1
|
Joshi S, Garlapati C, Nguyen T, Sharma S, Chandrashekar DS, Bhattarai S, Varambally S, Krishnamurti U, Li X, Aneja R. C/EBPβ increases tumor aggressiveness by enhancing KIFC1 expression in androgen receptor negative triple negative breast cancer. Cell Commun Signal 2025; 23:255. [PMID: 40448099 DOI: 10.1186/s12964-025-02243-7] [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: 03/02/2025] [Accepted: 05/09/2025] [Indexed: 06/02/2025] Open
Abstract
Quadruple-negative breast cancers (also known as AR-triple negative (TN) BC) lack the expression of estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), and androgen receptor (AR). AR-TNBC exhibits aggressive characteristics and a poor prognosis. Because of the lack of expression of therapeutic targets, limited therapeutic options exist for patients with AR-TNBC. Hence, new therapeutic targets and risk-predictive biomarkers are required for patients with AR-TNBC. In this study, we investigated the role of kinesin-like protein 1 (KIFC1) in AR-TNBC. We found that C/EBPβ binds to the KIFC1 promoter and induces its expression in the AR-TNBC cells. Notably, AR status was negatively correlated with KIFC1 levels. We also found that AR transcriptionally repressed the transcription factor C/EBPβ, which regulates the expression of KIFC1. The lack of AR expression in AR-TNBC led to C/EBPβ upregulation, thereby enhancing KIFC1 expression. Moreover, upregulation of KIFC1 in AR-TNBC increased cancer cell proliferation and promoted epithelial-mesenchymal transition (EMT), contributing to the aggressive characteristics of AR-TNBC. Inhibiting KIFC1 using the small molecule inhibitor CW069 significantly reduced tumor volume in mice bearing AR-TNBC xenografts, but not in those with triple-negative breast tumors. These data suggest that upregulation of C/EBPβ and KIFC1 contributes to the aggressive characteristics and poor prognosis of AR-TNBC, providing strong evidence that targeting KIFC1 using kinesin inhibitors could be a viable therapeutic approach for patients with AR-TNBC.
Collapse
Affiliation(s)
- Shriya Joshi
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
- Alkermes Inc, Waltham, MA, 02451, USA
- Discovery and Development Sciences, Leads Discovery and Optimization, Bristol Myers Squibb, Cambridge, MA, 02141, USA
| | - Chakravarthy Garlapati
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA.
- Alkermes Inc, Waltham, MA, 02451, USA.
| | - Thi Nguyen
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Shaligram Sharma
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
- Chemical Insights Research Institute of UL Research Institutes, Marietta, GA, 30067, USA
| | | | - Shristi Bhattarai
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA
| | - Sooryanarayana Varambally
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Uma Krishnamurti
- Department of Pathology, Yale University, New Haven, CT, 06510, USA
| | - Xiaoxian Li
- Department of Pathology & Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, 30303, USA.
- Department of Nutrition Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- O'Neal Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
2
|
Wu Q, Siddharth S, Verma D, Parida S, Sharma D. TRIM29 upregulation contributes to chemoresistance in triple negative breast cancer via modulating S100P-β-catenin axis. Cell Commun Signal 2025; 23:244. [PMID: 40420099 DOI: 10.1186/s12964-025-02233-9] [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: 11/20/2024] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Triple negative breast cancer, an inherently aggressive disease, is further impaired by the limited therapeutic options and chemotherapy-resistance; hence, elucidating the signaling nodes underlying chemotherapy resistance is of major interest. Focusing on the differentially expressed genes in recurrent TNBC, we identified TRIM29, a ubiquitin ligase belonging to TRIM family, as a uniquely enriched protein in chemoresistant TNBC. Here, we demonstrate that chemoresistant TNBC cells are inherently aggressive, exhibiting elevated growth and migration potential compared to chemosensitive cells, and in particular, they possess higher TRIM29 expression whose expression level modulation results in altered chemosensitivity. TRIM29 overexpression reduces chemotherapy response whereas TRIM29 knockout not only increases chemosensitivity but also reduces TNBC tumor growth. Tumor-dissociated cells maintain TRIM29 knockout status as well as exhibit similar functional alterations as chemoresistant TNBC cells. Mechanistically, RNA-sequencing of parental-chemosensitive, chemoresistant-inherently overexpressing TRIM29 and chemoresistant-TRIM29 knockout TNBC cells reveals a unique set of genes (S100P, SERPINB3, SERPINB4, CEACAM5, CEACAM6 and CDH6) showing significant upregulation with the acquisition of chemoresistance and downregulation with the TRIM29 knockout. Furthermore, an enrichment of β-catenin pathway in chemoresistant TNBC cells is observed. We uncovered a functional network where S100P, a metastasis inducing secretory factor, bidirectionally interacts with TRIM29, and modulates the expression of SERPINB3, SERPINB4, CEACAM5, CEACAM6 as well as β-catenin pathway genes. Showing the functional importance, S100P inhibitor reduces the growth and mammosphere formation in chemoresistant TNBC. Moreover, combining β-catenin inhibitor with chemotherapy shows synergistic inhibition of chemoresistant TNBC cells. Indeed, higher expression of TRIM29, S100P and β-catenin associates with reduced recurrence free survival. This work proposes TRIM29 as an important node that modulates a unique gene network in chemoresistant TNBC and whose biological impact is mediated by modulation of S100P and β-catenin.
