1
|
Callari M, Dugo M, Barreca M, Győrffy B, Galbardi B, Vigano L, Locatelli A, Dall'Ara C, Ferrarini M, Bisagni G, Colleoni M, Mansutti M, Zamagni C, Del Mastro L, Zambelli S, Frassoldati A, Biasi O, Pusztai L, Valagussa P, Viale G, Gianni L, Bianchini G. Determinants of response and molecular dynamics in HER2+ER+ breast cancers from the NA-PHER2 trial receiving HER2-targeted and endocrine therapies. Nat Commun 2025; 16:2195. [PMID: 40038334 DOI: 10.1038/s41467-025-57293-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 02/12/2025] [Indexed: 03/06/2025] Open
Abstract
Improved outcomes in HER2+ female breast cancer have resulted from chemotherapy and anti-HER2 therapies. However, HER2+ER+ cancers exhibit lower response rates. The phase 2 NA-PHER2 trial (NCT02530424) investigated chemo-free preoperative HER2 blockade (trastuzumab + pertuzumab) and CDK4/6 inhibition (palbociclib) with or without endocrine therapy (fulvestrant) in HER2+ER+ breast cancer. Clinical endpoints (i.e. Ki67 dynamics and pathological complete response) were previously reported. Here we report on the biomarker analysis, secondary objective of the study. Through RNA sequencing and tumour infiltrating lymphocytes (TIL) assessment in serial biopsies, we identified biomarkers predictive of pCR or Day14 Ki67 response and unveiled treatment-induced molecular changes. High immune infiltration and low ER signalling correlated with pCR, while TP53 mutations associated with high Day14 Ki67. Stratification based on Ki67 at Day14 and at surgery defined three response groups (Ki67 HighHigh, LowHigh, LowLow), with divergent tumour and stroma expression dynamics. The HighHigh group showed dysfunctional immune infiltration and overexpression of therapeutic targets like PAK4 at baseline. The LowLow group exhibited a Luminal A phenotype by the end of treatment. This study expands our understanding of drivers and dynamics of HER2+ER+ tumour response, towards treatment tailoring.
Collapse
MESH Headings
- Humans
- Female
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-2/genetics
- Breast Neoplasms/drug therapy
- Breast Neoplasms/genetics
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Receptors, Estrogen/metabolism
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/drug effects
- Biomarkers, Tumor/metabolism
- Biomarkers, Tumor/genetics
- Trastuzumab/therapeutic use
- Ki-67 Antigen/metabolism
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Treatment Outcome
- Middle Aged
- Mutation
- Antibodies, Monoclonal, Humanized
Collapse
Affiliation(s)
| | | | - Marco Barreca
- Fondazione Michelangelo, Milan, Italy
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
| | - Balázs Győrffy
- Dept. of Bioinformatics, Semmelweis University, Budapest, Hungary
- Dept. of Biophysics, Medical School, University of Pecs, Pecs, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, Budapest, Hungary
| | | | | | | | | | | | | | - Marco Colleoni
- IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | | | | | - Lucia Del Mastro
- Department of Internal Medicine and Medical Specialties (DiMI), School of Medicine, Università di Genova, Genoa, Italy
- Department of Medical Oncology, UO Clinica di Oncologia Medica, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | | | | | - Olivia Biasi
- IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | - Lajos Pusztai
- Department of Internal Medicine, Section of Medical Oncology, Yale School of Medicine, New Haven, CT, USA
| | | | - Giuseppe Viale
- Fondazione Michelangelo, Milan, Italy
- IEO, European Institute of Oncology, IRCCS, Milan, Italy
| | | | - Giampaolo Bianchini
- IRCCS San Raffaele Hospital, Milan, Italy.
- UniSR San Raffaele University, Milan, Italy.
| |
Collapse
|
2
|
McCoy M, Yeang CH, Bahnassy S, Tam S, Riggins RB, Parashar D, Beckman RA. Generalized Evolutionary Classifier for Evolutionary Guided Precision Medicine. JCO Precis Oncol 2025; 9:e2300714. [PMID: 40080755 PMCID: PMC11922188 DOI: 10.1200/po.23.00714] [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: 12/20/2023] [Revised: 01/13/2025] [Accepted: 02/03/2025] [Indexed: 03/15/2025] Open
Abstract
PURPOSE Current precision medicine (CPM) matches patients to therapies using traditional biomarkers, but inevitably resistance develops. Dynamic precision medicine (DPM) is a new evolutionary guided precision medicine (EGPM) approach undergoing translational development. It tracks intratumoral genetic heterogeneity and evolutionary dynamics, adapts as frequently as every 6 weeks, plans proactively for future resistance development, and incorporates multiple therapeutic agents. Simulations indicated DPM can significantly improve long-term survival and cure rates in a cohort of 3 million virtual patients representing a variety of clinical scenarios. Given the cost and invasiveness of monitoring subclones frequently, we sought to determine the value of a short DPM window of only two 6-week adaptations (moves). METHODS In a new simulation, nearly 3 million virtual patients, differing in DPM input parameters of initial subclone compositions, drug sensitivities, and growth and mutational kinetics, were simulated as previously described. Each virtual patient was treated with CPM, DPM, and DPM for two moves followed by CPM. RESULTS The first two DPM moves provide similar average benefit to a 5-year, 40-move sequence in the full virtual population. If the first two moves are identical for DPM and CPM, patients will not benefit from DPM (65% negative predictive value). A patient subset (20%) in which 2-move DPM and 40-move DPM provide closely similar outcomes has extraordinary predicted benefit (hazard ratio of DPM/CPM 0.03). CONCLUSION The first two DPM moves provide most of the clinical benefit of DPM, reducing the duration required for subclone monitoring. This also leads to an evolutionary classifier selecting patients who will benefit: those in whom DPM and CPM recommendations differ early. These advances bring DPM (and potentially other EGPM approaches) closer to potential clinical testing.
