1
|
Wilk SM, Lee K, Castillo CC, Haloul M, Gajda AM, Macias V, Wiley EL, Chen Z, Liu X, Wang X, Sverdlov M, Hoskins KF, Er EE. Multiplex imaging reveals novel patterns of MRTFA/B activation in the breast cancer microenvironment. J Transl Med 2025; 23:599. [PMID: 40448123 DOI: 10.1186/s12967-025-06559-3] [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: 01/30/2025] [Accepted: 05/02/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND Breast cancer progression and metastasis involve the action of multiple transcription factors in tumors and in the cells of the tumor microenvironment (TME) and understanding how these transcription factors are coordinated can guide novel therapeutic strategies. Myocardin-related transcription factors A and B (MRTFA/B also known as MKL1/2) are two related transcription factors that redundantly control cancer cell invasion and metastasis in mouse models of breast cancer, but their roles in human cancer are incompletely understood. Here, we investigated the expression and activation of these transcription factors to better assess their tumorigenic and metastatic impact on breast cancer and cells of the tumor microenvironment. METHODS We used a multiplexed immunofluorescence approach to label MRTFA, MRTFB, tumor cells by using pan Cytokeratin, endothelial cells by using CD31, and antigen presenting cells (APCs) by using HLA-DRA on two different breast cancer tissue microarrays (TMA): The breast cancer progression TMA provided by the Cooperative Human Tissue Network (CHTN_BrCaProg3) and the University of Illinois Breast Cancer Working Group (TMA BCWG UIC-001-TMA) that included primary tumor and lymph node metastases from patients residing in the West Side and South Side of Chicago. We also used bioinformatics analyses of the TCGA and METABRIC databases and the Broad Institute's single-cell RNA sequencing portal to investigate MRTFA/B expression patterns in the cells of the tumor microenvironment (TME). RESULTS We found that in human tumors, MRTFA/B are concurrently activated in cancer cells, but they show distinct patterns of expression across different histological subtypes and in the cells of the TME. Importantly, MRTFA expression was elevated in metastatic tumors of African American patients, who disproportionately die from breast cancer. Interestingly, in contrast to publicly available mRNA expression data, MRTFA was similarly expressed across estrogen receptor (ER) positive and negative breast tumors, while MRTFB expression was highest in ER+ breast tumors. Furthermore, MRTFA was specifically expressed in the perivascular antigen-presenting cells (APCs), which has been previously associated with immune suppression and breast cancer progression. We also found that MRTFA expression correlated with the expression of the immune checkpoint protein V-set immunoregulatory receptor (VSIR) in the TCGA data and found that MRTFA activity promotes VSIR expression in THP-1 monocytes and cultured HEK293 cells. CONCLUSIONS Our results provide unique insights into how MRTFA and MRTFB promote metastasis in human cancer, the differences of their expression patterns, and their immune suppressive function within the breast cancer TME. Our results will guide future studies on targeting MRTFA/B transcriptional activity and the resulting immune suppression in breast cancer.
Collapse
Affiliation(s)
- Stephanie M Wilk
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Kihak Lee
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Caitlyn C Castillo
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Mohamed Haloul
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Alexa M Gajda
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Virgilia Macias
- Department of Pathology, University of Illinois Chicago, Chicago, IL, USA
| | - Elizabeth L Wiley
- Department of Pathology, University of Illinois Chicago, Chicago, IL, USA
| | - Zhengjia Chen
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois Chicago, Chicago, IL, USA
- Biostatistics Shared Resource, University of Illinois Cancer Center, Chicago, IL, USA
| | - Xinyi Liu
- Department of Pharmacology & Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Xiaowei Wang
- Department of Pharmacology & Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Maria Sverdlov
- Research Histology Core, Research Resources Center, University of Illinois Chicago, Chicago, IL, USA
| | - Kent F Hoskins
- Division of Hematology/Oncology, College of Medicine, University of Illinois Chicago, Chicago, IL, USA
| | - Ekrem Emrah Er
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Dong C, Sun Y, Xu X, Li H, Song X, Wei W, Jiao C, Xu H, Liu Y, Mierzhakenmu Z, Li L, Ma B. c-Myc knockdown restores tamoxifen sensitivity in triple-negative breast cancer by reactivating the expression of ERα: the central role of miR-152 and miR-148a. Breast Cancer 2025; 32:529-542. [PMID: 40029493 DOI: 10.1007/s12282-025-01683-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Poor prognosis of triple-negative breast cancer (TNBC) is owing to its intrinsic heterogeneity and lack of targeted therapies. Emerging evidence has characterized that targeting c-Myc might be a promising way to treat TNBC. METHODS c-Myc knocked down TNBC cells were generated and the tamoxifen sensitivity was determined. Methylation-specific PCR analysis was used to detect the methylation status of ERα promoter, and c-Myc-mediated miRNA transcription was examined using chromatin immunoprecipitation and dual-luciferase reporter assays. The in vivo tamoxifen sensitivity was determined by mouse xenograft model. RESULTS c-Myc knockdown in TNBC cells leads to the reactivation of ERα and consequent acquisition its sensitivity to tamoxifen. c-Myc depletion decreased the methylation in the promoter of ERα and DNMT1 was identified as the main executor. c-Myc knockdown-induced tamoxifen sensitivity was reversed by DNMT1 overexpression. The expression of miR-152-3p and miR-148a-3p was largely induced in c-Myc knockdown TNBC cells, and both miR-152-3p and miR-148a-3p could target DNMT1 to regulate its expression. c-Myc binds to the promoter regions of miR-152-3p and miR-148a-3p to exert transcriptional suppression. By suppressing miR-152-3p or miR-148a-3p expression using inhibitors, enhanced sensitivity to tamoxifen induced by c-Myc knockdown was partially reversed. In vivo xenograft tumor model demonstrated that c-Myc knockdown mildly inhibits the growth of tumor, and a dramatic decline was observed when administrated with tamoxifen combined with c-Myc knockdown. CONCLUSION Our study first illustrated that c-Myc knockdown in TNBC cells reactivate ERα expression in a miR-152/miR-148a-DNMT1-dependent manner, and brought new sights into treating TNBC using hormonal therapies.
Collapse
Affiliation(s)
- Chao Dong
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Yonghong Sun
- Department of Central Operating Room, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Xiaoli Xu
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Huiling Li
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Xinyu Song
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Wenxin Wei
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Chong Jiao
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Haoyi Xu
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Yuanjing Liu
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Zuliyaer Mierzhakenmu
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China
| | - Li Li
- Department of Gynecological Oncology (First Ward), The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
| | - Binlin Ma
- Department of Breast and Thyroid Surgery, the Clinical Medical Research Center of Breast and Thyroid Tumor in Xinjiang, The Affiliated Tumor Hospital of Xinjiang Medical University, No. 789 Suzhou East Street, Urumqi, 830000, Xinjiang Uygur Autonomous Region, China.
| |
Collapse
|
3
|
Emara HM, Allam NK, Youness RA. A comprehensive review on targeted therapies for triple negative breast cancer: an evidence-based treatment guideline. Discov Oncol 2025; 16:547. [PMID: 40244488 PMCID: PMC12006628 DOI: 10.1007/s12672-025-02227-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive malignancy characterized by limited therapeutic options and poor prognosis. Despite advancements in precision oncology, conventional chemotherapy remains the cornerstone of TNBC treatment, often accompanied by debilitating side effects and suboptimal outcomes. This review presents a comprehensive analysis of clinical trials on targeted therapies, aiming to establish a novel, evidence-based treatment strategy exclusively leveraging molecularly targeted agents. By integrating patient-specific genetic profiles with therapeutic responses observed across various clinical trial phases, this approach seeks to optimize efficacy while minimizing toxicity. The proposed targeted therapy combinations hold significant potential to revolutionize TNBC treatment, offering a paradigm shift toward precision medicine and improved patient outcomes.
Collapse
Affiliation(s)
- Hadir M Emara
- Nanotechnology Program, School of Sciences & Engineering, The American University in Cairo, New Cairo, 11835, Egypt.
| | - Nageh K Allam
- Nanotechnology Program, School of Sciences & Engineering, The American University in Cairo, New Cairo, 11835, Egypt.
- Energy Materials Laboratory, Physics Department, School of Sciences & Engineering, The American University in Cairo, New Cairo, 11835, Egypt.
| | - Rana A Youness
- Department of Molecular Biology and Biochemistry, Faculty of Biotechnology, German International University, New Administrative Capital, Cairo, Egypt.
| |
Collapse
|
4
|
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
|
5
|
Yıldiz A, Bilici A, Acikgoz O, Hamdard J, Basim P, Cakir T, Cakir A, Olmez OF, Gezen C, Yildiz O. Prognostic implications of response to neoadjuvant chemotherapy in breast cancer subtypes. J Chemother 2025; 37:60-68. [PMID: 38351652 DOI: 10.1080/1120009x.2024.2314830] [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: 06/26/2023] [Revised: 01/23/2024] [Accepted: 02/01/2024] [Indexed: 01/21/2025]
Abstract
The current study was designed to assess the response to treatment, as well as clinical and survival outcomes, across different breast cancer subtypes in patients who underwent neoadjuvant chemotherapy (NAC). From 2014 to 2019, a total of 139 patients who were histologically confirmed to have breast cancer, underwent NAC, and subsequently received breast and axillary surgery, were retrospectively included in this study. The rates of pathological complete response (pCR) to NAC were significantly higher for HER2-positive and triple-negative subtypes than for luminal A and HER2-negative subtypes (p = 0.013). Multivariate analysis for disease-free survival (DFS) revealed that tumour grade and the presence of pCR were independent prognostic factors. The presence or absence of a pCR with NAC was an independent prognostic indicator in the multivariate analysis for overall survival (OS). Lastly, achieving a pCR was independently predicted by 18F-FDG PET/CT findings, the HER2-positive subtype, and the triple-negative subtype. Despite the inherent methodological limitations, our findings underscore the significance of identifying predictive markers to tailor NAC plans, with the aim of improving survival outcomes.
Collapse
Affiliation(s)
- Anil Yıldiz
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| | - Ahmet Bilici
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| | - Ozgur Acikgoz
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| | - Jamshid Hamdard
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| | - Pelin Basim
- Department of Breast Surgery, Istanbul Medipol University, Istanbul, Turkey
| | - Tansel Cakir
- Department of Nuclear Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Asli Cakir
- Department of Pathology, Istanbul Medipol University, Istanbul, Turkey
| | - Omer Fatih Olmez
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| | - Cem Gezen
- Department of Breast Surgery, Istanbul Medipol University, Istanbul, Turkey
| | - Ozcan Yildiz
- Department of Medical Oncology, Medical Faculty, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
6
|
East MP, Sprung RW, Okumu DO, Olivares-Quintero JF, Joisa CU, Chen X, Zhang Q, Erdmann-Gilmore P, Mi Y, Sciaky N, Malone JP, Bhatia S, McCabe IC, Xu Y, Sutcliffe MD, Luo J, Spears PA, Perou CM, Earp HS, Carey LA, Yeh JJ, Spector DL, Gomez SM, Spanheimer PM, Townsend RR, Johnson GL. Quantitative proteomic mass spectrometry of protein kinases to determine dynamic heterogeneity of the human kinome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.04.614143. [PMID: 39464086 PMCID: PMC11507871 DOI: 10.1101/2024.10.04.614143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The kinome is a dynamic system of kinases regulating signaling networks in cells and dysfunction of protein kinases contributes to many diseases. Regulation of the protein expression of kinases alters cellular responses to environmental changes and perturbations. We configured a library of 672 proteotypic peptides to quantify >300 kinases in a single LC-MS experiment using ten micrograms protein from human tissues including biopsies. This enables absolute quantitation of kinase protein abundance at attomole-femtomole expression levels, requiring no kinase enrichment and less than ten micrograms of starting protein from flash-frozen and formalin fixed paraffin embedded tissues. Breast cancer biopsies, organoids, and cell lines were analyzed using the SureQuant method, demonstrating the heterogeneity of kinase protein expression across and within breast cancer clinical subtypes. Kinome quantitation was coupled with nanoscale phosphoproteomics, providing a feasible method for novel clinical diagnosis and understanding of patient kinome responses to treatment.
Collapse
|
7
|
Hsieh RW, Symonds LK, Siu J, Cohen SA. Identification of circulating tumor DNA as a biomarker for diagnosis and response to therapies in cancer patients. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 391:43-93. [PMID: 39939078 DOI: 10.1016/bs.ircmb.2024.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
The sampling of circulating biomarkers provides an opportunity for non-invasive evaluation and monitoring of cancer activity. In modern day practice, this has typically been in the form of circulating tumor DNA (ctDNA) detected in plasma. The field of ctDNA has been a burgeoning technology, with prominent applications for blood-based cancer screening and in disease status assessment, especially after curative-intent surgery to evaluate for minimal residual disease (MRD). Clinical applications for the latter show an incredibly high sensitivity in certain cancer types with a need for additional studies to determine how much clinical decision-making should be adapted based on ctDNA results and which cancer types, stages, and treatments are best informed by ctDNA results. This chapter provides an overview of ctDNA detection as tool for cancer screening, detecting MRD, and/or molecularly characterizing a cancer, highlighting the rapidly amassing research as a prognostic biomarker and emerging data on ctDNA as a predictive biomarker.
Collapse
Affiliation(s)
- Ronan W Hsieh
- Division of Hematology/Oncology, University of Washington, Seattle, WA, United States; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Lynn K Symonds
- Division of Hematology/Oncology, University of Washington, Seattle, WA, United States; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States
| | - Jason Siu
- Department of Laboratory Medicine, University of Washington, Seattle, WA, United States
| | - Stacey A Cohen
- Division of Hematology/Oncology, University of Washington, Seattle, WA, United States; Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA, United States.
| |
Collapse
|
8
|
Jayaswal N, Srivastava S, Kumar S, Belagodu Sridhar S, Khalid A, Najmi A, Zoghebi K, Alhazmi HA, Mohan S, Tambuwala MM. Precision arrows: Navigating breast cancer with nanotechnology siRNA. Int J Pharm 2024; 662:124403. [PMID: 38944167 DOI: 10.1016/j.ijpharm.2024.124403] [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: 03/28/2024] [Revised: 06/26/2024] [Accepted: 06/26/2024] [Indexed: 07/01/2024]
Abstract
Nanotechnology-based drug delivery systems, including siRNA, present an innovative approach to treating breast cancer, which disproportionately affects women. These systems enable personalized and targeted therapies, adept at managing drug resistance and minimizing off-target effects. This review delves into the current landscape of nanotechnology-derived siRNA transport systems for breast cancer treatment, discussing their mechanisms of action, preclinical and clinical research, therapeutic applications, challenges, and future prospects. Emphasis is placed on the importance of targeted delivery and precise gene silencing in improving therapeutic efficacy and patient outcomes. The review addresses specific hurdles such as specificity, biodistribution, immunological reactions, and regulatory approval, offering potential solutions and avenues for future research. SiRNA drug delivery systems hold promise in revolutionizing cancer care and improving patient outcomes, but realizing their full potential necessitates ongoing research, innovation, and collaboration. Understanding the intricacies of siRNA delivery mechanisms is pivotal for designing effective cancer treatments, overcoming challenges, and advancing siRNA-based therapies for various diseases, including cancer. The article provides a comprehensive review of the methods involved in siRNA transport for therapeutic applications, particularly in cancer treatment, elucidating the complex journey of siRNA molecules from extracellular space to intracellular targets. Key mechanisms such as endocytosis, receptor-mediated uptake, and membrane fusion are explored, alongside innovative delivery vehicles and technologies that enhance siRNA delivery efficiency. Moreover, the article discusses challenges and opportunities in the field, including issues related to specificity, biodistribution, immune response, and clinical translation. By comprehending the mechanisms of siRNA delivery, researchers can design and develop more effective siRNA-based therapies for various diseases, including cancer.