Collapse
Affiliation(s)
- Qitong Wu
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA
| | - Sumit Siddharth
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA.
| | - Deepak Verma
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA
| | - Sheetal Parida
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA
| | - Dipali Sharma
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB 1, Rm 145, Baltimore, MD, 21231, USA.
| |
Collapse
|
3
|
Funasaka C, Kogawa T, Sakamoto N, Kusuhara S, Nakao T, Nakajima H, Kondoh C, Harano K, Matsubara N, Hosono A, Naito Y, Shimokawa M, Watanuki R, Yamashita Y, Yamauchi C, Onishi T, Ishii G, Mukohara T. The utility of immunohistochemistry-based biomarkers in predicting the pathological complete response in early-stage triple-negative breast cancer. Cancer Treat Res Commun 2025; 44:100941. [PMID: 40424708 DOI: 10.1016/j.ctarc.2025.100941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 04/17/2025] [Accepted: 05/02/2025] [Indexed: 05/29/2025]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of BC. A pathological complete response (pCR) to neoadjuvant chemotherapy is strongly associated with a favorable TNBC prognosis; however, established predictors of pCR, including tumor-infiltrating lymphocytes (TILs), are often not adequately reliable in the clinic. This study evaluated the utility of immunohistochemistry (IHC)-based markers and TILs in predicting pCR in early-stage TNBC. This study enrolled 61 women with stage I-III TNBC who were treated at our institution between January 2013 and December 2019. Pathological data were collected from electronic medical records, while IHC data were obtained from preoperative biopsy specimens. Fisher's test, multivariable logistic regression, and correlation analyses were used to identify biomarker candidates and their interactions. The majority of the patients had stage II or III invasive ductal TNBC. The pCR rate was 31 % (19/61). High TIL frequencies (≥ 40 %) and high Ki-67 (≥ 40 %) levels were associated with pCR. Among the patients with high TIL frequencies, AR-negative patients had a higher pCR rate than AR-positive patients (55.0 % versus 16.7 %; p = 0.71). Vimentin negativity correlated with high TIL frequencies (p = 0.02). High TIL frequencies and high Ki-67 levels were independently associated with an increased likelihood of achieving a pCR. The combination of high TIL frequencies and high Ki-67 levels was predictive of pCR in patients with primary TNBC, while AR and vimentin represent candidate markers that require further validation. Further studies should evaluate the performance of these markers in combination with other biomarkers and in the context of immune-checkpoint blockade.