Collapse
Affiliation(s)
- Matthew McCoy
- Department of Oncology, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC
| | | | - Shaymaa Bahnassy
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Stanley Tam
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Rebecca B Riggins
- Department of Oncology, Lombardi Comprehensive Cancer Center, Georgetown University, Washington, DC
| | - Deepak Parashar
- Division of Health Sciences, Warwick Applied Health, Warwick Medical School & Warwick Cancer Research Centre, University of Warwick, Coventry, United Kingdom
- The Alan Turing Institute for Data Science and Artificial Intelligence, The British Library, London, United Kingdom
| | - Robert A Beckman
- Department of Oncology, Lombardi Comprehensive Cancer Center and Innovation Center for Biomedical Informatics, Georgetown University Medical Center, Washington, DC
- Department of Biostatistics, Bioinformatics, and Biomathematics, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC
| |
Collapse
|
3
|
Wang Y, Haase S, Whitman A, Beltran A, Spanheimer PM, Brunk E. A Multimodal Framework to Uncover Drug-Responsive Subpopulations in Triple-Negative Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.14.638274. [PMID: 40027670 PMCID: PMC11870422 DOI: 10.1101/2025.02.14.638274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Understanding how individual cancer cells adapt to drug treatment is a fundamental challenge limiting precision medicine cancer therapy strategies. While single-cell technologies have advanced our understanding of cellular heterogeneity, efforts to connect the behavior of individual cells to broader tumor drug responses and uncover global trends across diverse systems remain limited. There is a growing availability of single-cell and bulk omics data, but a lack of centralized tools and repositories makes it difficult to study drug response globally, especially at the level of single-cell adaptation. To address this, we present a multimodal framework that integrates bulk and single-cell treated and untreated transcriptomics data to identify drug responsive cell populations in triple-negative breast cancer (TNBC). Our framework leverages population-scale bulk transcriptomics data from TNBC samples to define seven main "identities", each representing unique combinations of biologically relevant genes. These identities are dynamic and trackable, allowing us to map them onto single cells and uncover global patterns of how cell populations respond to drug treatment. Unlike static classifications, this approach captures the evolving nature of cellular states, revealing that a select few identities dominate and drive population-level responses during treatment. Crucially, our ability to decode these trends through the inherent noise of single-cell data provides a clearer picture of how heterogeneous cell populations adapt to therapy. By identifying the dominant identities and their dynamics, we can better predict how entire tumors respond to treatment. This insight is essential for designing precise combination therapies tailored to the unique heterogeneity of patient tumors, addressing the single-cell variations that ultimately determine therapeutic outcomes.
Collapse
|
4
|
Pellarin I, Dall'Acqua A, Favero A, Segatto I, Rossi V, Crestan N, Karimbayli J, Belletti B, Baldassarre G. Cyclin-dependent protein kinases and cell cycle regulation in biology and disease. Signal Transduct Target Ther 2025; 10:11. [PMID: 39800748 PMCID: PMC11734941 DOI: 10.1038/s41392-024-02080-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/16/2024] [Accepted: 11/13/2024] [Indexed: 01/18/2025] Open
Abstract
Cyclin Dependent Kinases (CDKs) are closely connected to the regulation of cell cycle progression, having been first identified as the kinases able to drive cell division. In reality, the human genome contains 20 different CDKs, which can be divided in at least three different sub-family with different functions, mechanisms of regulation, expression patterns and subcellular localization. Most of these kinases play fundamental roles the normal physiology of eucaryotic cells; therefore, their deregulation is associated with the onset and/or progression of multiple human disease including but not limited to neoplastic and neurodegenerative conditions. Here, we describe the functions of CDKs, categorized into the three main functional groups in which they are classified, highlighting the most relevant pathways that drive their expression and functions. We then discuss the potential roles and deregulation of CDKs in human pathologies, with a particular focus on cancer, the human disease in which CDKs have been most extensively studied and explored as therapeutic targets. Finally, we discuss how CDKs inhibitors have become standard therapies in selected human cancers and propose novel ways of investigation to export their targeting from cancer to other relevant chronic diseases. We hope that the effort we made in collecting all available information on both the prominent and lesser-known CDK family members will help in identify and develop novel areas of research to improve the lives of patients affected by debilitating chronic diseases.
Collapse
Affiliation(s)
- Ilenia Pellarin
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Alessandra Dall'Acqua
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Andrea Favero
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Ilenia Segatto
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Valentina Rossi
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Nicole Crestan
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Javad Karimbayli
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Barbara Belletti
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy
| | - Gustavo Baldassarre
- Division of Molecular Oncology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, National Cancer Institute, Aviano, Italy.
| |
Collapse
|
5
|
Bernal YA, Blanco A, Oróstica K, Delgado I, Armisén R. Integration of RNA Editing with Multiomics Data Improves Machine Learning Models for Predicting Drug Responses in Breast Cancer Patients. RESEARCH SQUARE 2024:rs.3.rs-5604105. [PMID: 39764127 PMCID: PMC11702790 DOI: 10.21203/rs.3.rs-5604105/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Background The integration of conventional omics data such as genomics and transcriptomics data into artificial intelligence models has advanced significantly in recent years; however, their low applicability in clinical contexts, due to the high complexity of models, has been limited in their direct use inpatients. We integrated classic omics, including DNA mutation and RNA gene expression, added a novel focus on promising omics methods based on A>I(G) RNA editing, and developed a drug response prediction model. Methods We analyzed 104 patients from the Breast Cancer Genome-Guided Therapy Study (NCT02022202). This study was used to train (70%) with 10-fold cross-validation and test (30%) the drug response classification models. We assess the performance of the random forest (RF), generalized linear model (GLM), and support vector machine (SVM) with the Caret package in classifying therapy response via various combinations of clinical data, tumoral and germline mutation data, gene expression data, and RNA editing data via the LASSO and PCA strategies. Results First, we characterized the cohort on the basis of clinical data, mutation landscapes, differential gene expression, and RNAediting sites in 69 nonresponders and 35 responders to therapy. Second, regarding the prediction models, we demonstrated that RNA editing data improved or maintained the performance of the RF model for predicting drug response across all combinations. To select the final model, we compared the F1 score between models with different data combinations, highlighting an F1 score of 0.96 (95% CI: 0.957--0.961) and an AUC of 0.922, using LASSO for feature selection. Finally, we developed a nonresponse risk score on the basis of features that contributed to the selected model, focusing on three RNA-edited sites in the genes KDM4B, miRNA200/TTLL10-AS1, and BEST1. The score was created to facilitate the clinical translation of our findings, presenting a probability of therapy response according to RNA editing site patterns. Conclusion Our study highlights the potential of RNA editing as a valuable addition to predictive modeling for drug response in patients with breast cancer. The nonresponse risk score could represent a tool for clinical translation, offering a probability-based assessment of therapy response. These findings suggest that incorporating RNA editing into predictive models could enhance personalized treatment strategies and improve decision-making in oncology.
