1
|
Du F, Ju J, Zheng F, Gao S, Yuan P. The Identification of Novel Prognostic and Predictive Biomarkers in Breast Cancer via the Elucidation of Tumor Ecotypes Using Ecotyper. CANCER INNOVATION 2025; 4:e70013. [PMID: 40432877 PMCID: PMC12107130 DOI: 10.1002/cai2.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 03/08/2025] [Accepted: 03/18/2025] [Indexed: 05/29/2025]
Abstract
Background Breast cancer is a highly heterogeneous disease, characterized by tumor and nontumor cells at various cell states. Ecotyper is an innovative machine learning framework that quantifies the tumor microenvironment and delineates the tumor ecosystem, demonstrating clinical significance. However, further validation is needed in breast cancer. Methods Ecotyper was applied to identify multiple cellular states and tumor ecotypes using large-scale breast cancer bulk sequencing data, followed by a detailed analysis of their associations with clinical classification, molecular subtypes, survival prognosis, and immunotherapy response. Identified subtypes were further characterized using single-cell and spatial data sets to reveal molecular profiles. Results In a comprehensive analysis of 6578 breast cancer samples from four data sets, Ecotyper identified 69 cellular states and 10 tumor ecotypes. Of these, 37 cellular states significantly correlated with overall survival. Notably, specific states within epithelial cells, macrophages/monocytes, and fibroblasts were linked to a worse prognosis. CE2 abundance was identified as the most significant marker indicating unfavorable prognosis and was further validated in an additional data set of 116 HER2-negative patients. These biomarkers also indicated the efficacy of neoadjuvant immunotherapy in breast cancer. CE2-high cancers were characterized by an abundance of basal-like epithelial cells, scant lymphocytic infiltration, and activation of hypoxia signaling. Single-cell analysis showed that CE2-high areas were rich in SPP1-positive tumor-associated macrophages(TAM), basal-like epithelial cells, and hypoxic cancer-associated fibroblasts(CAF). Spatially, these regions were often peripheral in triple-negative breast cancer, adjacent to fibrotic/necrotic zones. Multiplex immunofluorescence confirmed the enrichment of SPP1+CD68+TAM and HIF1A+SMA+CAF in hypoxic triple-negative breast cancer (TNBC) regions. Conclusions Ecotyper identified novel biomarkers for breast cancer prognosis and treatment prediction. The CE2-high region may represent a hypoxic immune-suppressive niche.
Collapse
Affiliation(s)
- Feng Du
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), The VIPII Gastrointestinal Cancer Division of Medical DepartmentPeking University Cancer Hospital and InstituteBeijingChina
| | - Jie Ju
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Day CarePeking University Cancer Hospital and InstituteBeijingChina
| | - Fangchao Zheng
- Department of Medical Oncology, Cancer Research Center, Shandong Cancer Hospital and InstituteShandong First Medical University and Shandong Academy of Medical SciencesJinanShandong ProvinceChina
| | - Songlin Gao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), The VIPII Gastrointestinal Cancer Division of Medical DepartmentPeking University Cancer Hospital and InstituteBeijingChina
| | - Peng Yuan
- Department of VIP Medical Services, National Cancer Centre/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
López-Velazco JI, Manzano S, Elorriaga K, Otaño M, Lahuerta A, Álvarez L, Etxabe I, Huarte M, Buch E, Gimenez J, Quiroga V, Fernandez M, Aragón S, Paré L, Prat A, Álvarez-López I, Caffarel MM, Urruticoechea A. Molecular characterisation of the residual disease after neoadjuvant endocrine therapy in ER+/HER2- breast cancer uncovers biomarkers of tumour response. Transl Oncol 2025; 57:102407. [PMID: 40349505 DOI: 10.1016/j.tranon.2025.102407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Neoadjuvant endocrine therapy (NET) in oestrogen receptor-positive /HER2-negative breast cancer (ER+/HER2- BC) allows real-time evaluation of treatment sensitivity by monitoring tumour response and offers the opportunity of personalised therapy. However, the lack of reproducible biomarkers to assess response and long-term prognosis after NET is a significant barrier to increase its indications. METHODS In this study we searched for clinically relevant molecular reporters of response to NET in a multicentre population of ER+/HER2- BC patients (n = 87) by using: PAM50 gene expression panel and immunohistochemical evaluation of key proteins involved in tumorigenesis. RESULTS Our PAM50 analyses show that tumours changing from luminal A to normal-like subtype after NET presented better radiological and pathological tumour responses, a significant larger decrease in Ki67 at surgery, lower preoperative endocrine prognostic index score (PEPI) and lower tumour cellularity size (TCS) than those with persistent luminal A status. Patients with the highest response to NET showed the largest decrease in PAM50-derived risk of recurrence (ROR) following NET. In addition, the percentage of p53 positive cells was associated with decreased response to NET. CONCLUSIONS Our findings highlight the change of intrinsic subtype from luminal A to normal-like after NET as a putative biomarker characterising the patient population that obtains the highest benefit from NET. Our study also suggests that changes in PAM50-derived ROR score and p53 evaluation could also help to identify those patients. Thus, this study uncovers potential biomarkers of response to NET and prognosis, which should be validated in independent cohorts, helping to the implementation of NET in the clinical practice.
Collapse
Affiliation(s)
- Joanna I López-Velazco
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain
| | - Sara Manzano
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain
| | - Kepa Elorriaga
- Gipuzkoa Pathology Unit, OSI Donostialdea - Onkologikoa, San Sebastián, Spain
| | - Maria Otaño
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gipuzkoa Cancer Unit/OSI Donostialdea - Onkologikoa, San Sebastián, Spain
| | - Ainhara Lahuerta
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gipuzkoa Cancer Unit/OSI Donostialdea - Onkologikoa, San Sebastián, Spain
| | - Luis Álvarez
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gynecology and General Surgery Departments - Breast Unit, Onkologikoa, San Sebastián, Spain
| | - Inge Etxabe
- Gynecology and General Surgery Departments - Breast Unit, Onkologikoa, San Sebastián, Spain
| | - Miren Huarte
- Gynecology and General Surgery Departments - Breast Unit, Onkologikoa, San Sebastián, Spain
| | - Elvira Buch
- Hospital Clínico Universitario de Valencia, Spain
| | | | | | - Marta Fernandez
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gynecology Department - Breast Unit, OSI Donostialdea, San Sebastián, Spain
| | | | - Laia Paré
- Hospital Clinic, Barcelona - IDIBAPS, Barcelona, Spain
| | - Aleix Prat
- Hospital Clinic, Barcelona - IDIBAPS, Barcelona, Spain
| | - Isabel Álvarez-López
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gipuzkoa Cancer Unit/OSI Donostialdea - Onkologikoa, San Sebastián, Spain
| | - Maria M Caffarel
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| | - Ander Urruticoechea
- Biogipuzkoa (previously known as Biodonostia) Health Research Institute, San Sebastián, Spain; Gipuzkoa Cancer Unit/OSI Donostialdea - Onkologikoa, San Sebastián, Spain.
| |
Collapse
|
3
|
Bhattacharya N, Rockstroh A, Deshpande SS, Thomas SK, Yadav A, Goswami C, Chawla S, Solomon P, Fourgeux C, Ahuja G, Hollier B, Kumar H, Roquilly A, Poschmann J, Lehman M, Nelson CC, Sengupta D. Artificial intelligence approaches for tumor phenotype stratification from single-cell transcriptomic data. eLife 2025; 13:RP98469. [PMID: 40511682 PMCID: PMC12165692 DOI: 10.7554/elife.98469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2025] Open
Abstract
Single-cell RNA-sequencing (scRNA-seq) coupled with robust computational analysis facilitates the characterization of phenotypic heterogeneity within tumors. Current scRNA-seq analysis pipelines are capable of identifying a myriad of malignant and non-malignant cell subtypes from single-cell profiling of tumors. However, given the extent of intra-tumoral heterogeneity, it is challenging to assess the risk associated with individual cell subpopulations, primarily due to the complexity of the cancer phenotype space and the lack of clinical annotations associated with tumor scRNA-seq studies. To this end, we introduce SCellBOW, a scRNA-seq analysis framework inspired by document embedding techniques from the domain of Natural Language Processing (NLP). SCellBOW is a novel computational approach that facilitates effective identification and high-quality visualization of single-cell subpopulations. We compared SCellBOW with existing best practice methods for its ability to precisely represent phenotypically divergent cell types across multiple scRNA-seq datasets, including our in-house generated human splenocyte and matched peripheral blood mononuclear cell (PBMC) dataset. For tumor cells, SCellBOW estimates the relative risk associated with each cluster and stratifies them based on their aggressiveness. This is achieved by simulating how the presence or absence of a specific cell subpopulation influences disease prognosis. Using SCellBOW, we identified a hitherto unknown and pervasive AR-/NElow (androgen-receptor-negative, neuroendocrine-low) malignant subpopulation in metastatic prostate cancer with conspicuously high aggressiveness. Overall, the risk-stratification capabilities of SCellBOW hold promise for formulating tailored therapeutic interventions by identifying clinically relevant tumor subpopulations and their impact on prognosis.
Collapse
Affiliation(s)
- Namrata Bhattacharya
- Australian Prostate Cancer Research Centre-Queensland, Faculty of Health, School of Biomedical Sciences, Centre for Genomics and Personalised Health, Queensland University of TechnologyBrisbaneAustralia
- Department of Computer Science and Engineering, Indraprastha Institute of Information Technology-Delhi (IIIT-Delhi), Okhla, Phase IIINew DelhiIndia
- Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
| | - Anja Rockstroh
- Australian Prostate Cancer Research Centre-Queensland, Faculty of Health, School of Biomedical Sciences, Centre for Genomics and Personalised Health, Queensland University of TechnologyBrisbaneAustralia
- Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
| | - Sanket Suhas Deshpande
- Department of Computational Biology, Indraprastha Institute of Information Technology-Delhi (IIIT-Delhi), Okhla, Phase IIINew DelhiIndia
| | - Sam Koshy Thomas
- School of Mathematical Sciences, The University of AdelaideAdelaideAustralia
| | - Anunay Yadav
- Department of Computer Science and Engineering, Indraprastha Institute of Information Technology-Delhi (IIIT-Delhi), Okhla, Phase IIINew DelhiIndia
| | - Chitrita Goswami
- Department of Computer Science and Engineering, Indraprastha Institute of Information Technology-Delhi (IIIT-Delhi), Okhla, Phase IIINew DelhiIndia
| | - Smriti Chawla
- Center for Computational Biomedicine, Harvard Medical SchoolBostonUnited States
| | - Pierre Solomon
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMRNantesFrance
| | - Cynthia Fourgeux
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMRNantesFrance
| | - Gaurav Ahuja
- Department of Computational Biology, Indraprastha Institute of Information Technology-Delhi (IIIT-Delhi), Okhla, Phase IIINew DelhiIndia
- Centre for Artificial Intelligence, Indraprastha Institute of Information Technology-Delhi (IIIT-Delhi), Okhla, Phase IIINew DelhiIndia
| | - Brett Hollier
- Australian Prostate Cancer Research Centre-Queensland, Faculty of Health, School of Biomedical Sciences, Centre for Genomics and Personalised Health, Queensland University of TechnologyBrisbaneAustralia
- Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
| | - Himanshu Kumar
- Laboratory of Immunology and Infectious Disease Biology, Department of Biological Sciences, Indian Institute of Science Education and Research (IISER)BhopalIndia
| | - Antoine Roquilly
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMRNantesFrance
| | - Jeremie Poschmann
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMRNantesFrance
| | - Melanie Lehman
- Australian Prostate Cancer Research Centre-Queensland, Faculty of Health, School of Biomedical Sciences, Centre for Genomics and Personalised Health, Queensland University of TechnologyBrisbaneAustralia
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British ColumbiaVancouverCanada
| | - Colleen C Nelson
- Australian Prostate Cancer Research Centre-Queensland, Faculty of Health, School of Biomedical Sciences, Centre for Genomics and Personalised Health, Queensland University of TechnologyBrisbaneAustralia
- Translational Research Institute, Princess Alexandra HospitalWoolloongabbaAustralia
| | - Debarka Sengupta
- Department of Computer Science and Engineering, Indraprastha Institute of Information Technology-Delhi (IIIT-Delhi), Okhla, Phase IIINew DelhiIndia
- Department of Computational Biology, Indraprastha Institute of Information Technology-Delhi (IIIT-Delhi), Okhla, Phase IIINew DelhiIndia
- Centre for Artificial Intelligence, Indraprastha Institute of Information Technology-Delhi (IIIT-Delhi), Okhla, Phase IIINew DelhiIndia
| |
Collapse
|
4
|
Hwang KW, Yun JW, Shin YJ, Lee HJ, Kim HS. Refining housekeeping genes and demonstrating their potential for clinical and experimental applications. Comput Biol Med 2025; 194:110546. [PMID: 40489916 DOI: 10.1016/j.compbiomed.2025.110546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 06/03/2025] [Accepted: 06/03/2025] [Indexed: 06/11/2025]
Abstract
BACKGROUND Housekeeping genes (HKGs) are crucial for maintaining basic cellular functions and are consistently expressed across various tissues and cell types, making them essential for normalizing gene expression. Their application is crucial in both basic research and clinical settings, such as breast cancer, where they help in accurate gene expression measurement and tumor subtype classification such as the PAM50 system. However, HKGs are often used without thorough assessment of their variability across different conditions, which may affect the reliability of normalization. METHODS AND FINDINGS We identified 16 candidate HKGs in breast tissue using TCGA RNA-seq data. These genes, along with previously known HKGs such as GAPDH, were evaluated across several breast cancer cell lines and experimental conditions that mimic clinical cancer treatment using quantitative real-time PCR (qPCR). The candidate HKGs were further validated using additional bulk and single cell RNA-seq datasets from the Gene Expression Omnibus (GEO) and by performing droplet digital PCR (ddPCR). We finally concluded that our candidate HKGs, especially EIF4H, GHITM, ATP5F1B, BRK1, and OS9, demonstrated greater stability than GAPDH and RPLP0. These genes were subsequently tested within the PAM50 breast cancer subtyping system, where they improved normalization performance over GAPDH. CONCLUSION We have identified novel HKGs useful in breast cancer research and proved that they exhibit more stable expression compared to previously known HKGs. These findings may offer researchers and clinicians a more reliable normalization standard for gene expression analysis, potentially enhancing the accuracy of breast cancer diagnosis and the selection of personalized treatments.
Collapse
Affiliation(s)
- Kyung Won Hwang
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae Won Yun
- Veterans Health Service Medical Research Institute, Veterans Health Service Medical Center, Seoul, 05368, Republic of Korea
| | - Ye Ji Shin
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hye Jung Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hong Sook Kim
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| |
Collapse
|
5
|
Sinha S, Roy R, Barman N, Sarkar P, Saha A, Biswas N. IL6 mediated cFLIP downregulation increases the migratory and invasive potential of triple negative breast cancer cell. Cell Signal 2025; 130:111679. [PMID: 39988287 DOI: 10.1016/j.cellsig.2025.111679] [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: 08/14/2024] [Revised: 02/03/2025] [Accepted: 02/16/2025] [Indexed: 02/25/2025]
Abstract
c-FLIP (cellular FLICE-Like Inhibitor of Apoptotic protein) alias CFLAR (Cellular FADD-like apoptosis regulator) is an inhibitor of Caspase 8 and thus plays a key role in the regulation of extrinsic apoptotic pathway. However, the mechanisms of cFLIP regulation during the course of progression of cancer and it's involvement in tumour cell migration and invasion is yet to be known. Our TCGA data analysis has shown that cFLIP is downregulated in many cancers, including breast cancer, especially at the later stages. Next, we have analysed the role of cFLIP in breast cancer progression in In-vitro study. In doing so, we have used luminal breast cancer cell line MCF7 as non-aggressive and non-invasive breast cancer model and triple negative breast cancer cell lines MDA-MB-231, MDA-MB-468 and MDA-MB-453 as highly aggressive and invasive breast cancer cell model. When, we analysed and compared MCF7 and triple negative cell lines, we found a negative correlation between cFLIP expression pattern and metastasis which supported our In-silico study. Moreover, we found that Il6, one of the most prominent cytokines inside tumour microenvironment, helped in cFLIP downregulation via activation of p38 in MDA-MB-231 cell line. Not only that we have shown that cFLIP negatively regulated autophagy and this autophagy down-regulation resulted in decrease in metastasis. Thus, we have shown in an In-vitro model, for the first time, a complete interconnecting pathway in which IL6 mediated p38 activation directly influences metastasis by regulating autophagy via cFLIP downregulation.
