1
|
Kiraz U, Rewcastle E, Fykse SK, Lundal I, Gudlaugsson EG, Skaland I, Søiland H, Baak JPA, Janssen EAM. Dual Functions of Androgen Receptor Overexpression in Triple-Negative Breast Cancer: A Complex Prognostic Marker. Bioengineering (Basel) 2025; 12:54. [PMID: 39851328 PMCID: PMC11761274 DOI: 10.3390/bioengineering12010054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/06/2025] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
A subset of triple-negative breast cancer (TNBC) expresses the androgen receptor (AR), but thresholds for AR positivity and its clinical significance vary. We hypothesize that objective assessment outperforms subjective methods, and that high AR negatively impacts prognosis. In a population-based TNBC cohort (n = 198) with long follow-up (4-383 months), AR expression was evaluated via subjective scoring (AR-Manual) and automated digital image analysis (AR-DIA). A 10% cut-off value via AR-DIA was the strongest negative prognostic threshold for distant metastases (p = 0.008). High AR-DIA correlated with lower grade (p = 0.014), and lower proliferation (p = 0.004) but also with larger tumors (p = 0.047), distant metastasis (p = 0.052), and lymph node (LN) positivity (p < 0.001), highlighting its dual roles. Multivariate analysis revealed interaction between LN status and AR-DIA (p < 0.001) as the strongest prognostic factor, followed by fibrotic focus (FF; p = 0.009), mitotic activity index (MAI; p = 0.018), and stromal tumor-infiltrating lymphocytes (sTILs; p = 0.041). AR-DIA had no additional prognostic value in favorable subgroups but was significant in unfavorable subgroups. In high AR-DIA patients with unfavorable characteristics, ACT did not improve survival, and patients may benefit from AR-targeted therapy. Overall, the DIA method provides reproducibility, high AR-DIA (≥10%) shows opposing survival effects in different TNBC subgroups, and AR evaluation is crucial for prognosis and AR-targeted therapies.
Collapse
Affiliation(s)
- Umay Kiraz
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021 Stavanger, Norway
| | - Emma Rewcastle
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Silja K. Fykse
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Ingrid Lundal
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Einar G. Gudlaugsson
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Ivar Skaland
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Håvard Søiland
- Department of Research, Stavanger University Hospital, 4011 Stavanger, Norway
| | - Jan P. A. Baak
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
| | - Emiel A. M. Janssen
- Department of Pathology, Stavanger University Hospital, 4011 Stavanger, Norway; (E.R.); (S.K.F.); (I.L.); (E.G.G.); (I.S.); (J.P.A.B.); (E.A.M.J.)
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, 4021 Stavanger, Norway
- Institute for Biomedicine and Glycomics, Griffith University, Queensland, QLD 4215, Australia
| |
Collapse
|
2
|
Rad SK, Yeo KKL, Li R, Wu F, Liu S, Nourmohammadi S, Murphy WM, Tomita Y, Price TJ, Ingman WV, Townsend AR, Smith E. Enhancement of Doxorubicin Efficacy by Bacopaside II in Triple-Negative Breast Cancer Cells. Biomolecules 2025; 15:55. [PMID: 39858449 PMCID: PMC11762400 DOI: 10.3390/biom15010055] [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: 12/04/2024] [Revised: 12/27/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is an aggressive subtype with limited treatment options and high resistance to chemotherapy. Doxorubicin is commonly used, but its efficacy is limited by variable sensitivity and resistance. Bacopaside II, a saponin compound, has shown anti-cancer potential. This study evaluates the effects of doxorubicin and bacopaside II, both individually and in combination, across TNBC subtypes to explore mechanisms of resistance and enhanced drug efficacy. METHODS The growth-inhibitory effects of doxorubicin and bacopaside II were assessed in four TNBC cell lines. IC50 values were determined using dose-response assays, and doxorubicin accumulation was measured via spectral flow cytometry. ATP-binding cassette (ABC) transporter expression (ABCB1, ABCC1, ABCC3, and ABCG2) was analyzed for correlations with drug sensitivity. In silico docking assessed the binding affinity of bacopaside II to ABC transporters. A 3D culture model simulated drug-resistant TNBC, and combination effects were evaluated with live-cell imaging. RESULTS Doxorubicin sensitivity varied across TNBC molecular subtypes, correlating to intracellular accumulation. Bacopaside II inhibited growth across subtypes, inducing apoptosis in sensitive cells and necrosis in resistant cells. Bacopaside II increased doxorubicin accumulation, independent of P-glycoprotein (ABCB1), possibly through interactions with other ABC transporters. In drug-resistant 3D cultures, bacopaside II maintained efficacy and enhanced doxorubicin accumulation, counteracting ABC transporter-mediated resistance. The doxorubicin and bacopaside II combination showed synergistic growth inhibition. CONCLUSIONS Bacopaside II enhances doxorubicin efficacy in TNBC by increasing drug accumulation and overcoming ABC transporter-mediated resistance, suggesting its potential as an adjuvant in TNBC treatment. These findings support further investigation of bacopaside II, particularly for resistant TNBC subtypes.
Collapse
Affiliation(s)
- Sima Kianpour Rad
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia; (S.K.R.); (K.K.L.Y.); (R.L.); (F.W.); (S.L.); (S.N.); (Y.T.); (T.J.P.); (A.R.T.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - Kenny K. L. Yeo
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia; (S.K.R.); (K.K.L.Y.); (R.L.); (F.W.); (S.L.); (S.N.); (Y.T.); (T.J.P.); (A.R.T.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - Runhao Li
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia; (S.K.R.); (K.K.L.Y.); (R.L.); (F.W.); (S.L.); (S.N.); (Y.T.); (T.J.P.); (A.R.T.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - Fangmeinuo Wu
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia; (S.K.R.); (K.K.L.Y.); (R.L.); (F.W.); (S.L.); (S.N.); (Y.T.); (T.J.P.); (A.R.T.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - Saifei Liu
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia; (S.K.R.); (K.K.L.Y.); (R.L.); (F.W.); (S.L.); (S.N.); (Y.T.); (T.J.P.); (A.R.T.)
| | - Saeed Nourmohammadi
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia; (S.K.R.); (K.K.L.Y.); (R.L.); (F.W.); (S.L.); (S.N.); (Y.T.); (T.J.P.); (A.R.T.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia;
| | - William M. Murphy
- Department of Surgery-Otolaryngology Head and Neck Surgery, The University of Adelaide and the Basil Hetzel Institute for Translational Health Research, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia;
| | - Yoko Tomita
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia; (S.K.R.); (K.K.L.Y.); (R.L.); (F.W.); (S.L.); (S.N.); (Y.T.); (T.J.P.); (A.R.T.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia;
- Medical Oncology, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia
| | - Timothy J. Price
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia; (S.K.R.); (K.K.L.Y.); (R.L.); (F.W.); (S.L.); (S.N.); (Y.T.); (T.J.P.); (A.R.T.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia;
- Medical Oncology, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia
| | - Wendy V. Ingman
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia;
- Robinson Research Institute, The University of Adelaide, Adelaide, SA 5005, Australia
| | - Amanda R. Townsend
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia; (S.K.R.); (K.K.L.Y.); (R.L.); (F.W.); (S.L.); (S.N.); (Y.T.); (T.J.P.); (A.R.T.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia;
- Medical Oncology, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia
| | - Eric Smith
- Solid Tumour Group, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia; (S.K.R.); (K.K.L.Y.); (R.L.); (F.W.); (S.L.); (S.N.); (Y.T.); (T.J.P.); (A.R.T.)
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia;
- Discipline of Surgery, Basil Hetzel Institute for Translational Health Research, The Queen Elizabeth Hospital, Central Adelaide Local Health Network, Woodville South, Adelaide, SA 5011, Australia
| |
Collapse
|
3
|
Garcia V, Gardecki E, Jou S, Li X, Shroyer KR, Saltz J, Acs B, Elfer K, Lennerz J, Salgado R, Gallas BD. Prioritizing cases from a multi-institutional cohort for a dataset of pathologist annotations. J Pathol Inform 2025; 16:100411. [PMID: 39720416 PMCID: PMC11667696 DOI: 10.1016/j.jpi.2024.100411] [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] [Received: 07/25/2024] [Revised: 11/07/2024] [Accepted: 11/12/2024] [Indexed: 12/26/2024] Open
Abstract
Objective With the increasing energy surrounding the development of artificial intelligence and machine learning (AI/ML) models, the use of the same external validation dataset by various developers allows for a direct comparison of model performance. Through our High Throughput Truthing project, we are creating a validation dataset for AI/ML models trained in the assessment of stromal tumor-infiltrating lymphocytes (sTILs) in triple negative breast cancer (TNBC). Materials and methods We obtained clinical metadata for hematoxylin and eosin-stained glass slides and corresponding scanned whole slide images (WSIs) of TNBC core biopsies from two US academic medical centers. We selected regions of interest (ROIs) from the WSIs to target regions with various tissue morphologies and sTILs densities. Given the selected ROIs, we implemented a hierarchical rank-sort method for case prioritization. Results We received 122 glass slides and clinical metadata on 105 unique patients with TNBC. All received cases were female, and the mean age was 63.44 years. 60% of all cases were White patients, and 38.1% were Black or African American. After case prioritization, the skewness of the sTILs density distribution improved from 0.60 to 0.46 with a corresponding increase in the entropy of the sTILs density bins from 1.20 to 1.24. We retained cases with less prevalent metadata elements. Conclusion This method allows us to prioritize underrepresented subgroups based on important clinical factors. In this manuscript, we discuss how we sourced the clinical metadata, selected ROIs, and developed our approach to prioritizing cases for inclusion in our pivotal study.
Collapse
Affiliation(s)
- Victor Garcia
- U.S. Food and Drug Administration, Center for Devices and Radiological Health, Office of Science and Engineering Laboratories, Division of Imaging, Diagnostics, and Software Reliability, Silver Spring, MD, United States of America
| | - Emma Gardecki
- U.S. Food and Drug Administration, Center for Devices and Radiological Health, Office of Science and Engineering Laboratories, Division of Imaging, Diagnostics, and Software Reliability, Silver Spring, MD, United States of America
| | - Stephanie Jou
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, United States of America
| | - Xiaoxian Li
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, GA, United States of America
| | - Kenneth R. Shroyer
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States of America
| | - Joel Saltz
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, NY, United States of America
| | - Balazs Acs
- Department of Oncology and Pathology, Cancer Centre Karolinska (CCK), Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Katherine Elfer
- U.S. Food and Drug Administration, Center for Devices and Radiological Health, Office of Science and Engineering Laboratories, Division of Imaging, Diagnostics, and Software Reliability, Silver Spring, MD, United States of America
- Division of Cancer Prevention, National Cancer Institute, National Institute of Health, Shady Grove, MD, United States of America
| | | | - Roberto Salgado
- Division of Research, Peter Mac Callum Cancer Centre, Melbourne, Australia
- Department of Pathology, ZAS Hospitals, Antwerp, Belgium
| | - Brandon D. Gallas
- U.S. Food and Drug Administration, Center for Devices and Radiological Health, Office of Science and Engineering Laboratories, Division of Imaging, Diagnostics, and Software Reliability, Silver Spring, MD, United States of America
| |
Collapse
|
4
|
Chaudhary A, Patil P, Raina P, Kaul-Ghanekar R. Matairesinol repolarizes M2 macrophages to M1 phenotype to induce apoptosis in triple-negative breast cancer cells. Immunopharmacol Immunotoxicol 2024:1-15. [PMID: 39722605 DOI: 10.1080/08923973.2024.2425028] [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: 06/05/2024] [Accepted: 10/27/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Triple-Negative Breast Cancer (TNBC), the most challenging subtype of Breast Cancer (BC), currently lacks targeted therapy, presenting a significant therapeutic gap in its management. Tumor Associated Macrophages (TAMs) play a significant role in TNBC progression and could be targeted by repolarizing them from M2 to M1 phenotype. Matairesinol (MAT), a plant lignan, has been shown to exhibit anticancer, anti-inflammatory and immunomodulatory activities. In this study, we explored how MAT-induced repolarization of THP-1-derived M2 macrophages towards the M1 phenotype, which could effectively target the TNBC cell line, MDA-MB-231. METHODS The differential expression of genes in THP-1-derived macrophages at mRNA levels was evaluated by RNAseq assay. An inverted microscope equipped with a CMOS camera was utilized to capture the morphological variations in THP-1 cells and THP-1-derived macrophages. Relative mRNA expression of M1 and M2 specific marker genes was quantified by qRT-PCR. Cell viability and induction of apoptosis were evaluated by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) and 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethylbenzimidazolylcarbocyanine iodide (JC-1 dye) assays, respectively. RESULTS MAT reduced the viability of M2a and M2d macrophages and repolarized them to M1 phenotype. Conditioned medium (CM) from MAT-treated M2a and M2d macrophages significantly reduced the viability of TNBC cells by apoptosis. CONCLUSION Targeting M2 macrophages is an important strategy to regulate cancer progression. Our study provides evidence that MAT may be a promising drug candidate for developing novel anti-TNBC therapy. However, further studies are warranted to thoroughly elucidate the molecular mechanism of action of MAT and evaluate its therapeutic potential in TNBC in vitro and in vivo models.
Collapse
Affiliation(s)
- Amol Chaudhary
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Prajakta Patil
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
| | - Prerna Raina
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Analytical Department (ADT), Lupin Limited, Pune, India
| | - Ruchika Kaul-Ghanekar
- Cancer Research Lab, Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth (Deemed to be University), Pune, India
- Symbiosis Centre for Research and Innovation (SCRI); Symbiosis International Deemed University (SIU), Pune, India
- Cancer Research Lab, Symbiosis School of Biological Sciences (SSBS), Symbiosis International Deemed University (SIU), Pune, India
| |
Collapse
|
5
|
Li Q, Teodoro G, Jiang Y, Kong J. NACNet: A histology context-aware transformer graph convolution network for predicting treatment response to neoadjuvant chemotherapy in Triple Negative Breast Cancer. Comput Med Imaging Graph 2024; 118:102467. [PMID: 39591709 DOI: 10.1016/j.compmedimag.2024.102467] [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: 06/18/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/28/2024]
Abstract
Neoadjuvant chemotherapy (NAC) response prediction for triple negative breast cancer (TNBC) patients is a challenging task clinically as it requires understanding complex histology interactions within the tumor microenvironment (TME). Digital whole slide images (WSIs) capture detailed tissue information, but their giga-pixel size necessitates computational methods based on multiple instance learning, which typically analyze small, isolated image tiles without the spatial context of the TME. To address this limitation and incorporate TME spatial histology interactions in predicting NAC response for TNBC patients, we developed a histology context-aware transformer graph convolution network (NACNet). Our deep learning method identifies the histopathological labels on individual image tiles from WSIs, constructs a spatial TME graph, and represents each node with features derived from tissue texture and social network analysis. It predicts NAC response using a transformer graph convolution network model enhanced with graph isomorphism network layers. We evaluate our method with WSIs of a cohort of TNBC patient (N=105) and compared its performance with multiple state-of-the-art machine learning and deep learning models, including both graph and non-graph approaches. Our NACNet achieves 90.0% accuracy, 96.0% sensitivity, 88.0% specificity, and an AUC of 0.82, through eight-fold cross-validation, outperforming baseline models. These comprehensive experimental results suggest that NACNet holds strong potential for stratifying TNBC patients by NAC response, thereby helping to prevent overtreatment, improve patient quality of life, reduce treatment cost, and enhance clinical outcomes, marking an important advancement toward personalized breast cancer treatment.
Collapse
Affiliation(s)
- Qiang Li
- Department of Mathematics and Statistics, Georgia State University, Atlanta, 30303, GA, USA.
| | - George Teodoro
- Department of Computer Science, Federal University of Minas Gerais, Belo Horizonte, 31270, Minas Gerais, Brazil.
| | - Yi Jiang
- Department of Mathematics and Statistics, Georgia State University, Atlanta, 30303, GA, USA; Winship Cancer Institute, Emory University, Atlanta, 30322, GA, USA.
| | - Jun Kong
- Department of Mathematics and Statistics, Georgia State University, Atlanta, 30303, GA, USA; Department of Computer Science, Georgia State University, Atlanta, 30303, GA, USA; Winship Cancer Institute, Emory University, Atlanta, 30322, GA, USA.
| |
Collapse
|
6
|
Kujala J, Tengström M, Heikkinen S, Taipale M, Kosma VM, Hartikainen JM, Mannermaa A. Circulating Micro-RNAs Predict the Risk of Recurrence in Triple-Negative Breast Cancer. Cells 2024; 13:1884. [PMID: 39594632 PMCID: PMC11593277 DOI: 10.3390/cells13221884] [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: 10/01/2024] [Revised: 11/06/2024] [Accepted: 11/10/2024] [Indexed: 11/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer with a high tendency for developing a recurrent disease. Circulating micro-RNAs (cmiRNAs) obtained through liquid biopsy are potential prognostic biomarkers for the assessment of TNBC recurrence risk. In this study, we sequenced cmiRNAs from the serum samples of 14 recurrent and 19 non-recurrent TNBC cases and compared expression profiles in relation to recurrence status, survival data and miRNA expression in matched tumor samples. Differential expression analysis between recurrent and non-recurrent cases identified ten differentially expressed (DE) cmiRNAs, of which cmiRNAs miR-21-5p (p = 0.030, HR = 1.87, 95% CI 1.06-3.30), miR-16-5p (p = 0.032, HR = 0.53, 95% CI 0.30-0.95), and miR-26b-5p (p = 0.023, HR = 0.52, 95% CI 0.29-0.91) were associated with recurrence-free survival in multivariable survival analysis. Expression profiles of matched tumor and serum samples were shown to correlate with each other. DE cmiRNAs were associated with common cancer-related signaling pathways and improved the overall predictive performance of the logistic regression model assessing the recurrence risk. Our results indicate that recurrent and non-recurrent TNBC differ in their cmiRNA expression profiles, and that three specific cmiRNAs can be used to assess the risk of recurrence in TNBC.