Collapse
Affiliation(s)
- Nandani Jayaswal
- Faculty of Pharmaceutical Sciences, Mahayogi Gorakhnath University, Gorakhpur, 273007, India
| | - Shriyansh Srivastava
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 273007, India; Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, India.
| | - Sachin Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 273007, India
| | | | - Asaad Khalid
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia.
| | - Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Khalid Zoghebi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Hassan A Alhazmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Syam Mohan
- Substance Abuse and Toxicology Research Centre, Jazan University, Jazan 45142, Saudi Arabia; School of Health Sciences, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India; Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India
| | - Murtaza M Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK; RAK College of Pharmacy, RAK Medical and Health Sciences University, Ras Al Khaimah, UAE.
| |
Collapse
|
9
|
Qattan A, Al-Tweigeri T, Suleman K, Alkhayal W, Tulbah A. Advanced Insights into Competitive Endogenous RNAs (ceRNAs) Regulated Pathogenic Mechanisms in Metastatic Triple-Negative Breast Cancer (mTNBC). Cancers (Basel) 2024; 16:3057. [PMID: 39272915 PMCID: PMC11394539 DOI: 10.3390/cancers16173057] [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: 08/01/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024] Open
Abstract
Triple-negative breast cancer is aggressive and challenging to treat because of a lack of targets and heterogeneity among tumors. A paramount factor in the mortality from breast cancer is metastasis, which is driven by genetic and phenotypic alterations that drive epithelial-mesenchymal transition, stemness, survival, migration and invasion. Many genetic and epigenetic mechanisms have been identified in triple-negative breast cancer that drive these metastatic phenotypes; however, this knowledge has not yet led to the development of effective drugs for metastatic triple-negative breast cancer (mTNBC). One that may not have received enough attention in the literature is post-translational regulation of broad sets of cancer-related genes through inhibitory microRNAs and the complex competitive endogenous RNA (ceRNA) regulatory networks they are influenced by. This field of study and the resulting knowledge regarding alterations in these networks is coming of age, enabling translation into clinical benefit for patients. Herein, we review metastatic triple-negative breast cancer (mTNBC), the role of ceRNA network regulation in metastasis (and therefore clinical outcomes), potential approaches for therapeutic exploitation of these alterations, knowledge gaps and future directions in the field.
Collapse
Affiliation(s)
- Amal Qattan
- Department of Molecular Oncology, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
- College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia
| | - Taher Al-Tweigeri
- Department of Medical Oncology, Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Kausar Suleman
- Department of Medical Oncology, Oncology Centre, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Wafa Alkhayal
- Department of Surgery, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| | - Asma Tulbah
- Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh 11211, Saudi Arabia
| |
Collapse
|
10
|
Schneider BP, Zhao F, Ballinger TJ, Garcia SF, Shen F, Virani S, Cella D, Bales C, Jiang G, Hayes L, Miller N, Srinivasiah J, Stringer-Reasor EM, Chitalia A, Davis AA, Makower DF, Incorvati J, Simon MA, Mitchell EP, DeMichele A, Miller KD, Sparano JA, Wagner LI, Wolff AC. ECOG-ACRIN EAZ171: Prospective Validation Trial of Germline Predictors of Taxane-Induced Peripheral Neuropathy in Black Women With Early-Stage Breast Cancer. J Clin Oncol 2024; 42:2899-2907. [PMID: 38828938 PMCID: PMC11670807 DOI: 10.1200/jco.24.00526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/16/2024] [Accepted: 05/02/2024] [Indexed: 06/05/2024] Open
Abstract
PURPOSE Black women experience higher rates of taxane-induced peripheral neuropathy (TIPN) compared with White women when receiving adjuvant once weekly paclitaxel for early-stage breast cancer, leading to more dose reductions and higher recurrence rates. EAZ171 aimed to prospectively validate germline predictors of TIPN and compare rates of TIPN and dose reductions in Black women receiving (neo)adjuvant once weekly paclitaxel and once every 3 weeks docetaxel for early-stage breast cancer. METHODS Women with early-stage breast cancer who self-identified as Black and had intended to receive (neo)adjuvant once weekly paclitaxel or once every 3 weeks docetaxel were eligible, with planned accrual to 120 patients in each arm. Genotyping was performed to determine germline neuropathy risk. Grade 2-4 TIPN by Common Terminology Criteria for Adverse Events (CTCAE) v5.0 was compared between high- versus low-risk genotypes and between once weekly paclitaxel versus once every 3 weeks docetaxel within 1 year. Patient-rated TIPN and patient-reported outcomes were compared using patient-reported outcome (PRO)-CTCAE and Functional Assessment of Cancer Therapy/Gynecologic Oncology Group-Neurotoxicity. RESULTS Two hundred and forty of 249 enrolled patients had genotype data, and 91 of 117 (77.8%) receiving once weekly paclitaxel and 87 of 118 (73.7%) receiving once every 3 weeks docetaxel were classified as high-risk. Physician-reported grade 2-4 TIPN was not significantly different in high- versus low-risk genotype groups with once weekly paclitaxel (47% v 35%; P = .27) or with once every 3 weeks docetaxel (28% v 19%; P = .47). Grade 2-4 TIPN was significantly higher in the once weekly paclitaxel versus once every 3 weeks docetaxel arm by both physician-rated CTCAE (45% v 29%; P = .02) and PRO-CTCAE (40% v 24%; P = .03). Patients receiving once weekly paclitaxel required more dose reductions because of TIPN (28% v 9%; P < .001) or any cause (39% v 25%; P = .02). CONCLUSION Germline variation did not predict risk of TIPN in Black women receiving (neo)adjuvant once weekly paclitaxel or once every 3 weeks docetaxel. Once weekly paclitaxel was associated with significantly more grade 2-4 TIPN and required more dose reductions than once every 3 weeks docetaxel.
Collapse
Affiliation(s)
| | - Fengmin Zhao
- Dana Farber Cancer Institute - ECOG-ACRIN Biostatistics Center
| | | | - Sofia F Garcia
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center
| | - Fei Shen
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
| | | | - David Cella
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center
| | - Casey Bales
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
| | - Guanglong Jiang
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
| | | | | | | | | | | | - Andrew A. Davis
- Washington University School of Medicine, St. Louis, Missouri
| | | | | | - Melissa A. Simon
- Northwestern University, Robert H. Lurie Comprehensive Cancer Center
| | | | | | - Kathy D. Miller
- Indiana University Melvin and Bren Simon Comprehensive Cancer Center
| | | | | | | |
Collapse
|
11
|
Ma J, Gong Y, Sun X, Liu C, Li X, Sun Y, Yang D, He J, Wang M, Du J, Zhang J, Xu W, Wang T, Chi X, Tang Y, Song J, Wang Y, Ma F, Chen C, Zhang H, Zhan J. Tumor suppressor FRMD3 controls mammary epithelial cell fate determination via notch signaling pathway. SCIENCE ADVANCES 2024; 10:eadk8958. [PMID: 38959315 PMCID: PMC11221522 DOI: 10.1126/sciadv.adk8958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 05/17/2024] [Indexed: 07/05/2024]
Abstract
The luminal-to-basal transition in mammary epithelial cells (MECs) is accompanied by changes in epithelial cell lineage plasticity; however, the underlying mechanism remains elusive. Here, we report that deficiency of Frmd3 inhibits mammary gland lineage development and induces stemness of MECs, subsequently leading to the occurrence of triple-negative breast cancer. Loss of Frmd3 in PyMT mice results in a luminal-to-basal transition phenotype. Single-cell RNA sequencing of MECs indicated that knockout of Frmd3 inhibits the Notch signaling pathway. Mechanistically, FERM domain-containing protein 3 (FRMD3) promotes the degradation of Disheveled-2 by disrupting its interaction with deubiquitinase USP9x. FRMD3 also interrupts the interaction of Disheveled-2 with CK1, FOXK1/2, and NICD and decreases Disheveled-2 phosphorylation and nuclear localization, thereby impairing Notch-dependent luminal epithelial lineage plasticity in MECs. A low level of FRMD3 predicts poor outcomes for breast cancer patients. Together, we demonstrated that FRMD3 is a tumor suppressor that functions as an endogenous activator of the Notch signaling pathway, facilitating the basal-to-luminal transformation in MECs.
Collapse
Affiliation(s)
- Ji Ma
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yuqing Gong
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Xiaoran Sun
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
- Department of Pathology, Peking University Health Science Center, Beijing 100191, China
| | - Cheng Liu
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Xueying Li
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yi Sun
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Decao Yang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Junming He
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Mengyuan Wang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Juan Du
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Jing Zhang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Weizhi Xu
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Tianzhuo Wang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Xiaochun Chi
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yan Tang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Jiagui Song
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Yunling Wang
- Institute of Cardiovascular Research, Peking University Health Science Center, Beijing 100191, China
| | - Fei Ma
- National Cancer Center, State Key Laboratory of Molecular Oncology, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ceshi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Hongquan Zhang
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| | - Jun Zhan
- Program for Cancer and Cell Biology, Department of Human Anatomy, Histology, and Embryology, School of Basic Medical Sciences, and Peking University International Cancer Institute, State Key Laboratory of Molecular Oncology, Peking University Health Science Center, Beijing 100191, China
| |
Collapse
|
12
|
Inampudi P, Yadlapalli DC, Gullipalli M. Clinicopathological Profiles of and Patterns of Recurrence in Triple-Negative Breast Cancer Patients at a Cancer Care Center in Southern India. Cureus 2024; 16:e63886. [PMID: 39099998 PMCID: PMC11298067 DOI: 10.7759/cureus.63886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2024] [Indexed: 08/06/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is characterized by the absence of expression of the estrogen receptor and the progesterone receptor by immunohistochemistry and human epidermal growth factor receptor overexpression absence either by immunohistochemistry or absence of amplification by fluorescence in-situ hybridization. TNBCs tend to have rapid growth when compared to other subtypes of breast cancer. TNBC is associated with higher histologic grade and more advanced disease at presentation. TNBC shows aggressive behavior and a high chance of recurrence. AIM The aim was to analyze the clinicopathological profiles of and recurrence patterns in TNBC patients at our institute where most patients are from rural areas. METHODS This retrospective study was done at a tertiary cancer care center in Southern India where most patients come from rural backgrounds. Institutional Ethics Committee approval was obtained before the study. Case files of all breast cancer patients registered and treated at our center from 2014 to 2019 were retrieved from the medical record department and reviewed. Data from patients diagnosed with triple-negative breast cancer were identified and analyzed. RESULTS Among the 841 breast cancer patients registered in our study, 150 (17.8%) were diagnosed with TNBC. The median age of diagnosis was 47 years. The majority of the patients, 89 (59.3%) presented with T2 tumors, and lymph node involvement was observed in 88 (58.6%) cases. Patient distribution based on cancer stage revealed that 77 (51.3%) had early-stage breast cancer (EBC), 70 (46.6%) had locally advanced breast cancer (LABC), and only three patients were categorized as having metastatic breast cancer (MBC). Modified radical mastectomy (MRM) was the preferred surgical approach in 144 (96%) cases, while only four patients underwent breast-conserving surgery (BCS). Adjuvant chemotherapy was administered to 119 (79.3%) patients, with 30 (20%) receiving both neoadjuvant and adjuvant chemotherapy (NACT/ACT). Among those who underwent NACT/ACT, a pathological complete response was observed in five (16.6%) patients out of 30 patients. The median duration of follow-up was 32.8 months. Among all patients, 36 (24%) experienced recurrence, with seven (19.4%) having local recurrence, 24 (66.6%) developing distant metastases, two patients experiencing both local and distant recurrence, and three patients developing contralateral breast cancer. Additionally, three patients experienced a second primary cancer. The most common sites of metastases were the lungs (14), followed by the bone (seven), the liver (four), and the brain (four). Recurrence rates were notably high within the first one to three years post-diagnosis. The median disease-free survival (DFS) of TNBC patients was estimated to be 65.6 months with no statistically significant difference (p=0.174) between EBC and LABC patients. CONCLUSION TNBC is known for its heterogeneity. While it is often regarded as being more responsive to chemotherapy compared to other subtypes of breast cancer, TNBCs tend to behave aggressively, basically due to the underlying aggressive tumor biology. Though there are many treatment options for different subtypes of breast cancer, therapeutic modalities are limited for TNBCs. Aggressive tumor biology with limited treatment options denotes a gap in the development of novel strategies to improve outcomes in this subset of breast cancer patients.
Collapse
Affiliation(s)
- Prudhvi Inampudi
- Medical Oncology, Ganni Subbulakshmi Garu Medical College, Rajahmundry, IND
| | | | | |
Collapse
|
13
|
Čeprnja T, Tomić S, Perić Balja M, Marušić Z, Blažićević V, Spagnoli GC, Juretić A, Čapkun V, Vuger AT, Pogorelić Z, Mrklić I. Prognostic Value of "Basal-like" Morphology, Tumor-Infiltrating Lymphocytes and Multi-MAGE-A Expression in Triple-Negative Breast Cancer. Int J Mol Sci 2024; 25:4513. [PMID: 38674098 PMCID: PMC11050590 DOI: 10.3390/ijms25084513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 04/16/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
"Basal-like" (BL) morphology and the expression of cancer testis antigens (CTA) in breast cancer still have unclear prognostic significance. The aim of our research was to explore correlations of the morphological characteristics and tumor microenvironment in triple-negative breast carcinomas (TNBCs) with multi-MAGE-A CTA expression and to determine their prognostic significance. Clinical records of breast cancer patients who underwent surgery between January 2017 and December 2018 in four major Croatian clinical centers were analyzed. A total of 97 non-metastatic TNBCs with available tissue samples and treatment information were identified. Cancer tissue sections were additionally stained with programmed death-ligand 1 (PD-L1) Ventana (SP142) and multi-MAGE-A (mAb 57B). BL morphology was detected in 47 (49%) TNBCs and was associated with a higher Ki-67 proliferation index and histologic grade. Expression of multi-MAGE-A was observed in 77 (79%) TNBCs and was significantly associated with BL morphology. Lymphocyte-predominant breast cancer (LPBC) status was detected in 11 cases (11.3%) and significantly correlated with the Ki-67 proliferation index, increased number of intratumoral lymphocytes (itTIL), and PD-L1 expression. No impact of BL morphology, multi-MAGE-A expression, histologic type, or LPBC status on disease-free survival was observed. Our data suggest that tumor morphology could help identify patients with potential benefits from CTA-targeting immunotherapy.