Collapse
Affiliation(s)
- Samraj Sinha
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Rajdeep Roy
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Nilesh Barman
- Department of Life Sciences, Presidency University, Kolkata, India
| | - Purandar Sarkar
- Institute of Health Sciences, Presidency University, Kolkata, India
| | - Abhik Saha
- Institute of Health Sciences, Presidency University, Kolkata, India
| | - Nabendu Biswas
- Department of Life Sciences, Presidency University, Kolkata, India.
| |
Collapse
|
6
|
Metsch JM, Hauschild AC. BenchXAI: Comprehensive benchmarking of post-hoc explainable AI methods on multi-modal biomedical data. Comput Biol Med 2025; 191:110124. [PMID: 40239236 DOI: 10.1016/j.compbiomed.2025.110124] [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: 12/20/2024] [Revised: 02/13/2025] [Accepted: 03/31/2025] [Indexed: 04/18/2025]
Abstract
The increasing digitalization of multi-modal data in medicine and novel artificial intelligence (AI) algorithms opens up a large number of opportunities for predictive models. In particular, deep learning models show great performance in the medical field. A major limitation of such powerful but complex models originates from their 'black-box' nature. Recently, a variety of explainable AI (XAI) methods have been introduced to address this lack of transparency and trust in medical AI. However, the majority of such methods have solely been evaluated on single data modalities. Meanwhile, with the increasing number of XAI methods, integrative XAI frameworks and benchmarks are essential to compare their performance on different tasks. For that reason, we developed BenchXAI, a novel XAI benchmarking package supporting comprehensive evaluation of fifteen XAI methods, investigating their robustness, suitability, and limitations in biomedical data. We employed BenchXAI to validate these methods in three common biomedical tasks, namely clinical data, medical image and signal data, and biomolecular data. Our newly designed sample-wise normalization approach for post-hoc XAI methods enables the statistical evaluation and visualization of performance and robustness. We found that the XAI methods Integrated Gradients, DeepLift, DeepLiftShap, and GradientShap performed well over all three tasks, while methods like Deconvolution, Guided Backpropagation, and LRP-α1-β0 struggled for some tasks. With acts such as the EU AI Act the application of XAI in the biomedical domain becomes more and more essential. Our evaluation study represents a first step towards verifying the suitability of different XAI methods for various medical domains.
Collapse
Affiliation(s)
| | - Anne-Christin Hauschild
- Institute for Medical Informatics, University Medical Center Göttingen, Germany; Institute for Predictive Deep Learning in Medicine and Healthcare, Justus-Liebig University, Gießen, Germany
| |
Collapse
|
7
|
Cascianelli S, Milojkovic I, Masseroli M. A novel machine learning-based workflow to capture intra-patient heterogeneity through transcriptional multi-label characterization and clinically relevant classification. J Biomed Inform 2025; 166:104817. [PMID: 40216371 DOI: 10.1016/j.jbi.2025.104817] [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: 11/06/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 05/25/2025]
Abstract
OBJECTIVES Patient classification into specific molecular subtypes is paramount in biomedical research and clinical practice to face complex, heterogeneous diseases. Existing methods, especially for gene expression-based cancer subtyping, often simplify patient molecular portraits, neglecting the potential co-occurrence of traits from multiple subtypes. Yet, recognizing intra-sample heterogeneity is essential for more precise patient characterization and improved personalized treatments. METHODS We developed a novel computational workflow, named MULTI-STAR, which addresses current limitations and provides tailored solutions for reliable multi-label patient subtyping. MULTI-STAR uses state-of-the-art subtyping methods to obtain promising machine learning-based multi-label classifiers, leveraging gene expression profiles. It modifies standard single-label similarity-based techniques to obtain multi-label patient characterizations. Then, it employs these characterizations to train single-sample predictors using different multi-label strategies and find the best-performing classifiers. RESULTS MULTI-STAR classifiers offer advanced multi-label recognition of all the subtypes contributing to the molecular and clinical traits of a patient, also distinguishing the primary from the additional relevant secondary subtype(s). The efficacy was demonstrated by developing multi-label solutions for breast and colorectal cancer subtyping that outperform existing methods in terms of prognostic value, primarily for overall survival predictions, and ability to work on a single sample at a time, as required in clinical practice. CONCLUSIONS This work emphasizes the importance of moving to multi-label subtyping to capture all the molecular traits of individual patients, considering also previously overlooked secondary assignments and paving the way for improved clinical decision-making processes in diverse heterogeneous disease contexts. Indeed, MULTI-STAR novel, reproducible and generalizable approach provides comprehensive representations of patient inner heterogeneity and clinically relevant insights, contributing to precision medicine and personalized treatments.
Collapse
Affiliation(s)
- Silvia Cascianelli
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Piazza Leonardo da Vinci, 32, Milano, 20133, Italy.
| | - Iva Milojkovic
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Piazza Leonardo da Vinci, 32, Milano, 20133, Italy
| | - Marco Masseroli
- Dipartimento di Elettronica, Informazione e Bioingegneria, Politecnico di Milano, Piazza Leonardo da Vinci, 32, Milano, 20133, Italy
| |
Collapse
|
8
|
Mondaca JM, Muñoz JMF, Barraza GA, Vanderhoeven F, Redondo AL, Flamini MI, Sanchez AM. Therapeutic potential of GNRHR analogs and SRC/FAK inhibitors to counteract tumor growth and metastasis in breast cancer. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167826. [PMID: 40189112 DOI: 10.1016/j.bbadis.2025.167826] [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: 11/21/2024] [Revised: 03/27/2025] [Accepted: 03/31/2025] [Indexed: 04/09/2025]
Abstract
Breast cancer (BC) is the leading cause of cancer death in women, with hormone-dependent BC accounting for about 80 % of cases, primarily affecting postmenopausal women with gonadotropins, luteinizing hormone (LH), and follicle-stimulating hormone (FSH) elevated. Treatments targeting the gonadotropin-releasing hormone receptor (GnRHR), such as the agonist leuprorelin (LEU) and antagonist degarelix (DEGA), are used for hormone-dependent tumors. While the functional role of gonadotropin receptors in extragonadal tissues remains uncertain, recent studies suggest LH contributes to tumor development and progression. Tumor progression involves reorganization in the actin cytoskeleton, induction of adhesion, and cell migration, driven by proteins such as Src and the focal adhesion kinase (FAK), which are related to invasive behaviors. The overexpression of both protein kinases generates an invasive and metastatic phenotype, then inhibitors targeting Src (PP2) and FAK (FAKi) have been developed to counteract this effect. This study combined GnRH analogs with Src and FAK inhibitors to target BC progression. We found that LH treatment influenced gene expression linked to tumor development. Examining the GnRHR-LEU and GnRHR-DEGA complexes revealed structural differences affecting ligand binding. In an orthotopic tumor model, DEGA reduced tumor growth, while LEU had the opposite effect. Combining DEGA with PP2 or FAKi enhanced tumor inhibition, improving mice survival. These findings provide valuable insights into the essential regulatory role of gonadotropins in genes involved in tumorigenic processes, highlighting the potential of GnRHR antagonists combined with Src or FAK inhibitors as a promising strategy to develop new drugs that interfere with the ability of breast tumor progression.
Collapse
Affiliation(s)
- Joselina Magali Mondaca
- Laboratorio de Transducción de Señales y Movimiento Celular, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Juan Manuel Fernandez Muñoz
- Departamento de Laboratorio de Salud Pública, Ministerio de Salud y Deportes, Gobierno de Mendoza, Mendoza, Argentina
| | - Gustavo Adolfo Barraza
- Laboratorio de Transducción de Señales y Movimiento Celular, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Fiorella Vanderhoeven
- Laboratorio de Biología Tumoral, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Analía Lourdes Redondo
- Laboratorio de Biología Tumoral, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Marina Inés Flamini
- Laboratorio de Biología Tumoral, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina.
| | - Angel Matias Sanchez
- Laboratorio de Transducción de Señales y Movimiento Celular, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Nacional de Cuyo, Mendoza, Argentina.
| |
Collapse
|
9
|
Aghebati M, Hossieni R, Makeh AS, Shirzadi A, Akbari ME. Ki-67 and 21-gene recurrence score assay in decision making for adjuvant chemotherapy in breast cancer patients. Discov Oncol 2025; 16:970. [PMID: 40448815 DOI: 10.1007/s12672-025-02233-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 03/25/2025] [Indexed: 06/02/2025] Open
Abstract
Although significant advances have been made in the molecular subtyping of breast cancers, identification of patients who do not benefit from the chemotherapy is a major challenge. Pioneer studies have examined the predictive value of the clinicopathological factors, such as tumor size, disease stage, the expression levels of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2) molecules and more importantly tumor cells proliferation index (Ki-67) to help guide patients' treatment and predict their outcome in the adjuvant chemotherapy setting. However, despite their clinical importance, no consensus is reached on their validity for chemotherapy decision. These challenges have ignited researchers to evaluate genomic signatures, which has led to the introduction of several genomic tests that can now help oncologists to include/exclude chemotherapy from the treatment regimen with more confidence. The present review aims to look back over the literature on the clinical significance of Ki-67 as well as the 21-gene recurrence score assay in identification of breast cancer patients who may benefit from the adjuvant chemotherapy.
Collapse
Affiliation(s)
- Mohammad Aghebati
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Hossieni
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Afsaneh Sadat Makeh
- Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Shirzadi
- Department of Surgery, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| | | |
Collapse
|
10
|
van Geel JJL, Jongbloed EM, Moustaquim J, van der Schoor G, van Leeuwen-Stok AE, Smid M, van Deurzen CHM, Wilting SM, Wesseling J, Sonke GS, Martens JWM, Schröder CP. Clinicopathological and molecular characterization of inflammatory breast cancer, the prospective INFLAME registry study. NPJ Breast Cancer 2025; 11:48. [PMID: 40442132 PMCID: PMC12122667 DOI: 10.1038/s41523-025-00764-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Accepted: 05/13/2025] [Indexed: 06/02/2025] Open
Abstract
Inflammatory breast cancer (IBC) is rare, with challenging diagnostics and unfavorable outcomes. Therefore, more molecular insight into IBC is needed. The comprehensive Dutch prospective INFLAME registry related IBC follow-up and treatment to histopathology and molecular analysis. Of consecutive patients, nationwide identified with newly diagnosed IBC, clinicopathological, treatment and outcome data were collected. Histopathology and RNA-sequencing were related to outcome. 125 IBC patients were enrolled. Forty-one (34%) patients had HER2 + , and 31 (25%) had triple-negative IBC. The estimated 3-year OS was 78% in M0 IBC and 29% in M1. PFS was worst in triple-negative IBC (median 7.9 vs 16.3 and 15.8 months in M1 HER2+ and HR + /HER2- IBC). DFS and OS in M0 IBC were better with guideline-concordant trimodal therapy than without (HR 0.15 and 0.15; p = 0.000005 and 0.00038). The unique prospective INFLAME confirms unfavorable IBC characteristics and outcomes. International efforts may support guideline adherence and identify IBC-specific targets.
Collapse
Affiliation(s)
- Jasper J L van Geel
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Elisabeth M Jongbloed
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jasmine Moustaquim
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | - Marcel Smid
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | | | - Saskia M Wilting
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Jelle Wesseling
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Divisions of Diagnostic Oncology and Molecular Pathology, Netherlands Cancer Institute, Amsterdam, The Netherlands & Department of Pathology, Leiden University Medical Center, Leiden, The Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - John W M Martens
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Carolina P Schröder
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands.
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands.
| |
Collapse
|
11
|
Antov GG, Gospodinova ZI, Novakovic M, Tesevic V, Krasteva NA, Pavlov DV, Valcheva-Kuzmanova SV. Molecular mechanisms of the anticancer action of fustin isolated from Cotinus coggygria Scop. in MDA-MB-231 triple-negative breast cancer cell line. Z NATURFORSCH C 2025; 80:233-250. [PMID: 39331583 DOI: 10.1515/znc-2024-0140] [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: 06/12/2024] [Accepted: 09/10/2024] [Indexed: 09/29/2024]
Abstract
The aim of the present work was to investigate some of the molecular mechanisms and targets of the anticancer action of the bioflavonoid fustin isolated from the heartwood of Cotinus coggygria Scop. in the triple-negative breast cancer cell line MDA-MB-231. For this purpose, we applied fluorescence microscopy analysis to evaluate apoptosis, necrosis, and mitochondrial integrity, wound healing assay to study fustin antimigratory potential and quantitative reverse transcription-polymerase chain reaction to analyze the expression of genes associated with cell cycle control, programmed cell death, metastasis, and epigenetic alterations. A complex network-based bioinformatic analysis was also employed for protein-protein network construction, hub genes identification, and functional enrichment. The results revealed a significant induction of early and late apoptotic and necrotic events, a slight alteration of the mitochondria-related fluorescence, and marked antimotility effect after fustin treatment. Of 34 analyzed genes, seven fustin targets were identified, of which CDKN1A, ATM, and MYC were significantly enriched in pathways such as cell cycle, intrinsic apoptotic signaling pathway in response to DNA damage and generic transcription pathway. Our findings outline some molecular mechanisms of the anticancer action of fustin pointing it out as a potential oncotherapeutic agent and provide directions for future in vivo research.
Collapse
Affiliation(s)
- Georgi G Antov
- Laboratory of Genome Dynamics and Stability, Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Zlatina I Gospodinova
- Laboratory of Genome Dynamics and Stability, Institute of Plant Physiology and Genetics, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Miroslav Novakovic
- Department of Chemistry, University of Belgrade - Institute of Chemistry, Technology and Metallurgy, National Institute of the Republic of Serbia, Belgrade, Serbia
| | - Vele Tesevic
- University of Belgrade - Faculty of Chemistry, Belgrade, Serbia
| | - Natalia A Krasteva
- Department of Electroinduced and Adhesive Properties, Institute of Biophysics and Biomedical Engineering, Bulgarian Academy of Sciences, Sofia, Bulgaria
| | - Danail V Pavlov
- Department of Biochemistry, Molecular Medicine and Nutrigenomics with Laboratory of Nutrigenomics, Functional Foods and Nutraceuticals, Faculty of Pharmacy, Medical University "Prof. Dr. Paraskev Stoyanov", Varna, Bulgaria
| | - Stefka V Valcheva-Kuzmanova
- Department of Pharmacology and Clinical Pharmacology and Therapeutics, Faculty of Medicine, Medical University "Prof. Dr. Paraskev Stoyanov", Varna, Bulgaria
| |
Collapse
|
12
|
Stucchi S, Borea R, Garcia-Recio S, Zingarelli M, Rädler PD, Camerini E, Marnata Pellegry C, O'Connor S, Earp HS, Carey LA, Perou CM, Savoldo B, Dotti G. B7-H3 and CSPG4 co-targeting as Pan-CAR-T cell treatment of triple-negative breast cancer. J Immunother Cancer 2025; 13:e011533. [PMID: 40425233 PMCID: PMC12107568 DOI: 10.1136/jitc-2025-011533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 05/12/2025] [Indexed: 05/29/2025] Open
Abstract
PURPOSE Chimeric antigen receptor T (CAR-T) cell therapy is under clinical investigation in patients with metastatic triple-negative breast cancer (TNBC). However, the identification of targetable antigens remains a high priority to avoid toxicity and prevent tumor escape. EXPERIMENTAL DESIGN Here we analyzed the gene expression of B7-H3 (CD276) and chondroitin sulfate proteoglycan 4 (CSPG4) in 98 TNBC samples identified in the AURORA US Network and Rapid Autopsy RNA sequencing data set at University of North Carolina (UNC). We then performed immunohistochemistry analysis for B7-H3 and CSPG4 protein expression in 151 TNBC samples collected at UNC. Finally, the validity of the proposed B7-H3 and CSGP4 co-targeting was tested in clinically relevant TNBC patient derived xenograft (PDX) models. RESULTS We observed that CD276 and CSPG4 genes are broadly and comparably expressed in TNBC samples, and gene expression is generally conserved in tumor metastases. None of the TNBC analyzed met the criteria for simultaneous low expression of CSPG4 and CD276 genes. Immunohistochemistry analysis showed a median H-score of 138 (105-168, lower and upper quartile, respectively) for B7-H3 expression and a median H-score of 33 (14-78 lower and upper quartile, respectively) for CSPG4 expression. Notably, 49% of the TNBC cores with B7-H3 H-score ≤105 exhibited a CSPG4 H-score exceeding its median value, and 37% and 18% of the TNBC cores with low B7-H3 expression scored CSPG4 expression above its median H-score or exceeded its upper quartile, respectively, confirming that at least one of these two proteins is expressed in 94% of the analyzed tumors. Finally, optimized dual-specific B7-H3 and CSPG4 CAR-T cells eradicated tumors with mixed antigen expression in TNBC PDX models. CONCLUSIONS These data highlight the clinical potential of the proposed approach that could be applicable to the great majority of patients with TNBC as well as most of patients with breast cancer in general.