Collapse
Affiliation(s)
- Jouni Kujala
- Institute of Clinical Medicine, Clinical Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (V.-M.K.)
| | - Maria Tengström
- Cancer Center, Department of Oncology, Kuopio University Hospital, FI-70029 Kuopio, Finland;
| | - Sami Heikkinen
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (S.H.); (M.T.)
| | - Mari Taipale
- Institute of Biomedicine, School of Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (S.H.); (M.T.)
- A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Veli-Matti Kosma
- Institute of Clinical Medicine, Clinical Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (V.-M.K.)
- Biobank of Eastern Finland, Kuopio University Hospital, FI-70029 Kuopio, Finland
| | - Jaana M. Hartikainen
- Institute of Clinical Medicine, Clinical Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (V.-M.K.)
- Genome Center of Eastern Finland, Institute of Clinical Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
- Multidisciplinary Cancer Research Community (Cancer RC), University of Eastern Finland, FI-70211 Kuopio, Finland
| | - Arto Mannermaa
- Institute of Clinical Medicine, Clinical Pathology and Forensic Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland; (J.K.); (V.-M.K.)
- Biobank of Eastern Finland, Kuopio University Hospital, FI-70029 Kuopio, Finland
- Multidisciplinary Cancer Research Community (Cancer RC), University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
7
|
Edmonds CE, O'Brien SR, McDonald ES, Mankoff DA, Pantel AR. PET Imaging of Breast Cancer: Current Applications and Future Directions. JOURNAL OF BREAST IMAGING 2024; 6:586-600. [PMID: 39401324 DOI: 10.1093/jbi/wbae053] [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: 04/23/2024] [Indexed: 11/07/2024]
Abstract
As molecular imaging use expands for patients with breast cancer, it is important for breast radiologists to have a basic understanding of molecular imaging, including PET. Although breast radiologists may not directly interpret such studies, basic knowledge of molecular imaging will enable the radiologist to better direct diagnostic workup of patients as well as discuss diagnostic imaging with the patient and other treating physicians. Several new tracers are now available to complement imaging glucose metabolism with FDG. Because it provides a noninvasive assessment of disease status across the whole body, PET offers specific advantages over tissue-based assays. Paired with targeted therapy, molecular imaging has the potential to guide personalized treatment of breast cancer, including guiding dosing during drug trials as well as predicting and assessing clinical response. This review discusses the current established applications of FDG, which remains the most widely used PET radiotracer for malignancy, including breast cancer, and highlights potential areas for expanded use based on recent research. It also summarizes research to date on the U.S. Food and Drug Administration (FDA)-approved PET tracer 16α-18F-fluoro-17β-estradiol (FES), which targets ER, including the current guidelines from the Society of Nuclear Medicine and Molecular Imaging on the appropriate use of FES-PET/CT for breast cancer as well as areas of active investigation for other potential applications. Finally, the review highlights several of the most promising novel PET tracers that are poised for clinical translation in the near future.
Collapse
Affiliation(s)
- Christine E Edmonds
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Sophia R O'Brien
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth S McDonald
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - David A Mankoff
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Austin R Pantel
- Department of Radiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
8
|
Jin LL, Lu HJ, Shao JK, Wang Y, Lu SP, Huang BF, Hu GN, Jin HC, Wang CQ. Relevance and mechanism of STAT3/miR-221-3p/Fascin-1 axis in EGFR TKI resistance of triple-negative breast cancer. Mol Cell Biochem 2024; 479:3037-3047. [PMID: 38145448 DOI: 10.1007/s11010-023-04907-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 11/25/2023] [Indexed: 12/26/2023]
Abstract
The epidermal growth factor receptor 1 (EGFR) plays a crucial role in the progression of various malignant tumors and is considered a potential target for treating triple-negative breast cancer (TNBC). However, the effectiveness of representative tyrosine kinase inhibitors (TKIs) used in EGFR-targeted therapy is limited in TNBC patients. In our study, we observed that the TNBC cell lines MDA-MB-231 and MDA-MB-468 exhibited resistance to Gefitinib. Treatment with Gefitinib caused an upregulation of Fascin-1 (FSCN1) protein expression and a downregulation of miR-221-3p in these cell lines. However, sensitivity to Gefitinib was significantly improved in both cell lines with either inhibition of FSCN1 expression or overexpression of miR-221-3p. Our luciferase reporter assay confirmed that FSCN1 is a target of miR-221-3p. Moreover, Gefitinib treatment resulted in an upregulation of phosphorylated signal transducer and activator of transcription 3 (p-STAT3) in MDA-MB-231 cells. Using Stattic, a small-molecule inhibitor of STAT3, we observed a significant enhancement in the inhibitory effect of Gefitinib on the growth, migration, and invasion of MDA-MB-231 cells. Additionally, Stattic treatment upregulated miR-221-3p expression and downregulated FSCN1 mRNA and protein expression. A strong positive correlation was noted between the expression of STAT3 and FSCN1 in breast cancer tissues. Furthermore, patients with high expression levels of both STAT3 and FSCN1 had a worse prognosis. Our findings suggest that elevated FSCN1 expression is linked to primary resistance to EGFR TKIs in TNBC. Moreover, we propose that STAT3 regulates the expression of miR-221-3p/FSCN1 and therefore modulates resistance to EGFR TKI therapy in TNBC. Combining EGFR TKI therapy with inhibition of FSCN1 or STAT3 may offer a promising new therapeutic option for TNBC.
Collapse
Affiliation(s)
- Lu-Lu Jin
- Department of Biomedical Sciences Laboratory, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Hua-Jun Lu
- Department of Oncological Radiotherapy, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Jun-Kang Shao
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, 60 Wu Ning Xi Road, Dongyang, Zhejiang, China
| | - Yan Wang
- Department of Medical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Shi-Ping Lu
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, 60 Wu Ning Xi Road, Dongyang, Zhejiang, China
| | - Bi-Fei Huang
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, 60 Wu Ning Xi Road, Dongyang, Zhejiang, China
| | - Gui-Nv Hu
- Department of Surgical Oncology, Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, Zhejiang, China
| | - Hong-Chuan Jin
- Laboratory of Cancer Biology, Key Laboratory of Biotherapy in Zhejiang Province, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, Hangzhou, Zhejiang, China
| | - Chao-Qun Wang
- Department of Pathology, Affiliated Dongyang Hospital of Wenzhou Medical University, 60 Wu Ning Xi Road, Dongyang, Zhejiang, China.
| |
Collapse
|
9
|
Jayan A, Sukumar JS, Fangman B, Patel T, Raghavendra AS, Liu D, Pasyar S, Rauch R, Basen-Engquist K, Tripathy D, Wang Y, Khan SS, Barcenas CH. Real-World Immune-Related Adverse Events in Patients With Early Triple-Negative Breast Cancer Who Received Pembrolizumab. JCO Oncol Pract 2024:OP2400371. [PMID: 39388649 DOI: 10.1200/op.24.00371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 08/24/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
PURPOSE The addition of pembrolizumab to chemotherapy in high-risk early triple-negative breast cancer (TNBC) improves cancer outcomes. However, pembrolizumab induces varied immune-related adverse events (irAEs) where some can be severe or lifelong. This retrospective study describes real-world patterns of irAEs in patients with TNBC who received pembrolizumab. METHODS We evaluated irAEs in patients with TNBC from a comprehensive cancer center and a community hospital who received pembrolizumab with chemotherapy between 2021 and 2023, excluding those enrolled in clinical trials. We used national guidelines to grade toxicities. Logistic regression assessed the effect of clinicopathologic variables on irAEs adjusting for covariates. RESULTS We identified 233 patients with a median age of 51 years, 62% had stage II TNBC, 35% had stage III TNBC, 25% were Hispanic, 21% were Black, and 42% were White. Eighty patients (34%) developed 100 separate irAEs. The most common irAEs were endocrinopathies (52%) and GI (23%); there were 26 grade ≥3 irAEs, which all resulted in hospitalization, the most common being GI (13 instances); 45 required systemic steroids, 16 required additional immunosuppressive therapy, and 32 patients discontinued pembrolizumab because of irAEs. Two patients who developed colitis eventually died due to complications. Most (67 instances) irAEs were unresolved at the time of last follow-up, but 55% (37/67) had improved to grade 1. No clinicopathologic factors were associated with the development or severity of irAEs. CONCLUSION In this real-world diverse population, we observed rates of irAEs comparable with KEYNOTE-522, where endocrinopathies were the most prevalent, but GI irAEs were also prevalent and severe. This emphasizes a critical issue as pembrolizumab is increasingly being used in early TNBC and could have long-term survivorship implications.
Collapse
Affiliation(s)
- Athira Jayan
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX
- Department of General Internal Medicine, MD Anderson Cancer Center, Houston, TX
| | - Jasmine S Sukumar
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX
| | - Benjamin Fangman
- Department of General Oncology, MD Anderson Cancer Center, Houston, TX
| | - Tejal Patel
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX
- Department of Biostatistics, MD Anderson Cancer Center, Houston,TX
| | | | - Diane Liu
- Department of Health Disparities Research, MD Anderson Cancer Center, Houston, TX
| | - Sarah Pasyar
- Department of Health Disparities Research, MD Anderson Cancer Center, Houston, TX
| | - Ronald Rauch
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX
| | | | - Debasish Tripathy
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX
| | - Yinghong Wang
- Department of Gastroenterology, Hepatology & Nutrition, MD Anderson Cancer Center, Houston, TX
| | - Sonya S Khan
- Department of Endocrine Neoplasia and HD, MD Anderson Cancer Center, Houston, TX
| | - Carlos H Barcenas
- Department of Breast Medical Oncology, MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
10
|
Qayoom H, Mir MA. Mutant P53 modulation by cryptolepine through cell cycle arrest and apoptosis in triple negative breast cancer. Biomed Pharmacother 2024; 179:117351. [PMID: 39216450 DOI: 10.1016/j.biopha.2024.117351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 08/13/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Triple Negative Breast cancer is an aggressive breast cancer subtype. It has a more aggressive clinical course, an earlier age of onset, a larger propensity for metastasis, and worse clinical outcomes as evidenced by a higher risk of recurrence and a shorter survival rate. Currently, the primary options for TNBC treatment are surgery, radiation, and chemotherapy. These treatments however remain ineffective due to recurrence. However, given that p53 mutations have been identified in more than 60-88 % of TNBC, translating p53 into the clinical situation is particularly important in TNBC. In this study, we screened and evaluated the therapeutic potential of cryptolepine (CRP) in TNBC in-vitro models being an anti-malarial drug it could be repurposed as an anti-cancer therapeutic targeting TNBC. Moreover, the cytotoxicity activity of cryptolepine to TNBC cells and a detailed anti-tumor mechanism in mutant P53 has not been reported before. METHODS MTT assays were used to examine the cytotoxicity and cell viability activity of Cryptolepine in TNBC, non-TNBC T47D and MCF-7 and non-malignant MCF10A cells. Scratch wound and clonogenic assay was used to evaluate the cryptolepine's effect on migration and colony forming ability of TNBC cells. Flow cytometry, MMP and DAPI was used to assess cell cycle arrest and cell apoptosis mechanism. The expression of proteins was detected by western blots. The differential expression of RNAs was evaluated by RT-PCR and the interaction between P53 and drug was evaluated computationally using in-silico approach and in-vitro using ChIP assay. RESULTS In this study, we found that cryptolepine has more preferential cytotoxicity in TNBC than non-TNBC cells. Notably, our studies revealed the mechanism by which cryptolepine induces intrinsic apoptosis and inhibit migration, colony formation ability, induce cell cycle arrest by inducing conformational change in the mutant P53 thereby increasing its DNA binding ability, hence activating its tumor suppressing potential significantly. CONCLUSION Our study revealed that CRP significantly reduced the proliferation, migration and colony forming ability of TNBC cells lines. Moreover, it was revealed that CRP induces cell cycle arrest and apoptosis by activating mutant P53 and enhancing its DNA binding ability to induce its tumor suppressing ability.
Collapse
Affiliation(s)
- Hina Qayoom
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India
| | - Manzoor A Mir
- Cancer Biology Lab, Department of Bioresources, School of Biological Sciences, University of Kashmir, Srinagar 190006, India.
| |
Collapse
|
11
|
Alaouna M, Hull R, Molefi T, Khanyile R, Mbodi L, Luvhengo TE, Chauke-Malinga N, Phakathi B, Penny C, Dlamini Z. Exploring Water-Soluble South African Tulbaghia violacea Harv Extract as a Therapeutic Approach for Triple-Negative Breast Cancer Metastasis. Curr Issues Mol Biol 2024; 46:10806-10828. [PMID: 39451522 PMCID: PMC11506433 DOI: 10.3390/cimb46100642] [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: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 10/26/2024] Open
Abstract
Triple-negative breast cancer (TNBC) accounts for approximately 20% of all breast cancer cases and is characterized by a lack of estrogen, progesterone, and human epidermal growth factor 2 receptors. Current targeted medicines have been unsuccessful due to this absence of hormone receptors. This study explored the efficacy of Tulbaghia violacea, a South African medicinal plant, for the treatment of TNBC metastasis. Extracts from T. violacea leaves were prepared using water and methanol. However, only the water-soluble extract showed anti-cancer activity and the effects of this water-soluble extract on cell adhesion, invasion, and migration, and its antioxidant activity were assessed using MCF-10A and MDA-MB-231 cells. The T. violacea extract that was soluble in water effectively decreased the movement and penetration of MDA-MB-231 cells through the basement membrane in scratch and invasion tests, while enhancing their attachment to a substance resembling an extracellular matrix. The sample showed mild-to-low antioxidant activity in the antioxidant assy. Nuclear magnetic resonance spectroscopy revealed 61 chemical components in the water-soluble extract, including DDMP, 1,2,4-triazine-3,5(2H,4H)-dione, vanillin, schisandrin, taurolidine, and α-pinene, which are known to have anti-cancer properties. An in-depth examination of the transcriptome showed alterations in genes linked to angiogenesis, metastasis, and proliferation post-treatment, with reduced activity in growth receptor signaling, angiogenesis, and cancer-related pathways, such as the Wnt, Notch, and PI3K pathways. These results indicate that T. violacea may be a beneficial source of lead chemicals for the development of potential therapeutic medicines that target TNBC metastasis. Additional studies are required to identify the precise bioactive chemical components responsible for the observed anti-cancer effects.
Collapse
Affiliation(s)
- Mohammed Alaouna
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; (M.A.); (C.P.)
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0084, South Africa; (R.H.); (T.M.); (R.K.); (N.C.-M.)
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0084, South Africa; (R.H.); (T.M.); (R.K.); (N.C.-M.)
| | - Thulo Molefi
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0084, South Africa; (R.H.); (T.M.); (R.K.); (N.C.-M.)
- Department of Medical Oncology, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0084, South Africa
| | - Richard Khanyile
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0084, South Africa; (R.H.); (T.M.); (R.K.); (N.C.-M.)
- Department of Medical Oncology, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0084, South Africa
| | - Langanani Mbodi
- Gynaecologic Oncology Unit, Department of Obstetrics and Gynaecology, Charlotte Maxeke Johannesburg Academic Hospital, University of the Witwatersrand, Johannesburg 2193, South Africa;
| | - Thifhelimbilu Emmanuel Luvhengo
- Department of Surgery, Charlotte Maxeke Johannesburg Academic Hospital, University of the Witwatersrand, Parktown, Johannesburg 2193, South Africa;
| | - Nkhensani Chauke-Malinga
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0084, South Africa; (R.H.); (T.M.); (R.K.); (N.C.-M.)
- Papillon Plastic Surgery, Suite 203B, 24 12th Avenue, Linksfield West, Johannesburg 2192, South Africa
| | - Boitumelo Phakathi
- Department of Surgery, Faculty of Health Sciences, University of Kwa-Zulu Natal, Durban 4041, South Africa;
| | - Clement Penny
- Department of Internal Medicine, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2193, South Africa; (M.A.); (C.P.)
