatment of breast cancer, medical imaging plays an essential role, but its interpretation relies on radiologists or clinical doctors. Radiomics can extract high-throughput quantitative imaging features from images of various modalities via traditional machine learning or deep learning methods following a series of standard processes. Hopefully, radiomic models may aid various processes in clinical practice. In this review, we summarize the current utilization of radiomics for predicting clinicopathological indices and clinical outcomes. We also focus on radio-multi-omics studies that bridge the gap between phenotypic and microscopic scale information. Acknowledging the deficiencies that currently hinder the clinical adoption of radiomic models, we discuss the underlying causes of this situation and propose future directions for advancing radiomics in breast cancer research.
Collapse
Affiliation(s)
- Ying-Jia Qi
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Guan-Hua Su
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xu Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
37
|
Majumdar S, Liu ST. Spatiotemporal regulation of MELK during mitosis. Front Cell Dev Biol 2024; 12:1406940. [PMID: 39355119 PMCID: PMC11443572 DOI: 10.3389/fcell.2024.1406940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/30/2024] [Indexed: 10/03/2024] Open
Abstract
Maternal Embryonic Leucine Zipper Kinase (MELK) has been studied intensively in recent years due to its overexpression in multiple cancers. However, the cell biology of MELK remains less characterized despite its well-documented association with mitosis. Here we report a distinctive pattern of human MELK that translocates from the cytoplasm to cell cortex within 3 min of anaphase onset. The cortex association lasts about 30 min till telophase. The spatiotemporal specific localization of MELK depends on the interaction between its Threonine-Proline (TP) rich domain and kinase associated 1 (KA1) domain, which is regulated by CDK1 kinase and PP4 protein phosphatase. KA1 domains are known to regulate kinase activities through various intramolecular interactions. Our results revealed a new role for KA1 domain to control subcellular localization of a protein kinase.
Collapse
Affiliation(s)
| | - Song-Tao Liu
- Department of Biological Sciences, University of Toledo, Toledo, OH, United States
| |
Collapse
|
38
|
Omar M, Harrell JC, Tamimi R, Marchionni L, Erdogan C, Nakshatri H, Ince TA. A triple hormone receptor ER, AR, and VDR signature is a robust prognosis predictor in breast cancer. Breast Cancer Res 2024; 26:132. [PMID: 39272208 PMCID: PMC11395215 DOI: 10.1186/s13058-024-01876-9] [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: 02/14/2024] [Accepted: 07/29/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Despite evidence indicating the dominance of cell-of-origin signatures in molecular tumor patterns, translating these genome-wide patterns into actionable insights has been challenging. This study introduces breast cancer cell-of-origin signatures that offer significant prognostic value across all breast cancer subtypes and various clinical cohorts, compared to previously developed genomic signatures. METHODS We previously reported that triple hormone receptor (THR) co-expression patterns of androgen (AR), estrogen (ER), and vitamin D (VDR) receptors are maintained at the protein level in human breast cancers. Here, we developed corresponding mRNA signatures (THR-50 and THR-70) based on these patterns to categorize breast tumors by their THR expression levels. The THR mRNA signatures were evaluated across 56 breast cancer datasets (5040 patients) using Kaplan-Meier survival analysis, Cox proportional hazard regression, and unsupervised clustering. RESULTS The THR signatures effectively predict both overall and progression-free survival across all evaluated datasets, independent of subtype, grade, or treatment status, suggesting improvement over existing prognostic signatures. Furthermore, they delineate three distinct ER-positive breast cancer subtypes with significant survival in differences-expanding on the conventional two subtypes. Additionally, coupling THR-70 with an immune signature identifies a predominantly ER-negative breast cancer subgroup with a highly favorable prognosis, comparable to ER-positive cases, as well as an ER-negative subgroup with notably poor outcome, characterized by a 15-fold shorter survival. CONCLUSIONS The THR cell-of-origin signature introduces a novel dimension to breast cancer biology, potentially serving as a robust foundation for integrating additional prognostic biomarkers. These signatures offer utility as a prognostic index for stratifying existing breast cancer subtypes and for de novo classification of breast cancer cases. Moreover, THR signatures may also hold promise in predicting hormone treatment responses targeting AR and/or VDR.
Collapse
Affiliation(s)
- Mohamed Omar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Dana-Farber Cancer Institute, Boston, MA, USA
| | - J Chuck Harrell
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23298, USA
| | - Rulla Tamimi
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Cihat Erdogan
- Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Harikrishna Nakshatri
- Departments of Surgery, Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Tan A Ince
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA.