Collapse
Affiliation(s)
- Simone Stucchi
- Lineberger Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Roberto Borea
- Lineberger Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Susana Garcia-Recio
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Manuela Zingarelli
- Lineberger Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Patrick D Rädler
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Elena Camerini
- Lineberger Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | - Siobhan O'Connor
- Pathology & Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - H Shelton Earp
- Lineberger Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Lisa A Carey
- Division of Oncology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Charles M Perou
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Barbara Savoldo
- Lineberger Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Pediatrics, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Gianpietro Dotti
- Lineberger Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
13
|
Cheung AM, Wang D, Quintayo MA, Yerofeyeva Y, Spears M, Bartlett JMS, Stein L, Bayani J, Yaffe MJ. Intra-tumoral spatial heterogeneity in breast cancer quantified using high-dimensional protein multiplexing and single cell phenotyping. Breast Cancer Res 2025; 27:88. [PMID: 40399910 PMCID: PMC12096620 DOI: 10.1186/s13058-025-02038-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/29/2025] [Indexed: 05/23/2025] Open
Abstract
BACKGROUND Breast cancer is a highly heterogeneous disease where variations of biomarker expression may exist between individual foci of a cancer (intra-tumoral heterogeneity). The extent of variation of biomarker expression in the cancer cells, distribution of cell types in the local tumor microenvironment and their spatial arrangement could impact on diagnosis, treatment planning and subsequent response to treatment. METHODS Using quantitative multiplex immunofluorescence (MxIF) imaging, we assessed the level of variations in biomarker expression levels among individual cells, density of cell cluster groups and spatial arrangement of immune subsets from regions sampled from 38 multi-focal breast cancers that were processed using whole-mount histopathology techniques. Molecular profiling was conducted to determine the intrinsic molecular subtype of each analysed region. RESULTS A subset of cancers (34.2%) showed intra-tumoral regions with more than one molecular subtype classification. High levels of intra-tumoral variations in biomarker expression levels were observed in the majority of cancers studied, particularly in Luminal A cancers. HER2 expression quantified with MxIF did not correlate well with HER2 gene expression, nor with clinical HER2 scores. Unsupervised clustering revealed the presence of various cell clusters with unique IHC4 protein co-expression patterns and the composition of these clusters were mostly similar among intra-tumoral regions. MxIF with immune markers and image patch analysis classified immune niche phenotypes and the prevalence of each phenotype in breast cancer subtypes was illustrated. CONCLUSIONS Our work illustrates the extent of spatial heterogeneity in biomarker expression and immune phenotypes, and highlights the importance of a comprehensive spatial assessment of the disease for prognosis and treatment planning.
Collapse
Affiliation(s)
- Alison M Cheung
- Biomarker Imaging Research Lab (BIRL), Sunnybrook Research Institute, Rm S658, 2075 Bayview Avenue, Toronto, ON, Canada
| | - Dan Wang
- Biomarker Imaging Research Lab (BIRL), Sunnybrook Research Institute, Rm S658, 2075 Bayview Avenue, Toronto, ON, Canada
| | - Mary Anne Quintayo
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Yulia Yerofeyeva
- Biomarker Imaging Research Lab (BIRL), Sunnybrook Research Institute, Rm S658, 2075 Bayview Avenue, Toronto, ON, Canada
| | - Melanie Spears
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - John M S Bartlett
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, ON, Canada
- University of Edinburgh, Edinburgh, UK
| | - Lincoln Stein
- Informatics and Bio-Computing, Ontario Institute for Cancer Research, Toronto, ON, Canada
| | - Jane Bayani
- Diagnostic Development, Ontario Institute for Cancer Research, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Martin J Yaffe
- Biomarker Imaging Research Lab (BIRL), Sunnybrook Research Institute, Rm S658, 2075 Bayview Avenue, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
14
|
Sridharan P, Ghosh M. Gene expression and agent-based modeling improve precision prognosis in breast cancer. Sci Rep 2025; 15:17059. [PMID: 40379718 DOI: 10.1038/s41598-025-01275-w] [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: 09/25/2024] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
Breast cancer survival is hard to predict because of the complex ways genes and cells interact. This study offers a new method to improve these predictions by combining gene expression profiling (GEP) with agent-based modeling (ABM). First, GEP will pinpoint genes that are important in breast cancer development. Then, a mathematical model will be built to show how these genes influence cell behavior. This data will be used in ABM to simulate tumor growth and treatment response. The ABM allows us to virtually test different treatments and see how they might affect patient survival. Finally, the model's accuracy will be checked against real patient data and compared to other models. By combining the strengths of GEP and ABM, this research could significantly improve breast cancer survival prediction. ABM's ability to analyze interactions mathematically could pave the way for more personalized and effective treatments.
Collapse
Affiliation(s)
- Padmasri Sridharan
- Department of Mathematics, School of Advanced Sciences, Vellore Institute of Technology, Chennai, 600127, India
| | - Mini Ghosh
- Department of Mathematics, School of Advanced Sciences, Vellore Institute of Technology, Chennai, 600127, India.
| |
Collapse
|
15
|
Van Swearingen AED, Lee MR, Rogers LW, Sibley AB, Shi P, Qin X, Goodin M, Seale K, Owzar K, Anders CK. Genomic and immune profiling of breast cancer brain metastases. Acta Neuropathol Commun 2025; 13:99. [PMID: 40355907 PMCID: PMC12070617 DOI: 10.1186/s40478-025-02001-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 04/06/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Brain metastases (BrM) arising from breast cancer (BC) are an increasing consequence of advanced disease, with up to half of patients with metastatic HER2 + or triple negative BC experiencing central nervous system (CNS) recurrence. The genomic alterations driving CNS recurrence, along with contributions of the immune microenvironment, particularly by intrinsic subtype, remain unclear. METHODS We characterized the genomic and immune landscape of BCBrM from a cohort of 42 patients by sequencing whole-exome DNA (WES) and total RNA libraries from frozen and FFPE BrM and FFPE extracranial tumors (ECT). Analyses included PAM50 intrinsic subtypes, somatic mutations, copy number variations (CNV), pathway alterations, immune cell type deconvolution, and associations with clinical outcomes RESULTS: Intrinsic subtype calls were concordant for the majority of BrM-ECT pairs (60%). Across all BrM and ECT samples, the most common somatic gene mutation was TP53 (64%, 30/47). For patients with matched FFPE BrM-FFPE ECT, alterations tended to be conserved across tissue type, although differential somatic mutations and CNV in specific genes were observed. Several genomic pathways were differentially expressed between patient-matched BrM-ECT; MYC targets, DNA damage repair, cholesterol homeostasis, and oxidative phosphorylation were higher in BrM, while immune-related pathways were lower in BrM. Deconvolution of immune populations between BrM-ECT demonstrated activated dendritic cell populations were higher in BrM compared to ECT. Increased expression of several oncogenic preselected pathways in BrM were associated with inferior survival, including DNA damage repair, inflammatory response, and oxidative phosphorylation CONCLUSIONS: Collectively, this study illustrates that while some genomic alterations are shared between BrM and ECT, there are also unique aspects of BrM including somatic mutations, CNV, pathway alterations, and immune landscape. A deeper understanding of differences inherent to BrM will contribute to the development of BrM-tailored therapeutic strategies. Additional analyses are warranted in larger cohorts, particularly with additional matched BrM-ECT.
Collapse
Affiliation(s)
| | - Marissa R Lee
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Layne W Rogers
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Alexander B Sibley
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Pixu Shi
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, USA
| | - Xiaodi Qin
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Michael Goodin
- Duke Center for Brain and Spine Metastasis, Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Katelyn Seale
- Duke Cancer Institute, Duke University Hospital, Durham, NC, USA
| | - Kouros Owzar
- Department of Biostatistics and Bioinformatics, Duke Center for Brain and Spine Metastasis, Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Carey K Anders
- Department of Medical Oncology, Duke Center for Brain and Spine Metastasis, Duke Cancer Institute, Duke University, 10 Searle Center Drive, Campus Box 3881, Durham, NC, 27710, USA.
| |
Collapse
|
16
|
Mouneimne G, Connors C, Watson A, Grant A, Campo D, Ring A, Kaur P, Lang JE. Mechanical Conditioning (MeCo) Score Progressively Increases Through the Metastatic Cascade in Breast Cancer via Circulating Tumor Cells. Cancers (Basel) 2025; 17:1632. [PMID: 40427129 PMCID: PMC12109637 DOI: 10.3390/cancers17101632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/15/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND The mechanical conditioning (MeCo) score is a multigene expression signature that is acquired by cancer cells in the primary breast tumor and is reflective of their responsiveness to ECM stiffness caused by tumor fibrosis. Chromatin remodeling downstream of mechanotransduction allows cancer cells to retain these acquired aggressive features even in the absence of mechanical stimulation from the primary tumor microenvironment, for instance, after dissemination through systemic circulation during metastasis. Importantly, patients who have high MeCo score tumors are at higher risk of developing metastatic breast cancer, compared to those with low MeCo scores. Moreover, circulating tumor cells (CTCs) are associated with a higher rate of metastatic dissemination, making CTC detection in the circulation of patients with breast cancer a significant prognostic biomarker for breast cancer metastasis. Beyond their enumeration per blood volume units, specific prognostic features of CTCs are not fully explored. We sought to determine whether MeCo scores increase stepwise along the metastatic cascade, from primary tumors to CTCs to distant metastatic colonization, using patient-matched biopsies. METHODS CTCs were isolated from the peripheral blood of two patient cohorts: patients with early-stage breast cancer using immunomagnetic enrichment/FACS methodology; and patients with late-stage breast cancer using the ANGLE Parsortix microfluidics system. Gene expression profiling using RNA-seq was performed on CTCs and matched primary tumors (PTs) in the early-stage cohort, and on CTCs and matched metastases (METs) for the late-stage cohorts. A quantile normalization approach was used to allow comparison across cohorts and MeCo scores were computed for all samples. The Wilcoxon matched-pairs signed rank test was performed for the comparison of MeCo scores from matching samples within each cohort; the Mann-Whitney unpaired test was used to compare MeCo scores of CTCs across cohorts. RESULTS In 12 pairs of patients with early-stage breast cancer, MeCo scores in CTCs were significantly higher than in their matched PTs (p = 0.026). Additionally, in 26 pairs of metastatic patient CTCs and METs, MeCo scores were significantly higher in METs compared to matched CTCs (p = 0.0004). MeCo scores of CTCs were similar between patients with early- and late-stage breast cancers, despite differing CTC isolation strategies (epitope-dependent and microfluidics size gradient). Notably, 98% of the genes in the MeCo score were present across evaluable CTC, MET, and PT samples. CONCLUSIONS Our results show that the MeCo score is higher in CTCs than in PTs, and higher in METs compared to CTCs, in early- and late-stage breast cancer, respectively (i.e., PT < CTC < MET). Therefore, the MeCo score is progressively higher throughout the metastatic cascade in breast cancer. These findings demonstrate that mechanical conditioning from primary tumors is retained during metastatic progression, after mechanical induction by ECM stiffness is lost, as cancer cells disseminate through systemic circulation. Additionally, these findings support that cancer cells with higher MeCo scores are more competent with-and potentially selected for-metastatic progression. Importantly, these findings provide a novel feature of CTCs, mechanical conditioning (MeCo), which is associated with higher capacity for metastasis. Furthermore, since the CTC MeCo score is elevated even in early-stage breast cancer, it could provide, in addition to CTC enumeration, a potential prognostic indicator to improve metastatic risk assessment in early disease.
Collapse
Affiliation(s)
- Ghassan Mouneimne
- University of Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Casey Connors
- Division of Breast Surgery, Cleveland Clinic, Lerner College of Medicine, 9500 Euclid Ave., Cleveland, OH 44195, USA
| | - Adam Watson
- MeCo Diagnostics, San Diego, CA 92103, USA; (A.W.)
| | - Adam Grant
- MeCo Diagnostics, San Diego, CA 92103, USA; (A.W.)
| | - Daniel Campo
- Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Ave., Los Angeles, CA 90033, USA (P.K.)
| | - Alexander Ring
- Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Ave., Los Angeles, CA 90033, USA (P.K.)
- Department of Medical Oncology and Hematology, University Hospital Zürich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Pushpinder Kaur
- Norris Comprehensive Cancer Center, University of Southern California, 1441 Eastlake Ave., Los Angeles, CA 90033, USA (P.K.)
| | - Julie E. Lang
- Division of Breast Surgery, Cleveland Clinic, Lerner College of Medicine, 9500 Euclid Ave., Cleveland, OH 44195, USA
| |
Collapse
|
17
|
Ciringione A, Rizzi F. Facing the Challenge to Mimic Breast Cancer Heterogeneity: Established and Emerging Experimental Preclinical Models Integrated with Omics Technologies. Int J Mol Sci 2025; 26:4572. [PMID: 40429718 PMCID: PMC12111172 DOI: 10.3390/ijms26104572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/05/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Breast cancer (BC) is among the most common neoplasms globally and is the leading cause of cancer-related mortality in women. Despite significant advancements in prevention, early diagnosis, and treatment strategies made over the past two decades, breast cancer continues to pose a significant global health challenge. One of the major obstacles in the clinical management of breast cancer patients is the high intertumoral and intratumoral heterogeneity that influences disease progression and therapeutic outcomes. The inability of preclinical experimental models to replicate this diversity has hindered the comprehensive understanding of BC pathogenesis and the development of new therapeutic strategies. An ideal experimental model must recapitulate every aspect of human BC to maintain the highest predictive validity. Therefore, a thorough understanding of each model's inherent characteristics and limitations is essential to bridging the gap between basic research and translational medicine. In this context, omics technologies serve as powerful tools for establishing comparisons between experimental models and human tumors, which may help address BC heterogeneity and vulnerabilities. This review examines the BC models currently used in preclinical research, including cell lines, patient-derived organoids (PDOs), organ-on-chip technologies, carcinogen-induced mouse models, genetically engineered mouse models (GEMMs), and xenograft mouse models. We emphasize the advantages and disadvantages of each model and outline the most important applications of omics techniques to aid researchers in selecting the most relevant model to address their specific research questions.
Collapse
Affiliation(s)
- Alessia Ciringione
- Laboratory of Biochemistry, Molecular Biology and Oncometabolism, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy;
| | - Federica Rizzi
- Laboratory of Biochemistry, Molecular Biology and Oncometabolism, Department of Medicine and Surgery, University of Parma, Via Volturno 39, 43125 Parma, Italy;
- National Institute of Biostructure and Biosystems (INBB), 00165 Rome, Italy
| |
Collapse
|
18
|
Lee YW, Lee SB, Chung IY, Kim J, Kim HJ, Ko BS, Son BH, Lee JW, Yoo TKR. Exploring the efficacy of extended endocrine therapy in pure mucinous breast carcinoma. Breast 2025; 82:104492. [PMID: 40349526 DOI: 10.1016/j.breast.2025.104492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 04/26/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND This study aims to compare the efficacy of 5- versus 10-year endocrine therapy in pure mucinous breast carcinoma (PMBC), focusing on late recurrence and related factors for personalized treatment. METHODS Patients with PMBC who underwent surgery from 1996 to 2014 at Asan Medical Center were included. Recurrence was categorized as early (<5 years) or late (≥5 years). The primary endpoint was disease-free survival in the 5- and 10- year endocrine groups. Subgroup analysis was performed focused on clinically high-risk patients (tumor ≥2 cm, nodal metastasis, or high histologic grade). RESULTS A total of 489 patients with PMBC were identified. During a follow-up time of 126 months, 35 (7.2 %) patients had an early recurrence, 25 (5.1 %) patients had a late recurrence, and 394 (87.7 %) patients had no recurrence. High histologic grade was the only factor significantly correlated to late recurrence (hazard ratio 6.92, 95 % confidence interval 1.53-31.3). Among the 5-year disease-free survivors (N = 416), 340 (81.7 %) and 76 (18.3 %) patients underwent 5-year and 10-year endocrine therapy, respectively. Endocrine therapy duration did not impact the 10-year disease-free survival rate (5-year [95.4 %] vs. 10-year [97.3 %] endocrine therapy, log-rank test p = 0.504). Subgroup analysis with clinically high-risk patients revealed no survival difference based on the endocrine therapy duration, too. CONCLUSION Extended endocrine therapy did not significantly reduce late recurrence in PMBC, even in high-risk groups, underscoring the importance of personalized strategies for sustained outcomes.