Collapse
Affiliation(s)
- Chikako Funasaka
- Departments of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan; Departments of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Takahiro Kogawa
- Departments of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan; Departments of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan; Department of Advanced Medical Development, Cancer Institute Hospital of Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| | - Naoya Sakamoto
- Departments of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Shota Kusuhara
- Departments of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Takehiro Nakao
- Departments of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Hiromichi Nakajima
- Departments of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Chihiro Kondoh
- Departments of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Kenichi Harano
- Departments of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan; Departments of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Nobuaki Matsubara
- Departments of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Ako Hosono
- Departments of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan; Departments of Pediatric Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Yoichi Naito
- Departments of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan; Departments of Experimental Therapeutics, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Mototsugu Shimokawa
- Department of Biostatistics, Yamaguchi University Graduate School of Medicine, 1-1-1, Minamikogushi, Ube City, Yamaguchi, 755-0046, Japan
| | - Rurina Watanuki
- Departments of Breast Surgery, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Yuji Yamashita
- Departments of Breast Surgery, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Chisako Yamauchi
- Departments of Breast Surgery, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Tatsuya Onishi
- Departments of Breast Surgery, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Genichiro Ishii
- Departments of Pathology and Clinical Laboratories, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| | - Toru Mukohara
- Departments of Medical Oncology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, 277-8577, Japan
| |
Collapse
|
4
|
Weng CC, Chiang CC, Chung YH, Ho YP, Chang YC, Hsiao IT, Mach RH. Evaluation of a simplified radiolabeling method for a PARP inhibitor in an animal model of breast cancer. EJNMMI Res 2025; 15:50. [PMID: 40301197 PMCID: PMC12040802 DOI: 10.1186/s13550-025-01236-4] [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: 12/27/2024] [Accepted: 04/05/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Several poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors were recently approved by the US Food and Drug Administration for use in cancer treatment. To facilitate the discovery of novel PARP-targeting ligands, a radioiodinated ligand, I-125-KX1, was developed and validated for its specificity to PARP-1; however, its preparation procedure is time-consuming. The present study employed a solid-phase extraction (SPE) method in the radiolabeling procedure of I-123/125-KX1 and evaluated its binding specificity by using receptor binding assays, autoradiography, and in vivo single photon emission computed tomography (SPECT) imaging technique. RESULTS Through the incorporation of the SPE purification method as the final step in the radioiodination procedure, the resultant product I-123/125-KX1 exhibited high radiochemical purity (> 99%) and an acceptable radiochemical yield (58.6% for I-123-KX1, 73.3% for I-125-KX1). The binding characteristics of this radiotracer were validated through saturation binding assays conducted on MDA-MB-231 and MCF-7 cells. The Kd values obtained for the tracer (~ 1.0 nM) was consistent with values reported in the literature, and the Bmax values of these two cell lines (2017 ± 178 fmol/mg on MDA-MB-231 vs. 1393 ± 105 fmol/mg on MCF-7) were in line with the results from Western blot analyses. To demonstrate the in vivo imaging ability of I-123-KX1 prepared in this study, an MDA-MB-231 tumor animal model was used and the tracer displayed a suitable uptake on the tumor tissues (6.9 ± 0.8%ID/mL). The binding specificity of the SPE-purified I-125-KX1 was further verified using in vitro autoradiography in conjunction with various PARP inhibitors. Additionally, an anti-PARP-1 immunohistochemistry experiment was conducted, which revealed that the autoradiograms of the radiotracer displayed a similar pattern. CONCLUSIONS This suggests that the I-123/125-KX1 prepared using the SPE method showed some comparable properties to those from the traditional method, indicating its potential suitability for future radioligand preparation in PARP studies. However, further characterization studies may be needed to confirm its efficacy.
Collapse
Affiliation(s)
- Chi-Chang Weng
- Department of Medical Imaging and Radiological Sciences, College of Medicine & Healthy Aging Center, Chang Gung University, Taoyuan, Taiwan.
- Department of Medical Research and Development, Research Division, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan.
- Department of Medical Imaging and Radiological Sciences, Chang Gung University, Taoyuan, Taiwan.