- New York-Presbyterian, Brooklyn Methodist Hospital, New York, NY, USA.
| |
Collapse
|
39
|
Liu Z, Fan Y, Cui M, Wang X, Zhao P. Investigation of tumour environments through advancements in microtechnology and nanotechnology. Biomed Pharmacother 2024; 178:117230. [PMID: 39116787 DOI: 10.1016/j.biopha.2024.117230] [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: 06/05/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
Cancer has a significant negative social and economic impact on both developed and developing countries. As a result, understanding the onset and progression of cancer is critical for developing therapies that can improve the well-being and health of individuals with cancer. With time, study has revealed, the tumor microenvironment has great influence on this process. Micro and nanoscale engineering techniques can be used to study the tumor microenvironment. Nanoscale and Microscale engineering use Novel technologies and designs with small dimensions to recreate the TME. Knowing how cancer cells interact with one another can help researchers develop therapeutic approaches that anticipate and counteract cancer cells' techniques for evading detection and fighting anti-cancer treatments, such as microfabrication techniques, microfluidic devices, nanosensors, and nanodevices used to study or recreate the tumor microenvironment. Nevertheless, a complicated action just like the growth and in cancer advancement, and their intensive association along the environment around it that has to be studied in more detail.
Collapse
Affiliation(s)
- Zhen Liu
- Department of Radiology, Shengjing Hospital of China Medical University, China
| | - Yan Fan
- Department of Pediatrics, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Mengyao Cui
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xu Wang
- Department of Surgical Oncology, Breast Surgery, General Surgery, The First Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Pengfei Zhao
- Department of Radiology, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
40
|
Vahed SZ, Khatibi SMH, Saadat YR, Emdadi M, Khodaei B, Alishani MM, Boostani F, Dizaj SM, Pirmoradi S. Introducing effective genes in lymph node metastasis of breast cancer patients using SHAP values based on the mRNA expression data. PLoS One 2024; 19:e0308531. [PMID: 39150915 PMCID: PMC11329117 DOI: 10.1371/journal.pone.0308531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 07/24/2024] [Indexed: 08/18/2024] Open
Abstract
OBJECTIVE Breast cancer, a global concern predominantly impacting women, poses a significant threat when not identified early. While survival rates for breast cancer patients are typically favorable, the emergence of regional metastases markedly diminishes survival prospects. Detecting metastases and comprehending their molecular underpinnings are crucial for tailoring effective treatments and improving patient survival outcomes. METHODS Various artificial intelligence methods and techniques were employed in this study to achieve accurate outcomes. Initially, the data was organized and underwent hold-out cross-validation, data cleaning, and normalization. Subsequently, feature selection was conducted using ANOVA and binary Particle Swarm Optimization (PSO). During the analysis phase, the discriminative power of the selected features was evaluated using machine learning classification algorithms. Finally, the selected features were considered, and the SHAP algorithm was utilized to identify the most significant features for enhancing the decoding of dominant molecular mechanisms in lymph node metastases. RESULTS In this study, five main steps were followed for the analysis of mRNA expression data: reading, preprocessing, feature selection, classification, and SHAP algorithm. The RF classifier utilized the candidate mRNAs to differentiate between negative and positive categories with an accuracy of 61% and an AUC of 0.6. During the SHAP process, intriguing relationships between the selected mRNAs and positive/negative lymph node status were discovered. The results indicate that GDF5, BAHCC1, LCN2, FGF14-AS2, and IDH2 are among the top five most impactful mRNAs based on their SHAP values. CONCLUSION The prominent identified mRNAs including GDF5, BAHCC1, LCN2, FGF14-AS2, and IDH2, are implicated in lymph node metastasis. This study holds promise in elucidating a thorough insight into key candidate genes that could significantly impact the early detection and tailored therapeutic strategies for lymph node metastasis in patients with breast cancer.