Collapse
Affiliation(s)
- Young-Won Lee
- Division of Breast and Endocrine Surgery, Department of Surgery, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea; Division of Breast Surgery, Department of Surgery, Ulsan University College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Sae-Byul Lee
- Division of Breast Surgery, Department of Surgery, Ulsan University College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Il Yong Chung
- Division of Breast Surgery, Department of Surgery, Ulsan University College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jisun Kim
- Division of Breast Surgery, Department of Surgery, Ulsan University College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Hee Jeong Kim
- Division of Breast Surgery, Department of Surgery, Ulsan University College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Beom Seok Ko
- Division of Breast Surgery, Department of Surgery, Ulsan University College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Byung Ho Son
- Division of Breast Surgery, Department of Surgery, Ulsan University College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Jong Won Lee
- Division of Breast Surgery, Department of Surgery, Ulsan University College of Medicine, Asan Medical Center, Seoul, Republic of Korea
| | - Tae-Kyung Robyn Yoo
- Division of Breast Surgery, Department of Surgery, Ulsan University College of Medicine, Asan Medical Center, Seoul, Republic of Korea.
| |
Collapse
|
19
|
Cheng Y, Liu B, Xin J, Wu X, Li W, Shang J, Wu J, Zhang Z, Xu B, Du M, Cheng G, Wang M. Single-cell and spatial RNA sequencing identify divergent microenvironments and progression signatures in early- versus late-onset prostate cancer. NATURE AGING 2025; 5:909-928. [PMID: 40211000 DOI: 10.1038/s43587-025-00842-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 02/26/2025] [Indexed: 04/12/2025]
Abstract
The clinical and pathological outcomes differ between early-onset (diagnosed in men ≤55 years of age) and late-onset prostate cancer, potentially attributed to the changes in hormone levels and immune activities associated with aging. Exploring the heterogeneity therein holds potential for developing age-specific precision interventions. Here, through single-cell and spatial transcriptomic analyses of prostate cancer tissues, we identified that an androgen response-related transcriptional meta-program (AR-MP) might underlie the age-related heterogeneity of tumor cells and microenvironment. APOE+ tumor-associated macrophages infiltrated AR-MP-activated tumor cells in early-onset prostate cancer, potentially facilitating tumor progression and immunosuppression. By contrast, inflammatory cancer-associated fibroblasts in late-onset prostate cancer correlated with downregulation of AR-MP of tumor cells and increased epithelial-to-mesenchymal transition and pre-existing castration resistance, which may also be linked to smoking. This study provides potential insights for tailoring precision treatments by age groups, emphasizing interventions that include targeting AR and tumor-associated macrophages in young patients but anchoring epithelial-to-mesenchymal transition and inflammatory cancer-associated fibroblasts in old counterparts.
Collapse
Affiliation(s)
- Yifei Cheng
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Urology, Southeast University Zhongda Hospital, Nanjing, China
| | - Bingxin Liu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Junyi Xin
- Department of Bioinformatics, School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, China
| | - Xiaobin Wu
- Department of Pathology, The Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, China
| | - Wenchao Li
- Department of Urology, Southeast University Zhongda Hospital, Nanjing, China
| | - Jinwei Shang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University & Jiangsu Province People's Hospital, Nanjing, China
| | - Jiajin Wu
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhengdong Zhang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Bin Xu
- Department of Urology, Southeast University Zhongda Hospital, Nanjing, China.
| | - Mulong Du
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, China.
| | - Gong Cheng
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University & Jiangsu Province People's Hospital, Nanjing, China.
| | - Meilin Wang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, China.
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China.
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China.
| |
Collapse
|
20
|
Sota Y, Seno S, Naoi Y, Honma K, Shimoda M, Tanei T, Matsuda H, Shimazu K. IRSN-23 gene diagnosis enhances breast cancer subtype classification and predicts response to neoadjuvant chemotherapy: new validation analyses. Breast Cancer 2025; 32:566-581. [PMID: 40128415 PMCID: PMC11993443 DOI: 10.1007/s12282-025-01687-6] [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: 10/09/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND This study evaluates the reproducibility of the IRSN-23 model, which classifies patients into highly chemotherapy-sensitive (Gp-R) or less-sensitive (Gp-NR) groups based on immune-related gene expression using DNA microarray analysis, and its impact on breast cancer subtype classification. METHODS Tumor tissues from 146 breast cancer patients receiving neoadjuvant chemotherapy (paclitaxel-FEC) ± trastuzumab at Osaka University Hospital (OUH) were used to classify patients into Gp-R or Gp-NR using IRSN-23. The ability to predict a pathological complete response (pCR) was assessed and the results were validated with independent public datasets (N = 1282). RESULTS In the OUH dataset, the pCR rate was significantly higher in the Gp-R group than in the Gp-NR group without trastuzumab (29 versus 1%, P = 1.70E-5). In all validation sets without anti-HER2 therapy, the pCR rate in the Gp-R group was significantly higher than that in the Gp-NR group. The pooled analysis of the validation set showed higher pCR rates in the Gp-R group than in the Gp-NR group, both without (N = 1103, 40 versus 12%, P = 2.02E-26) and with (N = 304, 49 versus 35%, P = 0.017) anti-HER2 therapy. Collaboration analyses of IRSN-23 and Oncotype Dx or PAM50 could identify highly chemotherapy-sensitive groups and refine breast cancer subtype classification based on the tumor microenvironment (offensive factor-PAM50 and defensive factor-IRSN-23), and the immune subtype was correlated with a better prognosis after NAC. CONCLUSIONS This study offers new validation analyses of IRSN-23 in predicting chemotherapy efficacy, showing high reproducibility. The findings indicate the clinical value of using IRSN-23 for refining breast cancer subtype classification, with implications for personalized treatment strategies and improved patient outcomes.
Collapse
Affiliation(s)
- Yoshiaki Sota
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan.
- Department of Breast and Endocrine Surgery, Osaka University Graduate School of Medicine, 2-2-E10 Yamadaoka, Suita, Osaka, 565-0871, Japan.
| | - Shigeto Seno
- Department of Bioinformatic Engineering, Graduateschool of Information Scienceand Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yasuto Naoi
- Department of Surgery, Divisionof Endocrineand BreastSurgery, Kyoto Prefectural University of Medicine, 465 Kawaramachi-hirokoji, Kamigyo-ku, Kyoto, 602-8566, Japan
| | - Keiichiro Honma
- Department of Diagnostic Pathology and Cytology, Osaka International Cancer Institute, 3-1-69 Otemae, Chuo-ku, Osaka, Osaka, 541-8567, Japan
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Tomonori Tanei
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hideo Matsuda
- Department of Bioinformatic Engineering, Graduateschool of Information Scienceand Technology, Osaka University, 1-5 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, 2-15 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
21
|
Winham SJ, McCarthy AM, Scott CG, Gastounioti A, Horng H, Norman AD, Mankowski WC, Pantalone L, Jensen MR, Acciavatti RJ, Maidment ADA, Cohen EA, Brandt KR, Conant EF, Kerlikowske KM, Kontos D, Vachon CM. Radiomic Parenchymal Phenotypes of Breast Texture from Mammography and Association with Risk of Breast Cancer. Radiology 2025; 315:e240281. [PMID: 40358450 DOI: 10.1148/radiol.240281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Background Parenchymal phenotypes reflect the intrinsic heterogeneity of both tissue structure and distribution on mammograms. Purpose To define parenchymal phenotypes on the basis of radiomic texture features derived from full-field digital mammography (FFDM) in breast screening populations and assess associations of parenchymal phenotypes with future risk of breast cancer and masking (false-negative [FN] findings or interval cancers), beyond breast density, and by race and ethnicity Materials and Methods A two-stage study design included a retrospective cross-sectional study of 30 000 randomly selected women with four-view FFDM (mean age, 57.4 years) and a nested case-control study of 1055 women with invasive breast cancer (151 Black and 893 White women) matched to 2764 women without breast cancer (411 Black and 2345 White women) (mean age, 60.4 years) sampled from April 2008 to September 2019 from three diverse breast screening practices. Radiomic features (n = 390) were extracted and standardized using an automated pipeline and adjusted for age and practice. Variation was classified using hierarchical clustering and principal component (PC) analysis. The resulting clusters and PCs were examined for association with invasive breast cancer risk, FN findings on mammograms, and symptomatic interval cancers beyond radiologist-reported Breast Imaging Reporting and Data System (BI-RADS) breast density using conditional logistic regression and likelihood ratio tests. Discrimination for breast cancer was assessed with area under the receiver operating characteristic curve (AUC). Results Six clusters and six PCs were defined, replicated, and associated with a higher risk of invasive breast cancer (P = .01 and P < .001, respectively) after adjustment for age, body mass index (calculated as weight in kilograms divided by height in meters squared), and BI-RADS breast density. PCs showed similar associations among Black and White women (P = .23). PCs were also positively associated with FN findings (P = .004) and symptomatic interval cancers (P = .006). AUC improved for all breast cancer end points when incorporating PCs, with the greatest improvement shown in prediction of FN findings (AUC with vs without PCs, 0.73 [95% CI: 0.68, 0.78] vs 0.66 [95% CI: 0.61, 0.71] , respectively; P = .004) and symptomatic interval cancers (AUC with vs without PCs, 0.77 [95% CI: 0.71, 0.82] vs 0.68 [95% CI: 0.62, 0.74], respectively; P = .006). Conclusion Parenchymal phenotypes based on radiomic features extracted from FFDM were associated with a higher risk of invasive breast cancer, specifically for FN findings and symptomatic interval cancer. © RSNA, 2025 Supplemental material is available for this article. See also the editorial by Mesurolle and El Khoury in this issue.
Collapse
Affiliation(s)
- Stacey J Winham
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First St SW, Biobusiness Bldg 5-81, Rochester, MN 55905
| | - Anne Marie McCarthy
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Philadelphia, Pa
| | - Christopher G Scott
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First St SW, Biobusiness Bldg 5-81, Rochester, MN 55905
| | | | - Hannah Horng
- Department of Radiology, University of Pennsylvania, Philadelphia, Pa
| | - Aaron D Norman
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First St SW, Biobusiness Bldg 5-81, Rochester, MN 55905
| | - Walter C Mankowski
- Department of Radiology, University of Pennsylvania, Philadelphia, Pa
- Department of Radiology, Columbia University, New York, NY
| | - Lauren Pantalone
- Department of Radiology, University of Pennsylvania, Philadelphia, Pa
| | - Matthew R Jensen
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First St SW, Biobusiness Bldg 5-81, Rochester, MN 55905
| | | | | | - Eric A Cohen
- Department of Radiology, University of Pennsylvania, Philadelphia, Pa
- Department of Radiology, Columbia University, New York, NY
| | | | - Emily F Conant
- Department of Radiology, University of Pennsylvania, Philadelphia, Pa
| | - Karla M Kerlikowske
- Departments of Medicine and Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, Calif
| | - Despina Kontos
- Department of Radiology, University of Pennsylvania, Philadelphia, Pa
- Department of Radiology, Columbia University, New York, NY
- Departments of Biomedical Informatics and Biomedical Engineering, Columbia University, New York, NY
| | - Celine M Vachon
- Department of Quantitative Health Sciences, Mayo Clinic, 200 First St SW, Biobusiness Bldg 5-81, Rochester, MN 55905
| |
Collapse
|
22
|
Cha YJ, O'Connell CE, Calhoun BC, Felsheim BM, Fernandez-Martinez A, Fan C, Brueffer C, Larsson C, Borg Å, Saal LH, Perou CM. Genomic Characteristics Related to Histology-Based Immune Features in Breast Cancer. Mod Pathol 2025; 38:100736. [PMID: 39956271 PMCID: PMC12103273 DOI: 10.1016/j.modpat.2025.100736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/18/2025]
Abstract
The immune cell component of the tumor microenvironment is an important modulator of tumor progression. In patients with breast cancer, tumor-infiltrating lymphocytes (TILs) and tertiary lymphoid structures (TLS) represent core aspects of antitumor immunity, both increasingly recognized for clinical relevance. In this study, we evaluated immune-related histology features using whole-slide hematoxylin and eosin (H&E) images of The Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA) data set (n = 1035) and analyzed these distinct features relative to gene expression, PAM50 subtypes, and patient survival. H&E images were evaluated for TILs, plasma cells (PCs), high-endothelial venule-associated lymphoid aggregates (HALA), and mature TLS. For HALA and TLS, location relative to the tumor (nontumor, peritumor, and intratumor) was determined. HER2-enriched (HER2E) and basal-like breast tumors exhibited the highest mean TILs and the presence of PCs. HALA were present in 35.1% of cases and TLS in 6.5% of cases, also predominantly in HER2E and basal-like tumors. We derived gene expression signatures for 10 histologically defined immune features and tested their clinical significance using transcriptomic and survival data from the Sweden Cancerome Analysis Network - Breast (SCAN-B) cohort. Signatures related to TILs, PCs, HALA/TLS, TLS, and specifically intratumor HALA and TLS were associated with better survival in HER2E and basal-like tumors. Peritumor HALA/TLS and nontumor signatures were nonsignificant or associated with worse outcomes. Furthermore, we compared the immune microenvironment of high-TIL (TILs > 10%) tumors from TCGA-BRCA by PAM50 subtype through supervised analyses of 200+ immune gene expression signatures, and unique immune features were identified for each subtype. In high-TIL luminal tumors, enriched immune signatures had little relation to prognosis. High-TIL HER2E and basal-like tumors had distinct immune signatures linked to improved survival, related to B and T cells, respectively. Overall, PAM50 subtypes of breast cancer exhibit distinct immune microenvironments, both histologically and molecularly. These differences in immune properties should be considered when developing precise treatment strategies to achieve optimal therapeutic efficacy for patients.
Collapse
Affiliation(s)
- Yoon Jin Cha
- Department of Pathology, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea; Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Constandina E O'Connell
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Benjamin C Calhoun
- Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Brooke M Felsheim
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Bioinformatics and Computational Biology Curriculum, Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Aranzazu Fernandez-Martinez
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif, France; Inserm, Gustave Roussy Cancer Campus, UMR981, Villejuif, France
| | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Christian Brueffer
- Division of Oncology, Department of Clinical Sciences, Lund University, Sweden; Lund University Cancer Center, Lund University, Sweden; Skåne University Hospital Comprehensive Cancer Center, Lund, Sweden
| | - Christer Larsson
- Lund University Cancer Center, Lund University, Sweden; Skåne University Hospital Comprehensive Cancer Center, Lund, Sweden; Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Sweden
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences, Lund University, Sweden; Lund University Cancer Center, Lund University, Sweden; Skåne University Hospital Comprehensive Cancer Center, Lund, Sweden
| | - Lao H Saal
- Division of Oncology, Department of Clinical Sciences, Lund University, Sweden; Lund University Cancer Center, Lund University, Sweden; Skåne University Hospital Comprehensive Cancer Center, Lund, Sweden
| | - Charles M Perou
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pathology and Laboratory Medicine, University of North Carolina School of Medicine, Chapel Hill, North Carolina; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
23
|
Tseng TY, Hsieh CH, Liu JY, Huang HC, Juan HF. Single-cell and multi-omics integration reveals cholesterol biosynthesis as a synergistic target with HER2 in aggressive breast cancer. Comput Struct Biotechnol J 2025; 27:1719-1731. [PMID: 40391299 PMCID: PMC12088767 DOI: 10.1016/j.csbj.2025.04.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 04/17/2025] [Accepted: 04/23/2025] [Indexed: 05/21/2025] Open
Abstract
Breast cancer stands as one of the most prevalent malignancies affecting women. Alterations in molecular pathways in cancer cells represent key regulatory disruptions that drive malignancy, influencing cancer cell survival, proliferation, and potentially modulating therapeutic responsiveness. Therefore, decoding the intricate molecular mechanisms and identifying novel therapeutic targets through systematic computational approaches are essential steps toward advancing effective breast cancer treatments. In this study, we developed an integrative computational framework that combines single-cell RNA sequencing (scRNA-seq) and multi-omics analyses to delineate the functional characteristics of malignant cell subsets in breast cancer patients. Our analyses revealed a significant correlation between cholesterol biosynthesis and HER2 expression in malignant breast cancer cells, supported by proteomics data, gene expression profiles, drug treatment scores, and cell-surface HER2 intensity measurements. Given previous evidence linking cholesterol biosynthesis to HER2 membrane dynamics, we proposed a combinatorial strategy targeting both pathways. Experimental validation through clonogenic and viability assays demonstrated that simultaneous inhibition of cholesterol biosynthesis (via statins) and HER2 (via Neratinib) synergistically reduced malignant breast cancer cells, even in HER2-negative contexts. Through systematic analysis of scRNA-seq and multi-omics data, our study computationally identified and experimentally validated cholesterol biosynthesis and HER2 as novel combinatorial therapeutic targets in breast cancer. This data-driven approach highlights the potential of leveraging multiple molecular profiling techniques to uncover previously unexplored treatment strategies.
Collapse
Affiliation(s)
- Tzu-Yang Tseng
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Chiao-Hui Hsieh
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Jie-Yu Liu
- Department of Life Science, National Taiwan University, Taipei, Taiwan
| | - Hsuan-Cheng Huang
- Institute of Biomedical Informatics, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hsueh-Fen Juan
- Department of Life Science, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Biomedical Electronics and Bioinformatics, National Taiwan University, Taipei, Taiwan
- Center for Computational and Systems Biology, National Taiwan University, Taipei, Taiwan
- Center for Advanced Computing and Imaging in Biomedicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
24
|
Ben Rabah C, Sattar A, Ibrahim A, Serag A. A Multimodal Deep Learning Model for the Classification of Breast Cancer Subtypes. Diagnostics (Basel) 2025; 15:995. [PMID: 40310373 PMCID: PMC12025686 DOI: 10.3390/diagnostics15080995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/22/2025] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Breast cancer is a heterogeneous disease with distinct molecular subtypes, each requiring tailored therapeutic strategies. Accurate classification of these subtypes is crucial for optimizing treatment and improving patient outcomes. While immunohistochemistry remains the gold standard for subtyping, it is invasive and may not fully capture tumor heterogeneity. Artificial Intelligence (AI), particularly Deep Learning (DL), offers a promising non-invasive alternative by analyzing medical imaging data. Methods: In this study, we propose a multimodal DL model that integrates mammography images with clinical metadata to classify breast lesions into five categories: benign, luminal A, luminal B, HER2-enriched, and triple-negative. Using the publicly available Chinese Mammography Database (CMMD), our model was trained and evaluated on a dataset of 4056 images from 1775 patients. Results: The proposed multimodal approach significantly outperformed a unimodal model based solely on mammography images, achieving an AUC of 88.87% for multiclass classification of these five categories, compared to 61.3% AUC for the unimodal model. Conclusions: These findings highlight the potential of multimodal AI-driven approaches for non-invasive breast cancer subtype classification, paving the way for improved diagnostic precision and personalized treatment strategies.