Collapse
Affiliation(s)
- Toni Čeprnja
- Department of Pathology, Forensic Medicine and Cytology, University Hospital of Split, 21000 Split, Croatia; (T.Č.); (S.T.); (I.M.)
| | - Snježana Tomić
- Department of Pathology, Forensic Medicine and Cytology, University Hospital of Split, 21000 Split, Croatia; (T.Č.); (S.T.); (I.M.)
- Department of Pathology, School of Medicine, University of Split, 21000 Split, Croatia
| | - Melita Perić Balja
- Department of Pathology, University Hospital Center “Sestre Milosrdnice”, 10000 Zagreb, Croatia
| | - Zlatko Marušić
- Department of Pathology, Zagreb University Hospital Center, 10000 Zagreb, Croatia
| | | | | | - Antonio Juretić
- Department of Oncology, University Hospital Dubrava, School of Medicine, University of Zagreb, 10000 Zagreb, Croatia;
| | - Vesna Čapkun
- Department of Nuclear Medicine, University Hospital of Split, 21000 Split, Croatia
| | - Ana Tečić Vuger
- Department of Oncology, University Hospital “Sestre Milosrdnice”, 10000 Zagreb, Croatia;
| | - Zenon Pogorelić
- Department of Pediatric Surgery, University Hospital of Split, 21000 Split, Croatia
- Department of Surgery, School of Medicine, University of Split, 21000 Split, Croatia
| | - Ivana Mrklić
- Department of Pathology, Forensic Medicine and Cytology, University Hospital of Split, 21000 Split, Croatia; (T.Č.); (S.T.); (I.M.)
- Department of Pathology, School of Medicine, University of Split, 21000 Split, Croatia
| |
Collapse
|
14
|
Fan T, Huang Y, Liu Z, Huang J, Ke B, Rong Y, Qiu H, Zhang B. Unveiling the Mechanism of the ChaiShao Shugan Formula Against Triple-Negative Breast Cancer. Drug Des Devel Ther 2024; 18:1115-1131. [PMID: 38618280 PMCID: PMC11016267 DOI: 10.2147/dddt.s394287] [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: 08/16/2023] [Accepted: 03/25/2024] [Indexed: 04/16/2024] Open
Abstract
Background The ChaiShao Shugan Formula (CSSGF) is a traditional Chinese medicine formula with recently identified therapeutic value in triple-negative breast cancer (TNBC). This study aimed to elucidate the underlying mechanism of CSSGF in TNBC treatment. Methods TNBC targets were analyzed using R and data were from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. The major ingredients and related protein targets of CSSGF were explored via the Traditional Chinese Medicine Systems Pharmacology database, and an ingredient-target network was constructed via Cytoscape to identify hub genes. The STRING database was used to construct the PPI network. GO and KEGG enrichment analyses were performed via R to obtain the main targets. The online tool Kaplan‒Meier plotter was used to identify the prognostic genes. Molecular docking was applied to the core target genes and active ingredients. MDA-MB-231 and MCF-7 cell lines were used to verify the efficacy of the various drugs. Results A total of 4562 genes were screened as TNBC target genes. The PPI network consisted of 89 nodes and 845 edges. Our study indicated that quercetin, beta-sitosterol, luteolin and catechin might be the core ingredients of CSSGF, and EGFR and c-Myc might be the latent therapeutic targets of CSSGF in the treatment of TNBC. GO and KEGG analyses indicated that the anticancer effect of CSSGF on TNBC was mainly associated with DNA binding, transcription factor binding, and other biological processes. The related signaling pathways mainly involved the TNF-a, IL-17, and apoptosis pathways. The molecular docking data indicated that quercetin, beta-sitosterol, luteolin, and catechin had high affinity for EGFR, JUN, Caspase-3 and ESR1, respectively. In vitro, we found that CSSGF could suppress the expression of c-Myc or promote the expression of EGFR. In addition, we found that quercetin downregulates c-Myc expression in two BC cell lines. Conclusion This study revealed the effective ingredients and latent molecular mechanism of action of CSSGF against TNBC and confirmed that quercetin could target c-Myc to induce anti-BC effects.
Collapse
Affiliation(s)
- Teng Fan
- TCM&VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Yuanyuan Huang
- TCM&VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Zeyu Liu
- TCM&VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Jinsheng Huang
- TCM&VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Bin Ke
- TCM&VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Yuming Rong
- TCM&VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Huijuan Qiu
- TCM&VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| | - Bei Zhang
- TCM&VIP Department, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
- Integrated Traditional Chinese and Western Medicine Research Center, Sun Yat-sen University Cancer Center, Guangzhou, People’s Republic of China
| |
Collapse
|
15
|
Alkhayyat R, Abbas A, Quinn CM, Rakha EA. Tumour 63 protein (p63) in breast pathology: biology, immunohistochemistry, diagnostic applications, and pitfalls. Histopathology 2024; 84:723-741. [PMID: 38012539 DOI: 10.1111/his.15101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/16/2023] [Accepted: 11/06/2023] [Indexed: 11/29/2023]
Abstract
Tumour protein 63 (p63) is a transcription factor of the p53 gene family, encoded by the TP63 gene located at chromosome 3q28, which regulates the activity of genes involved in growth and development of the ectoderm and derived tissues. p63 protein is normally expressed in the nuclei of the basal cell layer of glandular organs, including breast, in squamous epithelium and in urothelium. p63 immunohistochemical (IHC) staining has several applications in diagnostic breast pathology. It is commonly used to demonstrate myoepithelial cells at the epithelial stromal interface to differentiate benign and in situ lesions from invasive carcinoma and to characterize and classify papillary lesions including the distinction of breast intraduct papilloma from skin hidradenoma. p63 IHC is also used to identify and profile lesions showing myoepithelial cell and/or squamous differentiation, e.g. adenomyoepithelioma, salivary gland-like tumours including adenoid cystic carcinoma, and metaplastic breast carcinoma including low-grade adenosquamous carcinoma. This article reviews the applications of p63 IHC in diagnostic breast pathology and outlines a practical approach to the diagnosis and characterization of breast lesions through the identification of normal and abnormal p63 protein expression. The biology of p63, the range of available antibodies with emphasis on staining specificity and sensitivity, and pitfalls in interpretation are also discussed. The TP63 gene in humans, which shows a specific genomic structure, resulting in either TAp63 (p63) isoform or ΔNp63 (p40) isoform. As illustrated in the figure, both isoforms contain a DNA-binding domain (Orange box) and an oligomerization domain (Grey box). TAp63 contains an N-terminal transactivation (TA) domain (Green box), while ΔNp63 has an alternative terminus (Yellow box). Antibodies against conventional pan-p63 (TP63) bind to the DNA binding domain common to both isoforms (TAp63 and p40) and does not distinguish between them. Antibodies against TAp63 bind to the N-terminal TA domain, while antibodies specific to ΔNp63 (p40) bind to the alternative terminus. Each isoform has variant isotypes (α, β, γ, δ, and ε).
Collapse
Affiliation(s)
- Rabab Alkhayyat
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospitals, Nottingham, UK
- Department of Pathology, Salmaniya Medical Complex, Government Hospitals, Manama, Kingdom of Bahrain
| | - Areeg Abbas
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospitals, Nottingham, UK
| | - Cecily M Quinn
- Irish National Breast Screening Program, Department of Histopathology, St. Vincent's University Hospital, Dublin, School of Medicine, University College Dublin, Dublin, Ireland
| | - Emad A Rakha
- Department of Histopathology, Nottingham University Hospitals NHS Trust, Nottingham City Hospitals, Nottingham, UK
- Academic Unit for Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
- Department of Pathology, Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
| |
Collapse
|
16
|
Hoshi N, Uemura T, Tachibana K, Abe S, Murakami-Nishimagi Y, Okano M, Noda M, Saito K, Kono K, Ohtake T, Waguri S. Endosomal protein expression of γ1-adaptin is associated with tumor growth activity and relapse-free survival in breast cancer. Breast Cancer 2024; 31:305-316. [PMID: 38265632 PMCID: PMC10902087 DOI: 10.1007/s12282-023-01539-1] [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: 10/20/2022] [Accepted: 12/20/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND γ1-Adaptin is a subunit of adaptor protein complex-1 (AP-1), which regulates intracellular transport between the trans-Golgi network (TGN) and endosomes. Since expression levels of AP-1 subunits have been reported to be associated with cell proliferation and cancer malignancy, we investigated the relationships between the immunohistochemical expression of γ1-adaptin and both clinicopathological factors and relapse-free survival (RFS) in breast cancer tissue. MATERIALS AND METHODS SK-BR-3 cell line depleted of γ1-adaptin was used for cell proliferation, migration, and invasion assay. Intracellular localization of γ1-adaptin was examined with immunohistochemistry (IHC) using an antibody against γ1-adaptin, and with double immunohistofluorescence (IHF) microscopy using markers for the TGN and endosome. γ1-Adaptin intensities in IHC samples from 199 primary breast cancer patients were quantified and assessed in relation to clinicopathological factors and RFS. RESULTS Cell growth, migration, and invasion of SK-BR-3 cells were significantly suppressed by the depletion of γ1-adaptin. Although the staining patterns in the cancer tissues varied among cases by IHC, double IHF demonstrated that γ1-adaptin was mainly localized in EEA1-positive endosomes, but not in the TGN. γ1-Adaptin intensity was significantly higher in the tumor regions than in non-tumor regions. It was also higher in patients with Ki-67 (high), ER (-), PgR (-), and HER2 (+). Among subtypes of breast cancer, γ1-adaptin intensity was higher in HER2 than in luminal A or luminal B. The results of the survival analysis indicated that high γ1-adaptin intensity was significantly associated with worse RFS, and this association was also observed in group with ER (+), PgR (+), HER2 (-), Ki-67 (high), or luminal B. In addition, the Cox proportional hazards model showed that high γ1-adaptin intensity was an independent prognostic factor. CONCLUSION These results suggest that the endosomal expression of γ1-adaptin is positively correlated with breast cancer malignancy and could be a novel prognostic marker.
Collapse
Affiliation(s)
- Nobuhiro Hoshi
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takefumi Uemura
- Department of Anatomy and Histology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kazunoshin Tachibana
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Sadahiko Abe
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yuko Murakami-Nishimagi
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Maiko Okano
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Masaru Noda
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Katsuharu Saito
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tohru Ohtake
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Satoshi Waguri
- Department of Anatomy and Histology, Fukushima Medical University School of Medicine, Fukushima, Japan.
| |
Collapse
|
17
|
Piedra-Delgado L, Chambergo-Michilot D, Morante Z, Fairen C, Jerves-Coello F, Luque-Benavides R, Casas F, Bustamante E, Razuri-Bustamante C, Torres-Roman JS, Fuentes H, Gomez H, Narvaez-Rojas A, De la Cruz-Ku G, Araujo J. Survival according to the site of metastasis in triple-negative breast cancer patients: The Peruvian experience. PLoS One 2024; 19:e0293833. [PMID: 38300959 PMCID: PMC10833533 DOI: 10.1371/journal.pone.0293833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 10/19/2023] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Evidence regarding differences in survival associated with the site of metastasis in triple-negative breast cancer (TNBC) remains limited. Our aim was to analyze the overall survival (OS), distant relapse free survival (DRFS), and survival since the diagnosis of the relapse (MS), according to the side of metastasis. METHODS This was a retrospective study of TNBC patients with distant metastases at the Instituto Nacional de Enfermedades Neoplasicas (Lima, Peru) from 2000 to 2014. Prognostic factors were determined by multivariate Cox regression analysis. RESULTS In total, 309 patients were included. Regarding the type of metastasis, visceral metastasis accounted for 41% and the lung was the most frequent first site of metastasis (33.3%). With a median follow-up of 10.2 years, the 5-year DRFS and OS were 10% and 26%, respectively. N staging (N2-N3 vs. N0, HR = 1.49, 95%CI: 1.04-2.14), metastasis in visceral sites (vs. bone; HR = 1.55, 95%CI: 0.94-2.56), the central nervous system (vs. bone; HR = 1.88, 95% CI: 1.10-3.22), and multiple sites (vs. bone; HR = 2.55, 95%CI:1.53-4.25) were prognostic factors of OS whereas multiple metastasis (HR = 2.30, 95% CI: 1.42-3.72) was a predictor of MS. In terms of DRFS, there were no differences according to metastasis type or solid organ. CONCLUSION TNBC patients with multiple metastasis and CNS metastasis have an increased risk of death compared to those with bone metastasis in terms of OS and MS.
Collapse
Affiliation(s)
| | | | - Zaida Morante
- Departamento de Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas, Lima, Perú
| | - Carlos Fairen
- Boston Medical Center, Boston, Massachusetts, United States of America
| | | | | | - Fresia Casas
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | | | | | - Hugo Fuentes
- Departamento de Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas, Lima, Perú
| | - Henry Gomez
- Departamento de Oncología Médica, Instituto Nacional de Enfermedades Neoplásicas, Lima, Perú
| | - Alexis Narvaez-Rojas
- Department of Surgical Oncology, Miller School of Medicine, University Of Miami, Miami, Florida, United States of America
| | | | - Jhajaira Araujo
- Escuela Profesional de Medicina Humana, Universidad Privada San Juan Bautista, Chorrillos, Lima, Peru
| |
Collapse
|
18
|
Wilk SM, Lee K, Gajda AM, Haloul M, Macias V, Wiley EL, Chen Z, Liu X, Wang X, Sverdlov M, Hoskins KF, Emrah E. Multiplex Imaging Reveals Novel Subcellular, Microenvironmental, and Racial Patterns of MRTFA/B Activation in Invasive Breast Cancers and Metastases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.573909. [PMID: 38260321 PMCID: PMC10802460 DOI: 10.1101/2024.01.03.573909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Breast cancer progression and metastasis involve the action of multiple transcription factors in tumors and in the cells of the tumor microenvironment (TME) and understanding how these transcription factors are coordinated can guide novel therapeutic strategies. Myocardin related transcription factors A and B (MRTFA/B) are two related transcription factors that redundantly control cancer cell invasion and metastasis in mouse models of breast cancer, but their roles in human cancer are incompletely understood. Here, we used a combination of multiplexed immunofluorescence and bioinformatics analyses to show that MRTFA/B are concurrently activated in tumor cells, but they show distinct patterns of expression across different histological subtypes and in the TME. Importantly, MRTFA expression was elevated in metastatic tumors of African American patients, who disproportionately die from breast cancer. Interestingly, in contrast to publicly available mRNA expression data, MRTFA was similarly expressed across estrogen receptor (ER) positive and negative breast tumors, while MRTFB expression was highest in ER+ breast tumors. Furthermore, MRTFA was specifically expressed in the perivascular antigen presenting cells (APCs) and its expression correlated with the expression of the immune checkpoint protein V-set immunoregulatory receptor (VSIR). These results provide unique insights into how MRTFA and MRTFB can promote metastasis in human cancer, into the racial disparities of their expression patterns, and their function within the complex breast cancer TME.