Collapse
Affiliation(s)
| | - Seyed Mahdi Hosseiniyan Khatibi
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Rahat Breath and Sleep Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | | | - Manijeh Emdadi
- Department of Computer Engineering, Abadan Branch, Islamic Azad University, Abadan, Iran
| | - Bahareh Khodaei
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Matin Alishani
- Department of Computer Science, Faculty of Information Technology, University of Shahid Madani of Tabriz, Tabriz, Iran
| | - Farnaz Boostani
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Solmaz Maleki Dizaj
- Dental and Periodontal Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Saeed Pirmoradi
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
41
|
Diaz-Gimeno P, Sebastian-Leon P, Spath K, Marti-Garcia D, Sanchez-Reyes JM, Vidal MDC, Devesa-Peiro A, Sanchez-Ribas I, Martinez-Martinez A, Pellicer N, Wells D, Pellicer A. Predicting risk of endometrial failure: a biomarker signature that identifies a novel disruption independent of endometrial timing in patients undergoing hormonal replacement cycles. Fertil Steril 2024; 122:352-364. [PMID: 38518993 DOI: 10.1016/j.fertnstert.2024.03.015] [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: 05/17/2023] [Revised: 03/14/2024] [Accepted: 03/18/2024] [Indexed: 03/24/2024]
Abstract
OBJECTIVE To propose a new gene expression signature that identifies endometrial disruptions independent of endometrial luteal phase timing and predicts if patients are at risk of endometrial failure. DESIGN Multicentric, prospective study. SETTING Reproductive medicine research department in a public hospital affiliated with private fertility clinics and a reproductive genetics laboratory. PATIENTS Caucasian women (n = 281; 39.4 ± 4.8 years old with a body mass index of 22.9 ± 3.5 kg/m2) undergoing hormone replacement therapy between July 2018 and July 2021. Endometrial samples from 217 patients met RNA quality criteria for signature discovery and analysis. INTERVENTION(S) Endometrial biopsies collected in the mid-secretory phase. MAIN OUTCOME MEASURE(S) Endometrial luteal phase timing-corrected expression of 404 genes and reproductive outcomes of the first single embryo transfer (SET) after biopsy collection to identify prognostic biomarkers of endometrial failure. RESULTS Removal of endometrial timing variation from gene expression data allowed patients to be stratified into poor (n = 137) or good (n = 49) endometrial prognosis groups on the basis of their clinical and transcriptomic profiles. Significant differences were found between endometrial prognosis groups in terms of reproductive rates: pregnancy (44.6% vs. 79.6%), live birth (25.6% vs. 77.6%), clinical miscarriage (22.2% vs. 2.6%), and biochemical miscarriage (20.4% vs. 0%). The relative risk of endometrial failure for patients predicted as a poor endometrial prognosis was 3.3 times higher than those with a good prognosis. The differences in gene expression between both profiles were proposed as a biomarker, coined the endometrial failure risk (EFR) signature. Poor prognosis profiles were characterized by 59 upregulated and 63 downregulated genes mainly involved in regulation (17.0%), metabolism (8.4%), immune response, and inflammation (7.8%). This EFR signature had a median accuracy of 0.92 (min = 0.88, max = 0.94), median sensitivity of 0.96 (min = 0.91, max = 0.98), and median specificity of 0.84 (min = 0.77, max = 0.88), positioning itself as a promising biomarker for endometrial evaluation. CONCLUSION(S) The EFR signature revealed a novel endometrial disruption, independent of endometrial luteal phase timing, present in 73.7% of patients. This EFR signature stratified patients into 2 significantly distinct and clinically relevant prognosis profiles providing opportunities for personalized therapy. Nevertheless, further validations are needed before implementing this gene signature as an artificial intelligence (AI)-based tool to reduce the risk of patients experiencing endometrial failure.
Collapse
Affiliation(s)
- Patricia Diaz-Gimeno
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain.
| | - Patricia Sebastian-Leon
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | | | - Diana Marti-Garcia
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Josefa Maria Sanchez-Reyes
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| | - Maria Del Carmen Vidal
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Reproductive Medicine Center, IVI RMA Valencia, Valencia, Spain
| | - Almudena Devesa-Peiro
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain
| | - Immaculada Sanchez-Ribas
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Reproductive Medicine Center, IVI RMA Barcelona, Barcelona, Spain
| | - Asunta Martinez-Martinez
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain
| | - Nuria Pellicer
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; Reproductive Medicine Center, IVI RMA Valencia, Valencia, Spain
| | - Dagan Wells
- JUNO Genetics, Winchester House, Oxford, United Kingdom; Nuffield Department of Women's & Reproductive Health, University of Oxford, Women's Centre John Radcliffe Hospital, Oxford, United Kingdom
| | - Antonio Pellicer
- IVIRMA Global Research Alliance, IVI Foundation, Instituto de Investigación Sanitaria La Fe (IIS La Fe), Valencia, Spain; JUNO Genetics, Winchester House, Oxford, United Kingdom; Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia, Spain; Reproductive Medicine Center, IVI RMA Rome, Largo Il de brando Pizzetti, Roma, Italy
| |
Collapse
|
42
|
Gong EY, Jung D, Woo H, Song J, Choi E, Jo SG, Eyun SI, Kim S, Park YY. Genomic analysis uncovers that cold-inducible RNA binding protein is associated with estrogen receptor in breast cancer. Genes Genomics 2024; 46:899-907. [PMID: 38847971 DOI: 10.1007/s13258-024-01530-w] [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: 05/03/2024] [Accepted: 05/31/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND RNA-binding proteins (RBPs) perform various biological functions in humans and are associated with several diseases, including cancer. Therefore, RBPs have emerged as novel therapeutic targets. Although recent investigations have shown that RBPs have crucial functions in breast cancer (BC), detailed research is underway to determine the RBPs that are closely related to cancers. OBJECTIVE To provide an insight into estrogen receptor (ER) regulation by cold-inducible RNA binding protein (CIRBP) as a novel therapeutic target. RESULTS By analyzing the genomic data, we identified a potential RBP in BC. We found that CIRBP is highly correlated with ER function and influences clinical outcomes, such as patient survival and endocrine therapy responsiveness. In addition, CIRBP influences the proliferation of BC cells by directly binding to ER-RNA. CONCLUSION Our results suggest that CIRBP is a novel upstream regulator of ER and that the interplay between CIRBP and ER may be associated with the clinical relevance of BC.