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Pretoria 0084, South Africa; (R.H.); (T.M.); (R.K.); (N.C.-M.)
| |
Collapse
|
12
|
Manna M, Brabant M, Greene R, Chamberlain MD, Kumar A, Alimohamed N, Brezden-Masley C. Canadian Expert Recommendations on Safety Overview and Toxicity Management Strategies for Sacituzumab Govitecan Based on Use in Metastatic Triple-Negative Breast Cancer. Curr Oncol 2024; 31:5694-5708. [PMID: 39330050 PMCID: PMC11431578 DOI: 10.3390/curroncol31090422] [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: 08/10/2024] [Revised: 09/13/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024] Open
Abstract
Sacituzumab Govitecan (SG) is an antibody-drug conjugate (ADC) comprised of an anti-Trop-2 IgG1 molecule conjugated to SN-38, the active metabolite of irinotecan, via a pH-sensitive hydrolysable linker. As a result of recent Canadian funding for SG in advanced hormone receptor (HR)-positive breast cancer and triple-negative breast cancer (TNBC), experience with using SG and managing adverse events (AEs) has grown. This review presents a summary of evidence and adverse event recommendations derived from Canadian experience, with SG use in metastatic TNBC for extrapolation and guidance in all indicated settings. SG is dosed at 10 mg/kg on day 1 and day 8 of a 21-day cycle. Compared to treatment of physicians' choice (TPC) the phase III ASCENT and TROPiCS-02 studies demonstrated favorable survival data in unresectable locally advanced or metastatic TNBC and HR-positive HER2 negative metastatic breast cancer, respectively. The most common AEs were neutropenia, diarrhea, nausea, fatigue, alopecia, and anemia. This review outlines AE management recommendations for SG based on clinical trial protocols and Canadian guidelines, incorporating treatment delay, dose reductions, and the use of prophylactic and supportive medications.
Collapse
Affiliation(s)
- Mita Manna
- Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- Saskatoon Cancer Centre, Saskatchewan Cancer Agency, Saskatoon, SK S7N 4H4, Canada
| | - Michelle Brabant
- Department of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5A2, Canada
| | - Rowen Greene
- Department of Biochemistry, Microbiology, and Immunology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
| | - Michael Dean Chamberlain
- Department of Oncology, College of Medicine, University of Saskatchewan, Saskatoon, SK S7N 5E5, Canada
- Saskatoon Cancer Centre, Saskatchewan Cancer Agency, Saskatoon, SK S7N 4H4, Canada
| | - Aalok Kumar
- BC Cancer Surrey, University of British Columbia, Surrey, BC V3V 1Z2, Canada
| | - Nimira Alimohamed
- Department of Medicine, University of Calgary, Calgary, AB T2N 4N2, Canada
| | | |
Collapse
|
13
|
Im SA, Cortes J, Cescon DW, Yusof MM, Iwata H, Masuda N, Takano T, Huang CS, Chung CF, Tsugawa K, Park YH, Matsumoto K, Inoue K, Kwong A, Loi S, Fu W, Pan W, Karantza V, Rugo HS, Schmid P. Results from the randomized KEYNOTE-355 study of pembrolizumab plus chemotherapy for Asian patients with advanced TNBC. NPJ Breast Cancer 2024; 10:79. [PMID: 39266535 PMCID: PMC11393332 DOI: 10.1038/s41523-024-00679-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/19/2024] [Indexed: 09/14/2024] Open
Abstract
In the phase 3 KEYNOTE-355 study (NCT02819518), pembrolizumab plus chemotherapy demonstrated statistically significant and clinically meaningful improvements in progression-free survival (PFS) and overall survival (OS) versus placebo plus chemotherapy among patients with previously untreated locally recurrent inoperable or metastatic triple-negative breast cancer (TNBC) and programmed cell death ligand 1 (PD-L1) combined positive score (CPS) ≥ 10 tumors. We analyzed outcomes for the subgroup of patients enrolled in Asia in KEYNOTE-355. Patients received pembrolizumab 200 mg or placebo (2:1 randomization) every 3 weeks for 35 cycles plus investigator's choice chemotherapy. Primary endpoints were PFS per Response Evaluation Criteria in Solid Tumors version 1.1 and OS. Among patients enrolled in Hong Kong, Japan, Korea, Malaysia and Taiwan (pembrolizumab plus chemotherapy, n = 113; placebo plus chemotherapy, n = 47), 117 (73.1%) had PD-L1 CPS ≥ 1 and 56 (35.0%) had PD-L1 CPS ≥ 10. Median time from randomization to data cutoff (June 15, 2021) was 43.8 (range, 36.8‒53.2) months (intent-to-treat [ITT] population). Hazard ratios (HRs [95% CI]) for PFS in the CPS ≥ 10, CPS ≥ 1, and ITT populations were 0.48 (0.24‒0.98), 0.58 (0.37‒0.91), and 0.66 (0.44‒0.99), respectively. Corresponding HRs (95% CI) for OS were 0.54 (0.28‒1.04), 0.62 (0.40‒0.97), and 0.57 (0.39‒0.84). Grade 3/4 treatment-related adverse events (AEs) occurred in 77.9% versus 78.7% of patients with pembrolizumab plus chemotherapy versus placebo plus chemotherapy. No grade 5 AEs occurred. Clinically meaningful improvement in PFS and OS with manageable toxicity were observed with pembrolizumab plus chemotherapy versus placebo plus chemotherapy in patients enrolled in Asia with previously untreated, inoperable or metastatic TNBC.Trial registration: ClinicalTrials.gov, NCT02819518.
Collapse
Affiliation(s)
- Seock-Ah Im
- Seoul National University Hospital, Seoul National University College of Medicine, Cancer Research Institute, Seoul National University, Seoul, Republic of Korea.
| | - Javier Cortes
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Madrid and Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
| | - David W Cescon
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, ON, Canada
| | - Mastura Md Yusof
- Cancer Center at Pantai Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| | - Hiroji Iwata
- Aichi Cancer Center Hospital, Nagoya, Aichi, Japan
| | - Norikazu Masuda
- Nagoya University Graduate School of Medicine, Nagoya, Aichi, Japan
| | - Toshimi Takano
- The Cancer Institute Hospital of JFCR, Tokyo, Japan
- Toranomon Hospital, Tokyo, Japan
| | - Chiun-Sheng Huang
- National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan
| | - Chi-Feng Chung
- Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan
| | | | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | | | - Ava Kwong
- Division of Breast Surgery, The University of Hong Kong, Queen Mary and Tung Wah Hospital, Hong Kong, China
- The University of Hong Kong-ShenZhen Hospital, Shenzhen, China
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Centre, Melbourne, Australia
- The Sir Peter MacCallum Department of Medical Oncology, University of Melbourne, Parkville, Australia
| | - Wei Fu
- Merck & Co., Inc., Rahway, NJ, USA
| | | | | | - Hope S Rugo
- University of California San Francisco Comprehensive Cancer Center, San Francisco, CA, USA
| | - Peter Schmid
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, London, UK
| |
Collapse
|
14
|
Rossi V, Turati A, Rosato A, Carpanese D. Sacituzumab govitecan in triple-negative breast cancer: from bench to bedside, and back. Front Immunol 2024; 15:1447280. [PMID: 39211043 PMCID: PMC11357913 DOI: 10.3389/fimmu.2024.1447280] [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: 06/11/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Triple-negative breast cancer (TNBC) represents a major therapeutic challenge due to its heterogeneous and aggressive phenotype, and limited target-specific treatment options. The trophoblast cell surface antigen (Trop-2), a transmembrane glycoprotein overexpressed in various cancers, has emerged as a promising target for TNBC. Sacituzumab govitecan (SG), an antibody-drug conjugate (ADC) that targets Trop-2, has recently entered treatment algorithms for advanced and metastatic TNBC, independently from Trop-2 expression status, with manageable toxicity. Despite the impressive results, questions remain unsolved regarding its efficacy, safety profile, and Trop-2 biological role in cancer. Currently, Trop-2 cannot be designated as a predictive biomarker in SG treatment, albeit its expression correlates with disease outcome, yet its levels are not uniform across all TNBCs. Additionally, data regarding Trop-2 expression variations in primary and metastatic sites, and its interplay with other biomarkers are still ambiguous but mandatory in light of future applications of SG in other indications and settings. This poses the questions of a careful evaluation of the efficacy and toxicity profile of SG in such early stages of disease, and in personalized and combinatorial strategies. Research and clinical data are mandatory to address SG drawbacks and minimize its benefits, to realize its full potential as therapeutic agent in different epithelial tumors.
Collapse
Affiliation(s)
- Valentina Rossi
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
| | - Alessandra Turati
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
| | - Antonio Rosato
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padova, Italy
| | - Debora Carpanese
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology (IOV)-IRCCS, Padova, Italy
| |
Collapse
|
15
|
Hao Q, Dai L, Chang L, Song D, Liu D, Ma X, Wu H, Kang H. Evaluation of neoadjuvant chemotherapy for clinical T1 triple-negative breast cancer. Sci Rep 2024; 14:18055. [PMID: 39103475 PMCID: PMC11300443 DOI: 10.1038/s41598-024-68719-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/26/2024] [Indexed: 08/07/2024] Open
Abstract
The role of neoadjuvant chemotherapy and its benefits in patients with triple-negative breast cancer (TNBC) and small tumors are unclear. This study aims to compare survival differences between clinical T1 TNBC receiving neoadjuvant chemotherapy (NAC) and adjuvant chemotherapy (AC). Data for patients with clinical T1 TNBC were extracted from the Surveillance, Epidemiology, and End Results (SEER) database. Patients were categorized according to whether they received chemotherapy before or after surgery. Propensity Score Matching (PSM) was used to minimize the influence of confounding factors. OS and BCSS were compared between the two treatment sequences using Kaplan-Meier and univariate and multivariable Cox proportional hazards regression analyses. The study included 6249 women with T1 TNBC. In multivariate analysis, compared with that in the AC group, the hazard ratio for death in the NAC group was 1.54 (95% confidence interval 1.26-1.89, p < 0.001). NAC offers no additional benefits in any age group or T, N subgroups. Our findings suggest that NAC does not provide additional benefit to patients with clinical T1 TNBC, even in the presence of lymph node metastasis, or T1c.
Collapse
Affiliation(s)
- Qian Hao
- The Comprehensive Breast Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West 5th Road, Xi'an, 710061, China
| | - Luyao Dai
- The Comprehensive Breast Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West 5th Road, Xi'an, 710061, China
| | - Lidan Chang
- The Comprehensive Breast Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West 5th Road, Xi'an, 710061, China
| | - Dingli Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China
| | - Dandan Liu
- The Comprehensive Breast Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West 5th Road, Xi'an, 710061, China
| | - Xiaobin Ma
- The Comprehensive Breast Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West 5th Road, Xi'an, 710061, China.
| | - Hao Wu
- School of Basic Medical Sciences, Key Laboratory of Environment and Genes Related to Diseases of Ministry of Education, Xi'an Jiaotong University, 76 Yanta West Road, Xi'an, 710061, China.
| | - Huafeng Kang
- The Comprehensive Breast Center, The Second Affiliated Hospital of Xi'an Jiaotong University, 157 West 5th Road, Xi'an, 710061, China.
| |
Collapse
|
16
|
Duan F, Hua X, Bi X, Wang S, Shi Y, Xu F, Wang L, Huang J, Yuan Z, Huang Y. Screening optimal candidates with operable, early-stage triple-negative breast cancer benefitting from capecitabine maintenance: A post-hoc analysis of the SYSUCC-001 study. Breast 2024; 76:103740. [PMID: 38733700 PMCID: PMC11103481 DOI: 10.1016/j.breast.2024.103740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 04/01/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024] Open
Abstract
BACKGROUND To explore whether specific clinicopathological covariates are predictive for a benefit from capecitabine maintenance in early-stage triple-negative breast cancer (TNBC) in the SYSUCC-001 phase III clinical trial. METHODS Candidate covariates included age, menstrual status, type of surgery, postoperative chemotherapy regimen, Ki-67 percentage, histologic grade, primary tumor size, lymphovascular invasion, node status, and capecitabine medication. Their nonlinear effects were modeled by restricted cubic spline. The primary endpoint was disease-free survival (DFS). A survival prediction model was constructed using Cox proportional hazards regression analysis. RESULTS All 434 participants (306 in development cohort and 128 in validation cohort) were analyzed. The estimated 5-year DFS in development and validation cohorts were 77.8 % (95 % CI, 72.9%-82.7 %) and 78.2 % (95 % CI, 70.9%-85.5 %), respectively. Age and node status had significant nonlinear effects on DFS. The prediction model constructed using four covariates (node status, lymphovascular invasion, capecitabine maintenance, and age) demonstrated satisfactory calibration and fair discrimination ability, with C-index of 0.722 (95 % CI, 0.662-0.781) and 0.764 (95 % CI, 0.668-0.859) in development and validation cohorts, respectively. Moreover, patient classification was conducted according to their risk scores calculated using our model, in which, notable survival benefits were reported in low-risk subpopulations. An easy-to-use online calculator for predicting benefit of capecitabine maintenance was also designed. CONCLUSIONS The evidence-based prediction model can be readily assessed at baseline, which might help decision making in clinical practice and optimize patient stratification, especially for those with low-risk, capecitabine maintenance might be a potential strategy in the early-disease setting.
Collapse
Affiliation(s)
- Fangfang Duan
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xin Hua
- Department of Radiation Oncology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiwen Bi
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shusen Wang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yanxia Shi
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Fei Xu
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Li Wang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Jiajia Huang
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Zhongyu Yuan
- Department of Medical Oncology, Sun Yat-Sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Yuanyuan Huang
- Department of VIP Region, Sun Yat-Sen University Cancer Center, The State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
17
|
Wang M, Pan B, Hu Y, Gao J, Hou L, Yu Z, Li M, Zhao Z. STIL facilitates the development and malignant progression of triple-negative breast cancer through activation of Fanconi anemia pathway via interacting with KLF16. Transl Oncol 2024; 46:102010. [PMID: 38823260 PMCID: PMC11177054 DOI: 10.1016/j.tranon.2024.102010] [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: 07/30/2023] [Revised: 05/08/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND STIL is an important cell cycle-regulating protein specifically recruited to the mitotic centrosome to promote the replication of centrioles in dividing cells. However, the potential role of STIL in the regulation of the biological functions of triple-negative breast cancer remains still unclear. METHODS We screened for differentially expressed STIL in the Cancer Genome Atlas database. The expression of STIL protein in 10 pairs of breast cancer tissues and adjacent normal tissues was further assessed by western blotting. Functionally, the knockdown and overexpression of STIL have been used to explore the effects of STIL on breast cancer cell proliferation, migration, and invasion. Mechanistically, RNA-seq, dual-luciferase reporter assay, chromatin immunoprecipitation assay, mass spectrometry, immunoprecipitation assay, and DNA pull-down assay were performed. RESULTS Breast cancer tissues and cells have higher STIL expression than normal tissues and cells. STIL knockdown impairs breast cancer cell growth, migration, and invasion, whereas STIL overexpression accelerates these processes. STIL promotes breast cancer progression by regulating FANCD2 expression, and exploration of its molecular mechanism demonstrated that STIL interacts with KLF16 to regulate the expression of FANCD2. CONCLUSIONS Collectively, our findings identified STIL as a critical promoter of breast cancer progression that interacts with KLF16 to regulate Fanconi anemia pathway protein FANCD2. In summary, STIL is a potential novel biomarker and therapeutic target for breast cancer.
Collapse
Affiliation(s)
- Meiling Wang
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Bo Pan
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Ye Hu
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Jiyue Gao
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Lu Hou
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China
| | - Zhenlong Yu
- College of Pharmacy, Dalian Medical University, Dalian, China.
| | - Man Li
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China.
| | - Zuowei Zhao
- Department of Breast Surgery & Department of Oncology, The Second Hospital of Dalian Medical University, Dalian, 116023 China.
| |
Collapse
|
18
|
Shi Z, Liu Y, Fang X, Liu X, Meng J, Zhang J. Efficacy and prognosis of HER2-Low and HER2-Zero in triple-negative breast cancer after neoadjuvant chemotherapy. Sci Rep 2024; 14:16899. [PMID: 39043756 PMCID: PMC11266405 DOI: 10.1038/s41598-024-67795-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 07/16/2024] [Indexed: 07/25/2024] Open
Abstract
Mounting evidence showed that HER2-Low breast cancer patients could benefit from the novel anti-HER2 antibody-drug conjugates (ADCs) treatment, which pointed the way towards better therapy for HER2-Low patients. The purpose of this study was to describe the clinicopathological features, along with chemotherapeutic effects and survival outcomes of HER2-Low and HER2-Zero in TNBC who received neoadjuvant chemotherapy (NACT). We retrospectively evaluated 638 triple-negative breast cancer patients who were treated with neoadjuvant chemotherapy between August 2014 and August 2022. Pathologic complete response (pCR) and survival outcomes were analyzed in HER2-Low cohort, HER2-Zero cohort and the overall patients, respectively. In the entire cohort, 342 (53.6%) patients were HER2-Low and 296 (46.4%) patients were HER2-Zero. No significant difference was found between HER2-Low and HER2-Zero patients based on all the clinical-pathological characteristics. 143 cases (22.4%) achieved pCR after NACT in the overall TNBC patients. The pCR rate of the HER2-Low patients and the HER2-Zero patients was 21.3% and 23.6%, respectively, exhibiting no statistical difference (p = 0.487). The survival of pCR group after NACT significantly improved compared to non-pCR group either in HER2-Low patients or in HER2-Zero patients. Although we found that patients with HER2-Low had longer DFS than patients with HER2-Zero, there was no considerable difference (p = 0.068). However, HER2-Low patients had a dramatically longer OS than HER2-Zero patients (p = 0.012). The data from present study confirmed the clinical importance of HER2-Low expression in TNBC. Further effort is needed to determine whether HER2-Low could be a more favorable prognostic marker for individual treatment.
Collapse
Affiliation(s)
- Zhendong Shi
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Yingxue Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Xuan Fang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Xu Liu
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Jie Meng
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China
| | - Jin Zhang
- The Third Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, China.
- Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
- Tianjin's Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China.
- Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin, China.
| |
Collapse
|
19
|
Kim H, Son S, Ko Y, Lim H, Lee J, Lee KM, Shin I. CYR61 confers chemoresistance by upregulating survivin expression in triple-negative breast cancer. Carcinogenesis 2024; 45:510-519. [PMID: 38446998 DOI: 10.1093/carcin/bgae013] [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: 06/29/2023] [Revised: 02/07/2024] [Accepted: 03/05/2024] [Indexed: 03/08/2024] Open
Abstract
Cysteine-rich angiogenic inducer 61 (CYR61) is a protein from the CCN family of matricellular proteins that play diverse regulatory roles in the extracellular matrix. CYR61 is involved in cell adhesion, migration, proliferation, differentiation, apoptosis, and senescence. Here, we show that CYR61 induces chemoresistance in triple-negative breast cancer (TNBC). We observed that CYR61 is overexpressed in TNBC patients, and CYR61 expression correlates negatively with the survival of patients who receive chemotherapy. CYR61 knockdown reduced cell migration, sphere formation and the cancer stem cell (CSC) population and increased the chemosensitivity of TNBC cells. Mechanistically, CYR61 activated Wnt/β-catenin signaling and increased survivin expression, which are associated with chemoresistance, the epithelial-mesenchymal transition, and CSC-like phenotypes. Altogether, our study demonstrates a novel function of CYR61 in chemotherapy resistance in breast cancer.
Collapse
Affiliation(s)
- Hyungjoo Kim
- Department of Life Science, Hanyang University, Seoul 04763, Korea
| | - Seogho Son
- Department of Life Science, Hanyang University, Seoul 04763, Korea
| | - Yunhyo Ko
- Department of Life Science, Hanyang University, Seoul 04763, Korea
| | - Hogeun Lim
- Department of Life Science, Hanyang University, Seoul 04763, Korea
| | - Joohyung Lee
- Department of Life Science, Hanyang University, Seoul 04763, Korea
| | - Kyung-Min Lee
- Department of Life Science, Hanyang University, Seoul 04763, Korea
- Natural Science Institute, Hanyang University, Seoul 04763, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Korea
| | - Incheol Shin
- Department of Life Science, Hanyang University, Seoul 04763, Korea
- Natural Science Institute, Hanyang University, Seoul 04763, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul 04763, Korea
| |
Collapse
|
20
|
Greco C. Coexisting cancer regimes: Transformations of breast and lung cancer in the United Kingdom. Health (London) 2024; 28:615-632. [PMID: 37218210 PMCID: PMC11149387 DOI: 10.1177/13634593231176979] [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] [Indexed: 05/24/2023]
Abstract
Using in-depth interviews with medical professionals working in the UK, I explore the coexistence of two different cancer regimes in which the different innovations for breast and lung cancer can be located. Breast cancer treatment has seen a protracted series of significant innovations in the context of an emphasis on screening that coexists with a segmentation in subtypes that has allowed targeted therapies for most patients. Lung cancer has also seen the introduction of targeted therapies; however, these can only be used for small groups of patients. Consequently, some interviewees working on lung cancer have expressed a stronger focus on increasing the number of patients undergoing surgery, as well as introducing screening also for lung cancer. As a result, a cancer regime based on the promises of targeted therapies coexists with a more traditional approach that focuses on diagnosing and treating cancers in their early stages.
Collapse
|
21
|
Guo J, Meng W, Li Q, Zheng Y, Yin H, Liu Y, Zhao S, Ma J. Pretreatment Sarcopenia and MRI-Based Radiomics to Predict the Response of Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer. Bioengineering (Basel) 2024; 11:663. [PMID: 39061745 PMCID: PMC11274092 DOI: 10.3390/bioengineering11070663] [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: 05/20/2024] [Revised: 06/13/2024] [Accepted: 06/25/2024] [Indexed: 07/28/2024] Open
Abstract
The association between sarcopenia and the effectiveness of neoadjuvant chemotherapy (NAC) in triple-negative breast cancer (TNBC) remains uncertain. This study aims to examine the potential of sarcopenia as a predictive factor for the response to NAC in TNBC, and to assess whether its combination with MRI radiomic signatures can improve the predictive accuracy. We collected clinical and pathological information, as well as pretreatment breast MRI and abdominal CT images, of 121 patients with TNBC who underwent NAC at our hospital between January 2012 and September 2021. The presence of pretreatment sarcopenia was assessed using the L3 skeletal muscle index. Clinical models were constructed based on independent risk factors identified by univariate regression analysis. Radiomics data were extracted on breast MRI images and the radiomics prediction models were constructed. We integrated independent risk factors and radiomic features to build the combined models. The results of this study demonstrated that sarcopenia is an independent predictive factor for NAC efficacy in TNBC. The combination of sarcopenia and MRI radiomic signatures can further improve predictive performance.
Collapse
Affiliation(s)
- Jiamin Guo
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; (J.G.); (Y.Z.)
| | - Wenjun Meng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China;
| | - Qian Li
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; (Q.L.); (Y.L.)
| | - Yichen Zheng
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; (J.G.); (Y.Z.)
| | - Hongkun Yin
- Infervision Medical Technology Co., Ltd., No. 62 East Fourth Ring Middle Road, Chaoyang District, Beijing 100025, China;
| | - Ying Liu
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; (Q.L.); (Y.L.)
| | - Shuang Zhao
- Department of Radiology, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; (Q.L.); (Y.L.)
| | - Ji Ma
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu 610041, China; (J.G.); (Y.Z.)
| |
Collapse
|
22
|
Sommariva M, Dolci M, Triulzi T, Ambrogi F, Dugo M, De Cecco L, Le Noci V, Bernardo G, Anselmi M, Montanari E, Pupa SM, Signorini L, Gagliano N, Sfondrini L, Delbue S, Tagliabue E. Impact of in vitro SARS-CoV-2 infection on breast cancer cells. Sci Rep 2024; 14:13134. [PMID: 38849411 PMCID: PMC11161491 DOI: 10.1038/s41598-024-63804-3] [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: 07/23/2023] [Accepted: 06/03/2024] [Indexed: 06/09/2024] Open
Abstract
The pandemic of coronavirus disease 19 (COVID-19), caused by severe respiratory syndrome coronavirus 2 (SARS-CoV-2), had severe repercussions for breast cancer patients. Increasing evidence indicates that SARS-CoV-2 infection may directly impact breast cancer biology, but the effects of SARS-CoV-2 on breast tumor cells are still unknown. Here, we analyzed the molecular events occurring in the MCF7, MDA-MB-231 and HCC1937 breast cancer cell lines, representative of the luminal A, basal B/claudin-low and basal A subtypes, respectively, upon SARS-CoV-2 infection. Viral replication was monitored over time, and gene expression profiling was conducted. We found that MCF7 cells were the most permissive to viral replication. Treatment of MCF7 cells with Tamoxifen reduced the SARS-CoV-2 replication rate, suggesting an involvement of the estrogen receptor in sustaining virus replication in malignant cells. Interestingly, a metagene signature based on genes upregulated by SARS-CoV-2 infection in all three cell lines distinguished a subgroup of premenopausal luminal A breast cancer patients with a poor prognosis. As SARS-CoV-2 still spreads among the population, it is essential to understand the impact of SARS-CoV-2 infection on breast cancer, particularly in premenopausal patients diagnosed with the luminal A subtype, and to assess the long-term impact of COVID-19 on breast cancer outcomes.
Collapse
Affiliation(s)
- Michele Sommariva
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy.
- Microambiente e Biomarcatori dei Tumori Solidi, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy.
| | - Maria Dolci
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Via Pascal 36, 20133, Milan, Italy
| | - Tiziana Triulzi
- Microambiente e Biomarcatori dei Tumori Solidi, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Federico Ambrogi
- Dipartimento di Scienze Cliniche e di Comunità, Università degli Studi di Milano, Via Celoria 22, 20133, Milan, Italy
| | - Matteo Dugo
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Loris De Cecco
- Integrated Biology of Rare Tumors, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Valentino Le Noci
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
| | - Giancarla Bernardo
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
| | - Martina Anselmi
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
| | - Elena Montanari
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
| | - Serenella M Pupa
- Microambiente e Biomarcatori dei Tumori Solidi, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Lucia Signorini
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Via Pascal 36, 20133, Milan, Italy
| | - Nicoletta Gagliano
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
| | - Lucia Sfondrini
- Dipartimento di Scienze Biomediche per la Salute, Università degli Studi di Milano, Via Mangiagalli 31, 20133, Milan, Italy
- Microambiente e Biomarcatori dei Tumori Solidi, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| | - Serena Delbue
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Via Pascal 36, 20133, Milan, Italy
| | - Elda Tagliabue
- Microambiente e Biomarcatori dei Tumori Solidi, Dipartimento di Oncologia Sperimentale, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Via Amadeo 42, 20133, Milan, Italy
| |
Collapse
|
23
|
Cortes J, Winer EP, Lipatov O, Im S, Gonçalves A, Muñoz‐Couselo E, Lee KS, Schmid P, Tamura K, Testa L, Witzel I, Ohtani S, Hund S, Kulangara K, Karantza V, Mejia JA, Ma J, Jelinic P, Huang L, Pruitt SK, Emancipator K. Contribution of tumour and immune cells to PD-L1 expression as a predictive biomarker in metastatic triple-negative breast cancer: exploratory analysis from KEYNOTE-119. J Pathol Clin Res 2024; 10:e12371. [PMID: 38627977 PMCID: PMC11021797 DOI: 10.1002/2056-4538.12371] [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: 10/06/2023] [Revised: 02/27/2024] [Accepted: 03/06/2024] [Indexed: 04/19/2024]
Abstract
The efficacy of pembrolizumab monotherapy versus chemotherapy increased with increasing programmed death ligand 1 (PD-L1) expression, as quantified by combined positive score (CPS; PD-L1 expression on both tumour cells and immune cells) in patients with previously treated metastatic triple-negative breast cancer (mTNBC) in the phase 3 KEYNOTE-119 study. This exploratory analysis was conducted to determine whether the expression of PD-L1 on tumour cells contributes to the predictive value of PD-L1 CPS in mTNBC. PD-L1 expression in tumour samples was assessed using PD-L1 IHC 22C3 pharmDx and quantified using both CPS and tumour proportion score (TPS; PD-L1 expression on tumour cells alone). Calculated immune cell density (CID) was defined as CPS minus TPS. The ability of each scoring method (CPS, TPS, and CID) to predict clinical outcomes with pembrolizumab was evaluated. With pembrolizumab, the area under the receiver operating characteristic curve was 0.69 (95% CI = 0.58-0.80) for CPS, 0.55 (95% CI = 0.46-0.64) for TPS, and 0.67 (95% CI = 0.56-0.77) for CID. After correction for cutoff prevalence, CPS performed as well as, if not better than, CID with respect to predicting objective response rate, progression-free survival, and overall survival. Data from this exploratory analysis suggest that, although PD-L1 expression on immune cells alone is predictive of response to programmed death 1 blockade in mTNBC, adding tumour PD-L1 expression assessment (i.e. CPS, which combines immune cell and tumour cell PD-L1 expression) may improve prediction. PD-L1 CPS thus remains an effective and broadly applicable uniform scoring system for enriching response to programmed death 1 blockade with pembrolizumab in mTNBC as well as other tumour types.
Collapse
Affiliation(s)
- Javier Cortes
- Oncology DepartmentInternational Breast Cancer Center (BCC), Pangaea Oncology, QuirónsaludBarcelonaSpain
- Department of MedicineFaculty of Biomedical and Health Sciences, European University of MadridMadridSpain
| | - Eric P Winer
- Yale Cancer CenterYale School of MedicineNew HavenCTUSA
- Present address:
Yale Cancer CenterNew HavenCTUSA
| | - Oleg Lipatov
- Department of OncologyRepublican Clinical Oncology Dispensary of the Ministry of Public Health of Bashkortostan RepublicUfaRussia
| | - Seock‐Ah Im
- Department of Internal MedicineSeoul National University Cancer Research Institute, Seoul National University College of Medicine, Seoul National UniversitySeoulRepublic of Korea
| | - Anthony Gonçalves
- Aix Marseille University, CNRS, INSERM, Department of Medical OncologyInstitut Paoli‐Calmettes, CRCMMarseilleFrance
| | - Eva Muñoz‐Couselo
- Department of Medical OncologyVall d'Hebron University HospitalBarcelonaSpain
| | - Keun Seok Lee
- Department of Medical OncologyCenter for Breast Cancer, National Cancer CenterGoyangRepublic of Korea
| | - Peter Schmid
- Department of Cancer MedicineBarts ECMC, Barts Cancer Institute, Queen Mary University of London, and Barts Health NHS TrustLondonUK
| | - Kenji Tamura
- Department of Medical OncologyNational Cancer Center HospitalTokyoJapan
| | - Laura Testa
- Instituto do Câncer do Estado de São Paulo Octavio Frias de OliveiraHospital das Clínicas da Faculdade de Medicina da Universidade de Sao PauloSao PauloBrazil
| | - Isabell Witzel
- Department of GynecologyUniversity Medical Center Hamburg–EppendorfHamburgGermany
- Department of GynecologyUniversity of Zurich, University Hospital ZurichZurichSwitzerland
| | - Shoichiro Ohtani
- Division of Breast SurgeryHiroshima City Hiroshima Citizens HospitalHiroshimaJapan
| | - Stephanie Hund
- Diagnostics and Genomics GroupAgilent TechnologiesCarpinteriaCAUSA
| | - Karina Kulangara
- Diagnostics and Genomics GroupAgilent TechnologiesCarpinteriaCAUSA
| | | | - Jaime A Mejia
- Department of Medical OncologyMerck & Co., Inc.RahwayNJUSA
| | - Junshui Ma
- Early Development StatisticsMerck & Co., Inc.RahwayNJUSA
| | - Petar Jelinic
- Department of Medical OncologyMerck & Co., Inc.RahwayNJUSA
| | - Lingkang Huang
- Early Development StatisticsMerck & Co., Inc.RahwayNJUSA
| | - Scott K Pruitt
- Department of Medical OncologyMerck & Co., Inc.RahwayNJUSA
| | - Kenneth Emancipator
- Early Oncology DevelopmentMerck & Co., Inc.RahwayNJUSA
- Present address:
Precision Medicine, AbbVie, Inc.North ChicagoILUSA
| |
Collapse
|
24
|
Kim HY, Kim YM, Hong S. CK2α-mediated phosphorylation of GRP94 facilitates the metastatic cascade in triple-negative breast cancer. Cell Death Discov 2024; 10:185. [PMID: 38649679 PMCID: PMC11035675 DOI: 10.1038/s41420-024-01956-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Distant metastasis is a significant hallmark affecting to the high death rate of patients with triple-negative breast cancer (TNBC). Thus, it is crucial to identify and develop new therapeutic strategies to hinder cancer metastasis. While emerging studies have hinted a pivotal role of glucose-regulated protein 94 (GRP94) in tumorigenesis, the exact biological functions and molecular mechanisms of GRP94 in modulating cancer metastasis remain to be elucidated. Our study demonstrated an increased expression of GRP94 in TNBC correlated with metastatic progression and unfavorable prognosis in patients. Functionally, we identified that GRP94 depletion significantly diminished TNBC tumorigenesis and subsequent lung metastasis. In contrast, GRP94 overexpression exacerbated the invasiveness, migration, and lung metastasis of non-TNBC cells. Mechanistically, we found that casein kinase 2 alpha (CK2α) active in advanced breast cancer phosphorylated GRP94 at a conserved serine 306 (S306) residue. This phosphorylation increased the stability of GRP94 and enhanced its interaction with LRP6, leading to activation of canonical Wnt signaling. From a therapeutic standpoint, we found that benzamidine, a novel CK2α inhibitor, effectively suppressed GRP94 phosphorylation, LRP6 stabilization, and metastasis of TNBC. Our results point to the critical role of CK2α-mediated GRP94 phosphorylation in TNBC metastasis through activation of Wnt signaling, highlighting GRP94 as a therapeutic target to impede TNBC metastasis.
Collapse
Affiliation(s)
- Hye-Youn Kim
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon, 21999, Republic of Korea
| | - Young-Mi Kim
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon, 21999, Republic of Korea
| | - Suntaek Hong
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, Gachon University College of Medicine, Incheon, 21999, Republic of Korea.
| |
Collapse
|
25
|
Lewis K, La Selva R, Maldonado E, Annis MG, Najyb O, Cepeda Cañedo E, Totten S, Hébert S, Sabourin V, Mirabelli C, Ciccolini E, Lehuédé C, Choinière L, Russo M, Avizonis D, Park M, St-Pierre J, Kleinman CL, Siegel PM, Ursini-Siegel J. p66ShcA promotes malignant breast cancer phenotypes by alleviating energetic and oxidative stress. Redox Biol 2024; 70:103028. [PMID: 38211442 PMCID: PMC10821068 DOI: 10.1016/j.redox.2024.103028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 01/13/2024] Open
Abstract
Significant efforts have focused on identifying targetable genetic drivers that support the growth of solid tumors and/or increase metastatic ability. During tumor development and progression to metastatic disease, physiological and pharmacological selective pressures influence parallel adaptive strategies within cancer cell sub-populations. Such adaptations allow cancer cells to withstand these stressful microenvironments. This Darwinian model of stress adaptation often prevents durable clinical responses and influences the emergence of aggressive cancers with increased metastatic fitness. However, the mechanisms contributing to such adaptive stress responses are poorly understood. We now demonstrate that the p66ShcA redox protein, itself a ROS inducer, is essential for survival in response to physiological stressors, including anchorage independence and nutrient deprivation, in the context of poor outcome breast cancers. Mechanistically, we show that p66ShcA promotes both glucose and glutamine metabolic reprogramming in breast cancer cells, to increase their capacity to engage catabolic metabolism and support glutathione synthesis. In doing so, chronic p66ShcA exposure contributes to adaptive stress responses, providing breast cancer cells with sufficient ATP and redox balance needed to withstand such transient stressed states. Our studies demonstrate that p66ShcA functionally contributes to the maintenance of aggressive phenotypes and the emergence of metastatic disease by forcing breast tumors to adapt to chronic and moderately elevated levels of oxidative stress.