Collapse
|
6
|
Gao Y, Lin H, Tang T, Wang Y, Chen W, Li L. Circular RNAs in programmed cell death: Regulation mechanisms and potential clinical applications in cancer: A review. Int J Biol Macromol 2024; 280:135659. [PMID: 39288849 DOI: 10.1016/j.ijbiomac.2024.135659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/19/2024]
Abstract
Circular RNAs (circRNAs) are a novel class of non-coding RNAs with covalently closed structures formed by reverse splicing of precursor mRNAs. The widespread expression of circRNAs across species has been revealed by high-throughput sequencing and bioinformatics approaches, indicating their unique properties and diverse functions including acting as microRNA sponges and interacting with RNA-binding proteins. Programmed cell death (PCD), encompassing various forms such as apoptosis, necroptosis, pyroptosis, autophagy, and ferroptosis, is an essential process for maintaining normal development and homeostasis in the human body by eliminating damaged, infected, and aging cells. Many studies have demonstrated that circRNAs play crucial roles in tumourigenesis and development by regulating PCD in tumor cells, showing that circRNAs have the potential to be biomarkers and therapeutic targets in cancer. This review aims to comprehensively summarize the intricate associations between circRNAs and diverse PCD pathways in tumor cells, which play crucial roles in cancer development. Additionally, this review provides a detailed overview of the underlying mechanisms by which circRNAs modulate various forms of PCD for the first time. The ultimate objective is to offer valuable insights into the potential clinical significance of developing novel strategies based on circRNAs and PCD for cancer diagnosis, prognosis, and treatment.
Collapse
Affiliation(s)
- Yudi Gao
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Hong Lin
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Tiantian Tang
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yuanqiang Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China.
| | - Wanyi Chen
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| | - Lixian Li
- Department of Pharmacy, Chongqing University Cancer Hospital, Chongqing, 400030, China.
| |
Collapse
|
7
|
Barreca M, Dugo M, Galbardi B, Győrffy B, Valagussa P, Besozzi D, Viale G, Bianchini G, Gianni L, Callari M. Development and validation of a gene expression-based Breast Cancer Purity Score. NPJ Precis Oncol 2024; 8:242. [PMID: 39448787 PMCID: PMC11502849 DOI: 10.1038/s41698-024-00730-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 10/06/2024] [Indexed: 10/26/2024] Open
Abstract
The prevalence of malignant cells in clinical specimens, or tumour purity, is affected by both intrinsic biological factors and extrinsic sampling bias. Molecular characterization of large clinical cohorts is typically performed on bulk samples; data analysis and interpretation can be biased by tumour purity variability. Transcription-based strategies to estimate tumour purity have been proposed, but no breast cancer specific method is available yet. We interrogated over 6000 expression profiles from 10 breast cancer datasets to develop and validate a 9-gene Breast Cancer Purity Score (BCPS). BCPS outperformed existing methods for estimating tumour content. Adjusting transcriptomic profiles using the BCPS reduces sampling bias and aids data interpretation. BCPS-estimated tumour purity improved prognostication in luminal breast cancer, correlated with pathologic complete response in on-treatment biopsies from triple-negative breast cancer patients undergoing neoadjuvant treatment and effectively stratified the risk of relapse in HER2+ residual disease post-neoadjuvant treatment.
Collapse
Affiliation(s)
- Marco Barreca
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Milan, Italy
- Fondazione Michelangelo, Milan, Italy
| | | | | | - Balázs Győrffy
- Department of Bioinformatics, Semmelweis University, H-1094, Budapest, Hungary
- Department of Biophysics, Medical School, University of Pecs, H-7624, Pecs, Hungary
- Cancer Biomarker Research Group, Institute of Molecular Life Sciences, Research Centre for Natural Sciences, H-1117, Budapest, Hungary
| | | | - Daniela Besozzi
- Department of Informatics, Systems and Communication, University of Milano-Bicocca, Milan, Italy
- Bicocca Bioinformatics, Biostatistics and Bioimaging (B4) Research Centre, Milan, Italy
| | | | - Giampaolo Bianchini
- IRCCS San Raffaele Hospital, Milan, Italy.