Collapse
Affiliation(s)
- Eun-Yeung Gong
- Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Dana Jung
- Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Hyunmin Woo
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Jinhoo Song
- Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Eunjeong Choi
- Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Seo-Gyeong Jo
- Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan, 49315, Republic of Korea
| | - Seong-Il Eyun
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Seokho Kim
- Department of Medicinal Biotechnology, College of Health Science, Dong-A University, Busan, 49315, Republic of Korea.
| | - Yun-Yong Park
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
43
|
Fonseca VC, Sidiropoulou Z. Geriatric Breast Cancer: Staging, Molecular Surrogates, and Treatment. A Review & Meta-analysis. Aging Dis 2024; 15:1602-1618. [PMID: 37962462 PMCID: PMC11272193 DOI: 10.14336/ad.2023.1002] [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: 07/21/2023] [Accepted: 10/02/2023] [Indexed: 11/15/2023] Open
Abstract
Breast cancer (BC) is one of the most frequent cancers in females across the globe. Treatment recommendations for BC patients are primarily driven by patient age, staging and tumor molecular subtype. Thus, we updated the general overview of BC staging, molecular surrogates, and treatment choices for women >70 years based on a systematic study encompassing the years 2013-2023. A PRISMA guidelines and PICO framework were followed, and relevant research articles were searched using different data bases (Web of Sciences, PubMed, MEDLINE, and Scopus). Mixed Methods Appraisal Tool was used for studies quality assessment. The research articles that made it into the systematic review were compiled using qualitative criteria. In the meanwhile, heterogeneity was determined using meta-analysis with RevMan 5.4. We applied a random effects model with a 0.05 significance level. Overall, there were 4151 research articles, after screening only 17 articles with 39,906 patients were included. Conclusion: Elderly patients with breast cancer should be treated differently in an adapted way. The treatment should not be the same worldwide due to different health systems. Molecular surrogates are different in geriatric patients. Surgery is the best option for treatment in this subset of patients. We need to have therapeutic decision appointments for elderly patients with breast cancer. The guidelines and medical authority should be used in the best decision.
Collapse
Affiliation(s)
- Vasco C Fonseca
- Department of Oncology, Hospital Centre of West Lisbon, Portugal.
| | | |
Collapse
|
44
|
Liu W, Liu D, Zhang Z. Evaluating prognostic biomarkers for survival outcomes subject to informative censoring. Stat Methods Med Res 2024; 33:1342-1354. [PMID: 38841774 DOI: 10.1177/09622802241259170] [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] [Indexed: 06/07/2024]
Abstract
Prognostic biomarkers for survival outcomes are widely used in clinical research and practice. Such biomarkers are often evaluated using a C-index as well as quantities based on time-dependent receiver operating characteristic curves. Existing methods for their evaluation generally assume that censoring is uninformative in the sense that the censoring time is independent of the failure time with or without conditioning on the biomarker under evaluation. With focus on the C-index and the area under a particular receiver operating characteristic curve, we describe and compare three estimation methods that account for informative censoring based on observed baseline covariates. Two of them are straightforward extensions of existing plug-in and inverse probability weighting methods for uninformative censoring. By appealing to semiparametric theory, we also develop a doubly robust, locally efficient method that is more robust than the plug-in and inverse probability weighting methods and typically more efficient than the inverse probability weighting method. The methods are evaluated and compared in a simulation study, and applied to real data from studies of breast cancer and heart failure.