Collapse
Affiliation(s)
- Stephanie M. Wilk
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL
| | - Kihak Lee
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL
| | - Alexa M. Gajda
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL
| | - Mohamed Haloul
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL
| | - Virgilia Macias
- Department of Pathology, University of Illinois Chicago, Chicago, IL
| | | | - Zhengjia Chen
- Division of Epidemiology and Biostatistics, School of Public Health, University of Illinois Chicago, Chicago, IL
- Biostatistics Shared Resource, University of Illinois Cancer Center, Chicago, IL
| | - Xinyi Liu
- Department of Pharmacology & Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL
| | - Xiaowei Wang
- Department of Pharmacology & Regenerative Medicine, College of Medicine, University of Illinois Chicago, Chicago, IL
| | - Maria Sverdlov
- Research Histology Core, Research Resources Center, College of Medicine, University of Illinois Chicago, Chicago, IL
| | - Kent F. Hoskins
- Division of Hematology/Oncology, College of Medicine, University of Illinois Chicago, Chicago, IL
| | - Ekrem Emrah
- Department of Physiology and Biophysics, College of Medicine, University of Illinois Chicago, Chicago, IL
| |
Collapse
|
19
|
Abstract
Osteopontin (OPN) is a heavily post-translationally modified protein with a molecular weight of 44-70 kDa, depending on the degree of glycosylation. OPN is involved in various biological processes, including bone remodeling, immune response, cell adhesion, migration, and survival. It is essential for controlling osteoclast and osteoblast activity for maintaining bone mass and bone strength. Additionally, OPN has been linked to cardiovascular, inflammatory illnesses, as well as the onset and progression of cancer. OPN is a multifunctional protein that can interact with a variety of cell surface receptors, such as integrins, CD44, the urokinase-type plasminogen activator receptor (uPAR), as well as extracellular matrix (ECM) components (e.g. collagen and hydroxyapatite). These interactions contribute to its wide range of biological functions in general and has significant implications for bone biology, immunology and cancer, specifically. In this chapter, we summarize the structure of OPN with a focus on its molecular mechanisms of action in various cancers.
Collapse
Affiliation(s)
- Alpana Kumari
- Department of Optometry, University Institute of Allied Health Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Dharambir Kashyap
- Department of Medicine, The Brown Centre for Immunotherapy, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Vivek Kumar Garg
- Department of Medical Lab Technology, University Institute of Allied Health Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India.
| |
Collapse
|
20
|
Al Husban H, Al Rabadi A, Odeh AH, Abu Rumman K, Alkhawaldeh F, Noures H, Abo Ashoor M, Abu Rumman A, Atmeh M, Bawaneh M. Clinical Characteristics and Management of Triple-Negative Breast Cancer (TNBC) in Jordan: A Retrospective Analysis. Cureus 2024; 16:e53053. [PMID: 38410339 PMCID: PMC10896140 DOI: 10.7759/cureus.53053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2024] [Indexed: 02/28/2024] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) is known for its aggressive nature and poor prognosis. Despite its responsiveness to chemotherapy, TNBC presents challenges in terms of survival, recurrence, and mortality rates, particularly in diverse populations. Limited research in the Middle East hampers comprehensive understanding and tailored management. Methods A retrospective study at the King Hussein Medical Center in Jordan between the period 2009 to 2023 explored TNBC patients (n=110) who underwent adjuvant chemotherapy after local excision or modified radical mastectomy (MRM). Data encompassed demographics, clinical variables, and operative details. Statistical analysis employed Wilcoxon and chi-squared tests, examining mortality risks and associations between variables. Results Among 110 TNBC patients (mean age 52), 84% underwent MRM, 16% wide local excision and axillary clearance (WLE&AC). Lymphovascular invasion (LVI) was observed in 41%, linked to higher lymph node positivity. Neoadjuvant therapy preceded MRM in 25% of cases. While 75% had grade III tumors, the prevalence of invasive ductal carcinoma was 85%. Conclusions This study contributes crucial insights into TNBC characteristics and management in Jordan. Despite limitations such as retrospective design and sample size, the findings underscore the need for tailored interventions in TNBC patients, emphasizing the importance of neoadjuvant therapy and vigilant consideration of LVI status in treatment planning. Future longitudinal research should delve into disease progression and treatment outcomes in diverse populations, facilitating optimized TNBC management strategies.
Collapse
Affiliation(s)
- Hussein Al Husban
- Department of General Surgery, Jordanian Royal Medical Services, Amman, JOR
| | - Anas Al Rabadi
- Department of General Surgery, Jordanian Royal Medical Services, Amman, JOR
| | - Ala H Odeh
- Department of General Surgery, Jordanian Royal Medical Services, Amman, JOR
| | - Kahled Abu Rumman
- Department of General Surgery, Jordanian Royal Medical Services, Amman, JOR
| | - Feras Alkhawaldeh
- Department of General Surgery, Jordanian Royal Medical Services, Amman, JOR
| | - Haneen Noures
- Department of Pathology, Jordanian Royal Medical Services, Amman, JOR
| | - Mohammad Abo Ashoor
- Department of Clinical Oncology, Jordanian Royal Medical Services, Amman, JOR
| | - Anas Abu Rumman
- Department of General Surgery, Jordanian Royal Medical Services, Amman, JOR
| | - Mousa Atmeh
- Department of Clinical Oncology, Jordanian Royal Medical Services, Amman, JOR
| | - Mohannad Bawaneh
- Department of General Surgery, Jordanian Royal Medical Services, Amman, JOR
| |
Collapse
|
21
|
Kuburich NA, den Hollander P, Castaneda M, Pietilä M, Tang X, Batra H, Martínez-Peña F, Visal TH, Zhou T, Demestichas BR, Dontula RV, Liu JY, Maddela JJ, Padmanabhan RS, Phi LTH, Rosolen MJ, Sabapathy T, Kumar D, Giancotti FG, Lairson LL, Raso MG, Soundararajan R, Mani SA. Stabilizing vimentin phosphorylation inhibits stem-like cell properties and metastasis of hybrid epithelial/mesenchymal carcinomas. Cell Rep 2023; 42:113470. [PMID: 37979166 PMCID: PMC11062250 DOI: 10.1016/j.celrep.2023.113470] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 09/01/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023] Open
Abstract
Epithelial-mesenchymal transition (EMT) empowers epithelial cells with mesenchymal and stem-like attributes, facilitating metastasis, a leading cause of cancer-related mortality. Hybrid epithelial-mesenchymal (E/M) cells, retaining both epithelial and mesenchymal traits, exhibit heightened metastatic potential and stemness. The mesenchymal intermediate filament, vimentin, is upregulated during EMT, enhancing the resilience and invasiveness of carcinoma cells. The phosphorylation of vimentin is critical to its structure and function. Here, we identify that stabilizing vimentin phosphorylation at serine 56 induces multinucleation, specifically in hybrid E/M cells with stemness properties but not epithelial or mesenchymal cells. Cancer stem-like cells are especially susceptible to vimentin-induced multinucleation relative to differentiated cells, leading to a reduction in self-renewal and stemness. As a result, vimentin-induced multinucleation leads to sustained inhibition of stemness properties, tumor initiation, and metastasis. These observations indicate that a single, targetable phosphorylation event in vimentin is critical for stemness and metastasis in carcinomas with hybrid E/M properties.
Collapse
Affiliation(s)
- Nick A Kuburich
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Petra den Hollander
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Maria Castaneda
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mika Pietilä
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; The Janssen Pharmaceutical Companies of Johnson & Johnson, Espoo, Uusimaa, Finland
| | - Ximing Tang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Harsh Batra
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | - Tanvi H Visal
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tieling Zhou
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Breanna R Demestichas
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Ritesh V Dontula
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jojo Y Liu
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Joanna Joyce Maddela
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Reethi S Padmanabhan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Lan Thi Hanh Phi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matthew J Rosolen
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Thiru Sabapathy
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA
| | - Dhiraj Kumar
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Cancer Metastasis Initiative, Herbert Irving Comprehensive Cancer Center, Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Filippo G Giancotti
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA; Cancer Metastasis Initiative, Herbert Irving Comprehensive Cancer Center, Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Luke L Lairson
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Rama Soundararajan
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sendurai A Mani
- Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA; Legorreta Cancer Center, The Warren Alpert Medical School, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
22
|
Moon HR, Surianarayanan N, Singh T, Han B. Microphysiological systems as reliable drug discovery and evaluation tools: Evolution from innovation to maturity. BIOMICROFLUIDICS 2023; 17:061504. [PMID: 38162229 PMCID: PMC10756708 DOI: 10.1063/5.0179444] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Microphysiological systems (MPSs), also known as organ-on-chip or disease-on-chip, have recently emerged to reconstitute the in vivo cellular microenvironment of various organs and diseases on in vitro platforms. These microfluidics-based platforms are developed to provide reliable drug discovery and regulatory evaluation testbeds. Despite recent emergences and advances of various MPS platforms, their adoption of drug discovery and evaluation processes still lags. This delay is mainly due to a lack of rigorous standards with reproducibility and reliability, and practical difficulties to be adopted in pharmaceutical research and industry settings. This review discusses the current and potential use of MPS platforms in drug discovery processes while considering the context of several key steps during drug discovery processes, including target identification and validation, preclinical evaluation, and clinical trials. Opportunities and challenges are also discussed for the broader dissemination and adoption of MPSs in various drug discovery and regulatory evaluation steps. Addressing these challenges will transform long and expensive drug discovery and evaluation processes into more efficient discovery, screening, and approval of innovative drugs.
Collapse
Affiliation(s)
- Hye-Ran Moon
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | | | - Tarun Singh
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47907, USA
| | - Bumsoo Han
- Author to whom correspondence should be addressed:. Tel: +1-765-494-5626
| |
Collapse
|
23
|
Lai S, Liang F, Zhang W, Zhao Y, Li J, Zhao Y, Xu Y, Ding W, Zhan J, Zhen X, Yang R. Evaluation of molecular receptors status in breast cancer using an mpMRI-based feature fusion radiomics model: mimicking radiologists' diagnosis. Front Oncol 2023; 13:1219071. [PMID: 38074664 PMCID: PMC10698551 DOI: 10.3389/fonc.2023.1219071] [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: 05/08/2023] [Accepted: 10/31/2023] [Indexed: 08/31/2024] Open
Abstract
OBJECTIVE To investigate the performance of a novel feature fusion radiomics (RFF) model that incorporates features from multiparametric MRIs (mpMRI) in distinguishing different statuses of molecular receptors in breast cancer (BC) preoperatively. METHODS 460 patients with 466 pathology-confirmed BCs who underwent breast mpMRI at 1.5T in our center were retrospectively included hormone receptor (HR) positive (HR+) (n=336) and HR negative (HR-) (n=130). The HR- patients were further categorized into human epidermal growth factor receptor 2 (HER-2) enriched BC (HEBC) (n=76) and triple negative BC (TNBC) (n=54). All lesions were divided into a training/validation cohort (n=337) and a test cohort (n=129). Volumes of interest (VOIs) delineation, followed by radiomics feature extraction, was performed on T2WI, DWI600 (b=600 s/mm2), DWI800 (b=800 s/mm2), ADC map, and DCE1-6 (six continuous DCE-MRI) images of each lesion. Simulating a radiologist's work pattern, 150 classification base models were constructed and analyzed to determine the top four optimum sequences for classifying HR+ vs. HR-, TNBC vs. HEBC, TNBC vs. non-TNBC in a random selected training cohort (n=337). Building upon these findings, the optimal single sequence models (Rss) and combined sequences models (RFF) were developed. The AUC, sensitivity, accuracy and specificity of each model for subtype differentiation were evaluated. The paired samples Wilcoxon signed rank test was used for performance comparison. RESULTS During the three classification tasks, the optimal single sequence for classifying HR+ vs. HR- was DWI600, while the ADC map, derived from DWI800 performed the best in distinguishing TNBC vs. HEBC, as well as identifying TNBC vs. non-TNBC, with corresponding training AUC values of 0.787, 0.788, and 0.809, respectively. Furthermore, the integration of the top four sequences in RFF models yielded improved performance, achieving AUC values of 0.809, 0.805 and 0.847, respectively. Consistent results was observed in both the training/validation and testing cohorts, with AUC values of 0.778, 0.787, 0.818 and 0.726, 0.773, 0.773, respectively (all p < 0.05 except HR+ vs. HR-). CONCLUSION The RFF model, integrating mpMRI radiomics features, demonstrated promising ability to mimic radiologists' diagnosis for preoperative identification of molecular receptors of BC.
Collapse
Affiliation(s)
- Shengsheng Lai
- School of Medical Equipment, Guangdong Food and Drug Vocational College, Guangzhou, Guangdong, China
| | - Fangrong Liang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Wanli Zhang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Yue Zhao
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Jiamin Li
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Yandong Zhao
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Yongzhou Xu
- Department of Clinical & Technique Support, Philips Healthcare, Guangzhou, Guangdong, China
| | - Wenshuang Ding
- Department of Pathology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| | - Jie Zhan
- Department of Radiology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xin Zhen
- School of Biomedical Engineering, Southern Medical University, Guangzhou, Guangdong, China
| | - Ruimeng Yang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Department of Radiology, Guangzhou First People’s Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
24
|
Masci D, Naro C, Puxeddu M, Urbani A, Sette C, La Regina G, Silvestri R. Recent Advances in Drug Discovery for Triple-Negative Breast Cancer Treatment. Molecules 2023; 28:7513. [PMID: 38005235 PMCID: PMC10672974 DOI: 10.3390/molecules28227513] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is one of the most heterogeneous and aggressive breast cancer subtypes with a high risk of death on recurrence. To date, TNBC is very difficult to treat due to the lack of an effective targeted therapy. However, recent advances in the molecular characterization of TNBC are encouraging the development of novel drugs and therapeutic combinations for its therapeutic management. In the present review, we will provide an overview of the currently available standard therapies and new emerging therapeutic strategies against TNBC, highlighting the promises that newly developed small molecules, repositioned drugs, and combination therapies have of improving treatment efficacy against these tumors.