- Università Vita-Salute San Raffaele, Milan, Italy.
| | | | | |
Collapse
|
8
|
Martello SE, Xia J, Kusunose J, Hacker BC, Mayeaux MA, Lin EJ, Hawkes A, Singh A, Caskey CF, Rafat M. Ultrafast power doppler ultrasound enables longitudinal tracking of vascular changes that correlate with immune response after radiotherapy. Theranostics 2024; 14:6883-6896. [PMID: 39629131 PMCID: PMC11610147 DOI: 10.7150/thno.97759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 10/09/2024] [Indexed: 12/06/2024] Open
Abstract
Rationale: While immunotherapy shows great promise in patients with triple negative breast cancer, many will not respond to treatment. Radiotherapy has the potential to prime the tumor-immune microenvironment for immunotherapy. However, predicting response is difficult due to tumor heterogeneity across patients, which necessitates personalized medicine strategies that incorporate tumor tracking into the therapeutic approach. Here, we investigated the use of ultrasound (US) imaging of the tumor vasculature to monitor the tumor response to treatment. Methods: We utilized ultrafast power doppler US to track the vascular response to radiotherapy over time. We used 4T1 (metastatic) and 67NR (non-metastatic) breast cancer models to determine if US measurements corroborate conventional immunostaining analysis of the tumor vasculature. To evaluate the effects of radiation, tumor volume and vascular index were calculated using US, and the correlation between vascular changes and immune cell infiltration was determined. Results: US tumor measurements and the quantified vascular response to radiation were confirmed with caliper measurements and immunostaining, respectively, demonstrating a proof-of-principle method for non-invasive vascular monitoring. Additionally, we found significant infiltration of CD8+ T cells into irradiated tumors 10 days after radiation, which followed a sustained decline in vascular index and an increase in splenic CD8+ T cells that was first observed 1 day post-radiation. Conclusions: Our findings reveal that ultrafast power doppler US can evaluate changes in tumor vasculature that are indicative of shifts in the tumor-immune microenvironment. This work may lead to improved patient outcomes through observing and predicting response to therapy.
Collapse
Affiliation(s)
- Shannon E. Martello
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jixin Xia
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jiro Kusunose
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Benjamin C. Hacker
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
| | - McKenzie A. Mayeaux
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Erica J. Lin
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Adrienne Hawkes
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Aparna Singh
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Charles F. Caskey
- Department of Radiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Marjan Rafat
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
- Department of Radiation Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
9
|
Mazzeo R, Sears J, Palmero L, Bolzonello S, Davis AA, Gerratana L, Puglisi F. Liquid biopsy in triple-negative breast cancer: unlocking the potential of precision oncology. ESMO Open 2024; 9:103700. [PMID: 39288656 PMCID: PMC11421323 DOI: 10.1016/j.esmoop.2024.103700] [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: 03/05/2024] [Revised: 06/29/2024] [Accepted: 08/06/2024] [Indexed: 09/19/2024] Open
Abstract
In the era of precision oncology, the management of triple-negative breast cancer (TNBC) is rapidly changing and becoming more complicated with a variety of chemotherapy, immunotherapy, and targeted treatment options. Currently, TNBC treatment is based on prognostic and predictive factors including immunohistochemical biomarkers [e.g. programmed death-ligand 1 (PD-L1)] and germline BRCA mutations. Given the current limitation of existing biomarkers, liquid biopsies may serve as clinically useful tools to determine treatment efficacy and response in both the (neo)adjuvant and metastatic settings, for detecting early relapse, and for monitoring clonal evolution during treatment. In this review, we comprehensively summarize current and future liquid biopsy applications. Specifically, we highlight the role of circulating tumor cell characterization, circulating tumor DNA, and other preclinical liquid biopsy technologies including circulating exosomes, RNA liquid biopsy, and circulating immune-based biomarkers. In the near future, these biomarkers may serve to identify early disease relapse, therapeutic targets, and disease clonality for patients with TNBC in the clinical setting.
Collapse
Affiliation(s)
- R Mazzeo
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano; Department of Medicine, University of Udine, Udine, Italy
| | - J Sears
- Department of Medicine, Washington University in St. Louis, St. Louis
| | - L Palmero
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano; Department of Medicine, University of Udine, Udine, Italy
| | - S Bolzonello
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano
| | - A A Davis
- Department of Medicine, Division of Oncology, Washington University School of Medicine, St. Louis, USA
| | - L Gerratana
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano; Department of Medicine, University of Udine, Udine, Italy.
| | - F Puglisi
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Aviano; Department of Medicine, University of Udine, Udine, Italy
| |
Collapse
|
10
|
Lee S, Kim JY, Lee SJ, Hwang CS, Lee HJ, Kim KB, Lee JH, Shin DH, Choi KU, Lee CH, Huh GY, Kim A. Impact of Neoadjuvant Chemotherapy (NAC) on Biomarker Expression in Breast Cancer. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:737. [PMID: 38792920 PMCID: PMC11123214 DOI: 10.3390/medicina60050737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 04/24/2024] [Accepted: 04/28/2024] [Indexed: 05/26/2024]
Abstract
Background and Objectives: This study aimed to explore biomarker change after NAC (neoadjuvant chemotherapy) and to investigate biomarker expression as a prognostic factor in patients with residual disease (RD) after NAC. Materials and Methods: We retrospectively evaluated 104 patients with invasive breast cancer, who underwent NAC and surgery at Pusan National University Hospital from 2015 to July 2022. The expression of the biomarker was assessed, and the overall survival (OS) and disease-free survival (DFS) were investigated. Results: After NAC, 24 patients (23.1%) out of 104 total patients had a pathological complete response (pCR). We found that changes in at least one biomarker were observed in 41 patients (51.2%), among 80 patients with RD. In patients with RD after NAC (n = 80), a subtype change was identified in 20 patients (25.0%). Any kind of change in the HER2 status was present 19 (23.7%) patients. The hormone receptor (HR)+/HER2+ subtype was significantly associated with better disease-free survival (DFS) (HR, 0.13; 95% CI, 0.02-0.99; p = 0.049). No change in p53 was associated with better DFS, and negative-to-positive change in p53 expression after NAC was correlated with worse DFS (p < 0.001). Negative-to-positive change in p53 was an independent, worse DFS factor in the multivariate analysis (HR,18.44; 95% CI, 1.86-182.97; p = 0.013). Conclusions: Biomarker change and subtype change after NAC were not infrequent, which can affect the further treatment strategy after surgery. The expression change of p53 might have a prognostic role. Overall, we suggest that the re-evaluation of biomarkers after NAC can provide a prognostic role and is needed for the best decision to be made on further treatment.