Collapse
Affiliation(s)
- Kyle Lewis
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Rachel La Selva
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Elias Maldonado
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Matthew G Annis
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Ouafa Najyb
- Department of Biochemistry, McGill University, Montreal, QC, Canada; Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Eduardo Cepeda Cañedo
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Stephanie Totten
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Steven Hébert
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Valérie Sabourin
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada
| | - Caitlynn Mirabelli
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Emma Ciccolini
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada
| | - Camille Lehuédé
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Luc Choinière
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Mariana Russo
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Daina Avizonis
- Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | - Morag Park
- Department of Biochemistry, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada; Goodman Cancer Institute, McGill University, Montreal, QC, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| | - Julie St-Pierre
- Department of Biochemistry, Microbiology and Immunology and Ottawa Institute of Systems Biology, University of Ottawa, ON, Canada
| | - Claudia L Kleinman
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Peter M Siegel
- Department of Biochemistry, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada; Goodman Cancer Institute, McGill University, Montreal, QC, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| | - Josie Ursini-Siegel
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, QC, Canada; Department of Biochemistry, McGill University, Montreal, QC, Canada; Division of Experimental Medicine, McGill University, Montreal, QC, Canada; Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
26
|
Kashyap D, Salman H. Targeting Interleukin-13 Receptor α2 and EphA2 in Aggressive Breast Cancer Subtypes with Special References to Chimeric Antigen Receptor T-Cell Therapy. Int J Mol Sci 2024; 25:3780. [PMID: 38612592 PMCID: PMC11011362 DOI: 10.3390/ijms25073780] [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: 03/15/2024] [Revised: 03/24/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
Breast cancer (BCA) remains the leading cause of cancer-related mortality among women worldwide. This review delves into the therapeutic challenges of BCA, emphasizing the roles of interleukin-13 receptor α2 (IL-13Rα2) and erythropoietin-producing hepatocellular receptor A2 (EphA2) in tumor progression and resistance. Highlighting their overexpression in BCA, particularly in aggressive subtypes, such as Her-2-enriched and triple-negative breast cancer (TNBC), we discuss the potential of these receptors as targets for chimeric antigen receptor T-cell (CAR-T) therapies. We examine the structural and functional roles of IL-13Rα2 and EphA2, their pathological significance in BCA, and the promising therapeutic avenues their targeting presents. With an in-depth analysis of current immunotherapeutic strategies, including the limitations of existing treatments and the potential of dual antigen-targeting CAR T-cell therapies, this review aims to summarize potential future novel, more effective therapeutic interventions for BCA. Through a thorough examination of preclinical and clinical studies, it underlines the urgent need for targeted therapies in combating the high mortality rates associated with Her-2-enriched and TNBC subtypes and discusses the potential role of IL-13Rα2 and EphA2 as promising candidates for the development of CAR T-cell therapies.
Collapse
Affiliation(s)
| | - Huda Salman
- Brown Center for Immunotherapy, Melvin and Bren Simon Comprehensive Cancer Center, Division of Hematology and Oncology, School of Medicine, Indiana University, Indianapolis, IN 46202, USA;
| |
Collapse
|
27
|
Cavanagh RJ, Monteiro PF, Moloney C, Travanut A, Mehradnia F, Taresco V, Rahman R, Martin SG, Grabowska AM, Ashford MB, Alexander C. Free drug and ROS-responsive nanoparticle delivery of synergistic doxorubicin and olaparib combinations to triple negative breast cancer models. Biomater Sci 2024; 12:1822-1840. [PMID: 38407276 DOI: 10.1039/d3bm01931d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Combinations of the topoisomerase II inhibitor doxorubicin and the poly (ADP-ribose) polymerase inhibitor olaparib offer potential drug-drug synergy for the treatment of triple negative breast cancers (TNBC). In this study we performed in vitro screening of combinations of these drugs, administered directly or encapsulated within polymer nanoparticles, in both 2D and in 3D spheroid models of breast cancer. A variety of assays were used to evaluate drug potency, and calculations of combination index (CI) values indicated that synergistic effects of drug combinations occurred in a molar-ratio dependent manner. It is suggested that the mechanisms of synergy were related to enhancement of DNA damage as shown by the level of double-strand DNA breaks, and mechanisms of antagonism associated with mitochondrial mediated cell survival, as indicated by reactive oxygen species (ROS) generation. Enhanced drug delivery and potency was observed with nanoparticle formulations, with a greater extent of doxorubicin localised to cell nuclei as evidenced by microscopy, and higher cytotoxicity at the same time points compared to free drugs. Together, the work presented identifies specific combinations of doxorubicin and olaparib which were most effective in a panel of TNBC cell lines, explores the mechanisms by which these combined agents might act, and shows that formulation of these drug combinations into polymeric nanoparticles at specific ratios conserves synergistic action and enhanced potency in vitro compared to the free drugs.
Collapse
Affiliation(s)
| | - Patrícia F Monteiro
- School of Pharmacy, University of Nottingham, NG7 2RD, UK.
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | - Cara Moloney
- School of Pharmacy, University of Nottingham, NG7 2RD, UK.
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | | | | | | | - Ruman Rahman
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | - Stewart G Martin
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | - Anna M Grabowska
- School of Medicine, BioDiscovery Institute, University of Nottingham, NG7 2RD, UK
| | - Marianne B Ashford
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, Macclesfield, UK
| | | |
Collapse
|
28
|
Woodward ER, Lalloo F, Forde C, Pugh S, Burghel GJ, Schlecht H, Harkness EF, Howell A, Howell SJ, Gandhi A, Evans DG. Germline testing of BRCA1, BRCA2, PALB2 and CHEK2 c.1100delC in 1514 triple negative familial and isolated breast cancers from a single centre, with extended testing of ATM, RAD51C and RAD51D in over 400. J Med Genet 2024; 61:385-391. [PMID: 38123987 DOI: 10.1136/jmg-2023-109671] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 11/14/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND The identification of germline pathogenic gene variants (PGVs) in triple negative breast cancer (TNBC) is important to inform further primary cancer risk reduction and TNBC treatment strategies. We therefore investigated the contribution of breast cancer associated PGVs to familial and isolated invasive TNBC. METHODS Outcomes of germline BRCA1, BRCA2 and CHEK2_c.1100delC testing were recorded in 1514 women (743-isolated, 771-familial), and for PALB2 in 846 women (541-isolated, 305-familial), with TNBC and smaller numbers for additional genes. Breast cancer free controls were identified from Predicting Risk Of Cancer At Screening and BRIDGES (Breast cancer RIsk after Diagnostic GEne Sequencing) studies. RESULTS BRCA1_PGVs were detected in 52 isolated (7.0%) and 195 (25.3%) familial cases (isolated-OR=58.9, 95% CI: 16.6 to 247.0), BRCA2_PGVs in 21 (2.8%) isolated and 67 (8.7%) familial cases (isolated-OR=5.0, 95% CI: 2.3 to 11.2), PALB2_PGVs in 9 (1.7%) isolated and 12 (3.9%) familial cases (isolated-OR=8.8, 95% CI: 2.5 to 30.4) and CHEK2_c.1100delC in 0 isolated and 3 (0.45%) familial cases (isolated-OR=0.0, 95% CI: 0.00 to 2.11). BRCA1_PGV detection rate was >10% for all familial TNBC age groups and significantly higher for younger diagnoses (familial: <50 years, n=165/538 (30.7%); ≥50 years, n=30/233 (12.9%); p<0.0001). Women with a G3_TNBC were more likely to have a BRCA1_PGV as compared with a BRCA2 or PALB2_PGV (p<0.0001). 0/743 isolated TNBC had the CHEK2_c.1100delC PGV and 0/305 any ATM_PGV, but 2/240 (0.83%) had a RAD51D_PGV. CONCLUSION PGVs in BRCA1 are associated with G3_TNBCs. Familial TNBCs and isolated TNBCs <30 years have a >10% likelihood of a PGV in BRCA1. BRCA1_PGVs are associated with younger age of familial TNBC. There was no evidence for any increased risk of TNBC with CHEK2 or ATM PGVs.
Collapse
Affiliation(s)
- Emma R Woodward
- Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Manchester Breast Centre, The Christie NHS Foundation Trust, Manchester, UK
| | - Fiona Lalloo
- Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Claire Forde
- Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Sarah Pugh
- Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - George J Burghel
- Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Helene Schlecht
- Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| | - Elaine F Harkness
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Anthony Howell
- Manchester Breast Centre, The Christie NHS Foundation Trust, Manchester, UK
- Prevent Breast Cancer Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Sacha J Howell
- Manchester Breast Centre, The Christie NHS Foundation Trust, Manchester, UK
- Prevent Breast Cancer Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Ashu Gandhi
- Prevent Breast Cancer Unit, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester, UK
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - D Gareth Evans
- Manchester Centre for Genomic Medicine, Manchester University Hospitals NHS Foundation Trust, Manchester, UK
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
- Manchester Breast Centre, The Christie NHS Foundation Trust, Manchester, UK
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
29
|
Bidany-Mizrahi T, Shweiki A, Maroun K, Abu-Tair L, Mali B, Aqeilan RI. Unveiling the relationship between WWOX and BRCA1 in mammary tumorigenicity and in DNA repair pathway selection. Cell Death Discov 2024; 10:145. [PMID: 38499540 PMCID: PMC10948869 DOI: 10.1038/s41420-024-01878-8] [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: 11/29/2023] [Revised: 02/07/2024] [Accepted: 02/20/2024] [Indexed: 03/20/2024] Open
Abstract
Breast cancer is the leading cause of cancer-related deaths in women worldwide, with the basal-like or triple-negative breast cancer (TNBC) subtype being particularly aggressive and challenging to treat. Understanding the molecular mechanisms driving the development and progression of TNBC is essential. We previously showed that WW domain-containing oxidoreductase (WWOX) is commonly inactivated in TNBC and is implicated in the DNA damage response (DDR) through ATM and ATR activation. In this study, we investigated the interplay between WWOX and BRCA1, both frequently inactivated in TNBC, on mammary tumor development and on DNA double-strand break (DSB) repair choice. We generated and characterized a transgenic mouse model (K14-Cre;Brca1fl/fl;Wwoxfl/fl) and observed that mice lacking both WWOX and BRCA1 developed basal-like mammary tumors and exhibited a decrease in 53BP1 foci and an increase in RAD51 foci, suggesting impaired DSB repair. We examined human TNBC cell lines harboring wild-type and mutant BRCA1 and found that WWOX expression promoted NHEJ repair in cells with wild-type BRCA1. Our findings suggest that WWOX and BRCA1 play an important role in DSB repair pathway choice in mammary epithelial cells, underscoring their functional interaction and significance in breast carcinogenesis.
Collapse
Affiliation(s)
- Tirza Bidany-Mizrahi
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Aya Shweiki
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Kian Maroun
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Lina Abu-Tair
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Bella Mali
- Department of Pathology, Hadassah University Hospital, Jerusalem, Israel
| | - Rami I Aqeilan
- The Concern Foundation Laboratories, The Lautenberg Center for Immunology and Cancer Research, Department of Immunology and Cancer Research-IMRIC, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
- Cyprus Cancer Research Institute (CCRI), Nicosia, Cyprus.
| |
Collapse
|
30
|
Khanna M, Singh LK, Shrivastava K, Singh R. An enhanced and efficient approach for feature selection for chronic human disease prediction: A breast cancer study. Heliyon 2024; 10:e26799. [PMID: 38463826 PMCID: PMC10920178 DOI: 10.1016/j.heliyon.2024.e26799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 01/15/2024] [Accepted: 02/20/2024] [Indexed: 03/12/2024] Open
Abstract
Computer-aided diagnosis (CAD) systems play a vital role in modern research by effectively minimizing both time and costs. These systems support healthcare professionals like radiologists in their decision-making process by efficiently detecting abnormalities as well as offering accurate and dependable information. These systems heavily depend on the efficient selection of features to accurately categorize high-dimensional biological data. These features can subsequently assist in the diagnosis of related medical conditions. The task of identifying patterns in biomedical data can be quite challenging due to the presence of numerous irrelevant or redundant features. Therefore, it is crucial to propose and then utilize a feature selection (FS) process in order to eliminate these features. The primary goal of FS approaches is to improve the accuracy of classification by eliminating features that are irrelevant or less informative. The FS phase plays a critical role in attaining optimal results in machine learning (ML)-driven CAD systems. The effectiveness of ML models can be significantly enhanced by incorporating efficient features during the training phase. This empirical study presents a methodology for the classification of biomedical data using the FS technique. The proposed approach incorporates three soft computing-based optimization algorithms, namely Teaching Learning-Based Optimization (TLBO), Elephant Herding Optimization (EHO), and a proposed hybrid algorithm of these two. These algorithms were previously employed; however, their effectiveness in addressing FS issues in predicting human diseases has not been investigated. The following evaluation focuses on the categorization of benign and malignant tumours using the publicly available Wisconsin Diagnostic Breast Cancer (WDBC) benchmark dataset. The five-fold cross-validation technique is employed to mitigate the risk of over-fitting. The evaluation of the proposed approach's proficiency is determined based on several metrics, including sensitivity, specificity, precision, accuracy, area under the receiver-operating characteristic curve (AUC), and F1-score. The best value of accuracy computed through the suggested approach is 97.96%. The proposed clinical decision support system demonstrates a highly favourable classification performance outcome, making it a valuable tool for medical practitioners to utilize as a secondary opinion and reducing the overburden of expert medical practitioners.
Collapse
Affiliation(s)
- Munish Khanna
- School of Computing Science and Engineering, Galgotias University, Greater Noida, Gautam Buddh Nagar, India
| | - Law Kumar Singh
- Department of Computer Engineering and Applications, GLA University, Mathura, India
| | - Kapil Shrivastava
- Department of Computer Engineering and Applications, GLA University, Mathura, India
| | - Rekha Singh
- Department of Physics, Uttar Pradesh Rajarshi Tandon Open University, Prayagraj, Uttar Pradesh, India
| |
Collapse
|
31
|
Scheffges C, Devy J, Giustiniani J, Francois S, Cartier L, Merrouche Y, Foussat A, Potteaux S, Bensussan A, Marie-Cardine A. Identification of CD160-TM as a tumor target on triple negative breast cancers: possible therapeutic applications. Breast Cancer Res 2024; 26:28. [PMID: 38360636 PMCID: PMC10870674 DOI: 10.1186/s13058-024-01785-x] [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: 11/13/2023] [Accepted: 02/12/2024] [Indexed: 02/17/2024] Open
Abstract
BACKGROUND Despite major therapeutic advances, triple-negative breast cancer (TNBC) still presents a worth prognosis than hormone receptors-positive breast cancers. One major issue relies in the molecular and mutational heterogeneity of TNBC subtypes that is reinforced by the absence of reliable tumor-antigen that could serve as a specific target to further promote efficient tumor cell recognition and depletion. CD160 is a receptor mainly expressed by NK lymphocytes and presenting two isoforms, namely the GPI-anchored form (CD160-GPI) and the transmembrane isoform (CD160-TM). While CD160-GPI is constitutively expressed on resting cells and involved in the generation of NK cells' cytotoxic activity, CD160-TM is neo-synthesized upon activation and promotes the amplification of NK cells' killing ability. METHODS CD160 expression was assessed by immunohistochemistry (IHC) and flow cytometry on TNBC patient biopsies or cell lines, respectively. Antibody (Ab)-mediated tumor depletion was tested in vitro by performing antibody-dependent cell cytotoxicity (ADCC) and phagocytosis (ADCP) assays, and in vivo on a TNBC mouse model. RESULTS Preliminary data obtained by IHC on TNBC patients' tumor biopsies revealed an unconventional expression of CD160 by TNBC tumor cells. By using a specific but conformation-dependent anti-CD160-TM Ab, we established that CD160-TM, but not CD160-GPI, was expressed by TNBC tumor cells. A conformation-independent anti-CD160-TM mAb (22B12; muIgG2a isotype) was generated and selected according to pre-defined specificity and functional criterions. In vitro functional assays demonstrated that ADCC and ADCP could be induced in the presence of 22B12, resulting in TNBC cell line apoptosis. The ability of 22B12 to exert an in vivo anti-tumor activity was also demonstrated on a TNBC murine model. CONCLUSIONS Our data identify CD160-TM as a tumor marker for TNBC and provide a rational for the use of anti-CD160-TM antibodies as therapeutic tools in this tumor context.