Collapse
Affiliation(s)
- Wei Liu
- School of Management, Harbin Institute of Technology, Harbin, China
| | - Danping Liu
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zhiwei Zhang
- Biostatistics Innovation Group, Gilead Sciences Inc, Foster City, CA, USA
| |
Collapse
|
45
|
Hyams DM, Bareket-Samish A, Rocha JEB, Diaz-Botero S, Franco S, Gagliato D, Gomez HL, Korbenfeld E, Krygier G, Mattar A, De Pierro AN, Borrego MR, Villarreal C. Selecting postoperative adjuvant systemic therapy for early-stage breast cancer: An updated assessment and systematic review of leading commercially available gene expression assays. J Surg Oncol 2024; 130:166-187. [PMID: 38932668 DOI: 10.1002/jso.27692] [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: 04/30/2024] [Accepted: 05/05/2024] [Indexed: 06/28/2024]
Abstract
Gene expression assays (GEAs) can guide treatment for early-stage breast cancer. Several large prospective randomized clinical trials, and numerous additional studies, now provide new information for selecting an appropriate GEA. This systematic review builds upon prior reviews, with a focus on five widely commercialized GEAs (Breast Cancer Index®, EndoPredict®, MammaPrint®, Oncotype DX®, and Prosigna®). The comprehensive dataset available provides a contemporary opportunity to assess each GEA's utility as a prognosticator and/or predictor of adjuvant therapy benefit.
Collapse
Affiliation(s)
- David M Hyams
- Medical Director, Desert Surgical Oncology, Eisenhower Medical Center, Rancho Mirage, California, USA
| | | | - Juan Enrique Bargallo Rocha
- Breast Cancer Department, Instituto Nacional de Cancerología Mexico and Centro Medico ABC, Mexico City, Mexico
| | - Sebastian Diaz-Botero
- Breast Surgical Oncology Unit, Cancer Center at Clínica Universidad de Navarra, Madrid, Spain
| | - Sandra Franco
- Medical Director, Centro de Tratamiento e Investigación sobre el Cáncer, CTIC, Bogotá, Colombia
| | - Debora Gagliato
- Department of Clinical Oncology, Beneficencia Portuguesa de Sao Paulo, San Paulo, Brazil
| | - Henry L Gomez
- Breast Unit Director, OncoSalud, Clinica Delgado, AUNA, Universidad Ricardo Palma, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Ernesto Korbenfeld
- Department of Oncology, Hospital Británico de Buenos Aires, Buenos Aires, Argentina
| | - Gabriel Krygier
- Department of Oncology, Universitary Hospital de Clínicas, Montevideo, Uruguay
| | - Andre Mattar
- Director of Mastology Center, Centro de Referência da Saúde da Mulher, Hospital da Mulher, São Paulo, Brazil
| | - Aníbal Nuñez De Pierro
- Department of Surgery, Unit of Mastology, Hospital J.A. Fernandez, Buenos Aires City, Argentina
| | - Manuel Ruiz Borrego
- Medical Oncology Service, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Cynthia Villarreal
- Head, Department of Medical Oncology, Breast Cancer Center, Hospital Zambrano Hellion TecSalud, Tecnologico de Monterrey, Monterrey, Nuevo Leon, Mexico
| |
Collapse
|
46
|
Coombes RC, Angelou C, Al-Khalili Z, Hart W, Francescatti D, Wright N, Ellis I, Green A, Rakha E, Shousha S, Amrania H, Phillips CC, Palmieri C. Performance of a novel spectroscopy-based tool for adjuvant therapy decision-making in hormone receptor-positive breast cancer: a validation study. Breast Cancer Res Treat 2024; 205:349-358. [PMID: 38244167 PMCID: PMC11101376 DOI: 10.1007/s10549-023-07229-y] [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: 08/02/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024]
Abstract
PURPOSE Digistain Index (DI), measured using an inexpensive mid-infrared spectrometer, reflects the level of aneuploidy in unstained tissue sections and correlates with tumor grade. We investigated whether incorporating DI with other clinicopathological variables could predict outcomes in patients with early breast cancer. METHODS DI was calculated in 801 patients with hormone receptor-positive, HER2-negative primary breast cancer and ≤ 3 positive lymph nodes. All patients were treated with systemic endocrine therapy and no chemotherapy. Multivariable proportional hazards modeling was used to incorporate DI with clinicopathological variables to generate the Digistain Prognostic Score (DPS). DPS was assessed for prediction of 5- and 10-year outcomes (recurrence, recurrence-free survival [RFS] and overall survival [OS]) using receiver operating characteristics and Cox proportional hazards regression models. Kaplan-Meier analysis evaluated the ability of DPS to stratify risk. RESULTS DPS was consistently highly accurate and had negative predictive values for all three outcomes, ranging from 0.96 to 0.99 at 5 years and 0.84 to 0.95 at 10 years. DPS demonstrated statistically significant prognostic ability with significant hazard ratios (95% CI) for low- versus high-risk classification for RFS, recurrence and OS (1.80 [CI 1.31-2.48], 1.83 [1.32-2.52] and 1.77 [1.28-2.43], respectively; all P < 0.001). CONCLUSION DPS showed high accuracy and predictive performance, was able to stratify patients into low or high-risk, and considering its cost and rapidity, has the potential to offer clinical utility.