Collapse
Affiliation(s)
- Suji Lee
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, 179 Gudeok-ro, Seo-gu, Busan 49241, Republic of Korea
- Department of Pathology, School of Medicine, Pusan National University, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
| | - Jee Yeon Kim
- Department of Pathology, School of Medicine, Pusan National University, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
- Department of Pathology, Yangsan Pusan National University Hospital, Medical Research Institute, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
| | - So Jeong Lee
- Department of Pathology, Seegene Medial Foundation Busan, Joongangdaero 297, Busan 48792, Republic of Korea
| | - Chung Su Hwang
- Department of Pathology, School of Medicine, Pusan National University, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
- Department of Pathology, Yangsan Pusan National University Hospital, Medical Research Institute, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
| | - Hyun Jung Lee
- Department of Pathology, School of Medicine, Pusan National University, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
- Department of Pathology, Yangsan Pusan National University Hospital, Medical Research Institute, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
| | - Kyung Bin Kim
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, 179 Gudeok-ro, Seo-gu, Busan 49241, Republic of Korea
- Department of Pathology, School of Medicine, Pusan National University, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
| | - Jung Hee Lee
- Department of Pathology, School of Medicine, Pusan National University, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
- Department of Pathology, Yangsan Pusan National University Hospital, Medical Research Institute, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
| | - Dong Hoon Shin
- Department of Pathology, School of Medicine, Pusan National University, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
- Department of Pathology, Yangsan Pusan National University Hospital, Medical Research Institute, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
| | - Kyung Un Choi
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, 179 Gudeok-ro, Seo-gu, Busan 49241, Republic of Korea
- Department of Pathology, School of Medicine, Pusan National University, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
| | - Chang Hun Lee
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, 179 Gudeok-ro, Seo-gu, Busan 49241, Republic of Korea
- Department of Pathology, School of Medicine, Pusan National University, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
| | - Gi Yeong Huh
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, 179 Gudeok-ro, Seo-gu, Busan 49241, Republic of Korea
- Department of Pathology, School of Medicine, Pusan National University, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
| | - Ahrong Kim
- Department of Pathology, Pusan National University Hospital, Biomedical Research Institution, 179 Gudeok-ro, Seo-gu, Busan 49241, Republic of Korea
- Department of Pathology, School of Medicine, Pusan National University, Beomeori, Mulgeum-eop, Yangsan 50612, Republic of Korea
| |
Collapse
|
11
|
Morrison L, Loibl S, Turner NC. The CDK4/6 inhibitor revolution - a game-changing era for breast cancer treatment. Nat Rev Clin Oncol 2024; 21:89-105. [PMID: 38082107 DOI: 10.1038/s41571-023-00840-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 79.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2023] [Indexed: 01/27/2024]
Abstract
Cyclin-dependent kinase (CDK) 4/6 inhibition in combination with endocrine therapy is the standard-of-care treatment for patients with advanced-stage hormone receptor-positive, HER2 non-amplified (HR+HER2-) breast cancer. These agents can also be administered as adjuvant therapy to patients with higher-risk early stage disease. Nonetheless, the clinical success of these agents has created several challenges, such as how to address acquired resistance, identifying which patients are most likely to benefit from therapy prior to treatment, and understanding the optimal timing of administration and sequencing of these agents. In this Review, we describe the rationale for targeting CDK4/6 in patients with breast cancer, including a summary of updated clinical evidence and how this should inform clinical practice. We also discuss ongoing research efforts that are attempting to address the various challenges created by the widespread implementation of these agents.
Collapse
Affiliation(s)
- Laura Morrison
- Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK
- Breast Unit, The Royal Marsden Hospital, London, UK
| | - Sibylle Loibl
- German Breast Group, Goethe University, Frankfurt, Germany
| | - Nicholas C Turner
- Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK.
- Breast Unit, The Royal Marsden Hospital, London, UK.
| |
Collapse
|
12
|
Araghi M, Gharebakhshi F, Faramarzi F, Mafi A, Mousavi T, Alimohammadi M, Soleimantabar H. Efficacy and Safety of Pembrolizumab Monotherapy or Combined Therapy in Patients with Metastatic Triple-negative Breast Cancer: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Curr Gene Ther 2024; 25:72-88. [PMID: 39468438 DOI: 10.2174/0115665232283880240301035621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 10/30/2024]
Abstract
BACKGROUND Metastatic Triple-negative Breast Cancer (mTNBC) is the most aggressive form of breast cancer, with a greater risk of metastasis and recurrence. Research studies have published in-depth analyses of the advantages and disadvantages of pembrolizumab, and early data from numerous trials suggests that patients with mTNBC have had remarkable outcomes. This meta-analysis compares the data from numerous relevant studies in order to evaluate the safety and efficacy of pembrolizumab monotherapy or combination therapies for mTNBC. METHODS To identify eligible RCTs, a thorough literature search was carried out using electronic databases. CMA software was utilized to perform heterogeneity tests using fixed and random-effects models. RESULTS According to our pooled data, the median Progression-free Survival (PFS) was 2.66 months, and the median overall survival (OS) was 12.26 months. Furthermore, by comparing efficacy indicators between PD-L1-positive and PD-L1-negative groups, a correlation was found between the overexpression of PD-L1 with OS, PFS, and ORR. Patients with PD-L1-positive tumors had a higher response rate, with an ORR of 21.1%, compared to the patients with PD-L1-negative tumors. The ORR for first-line immunotherapy was higher than that of ≥second-line immunotherapy. In addition, pembrolizumab plus combination treatment resulted in a pooled incidence of immune- related adverse events of 22.7%. CONCLUSION A modest response to pembrolizumab monotherapy was detected in the mTNBC patients. Furthermore, a better outcome from pembrolizumab treatment may be predicted by PD-L1-- positive status, non-liver/lung metastases, combination therapy, and first-line immunotherapy. Pembrolizumab, in combination with chemotherapy, may be more beneficial for patients whose tumors are PD-L1 positive.