Collapse
Affiliation(s)
- Claire Scheffges
- INSERM U976, HIPI, Team 1, 75010, Paris, France
- Université Paris Cité, IRSL, 75010, Paris, France
- Alderaan Biotechnology, 75005, Paris, France
| | - Jérôme Devy
- UMR CNRS/URCA 7369, MEDyC, Université de Reims-Champagne-Ardennes, 51100, Reims, France
| | | | | | - Lucille Cartier
- Département de Recherche, Institut Godinot, 51100, Reims, France
- UR7509, IRMAIC, Université de Reims-Champagne-Ardennes, 51097, Reims, France
| | - Yacine Merrouche
- Département de Recherche, Institut Godinot, 51100, Reims, France
- UR7509, IRMAIC, Université de Reims-Champagne-Ardennes, 51097, Reims, France
| | | | - Stéphane Potteaux
- UR7509, IRMAIC, Université de Reims-Champagne-Ardennes, 51097, Reims, France
| | - Armand Bensussan
- INSERM U976, HIPI, Team 1, 75010, Paris, France
- Université Paris Cité, IRSL, 75010, Paris, France
| | - Anne Marie-Cardine
- INSERM U976, HIPI, Team 1, 75010, Paris, France.
- Université Paris Cité, IRSL, 75010, Paris, France.
| |
Collapse
|
32
|
Pan WK, Ren SY, Zhu LX, Lin BC. A Web-based Prediction Model for Early Death in Patients With Metastatic Triple-negative Breast Cancer. Am J Clin Oncol 2024; 47:71-80. [PMID: 37871164 PMCID: PMC10805354 DOI: 10.1097/coc.0000000000001058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is a subtype of breast cancer characterized by the absence of expression of estrogen receptor, progesterone receptor, or human epidermal growth factor receptor 2. This subtype of breast cancer is known for its high aggressiveness, high metastatic potential, tendency for recurrence, and poor prognosis. Patients with metastatic TNBC (mTNBC) have a poorer prognosis and a higher likelihood of early death (survival time ≤3 months). Therefore, the development of effective individualized survival prediction tools, such as prediction nomograms and web-based survival calculators, is of great importance for predicting the probability of early death in patients with metastatic TNBC. METHODS Patients diagnosed with mTNBC in the Surveillance, Epidemiology, and End Results database between 2010 and 2015 were included in the model construction. Univariate and multivariate logistic regression analysis was performed to identify risk factors associated with early death in patients with mTNBC and predictive prognostic nomograms were constructed. The accuracy of the nomograms was verified using receiver operating characteristic curves, and GiViTi Calibration belt plots were used to evaluate the model consistency. The clinical applicability of the nomograms was evaluated using decision curve analysis. On the basis of the predictive prognostic nomograms, a network survival rate calculator was developed for individualized survival prediction in patients with mTNBC. RESULTS A total of 2230 patients diagnosed with mTNBC were included in the Surveillance, Epidemiology, and End Results database for this study. After strict exclusion criteria, 1428 patients were found to be eligible for the study. All the patients were randomly divided into a training cohort and a validation cohort in a ratio of 7:3. Independent risk factors for mTNBC, including age, tumor size, brain metastasis, liver metastasis, surgery, and chemotherapy, were identified and integrated to construct the prediction nomogram and survival calculator. Results of receiver operating characteristic curves, calibration curves, and decision curve analysis curves from the training and validation cohort confirmed that the developed nomogram and web-based survival calculator in this study could accurately predict the probability of early death in patients with mTNBC. CONCLUSIONS In this study, we developed a reliable prediction nomogram and web-based survival calculator for predicting the probability of early death in patients with mTNBC. These tools can assist clinical physicians in identifying high-risk patients and developing personalized treatment plans as early as possible.
Collapse
Affiliation(s)
- Wen-kai Pan
- Department of Radiotherapy, The First Affiliated Hospital of Wenzhou Medical University
| | - Si-yan Ren
- Medical and Radiation Oncology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Liao-xiang Zhu
- Department of Radiotherapy, The First Affiliated Hospital of Wenzhou Medical University
| | - Bao-chai Lin
- Department of Radiotherapy, The First Affiliated Hospital of Wenzhou Medical University
| |
Collapse
|
33
|
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
|
34
|
Li X, Li J, Hu Q, Zhang X, Chen F. Association of physical weight statuses defined by body mass index (BMI) with molecular subtypes of premenopausal breast cancer: a systematic review and meta-analysis. Breast Cancer Res Treat 2024; 203:429-447. [PMID: 37882920 DOI: 10.1007/s10549-023-07139-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 09/25/2023] [Indexed: 10/27/2023]
Abstract
BACKGROUND AND PURPOSE The association between overweight/obesity and postmenopausal breast cancer has been proven. However, uncertainty exists regarding the association between physical weight statuses and premenopausal breast cancer subtypes. This study aimed to explore the association of body weight statuses with molecular subtypes of premenopausal breast cancer. METHOD A systematic search of Medline, PubMed, Embase, and Web of Science was performed. The Newcastle-Ottawa Scale (NOS) and the Joanna Briggs Institute (JBI) Critical Appraisal tools were used to evaluate the quality of the literature. STATA and R software were used to analyze the extracted data. RESULT The meta-analysis included 35 observational studies with a total of 41,049 premenopausal breast cancer patients. The study showed that the proportion of underweight patients was 4.8% (95% CI = 3.9-5.8%, P = 0.01), overweight was 29% (95%CI = 27.1-30.9%, P < 0.01), obesity was 17.8% (95% CI = 14.9-21.2%, P < 0.0001), and normal weight was 51.6% (95% CI = 46.7-56.5%, P < 0.0001). The pooled results showed that in comparison to the normal weight group, being physically underweight is related to a 1.44-fold risk (OR = 1.44, 95%CI = 1.28-1.63, P < 0.0001) of HER2 + breast cancer. Overweight is related to a 1.16-fold risk (OR = 1.16, 95%CI = 1.06-1.26, P = 0.002) of TNBC and a 16% lower risk (OR = 0.84, 95%CI = 0.75-0.93, P = 0.001) of ER + breast cancer. When compared to underweight/normal weight populations, both overweight (OR = 0.74, 95%CI = 0.56-0.97, P = 0.032) and obesity (OR = 0.70, 95%CI = 0.50-0.98, P = 0.037) can reduce the risk of ER + PR + breast cancer. CONCLUSION In the premenopausal breast cancer population, the distribution of patients' numbers with different weight statuses was significantly distinct among the various breast cancer subtypes. Additionally, the associations between physical weight statuses and the risk of premenopausal breast cancer subtypes are divergent.
Collapse
Affiliation(s)
- Xuchu Li
- Department of Medical, Queen Mary School, Nanchang University, 461 Bayi Avenue, Donghu District, Nanchang City, 330006, Jiangxi Province, China
| | - Jinping Li
- Department of General Medical, People's Hospital of Fu City, Yan'an, 727505, Shaanxi Province, China
| | - Qirui Hu
- College of Food Science, Nanchang University, Nanchang, 330047, Jiangxi Province, China
| | - Xu Zhang
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Fang Chen
- College of Food Science, Nanchang University, Nanchang, 330047, Jiangxi Province, China.
- Jiangxi Provincial Key Laboratory of Preventive Medicine, School of Public Health, Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
35
|
Dubey R, Sharma A, Gupta S, Gupta GD, Asati V. A comprehensive review of small molecules targeting PI3K pathway: Exploring the structural development for the treatment of breast cancer. Bioorg Chem 2024; 143:107077. [PMID: 38176377 DOI: 10.1016/j.bioorg.2023.107077] [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: 10/04/2023] [Revised: 11/28/2023] [Accepted: 12/27/2023] [Indexed: 01/06/2024]
Abstract
Cancer stands as one of the deadliest diseases, ranking second in terms of its global impact. Despite the presence of numerous compelling theories concerning its origins, none have succeeded in fully elucidating the intricate nature of this ailment. Among the prevailing concerns in today's world, breast cancer proliferation remains a significant issue, particularly affecting females. The abnormal proliferation of the PI3K pathway emerges as a prominent driver of breast cancer, underscoring its role in cellular survival and proliferation. Consequently, targeting this pathway has emerged as a leading strategy in breast cancer therapeutics. Within this context, the present article explores the current landscape of anti-tumour drug development, focusing on structural activity relationships (SAR) in PI3K targeting breast cancer treatment. Notably, certain moieties like triazines, pyrimidine, quinazoline, quinoline, and pyridoxine have been explored as potential PI3K inhibitors for combating breast cancer. Various heterocyclic small molecules are undergoing clinical trials, such as Alpelisib, the first orally available FDA-approved drug targeting PI3K; others include buparlisib, pictilisib, and taselisib, which inhibit class I PI3K. These drugs are used for the treatment of breast cancer but still have various side effects with their high cost. Therefore, the primary goal of this review is to include all current advances in the development of anticancer medicines that target PI3K over-activation in the treatment of breast cancer.
Collapse
Affiliation(s)
- Rahul Dubey
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Anushka Sharma
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - Shankar Gupta
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India
| | - G D Gupta
- Department of Pharmaceutics, ISF College of Pharmacy, Moga, Punjab, India
| | - Vivek Asati
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
36
|
Ding JH, Xiao Y, Yang F, Song XQ, Xu Y, Ding XH, Ding R, Shao ZM, Di GH, Jiang YZ. Guanosine diphosphate-mannose suppresses homologous recombination repair and potentiates antitumor immunity in triple-negative breast cancer. Sci Transl Med 2024; 16:eadg7740. [PMID: 38170790 DOI: 10.1126/scitranslmed.adg7740] [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: 01/22/2023] [Accepted: 11/29/2023] [Indexed: 01/05/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer with poor prognosis. TNBCs with high homologous recombination deficiency (HRD) scores benefit from DNA-damaging agents, including platinum drugs and poly(ADP-ribose) polymerase (PARP) inhibitors, whereas those with low HRD scores still lack therapeutic options. Therefore, we sought to exploit metabolic alterations to induce HRD and sensitize DNA-damaging agents in TNBCs with low HRD scores. We systematically analyzed TNBC metabolomics and identified a metabolite, guanosine diphosphate (GDP)-mannose (GDP-M), that impeded homologous recombination repair (HRR). Mechanistically, the low expression of the upstream enzyme GDP-mannose-pyrophosphorylase-A (GMPPA) led to the endogenous up-regulation of GDP-M in TNBC. The accumulation of GDP-M in tumor cells further reduced the interaction between breast cancer susceptibility gene 2 (BRCA2) and ubiquitin-specific peptidase 21 (USP21), which promoted the ubiquitin-mediated degradation of BRCA2 to inhibit HRR. Therapeutically, we illustrated that the supplementation of GDP-M sensitized DNA-damaging agents to impair tumor growth in both in vitro (cancer cell line and patient-derived organoid) and in vivo (xenograft in immunodeficient mouse) models. Moreover, the combination of GDP-M with DNA-damaging agents activated STING-dependent antitumor immunity in immunocompetent syngeneic mouse models. Therefore, GDP-M supplementation combined with PARP inhibition augmented the efficacy of anti-PD-1 antibodies. Together, these findings suggest that GDP-M is a crucial HRD-related metabolite and propose a promising therapeutic strategy for TNBCs with low HRD scores using the combination of GDP-M, PARP inhibitors, and anti-PD-1 immunotherapy.
Collapse
Affiliation(s)
- Jia-Han Ding
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
- Human Phenome Institute, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai 201203, P. R. China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Fan Yang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Xiao-Qing Song
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Ying Xu
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Xiao-Hong Ding
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Rui Ding
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Gen-Hong Di
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, P. R. China
| |
Collapse
|
37
|
Damaskos C, Psilopatis I, Garmpi A, Dimitroulis D, Nikolettos K, Vrettou K, Sarantis P, Koustas E, Kouraklis G, Antoniou EA, Karamouzis MV, Nikolettos N, Tsikouras P, Marinos G, Kontomanolis E, Kontzoglou K, Garmpis N. Evaluation of the Histone Deacetylase 2 (HDAC-2) Expression in Human Breast Cancer. Cancers (Basel) 2024; 16:209. [PMID: 38201636 PMCID: PMC10777907 DOI: 10.3390/cancers16010209] [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: 11/18/2023] [Revised: 12/20/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND/AIM Triple negative breast cancer belongs to the most aggressive breast cancer forms. Histone deacetylases (HDACs) constitute a class of enzymes that exhibit a significant role in breast cancer genesis and progression. In this study, we aimed at assessing the clinical importance of HDAC-2 in triple negative breast cancer. MATERIALS AND METHODS A total of 138 breast cancer specimens were examined on an immunohistochemical basis. A statistical analysis was performed in order to examine the association between HDAC-2 and the survival and clinicopathological features of the patients. RESULTS Increased HDAC-2 expression was observed in every fourth case of triple negative breast cancer with positive HDAC-2 staining, whereas only 12 out of 98 non-triple negative breast cancer samples showed high HDAC-2 expression. HDAC-2 overexpression correlated with prolonged overall survival (OS) and disease-free survival (DFS) in triple negative breast cancer. CONCLUSIONS High HDAC-2 levels in triple negative breast cancer seem to positively influence patient survival, disease stage and recurrence.
Collapse
Affiliation(s)
- Christos Damaskos
- Renal Transplantation Unit, Laiko General Hospital, 11527 Athens, Greece
- N.S. Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece (N.G.)
| | - Iason Psilopatis
- Department of Obstetrics and Gynecology, University Erlangen, Universitaetsstrasse 21–23, 91054 Erlangen, Germany
| | - Anna Garmpi
- First Department of Propedeutic Internal Medicine, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Dimitrios Dimitroulis
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Konstantinos Nikolettos
- Department of of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece (N.N.)
| | - Kleio Vrettou
- Department of Cytopathology, Sismanogleio General Hospital, 15126 Athens, Greece
| | - Panagiotis Sarantis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (E.K.)
| | - Evangelos Koustas
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (E.K.)
| | - Gregory Kouraklis
- Department of Surgery, Evgenideio Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Efstathios A. Antoniou
- N.S. Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece (N.G.)
- Department of General Surgery and HPB Surgery of Adults and Children, Hygeia Hospital, 15123 Athens, Greece
| | - Michail V. Karamouzis
- Molecular Oncology Unit, Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (P.S.); (E.K.)
| | - Nikolaos Nikolettos
- Department of of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece (N.N.)
| | - Panagiotis Tsikouras
- Department of of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece (N.N.)
| | - Georgios Marinos
- Department of Hygiene, Epidemiology and Medical Statistics, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Emmanouil Kontomanolis
- Department of of Obstetrics and Gynecology, Democritus University of Thrace, 68100 Alexandroupolis, Greece (N.N.)
| | - Konstantinos Kontzoglou
- N.S. Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece (N.G.)
- Second Department of Propedeutic Surgery, Laiko General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Nikolaos Garmpis
- N.S. Christeas Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece (N.G.)
- Department of General Surgery and HPB Surgery of Adults and Children, Hygeia Hospital, 15123 Athens, Greece
| |
Collapse
|
38
|
Yu B, Luo J, Yang Y, Zhen K, Shen B. Novel molecular insights into pyroptosis in triple-negative breast cancer prognosis and immunotherapy. J Gene Med 2024; 26:e3645. [PMID: 38041540 DOI: 10.1002/jgm.3645] [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: 08/31/2023] [Revised: 10/31/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND Patients with triple-negative breast cancer (TNBC) often have a poor prognostic outcome. Current treatment strategies cannot benefit all TNBC patients. Previous findings suggested pyroptosis as a novel target for suppressing cancer development, although the relationship between TNBC and pyroptosis-related genes (PRGs) was still unclear. METHODS Gene expression data and clinical follow-up of TNBC patients were collected from the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) and Gene Expression Omnibus (GEO). PRGs were screened using weighted gene co-expression network analysis. Cox regression analysis and the least absolute shrinkage and selection operator (i.e. LASSO) technique were applied to construct a pyroptosis-related prognostic risk score (PPRS) model, which was further combined with the clinicopathological characteristics of TNBC patients to develop a survival decision tree and a nomogram. The model was used to calculate the PPRS, and then the overall survival, immune infiltration, immunotherapy response and drug sensitivity of TNBC patients were analyzed based on the PPRS. RESULTS The PPRS model was closely related to clinicopathological features and can independently and accurately predict the prognosis of TNBC. According to normalized PPRS, patients in different cohorts were divided into two groups. Compared with the high-PPRS group, the low-PPRS group had significantly higher ESTIMATE (i.e. Estimation of STromal and Immune cells in MAlignantTumours using Expression data) score, immune score and stromal score, and it also had overexpressed immune checkpoints and significantly reduced Tumor Immune Dysfunction and Exclusion (TIDE) score, as well as higher sensitivity to paclitaxel, veliparib, olaparib and talazoparib. A decision tree and nomogram based on PPRS and clinical characteristics can improve the prognosis stratification and survival prediction for TNBC patients. CONCLUSIONS A PPRS model was developed to predict TNBC patients' immune characteristics and response to immunotherapy, chemotherapy and targeted therapy, as well as their survival outcomes.