Collapse
Affiliation(s)
| | - Aamenah Sattar
- Department of Medicine, New Vision University, 0159 Tbilisi, Georgia
| | - Ahmed Ibrahim
- AI Innovation Lab, Weill Cornell Medicine, Doha 24144, Qatar
| | - Ahmed Serag
- AI Innovation Lab, Weill Cornell Medicine, Doha 24144, Qatar
| |
Collapse
|
25
|
Binder A, Tendl-Schulz K, Marhold M, Wimmer K, Rudas M, Bartsch R, Bago-Horvath Z, Gruber ES, Exner R. Risk Assessment Using Gene Expression Profiling Correlates with Clinical Prognosis Estimation in Hormone Receptor-Positive/HER2-Negative Early Breast Cancer. Breast Care (Basel) 2025:1-8. [PMID: 40406379 PMCID: PMC12094704 DOI: 10.1159/000545785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 04/03/2025] [Indexed: 05/26/2025] Open
Abstract
Background Gene expression profiles (GEPs) are recommended for tailoring adjuvant treatment in patients with hormone receptor (HR)-positive/HER2-negative breast cancer (BC) with intermediate clinical and pathological risk. This single-center retrospective study aimed at evaluating the clinical relevance of the additive information provided by GEPs in clinical routine at a tertiary care center. Methods From 03/2010 to 07/2019, GEPs by either MammaPrint (MP) or PAM50 of HR-positive/HER2-negative early-stage BC were retrospectively included in the study. Pseudonymized data were processed for statistical analysis. Correlations between clinical and molecular risk markers were calculated. Survival was estimated using the Kaplan-Meier method. Results Clinical and molecular risk data were available for 213 patients; complete follow-up data were available for 189 patients. According to GEPs by either MP (n = 69) or PAM50 (n = 144), 67 patients (31.5%) had low, 58 (27.2%) intermediate only in PAM50, and 88 (41.3%) high-risk BC. The MP group showed a higher rate of molecular low-risk tumors, while tumors analyzed by PAM50 were more frequent in a molecular high-risk situation. A significant correlation of proliferation rate and grading with the molecular risk score was observed (p < 0.001 each). Adjuvant chemotherapy was recommended in 87.5% of molecular high-risk tumors but administered in 64.8% only. Interestingly, a worse DFS was detected in the molecular low-risk group compared to the high-risk group (p = 0.55). It may be assumed that this is associated with an advanced tumor stage in these patients. Conclusion In HR-positive/HER2-negative BC, proliferation rate as well as tumor grade correlated significantly with risk assessment by GEPs. Despite a high-risk result, chemotherapy is often omitted due to patient-specific factors such as age, comorbidities, or patients' preference. On the other hand, survival with genomic low-risk tumors is likely to be compromised to more advanced stage, questioning the clinical validity of GEPs in these cases.
Collapse
Affiliation(s)
- Alexa Binder
- Department of General Surgery, Medical University Vienna and Comprehensive Cancer Center Vienna, Vienna, Austria
| | - Kristina Tendl-Schulz
- Clinical Institute of Pathology, Medical University Vienna and Comprehensive Cancer Center Vienna, Vienna, Austria
| | - Maximilian Marhold
- Division of Oncology, Department of Medicine I, Medical University Vienna and Comprehensive Cancer Center Vienna, Vienna, Austria
| | - Kerstin Wimmer
- Department of General Surgery, Medical University Vienna and Comprehensive Cancer Center Vienna, Vienna, Austria
| | - Margaretha Rudas
- Clinical Institute of Pathology, Medical University Vienna and Comprehensive Cancer Center Vienna, Vienna, Austria
| | - Rupert Bartsch
- Division of Oncology, Department of Medicine I, Medical University Vienna and Comprehensive Cancer Center Vienna, Vienna, Austria
| | - Zsuzsanna Bago-Horvath
- Clinical Institute of Pathology, Medical University Vienna and Comprehensive Cancer Center Vienna, Vienna, Austria
| | - Elisabeth S. Gruber
- Department of General Surgery, Medical University Vienna and Comprehensive Cancer Center Vienna, Vienna, Austria
| | - Ruth Exner
- Department of General Surgery, Medical University Vienna and Comprehensive Cancer Center Vienna, Vienna, Austria
| |
Collapse
|
26
|
Xiang L, Yang J, Rao J, Ma A, Liu C, Zhang Y, Huang A, Xie T, Xue H, Chen Z, Yuan J, Yan H. Integrating Machine Learning and Bulk and Single-Cell RNA Sequencing to Decipher Diverse Cell Death Patterns for Predicting the Prognosis of Neoadjuvant Chemotherapy in Breast Cancer. Int J Mol Sci 2025; 26:3682. [PMID: 40332226 PMCID: PMC12027272 DOI: 10.3390/ijms26083682] [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: 02/24/2025] [Revised: 04/01/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Breast cancer (BRCA) continues to pose a serious risk to women's health worldwide. Neoadjuvant chemotherapy (NAC) is a critical treatment strategy. Nevertheless, the heterogeneity in treatment outcomes necessitates the identification of reliable biomarkers and prognostic models. Programmed cell death (PCD) pathways serve as a critical factor in tumor development and treatment response. However, the relationship between the diverse patterns of PCD and NAC in BRCA remains unclear. We integrated machine learning and multiple bioinformatics tools to explore the association between 19 PCD patterns and the prognosis of NAC within a cohort of 921 BRCA patients treated with NAC from seven multicenter cohorts. A prognostic risk model based on PCD-related genes (PRGs) was constructed and evaluated using a combination of 117 machine learning algorithms. Immune infiltration analysis, mutation analysis, pharmacological analysis, and single-cell RNA sequencing (scRNA-seq) were conducted to explore the genomic profile and clinical significance of these model genes in BRCA. Immunohistochemistry (IHC) was employed to validate the expression of select model genes (UGCG, BTG22, TNFRSF21, and MYB) in BRCA tissues. We constructed a PRGs prognostic risk model by using a signature comprising 20 PCD-related DEGs to forecast the clinical outcomes of NAC in BRCA patients. The prognostic model demonstrated excellent predictive accuracy, with a high concordance index (C-index) of 0.772, and was validated across multiple independent datasets. Our results demonstrated a strong association between the developed model and the survival prognosis, clinical pathological features, immune infiltration, tumor microenvironment (TME), gene mutations, and drug sensitivity of NAC for BRCA patients. Moreover, IHC studies further demonstrated that the expression of certain model genes in BRCA tissues was significantly associated with the efficacy of NAC and emerged as an autonomous predictor of outcomes influencing the outcome of patients. We are the first to integrate machine learning and bulk and scRNA-seq to decode various cell death mechanisms for the prognosis of NAC in BRCA. The developed unique prognostic model, based on PRGs, provides a novel and comprehensive strategy for predicting the NAC outcomes of BRCA patients. This model not only aids in understanding the mechanisms underlying NAC efficacy but also offers insights into personalized treatment strategies, potentially improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Honglin Yan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China; (L.X.); (J.Y.); (J.R.); (A.M.); (C.L.); (Y.Z.); (A.H.); (T.X.); (H.X.); (Z.C.); (J.Y.)
| |
Collapse
|
27
|
Pérez Malla CU, Kalla J, Tiefenbacher A, Wasinger G, Kluge K, Egger G, Sheibani-Tezerji R. Goistrat: gene-of-interest-based sample stratification for the evaluation of functional differences. BMC Bioinformatics 2025; 26:97. [PMID: 40188042 PMCID: PMC11971790 DOI: 10.1186/s12859-025-06109-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 03/10/2025] [Indexed: 04/07/2025] Open
Abstract
PURPOSE Understanding the impact of gene expression in pathological processes, such as carcinogenesis, is crucial for understanding the biology of cancer and advancing personalised medicine. Yet, current methods lack biologically-informed-omics approaches to stratify cancer patients effectively, limiting our ability to dissect the underlying molecular mechanisms. RESULTS To address this gap, we present a novel workflow for the stratification and further analysis of multi-omics samples with matched RNA-Seq data that relies on MSigDB curated gene sets, graph machine learning and ensemble clustering. We compared the performance of our workflow in the top 8 TCGA datasets and showed its clear superiority in separating samples for the study of biological differences. We also applied our workflow to analyse nearly a thousand prostate cancer samples, focusing on the varying expression of the FOLH1 gene, and identified specific pathways such as the PI3K-AKT-mTOR gene sets as well as signatures linked to prostate tumour aggressiveness. CONCLUSION Our comprehensive approach provides a novel tool to identify disease-relevant functions of genes of interest (GOI) in large datasets. This integrated approach offers a valuable framework for understanding the role of the expression variation of a GOI in complex diseases and for informing on targeted therapeutic strategies.
Collapse
Affiliation(s)
- Carlos Uziel Pérez Malla
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Ludwig Boltzmann Gesellschaft, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Jessica Kalla
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Andreas Tiefenbacher
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Gabriel Wasinger
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Kilian Kluge
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Department of Biomedical Imaging and Image-Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Christian Doppler Laboratory for Applied Metabolomics, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Ludwig Boltzmann Gesellschaft, Währinger Gürtel 18-20, Vienna, 1090, Austria
- Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria
| | - Raheleh Sheibani-Tezerji
- Department of Pathology, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria.
- Ludwig Boltzmann Institute Applied Diagnostics, Ludwig Boltzmann Gesellschaft, Währinger Gürtel 18-20, Vienna, 1090, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Währinger Gürtel 18-20, Vienna, 1090, Austria.
| |
Collapse
|
28
|
Vidal M, Falato C, Pascual T, Sanchez-Bayona R, Muñoz-Mateu M, Cebrecos I, Gonzalez-Farré X, Cortadellas T, Margelí Vila M, Luna MA, Siso C, Amillano K, Galván P, Bergamino MA, Ferrero-Cafiero JM, Salvador F, Espinosa Guerrero A, Pare L, Sanfeliu E, Prat A, Bellet M. Elacestrant in Women with Estrogen Receptor-Positive and HER2-Negative Early Breast Cancer: Results from the Preoperative Window-of-Opportunity ELIPSE Trial. Clin Cancer Res 2025; 31:1223-1232. [PMID: 39820652 PMCID: PMC11959270 DOI: 10.1158/1078-0432.ccr-24-2460] [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: 07/30/2024] [Revised: 10/04/2024] [Accepted: 01/14/2025] [Indexed: 01/19/2025]
Abstract
PURPOSE Elacestrant has shown significantly prolonged progression-free survival compared with standard-of-care endocrine therapy in estrogen receptor-positive (ER-positive), HER2-negative metastatic breast cancer, whereas potential benefit in early-stage disease requires further exploration. The SOLTI-ELIPSE window-of-opportunity trial investigated the biological changes induced by a short course of preoperative elacestrant in postmenopausal women with early breast cancer. PATIENTS AND METHODS Eligible patients with untreated T1c (≥1.5 cm)-T3, N0, ER-positive/HER2-negative breast cancer with locally assessed Ki67 ≥10% received elacestrant at a daily dose of 345 mg for 4 weeks. The primary efficacy endpoint was complete cell cycle arrest, defined as Ki67 ≤2.7%, on day 28. RESULTS Overall, 22 patients were evaluable for the primary endpoint. Elacestrant was associated with a complete cell cycle arrest rate of 27.3% and a statistically significant Ki67 geometric mean change of -52.9% (P = 0.007; 95% confidence interval, -67.4 to -32.1). Notably, the treatment with elacestrant led to a shift toward a more endocrine-sensitive and less proliferative tumor phenotype based on PAM50-based gene signatures. Elacestrant increased the expression of immune-response genes (GZMB, CD4, and CD8A) and suppressed proliferation and estrogen-signaling genes (MKI67, ESR1, and AR). These biological changes were independent of the levels of Ki67 suppression on day 28. The most common adverse events were grade 1 anemia (21.7%), hot flushes (8.7%), constipation (8.7%), and abdominal pain (8.7%). One patient experienced a grade 3 cutaneous rash, leading to treatment discontinuation. No other serious adverse events were reported. CONCLUSIONS Preoperative treatment with elacestrant in early breast cancer demonstrated relevant biological and molecular responses and exhibited a manageable safety profile. These findings support further investigation of elacestrant in the early setting.
Collapse
Affiliation(s)
- Maria Vidal
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Claudette Falato
- SOLTI Cancer Research Group, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Tomás Pascual
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Rodrigo Sanchez-Bayona
- SOLTI Cancer Research Group, Barcelona, Spain
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Montserrat Muñoz-Mateu
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Isaac Cebrecos
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
| | | | - Tomás Cortadellas
- Breast Unit, Department of Obstetrics and Gynaecology, Hospital Universitari General de Catalunya, Barcelona, Spain
| | - Mireia Margelí Vila
- SOLTI Cancer Research Group, Barcelona, Spain
- B-ARGO Group, Medical Oncology Department, ICO Badalona, Germans Trias I Pujol Institute, Badalona, Spain
- Medicine Department, Autonomous University, Barcelona, Spain
| | - Miguel A. Luna
- B-ARGO Group, Medical Oncology Department, ICO Badalona, Germans Trias I Pujol Institute, Badalona, Spain
| | | | - Kepa Amillano
- Hospital Universitari Sant Joan de Reus, Barcelona, Spain
| | - Patricia Galván
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
| | - Milana A. Bergamino
- SOLTI Cancer Research Group, Barcelona, Spain
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- B-ARGO Group, Medical Oncology Department, ICO Badalona, Germans Trias I Pujol Institute, Badalona, Spain
| | | | | | | | - Laia Pare
- SOLTI Cancer Research Group, Barcelona, Spain
| | - Esther Sanfeliu
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Department of Pathology, Hospital Clinic de Barcelona, Barcelona, Spain
| | - Aleix Prat
- Cancer Institute and Blood Disorders, Hospital Clinic de Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumor, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Medicine Department, University of Barcelona, Barcelona, Spain
| | - Meritxell Bellet
- SOLTI Cancer Research Group, Barcelona, Spain
- Medicine Department, Autonomous University, Barcelona, Spain
- Vall d’Hebron University Hospital, Barcelona, Spain
- Vall d’Hebron Institute of Oncology (VHIO), Barcelona, Spain
| |
Collapse
|
29
|
Zhu X, Hu M, Huang X, Li L, Lin X, Shao X, Li J, Du X, Zhang X, Sun R, Tong T, Ma Y, Ning L, Jiang Y, Zhang Y, Shao Y, Wang Z, Zhou Y, Ding J, Zhao Y, Xuan B, Zhang H, Zhang Y, Hong J, Fang JY, Xiao X, Shen B, He S, Chen H. Interplay between gut microbial communities and metabolites modulates pan-cancer immunotherapy responses. Cell Metab 2025; 37:806-823.e6. [PMID: 39909032 DOI: 10.1016/j.cmet.2024.12.013] [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: 04/14/2024] [Revised: 10/20/2024] [Accepted: 12/21/2024] [Indexed: 02/07/2025]
Abstract
Immune checkpoint blockade (ICB) therapy has revolutionized cancer treatment but remains effective in only a subset of patients. Emerging evidence suggests that the gut microbiome and its metabolites critically influence ICB efficacy. In this study, we performed a multi-omics analysis of fecal microbiomes and metabolomes from 165 patients undergoing anti-programmed cell death protein 1 (PD-1)/programmed death ligand 1 (PD-L1) therapy, identifying microbial and metabolic entities associated with treatment response. Integration of data from four public metagenomic datasets (n = 568) uncovered cross-cohort microbial and metabolic signatures, validated in an independent cohort (n = 138). An integrated predictive model incorporating these features demonstrated robust performance. Notably, we characterized five response-associated enterotypes, each linked to specific bacterial taxa and metabolites. Among these, the metabolite phenylacetylglutamine (PAGln) was negatively correlated with response and shown to attenuate anti-PD-1 efficacy in vivo. This study sheds light on the interplay among the gut microbiome, the gut metabolome, and immunotherapy response, identifying potential biomarkers to improve treatment outcomes.