Collapse
Affiliation(s)
- Mahmood Araghi
- Department of Pathology, School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Farshad Gharebakhshi
- Department of Radiology, School of Medicine, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Faramarzi
- Department of Immunology, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Mafi
- Department of Clinical Biochemistry, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Tahoora Mousavi
- Molecular and Cell Biology Research Center (MCBRC), Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
- Medical Sciences Technologies, Molecular and Cell Biology Research Center (MCBRC), Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mina Alimohammadi
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hussein Soleimantabar
- Department of Radiology, School of Medicine, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Jiang M, Liu J, Li Q, Xu B. The trichotomy of HER2 expression confers new insights into the understanding and managing for breast cancer stratified by HER2 status. Int J Cancer 2023; 153:1324-1336. [PMID: 37314204 DOI: 10.1002/ijc.34570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/26/2023] [Accepted: 04/28/2023] [Indexed: 06/15/2023]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a tyrosine kinase receptor that plays a carcinogenic role in breast cancer (BC) through gene amplification, mutation, or overexpression. Traditional methods of HER2 detection were divided into positive (immunohistochemistry (IHC) 3+/fluorescence in situ hybridization (FISH) amplification) and negative (IHC 2+/FISH-, IHC 1+, IHC 0) according to the dichotomy method. Anti-HER2-targeted therapies, such as trastuzumab and pertuzumab, have significantly improved the prognosis of HER2-positive patients. However, up to 75% to 85% of patients remain HER2-negative. In recent years, with the rapid development of molecular biology, gene detection technology, targeted therapy, and immunotherapy, researchers have actively explored the clinicopathological characteristics, molecular biological characteristics, treatment methods, and HER2 detection methods of HER2-low/zero breast cancer. With the clinical efficacy of new anti-HER2 targeted drugs, accurate classification of breast cancer is very important for the treatment choice. Therefore, the following review summarizes the necessity of developing HER2 detection methods, and the clinicopathological and drug treatment characteristics of patients with HER2-low/zero, to light the dawn of the treatment of breast cancer patients with HER2-low/zero expression.
Collapse
Affiliation(s)
- Mingxia Jiang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiaxuan Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiao Li
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
14
|
D’Arienzo A, Verrazzo A, Pagliuca M, Napolitano F, Parola S, Viggiani M, Caputo R, Puglisi F, Giuliano M, Del Mastro L, Arpino G, De Laurentiis M, Montemurro F. Toxicity profile of antibody-drug conjugates in breast cancer: practical considerations. EClinicalMedicine 2023; 62:102113. [PMID: 37554126 PMCID: PMC10404866 DOI: 10.1016/j.eclinm.2023.102113] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 08/10/2023] Open
Abstract
Antibody-drug conjugates (ADCs) represent a novel and evolving class of antineoplastic agents, constituted by monoclonal antibody linked to biologically active drugs, delivering cytotoxic compounds at the tumor site, reducing the likelihood of systemic exposure and toxicity. They are generally well tolerated, nevertheless some predictable adverse reactions need careful monitoring and timely approach. These include neutropenia, nausea and vomiting, alopecia, diarrhea, left ventricular dysfunction, ILD/pneumonitis. The mechanisms leading to drug-associated toxicities are summarized, and prophylaxis protocols and appropriate management strategies are proposed, based on current literature. This review aims to collect the most updated evidence on toxicities potentially occurring during breast cancer treatment with approved or under clinical investigation (advanced stage) ADCs. A focus is dedicated to monitoring protocols and clinical management, aimed at preventing and/or promptly address relevant problems, in order to avoid premature discontinuation or improper dose reduction.
Collapse
Affiliation(s)
- Andrea D’Arienzo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Annarita Verrazzo
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Via Mezzocannone 4, Naples 80138, Italy
| | - Martina Pagliuca
- Scuola Superiore Meridionale (SSM), Clinical and Translational Oncology, Via Mezzocannone 4, Naples 80138, Italy
- Molecular Predictors and New Targets in Oncology Unit 981, Gustave Roussy, 114 Rue Édouard-Vaillant, Villejuif 94805, France
| | - Fabiana Napolitano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Sara Parola
- Oncology Unit, PO di San Felice a Cancello, Via Roma 349, San Felice a Cancello, Caserta 81021, Italy
| | - Martina Viggiani
- Department of Oncology, HFR Fribourg-Cantonal Hospital, Chemin des Pensionnats 2-6, Fribourg 1708, Switzerland
| | - Roberta Caputo
- Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Via Mariano Semmola 52, Naples 80131, Italy
| | - Fabio Puglisi
- Department of Medicine, University of Udine, Via Palladio 8, Udine 33100, Italy
- Department of Medical Oncology, CRO Aviano, National Cancer Institute, IRCCS, Via Franco Gallini 2, Aviano, Pordenone 33081, Italy
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Lucia Del Mastro
- Department of Medical Oncology, UO Oncologia Medica 2, IRCCS Ospedale Policlinico San Martino, Largo R. Benzi 10, Genova 16132, Italy
| | - Grazia Arpino
- Department of Clinical Medicine and Surgery, University of Naples “Federico II”, Via Sergio Pansini 5, Naples 80131, Italy
| | - Michelino De Laurentiis
- Division of Breast Medical Oncology, Istituto Nazionale Tumori IRCCS “Fondazione G. Pascale”, Via Mariano Semmola 52, Naples 80131, Italy
| | - Filippo Montemurro
- Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142 -KM 3.95, Candiolo, Torino 10060, Italy
| |
Collapse
|
15
|
Yang C, Brezden-Masley C, Joy AA, Sehdev S, Modi S, Simmons C, Henning JW. Targeting HER2-low in metastatic breast cancer: an evolving treatment paradigm. Ther Adv Med Oncol 2023; 15:17588359231175440. [PMID: 37323186 PMCID: PMC10262633 DOI: 10.1177/17588359231175440] [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: 01/19/2023] [Accepted: 04/25/2023] [Indexed: 06/17/2023] Open
Abstract
The results of the Phase III DESTINY-Breast04 trial of trastuzumab deruxtecan (T-DXd) are leading to a shift in both the classification and treatment of human epidermal growth factor receptor 2 (HER2)-negative metastatic breast cancer. In this trial, T-DXd was associated with a substantial survival benefit among patients with hormone receptor-positive and hormone receptor-negative disease and low expression of HER2, a biomarker previously considered unactionable in this treatment setting. Herein, we discuss the evolving therapeutic pathway for HER2-low disease, ongoing clinical trials, and the potential challenges and evidence gaps arising with treatment of this patient population.