Collapse
Affiliation(s)
- Domiziana Masci
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (D.M.); (A.U.)
| | - Chiara Naro
- Department of Neurosciences, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (C.N.); (C.S.)
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Michela Puxeddu
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (D.M.); (A.U.)
| | - Claudio Sette
- Department of Neurosciences, Section of Human Anatomy, Catholic University of the Sacred Heart, Largo Francesco Vito 1, 00168 Rome, Italy; (C.N.); (C.S.)
- GSTeP-Organoids Research Core Facility, Fondazione Policlinico Universitario A. Gemelli, IRCCS, 00168 Rome, Italy
| | - Giuseppe La Regina
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| | - Romano Silvestri
- Laboratory Affiliated to Istituto Pasteur Italia—Fondazione Cenci Bolognetti, Department of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy; (M.P.); (G.L.R.)
| |
Collapse
|
25
|
Tariq M, Richard V, Kerin MJ. MicroRNAs as Molecular Biomarkers for the Characterization of Basal-like Breast Tumor Subtype. Biomedicines 2023; 11:3007. [PMID: 38002007 PMCID: PMC10669494 DOI: 10.3390/biomedicines11113007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/03/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Breast cancer is a heterogeneous disease highlighted by the presence of multiple tumor variants and the basal-like breast cancer (BLBC) is considered to be the most aggressive variant with limited therapeutics and a poor prognosis. Though the absence of detectable protein and hormonal receptors as biomarkers hinders early detection, the integration of genomic and transcriptomic profiling led to the identification of additional variants in BLBC. The high-throughput analysis of tissue-specific micro-ribonucleic acids (microRNAs/miRNAs) that are deemed to have a significant role in the development of breast cancer also displayed distinct expression profiles in each subtype of breast cancer and thus emerged to be a robust approach for the precise characterization of the BLBC subtypes. The classification schematic of breast cancer is still a fluid entity that continues to evolve alongside technological advancement, and the transcriptomic profiling of tissue-specific microRNAs is projected to aid in the substratification and diagnosis of the BLBC tumor subtype. In this review, we summarize the current knowledge on breast tumor classification, aim to collect comprehensive evidence based on the microRNA expression profiles, and explore their potential as prospective biomarkers of BLBC.
Collapse
Affiliation(s)
| | - Vinitha Richard
- Discipline of Surgery, Lambe Institute for Translational Research, H91 TK33 Galway, Ireland;
| | - Michael J. Kerin
- Discipline of Surgery, Lambe Institute for Translational Research, H91 TK33 Galway, Ireland;
| |
Collapse
|
26
|
Shakya R, Byun MR, Joo SH, Chun KS, Choi JS. Domperidone Exerts Antitumor Activity in Triple-Negative Breast Cancer Cells by Modulating Reactive Oxygen Species and JAK/STAT3 Signaling. Biomol Ther (Seoul) 2023; 31:692-699. [PMID: 37899746 PMCID: PMC10616512 DOI: 10.4062/biomolther.2023.173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/08/2023] [Accepted: 10/10/2023] [Indexed: 10/31/2023] Open
Abstract
The lack of molecular targets hampers the treatment of triple-negative breast cancer (TNBC). In this study, we determined the cytotoxicity of domperidone, a dopamine D2 receptor (DRD2) antagonist in human TNBC BT-549 and CAL-51 cells. Domperidone inhibited cell growth in a dose- and time-dependent manner. The annexin V/propidium iodide staining showed that domperidone induced apoptosis. The domperidone-induced apoptosis was accompanied by the generation of mitochondrial superoxide and the down-regulation of cyclins and CDKs. The apoptotic effect of domperidone on TNBC cells was prevented by pre-treatment with Mito-TEMPO, a mitochondria-specific antioxidant. The prevention of apoptosis with Mito-TEMPO even at concentrations as low as 100 nM, implies that the generation of mitochondrial ROS mediated the domperidone-induced apoptosis. Immunoblot analysis showed that domperidone-induced apoptosis occurred through the down-regulation of the phosphorylation of JAK2 and STAT3. Moreover, domperidone downregulated the levels of D2-like dopamine receptors including DRD2, regardless of their mRNA levels. Our results support further development of DRD2 antagonists as potential therapeutic strategy treating TNBC.
Collapse
Affiliation(s)
- Rajina Shakya
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Mi Ran Byun
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Sang Hoon Joo
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| | - Kyung-Soo Chun
- College of Pharmacy, Keimyung University, Daegu 42601, Republic of Korea
| | - Joon-Seok Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan 38430, Republic of Korea
| |
Collapse
|
27
|
Al-Kabariti AY, Abbas MA. Progress in the Understanding of Estrogen Receptor Alpha Signaling in Triple-Negative Breast Cancer: Reactivation of Silenced ER-α and Signaling through ER-α36. Mol Cancer Res 2023; 21:1123-1138. [PMID: 37462782 DOI: 10.1158/1541-7786.mcr-23-0321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/21/2023] [Accepted: 07/14/2023] [Indexed: 11/02/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive tumor that accounts for approximately 15% of total breast cancer cases. It is characterized by poor prognosis and high rate of recurrence compared to other types of breast cancer. TNBC has a limited range of treatment options that include chemotherapy, surgery, and radiation due to the absence of estrogen receptor alpha (ER-α) rendering hormonal therapy ineffective. However, possible targets for improving the clinical outcomes in TNBC exist, such as targeting estrogen signaling through membranous ER-α36 and reactivating silenced ER-α. It has been shown that epigenetic drugs such as DNA methyltransferase and histone deacetylase inhibitors can restore the expression of ER-α. This reactivation of ER-α, presents a potential strategy to re-sensitize TNBC to hormonal therapy. Also, this review provides up-to-date information related to the direct involvement of miRNA in regulating the translation of ER-α mRNA. Specific epi-miRNAs can regulate ER-α expression indirectly by post-transcriptional targeting of mRNAs of enzymes that are involved in DNA methylation and histone deacetylation. Furthermore, ER-α36, an alternative splice variant of ER-α66, is highly expressed in ER-negative breast tumors and activates MAPK/ERK pathway, promoting cell proliferation, escaping apoptosis, and enhancing metastasis. In the future, these recent advances may be helpful for researchers working in the field to obtain novel treatment options for TNBC, utilizing epigenetic drugs and epi-miRNAs that regulate ER-α expression. Also, there is some evidence to suggest that drugs that decrease the expression of ER-α36 may be effective in treating TNBC.
Collapse
Affiliation(s)
- Aya Y Al-Kabariti
- Department of Biopharmaceutics and Clinical Pharmacy, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman, Jordan
| | - Manal A Abbas
- Pharmacological and Diagnostic Research Centre, Al-Ahliyya Amman University, Amman, Jordan
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
| |
Collapse
|
28
|
Chen YT, Luo YX, Chan SH, Chiu WY, Yang HW. Dual antibody-aided mesoporous nanoreactor for H 2O 2 self-supplying chemodynamic therapy and checkpoint blockade immunotherapy in triple-negative breast cancer. J Nanobiotechnology 2023; 21:385. [PMID: 37875918 PMCID: PMC10594761 DOI: 10.1186/s12951-023-02154-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 10/11/2023] [Indexed: 10/26/2023] Open
Abstract
Triple-negative breast cancer (TNBC) represents a formidable challenge due to the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression, rendering it unresponsive to conventional hormonal and targeted therapies. This study introduces the development of mesoporous nanoreactors (NRs), specifically mPDA@CuO2 NRs, as acid-triggered agents capable of self-supplying H2O2 for chemodynamic therapy (CDT). To enhance therapeutic efficacy, these NRs were further modified with immune checkpoint antagonists, specifically anti-PD-L1 and anti-CD24 antibodies, resulting in the formation of dual antibody-aided mesoporous nanoreactors (dAbPD-L1/CD24-mPDA@CuO2 NRs). These NRs were designed to combine CDT and checkpoint blockade immunotherapy (CBIT) for precise targeting of 4T1 TNBC cells. Remarkably, dAbPD-L1/CD24-mPDA@CuO2 NRs exhibited tumor-targeted CDT triggered by H2O2 and successfully activated immune cells including T cells and macrophages. This integrated approach led to a remarkable inhibition of tumor growth by leveraging the collaborative effects of the therapies. The findings of this study introduce a novel and promising strategy for the integrative and collaborative treatment of refractory cancers, providing valuable insights into addressing the challenges posed by aggressive breast cancer, particularly TNBC.
Collapse
Affiliation(s)
- Ying-Tzu Chen
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
- Department of Neurosurgery, Neuroscience Research Center, Chang Gung Memorial Hospital, Linkou, 33305, Taoyuan, Taiwan
| | - Ying-Xiang Luo
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan
| | - Shih-Hsuan Chan
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, 40402, Taiwan
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 40402, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, 40402, Taiwan
| | - Wen-Yi Chiu
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung, 80424, Taiwan.
- Department of Family Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung, 80284, Taiwan.
| | - Hung-Wei Yang
- Department of Biomedical Engineering, National Cheng Kung University, Tainan, 70101, Taiwan.
- Medical Device Innovation Center, National Cheng Kung University, Tainan, 70101, Taiwan.
| |
Collapse
|
29
|
Zhang Z, Zhang R, Li D. Molecular Biology Mechanisms and Emerging Therapeutics of Triple-Negative Breast Cancer. Biologics 2023; 17:113-128. [PMID: 37767463 PMCID: PMC10520847 DOI: 10.2147/btt.s426392] [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: 07/12/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023]
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer that is conventionally characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2), accounting for approximately 15-20% of all breast cancers. Compared to other molecular phenotypes, TNBC is typically associated with high malignancy and poor prognosis. Cytotoxic agents have been the mainstay of treatment for the past few decades due to the lack of definitive targets and limited therapeutic interventions. However, recent developments have demonstrated that TNBC has peculiar molecular classifications and biomarkers, which provide the possibility of evolving treatment from basic cytotoxic chemotherapy to an expanding domain of targeted therapies. This review presents a framework for understanding the current clinical experience surrounding molecular biology mechanisms in TNBC (Figure 1). Including immunotherapy, polymerase (PARP) and PI3K/AKT pathway inhibitors, antibody-drug conjugates, and androgen receptor (AR) blockade. Additionally, the role of miRNA therapeutics targeting TNBC and potential strategies targeting cancer stem cells (CSCs) are discussed and highlighted. As more and more treatments arise on the horizon, we believe that patients with TNBC will have a new sense of hope.
Collapse
Affiliation(s)
- Zhiying Zhang
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| | - Rui Zhang
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| | - Donghai Li
- Inner Mongolia Medical University, Department of Thyroid Breast Surgery, Affiliated Hospital of Inner Mongolia Medical University, Inner Mongolia, 010050, People’s Republic of China
| |
Collapse
|
30
|
Al-Refai R, Bendari A, Morrar D, Sham S, Kataw L, Garajayev A, Hajiyeva S. Immunohistochemical Staining Characteristics of Low-Grade Invasive Ductal Carcinoma Using the ADH5 Cocktail (CK5/14, P63, and CK7/18): A Potential Interpretative Pitfall. Diagnostics (Basel) 2023; 13:2966. [PMID: 37761331 PMCID: PMC10527570 DOI: 10.3390/diagnostics13182966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/21/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Background: In our practice, the antibody cocktail ADH5 (CK5/14, p63, and CK7/18) helps with diagnostic challenges, such as identifying microinvasion and foci of invasive carcinoma, differentiating atypical ductal hyperplasia from hyperplasia of the usual type, and distinguishing basal phenotypes in triple-negative carcinomas. However, the ADH5 cocktail does have pitfalls and caveats. Methods: We describe our experience with the ADH5 cocktail of antibodies in breast pathology. Institutional knowledge and a literature search form our data sources. Results: We analyzed 44 cases. Four out of a total of 44 cases (9.1%)-two tubular carcinomas and two low-grade invasive breast carcinomas of no special type (ductal) with tubular features-showed an expected pattern of staining for ADH5 with a loss of brown (P63, CK5/14) staining around invasive glands and diffuse red (CK7/18) expression. Forty out of 44 (90.9%) cases showed an unexpected staining pattern (mixture of cytoplasmic brown and red). All 44 cases (100%) showed negative myoepithelial staining around invasive foci when separately stained for P63 and SMMH (Smooth Muscle Myosin Heavy). Conclusions: The unexpected staining pattern of ADH5 in low-grade invasive ductal carcinomas can be challenging to interpret in these lesions with low-grade cytology. The occurrence can cause confusion among users who employ multiplex stains, and it is important for users to be aware of this potential pitfall.