Collapse
Affiliation(s)
- Xiaoqiang Zhu
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Department of Gastroenterology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Muni Hu
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaowen Huang
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lingxi Li
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaolin Lin
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoyan Shao
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Jiantao Li
- Shanghai Lung Cancer Center, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoyue Du
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China
| | - Xinjia Zhang
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
| | - Rongrong Sun
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Tianying Tong
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yanru Ma
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lijun Ning
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Jiang
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Zhang
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yuqi Shao
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhenyu Wang
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yilu Zhou
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jinmei Ding
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ying Zhao
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Baoqin Xuan
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Hongyang Zhang
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, China
| | - Youwei Zhang
- Department of Medical Oncology, Xuzhou Central Hospital, Clinical School of Xuzhou Medical University, Xuzhou, China
| | - Jie Hong
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-Yuan Fang
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xiuying Xiao
- Department of Oncology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Bo Shen
- Department of Oncology, The Affiliated Cancer Hospital of Nanjing Medical University, Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, Nanjing, China.
| | - Songbing He
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| | - Haoyan Chen
- State Key Laboratory of Systems Medicine for Cancer, Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Shanghai Institute of Digestive Disease, NHC Key Laboratory of Digestive Diseases, Renji Hospital, Shanghai Cancer Institute, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
30
|
Jayachandran P, Deshmukh SK, Wu S, Ribeiro JR, Kang I, Xiu J, Farrell A, Battaglin F, Spicer DV, Soni S, Zhang W, Ashouri K, Millstein J, Ma CX, Graff SL, Radovich M, Sledge GW, Lenz HJ, Roussos Torres ET. Association of Androgen Receptor Expression With Tumor Immune Landscape and Treatment Outcomes of Patients With Breast Cancer. JCO Precis Oncol 2025; 9:e2400459. [PMID: 40294352 DOI: 10.1200/po-24-00459] [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: 12/07/2024] [Revised: 02/11/2025] [Accepted: 03/04/2025] [Indexed: 04/30/2025] Open
Abstract
PURPOSE Although estrogen receptor is well studied in breast cancer (BC), the role of androgen receptor (AR) in prognosis and therapy response is less understood. Here, we characterized the clinicopathologic and molecular features of AR gene expression in BC subtypes. METHODS Ten thousand seven hundred twenty-eight BC samples were tested by next-generation DNA sequencing, whole-transcriptome sequencing, and immunohistochemistry at Caris Life Sciences (Phoenix, AZ). Tumors with AR-high and AR-low RNA expression were stratified by top and bottom quartiles, respectively. Treatment-associated survival was obtained from insurance claims and calculated from treatment start to last contact using Kaplan-Meier estimates. Statistical significance was determined by chi-square and Mann-Whitney U test with P values adjusted for multiple comparisons (q < .05). RESULTS AR-low was associated with basal-like tumors. AR-high tumors were associated with increased mutation rates in several genes-namely PIK3CA and CDH1-across all subtypes, while other associations such as RB1 and MAP3K1 were subtype-dependent. The immune landscape was differentially affected by AR expression in each subtype, but these differences did not correspond to differential responses to immune checkpoint blockade. Patients with AR-high tumors had a longer therapy response for most subtypes, but those with AR-high tumors that were human epidermal growth factor receptor 2-enriched and luminal B trended toward worse chemotherapy or hormone therapy response, respectively. CONCLUSION Our data suggest a unique molecular profile of AR-high BC that is subtype-specific and generally associated with improved outcomes. Exploration of specific mutations and immune-oncology markers associated with AR-high may aid in molecularly selected clinical trial design for patients with advanced BC.
Collapse
Affiliation(s)
- Priya Jayachandran
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | | | | | | | - Irene Kang
- Department of Medical Oncology & Therapeutics Research, City of Hope Orange County, Lennar Foundation Cancer Center, Irvine, CA
| | | | | | - Francesca Battaglin
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Darcy V Spicer
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Shivani Soni
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Wu Zhang
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Karam Ashouri
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Joshua Millstein
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Cynthia X Ma
- Division of Oncology, Washington University, St Louis, MO
| | - Stephanie L Graff
- Legorreta Cancer Center, Brown University, Providence, RI
- Lifespan Cancer Institute, Providence, RI
| | | | | | - Heinz-Josef Lenz
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Evanthia T Roussos Torres
- Division of Medical Oncology, Keck School of Medicine, University of Southern California, Los Angeles, CA
| |
Collapse
|
31
|
Llinas-Bertran A, Butjosa-Espín M, Barberi V, Seoane JA. Multimodal data integration in early-stage breast cancer. Breast 2025; 80:103892. [PMID: 39922065 PMCID: PMC11973824 DOI: 10.1016/j.breast.2025.103892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/13/2024] [Accepted: 01/27/2025] [Indexed: 02/10/2025] Open
Abstract
The use of biomarkers in breast cancer has significantly improved patient outcomes through targeted therapies, such as hormone therapy anti-Her2 therapy and CDK4/6 or PARP inhibitors. However, existing knowledge does not fully encompass the diverse nature of breast cancer, particularly in triple-negative tumors. The integration of multi-omics and multimodal data has the potential to provide new insights into biological processes, to improve breast cancer patient stratification, enhance prognosis and response prediction, and identify new biomarkers. This review presents a comprehensive overview of the state-of-the-art multimodal (including molecular and image) data integration algorithms developed and with applicability to breast cancer stratification, prognosis, or biomarker identification. We examined the primary challenges and opportunities of these multimodal data integration algorithms, including their advantages, limitations, and critical considerations for future research. We aimed to describe models that are not only academically and preclinically relevant, but also applicable to clinical settings.
Collapse
Affiliation(s)
- Arnau Llinas-Bertran
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Maria Butjosa-Espín
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Vittoria Barberi
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jose A Seoane
- Cancer Computational Biology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
32
|
Nguyen Van Long F, Poirier B, Desbiens C, Perron M, Paquet C, Ouellet C, Diorio C, Lemieux J, Nabi H. First versus second-generation molecular profiling tests: How both can guide decision-making in early-stage hormone-receptor positive breast cancers? Cancer Treat Rev 2025; 135:102909. [PMID: 40054315 DOI: 10.1016/j.ctrv.2025.102909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 04/08/2025]
Abstract
Hormone receptor-positive (HR+) and human epidermal growth factor receptor 2-negative (HER2-) tumors represent the most common types of early-stage breast cancer. However, their response to adjuvant systemic treatments varies widely due to tumor heterogeneity. Current decisions for adjuvant treatment rely heavily on clinical and pathological characteristics, which can sometimes lead to overtreatment. Accurately identifying patients who will benefit from adjuvant chemotherapy at an individual level remains a challenge. Multigene profiling assays are now widely used in clinics to better assess recurrence risk and chemotherapy response for HR+ disease. In this report, we examine the advantages and limitations of two widely used molecular profiling tests-Oncotype DX and Prosigna. Both Oncotype DX and Prosigna have been demonstrated to be effective prognostic tools in early breast cancer, with Oncotype DX also being validated as a predictive tool to guide chemotherapy decisions. We focus on studies that directly compare these molecular tests and discuss how their strengths can be leveraged to improve clinical decision-making for early-stage HR+ breast cancers. Finally, we highlight remaining knowledge gaps and propose directions for future research.
Collapse
Affiliation(s)
- Flora Nguyen Van Long
- Oncology Axis, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc-UL), Quebec city, QC G1V 4G2, Canada
| | - Brigitte Poirier
- Oncology Axis, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc-UL), Quebec city, QC G1V 4G2, Canada; Centre des maladies du sein, CHU de Québec-Université Laval, Quebec city, QC G1V 4G2, Canada
| | - Christine Desbiens
- Oncology Axis, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc-UL), Quebec city, QC G1V 4G2, Canada; Centre des maladies du sein, CHU de Québec-Université Laval, Quebec city, QC G1V 4G2, Canada
| | - Marjorie Perron
- Pathology department, CHU de Québec-Université Laval, Quebec city, QC G1V 4G2, Canada
| | - Claudie Paquet
- Pathology department, CHU de Québec-Université Laval, Quebec city, QC G1V 4G2, Canada
| | - Cathie Ouellet
- Pathology department, CHU de Québec-Université Laval, Quebec city, QC G1V 4G2, Canada
| | - Caroline Diorio
- Oncology Axis, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc-UL), Quebec city, QC G1V 4G2, Canada; Department of Social and Preventive Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada
| | - Julie Lemieux
- Oncology Axis, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc-UL), Quebec city, QC G1V 4G2, Canada; Centre des maladies du sein, CHU de Québec-Université Laval, Quebec city, QC G1V 4G2, Canada
| | - Hermann Nabi
- Oncology Axis, Centre Hospitalier Universitaire de Québec Research Center - Université Laval (CRCHUQc-UL), Quebec city, QC G1V 4G2, Canada; Department of Social and Preventive Medicine, Faculty of Medicine, Université Laval, Quebec City, QC G1V 0A6, Canada.
| |
Collapse
|
33
|
Muñoz DP, Arcuschin CD, Kahrizi K, Sayaman RW, DiBenedetto C, Salaberry PJ, Shen Y, Zakroui O, Schwarzer C, Scapozza A, Betancur P, Saba JD, Coppé JP, Barcellos-Hoff MH, Kappes D, Veer LV', Schor IE. Super-enhancer profiling reveals ThPOK/ZBTB7B, a CD4+ cell lineage commitment factor, as a master regulator that restricts breast cancer cells to a luminal non-migratory phenotype. RESEARCH SQUARE 2025:rs.3.rs-6240646. [PMID: 40235471 PMCID: PMC11998796 DOI: 10.21203/rs.3.rs-6240646/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Despite efforts to understand breast cancer biology, metastatic disease remains a clinical challenge. Identifying suppressors of breast cancer progression and mechanisms of transition to more invasive phenotypes could provide game changing therapeutic opportunities. Transcriptional deregulation is central to all malignancies, highlighted by the extensive reprogramming of regulatory elements that underlie oncogenic programs. Among these, super-enhancers (SEs) stand out due to their enrichment in genes controlling cancer hallmarks. To reveal novel breast cancer dependencies, we integrated the analysis of the SE landscape with master regulator activity inference for a series of breast cancer cell lines. As a result, we identified T-helper-inducing Poxviruses and Zinc-finger (POZ)/Krüppel-like factor (ThPOK, ZBTB7B), a CD4+ cell lineage commitment factor, as a breast cancer master regulator that is recurrently associated with a SE. ThPOK expression is highest in luminal breast cancer but is significantly reduced in the basal subtype. Manipulation of ThPOK levels in cell lines shows that its repressive function restricts breast cancer cells to an epithelial phenotype by suppressing the expression of genes involved in the epithelial-mesenchymal transition (EMT), WNT/b-catenin target genes, and the pro-metastatic TGFb pathway. Our study reveals ThPOK as a master transcription factor that restricts the acquisition of metastatic features in breast cancer cells.
Collapse
|
34
|
Iacovacci J, Brough R, Moughari FA, Alexander J, Kemp H, Tutt ANJ, Natrajan R, Lord CJ, Haider S. Proteogenomic discovery of RB1-defective phenocopy in cancer predicts disease outcome, response to treatment, and therapeutic targets. SCIENCE ADVANCES 2025; 11:eadq9495. [PMID: 40138429 PMCID: PMC11939072 DOI: 10.1126/sciadv.adq9495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 02/11/2025] [Indexed: 03/29/2025]
Abstract
Genomic defects caused by truncating mutations or deletions in the Retinoblastoma tumor suppressor gene (RB1) are frequently observed in many cancer types leading to dysregulation of the RB pathway. Here, we propose an integrative proteogenomic approach that predicts cancers with dysregulation in the RB pathway. A subset of these cancers, which we term as "RBness," lack RB1 genomic defects and yet phenocopy the transcriptional profile of RB1-defective cancers. We report RBness as a pan-cancer phenomenon, associated with patient outcome and chemotherapy response in multiple cancer types, and predictive of CDK4/6 inhibitor response in estrogen-positive breast cancer. Using RNA interference and a CRISPR-Cas9 screen in isogenic models, we find that RBness cancers also phenocopy synthetic lethal vulnerabilities of cells with RB1 genomic defects. In summary, our findings suggest that dysregulation of the RB pathway in cancers lacking RB1 genomic defects provides a molecular rationale for how these cancers could be treated.
Collapse
Affiliation(s)
- Jacopo Iacovacci
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- Data Science Unit, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milano 20133, Italy
| | - Rachel Brough
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Fatemeh Ahmadi Moughari
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - John Alexander
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Harriet Kemp
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Andrew N. J. Tutt
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Rachael Natrajan
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| | - Christopher J. Lord
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
- CRUK Gene Function Laboratory, The Institute of Cancer Research, London SW3 6JB, UK
| | - Syed Haider
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
35
|
Aine M, Nacer DF, Arbajian E, Veerla S, Karlsson A, Häkkinen J, Johansson HJ, Rosengren F, Vallon-Christersson J, Borg Å, Staaf J. The DNA methylation landscape of primary triple-negative breast cancer. Nat Commun 2025; 16:3041. [PMID: 40155623 PMCID: PMC11953470 DOI: 10.1038/s41467-025-58158-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a clinically challenging and molecularly heterogenous breast cancer subgroup. Here, we investigate the DNA methylation landscape of TNBC. By analyzing tumor methylome profiles and accounting for the genomic context of CpG methylation, we divide TNBC into two epigenetic subtypes corresponding to a Basal and a non-Basal group, in which characteristic transcriptional patterns are correlated with DNA methylation of distal regulatory elements and epigenetic regulation of key steroid response genes and developmental transcription factors. Further subdivision of the Basal and non-Basal subtypes identifies subgroups transcending genetic and proposed TNBC mRNA subtypes, demonstrating widely differing immunological microenvironments, putative epigenetically-mediated immune evasion strategies, and a specific metabolic gene network in older patients that may be epigenetically regulated. Our study attempts to target the epigenetic backbone of TNBC, an approach that may inform future studies regarding tumor origins and the role of the microenvironment in shaping the cancer epigenome.
Collapse
Affiliation(s)
- Mattias Aine
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Deborah F Nacer
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Elsa Arbajian
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Srinivas Veerla
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Anna Karlsson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Jari Häkkinen
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Henrik J Johansson
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - Frida Rosengren
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Johan Vallon-Christersson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden.
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE 22381, Lund, Sweden.
| |
Collapse
|
36
|
Lin Z, Wang X, Hua G, Zhong F, Cheng W, Qiu Y, Chi Z, Zeng H, Wang X. Identification of mitochondrial permeability transition-related lncRNAs as quantitative biomarkers for the prognosis and therapy of breast cancer. Front Genet 2025; 16:1510154. [PMID: 40206506 PMCID: PMC11979797 DOI: 10.3389/fgene.2025.1510154] [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: 11/07/2024] [Accepted: 03/05/2025] [Indexed: 04/11/2025] Open
Abstract
Breast cancer (BC) continues to pose a global health threat and presents challenges for treatment due to its high heterogeneity. Recent advancements in the understanding of mitochondrial permeability transition (MPT) and the regulatory roles of long non-coding RNAs (lncRNAs) offer potential insights for the stratification and personalized treatment of BC. Although the association between MPT and lncRNAs has not been widely studied, a few research studies have indicated a regulatory impact of lncRNAs on MPT, further deepening the understanding of the tumor. To identify reliable biomarkers associated with MPT for managing BC, bulk RNA-seq data of MPT-related lncRNAs acquired from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) project were utilized to assess BC patients. A scoring system, termed the MPT-related score (MPTRscore), was developed using LASSO-Cox regression on data from 1,029 BC patients from TCGA-BRCA. Meanwhile, the superior prognostic accuracy of the MPTRscore was demonstrated by comparing it with biomarkers, including PAM50 subtyping for standardization. Subsequently, a clinical prediction model was created by incorporating the MPTRscore and clinical variables. This analysis revealed two distinct MPTRscore groups characterized by different biomolecular processes, tumor microenvironment (TME) patterns, and clinical outcomes. The MPTRscore was further investigated through unsupervised consensus clustering of TCGA-BRCA based on MPTRscore-related prognostic genes. Additionally, the MPTRscore was identified as an independent prognostic factor for BC and showed guiding utility in immunotherapy and chemotherapy response. Specifically, patients with a low MPTRscore exhibited better prognosis and treatment responses compared to those with a high MPTRscore. Significantly, the relevance of clustering results and MPTRscore was found to be mediated by lncRNA transcript RP11-573D15.8-018. In conclusion, MPTRscore-related clusters were identified in BC, and an integrative score was developed as a biomarker for predicting BC prognosis and therapeutic response. Additionally, molecular interactions underlying the relationship between MPTRscore-related clusters and MPTRscore were uncovered, proving insights for BC stratification. These findings may aid in prognosis determination and therapeutic decision-making for BC patients.