Collapse
Affiliation(s)
- Charlie Yang
- Tom Baker Cancer Centre, University of Calgary, 1331 29 Street NW, Calgary, AB T2N 4N2, Canada
| | | | - Anil Abraham Joy
- Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| | | | - Shanu Modi
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christine Simmons
- BC Cancer Agency – Vancouver Centre, University of British Columbia, Vancouver, BC, Canada
| | | |
Collapse
|
16
|
Wang J, Liao D, Zhang X, Miao C, Chen K. Can Patients with HER2-Low Breast Cancer Benefit from Anti-HER2 Therapies? A Review. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:281-294. [PMID: 37113514 PMCID: PMC10128871 DOI: 10.2147/bctt.s407181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023]
Abstract
Breast cancer (BC) poses a severe threat to the health of women worldwide. Currently, different therapeutic regimens are used for BC according to the pathological classification of HER2-positive or HER2-negative. Clinical reports of HER2-low expression indicate that the condition is HER2-negative, which was ineligible for HER2-targeted therapy. In contrast to HER2-zero tumors, however, HER2-low BC is a heterogeneous disease with unique genetic characteristics, prognoses, and different therapeutic responses. Clinical efficacy has been demonstrated by numerous potent and innovative anti-HER2 medications, particularly antibody-drug conjugates (ADCs). Certain ADCs, including T-DXd, have demonstrated good efficacy in some trials either used alone or in conjunction with other medications. To enhance outcomes in individuals with HER2-low BC, immunotherapy and other treatments are frequently combined with HER2-targeted therapy. There are also alternative strategies that target both HER2 and HER3 or other antigenic sites. We hope more individuals with HER2-low BC will benefit from more precise treatment regimens in the future. This article provides a review of existing research and clinical trials.
Collapse
Affiliation(s)
- Jin Wang
- Department of Emergency, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Dongying Liao
- Department of Emergency, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Xuemin Zhang
- Department of Emergency, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Changhong Miao
- Department of Emergency, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| | - Kuang Chen
- Department of Emergency, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People’s Republic of China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, People’s Republic of China
- Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, People’s Republic of China
| |
Collapse
|
17
|
LINC00589-dominated ceRNA networks regulate multiple chemoresistance and cancer stem cell-like properties in HER2 + breast cancer. NPJ Breast Cancer 2022; 8:115. [PMID: 36309503 PMCID: PMC9617889 DOI: 10.1038/s41523-022-00484-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 10/12/2022] [Indexed: 11/29/2022] Open
Abstract
Resistance to human epidermal growth factor receptor 2 (HER2)-targeted therapy (trastuzumab), cancer stem cell (CSC)-like properties and multiple chemoresistance often concur and intersect in breast cancer, but molecular links that may serve as effective therapeutic targets remain largely unknown. Here, we identified the long noncoding RNA, LINC00589 as a key regulatory node for concurrent intervention of these processes in breast cancer cells in vitro and in vivo. We demonstrated that the expression of LINC00589 is clinically valuable as an independent prognostic factor for discriminating trastuzumab responders. Mechanistically, LINC00589 serves as a ceRNA platform that simultaneously sponges miR-100 and miR-452 and relieves their repression of tumor suppressors, including discs large homolog 5 (DLG5) and PR/SET domain 16 (PRDM16, a transcription suppressor of mucin4), thereby exerting multiple cancer inhibitory functions and counteracting drug resistance. Collectively, our results disclose two LINC00589-initiated ceRNA networks, the LINC00589-miR-100-DLG5 and LINC00589-miR-452-PRDM16- mucin4 axes, which regulate trastuzumab resistance, CSC-like properties and multiple chemoresistance of breast cancer, thus providing potential diagnostic and prognostic markers and therapeutic targets for HER2-positive breast cancer.
Collapse
|
18
|
Li Y, Liao X, Ma L. ERCC1 is a potential biomarker for predicting prognosis, immunotherapy, chemotherapy efficacy, and expression validation in HER2 over-expressing breast cancer. Front Oncol 2022; 12:955719. [PMID: 36338712 PMCID: PMC9631216 DOI: 10.3389/fonc.2022.955719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/10/2022] [Indexed: 11/22/2022] Open
Abstract
Objective To investigate the relationship between Excision repair cross-complementation 1 (ERCC1) expression, clinicopathological features, and breast cancer prognosis in patients treated with trastuzumab. Further, we aim to explore the immune status of ERCC1 in breast cancer. Methods The data were retrieved from publicly available databases like the Cancer Genome Atlas, Therapeutically Applicable Research to Generate Effective Treatments, and the Genotype-Tissue Expression. The data was used to perform differential expression analyses between tumor and normal tissues in pan-cancers, immune-related analysis, homologous recombination deficiency (HRD), tumor mutation burden, and microsatellite instability. A total of 210 patients with HER2 over-expressing breast cancer from the Fourth Hospital of Hebei Medical University between January 2013 to December 2015 were enrolled in the study. Ten adjacent normal tissues were used to study the expression pattern of ERCC1 in normal tissues. Immunohistochemistry was performed to study ERCC1 expression and immune cell infiltration in different status of ERCC1 expression. Further, the correlation between ERCC1 expression, immune cell infiltration clinicopathological features, and the prognosis of patients with breast cancer was analyzed. Results The immune analysis revealed a significant correlation between CD8+ T cell, CD4+ T cell, T helper cell, macrophages, mast cells, and ERCC1 expression. Spearman analysis show that ERCC1 expression is related to macrophages and T cells. A close correlation was observed between increased ERCC1 expression and high tumor immune dysfunction and exclusion (TIDE) score as well as HRD. The results revealed a significant correlation among ERCC1, chemotherapy and estrogen receptor (ER; P < 0.05) expression. Univariate survival analysis revealed a significant correlation (P < 0.05) between that ERCC1 and ER expression, blood vessel invasion, and disease-free survival (DFS). ERCC1 and ER expression, tumor size, blood vessel invasion, pathological type, and lymph node metastases significantly correlated (P < 0.05) with overall survival in patients. Multivariate regression analysis revealed that ERCC1 expression and chemotherapy were independent factors that influence DFS. ERCC1 expression and vascular tumor thrombus were independent influencing factors that influence OS. Conclusion A correlation was observed between high ERCC1 expression and poor patient prognosis. High ERCC1 expression also influences the efficacy of immunotherapy and chemotherapy.