Collapse
Affiliation(s)
- Reham Al-Refai
- Department of Pathology and Laboratory Medicine, Northwell Health Lenox Hill Hospital, New York, NY 10075, USA; (A.B.); (D.M.); (S.S.); (L.K.); (S.H.)
| | - Ahmed Bendari
- Department of Pathology and Laboratory Medicine, Northwell Health Lenox Hill Hospital, New York, NY 10075, USA; (A.B.); (D.M.); (S.S.); (L.K.); (S.H.)
| | - Doaa Morrar
- Department of Pathology and Laboratory Medicine, Northwell Health Lenox Hill Hospital, New York, NY 10075, USA; (A.B.); (D.M.); (S.S.); (L.K.); (S.H.)
| | - Sunder Sham
- Department of Pathology and Laboratory Medicine, Northwell Health Lenox Hill Hospital, New York, NY 10075, USA; (A.B.); (D.M.); (S.S.); (L.K.); (S.H.)
| | - Layth Kataw
- Department of Pathology and Laboratory Medicine, Northwell Health Lenox Hill Hospital, New York, NY 10075, USA; (A.B.); (D.M.); (S.S.); (L.K.); (S.H.)
| | - Azar Garajayev
- Baku Health Center, Azerbaijan Medical University, Baku AZ1022, Azerbaijan;
| | - Sabina Hajiyeva
- Department of Pathology and Laboratory Medicine, Northwell Health Lenox Hill Hospital, New York, NY 10075, USA; (A.B.); (D.M.); (S.S.); (L.K.); (S.H.)
| |
Collapse
|
31
|
Bae K, Jeon YS, Hwangbo Y, Yoo CW, Han N, Feng M. Data-Efficient Computational Pathology Platform for Faster and Cheaper Breast Cancer Subtype Identifications: Development of a Deep Learning Model. JMIR Cancer 2023; 9:e45547. [PMID: 37669090 PMCID: PMC10509735 DOI: 10.2196/45547] [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: 01/15/2023] [Revised: 07/07/2023] [Accepted: 07/21/2023] [Indexed: 09/06/2023] Open
Abstract
BACKGROUND Breast cancer subtyping is a crucial step in determining therapeutic options, but the molecular examination based on immunohistochemical staining is expensive and time-consuming. Deep learning opens up the possibility to predict the subtypes based on the morphological information from hematoxylin and eosin staining, a much cheaper and faster alternative. However, training the predictive model conventionally requires a large number of histology images, which is challenging to collect by a single institute. OBJECTIVE We aimed to develop a data-efficient computational pathology platform, 3DHistoNet, which is capable of learning from z-stacked histology images to accurately predict breast cancer subtypes with a small sample size. METHODS We retrospectively examined 401 cases of patients with primary breast carcinoma diagnosed between 2018 and 2020 at the Department of Pathology, National Cancer Center, South Korea. Pathology slides of the patients with breast carcinoma were prepared according to the standard protocols. Age, gender, histologic grade, hormone receptor (estrogen receptor [ER], progesterone receptor [PR], and androgen receptor [AR]) status, erb-B2 receptor tyrosine kinase 2 (HER2) status, and Ki-67 index were evaluated by reviewing medical charts and pathological records. RESULTS The area under the receiver operating characteristic curve and decision curve were analyzed to evaluate the performance of our 3DHistoNet platform for predicting the ER, PR, AR, HER2, and Ki67 subtype biomarkers with 5-fold cross-validation. We demonstrated that 3DHistoNet can predict all clinically important biomarkers (ER, PR, AR, HER2, and Ki67) with performance exceeding the conventional multiple instance learning models by a considerable margin (area under the receiver operating characteristic curve: 0.75-0.91 vs 0.67-0.8). We further showed that our z-stack histology scanning method can make up for insufficient training data sets without any additional cost incurred. Finally, 3DHistoNet offered an additional capability to generate attention maps that reveal correlations between Ki67 and histomorphological features, which renders the hematoxylin and eosin image in higher fidelity to the pathologist. CONCLUSIONS Our stand-alone, data-efficient pathology platform that can both generate z-stacked images and predict key biomarkers is an appealing tool for breast cancer diagnosis. Its development would encourage morphology-based diagnosis, which is faster, cheaper, and less error-prone compared to the protein quantification method based on immunohistochemical staining.
Collapse
Affiliation(s)
- Kideog Bae
- Healthcare AI Team, Healthcare Platform Center, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Young Seok Jeon
- Institute of Data Science, National University of Singapore, Singapore, Singapore
| | - Yul Hwangbo
- Healthcare AI Team, Healthcare Platform Center, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
- Department of Cancer AI & Digital Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Chong Woo Yoo
- Department of Pathology, National Cancer Center Hospital, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Nayoung Han
- Healthcare AI Team, Healthcare Platform Center, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
- Department of Cancer AI & Digital Health, Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
- Department of Pathology, National Cancer Center Hospital, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Mengling Feng
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| |
Collapse
|
32
|
Tong L, Yu X, Wang S, Chen L, Wu Y. Research Progress on Molecular Subtyping and Modern Treatment of Triple-Negative Breast Cancer. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:647-658. [PMID: 37644916 PMCID: PMC10461741 DOI: 10.2147/bctt.s426121] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 08/15/2023] [Indexed: 08/31/2023]
Abstract
Breast cancer has become the most common malignant tumor worldwide. Triple-negative breast cancer (TNBC) is a type of breast cancer that is negative for estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2). Compared with other molecular subtypes of breast cancer, TNBC is the most aggressive and highly heterogeneous. TNBC is insensitive to endocrine and anti-HER2 therapy, and chemotherapy is currently the main systemic treatment. With the continuous development of detection techniques and deepening research on TNBC molecular subtypes, drugs targeting immune checkpoints and different targets have emerged, such as atezolizumab, pembrolizumab, poly (ADP-ribose) polymerase (PARP) inhibitors, trophoblast cell-surface antigen 2 (TROP-2), and antibody-drug conjugates. These therapies provide new hope for TNBC treatment. Based on the analysis and classification of TNBC, this article summarizes the immunotherapy, targeted therapy, and new treatment combinations, providing references for the precise treatment of TNBC in the future.
Collapse
Affiliation(s)
- Ling Tong
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
- Department of Breast Surgery, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
| | - Xiangling Yu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
| | - Shan Wang
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
- Department of Breast Surgery, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
| | - Ling Chen
- Department of Breast Surgery, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
| | - Yibo Wu
- Human Reproductive and Genetic Center, Affiliated Hospital of Jiangnan University, Wuxi, People’s Republic of China
| |
Collapse
|
33
|
Liu X, Bai F, Wang Y, Wang C, Chan HL, Zheng C, Fang J, Zhu WG, Pei XH. Loss of function of GATA3 regulates FRA1 and c-FOS to activate EMT and promote mammary tumorigenesis and metastasis. Cell Death Dis 2023; 14:370. [PMID: 37353480 PMCID: PMC10290069 DOI: 10.1038/s41419-023-05888-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/25/2023]
Abstract
Basal-like breast cancers (BLBCs) are among the most aggressive cancers, partly due to their enrichment of cancer stem cells (CSCs). Breast CSCs can be generated from luminal-type cancer cells via epithelial-mesenchymal transition (EMT). GATA3 maintains luminal cell fate, and its expression is lost or reduced in BLBCs. However, deletion of Gata3 in mice or cells results in early lethality or proliferative defects. It is unknown how loss-of-function of GATA3 regulates EMT and CSCs in breast cancer. We report here that haploid loss of Gata3 in mice lacking p18Ink4c, a cell cycle inhibitor, up-regulates Fra1, an AP-1 family protein that promotes mesenchymal traits, and downregulates c-Fos, another AP-1 family protein that maintains epithelial fate, leading to activation of EMT and promotion of mammary tumor initiation and metastasis. Depletion of Gata3 in luminal tumor cells similarly regulates Fra1 and c-Fos in activation of EMT. GATA3 binds to FOSL1 (encoding FRA1) and FOS (encoding c-FOS) loci to repress FOSL1 and activate FOS transcription. Deletion of Fra1 or reconstitution of Gata3, but not reconstitution of c-Fos, in Gata3 deficient tumor cells inhibits EMT, preventing tumorigenesis and/or metastasis. In human breast cancers, GATA3 expression is negatively correlated with FRA1 and positively correlated with c-FOS. Low GATA3 and FOS, but high FOSL1, are characteristics of BLBCs. Together, these data provide the first genetic evidence indicating that loss of function of GATA3 in mammary tumor cells activates FOSL1 to promote mesenchymal traits and CSC function, while concurrently repressing FOS to lose epithelial features. We demonstrate that FRA1 is required for the activation of EMT in GATA3 deficient tumorigenesis and metastasis.
Collapse
Affiliation(s)
- Xiong Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Feng Bai
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, 518060, China.
- Department of Pathology, Shenzhen University Health Science Center, Shenzhen, 518060, China.
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA.
| | - Yuchan Wang
- Gansu Dian Medical Laboratory, Lanzhou, 730000, China
| | - Chuying Wang
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
- The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Ho Lam Chan
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA
| | - Chenglong Zheng
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Jian Fang
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Wei-Guo Zhu
- Department of Biochemistry and Molecular Biology, International Cancer Center, Shenzhen University Health Science Center, Shenzhen, 518060, China
| | - Xin-Hai Pei
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, International Cancer Center, Marshall Laboratory of Biomedical Engineering, The First Affiliated Hospital, Shenzhen University Health Science Center, Shenzhen, 518060, China.
- Dewitt Daughtry Family Department of Surgery, University of Miami, Miami, FL, 33136, USA.
- Department of Anatomy and Histology, Shenzhen University Health Science Center, Shenzhen, 518060, China.
| |
Collapse
|
34
|
Almahari SAI, Chandran N, Maki RJ. Deceptive Triple-Negative Breast Cancer of Intermediate Grade: A Case of Rare Microglandular Adenosis-Associated Carcinoma. Cureus 2023; 15:e39531. [PMID: 37250605 PMCID: PMC10217789 DOI: 10.7759/cureus.39531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/26/2023] [Indexed: 05/31/2023] Open
Abstract
Breast cancer is the most common cancer among women and the leading cause of cancer-related deaths globally. Ductal carcinoma of no special type is the most prevalent, followed by lobular carcinoma. Finding a triple-negative breast cancer of intermediate grade on core biopsies should raise the possibility of dealing with one of the rare subtypes such as microglandular adenosis (MGA)-associated carcinoma. Here, we present a case of a 40-year-old female, who presented with bilateral breast masses, in which one of them was a high-grade carcinoma and the other turned out to be an MGA-associated carcinoma, which was misdiagnosed initially on the core biopsy as a grade II triple-negative ductal carcinoma of no special type. Such diagnosis is challenging to pathologists, especially on small biopsies where the full morphological spectrum is not evident.
Collapse
Affiliation(s)
- Sayed Ali I Almahari
- Department of Pathology: Anatomical Pathology, Salmaniya Medical Complex, Manama, BHR
| | - Nisha Chandran
- Department of Pathology: Anatomical Pathology, Salmaniya Medical Complex, Manama, BHR
| | - Reem J Maki
- Department of Radiology, Salmaniya Medical Complex, Manama, BHR
| |
Collapse
|
35
|
Das C, Adhikari S, Bhattacharya A, Chakraborty S, Mondal P, Yadav SS, Adhikary S, Hunt CR, Yadav K, Pandita S, Roy S, Tainer JA, Ahmed Z, Pandita TK. Epigenetic-Metabolic Interplay in the DNA Damage Response and Therapeutic Resistance of Breast Cancer. Cancer Res 2023; 83:657-666. [PMID: 36661847 PMCID: PMC11285093 DOI: 10.1158/0008-5472.can-22-3015] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/30/2022] [Accepted: 01/04/2023] [Indexed: 01/21/2023]
Abstract
Therapy resistance is imposing a daunting challenge on effective clinical management of breast cancer. Although the development of resistance to drugs is multifaceted, reprogramming of energy metabolism pathways is emerging as a central but heterogenous regulator of this therapeutic challenge. Metabolic heterogeneity in cancer cells is intricately associated with alterations of different signaling networks and activation of DNA damage response pathways. Here we consider how the dynamic metabolic milieu of cancer cells regulates their DNA damage repair ability to ultimately contribute to development of therapy resistance. Diverse epigenetic regulators are crucial in remodeling the metabolic landscape of cancer. This epigenetic-metabolic interplay profoundly affects genomic stability of the cancer cells as well as their resistance to genotoxic therapies. These observations identify defining mechanisms of cancer epigenetics-metabolism-DNA repair axis that can be critical for devising novel, targeted therapeutic approaches that could sensitize cancer cells to conventional treatment strategies.
Collapse
Affiliation(s)
- Chandrima Das
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| | - Swagata Adhikari
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| | - Apoorva Bhattacharya
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
| | | | - Payel Mondal
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Homi Bhaba National Institute, Mumbai 400094, India
| | - Shalini S. Yadav
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Santanu Adhikary
- Biophysics and Structural Genomics Division, Saha Institute of Nuclear Physics, 1/AF Bidhannagar, Kolkata 700064, India
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - Clayton R Hunt
- Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Kamlesh Yadav
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, 77030, USA
| | - Shruti Pandita
- University of Texas Health San Antonio MD Anderson Cancer Center, San Antonio, Texas, 78229, USA
| | - Siddhartha Roy
- Structural Biology and Bioinformatics Division, Council of Scientific and Industrial Research (CSIR)-Indian Institute of Chemical Biology, Kolkata, India
| | - John A Tainer
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zamal Ahmed
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Tej K. Pandita
- Houston Methodist Research Institute, Houston, TX, 77030, USA
- Center for Genomics and Precision Medicine, Texas A&M College of Medicine, Houston, Texas, 77030, USA
| |
Collapse
|
36
|
Svyatenko T, Prokhach A, Antoniuk S, Hurtovyy V, Tereshkov D, Prokhach A. MCATs: Case report of adnexal adenocarcinoma of not otherwise specified type. Clin Case Rep 2023; 11:e7097. [PMID: 36937642 PMCID: PMC10017407 DOI: 10.1002/ccr3.7097] [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: 07/13/2022] [Revised: 10/14/2022] [Accepted: 02/20/2023] [Indexed: 03/18/2023] Open
Abstract
Malignant cutaneous adnexal tumors are rare dermatosis that can have extradermal twins. Diagnosis of adnexal skin tumors is sometimes difficult. Immunohistochemistry has limited value in the diagnosis. Most adnexal skin tumors are localized on the head and neck. Difficulties in their diagnosis associated with absence of strict clinical criteria.
Collapse
|
37
|
[Triple-negative breast cancer : Classification, current concepts, and therapy-related factors]. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:32-38. [PMID: 36595080 DOI: 10.1007/s00292-022-01177-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 01/04/2023]
Abstract
Triple-negative breast cancer (TNBC) accounts for about 10% of all breast cancer cases and is defined by the lack of expression of estrogen and progesterone receptors and the lack of overexpression or amplification of HER2. It differs with regard to the younger age of the patients, an increased association with a mutation of BRCA1 and a mostly low differentiation from hormone receptor-positive breast cancer. The spectrum of triple-negative breast cancer shows considerable heterogeneity both at the morphological and at the molecular level. It includes most commonly TNBC of no special type, with and without basal phenotype, triple-negative metaplastic breast carcinomas, triple-negative breast carcinomas with apocrine differentiation and rare triple-negative tumor types. At the gene-expression level, TNBC most commonly is associated with a basal phenotype, with rarer molecular variants of TNBC involving the Claudin-low, molecular apocrine types, and other rarer subtypes. Therefore, a critical use of the term TNBC, considering the histopathological tumor differentiation, is recommended.
Collapse
|
38
|
Classifying Breast Cancer Metastasis Based on Imaging of Tumor Primary and Tumor Biology. Diagnostics (Basel) 2023; 13:diagnostics13030437. [PMID: 36766541 PMCID: PMC9914718 DOI: 10.3390/diagnostics13030437] [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: 11/24/2022] [Revised: 01/14/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
The molecular classification of breast cancer has allowed for a better understanding of both prognosis and treatment of breast cancer. Imaging of the different molecular subtypes has revealed that biologically different tumors often exhibit typical features in mammography, ultrasound, and MRI. Here, we introduce the molecular classification of breast cancer and review the typical imaging features of each subtype, examining the predictive value of imaging with respect to distant metastases.