| | - Chao-Chih Chiang
- Department of Isotope Application Research, National Atomic Research Institute, Taoyuan, Taiwan
| | - Yi-Hsiu Chung
- Department of Medical Research and Development, Research Division, Chang Gung Memorial Hospital at Linkou, Taoyuan, 333, Taiwan
| | - Yi-Pei Ho
- Department of Medical Imaging and Radiological Sciences, College of Medicine & Healthy Aging Center, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Chuan Chang
- Department of Medical Imaging and Radiological Sciences, College of Medicine & Healthy Aging Center, Chang Gung University, Taoyuan, Taiwan
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Ing-Tsung Hsiao
- Department of Medical Imaging and Radiological Sciences, College of Medicine & Healthy Aging Center, Chang Gung University, Taoyuan, Taiwan
- Department of Nuclear Medicine and Molecular Imaging Center, Chang Gung Memorial Hospital, Linkou, Taoyuan, Taiwan
| | - Robert H Mach
- Division of Nuclear Medicine and Clinical Molecular Imaging, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
5
|
Ashrafi P, Sari S, Javani Jouni F, Zafari J, Asgari F. Potentiated Effects of Photobiomodulation and Celecoxib on the Epithelial-Mesenchymal Transition Signaling of E-Cadherin, N-Cadherin, α-SMA in Breast Cancer Cells, MCF7, and MDA-MB-231. Photobiomodul Photomed Laser Surg 2025; 43:115-123. [PMID: 39992209 DOI: 10.1089/photob.2024.0155] [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: 02/25/2025] Open
Abstract
Background and Objective: Breast cancer (BC) is one of the most common cancers among women, with a high potential for metastasis. The epithelial-mesenchymal transition (EMT) is crucial in the invasion and metastasis of cancer cells. This research was designed to examine the efficacy of photobiomodulation therapy in combination with celecoxib in inhibiting the EMT process. We also analyzed the changes in the expression of E-cadherin, N-cadherin, and α-SMA genes in BC cell lines MCF-7 and MDA-MB-231. Material and Methods: In this study, the IC50 of celecoxib was first determined using the 3-(4,5-dimethylthiazol-2-yl)2,5-diphenyltetrazolium bromide assay for both cell lines. The cells were then treated with celecoxib, laser irradiation, and their combination. A migration assay was performed to evaluate the cell migration. Real-time polymerase chain reaction also assessed the changes in the expression of the abovementioned genes. Results: A combination of celecoxib and laser therapy significantly reduced the migration of cancer cells. Additionally, the potentiated effect of the combined therapy altered the expression levels of the aforementioned genes, indicating the potential role of the combination treatment in regulating EMT. Conclusions: Our research discloses that combining laser therapy with celecoxib could serve as an effective therapeutic approach to inhibit BC invasion and metastasis by targeting the EMT process and decelerating disease progression. Further investigations are essential to validate these results in clinical environments.
Collapse
Affiliation(s)
- Parisa Ashrafi
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Soyar Sari
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Javani Jouni
- Department of Biochemistry and Biophysics, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Jaber Zafari
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fatemeh Asgari
- Department of Cellular and Molecular Biology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Laser Application in Medical Sciences Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Chen YZ, Xu S, Ren H, Zhang J, Jia Y, Sun H. Characterization of novel sialylation-associated microRNA signature for prognostic assessment in breast cancer and its implications for the tumor microenvironment. J Steroid Biochem Mol Biol 2025; 248:106683. [PMID: 39900230 DOI: 10.1016/j.jsbmb.2025.106683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/05/2025]
Abstract
Sialylation, a key post-translational modification essential for protein function, is regulated by steroid hormones, along with other glycosylations like fucosylation. These modifications influence tumor growth and metastasis by modulating immune activation. MicroRNAs (miRNAs), crucial in gene expression, affect sialylation and are emerging as promising biomarkers in breast cancer, though their prognostic value remains unclear. Sialylation-related miRNAs were identified through Pearson correlation analysis, and an eight-miRNA risk signature was developed using univariate and Least Absolute Shrinkage and Selection Operator (LASSO) regression in the TCGA dataset. The prognostic value was validated in two independent GEO datasets. Multivariate analysis confirmed that the miRNA risk score is an independent predictor of overall survival (OS). A nomogram integrating clinical characteristics and the risk score was created to predict 1-, 3-, and 5-year OS, assessed through calibration curves, ROC curves, and area under the ROC curve (AUC). Biological pathways were explored using GSEA and GSVA, while immune infiltrates were identified through CIBERSORT and TIMER. The eight-miRNA signature effectively predicted OS, recurrence-free survival, and disease-free survival. High-risk patients exhibited increased macrophage and neutrophil levels, indicative of a poor prognosis. High-risk patients, especially those with triple-negative breast cancer, had significantly worse outcomes. This risk score could inform personalized treatment strategies in breast cancer management.