Collapse
|
19
|
Michmerhuizen AR, Lerner LM, Ward C, Pesch AM, Zhang A, Schwartz R, Wilder-Romans K, Eisner JR, Rae JM, Pierce LJ, Speers CW. Androgen and oestrogen receptor co-expression determines the efficacy of hormone receptor-mediated radiosensitisation in breast cancer. Br J Cancer 2022; 127:927-936. [PMID: 35618789 PMCID: PMC9427858 DOI: 10.1038/s41416-022-01849-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 04/26/2022] [Accepted: 05/06/2022] [Indexed: 11/08/2022] Open
Abstract
PURPOSE Radiation therapy (RT) and hormone receptor (HR) inhibition are used for the treatment of HR-positive breast cancers; however, little is known about the interaction of the androgen receptor (AR) and estrogen receptor (ER) in response to RT in AR-positive, ER-positive (AR+/ER+) breast cancers. Here we assessed radiosensitisation of AR+/ER+ cell lines using pharmacologic or genetic inhibition/degradation of AR and/or ER. METHODS Radiosensitisation was assessed with AR antagonists (enzalutamide, apalutamide, darolutamide, seviteronel, ARD-61), ER antagonists (tamoxifen, fulvestrant) or using knockout of AR. RESULTS Treatment with AR antagonists or ER antagonists in combination with RT did not result in radiosensitisation changes (radiation enhancement ratios [rER]: 0.76-1.21). Fulvestrant treatment provided significant radiosensitisation of CAMA-1 and BT-474 cells (rER: 1.06-2.0) but not ZR-75-1 cells (rER: 0.9-1.11). Combining tamoxifen with enzalutamide did not alter radiosensitivity using a 1 h or 1-week pretreatment (rER: 0.95-1.14). Radiosensitivity was unchanged in AR knockout compared to Cas9 cells (rER: 1.07 ± 0.11), and no additional radiosensitisation was achieved with tamoxifen or fulvestrant compared to Cas9 cells (rER: 0.84-1.19). CONCLUSION While radiosensitising in AR + TNBC, AR inhibition does not modulate radiation sensitivity in AR+/ER+ breast cancer. The efficacy of ER antagonists in combination with RT may also be dependent on AR expression.
Collapse
Affiliation(s)
- Anna R Michmerhuizen
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
| | - Lynn M Lerner
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Connor Ward
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Andrea M Pesch
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
| | - Amanda Zhang
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Rachel Schwartz
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Kari Wilder-Romans
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | | | - James M Rae
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
- Department of Pharmacology, University of Michigan, Ann Arbor, MI, USA
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Lori J Pierce
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA
| | - Corey W Speers
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA.
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
20
|
Koirala N, Dey N, Aske J, De P. Targeting Cell Cycle Progression in HER2+ Breast Cancer: An Emerging Treatment Opportunity. Int J Mol Sci 2022; 23:6547. [PMID: 35742993 PMCID: PMC9224522 DOI: 10.3390/ijms23126547] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 06/09/2022] [Accepted: 06/09/2022] [Indexed: 11/16/2022] Open
Abstract
The development of HER2-targeted therapies has dramatically improved patient survival and patient management and increased the quality of life in the HER2+ breast cancer patient population. Due to the activation of compensatory pathways, patients eventually suffer from resistance to HER2-directed therapies and develop a more aggressive disease phenotype. One of these mechanisms is the crosstalk between ER and HER2 signaling, especially the CDK4/6-Cyclin D-Rb signaling axis that is commonly active and has received attention for its potential role in regulating tumor progression. CDK 4/6 inhibitors interfere with the binding of cell-cycle-dependent kinases (CDKs) with their cognate partner cyclins, and forestall the progression of the cell cycle by preventing Rb phosphorylation and E2F release that consequentially leads to cancer cell senescence. CDK 4/6 inhibitors, namely, palbociclib, ribociclib, and abemaciclib, in combination with anti-estrogen therapies, have shown impressive outcomes in hormonal receptor-positive (HR+) disease and have received approval for this disease context. As an extension of this concept, preclinical/clinical studies incorporating CDK 4/6 inhibitors with HER2-targeted drugs have been evaluated and have shown potency in limiting tumor progression, restoring therapeutic sensitivity, and may improving the management of the disease. Currently, several clinical trials are examining the synergistic effects of CDK 4/6 inhibitors with optimized HER2-directed therapies for the (ER+/-) HER2+ population in the metastatic setting. In this review, we aim to interrogate the burden of HER2+ disease in light of recent treatment progress in the field and examine the clinical benefit of CDK 4/6 inhibitors as a replacement for traditional chemotherapy to improve outcomes in HER2+ breast cancer.
Collapse
Affiliation(s)
| | | | | | - Pradip De
- Translational Oncology Laboratory, Avera Cancer Institute, Sioux Falls, SD 57105, USA; (N.K.); (N.D.); (J.A.)
| |
Collapse
|