Collapse
Affiliation(s)
- Bin Yu
- Linping Campus, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junjie Luo
- Linping Campus, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yifei Yang
- Linping Campus, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Zhen
- Linping Campus, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Binjie Shen
- Linping Campus, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
39
|
Kumari L, Mishra L, Patel P, Sharma N, Gupta GD, Kurmi BD. Emerging targeted therapeutic strategies for the treatment of triple-negative breast cancer. J Drug Target 2023; 31:889-907. [PMID: 37539789 DOI: 10.1080/1061186x.2023.2245579] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 07/30/2023] [Indexed: 08/05/2023]
Abstract
Triple-negative breast cancer (TNBC), a subtype of breast cancer that lacks expression of oestrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER-2), has clinical features including a high degree of invasiveness, an elevated risk of metastasis, tendency to relapse, and poor prognosis. It constitutes around 10-15% of all breast cancer, and having heredity of BRCA1 mutated breast cancer could be a reason for the occurrence of TNBC in women. Overexpression of cellular and molecular targets, i.e. CD44 receptor, EGFR receptor, Folate receptor, Transferrin receptor, VEGF receptor, and Androgen receptor, have emerged as promising targets for treating TNBC. Signalling pathways such as Notch signalling and PI3K/AKT/mTOR also play a significant role in carrying out and managing crucial pro-survival and pro-growth cellular processes that can be utilised for targeted therapy against triple-negative breast cancer. This review sheds light on various targeting strategies, including cellular and molecular targets, signalling pathways, poly (ADP-ribose) polymerase inhibitors, antibody-drug conjugates, and immune checkpoint inhibitors PARP, immunotherapy, ADCs have all found a place in the current TNBC therapeutic paradigm. The role of photothermal therapy (PTT) and photodynamic therapy (PDT) has also been explored briefly.
Collapse
Affiliation(s)
- Lakshmi Kumari
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| | - Lopamudra Mishra
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| | - Preeti Patel
- Department of Pharmaceutical Chemistry, ISF College Pharmacy, Moga, Punjab, India
| | - Nitin Sharma
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| | | | - Balak Das Kurmi
- Department of Pharmaceutics, ISF College Pharmacy, Moga, Punjab, India
| |
Collapse
|
40
|
Arvandi S, Razmjoo S, Zaheri Abdevand P. Risk factors and survival of triple-negative breast cancer among breast cancer patients: Ten-year cross-sectional study in the southwestern Iranian population. Health Sci Rep 2023; 6:e1767. [PMID: 38111745 PMCID: PMC10726015 DOI: 10.1002/hsr2.1767] [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] [Received: 09/15/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/20/2023] Open
Abstract
Background Breast cancer results from genetic and epigenetic mutations, contributing significantly to cancer-related morbidity and mortality. This study aimed to determine the prevalence and survival rates of triple-negative breast cancer (TNBC) among breast cancer patients in southwestern Iran over a ten-year period. Methods This retrospective cross-sectional study aims to assess prognostic factors associated with survival in women diagnosed with breast cancer in Iran's southwestern region over a ten-year period (2007-2017). Data were collected from patients who visited the Clinical Oncology Department at Golestan Hospital in Ahvaz (the breast cancer center of the Southwestern country). The study enrolled women diagnosed with TNBC using a census method and data from medical records. The primary outcome (survival rates) and secondary outcomes (demographic data, diagnostic stages, and three receptors estrogen receptors [ER], progesterone receptor [PR], human epidermal growth factor receptor 2 [HER2] status) were collected. Results Breast cancer was diagnosed in 2641 women over ten years; TNBC was diagnosed in 227 individuals (8.59%). Statistical analysis revealed a significant correlation between negative ER status and TNBC (p > 0.05). Furthermore, the prevalence of TNBC differed significantly from that of other types of breast cancer (p = 0.0001). The variables of age, HER2, PR, and TNBC grade did not differ significantly (p > 0.05). The overall disease-free survival rate over 5 years was 88.1%, while the rate for individuals without recurrence was 77.97%. Conclusion This study highlights a differentially low incidence of TNBC in the southwestern part of Iran when compared to other regions; genetic or epigenetic influences may explain this discrepancy. ER-negative status is a crucial prognostic indicator in diagnosing TNBC. The incidence of this disease is expected to rise by 100% in 2 years and 77.97% in 5 years.
Collapse
Affiliation(s)
- Sholeh Arvandi
- Department of Radiotherapy, Faculty of Medicine, Golestan HospitalAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Sasan Razmjoo
- Department of Radiotherapy, Faculty of Medicine, Golestan HospitalAhvaz Jundishapur University of Medical SciencesAhvazIran
| | | |
Collapse
|
41
|
Xing Y, Ba-Tu J, Dong C, Cao X, Li B, Jia X, Juan Y, Lv X, Zhang H, Qin N, Han W, Wang D, Qi X, Wang Y, Hao X, Zhang S, Du X, Wang H, Wang M. Phosphorylation of USP27X by GSK3β maintains the stability and oncogenic functions of CBX2. Cell Death Dis 2023; 14:782. [PMID: 38030604 PMCID: PMC10687032 DOI: 10.1038/s41419-023-06304-y] [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: 07/05/2023] [Revised: 10/25/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023]
Abstract
Chromobox protein homolog 2 (CBX2) exerts a multifaceted impact on the progression of aggressive cancers. The proteasome-dependent pathway is crucial for modulating CBX2 regulation, while the specific regulatory roles and mechanisms of deubiquitinating enzymes targeting CBX2 remain poorly understood. Mass spectrometry analysis identified ubiquitin-specific peptidase 27X (USP27X) as a deubiquitinating enzyme that targets CBX2. Overexpression of USP27X significantly enhances CBX2 levels by promoting deubiquitination, while deficiency of USP27X leads to CBX2 degradation, thereby inhibiting tumorigenesis. Furthermore, it has been revealed that glycogen synthase kinase 3 beta (GSK3β) can directly bind to and phosphorylate USP27X, thereby enhancing the interaction between USP27X and CBX2 and leading to further stabilization of the CBX2 protein. Clinically, the co-expression of high levels of USP27X and CBX2 in breast cancer tissues is indicative of a poor prognosis for patients with this disease. These findings collectively underscore the critical regulatory role played by USP27X in modulating CBX2, thereby establishing the GSK3β-USP27X-CBX2 axis as a pivotal driver of malignant progression in breast cancer.
Collapse
Affiliation(s)
- Yushu Xing
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- The Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Jirimu Ba-Tu
- Medical Innovation Center for Nationalities, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Chongyang Dong
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiaodong Cao
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- The Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Bing Li
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xin Jia
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yu Juan
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiaojie Lv
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Huiwen Zhang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Na Qin
- College of Mongolian Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Wuri Han
- College of Mongolian Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Dongfeng Wang
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiao Qi
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Yutong Wang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xulu Hao
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Shuang Zhang
- College of Traditional Chinese Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
| | - Xiaoli Du
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
- The Center for New Drug Safety Evaluation and Research, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| | - Huanyun Wang
- College of Pharmacy, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| | - Minjie Wang
- Medical Experimental Center of Basic Medical School, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China.
| |
Collapse
|
42
|
Kim L, Coman M, Pusztai L, Park TS. Neoadjuvant Immunotherapy in Early, Triple-Negative Breast Cancers: Catching Up with the Rest. Ann Surg Oncol 2023; 30:6441-6449. [PMID: 37349612 DOI: 10.1245/s10434-023-13714-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 05/23/2023] [Indexed: 06/24/2023]
Abstract
Despite breast cancer being long thought to be "immunologically cold," within early, triple-negative breast cancer (TNBC), there has been exciting advances with the use of immune checkpoint modulation combined with neoadjuvant chemotherapy. We review the major trials that have investigated combination immunochemotherapy in the neoadjuvant setting, reviewing both the pathological complete response rates and the maturing data regarding event-free and overall survival. Strategies to deescalate adjuvant therapy in patients with preserving excellent clinical outcome, as well as exploration of combinatorial adjuvant therapies to improve outcome in those with extensive residual are the next-generation challenges. In addition to refinement of existing biomarkers, such as PD-L1, TILs, and tumor mutational burden (TMB), exploration of topics like the microbiome as both a biomarker and a therapeutic has shown promise in other cancer types, which motivates investigating these in breast cancer.
Collapse
Affiliation(s)
- Leah Kim
- Section of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Magdalena Coman
- Section of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven, CT, USA
| | - Lajos Pusztai
- Yale School of Medicine, Yale Cancer Center, New Haven, CT, USA
| | - Tristen S Park
- Section of Surgical Oncology, Department of Surgery, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
43
|
Shao G, Fan X, Zhang P, Liu X, Huang L, Ji S. Circ_0004676 exacerbates triple-negative breast cancer progression through regulation of the miR-377-3p/E2F6/PNO1 axis. Cell Biol Toxicol 2023; 39:2183-2205. [PMID: 35870038 DOI: 10.1007/s10565-022-09704-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 02/23/2022] [Indexed: 12/09/2022]
Abstract
BACKGROUND The significant roles of circular RNAs (circRNAs) in different cancers and diseases have been reported. We now focused on the possible role of a newly recognized circRNA, circ_0004674 in triple-negative breast cancer (TNBC), and the related downstream mechanism. METHODS The expression of circ_0004674 in TNBC tissues and cells was determined followed by analysis of the correlation between circ_0004674 and TNBC patients' prognosis. The interaction between circ_0004674, miR-377-3p, E2F6, and PNO1 was then identified using bioinformatics analysis combined with FISH, RIP, RNA pull-down, RT-qPCR, and Western blot analysis. Using gain-of-function and loss-of-function methods, we analyzed the effect of circ_0004674, miR-377-3p, E2F6, and PNO1 on TNBC in vivo and in vitro. RESULTS Increased circ_0004674 and E2F6 but decreased miR-377-3p were observed in TNBC tissues and MDA-MB-231 TNBC cells, all of which findings were associated with poor prognosis in patients with TNBC. Silencing of circ_0004676 remarkably suppressed the proliferation, cell cycle progression, and migration of TNBC cells in vitro, as well as inhibiting tumorigenesis and metastasis in vivo. Additionally, circ_0004676 served as a sponge of miR-377-3p which bound to the transcription factor E2F6. In the presence of overexpression of circ_0004676, E2F6 expression and its target PNO1 expression were elevated, while miR-377-3p expression was decreased. Interestingly, overexpression of E2F6 could reverse the inhibitory effect on tumor growth caused by downregulation of circ_0004676. CONCLUSION Our study highlighted the carcinogenic effect of circ_0004676 on TNBC through regulation of the miR-377-3p/E2F6/PNO1 axis. 1. Circ_0004674 is highly expressed in TNBC tissues and cells. 2. Circ_0004674 upregulates the expression of E2F6 by sponging miR-377-3p. 3. E2F6 upregulates PNO1 by binding to the PNO1 promoter. 4. Circ_0004674 favors TNBC progression by regulating the miR-377-3p/E2F6/PNO1 axis. 5. This study provides a new target for the treatment of TNBC.
Collapse
Affiliation(s)
- Guoli Shao
- Special Medical Service Center, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Road, Haizhu District, Guangzhou, 510282, People's Republic of China
| | - Xulong Fan
- Department of Breast Surgery, Maternity and Children's Healthcare Hospital of Foshan, Foshan, 528000, People's Republic of China
| | - Pusheng Zhang
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Xuewen Liu
- Special Medical Service Center, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Road, Haizhu District, Guangzhou, 510282, People's Republic of China
| | - Lei Huang
- Department of General Surgery, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, People's Republic of China
| | - Shufeng Ji
- Special Medical Service Center, Zhujiang Hospital of Southern Medical University, No. 253, Middle Gongye Road, Haizhu District, Guangzhou, 510282, People's Republic of China.
| |
Collapse
|
44
|
Adrada BE, Moseley TW, Kapoor MM, Scoggins ME, Patel MM, Perez F, Nia ES, Khazai L, Arribas E, Rauch GM, Guirguis MS. Triple-Negative Breast Cancer: Histopathologic Features, Genomics, and Treatment. Radiographics 2023; 43:e230034. [PMID: 37792593 PMCID: PMC10560981 DOI: 10.1148/rg.230034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 05/09/2023] [Accepted: 06/01/2023] [Indexed: 10/06/2023]
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous and aggressive group of tumors that are defined by the absence of estrogen and progesterone receptors and lack of ERBB2 (formerly HER2 or HER2/neu) overexpression. TNBC accounts for 8%-13% of breast cancers. In addition, it accounts for a higher proportion of breast cancers in younger women compared with those in older women, and it disproportionately affects non-Hispanic Black women. TNBC has high metastatic potential, and the risk of recurrence is highest during the 5 years after it is diagnosed. TNBC exhibits benign morphologic imaging features more frequently than do other breast cancer subtypes. Mammography can be suboptimal for early detection of TNBC owing to factors that include the fast growth of this cancer, increased mammographic density in young women, and lack of the typical features of malignancy at imaging. US is superior to mammography for TNBC detection, but benign-appearing features can lead to misdiagnosis. Breast MRI is the most sensitive modality for TNBC detection. Most cases of TNBC are treated with neoadjuvant chemotherapy, followed by surgery and radiation. MRI is the modality of choice for evaluating the response to neoadjuvant chemotherapy. Survival rates for individuals with TNBC are lower than those for persons with hormone receptor-positive and human epidermal growth factor receptor 2-positive cancers. The 5-year survival rates for patients with localized, regional, and distant disease at diagnosis are 91.3%, 65.8%, and 12.0%, respectively. The early success of immunotherapy has raised hope regarding the development of personalized strategies to treat TNBC. Imaging and tumor biomarkers are likely to play a crucial role in the prediction of TNBC treatment response and TNBC patient survival in the future. ©RSNA, 2023 Quiz questions for this article are available in the supplemental material.
Collapse
Affiliation(s)
- Beatriz E. Adrada
- From the Departments of Breast Imaging (B.E.A., T.W.M., M.M.K.,
M.E.S., M.M.P., F.P., E.S.N., E.A., G.M.R., M.S.G.), Breast Surgical Oncology
(T.W.M.), Pathology-Anatomical (L.K.), and Abdominal Imaging (G.M.R.), The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1350,
Houston, TX 77030
| | - Tanya W. Moseley
- From the Departments of Breast Imaging (B.E.A., T.W.M., M.M.K.,
M.E.S., M.M.P., F.P., E.S.N., E.A., G.M.R., M.S.G.), Breast Surgical Oncology
(T.W.M.), Pathology-Anatomical (L.K.), and Abdominal Imaging (G.M.R.), The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1350,
Houston, TX 77030
| | - Megha M. Kapoor
- From the Departments of Breast Imaging (B.E.A., T.W.M., M.M.K.,
M.E.S., M.M.P., F.P., E.S.N., E.A., G.M.R., M.S.G.), Breast Surgical Oncology
(T.W.M.), Pathology-Anatomical (L.K.), and Abdominal Imaging (G.M.R.), The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1350,
Houston, TX 77030
| | - Marion E. Scoggins
- From the Departments of Breast Imaging (B.E.A., T.W.M., M.M.K.,
M.E.S., M.M.P., F.P., E.S.N., E.A., G.M.R., M.S.G.), Breast Surgical Oncology
(T.W.M.), Pathology-Anatomical (L.K.), and Abdominal Imaging (G.M.R.), The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1350,
Houston, TX 77030
| | - Miral M. Patel
- From the Departments of Breast Imaging (B.E.A., T.W.M., M.M.K.,
M.E.S., M.M.P., F.P., E.S.N., E.A., G.M.R., M.S.G.), Breast Surgical Oncology
(T.W.M.), Pathology-Anatomical (L.K.), and Abdominal Imaging (G.M.R.), The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1350,
Houston, TX 77030
| | - Frances Perez
- From the Departments of Breast Imaging (B.E.A., T.W.M., M.M.K.,
M.E.S., M.M.P., F.P., E.S.N., E.A., G.M.R., M.S.G.), Breast Surgical Oncology
(T.W.M.), Pathology-Anatomical (L.K.), and Abdominal Imaging (G.M.R.), The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1350,
Houston, TX 77030
| | - Emily S. Nia
- From the Departments of Breast Imaging (B.E.A., T.W.M., M.M.K.,
M.E.S., M.M.P., F.P., E.S.N., E.A., G.M.R., M.S.G.), Breast Surgical Oncology
(T.W.M.), Pathology-Anatomical (L.K.), and Abdominal Imaging (G.M.R.), The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1350,
Houston, TX 77030
| | - Laila Khazai
- From the Departments of Breast Imaging (B.E.A., T.W.M., M.M.K.,
M.E.S., M.M.P., F.P., E.S.N., E.A., G.M.R., M.S.G.), Breast Surgical Oncology
(T.W.M.), Pathology-Anatomical (L.K.), and Abdominal Imaging (G.M.R.), The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1350,
Houston, TX 77030
| | - Elsa Arribas
- From the Departments of Breast Imaging (B.E.A., T.W.M., M.M.K.,
M.E.S., M.M.P., F.P., E.S.N., E.A., G.M.R., M.S.G.), Breast Surgical Oncology
(T.W.M.), Pathology-Anatomical (L.K.), and Abdominal Imaging (G.M.R.), The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1350,
Houston, TX 77030
| | - Gaiane M. Rauch
- From the Departments of Breast Imaging (B.E.A., T.W.M., M.M.K.,
M.E.S., M.M.P., F.P., E.S.N., E.A., G.M.R., M.S.G.), Breast Surgical Oncology
(T.W.M.), Pathology-Anatomical (L.K.), and Abdominal Imaging (G.M.R.), The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1350,
Houston, TX 77030
| | - Mary S. Guirguis
- From the Departments of Breast Imaging (B.E.A., T.W.M., M.M.K.,
M.E.S., M.M.P., F.P., E.S.N., E.A., G.M.R., M.S.G.), Breast Surgical Oncology
(T.W.M.), Pathology-Anatomical (L.K.), and Abdominal Imaging (G.M.R.), The
University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1350,
Houston, TX 77030
| |
Collapse
|
45
|
Wu J, Li J, Xu H, Qiu N, Huang X, Li H. Periostin drives extracellular matrix degradation, stemness, and chemoresistance by activating the MAPK/ERK signaling pathway in triple-negative breast cancer cells. Lipids Health Dis 2023; 22:153. [PMID: 37716956 PMCID: PMC10504790 DOI: 10.1186/s12944-023-01912-1] [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: 06/19/2023] [Accepted: 08/29/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND Adipose tissue, which is mainly composed of adipocytes, is a crucial component of the tumor microenvironment, particularly in breast cancer. Adipocytes surround breast cancer cells and may participate in cell‒cell interactions in the breast microenvironment. However, little is currently known about how adipocytes influence the biological behavior of the surrounding breast cancer cells. Hence, this study sought to investigate the role and underlying mechanisms of periostin in triple-negative breast cancer (TNBC) cells cocultured with adipogenic conditioned medium (ACM) and palmitic acid (PA). METHODS Human TNBC cell lines (MDA‒MB‒231 and SUM159PT) were treated with ACM and PA, then the expression of periostin, matrix metalloproteinases (MMPs) and stemness-related molecules were assessed by Western blotting and RT‒qPCR. The cellular viability was assessed using CCK‒8 assay. Plasmid transfection, RNA sequencing, and pathway inhibitor were used to explore the specific mechanisms of periostin. RESULTS ACM and PA elevated the expression of both MMPs and stemness-related molecules in TNBCs. MMPs can promote tumor cell infiltration and migration by degrading the extracellular matrix, and stemness expression increases the development of tumor chemoresistance. Additionally, ACM and PA increased periostin expression, while inhibiting periostin disrupted the involvement of ACM and PA in promoting extracellular matrix degradation, stemness, and chemoresistance in TNBCs. Furthermore, this study found that periostin promoted TNBC progression by activating the MAPK/ERK signaling pathway and that inhibition of MAPK/ERK signaling reduced the phenotype caused by periostin upregulation in TNBCs treated with ACM or PA. Finally, the present results showed that the high expression of POSTN, which encodes periostin, was substantially related to worse survival in TNBC patients. CONCLUSIONS The results of the study elucidated for the first time how periostin is the key protein secreted in TNBCs in response to the adipocyte-regulated tumor microenvironment, while periostin-neutralizing antibodies may serve as potential therapeutic agents in relation to TNBC progression.