Collapse
Affiliation(s)
- Yong-Zi Chen
- Laboratory of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Shilei Xu
- Laboratory of Tumor Cell Biology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Hailing Ren
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Jun Zhang
- Department of Breast Cancer, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Yongsheng Jia
- Department of Breast Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Haiyan Sun
- Department of Integrative Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education; Key Laboratory of Cancer Prevention and Therapy, Tianjin; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| |
Collapse
|
7
|
Chang CC, Li HJ, Satange R, Lin SM, Chen TL, Lin CC, Neidle S, Hou MH. Structural and Functional Insights into Targeting GCCG Sites in the EGFR Promoter by Two DNA Intercalators to Inhibit Breast Cancer Metastasis. J Med Chem 2025; 68:6601-6615. [PMID: 40032551 PMCID: PMC11956004 DOI: 10.1021/acs.jmedchem.4c03203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 02/10/2025] [Accepted: 02/25/2025] [Indexed: 03/05/2025]
Abstract
Chemotherapeutic drugs are commonly used to treat cancers lacking targeted therapy options. However, their low specificity limits their treatment effectiveness. We report here that the cooperative binding of doxorubicin (Dox) with actinomycin D (ActD) enhances the specificity for consecutive GCCG sites in DNA. Using X-ray crystallography, we determined the crystal structure of ActD and Dox bound to d(AGCCGT)2 DNA. ActD intercalation at the GpC site induces a novel Dox binding mode at the adjacent CpG step. This ensures a snug fit, avoids steric clashes, and enhances the specificity. Transcriptome analysis revealed that combining Dox with ActD synergistically down-regulates EGFR in TNBC cells. Additionally, it reduces EGFR promoter activity. In vivo, the combination significantly suppresses tumor growth and outperforms the standard Dox and cyclophosphamide regimen in inhibiting metastasis. This study highlights targeting the activated EGFR pathway with sequence-specific DNA-targeting drug combinations as a potential TNBC treatment.
Collapse
Affiliation(s)
- Chih-Chun Chang
- Graduate
Institute of Biotechnology, National Chung
Hsing University, Taichung 402, Taiwan
| | - Hsin-Ju Li
- Graduate
Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Roshan Satange
- Graduate
Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Shan-Meng Lin
- Graduate
Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
| | - Tai-Lin Chen
- Post
Baccalaureate Medicine, School of Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Chi-Chien Lin
- Institute
of Biomedical Science, National Chung Hsing
University, Taichung 402, Taiwan
| | - Stephen Neidle
- The
School of Pharmacy, University College London, London WC1N 1AX, U.K.
| | - Ming-Hon Hou
- Graduate
Institute of Biotechnology, National Chung
Hsing University, Taichung 402, Taiwan
- Graduate
Institute of Genomics and Bioinformatics, National Chung Hsing University, Taichung 402, Taiwan
- Biotechnology
Center, National Chung Hsing University, Taichung 402, Taiwan
| |
Collapse
|
8
|
De K, Jana M, Chowdhury B, Calaf GM, Roy D. Role of PARP Inhibitors: A New Hope for Breast Cancer Therapy. Int J Mol Sci 2025; 26:2773. [PMID: 40141415 PMCID: PMC11942994 DOI: 10.3390/ijms26062773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/05/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Tumors formed by the unchecked growth of breast cells are known as breast cancer. The second most frequent cancer in the world is breast cancer. It is the most common cancer among females. In 2022, 2,296,840 women were diagnosed with breast cancer. The therapy of breast cancer is evolving through the development of Poly (ADP-ribose) polymerase (PARP) inhibitors, which are offering people with specific genetic profiles new hope as research into the disease continues. It focuses on patients with BRCA1 and BRCA2 mutations. This review summarizes the most recent research on the mechanisms of action of PARP inhibitors and their implications for breast cancer therapy. We review how therapeutic applications are developing and highlight recent studies showing the effectiveness of these medicines whether used alone or in combination. Furthermore, the significance of customized therapy is highlighted in enhancing patient outcomes as we address the function of genetic testing in identifying candidates for PARP inhibition. Recommendations for future research areas to maximize the therapeutic potential of PARP inhibitors are also included, along with challenges and limits in their clinical usage. The objective of this review is to improve our comprehension of the complex interaction between breast cancer biology and PARP inhibition. This knowledge will help to guide screening approaches, improve clinical practice, and support preventive initiatives for people at risk.