Collapse
Affiliation(s)
- R Charles Coombes
- Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Christina Angelou
- Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Zamzam Al-Khalili
- Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - William Hart
- Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | | | | | - Ian Ellis
- Nottingham University Hospital, Nottingham, UK
| | | | - Emad Rakha
- Nottingham University Hospital, Nottingham, UK
| | - Sami Shousha
- Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Hemmel Amrania
- Imperial College London, South Kensington Campus, London, SW7 2AZ, UK.
| | - Chris C Phillips
- Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | | |
Collapse
|
47
|
Lv Y, Wang Y, Zhang Y, Chen S, Yao Y. Predicting the Risk of Breast Cancer Recurrence and Metastasis based on
miRNA Expression. Curr Bioinform 2024; 19:482-489. [DOI: 10.2174/1574893618666230914105741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/18/2023] [Accepted: 08/09/2023] [Indexed: 01/04/2025]
Abstract
Background:
Even after surgery, breast cancer patients still suffer from recurrence and
metastasis. Thus, it is critical to predict accurately the risk of recurrence and metastasis for individual
patients, which can help determine the appropriate adjuvant therapy.
Methods:
The purpose of this study is to investigate and compare the performance of several categories of molecular biomarkers, i.e., microRNA (miRNA), long non-coding RNA (lncRNA), messenger RNA (mRNA), and copy number variation (CNV), in predicting the risk of breast cancer recurrence and metastasis. First, the molecular data (miRNA, lncRNA, mRNA, and CNV) of 483 breast
cancer patients were downloaded from the Cancer Genome Atlas, which were then randomly divided
into the training and test sets with a ratio of 7:3. Second, the feature selection process was applied by
univariate Cox and multivariate Cox variance analysis on the training set (e.g., 15 miRNAs). According to the selected features (e.g., 15 miRNAs), a random forest classifier and several other classification methods were established according to the label of recurrence and metastasis. Finally, the performances of the classification models were compared and evaluated on the test set.
Results:
The area under the ROC curve was 0.70 for miRNA, better than those using other biomarkers.
Conclusion:
These results indicated that miRNA has important guiding significance in predicting
recurrence and metastasis of breast cancer.
Collapse
Affiliation(s)
- Yaping Lv
- School of Mathematics and Statistics, Hainan Normal University, Haikou 570100, China
- Genies Beijing Co., Ltd.,
Beijing 100102, China
| | - Yanfeng Wang
- Department of Pathology, Beidahuang Industry Group General Hospital, Haerbin 150088,
China
| | - Yumeng Zhang
- School of Mathematics and Statistics, Hainan Normal University, Haikou 570100, China
| | - Shuzhen Chen
- School of Mathematics and Statistics, Hainan Normal University, Haikou 570100, China
| | - Yuhua Yao
- School of Mathematics and Statistics, Hainan Normal University, Haikou 570100, China
- Key Laboratory of Data Science and Intelligence Education, Ministry of Education, Hainan Normal University,
Haikou, China
- Key Laboratory of Computational Science and Application of Hainan Province, Hainan Normal University, Haikou, China
| |
Collapse
|
48
|
Qian X, Tan H, Liu X, Zhao W, Chan MD, Kim P, Zhou X. Radiogenomics-Based Risk Prediction of Glioblastoma Multiforme with Clinical Relevance. Genes (Basel) 2024; 15:718. [PMID: 38927654 PMCID: PMC11202835 DOI: 10.3390/genes15060718] [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: 04/12/2024] [Revised: 05/20/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024] Open
Abstract
Glioblastoma multiforme (GBM)is the most common and aggressive primary brain tumor. Although temozolomide (TMZ)-based radiochemotherapy improves overall GBM patients' survival, it also increases the frequency of false positive post-treatment magnetic resonance imaging (MRI) assessments for tumor progression. Pseudo-progression (PsP) is a treatment-related reaction with an increased contrast-enhancing lesion size at the tumor site or resection margins miming tumor recurrence on MRI. The accurate and reliable prognostication of GBM progression is urgently needed in the clinical management of GBM patients. Clinical data analysis indicates that the patients with PsP had superior overall and progression-free survival rates. In this study, we aimed to develop a prognostic model to evaluate the tumor progression potential of GBM patients following standard therapies. We applied a dictionary learning scheme to obtain imaging features of GBM patients with PsP or true tumor progression (TTP) from the Wake dataset. Based on these radiographic features, we conducted a radiogenomics analysis to identify the significantly associated genes. These significantly associated genes were used as features to construct a 2YS (2-year survival rate) logistic regression model. GBM patients were