Collapse
Affiliation(s)
- Jinna Wu
- Department of Breast Oncology Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Jia Li
- Department of Breast Oncology Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Huiya Xu
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Ni Qiu
- Department of Breast Oncology Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Xiaojia Huang
- Department of Breast Oncology Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China
| | - Hongsheng Li
- Department of Breast Oncology Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, 510095, China.
| |
Collapse
|
46
|
Gupta SRR, Ta TM, Khan M, Singh A, Singh IK, Peethambaran B. Identification and validation of a small molecule targeting ROR1 for the treatment of triple negative breast cancer. Front Cell Dev Biol 2023; 11:1243763. [PMID: 37779899 PMCID: PMC10534069 DOI: 10.3389/fcell.2023.1243763] [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] [Received: 06/21/2023] [Accepted: 08/11/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction: Breast cancer is the most common cancer in women, with roughly 10-15% of new cases classified as triple-negative breast cancer (TNBC). Traditional chemotherapies are often toxic to normal cells. Therefore, it is important to discover new anticancer compounds that target TNBC while causing minimal damage to normal cells. Receptor tyrosine kinase-like Orphan Receptor 1 (ROR1) is an oncofetal protein overexpressed in numerous human malignancies, including TNBC. This study investigated potential small molecules targeting ROR1. Methodology: Using AutoDock Vina and Glide, we screened 70,000 chemicals for our investigation. We obtained 10 representative compounds via consensus voting, deleting structural alerts, and clustering. After manual assessment, compounds 2 and 4 were chosen for MD simulation and cell viability experiment. Compound 4 showed promising results in the viability assay, which led us to move further with the apoptosis assay and immunoblotting. Results: Compound 4 (CID1261330) had docking scores of -6.635 and -10.8. It fits into the pocket and shows interactions with GLU64, ASP174, and PHE93. Its RMSD fluctuates around 0.20 nm and forms two stable H-bonds indicating compound 4 stability. It inhibits cell proliferation in MDA-MB-231, HCC1937, and HCC1395 cell lines, with IC50 values of approximately 2 μM to 10 μM, respectively. Compound 4 did not kill non-malignant epithelial breast cells MCF-10A (IC50 > 27 μM). These results were confirmed by the significant number of apoptotic cells in MDA-MB-231 cells (47.6%) but not in MCF-10A cells (7.3%). Immunoblot analysis provided additional support in the same direction. Discussion: These findings collectively suggest that compound 4 has the potential to effectively eliminate TNBC cells while causing minimal harm to normal breast cells. The promising outcomes of this study lay the groundwork for further testing of compound 4 in other malignancies characterized by ROR1 upregulation, serving as a proof-of-concept for its broader applicability.
Collapse
Affiliation(s)
- Shradheya R. R. Gupta
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India
| | - Tram M. Ta
- Department of Biology, Saint Joseph’s University, Philadelphia, PA, United States
| | - Maryam Khan
- Department of Biology, Saint Joseph’s University, Philadelphia, PA, United States
| | - Archana Singh
- Department of Botany, Hans Raj College, University of Delhi, New Delhi, India
| | - Indrakant K. Singh
- Molecular Biology Research Laboratory, Department of Zoology, Deshbandhu College, University of Delhi, New Delhi, India
- Delhi School of Public Health, Institute of Eminence, University of Delhi, New Delhi, India
- Norris Comprehensive Cancer Center, Division of Medical Oncology, University of Southern California, Los Angeles, CA, United States
| | - Bela Peethambaran
- Department of Biology, Saint Joseph’s University, Philadelphia, PA, United States
| |
Collapse
|
47
|
Li L, Lin Z, Xu X, Wang W, Chen H, Feng Z, Yang Z, Hao J. A pH/GSH/Glucose Responsive Nanozyme for Tumor Cascade Amplified Starvation and Chemodynamic Theranostics. ACS APPLIED MATERIALS & INTERFACES 2023; 15:41224-41236. [PMID: 37615578 DOI: 10.1021/acsami.3c05412] [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: 08/25/2023]
Abstract
Nanozymes have brought enormous opportunities for disease theranostics. Here, a self-enhanced catalytic nanocrystal based on a bismuth-manganese core-shell nanoflower containing glucose oxide (GOx), termed BDS-GOx@MnOx, was designed for 4T1 tumor theranostics in vitro and in vivo. The BDS-GOx@MnOx nanozymes enable enhanced starvation treatment (ST) and chemotherapy (CDT) with high efficacy and exhibit sensitive tumor microenvironment (TME) responsive character for tumor therapy as well as for tumor-enhanced computer tomography (CT) and magnetic resonance (MR) diagnostic imaging. The characters and mechanism of the BDS-GOx@MnOx nanozymes have also been systematically studied and revealed.
Collapse
Affiliation(s)
- Lihua Li
- Future Institute of Technology, School of Information and Optoelectronic Science and Engineering, South China Normal University, Guangzhou 510515, P. R. China
- Department of Applied Physics, The Hong Kong Polytechnic University, 999077 Hong Kong, P. R. China
| | - Zefeng Lin
- The State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, School of Materials Science and Technology, South China University of Technology, Guangzhou, Guangdong 510640, P. R. China
- Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Orthopedic Center, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, Guangdong, P. R. China
| | - Xingyi Xu
- The State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Fiber Laser Materials and Applied Techniques, School of Materials Science and Technology, South China University of Technology, Guangzhou, Guangdong 510640, P. R. China
| | - Wanshun Wang
- Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Orthopedic Center, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, Guangdong, P. R. China
| | - Hu Chen
- Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Orthopedic Center, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, Guangdong, P. R. China
| | - Zhibin Feng
- Key Laboratory of Trauma & Tissue Repair of Tropical Area of PLA, Orthopedic Center, General Hospital of Southern Theater Command of PLA, Guangzhou 510010, Guangdong, P. R. China
| | - Zhongmin Yang
- Future Institute of Technology, South China Normal University, Guangzhou 510631, P. R. China
| | - Jianhua Hao
- Department of Applied Physics, The Hong Kong Polytechnic University, 999077 Hong Kong, P. R. China
| |
Collapse
|
48
|
Gonzàlez-Farré M, Gibert J, Santiago-Díaz P, Menéndez S, Monzonis X, Olivares F, Riera X, López D, Torner A, Casado B, Bellosillo B, Lloveras B, Casadevall D, Rovira A, Servitja S, Albanell J, Vázquez I, Comerma L. Characterization and spatial distribution of the immune cell infiltrate in triple-negative breast cancer: a novel classification based on plasma cells and CD8+ T cells. Hum Pathol 2023; 139:91-105. [PMID: 37517596 DOI: 10.1016/j.humpath.2023.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/01/2023]
Abstract
Stromal tumor-infiltrating lymphocytes (sTILs) are a robust prognostic and predictive biomarker in triple-negative breast carcinoma. However, the sTIL compartment comprises different cell populations. The aim of the study is to characterize the distribution of T cells (CD3+ and CD8+), B cells, and plasma cells and explore their association with outcome in the surgical specimen of 62 patients. Furthermore, programmed death ligand 1 expression and the presence of tertiary lymphoid structures (TLSs) are explored. Patients with higher sTILs achieve better progression-free survival (PFS) (P = .0013), and tumors have more plasma cells in the infiltrate. Specifically, higher counts of T cells (both CD3+ and CD8+) have better PFS (P = .002 and P = .0086, respectively) as it is observed in tumors with higher infiltration of CD8+ T cells in the tumor core (P = .035). Higher infiltration by B cells and plasma cells shows a positive tendency toward increased PFS (P = .06 and P = .058). Programmed death ligand 1 (SP142) is positive in 56% of tumors. Tumors with at least 1 TLS (42%) show higher CD8+ T cell infiltration in the tumor core and the sTIL value doubles compared to tumors devoid of TLSs [sTIL mean: 36 ± 11% and 18 ± 5% (CI [Confidence Interval]: 95%), respectively]. Our study demonstrates that the characterization of the immune cell infiltration is as relevant as its distribution. Moreover, the importance of considering different immune cell types for classification is emphasized. Therefore, a new classification of triple-negative breast carcinoma immune infiltration with CD8+ T cell and plasma cell densities in the tumor core and infiltrative margin is proposed.
Collapse
Affiliation(s)
- Mònica Gonzàlez-Farré
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain; Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain.
| | - Joan Gibert
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain; Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
| | | | - Silvia Menéndez
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
| | - Xavier Monzonis
- Department of Medical Oncology, Hospital del Mar, 08003 Barcelona, Spain
| | | | - Xènia Riera
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain
| | - David López
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain
| | - Ariadna Torner
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain
| | - Beatriz Casado
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain
| | - Beatriz Bellosillo
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain; Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain; Department of Medicine and Life Sciences (MELIS), University Pompeu Fabra, Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Belén Lloveras
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain; Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain; Department of Medicine and Life Sciences (MELIS), University Pompeu Fabra, Doctor Aiguader 88, 08003 Barcelona, Spain
| | - David Casadevall
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
| | - Ana Rovira
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain; Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain
| | - Sònia Servitja
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain; Department of Medical Oncology, Hospital del Mar, 08003 Barcelona, Spain
| | - Joan Albanell
- Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain; Department of Medical Oncology, Hospital del Mar, 08003 Barcelona, Spain; Center for Biomedical Network Research on Cancer (CIBERONC), 28029 Madrid, Spain; Department of Medicine and Life Sciences (MELIS), University Pompeu Fabra, Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Ivonne Vázquez
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain
| | - Laura Comerma
- Department of Pathology, Hospital del Mar, 08003 Barcelona, Spain; Cancer Research Program, IMIM (Hospital del Mar Medical Research Institute), 08003 Barcelona, Spain
| |
Collapse
|
49
|
Song SE, Woo OH, Cho Y, Cho KR, Park KH, Kim JW. Prediction of Axillary Lymph Node Metastasis in Early-stage Triple-Negative Breast Cancer Using Multiparametric and Radiomic Features of Breast MRI. Acad Radiol 2023; 30 Suppl 2:S25-S37. [PMID: 37331865 DOI: 10.1016/j.acra.2023.05.025] [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: 04/24/2023] [Revised: 05/21/2023] [Accepted: 05/22/2023] [Indexed: 06/20/2023]
Abstract
RATIONALE AND OBJECTIVES To investigate whether machine learning (ML) approaches using breast magnetic resonance imaging (MRI)-derived multiparametric and radiomic features could predict axillary lymph node metastasis (ALNM) in stage I-II triple-negative breast cancer (TNBC). MATERIALS AND METHODS Between 2013 and 2019, 86 consecutive patients with TNBC who underwent preoperative MRI and surgery were enrolled and divided into ALNM (N = 27) and non-ALNM (n = 59) groups according to histopathologic results. For multiparametric features, kinetic features using computer-aided diagnosis (CAD), morphologic features, and apparent diffusion coefficient (ADC) values at diffusion-weighted images were evaluated. For extracting radiomic features, three-dimensional segmentation of tumors using T2-weighted images (T2WI) and T1-weighted subtraction images were respectively performed by two radiologists. Each predictive model using three ML algorithms was built using multiparametric features or radiomic features, or both. The diagnostic performances of models were compared using the DeLong method. RESULTS Among multiparametric features, non-circumscribed margin, peritumoral edema, larger tumor size, and larger angio-volume at CAD were associated with ALNM in univariate analysis. In multivariate analysis, larger angio-volume was the sole statistically significant predictor for ALNM (odds ratio = 1.33, P = 0.008). Regarding ADC values, there were no significant differences according to ALNM status. The area under the receiver operating characteristic curve for predicting ALNM was 0.74 using multiparametric features, 0.77 using radiomic features from T1-weighted subtraction images, 0.80 using radiomic features from T2WI, and 0.82 using all features. CONCLUSION A predictive model incorporating breast MRI-derived multiparametric and radiomic features may be valuable in predicting ALNM preoperatively in patients with TNBC.
Collapse
Affiliation(s)
- Sung Eun Song
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea (S.E.S., Y.C., KRC)
| | - Ok Hee Woo
- Department of Radiology, Korea University Guro Hospital, Korea University College of Medicine, Seoul, Korea (O.H.W.).
| | - Yongwon Cho
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea (S.E.S., Y.C., KRC)
| | - Kyu Ran Cho
- Department of Radiology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea (S.E.S., Y.C., KRC)
| | - Kyong Hwa Park
- Department of Oncology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea (K.H.P., J.W.K.)
| | - Ju Won Kim
- Department of Oncology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Korea (K.H.P., J.W.K.)
| |
Collapse
|
50
|
Michlewska S, Wójkowska D, Watala C, Skiba E, Ortega P, de la Mata FJ, Bryszewska M, Ionov M. Ruthenium metallodendrimer against triple-negative breast cancer in mice. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 53:102703. [PMID: 37591367 DOI: 10.1016/j.nano.2023.102703] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 07/11/2023] [Accepted: 08/05/2023] [Indexed: 08/19/2023]
Abstract
Carbosilane metallodendrimers, based on the arene Ru(II) complex (CRD13) and integrated to imino-pyridine surface groups have been investigated as an anticancer agent in a mouse model with triple-negative breast cancer. The dendrimer entered into the cells efficiently, and exhibited selective toxicity for 4T1 cells. In vivo investigations proved that a local injection of CRD13 caused a reduction of tumour mass and was non-toxic. ICP analyses indicated that Ru(II) accumulated in all tested tissues with a greater content detected in the tumour.
Collapse
Affiliation(s)
- Sylwia Michlewska
- Laboratory of Microscopic Imaging and Specialized Biological Techniques, Faculty of Biology and Environmental Protection, University of Lodz, Poland; Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland.
| | - Dagmara Wójkowska
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Poland
| | - Cezary Watala
- Department of Haemostatic Disorders, Faculty of Health Sciences, Medical University of Lodz, Poland
| | - Elżbieta Skiba
- Institute of General and Ecological Chemistry, Lodz University of Technology, Poland
| | - Paula Ortega
- Universidad de Alcalá, Department of Organic and Inorganic Chemistry, Research Institute in Chemistry "Andrés M. del Río" (IQAR), Madrid, Spain; Instituto de Investigación Sanitaria Ramón y Cajal, IRYCIS, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Francisco Javier de la Mata
- Universidad de Alcalá, Department of Organic and Inorganic Chemistry, Research Institute in Chemistry "Andrés M. del Río" (IQAR), Madrid, Spain; Instituto de Investigación Sanitaria Ramón y Cajal, IRYCIS, Spain; Networking Research Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Spain
| | - Maria Bryszewska
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland
| | - Maksim Ionov
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Poland
| |
Collapse
|