Collapse
Affiliation(s)
- Zhongshu Lin
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- School of Biological and Behavioural Science, Queen Mary University of London, London, United Kingdom
- Queen Mary College, Nanchang University, Nanchang, China
| | - Xinlu Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
- School of Public Health, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Guanxiang Hua
- School of Biological and Behavioural Science, Queen Mary University of London, London, United Kingdom
- Queen Mary College, Nanchang University, Nanchang, China
| | - Fangmin Zhong
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Wangxinjun Cheng
- School of Biological and Behavioural Science, Queen Mary University of London, London, United Kingdom
- Queen Mary College, Nanchang University, Nanchang, China
| | - Yuxiang Qiu
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Zhe Chi
- School of Biological and Behavioural Science, Queen Mary University of London, London, United Kingdom
- Queen Mary College, Nanchang University, Nanchang, China
| | - Huan Zeng
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
37
|
Guo J, Zhang Z. Integrated multi-omics unveils novel immune signature for predicting prognosis in colon cancer patients. Sci Rep 2025; 15:9788. [PMID: 40118975 PMCID: PMC11928561 DOI: 10.1038/s41598-025-85390-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 01/02/2025] [Indexed: 03/24/2025] Open
Abstract
Colon cancer, a prevalent malignancy, is subject to intricate immune modulation, which substantially affects both treatment efficacy and prognostic outcomes. Furthermore, colon cancer is highly heterogeneous, and our comprehensive understanding of its immune microenvironment has not yet been fully realized. There is still ample opportunity for in-depth investigation into the composition and interactions of immune cells within colon cancer, as well as their implications for disease prognosis. In this study, we employed single-cell data from colon cancer to distinguish immune cells from non-immune cells through cluster analysis. Furthermore, we conducted an in-depth analysis of myeloid and T cells, which were categorized into 20 distinct cell subpopulations. Functional enrichment analysis revealed T cells' active involvement in the Fatty Acid Metabolism and Adipogenesis pathways, while immune checkpoint-associated genes (ICGs) were notably upregulated in CD8+ T cells. Subsequent analysis involved calculating gene scores to characterize cell subpopulations, which, when combined with patient survival time analysis, revealed a significant association between the gene characterization score (referred to as "imm-score") and the survival of colon cancer patients. Specifically, the presence of CD8+-ANXA1hi-T cells was linked to shortened overall survival in the high imm-score subgroup. Subsequently, combined with genomic analysis, patients in the high imm-score subgroup exhibited elevated tumor mutation burden (TMB) and heightened activity in both the epithelial-mesenchymal transition (EMT) and Notch signaling pathway. Finally, according to our new algorithm, scores calculated predicted the effectiveness of immunotherapy for patients. The results revealed that patients with lower scores could achieve better therapeutic outcomes with immunotherapy. This study offers an extensive analysis of the interplay between T cells and myeloid cells within colon cancer tissues, exploring their impact on the survival and prognosis of colon cancer patients. Additionally, it unveils the potential significance of the imm-score in colon cancer, potentially indicating a poor prognosis and providing novel insights into the immune-regulatory mechanisms underlying the disease.
Collapse
Affiliation(s)
- Jing Guo
- Department of Gastrointestinal and Anorectal Surgery, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
| | - Zili Zhang
- Department of Gastrointestinal and Anorectal Surgery, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China.
| |
Collapse
|
38
|
Loughrey CF, Maguire S, Dłotko P, Bai L, Orr N, Jurek-Loughrey A. A novel method for subgroup discovery in precision medicine based on topological data analysis. BMC Med Inform Decis Mak 2025; 25:139. [PMID: 40102808 PMCID: PMC11921513 DOI: 10.1186/s12911-025-02852-9] [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: 08/06/2024] [Accepted: 01/03/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND The Mapper algorithm is a data mining topological tool that can help us to obtain higher level understanding of disease by visualising the structure of patient data as a similarity graph. It has been successfully applied for exploratory analysis of cancer data in the past, delivering several significant subgroup discoveries. Using the Mapper algorithm in practice requires setting up multiple parameters. The graph then needs to be manually analysed according to a research question at hand. It has been highlighted in the literature that Mapper's parameters have significant impact on the output graph shape and there is no established way to select their optimal values. Hence while using the Mapper algorithm, different parameter values and consequently different output graphs need to be studied. This prevents routine application of the Mapper algorithm in real world settings. METHODS We propose a new algorithm for subgroup discovery within the Mapper graph. We refer to the task as hotspot detection as it is designed to identify homogenous and geometrically compact subsets of patients, which are distinct with respect to their clinical or molecular profiles (e.g. survival). Furthermore, we propose to include the existence of a hotspot as a criterion while searching the parameter space, addressing one of the key limitations of the Mapper algorithm (i.e. parameter selection). RESULTS Two experiments were performed to demonstrate the efficacy of the algorithm, including an artificial hotspot in the Two Circles dataset and a real world case study of subgroup discovery in oestrogen receptor-positive breast cancer. Our hotspot detection algorithm successfully identified graphs containing homogenous communities of nodes within the Two Circles dataset. When applied to gene expression data of ER+ breast cancer patients, appropriate parameters were identified to generate a Mapper graph revealing a hotspot of ER+ patients with poor prognosis and characteristic patterns of gene expression. This was subsequently confirmed in an independent breast cancer dataset. CONCLUSIONS Our proposed method can be effectively applied for subgroup discovery with pathology data. It allows us to find optimal parameters of the Mapper algorithm, bridging the gap between its potential and the translational research.
Collapse
Affiliation(s)
- Ciara F Loughrey
- School of Electronics, Electrical Engineering and Computer Science, Queen's University Belfast, Belfast, UK
| | - Sarah Maguire
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Paweł Dłotko
- Dioscuri Centre in Topological Data Analysis, Mathematical Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Lu Bai
- School of Electronics, Electrical Engineering and Computer Science, Queen's University Belfast, Belfast, UK
| | - Nick Orr
- Patrick G Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Anna Jurek-Loughrey
- School of Electronics, Electrical Engineering and Computer Science, Queen's University Belfast, Belfast, UK.
| |
Collapse
|
39
|
De K, Jana M, Chowdhury B, Calaf GM, Roy D. Role of PARP Inhibitors: A New Hope for Breast Cancer Therapy. Int J Mol Sci 2025; 26:2773. [PMID: 40141415 PMCID: PMC11942994 DOI: 10.3390/ijms26062773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Tumors formed by the unchecked growth of breast cells are known as breast cancer. The second most frequent cancer in the world is breast cancer. It is the most common cancer among females. In 2022, 2,296,840 women were diagnosed with breast cancer. The therapy of breast cancer is evolving through the development of Poly (ADP-ribose) polymerase (PARP) inhibitors, which are offering people with specific genetic profiles new hope as research into the disease continues. It focuses on patients with BRCA1 and BRCA2 mutations. This review summarizes the most recent research on the mechanisms of action of PARP inhibitors and their implications for breast cancer therapy. We review how therapeutic applications are developing and highlight recent studies showing the effectiveness of these medicines whether used alone or in combination. Furthermore, the significance of customized therapy is highlighted in enhancing patient outcomes as we address the function of genetic testing in identifying candidates for PARP inhibition. Recommendations for future research areas to maximize the therapeutic potential of PARP inhibitors are also included, along with challenges and limits in their clinical usage. The objective of this review is to improve our comprehension of the complex interaction between breast cancer biology and PARP inhibition. This knowledge will help to guide screening approaches, improve clinical practice, and support preventive initiatives for people at risk.
Collapse
Affiliation(s)
- Kamalendu De
- Department of Biological Sciences (Botany), Midnapore City College, Midnapore 721129, West Bengal, India;
| | - Malabendu Jana
- Department of Neurological Science, Rush University School of Medicine, Chicago, IL 773, USA;
| | - Bhabadeb Chowdhury
- HIV Dynamics and Replication Program, National Institute of Health, Frederick, MD 21702, USA;
| | - Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile
| | - Debasish Roy
- Department of Natural Sciences, Hostos College of The City University of New York, Bronx, NY 718, USA;
| |
Collapse
|
40
|
MacGrogan G. [Apocrine lesions of the breast]. Ann Pathol 2025:S0242-6498(25)00031-8. [PMID: 40107901 DOI: 10.1016/j.annpat.2025.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 03/01/2025] [Indexed: 03/22/2025]
Abstract
Apocrine breast lesions encompass a spectrum of histopathological abnormalities, ranging from benign apocrine metaplasia to invasive apocrine carcinomas. Their defining feature lies in cells with abundant eosinophilic cytoplasm and round nuclei with prominent nucleoli. These cells strongly express the androgen receptor while lacking estrogen receptor-alpha and progesterone receptor expression. Benign lesions, frequently associated with mammary cysts or papillomas, lack nuclear and architectural atypia. In contrast, atypical apocrine lesions exhibit significant nuclear and structural abnormalities, posing diagnostic challenges when distinguishing them from apocrine ductal or lobular carcinoma in situ. Diagnosis relies on the extent of atypia and the presence of tumor necrosis. Invasive apocrine carcinomas are rare, accounting for less than 1% of all breast cancers, and predominantly occur in postmenopausal women. Histologically, they are often grade 1 or 2 tumors. Approximately 50% exhibit HER2 amplification and overexpression. Immunohistochemically, they are characterized by positivity for FOXA1 and GATA3, and negativity for FOXC1 and SOX10, and variable expression of TRPS1. These carcinomas belong to the molecular apocrine carcinoma family, which includes HER2-enriched tumors driven by HER2 addiction and androgen receptor-positive luminal tumors, a subtype of triple-negative breast cancers. The latter are defined by androgen receptor pathway activation and are frequently associated with PI3K pathway alterations and cell cycle dysregulation, suggesting potential therapeutic targets.
Collapse
Affiliation(s)
- Gaëtan MacGrogan
- Département de biopathologie, institut Bergonié, 229, cours de l'Argonne, 33076 Bordeaux cedex, France.
| |
Collapse
|
41
|
Rao J, Kirk PDW. VICatMix: variational Bayesian clustering and variable selection for discrete biomedical data. BIOINFORMATICS ADVANCES 2025; 5:vbaf055. [PMID: 40206332 PMCID: PMC11981716 DOI: 10.1093/bioadv/vbaf055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 03/13/2025] [Indexed: 04/11/2025]
Abstract
Summary Effective clustering of biomedical data is crucial in precision medicine, enabling accurate stratification of patients or samples. However, the growth in availability of high-dimensional categorical data, including 'omics data, necessitates computationally efficient clustering algorithms. We present VICatMix, a variational Bayesian finite mixture model designed for the clustering of categorical data. The use of variational inference (VI) in its training allows the model to outperform competitors in terms of computational time and scalability, while maintaining high accuracy. VICatMix furthermore performs variable selection, enhancing its performance on high-dimensional, noisy data. The proposed model incorporates summarization and model averaging to mitigate poor local optima in VI, allowing for improved estimation of the true number of clusters simultaneously with feature saliency. We demonstrate the performance of VICatMix with both simulated and real-world data, including applications to datasets from The Cancer Genome Atlas, showing its use in cancer subtyping and driver gene discovery. We demonstrate VICatMix's potential utility in integrative cluster analysis with different 'omics datasets, enabling the discovery of novel disease subtypes. Availability and implementation VICatMix is freely available as an R package via CRAN, incorporating C++ for faster computation, at https://CRAN.R-project.org/package=VICatMix.
Collapse
Affiliation(s)
- Jackie Rao
- MRC Biostatistics Unit, University of Cambridge, Cambridge, CB2 0SR, United Kingdom
| | - Paul D W Kirk
- MRC Biostatistics Unit, University of Cambridge, Cambridge, CB2 0SR, United Kingdom
- CRUK Cambridge Centre Ovarian Programme, University of Cambridge, Cambridge, CB2 0RE, United Kingdom
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), University of Cambridge, Cambridge, CB2 0AW, United Kingdom
| |
Collapse
|
42
|
Jensen MB, Nielsen TO, Bartlett J, Lænkholm AV, Shepherd L, Ejlertsen B. Prosigna Risk of Recurrence score and intrinsic subtypes are associated with adjuvant anthracycline chemotherapy benefit in high-risk breast cancer. NPJ Breast Cancer 2025; 11:26. [PMID: 40064871 PMCID: PMC11894038 DOI: 10.1038/s41523-025-00738-7] [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: 09/06/2024] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
NCIC-CTG MA.5 and DBCG 89D are symmetrically designed randomized trials comparing adjuvant cyclophosphamide, epirubicin, and fluorouracil with cyclophosphamide, methotrexate, and fluorouracil in high-risk breast cancer patients. In a joint analysis we evaluate the predictive value in terms of anthracycline benefit of molecular subtyping by PAM50. A statistically significant interaction (P = 0.008) between continuous Risk of Recurrence (ROR) score and treatment regimen is evident, translating into a clear distinct treatment effect according to ROR score category with HR 0.51 for ROR score ≥ 72 and HR 1.10 for ROR score < 52 (Pinteraction = 0.004). The analysis provides evidence of the benefit from anthracycline in HER2-enriched subtype; for patients with discordance of HER2 subtype and clinical HER2 status, HER2-enriched subtype was predictive of anthracycline benefit whereas clinical HER2 positive status was not. Anthracycline-based adjuvant chemotherapy may safely be withheld for patients with a low ROR score while the benefit increases with increasing ROR score.
Collapse
Affiliation(s)
- Maj-Britt Jensen
- Danish Breast Cancer Cooperative Group, Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark.
| | - Torsten O Nielsen
- Department of Pathology and Laboratory Medicine, Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, Canada
| | - John Bartlett
- Cancer Research UK Scotland Centre, University of Edinburgh, Edinburgh, United Kingdom
| | - Anne-Vibeke Lænkholm
- Department of Surgical Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Lois Shepherd
- Canadian Cancer Trials Group, Queen's University, Kingston, ON, Canada
| | - Bent Ejlertsen
- Danish Breast Cancer Cooperative Group, Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
43
|
Felsheim BM, Fernandez-Martinez A, Fan C, Pfefferle AD, Hayward MC, Hoadley KA, Rashid NU, Tolaney SM, Somlo G, Carey LA, Sikov WM, Perou CM. Prognostic and molecular multi-platform analysis of CALGB 40603 (Alliance) and public triple-negative breast cancer datasets. NPJ Breast Cancer 2025; 11:24. [PMID: 40057511 PMCID: PMC11890565 DOI: 10.1038/s41523-025-00740-z] [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: 10/04/2024] [Accepted: 02/25/2025] [Indexed: 03/30/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and heterogeneous disease that remains challenging to target with traditional therapies and to predict risk. We provide a comprehensive characterization of 238 stage II-III TNBC tumors with paired RNA and DNA sequencing data from the CALGB 40603 (Alliance) clinical trial, along with 448 stage II-III TNBC tumors with paired RNA and DNA data from three additional datasets. We identify DNA mutations associated with RNA-based subtypes, specific TP53 missense mutations compatible with potential neoantigen activity, and a consistently highly altered copy number landscape. We train exploratory multi-modal elastic net models of TNBC patient overall survival to determine the added impact of DNA-based features to RNA and clinical features. We find that mutations and copy number show little to no prognostic value, while RNA expression features, including signatures of T cell and B cell activity, along with stage, improve stratification of TNBC survival risk.
Collapse
Affiliation(s)
- Brooke M Felsheim
- Bioinformatics and Computational Biology Curriculum, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Cheng Fan
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam D Pfefferle
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Michele C Hayward
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katherine A Hoadley
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Naim U Rashid
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA
| | | | - George Somlo
- City of Hope Comprehensive Cancer Center, Duarte, CA, USA
| | - Lisa A Carey
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Division of Hematology-Oncology, Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - William M Sikov
- Program in Women's Oncology, Women and Infants Hospital of Rhode Island, Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
44
|
Hohmann L, Sigurjonsdottir K, Campos AB, Nacer DF, Veerla S, Rosengren F, Reddy PT, Häkkinen J, Nordborg N, Vallon-Christersson J, Memari Y, Black D, Bowden R, Davies HR, Borg Å, Nik-Zainal S, Staaf J. Genomic characterization of the HER2-enriched intrinsic molecular subtype in primary ER-positive HER2-negative breast cancer. Nat Commun 2025; 16:2208. [PMID: 40044693 PMCID: PMC11882987 DOI: 10.1038/s41467-025-57419-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 02/18/2025] [Indexed: 03/09/2025] Open
Abstract
ER-positive/HER2-negative (ERpHER2n) breast cancer classified as PAM50 HER2-enriched (ERpHER2n-HER2E) represents a small high-risk patient subgroup. In this study, we investigate genomic, transcriptomic, and clinical features of ERpHER2n-HER2E breast tumors using two primary ERpHER2n cohorts comprising a total of 5640 patients. We show that ERpHER2n-HER2E tumors exhibit aggressive clinical features and poorer clinical outcomes compared to Luminal A and Luminal B tumors. Furthermore, ERpHER2n-HER2E breast cancer does not consist of misclassified or HER2-low cases, has little impact of ERBB2, is highly proliferative and less ER dependent than other luminal subtypes. It is not an obvious biological entity but is nevertheless associated with potentially targetable molecular features, notably a high immune response and high FGFR4 expression. Strikingly, molecular features that define the HER2E subtype in luminal disease are also consistent in HER2-positive disease, including an epigenetic mechanism for high FGFR4 expression in breast cancer.