classified into low- and high-survival risk groups based on the individual 2YS scores derived from this model. We tested our model using an independent The Cancer Genome Atlas Program (TCGA) dataset and found that 2YS scores were significantly associated with the patient's overall survival. We used two cohorts of the TCGA data to train and test our model. Our results show that the 2YS scores-based classification results from the training and testing TCGA datasets were significantly associated with the overall survival of patients. We also analyzed the survival prediction ability of other clinical factors (gender, age, KPS (Karnofsky performance status), normal cell ratio) and found that these factors were unrelated or weakly correlated with patients' survival. Overall, our studies have demonstrated the effectiveness and robustness of the 2YS model in predicting the clinical outcomes of GBM patients after standard therapies.
Collapse
Affiliation(s)
- Xiaohua Qian
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
- Department of Bioinformatics and Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA (X.L.); (P.K.)
| | - Hua Tan
- Department of Bioinformatics and Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA (X.L.); (P.K.)
| | - Xiaona Liu
- Department of Bioinformatics and Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA (X.L.); (P.K.)
| | - Weiling Zhao
- Department of Bioinformatics and Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA (X.L.); (P.K.)
| | - Michael D. Chan
- Department of Radiation Oncology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Pora Kim
- Department of Bioinformatics and Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA (X.L.); (P.K.)
| | - Xiaobo Zhou
- Department of Bioinformatics and Systems Medicine, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA (X.L.); (P.K.)
| |
Collapse
|
49
|
Bleaney CW, Abdelaal H, Reardon M, Anandadas C, Hoskin P, Choudhury A, Forker L. Clinical Biomarkers of Tumour Radiosensitivity and Predicting Benefit from Radiotherapy: A Systematic Review. Cancers (Basel) 2024; 16:1942. [PMID: 38792019 PMCID: PMC11119069 DOI: 10.3390/cancers16101942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/18/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024] Open
Abstract
Modern advanced radiotherapy techniques have improved the precision and accuracy of radiotherapy delivery, with resulting plans being highly personalised based on individual anatomy. Adaptation for individual tumour biology remains elusive. There is an unmet need for biomarkers of intrinsic radiosensitivity that can predict tumour response to radiation to facilitate individualised decision-making, dosing and treatment planning. Over the last few decades, the use of high throughput molecular biology technologies has led to an explosion of newly discovered cancer biomarkers. Gene expression signatures are now used routinely in clinic to aid decision-making regarding adjuvant systemic therapy. They have great potential as radiotherapy biomarkers. A previous systematic review published in 2015 reported only five studies of signatures evaluated for their ability to predict radiotherapy benefits in clinical cohorts. This updated systematic review encompasses the expanded number of studies reported in the last decade. An additional 27 studies were identified. In total, 22 distinct signatures were recognised (5 pre-2015, 17 post-2015). Seventeen signatures were 'radiosensitivity' signatures and five were breast cancer prognostic signatures aiming to identify patients at an increased risk of local recurrence and therefore were more likely to benefit from adjuvant radiation. Most signatures (15/22) had not progressed beyond the discovery phase of development, with no suitable validated clinical-grade assay for application. Very few signatures (4/17 'radiosensitivity' signatures) had undergone any laboratory-based biological validation of their ability to predict tumour radiosensitivity. No signatures have been assessed prospectively in a phase III biomarker-led trial to date and none are recommended for routine use in clinical guidelines. A phase III prospective evaluation is ongoing for two breast cancer prognostic signatures. The most promising radiosensitivity signature remains the radiosensitivity index (RSI), which is used to calculate a genomic adjusted radiation dose (GARD). There is an ongoing phase II prospective biomarker-led study of RSI/GARD in triple negative breast cancer. The results of these trials are eagerly anticipated over the coming years. Future work in this area should focus on (1) robust biological validation; (2) building biobanks alongside large radiotherapy randomised controlled trials with dose variance (to demonstrate an interaction between radiosensitivity signature and dose); (3) a validation of clinical-grade cost-effective assays that are deliverable within current healthcare infrastructure; and (4) an integration with biomarkers of other determinants of radiation response.