Collapse
|
39
|
Sialyl Lewis X/A and Cytokeratin Crosstalk in Triple Negative Breast Cancer. Cancers (Basel) 2023; 15:cancers15030731. [PMID: 36765690 PMCID: PMC9913872 DOI: 10.3390/cancers15030731] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/31/2022] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Triple-negative breast cancer (TNBC) encompasses multiple entities and is generally highly aggressive and metastatic. We aimed to determine the clinical and biological relevance of Sialyl-Lewis X and A (sLeX/A)-a fucosylated glycan involved in metastasis-in TNBC. Here, we studied tissues from 50 TNBC patients, transcripts from a TNBC dataset from The Cancer Genome Atlas (TCGA) database, and a primary breast cancer cell line. All 50 TNBC tissue samples analysed expressed sLeX/A. Patients with high expression of sLeX/A had 3 years less disease-free survival than patients with lower expression. In tissue, sLeX/A negatively correlated with cytokeratins 5/6 (CK5/6, which was corroborated by the inverse correlation between fucosyltransferases and CK5/6 genes. Our observations were confirmed in vitro when inhibition of sLeX/A remarkably increased expression of CK5/6, followed by a decreased proliferation and invasion capacity. Among the reported glycoproteins bearing sLeX/A and based on the STRING tool, α6 integrin showed the highest interaction score with CK5/6. This is the first report on the sLeX/A expression in TNBC, highlighting its association with lower disease-free survival and its inverse crosstalk with CK5/6 with α6 integrin as a mediator. All in all, sLeX/A is critical for TNBC malignancy and a potential prognosis biomarker and therapeutic target.
Collapse
|
40
|
Tumorigenicity of EGFR- and/or HER2-Positive Breast Cancers Is Mediated by Recruitment of Tumor-Associated Macrophages. Int J Mol Sci 2023; 24:ijms24021443. [PMID: 36674955 PMCID: PMC9866454 DOI: 10.3390/ijms24021443] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 12/27/2022] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Basal-like breast cancer (BLBC) has a clinically aggressive nature. It is prevalent in young women and is known to often relapse rapidly. To date, the molecular mechanisms regarding the aggressiveness of BLBC have not been fully understood. In the present study, mechanisms of aggressiveness of BLBC involving EGFR and/or HER2 expression and interactions between tumor and tumor-associated macrophages (TAMs) were explored. The prognosis of breast cancer patients who underwent surgery at Samsung Medical Center was analyzed. It was found that the co-expression of EGFR and HER2 was associated with a worse prognosis. Therefore, we generated EGFR-positive BLBC cells with stable HER2 overexpression and analyzed the profile of secretory cytokines. Chemokine (C-C motif) ligand 2 (CCL2) expression was increased in HER2-overexpressed BLBC cells. Recombinant human CCL2 treatment augmented the motility of TAMs. In addition, the conditioned culture media of HER2-overexpressed BLBC cells increased the motility of TAMs. Furthermore, activation of TAMs by CCL2 or the conditioned culture media of HER2-overexpressed cells resulted in the production of pro-inflammatory cytokines, such as IL-8 and IL-1β. These observations reveal that CCL2 derived from EGFR and HER2 co-expressed BLBC cells can lead to increased TAM recruitment and the induction of IL-8 and IL-1β from recruited TAMs, triggering the tumorigenesis of breast cancer with the expression of both EGFR and HER2. Our findings demonstrate that EGFR+ and HER2+ BLBC aggressiveness is partially mediated through the interaction between BLBC and TAMs recruited by CCL2.
Collapse
|
41
|
Li F, Niu M, Qin K, Guo R, Yi Y, Xu J, Li L, Xie S, Fu M, Wen N, Liao W, Xiao ZXJ. FBXL2 promotes E47 protein instability to inhibit breast cancer stemness and paclitaxel resistance. Oncogene 2023; 42:339-350. [PMID: 36460773 DOI: 10.1038/s41388-022-02559-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 12/05/2022]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with a high risk of metastasis and recurrence. Although chemotherapy has greatly improved the clinical outcome of TNBC patients, acquired drug resistance remains a huge challenge for TNBC treatment. Breast cancer stem cells (BCSCs) play a critical role in breast cancer development, metastasis, recurrence, and chemotherapy resistance. Thus, it is of great importance to decipher the underlying molecular mechanism of BCSCs regulation for TNBC drug resistance. In this study, we demonstrate that the F-box protein FBXL2 is a critical negative regulator of BCSCs stemness and that downregulation of FBXL2 plays a causal role in TNBC drug resistance. We show that expression levels of FBXL2 significantly influence CD44high/CD24low subpopulation and the mammosphere formation ability of TNBC cells. Ectopic expression of FBXL2 inhibits initiation of TNBC and overcomes paclitaxel resistance in vivo. In addition, activation of FBXL2 by nebivolol, a clinically used small-molecule inhibitor of the beta-1 receptor, markedly overcomes BCSCs-induced paclitaxel resistance. Mechanistically, we show that FBXL2 targets transcriptional factor E47 for polyubiquitin- and proteasome-mediated degradation, resulting in inhibition of BCSC stemness. Clinical analyses indicate that low expression of FBXL2 correlates with high expression of E47 as well as with high stemness features, and is associated with poor clinical outcomes of breast cancer patients. Taken together, these results highlight that the FBXL2-E47 axis plays a critical role in the regulation of BCSC stemness and paclitaxel resistance. Thus, targeting FBXL2 might be a potential therapeutic strategy for drug-resistant TNBC.
Collapse
Affiliation(s)
- Fengtian Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, P.R. China
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Mengmeng Niu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, P.R. China.
| | - Kewei Qin
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, P.R. China
| | - Rongtian Guo
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, P.R. China
| | - Yong Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, P.R. China
| | - Jing Xu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, P.R. China
| | - Luping Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, P.R. China
| | - Siyi Xie
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, P.R. China
| | - Mengyuan Fu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, P.R. China
| | - Nasi Wen
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, P.R. China
| | - Wenting Liao
- State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, 510060, P.R. China
| | - Zhi-Xiong Jim Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610065, P.R. China.
| |
Collapse
|
42
|
Balkrishna A, Umar Zango U, Kauser Nasir S, Arya V. A Clinical Cognizance of Molecular and Pathological Diagnostic Approach of TNBC. THERAPEUTIC DRUG TARGETS AND PHYTOMEDICINE FOR TRIPLE NEGATIVE BREAST CANCER 2023:26-46. [DOI: 10.2174/9789815079784123010005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Genetic, transcriptional, and clinical heterogeneity of disease has remained
to be a prominent obstacle to the development of a targeted therapeutic approach
against TNBC. So far, based on tumor size, lymph node status, and histologic features
TNBC subtypes were stratified. Insights into inter and intratumoral heterogeneity of
TNBC were gained by next-generation sequencing, genomic, transcriptomic,
proteomic, and clinicopathological characterization. To depict tumor response to
neoadjuvant chemotherapy, radiological characterization may also a play significant
role. Biomarkers for subtyping TNBC were highly needed to depict the survival
outcome. This chapter discussed the available and possible molecular and pathological
diagnostic approaches to TNBC. Furthermore, the integration of morphological and
genomic data may emerge as a promising approach for the identification of new
therapeutic and prognostic markers to predict the likely outcome of the disease. This
chapter aims to highlight the molecular and pathological diagnostic approaches to
depict both metastatic and non-metastatic TNBC. <br>
Collapse
Affiliation(s)
- Acharya Balkrishna
- Patanjali Research Institute,Patanjali Herbal Research Department,Haridwar,India,
| | - Usman Umar Zango
- Sa'adatu Rimi College of Education,Department of Biology,Kumbotso,Nigeria,
| | - Saima Kauser Nasir
- Indian Institute of Science Bangalore,Department of Microbiology and Cell Biology (MCB),Bangalore,India,
| | - Vedpriya Arya
- Patanjali Research Institute,Patanjali Herbal Research Department,Haridwar,India,
| |
Collapse
|
43
|
Araki M, Noguchi S, Kubo Y, Yasuda A, Koh M, Otsuka H, Yokosuka M, Soeta S. Expression of receptor-type tumour endothelial marker 8 in carcinoma cells showing luminal progenitor-like phenotypes in canine mammary gland carcinomas. J Comp Pathol 2023; 200:35-45. [PMID: 36641985 DOI: 10.1016/j.jcpa.2022.12.001] [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: 07/13/2022] [Revised: 09/05/2022] [Accepted: 11/05/2022] [Indexed: 01/15/2023]
Abstract
This study aimed to investigate the expression of receptor-type tumour endothelial marker 8 (TEM8RT) in canine mammary gland carcinomas (CMGCs) using immunohistochemistry and to evaluate the association between carcinoma cell TEM8RT expression and tumour histological features, histological grades and the differentiation status of neoplastic epithelial cells. TEM8RT expression was more frequently detected in simple carcinomas (tubular and tubulopapillary) than in solid carcinomas, and it was significantly correlated with histological grade Ⅰ tumours and a low mitotic index. Additionally, TEM8RT+ carcinoma cells were more frequently found in CMGCs showing luminal progenitor-like phenotypes, such as Notch1+, CK19+/CK5+/CD49f+ and CK19+/CK5-/CD49f+. Double-labelling immunofluorescence detection techniques confirmed that most TEM8RT+ carcinoma cells expressed CD49f, Notch1 and CK19. However, TEM8RT immunoreactivity was not found in carcinoma cells expressing GATA3, which upregulates mature luminal cell differentiation. Furthermore, TEM8RT+ carcinoma cells were detected in a few CMGCs showing basal/stem cell-like phenotypes such as CK19-/CK5+/CD49f+ and CK19-/CK5+/CD49f-. These findings indicate that TEM8RT is expressed in luminal progenitor-like carcinoma cells in CMGCs. Since TEM8 enhances self-renewal in human mammary stem/progenitor cells, it also may be involved in maintenance of luminal progenitor-like carcinoma cells, resulting in prevention of their transition to basal/stem cell-like carcinoma cells and development of less malignant CMGCs. Therefore, TEM8RT may be useful for indicating prognostic outcomes and identifying the possible ontogeny of carcinoma cells in mammary gland tumours.
Collapse
Affiliation(s)
- Mami Araki
- Laboratory of Veterinary Anatomy, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Syunya Noguchi
- Department of Molecular Medicine and Anatomy, Nippon Medical School, Sendagi, Bunkyo-ku, Tokyo, Japan
| | - Yoshiaki Kubo
- Veterinary Medical Teaching Hospital, Attached Facility, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Akiko Yasuda
- Veterinary Medical Teaching Hospital, Attached Facility, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Miki Koh
- Laboratory of Veterinary Anatomy, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Hirotada Otsuka
- Laboratory of Veterinary Anatomy, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Makoto Yokosuka
- Laboratory of Comparative and Behavioral Medicine, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan
| | - Satoshi Soeta
- Laboratory of Veterinary Anatomy, Faculty of Veterinary Science, Nippon Veterinary and Life Science University, Musashino-shi, Tokyo, Japan.
| |
Collapse
|
44
|
Lien K, Mayer W, Herrera R, Padilla NT, Cai X, Lin V, Pholcharoenchit R, Palefsky J, Tugizov SM. HIV-1 Proteins gp120 and Tat Promote Epithelial-Mesenchymal Transition and Invasiveness of HPV-Positive and HPV-Negative Neoplastic Genital and Oral Epithelial Cells. Microbiol Spectr 2022; 10:e0362222. [PMID: 36314970 PMCID: PMC9770004 DOI: 10.1128/spectrum.03622-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022] Open
Abstract
The incidence of human papillomavirus (HPV)-associated anogenital and oropharyngeal cancer in human immunodeficiency virus (HIV)-infected individuals is substantially higher than in HIV-uninfected individuals. HIV may also be a risk factor for the development of HPV-negative head and neck, liver, lung, and kidney cancer. However, the molecular mechanisms underlying HIV-1-associated increase of epithelial malignancies are not fully understood. Here, we showed that HPV-16-immortalized anal AKC-2 and cervical CaSki epithelial cells that undergo prolonged exposure to cell-free HIV-1 virions or HIV-1 viral proteins gp120 and tat respond with the epithelial-mesenchymal transition (EMT) and increased invasiveness. Similar responses were observed in HPV-16-infected SCC-47 and HPV-16-negative HSC-3 oral epithelial cancer cells that were cultured with these viral proteins. EMT induced by gp120 and tat led to detachment of poorly adherent cells from the culture substratum; these cells remained capable of reattachment, upon which they coexpressed both E-cadherin and vimentin, indicative of an intermediate stage of EMT. The reattached cells also expressed stem cell markers CD133 and CD44, which may play a critical role in cancer cell invasion and metastasis. Inhibition of transforming growth factor (TGF)-β1 and MAPK signaling and vimentin expression, and restoration of E-cadherin expression reduced HIV-induced EMT and the invasive activity of HPV-16-immortalized anal and cervical epithelial cells. Collectively, our results suggest that these approaches along with HIV viral suppression with antiretroviral therapy (ART) might be useful to limit the role of HIV-1 infection in the acceleration of HPV-associated or HPV-independent epithelial neoplasia. IMPORTANCE HPV-16-immortalized genital and oral epithelial cells and HPV-negative oral cancer cells that undergo prolonged contact with cell-free HIV-1 virions or with viral proteins gp120 and tat respond by becoming more invasive. EMT cells induced by HIV-1 in cultures of HPV-16-immortalized anal and cervical epithelial cells express the stem cell markers CD133 and CD44. These results suggest that the interaction of HIV-1 with neoplastic epithelial cells may lead to their de-differentiation into cancer stem cells that are resistant to apoptosis and anti-cancer drugs. Thus, this pathway may play a critical role in the development of invasive cancer. Inhibition of TGF-β1 and MAPK signaling and vimentin expression, and restoration of E-cadherin expression reduced HIV-induced EMT and the invasiveness of HPV-16-immortalized anal and cervical epithelial cells. Taken together, these results suggest that these approaches might be exploited to limit the role of HIV-1 infection in the acceleration of HPV-associated or HPV-independent epithelial neoplasia.