Collapse
Affiliation(s)
- Lennart Hohmann
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Kristin Sigurjonsdottir
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ana Bosch Campos
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Deborah F Nacer
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Srinivas Veerla
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Frida Rosengren
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | - Jari Häkkinen
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Nicklas Nordborg
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | | | - Yasin Memari
- Academic Department of Medical Genetics, School of Clinical Medicine & Early Cancer Institute, University of Cambridge, Cambridge, UK
| | - Daniella Black
- Academic Department of Medical Genetics, School of Clinical Medicine & Early Cancer Institute, University of Cambridge, Cambridge, UK
| | - Ramsay Bowden
- Academic Department of Medical Genetics, School of Clinical Medicine & Early Cancer Institute, University of Cambridge, Cambridge, UK
| | - Helen R Davies
- Academic Department of Medical Genetics, School of Clinical Medicine & Early Cancer Institute, University of Cambridge, Cambridge, UK
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden
| | - Serena Nik-Zainal
- Academic Department of Medical Genetics, School of Clinical Medicine & Early Cancer Institute, University of Cambridge, Cambridge, UK
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Lund, Sweden.
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
45
|
Callari M, Dugo M, Barreca M, Győrffy B, Galbardi B, Vigano L, Locatelli A, Dall'Ara C, Ferrarini M, Bisagni G, Colleoni M, Mansutti M, Zamagni C, Del Mastro L, Zambelli S, Frassoldati A, Biasi O, Pusztai L, Valagussa P, Viale G, Gianni L, Bianchini G. Determinants of response and molecular dynamics in HER2+ER+ breast cancers from the NA-PHER2 trial receiving HER2-targeted and endocrine therapies. Nat Commun 2025; 16:2195. [PMID: 40038334 PMCID: PMC11880565 DOI: 10.1038/s41467-025-57293-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 02/12/2025] [Indexed: 03/06/2025] Open
Abstract
Improved outcomes in HER2+ female breast cancer have resulted from chemotherapy and anti-HER2 therapies. However, HER2+ER+ cancers exhibit lower response rates. The phase 2 NA-PHER2 trial (NCT02530424) investigated chemo-free preoperative HER2 blockade (trastuzumab + pertuzumab) and CDK4/6 inhibition (palbociclib) with or without endocrine therapy (fulvestrant) in HER2+ER+ breast cancer. Clinical endpoints (i.e. Ki67 dynamics and pathological complete response) were previously reported. Here we report on the biomarker analysis, secondary objective of the study. Through RNA sequencing and tumour infiltrating lymphocytes (TIL) assessment in serial biopsies, we identified biomarkers predictive of pCR or Day14 Ki67 response and unveiled treatment-induced molecular changes. High immune infiltration and low ER signalling correlated with pCR, while TP53 mutations associated with high Day14 Ki67. Stratification based on Ki67 at Day14 and at surgery defined three response groups (Ki67 HighHigh, LowHigh, LowLow), with divergent tumour and stroma expression dynamics. The HighHigh group showed dysfunctional immune infiltration and overexpression of therapeutic targets like PAK4 at baseline. The LowLow group exhibited a Luminal A phenotype by the end of treatment. This study expands our understanding of drivers and dynamics of HER2+ER+ tumour response, towards treatment tailoring.
Collapse
MESH Headings
- Humans
- Female
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/genetics
- Ki-67 Antigen/metabolism
- Ki-67 Antigen/genetics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Receptors, Estrogen/metabolism
- Receptors, Estrogen/genetics
- Trastuzumab/therapeutic use
- Trastuzumab/administration & dosage
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/drug effects
- Pyridines/therapeutic use
- Pyridines/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Tumor Suppressor Protein p53/genetics
- Treatment Outcome
- Middle Aged
- Antineoplastic Agents, Hormonal/therapeutic use
- Piperazines
Collapse
Affiliation(s)
| | | | - Marco Barreca
- Fondazione Michelangelo, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Balázs Győrffy
- Dept. of Bioinformatics, Semmelweis University, Budapest, Hungary
- Dept. of Biophysics, Medical School, University of Pecs, Pecs, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, Budapest, Hungary
| | | | | | | | | | | | | | - Marco Colleoni
- IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | | | | | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, Università di Genova, Genoa, Italy
- Department of Medical Oncology, UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | | | - Olivia Biasi
- IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Lajos Pusztai
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, New Haven, CT, USA
| | | | - Giuseppe Viale
- Fondazione Michelangelo, Milan, Italy
- IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | | | - Giampaolo Bianchini
- IRCCS San Raffaele Hospital, Milan, Italy.
- UniSR San Raffaele University, Milan, Italy.
| |
Collapse
|
46
|
Yang S, Hu L, Chen P, Zeng X, Mao S. AJGM: joint learning of heterogeneous gene networks with adaptive graphical model. Bioinformatics 2025; 41:btaf096. [PMID: 40073230 PMCID: PMC11937957 DOI: 10.1093/bioinformatics/btaf096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/29/2025] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
MOTIVATION Inferring gene networks provides insights into biological pathways and functional relationships among genes. When gene expression samples exhibit heterogeneity, they may originate from unknown subtypes, prompting the utilization of mixture Gaussian graphical model (GGM) for simultaneous subclassification and gene network inference. However, this method overlooks the heterogeneity of network relationships across subtypes and does not sufficiently emphasize shared relationships. Additionally, GGM assumes data follows a multivariate Gaussian distribution, which is often not the case with zero-inflated scRNA-seq data. RESULTS We propose an Adaptive Joint Graphical Model (AJGM) for estimating multiple gene networks from single-cell or bulk data with unknown heterogeneity. In AJGM, an overall network is introduced to capture relationships shared by all samples. The model establishes connections between the subtype networks and the overall network through adaptive weights, enabling it to focus more effectively on gene relationships shared across all networks, thereby enhancing the accuracy of network estimation. On synthetic data, the proposed approach outperforms existing methods in terms of sample classification and network inference, particularly excelling in the identification of shared relationships. Applying this method to gene expression data from triple-negative breast cancer confirms known gene pathways and hub genes, while also revealing novel biological insights. AVAILABILITY AND IMPLEMENTATION The Python code and demonstrations of the proposed approaches are available at https://github.com/yyytim/AJGM, and the software is archived in Zenodo with DOI: 10.5281/zenodo.14740972.
Collapse
Affiliation(s)
- Shunqi Yang
- Department of Statistics, Hunan University, Changsha, Hunan, 410006, China
| | - Lingyi Hu
- Department of Statistics, Hunan University, Changsha, Hunan, 410006, China
| | - Pengzhou Chen
- Department of Statistics, Hunan University, Changsha, Hunan, 410006, China
| | - Xiangxiang Zeng
- College of Computer Science and Electronic Engineering, Hunan University, Changsha, Hunan, 410082, China
| | - Shanjun Mao
- Department of Statistics, Hunan University, Changsha, Hunan, 410006, China
| |
Collapse
|
47
|
Kallah-Dagadu G, Mohammed M, Nasejje JB, Mchunu NN, Twabi HS, Batidzirai JM, Singini GC, Nevhungoni P, Maposa I. Breast cancer prediction based on gene expression data using interpretable machine learning techniques. Sci Rep 2025; 15:7594. [PMID: 40038307 PMCID: PMC11880515 DOI: 10.1038/s41598-025-85323-5] [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: 06/03/2024] [Accepted: 01/01/2025] [Indexed: 03/06/2025] Open
Abstract
Breast cancer remains a global health burden, with an increase in deaths related to this particular cancer. Accurately predicting and diagnosing breast cancer is important for treatment development and survival of patients. This study aimed to accurately predict breast cancer using a dataset comprising 1208 observations and 3602 genes. The study employed feature selection techniques to identify the most influential predictive genes for breast cancer using machine learning (ML) models. The study used K-nearest Neighbors (KNN), random forests (RF), and a support vector machine (SVM). Furthermore, the study employed feature- and model-based importance and explainable ML methods, including Shapley values, Partial dependency (PDPS), and Accumulated Local Effects (ALE) plots, to explain the genes' importance ranking from the ML methods. Shapley values highlighted the significance of some of the genes in predicting cancer presence. Model-based feature ranking techniques, particularly the Leaving-One-Covariate-In (LOCI) method, identified the ten most critical genes for predicting tumor cases. The LOCI rankings from the SVM and RF methods were aligned. Additionally, visualization methods such as PDPS and ALE plots demonstrated how individual feature changes affect predictions and interactions with other genes. By combining feature selection techniques and explainable ML methods, this study has demonstrated the interpretability and reliability of machine learning models for breast cancer prediction, emphasizing the importance of incorporating explainable ML approaches for medical decision-making.
Collapse
Affiliation(s)
- Gabriel Kallah-Dagadu
- Department of Statistics and Actuarial Science, University of Ghana, Accra, Ghana
- School of Mathematics, Statistics, and Computer Science, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Mohanad Mohammed
- School of Mathematics, Statistics, and Computer Science, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | - Justine B Nasejje
- School of Statistics and Actuarial Science, University of the Witwatersrand, Johannesburg-Braamfontein, South Africa
| | | | - Halima S Twabi
- Department of Mathematical Sciences, University of Malawi, Zomba, Malawi
| | - Jesca Mercy Batidzirai
- School of Mathematics, Statistics, and Computer Science, University of KwaZulu-Natal, Pietermaritzburg, South Africa
| | | | - Portia Nevhungoni
- Biostatistics Research Unit, South African Medical Research Council, Pretoria, South Africa
| | - Innocent Maposa
- Division of Epidemiology and Biostatistics, Department of Global Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, Cape Town, South Africa.
| |
Collapse
|
48
|
McBean B, Abou Zeidane R, Lichtman-Mikol S, Hauk B, Speers J, Tidmore S, Flores CL, Rana PS, Pisano C, Liu M, Santola A, Montero A, Boyle AP, Speers CW. MELK as a Mediator of Stemness and Metastasis in Aggressive Subtypes of Breast Cancer. Int J Mol Sci 2025; 26:2245. [PMID: 40076867 PMCID: PMC11900306 DOI: 10.3390/ijms26052245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the breast cancer subtype with the poorest prognosis and lacks actionable molecular targets for treatment. Maternal embryonic leucine zipper kinase (MELK) is highly expressed in TNBC and has been implicated in poor clinical outcomes, though its mechanistic role in the aggressive biology of TNBC is poorly understood. Here, we demonstrate a role of MELK in TNBC progression and metastasis. Analysis of publicly available datasets revealed that high MELK expression correlates with worse overall survival, recurrence-free survival, and distant metastasis-free survival, and MELK is co-expressed with metastasis-related genes. Functional studies demonstrated that MELK inhibition, using genomic or pharmacologic inhibition, reduces mammosphere formation, migration, and invasion in high-MELK-expressing TNBC cell lines. Conversely, MELK overexpression in low-MELK-expressing cell lines significantly increased invasive capacity in vitro and metastatic potential in vivo, as evidenced by enhanced metastasis to the liver and lungs in a chorioallantoic membrane assay. These findings highlight MELK as a key regulator of TNBC aggressiveness and support its potential as a therapeutic target to mitigate metastasis and improve patient outcomes.
Collapse
Affiliation(s)
- Breanna McBean
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA; (B.M.); (A.P.B.)
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Reine Abou Zeidane
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Samuel Lichtman-Mikol
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Benjamin Hauk
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Johnathan Speers
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Savannah Tidmore
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Citlally Lopez Flores
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Priyanka S. Rana
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Courtney Pisano
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Meilan Liu
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA; (M.L.); (A.S.)
| | - Alyssa Santola
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA; (M.L.); (A.S.)
| | - Alberto Montero
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alan P. Boyle
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109, USA; (B.M.); (A.P.B.)
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Corey W. Speers
- Department of Radiation Oncology, University Hospitals Case Medical Center, Cleveland, OH 44106, USA; (R.A.Z.); (S.L.-M.); (B.H.); (J.S.); (S.T.); (C.L.F.); (P.S.R.); (C.P.); (A.M.)
- Department of Radiation Oncology, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI 48109, USA; (M.L.); (A.S.)
| |
Collapse
|
49
|
Meng M, Wang J, Yang J, Zhang Y, Tu X, Hu P. PRR13 expression as a prognostic biomarker in breast cancer: correlations with immune infiltration and clinical outcomes. Front Mol Biosci 2025; 12:1518031. [PMID: 40099041 PMCID: PMC11911201 DOI: 10.3389/fmolb.2025.1518031] [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: 10/27/2024] [Accepted: 01/08/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Breast cancer continues to be a primary cause of cancer-related mortality among women globally. Identifying novel biomarkers is essential for enhancing patient prognosis and informing therapeutic decisions. The PRR13 gene, associated with taxol resistance and the progression of various cancers, remains under-characterized in breast cancer. This study aimed to investigate the role of PRR13 in breast cancer and its potential as a prognostic biomarker. Methods We performed a comparative analysis of PRR13 gene expression utilizing the TCGA database against non-cancerous tissues and employed STRING to evaluate PRR13's protein-protein interactions and associated pathways. Additionally, we investigated the relationship between PRR13 mRNA expression and immune cell infiltration in breast cancer (BRCA) using two methodologies. Furthermore, a retrospective analysis of 160 patients was conducted, wherein clinical data were collected and PRR13 expression was evaluated through immunohistochemistry and qRT-PCR to determine its association with clinicopathological features and patient survival. Results Analysis of the TCGA database revealed significant upregulation of PRR13 expression across 12 different cancer types, including breast cancer. High PRR13 expression was positively correlated with various immune cells, including NK cells, eosinophils, Th17 cells, and mast cells, whereas a negative correlation was observed with B cells, macrophages, and other immune subsets. Enrichment analysis of PRR13 and its 50 interacting proteins revealed significant associations with biological processes such as cell adhesion and migration, and pathways including ECMreceptor interaction and PI3K-Akt signaling. Single-cell analysis demonstrated associations between PRR13 and pathways pertinent to inflammation and apoptosis. Validation studies confirmed elevated PRR13 expression in tumor tissue compared to adjacent non-cancerous tissue. Immunohistochemistry demonstrated high PRR13 expression in 55.6% of cancer cases, particularly associated with advanced clinical stage and lymph node metastasis. Moreover, high PRR13 expression significantly correlated with shorter overall survival and served as an independent prognostic factor. Subgroup analysis underscored the prognostic significance of PRR13 in aggressive tumor subtypes, with particularly strong associations observed in T3, N1-3, and moderately to poorly differentiated tumors. Discussion In conclusion, PRR13 expression is upregulated in breast cancer tissues and may serve as a valuable prognostic indicator for breast cancer patients, potentially impacting patient survival and therapeutic strategies.
Collapse
Affiliation(s)
- Mingjing Meng
- Department of Research and Foreign Affairs, The Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China
| | - Jiani Wang
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jiumei Yang
- Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, China
| | - Yangming Zhang
- Equipment Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xusheng Tu
- Emergency Department, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Pan Hu
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
50
|
Lin S, Nguyen LL, McMellen A, Leibowitz MS, Davidson N, Spinosa D, Bitler BG. Leveraging Multi-omics to Disentangle the Complexity of Ovarian Cancer. Mol Diagn Ther 2025; 29:145-151. [PMID: 39557776 DOI: 10.1007/s40291-024-00757-3] [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] [Accepted: 10/29/2024] [Indexed: 11/20/2024]
Abstract
To better understand ovarian cancer lethality and treatment resistance, sophisticated computational approaches are required that address the complexity of the tumor microenvironment, genomic heterogeneity, and tumor evolution. The ovarian cancer tumor ecosystem consists of multiple tumors and cell types that support disease growth and progression. Over the last two decades, there has been a revolution in -omic methodologies to broadly define components and essential processes within the tumor microenvironment, including transcriptomics, metabolomics, proteomics, genome sequencing, and single-cell analyses. While most of these technologies comprehensively characterize a single biological process, there is a need to understand the biological and clinical impact of integrating multiple -omics platforms. Overall, multi-omics is an intriguing analytic framework that can better approximate biological complexity; however, data aggregation and integration pipelines are not yet sufficient to reliably glean insights that affect clinical outcomes.
Collapse
Affiliation(s)
- Shijuan Lin
- Divisions of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver, Anschutz Medical Campus, 12700 East 19th Avenue, MS 8613, Aurora, CO, 80045, USA
| | - Lily L Nguyen
- Divisions of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver, Anschutz Medical Campus, 12700 East 19th Avenue, MS 8613, Aurora, CO, 80045, USA
| | - Alexandra McMellen
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| | - Michael S Leibowitz
- Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| | - Natalie Davidson
- Divisions of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver, Anschutz Medical Campus, 12700 East 19th Avenue, MS 8613, Aurora, CO, 80045, USA
| | - Daniel Spinosa
- Gynecologic Oncology, Department of Obstetrics and Gynecology, University of Colorado Denver, Anschutz Medical Campus, Aurora, CO, USA
| | - Benjamin G Bitler
- Divisions of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver, Anschutz Medical Campus, 12700 East 19th Avenue, MS 8613, Aurora, CO, 80045, USA.
| |
Collapse
|