Collapse
Affiliation(s)
- Kamalendu De
- Department of Biological Sciences (Botany), Midnapore City College, Midnapore 721129, West Bengal, India;
| | - Malabendu Jana
- Department of Neurological Science, Rush University School of Medicine, Chicago, IL 773, USA;
| | - Bhabadeb Chowdhury
- HIV Dynamics and Replication Program, National Institute of Health, Frederick, MD 21702, USA;
| | - Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile
| | - Debasish Roy
- Department of Natural Sciences, Hostos College of The City University of New York, Bronx, NY 718, USA;
| |
Collapse
|
9
|
Bouzid RS, Bouzid R, Labed H, Serhani I, Hellal D, Oumeddour L, Boudhiaf I, Ibrir M, Khadraoui H, Belaaloui G. Molecular subtyping and target identification in triple negative breast cancer through immunohistochemistry biomarkers. BMC Cancer 2025; 25:454. [PMID: 40082760 PMCID: PMC11905517 DOI: 10.1186/s12885-025-13832-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 02/27/2025] [Indexed: 03/16/2025] Open
Abstract
BACKGROUND The Triple-Negative Breast Cancer (TNBC) molecular subtyping and target identification based on Immunohistochemistry (IHC) is of considerable worth for routine use. Yet, literature on this topic is limited worldwide and needs to be enriched with data from different populations. METHODS We assessed the IHC expression of subtyping biomarkers (Cytokeratins 5, 14 and 17, Epidermal Growth Factor Receptor, Claudins 3 and 7, E-cadherin, Vimentin and Androgen receptor) and predictive biomarkers (Tumor-infiltrating lymphocytes (TILs) density, Breast Cancer Antigen 1 (BRCA1) and P53) in a cohort of TNBC patients. Clinicopathologic parameters and overall survival (OS) were investigated as well. RESULTS The patients were aged 50.11 ± 12.13y (more than 40y in 76.56% of patients), and 23.44% had a BC family history. They were in a non-advanced stage: 51.6% T2 stage, 56.2% negative lymph node involvement, 76.6% without metastasis and 64.1% grade II Scarff-Bloom-Richardson classification (SBR). The IHC subtypes were: 53.1% Basal-like1 (BL1), 6.3% Basal-like2 (BL2), 17.2% Mesenchymal (MES), 9.4% Luminal Androgen Receptor (LAR), 4.7% Mixed subtype and 9.4% "Unclassified" type. The LAR subtype involved the youngest patients (40.17 ± 8.68y, p = 0.02). The "Unclassified" subtype expressed the p53 mutated-type pattern more frequently (100%, p = 0.07). The BRCA1 mutated pattern and TILs infiltration were present in (23.44% and 37.5% of patients, respectively). The OS of the subtypes differed significantly (p = 0.007, log-rank test). The subtypes median OS were, respectively, 15.47 mo. (Unclassified), 18.94 mo. (BL2), 27.23 mo. (MES), 27.28 mo. (Mixed), 30.88 mo. (BL1), and 45.07 mo. (LAR). There was no difference in the OS following age, BRCA1 expression, p53 pattern and TILs density. Though, the OS following the TNM stage was different (p = 0.001). A multivariable Cox proportional hazards regression analysis showed that TNM staging and TNBC subtypes, independently influence the OS (p < 0.001 and p = 0.017, respectively). Hence, IHC is useful in TNBC subtyping for prognostic purposes and in the identification of therapeutic biomarkers. Further investigation is required to confirm our results and to implement IHC as a routine tool to improve patient's care.
Collapse
Affiliation(s)
- Rima Saad Bouzid
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Department of Biology of Organisms, Faculty of Natural and Life Sciences, University of Batna 2, 05000, Batna, Algeria
| | - Radhia Bouzid
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Department of Biology of Organisms, Faculty of Natural and Life Sciences, University of Batna 2, 05000, Batna, Algeria
| | - Housna Labed
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Department of Biology of Organisms, Faculty of Natural and Life Sciences, University of Batna 2, 05000, Batna, Algeria
| | - Iman Serhani
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Dounia Hellal
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Leilia Oumeddour
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Ines Boudhiaf
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Massouda Ibrir
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, University Hospital, Batna, Algeria
| | - Hachani Khadraoui
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria
- Faculty of Medicine, University Batna 2, Batna, Algeria
- Pathology Department, Cancer Control Center (CLCC), Batna, Algeria
| | - Ghania Belaaloui
- Laboratory of Acquired and Constitutional Genetic Diseases (MAGECA), Faculty of Medicine, University of Batna 2, 05000, Batna, Algeria.