Collapse
Affiliation(s)
- Christopher W. Bleaney
- Translational Radiobiology Group, Division of Cancer Sciences, The Oglesby Cancer Research Building, The University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, UK (L.F.)
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Hebatalla Abdelaal
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Mark Reardon
- Translational Radiobiology Group, Division of Cancer Sciences, The Oglesby Cancer Research Building, The University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, UK (L.F.)
| | - Carmel Anandadas
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Peter Hoskin
- Translational Radiobiology Group, Division of Cancer Sciences, The Oglesby Cancer Research Building, The University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, UK (L.F.)
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Ananya Choudhury
- Translational Radiobiology Group, Division of Cancer Sciences, The Oglesby Cancer Research Building, The University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, UK (L.F.)
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| | - Laura Forker
- Translational Radiobiology Group, Division of Cancer Sciences, The Oglesby Cancer Research Building, The University of Manchester, 555 Wilmslow Road, Manchester M20 4GJ, UK (L.F.)
- Department of Clinical Oncology, The Christie NHS Foundation Trust, 550 Wilmslow Road, Manchester M20 4BX, UK
| |
Collapse
|
50
|
Asghar N, Khalil U, Ahmad B, Alshanbari HM, Hamraz M, Ahmad B, Khan DM. Improved nonparametric survival prediction using CoxPH, Random Survival Forest & DeepHit Neural Network. BMC Med Inform Decis Mak 2024; 24:120. [PMID: 38715002 PMCID: PMC11531126 DOI: 10.1186/s12911-024-02525-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/30/2024] [Indexed: 11/03/2024] Open
Abstract
In recent times, time-to-event data such as time to failure or death is routinely collected alongside high-throughput covariates. These high-dimensional bioinformatics data often challenge classical survival models, which are either infeasible to fit or produce low prediction accuracy due to overfitting. To address this issue, the focus has shifted towards introducing a novel approaches for feature selection and survival prediction. In this article, we propose a new hybrid feature selection approach that handles high-dimensional bioinformatics datasets for improved survival prediction. This study explores the efficacy of four distinct variable selection techniques: LASSO, RSF-vs, SCAD, and CoxBoost, in the context of non-parametric biomedical survival prediction. Leveraging these methods, we conducted comprehensive variable selection processes. Subsequently, survival analysis models-specifically CoxPH, RSF, and DeepHit NN-were employed to construct predictive models based on the selected variables. Furthermore, we introduce a novel approach wherein only variables consistently selected by a majority of the aforementioned feature selection techniques are considered. This innovative strategy, referred to as the proposed method, aims to enhance the reliability and robustness of variable selection, subsequently improving the predictive performance of the survival analysis models. To evaluate the effectiveness of the proposed method, we compare the performance of the proposed approach with the existing LASSO, RSF-vs, SCAD, and CoxBoost techniques using various performance metrics including integrated brier score (IBS), concordance index (C-Index) and integrated absolute error (IAE) for numerous high-dimensional survival datasets. The real data applications reveal that the proposed method outperforms the competing methods in terms of survival prediction accuracy.
Collapse
Affiliation(s)
- Naseem Asghar
- Department of Statistics, Abdul Wali Khan University Mardan, Mardan, KP, Pakistan
- Department of Statistics, University of Swat, Swat, KP, Pakistan
| | - Umair Khalil
- Department of Statistics, Abdul Wali Khan University Mardan, Mardan, KP, Pakistan
| | - Basheer Ahmad
- Department of Statistics, Abdul Wali Khan University Mardan, Mardan, KP, Pakistan
| | - Huda M Alshanbari
- Department of Mathematical Sciences, College of Science, Princess Nourah bint Abdulrahman University, P.O.Box 84428, Riyadh, 11671, Saudi Arabia
| | - Muhammad Hamraz
- Department of Statistics, Abdul Wali Khan University Mardan, Mardan, KP, Pakistan
| | | | - Dost Muhammad Khan
- Department of Statistics, Abdul Wali Khan University Mardan, Mardan, KP, Pakistan
| |
Collapse
|