Collapse
Affiliation(s)
- Kathy Lien
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Wasima Mayer
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Rossana Herrera
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Nicole T. Padilla
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Xiaodan Cai
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Vicky Lin
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | | | - Joel Palefsky
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| | - Sharof M. Tugizov
- Department of Medicine, University of California-San Francisco, San Francisco, California, USA
| |
Collapse
|
45
|
Rashid NS, Boyd DC, Olex AL, Grible JM, Duong AK, Alzubi MA, Altman JE, Leftwich TJ, Valentine AD, Hairr NS, Zboril EK, Smith TM, Pfefferle AD, Dozmorov MG, Harrell JC. Transcriptomic changes underlying EGFR inhibitor resistance in human and mouse models of basal-like breast cancer. Sci Rep 2022; 12:21248. [PMID: 36482068 PMCID: PMC9731984 DOI: 10.1038/s41598-022-25541-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 11/30/2022] [Indexed: 12/13/2022] Open
Abstract
The goals of this study were to identify transcriptomic changes that arise in basal-like breast cancer cells during the development of resistance to epidermal growth factor receptor inhibitors (EGFRi) and to identify drugs that are cytotoxic once EGFRi resistance occurs. Human patient-derived xenografts (PDXs) were grown in immunodeficient mice and treated with a set of EGFRi; the EGFRi erlotinib was selected for more expansive in vivo studies. Single-cell RNA sequencing was performed on mammary tumors from the basal-like PDX WHIM2 that was treated with vehicle or erlotinib for 9 weeks. The PDX was then subjected to long-term erlotinib treatment in vivo. Through serial passaging, an erlotinib-resistant subline of WHIM2 was generated. Bulk RNA-sequencing was performed on parental and erlotinib-resistant tumors. In vitro high-throughput drug screening with > 500 clinically used compounds was performed on parental and erlotinib-resistant cells. Previously published bulk gene expression microarray data from MMTV-Wnt1 tumors were contrasted with the WHIM2 PDX data. Erlotinib effectively inhibited WHIM2 tumor growth for approximately 4 weeks. Compared to untreated cells, single-cell RNA sequencing revealed that a greater proportion of erlotinib-treated cells were in the G1 phase of the cell cycle. Comparison of WHIM2 and MMTV-Wnt1 gene expression data revealed a set of 38 overlapping genes that were differentially expressed in the erlotinib-resistant WHIM2 and MMTV-Wnt1 tumors. Comparison of all three data types revealed five genes that were upregulated across all erlotinib-resistant samples: IL19, KLK7, LCN2, SAA1, and SAA2. Of these five genes, LCN2 was most abundantly expressed in triple-negative breast cancers, and its knockdown restored erlotinib sensitivity in vitro. Despite transcriptomic differences, parental and erlotinib-resistant WHIM2 displayed similar responses to the majority of drugs assessed for cytotoxicity in vitro. This study identified transcriptomic changes arising in erlotinib-resistant basal-like breast cancer. These data could be used to identify a biomarker or develop a gene signature predictive of patient response to EGFRi. Future studies should explore the predictive capacity of these gene signatures as well as how LCN2 contributes to the development of EGFRi resistance.
Collapse
Affiliation(s)
- Narmeen S Rashid
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
- Department of Biology, University of Richmond, Richmond, VA, 23173, USA
| | - David C Boyd
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
- Program in Integrative Life Sciences, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Amy L Olex
- C. Kenneth and Diane Wright Center for Clinical and Translational Research, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Jacqueline M Grible
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Alex K Duong
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Mohammad A Alzubi
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
- Oncology Center-Division of Pediatric Oncology, Johns Hopkins University, Baltimore, MD, 21287, USA
| | - Julia E Altman
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Tess J Leftwich
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Aaron D Valentine
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Nicole S Hairr
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Emily K Zboril
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Timothy M Smith
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - Adam D Pfefferle
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27514, USA
| | - Mikhail G Dozmorov
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA
- Department of Biostatistics, Virginia Commonwealth University, Richmond, VA, 23220, USA
| | - J Chuck Harrell
- Department of Pathology, Virginia Commonwealth University, Richmond, VA, 23220, USA.
- Massey Cancer Center, Virginia Commonwealth University, Richmond, VA, 23220, USA.
| |
Collapse
|
46
|
Lin PH, Tseng LM, Lee YH, Chen ST, Yeh DC, Dai MS, Liu LC, Wang MY, Lo C, Chang S, Tan KT, Chen SJ, Kuo SH, Huang CS. Neoadjuvant afatinib with paclitaxel for triple-negative breast cancer and the molecular characteristics in responders and non-responders. J Formos Med Assoc 2022; 121:2538-2547. [PMID: 35752529 DOI: 10.1016/j.jfma.2022.05.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/25/2022] [Accepted: 05/30/2022] [Indexed: 10/17/2022] Open
Abstract
BACKGROUND The prognosis of triple-negative breast cancer (TNBC) is worse and a major proportion of TNBC expresses epidermal growth factor receptor (EGFR). Afatinib can inhibit EGFR signal pathway; however, its treatment effect for TNBC is unknown. Thus, we aimed to assess the efficacy and biomarkers of afatinib in combination with paclitaxel in a neoadjuvant setting. METHODS Patients with stage II to III TNBC were enrolled. They received 40 mg of afatinib daily for 14 days, followed by daily afatinib and weekly paclitaxel (80 mg/m2) every 21 days for four to six cycles. To explore the mechanisms of responsiveness and non-responsiveness, 409 cancer-associated genes were sequenced. RESULTS Twenty-one patients were enrolled and one patient achieved a complete clinical response; however, a 2 mm residual tumor was noted in the surgical specimen. Overall, 33.0% patients were responders. Fifteen patients received molecular testing. No activated mutation of EGFR or Her2 were found. Activated PI3K or JAK2 pathway were trended to associate with non-responder (p = 0.057). Mutation of homologous recombination (HR) genes were correlated with non-responsiveness (p = 0.005). Seven patients did not have altered PI3K, JAK2 or HR pathway; six (85.7%) of them were responder. Patients with the amplified DAXX gene was associated with a favorable trend of response (p = 0.109). CONCLUSION Adding afatinib to neoadjuvant paclitaxel generated a modest effect in TNBC. Exploratory molecular analysis suggested that activated PI3K, JAK2 pathways and mutation of HR genes were associated with therapeutic non-responsiveness, and amplification of DAXX genes was associated with responsiveness to afatinib in combination with paclitaxel.
Collapse
Affiliation(s)
- Po-Han Lin
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan; Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Ling-Ming Tseng
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Hsuan Lee
- Department of Pathology, National Taiwan University Hospital, Taipei, Taiwan
| | - Shou-Tung Chen
- Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | - Dah-Cherng Yeh
- Department of Surgery, Chung Kang Branch, Cheng Ching Hospital, Taichung, Taiwan
| | - Ming-Shen Dai
- Hematology and Oncology, Department of Internal Medicine, Tri-service General Hospital, Taipei, Taiwan
| | - Liang-Chih Liu
- Department of Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Ming-Yang Wang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | - Chiao Lo
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan
| | | | | | | | - Sung-Hsin Kuo
- Department of Medical Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chiun-Sheng Huang
- Department of Surgery, National Taiwan University Hospital, Taipei, Taiwan; Department of Surgery, College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
47
|
Williams SD, Smith TM, Stewart LV, Sakwe AM. Hypoxia-Inducible Expression of Annexin A6 Enhances the Resistance of Triple-Negative Breast Cancer Cells to EGFR and AR Antagonists. Cells 2022; 11:3007. [PMID: 36230969 PMCID: PMC9564279 DOI: 10.3390/cells11193007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 09/16/2022] [Accepted: 09/21/2022] [Indexed: 11/29/2022] Open
Abstract
Physiological changes such as hypoxia in the tumor microenvironment (TME) endow cancer cells with malignant properties, leading to tumor recurrence and rapid progression. Here, we assessed the effect of hypoxia (1% Oxygen) on the tumor suppressor Annexin A6 (AnxA6) and the response of triple-negative breast cancer (TNBC) cells to epidermal growth factor receptor (EGFR) and androgen receptor (AR) targeted therapies. We demonstrate that brief exposure of TNBC cells to hypoxia (within 24 h) is associated with down regulation of AnxA6 while > 24 h exposure cell type dependently stimulated the expression of AnxA6. Hypoxia depicted by the expression and stability of HIF-1/2α led to up regulation of the HIF target genes SLC2A1, PGK1 as well as AR and the AR target genes FABP-4 and PPAR-γ, but the cellular levels of AnxA6 protein decreased under prolonged hypoxia. Down regulation of AnxA6 in TNBC cells inhibited, while AnxA6 over expression enhanced the expression and cellular levels of HIF-1/2α, SLC2A1 and PGK1. RNAi mediated inhibition of hypoxia induced AnxA6 expression also strongly inhibited glucose uptake and ROS production in AnxA6 expressing TNBC cells. Using a luciferase reporter assay, we confirm that short-term exposure of cells to hypoxia inhibits while prolonged exposure of cells to hypoxia enhances AnxA6 promoter activity in HEK293T cells. Compared to cells cultured under normoxia, TNBC cells were more resistant to lapatinib under hypoxic conditions, and the downregulation of AnxA6 sensitized the cells to EGFR as well as AR antagonists. These data suggest that AnxA6 is a hypoxia inducible gene and that targeting AnxA6 upregulation may be beneficial in overcoming TNBC resistance to EGFR and/or AR targeted therapies.
Collapse
Affiliation(s)
- Stephen D. Williams
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA
| | - Tunde M. Smith
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA
| | - LaMonica V. Stewart
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA
- School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Amos M. Sakwe
- Department of Biochemistry, Cancer Biology, Neuroscience, and Pharmacology, School of Graduate Studies and Research, Meharry Medical College, Nashville, TN 37208, USA
| |
Collapse
|
48
|
Li Y, Zhang H, Merkher Y, Chen L, Liu N, Leonov S, Chen Y. Recent advances in therapeutic strategies for triple-negative breast cancer. J Hematol Oncol 2022; 15:121. [PMID: 36038913 PMCID: PMC9422136 DOI: 10.1186/s13045-022-01341-0] [Citation(s) in RCA: 392] [Impact Index Per Article: 130.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/18/2022] [Indexed: 01/03/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is the most malignant subtype of breast cancer (BC) with a poor prognosis. Current treatment options are limited to surgery, adjuvant chemotherapy and radiotherapy; however, a proportion of patients have missed the surgical window at the time of diagnosis. TNBC is a highly heterogeneous cancer with specific mutations and aberrant activation of signaling pathways. Hence, targeted therapies, such as those targeting DNA repair pathways, androgen receptor signaling pathways, and kinases, represent promising treatment options against TNBC. In addition, immunotherapy has also been demonstrated to improve overall survival and response in TNBC. In this review, we summarize recent key advances in therapeutic strategies based on molecular subtypes in TNBC.
Collapse
Affiliation(s)
- Yun Li
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Huajun Zhang
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Yulia Merkher
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia, 141700
| | - Lin Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Na Liu
- Department of Endocrinology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Sergey Leonov
- School of Biological and Medical Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia, 141700. .,Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia, 142290.
| | - Yongheng Chen
- Department of Oncology, NHC Key Laboratory of Cancer Proteomics, Laboratory of Structural Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
49
|
Cheng T, Wu Y, Liu Z, Yu Y, Sun S, Guo M, Sun B, Huang C. CDKN2A-mediated molecular subtypes characterize the hallmarks of tumor microenvironment and guide precision medicine in triple-negative breast cancer. Front Immunol 2022; 13:970950. [PMID: 36052076 PMCID: PMC9424905 DOI: 10.3389/fimmu.2022.970950] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/22/2022] [Indexed: 11/13/2022] Open
Abstract
Currently, breast cancer (BRCA) has become the most common cancer in the world, whose pathological mechanism is complex. Among its subtypes, triple-negative breast cancer (TNBC) has the worst prognosis. With the increasing number of diagnosed TNBC patients, the urgent need of novel biomarkers is also rising. Cyclin-dependent kinase inhibitor 2A (CDKN2A) has recently emerged as a key regulator associated with ferroptosis and cuproptosis (FAC) and has exhibited a significant effect on BRCA, but its detailed mechanism remains elusive. Herein, we conducted the first converge comprehensive landscape analysis of FAC-related gene CDKN2A in BRCA and disclosed its prognostic value in BRCA. Then, an unsupervised cluster analysis based on CDKN2A-correlated genes unveiled three subtypes, namely cold-immune subtype, IFN-γ activated subtype and FTL-dominant subtype. Subsequent analyses depicting hallmarks of tumor microenvironment (TME) among three subtypes suggested strong association between TNBC and CDKN2A. Given the fact that the most clinically heterogeneous TNBC always displayed the most severe outcomes and lacked relevant drug targets, we further explored the potential of immunotherapy for TNBC by interfering CDKN2A and constructed the CDKN2A-derived prognostic model for TNBC patients by Lasso-Cox. The 21-gene–based prognostic model showed high accuracy and was verified in external independent validation cohort. Moreover, we proposed three drugs for TNBC patients based on our model via targeting epidermal growth factor receptor. In summary, our study indicated the potential of CDKN2A as a pioneering prognostic predictor for TNBC and provided a rationale of immunotherapy for TNBC, and offered fresh perspectives and orientations for cancer treatment via inducing ferroptosis and cuproptosis to develop novel anti-cancer treatment strategies.
Collapse
Affiliation(s)
- Tianyi Cheng
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Yingyi Wu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, Macao SAR, China
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Zhiyu Liu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Yi Yu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, Macao SAR, China
| | - Shixue Sun
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Min Guo
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, China
| | - Baoqing Sun
- Department of Allergy and Clinical Immunology, Department of Laboratory, National Center for Respiratory Medicine, National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Baoqing Sun, ; Chen Huang,
| | - Chen Huang
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macao, Macao SAR, China
- Dr. Neher’s Biophysics Laboratory for Innovative Drug Discovery, Macau University of Science and Technology, Macao, Macao SAR, China
- *Correspondence: Baoqing Sun, ; Chen Huang,
| |
Collapse
|
50
|
Meng L, Song K, Li S, Kang Y. Exosomes: Small Vesicles with Important Roles in the Development, Metastasis and Treatment of Breast Cancer. MEMBRANES 2022; 12:membranes12080775. [PMID: 36005690 PMCID: PMC9414313 DOI: 10.3390/membranes12080775] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 05/12/2023]
Abstract
Breast cancer (BC) has now overtaken lung cancer as the most common cancer, while no biopredictive marker isolated from biological fluids has yet emerged clinically. After traditional chemotherapy, with the huge side effects brought by drugs, patients also suffer from the double affliction of drugs to the body while fighting cancer, and they often quickly develop drug resistance after the drug, leading to a poor prognosis. And the treatment of some breast cancer subtypes, such as triple negative breast cancer (TNBC), is even more difficult. Exosomes (Exos), which are naturally occurring extracellular vesicles (EVs) with nanoscale acellular structures ranging in diameter from 40 to 160 nm, can be isolated from various biological fluids and have been widely studied because they are derived from the cell membrane, have extremely small diameter, and are widely involved in various biological activities of the body. It can be used directly or modified to make derivatives or to make some analogs for the treatment of breast cancer. This review will focus on the involvement of exosomes in breast cancer initiation, progression, invasion as well as metastasis and the therapeutic role of exosomes in breast cancer.
Collapse
Affiliation(s)
- Ling’ao Meng
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
| | - Kedong Song
- State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
- Correspondence: (S.L.); (Y.K.)
| | - Yue Kang
- Department of Breast Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, China
- Correspondence: (S.L.); (Y.K.)
| |
Collapse
|