- Faculty of Medicine, University Batna 2, Batna, Algeria.
| |
Collapse
|
10
|
Spurnić I, Šušnjar S, Jovanić I, Medić-Miljić N, Milovanović Z, Popović Krneta M, Bukumirić Z, Gavrilović D, Rajšić S, Marković I. The Diagnostics of Disease Relapse Within Five-Year Follow-Up and the Role of Androgen Receptor Expression in Patients with Early Triple-Negative Breast Cancer. Diagnostics (Basel) 2025; 15:692. [PMID: 40150035 PMCID: PMC11941219 DOI: 10.3390/diagnostics15060692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Triple-negative breast cancer (TNBC) is characterized by the absence of the expression of estrogen receptors, progesterone receptors, and human epidermal growth factor receptor 2. As there are no specific targeted therapies, TNBC patients often face an aggressive clinical course. The expression of the androgen receptor (AR) has been found in up to 30% of TNBC cases, but the association between the AR status and survival rates in TNBC remains controversial. The aim of this study was to explore the association of AR expression with the disease outcome in patients with early TNBC within a 5-year follow-up. Methods: AR expression was determined by immunohistochemistry in a cohort of 124 early-TNBC patients treated at the Institute for Oncology and Radiology of Serbia. The cut-off value used for the positive AR status was >10% tumor cells. The association of the AR status with clinicopathological factors (age, stage, tumor diameter, lymph node invasion, metastatic spread, Ki-67 score, EGFR score, and cytokeratin 5/6 score) and the disease outcome (disease-free survival-DFS-and overall survival-OS) was investigated. Results: Our analysis showed that the AR-positive status was associated with a significantly lower Ki-67 score compared to the AR-negative samples. A univariate analysis indicated that the age, tumor size, nodal status, and EGFR score significantly influenced both 5-year DFS and OS. Multivariate Cox analysis suggested that a smaller tumor size, lower nodal status, and AR expression were independent predictors of longer survival rates in TNBC patients. Conclusions: The results of this study suggest that the positive AR status may be a favorable prognostic factor in TNBC patients within the first five years after surgery.
Collapse
Affiliation(s)
- Igor Spurnić
- Surgical Oncology Clinic, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Snežana Šušnjar
- Clinic for Medical Oncology, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia
| | - Irena Jovanić
- Department of Pathology, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (I.J.); (N.M.-M.); (Z.M.)
| | - Nataša Medić-Miljić
- Department of Pathology, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (I.J.); (N.M.-M.); (Z.M.)
| | - Zorka Milovanović
- Department of Pathology, Institute of Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (I.J.); (N.M.-M.); (Z.M.)
| | - Marina Popović Krneta
- Department of Nuclear Medicine, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia;
| | - Zoran Bukumirić
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Institute of Medical Statistics and Informatics, 11000 Belgrade, Serbia
| | - Dušica Gavrilović
- Data Center, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia;
| | - Saša Rajšić
- Department of Anesthesiology and Intensive Care Medicine, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Ivan Marković
- Surgical Oncology Clinic, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| |
Collapse
|
11
|
Wang TT, Zhang LL, Li FB, Zhang J, Zhang ZB, Mi DZ, Sun J, Zhang HY, Wang CY, Chen YH, Chen CS. LN-439A, a novel BAP1 inhibitor, suppresses the growth of basal-like breast cancer by degrading KLF5. Acta Pharmacol Sin 2025; 46:715-727. [PMID: 39379684 PMCID: PMC11845570 DOI: 10.1038/s41401-024-01361-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 07/17/2024] [Indexed: 10/10/2024]
Abstract
Basal-like breast cancer (BLBC) is the most malignant subtype of breast cancer because of its aggressive clinical behaviour and lack of effective targeted agents. Krüppel-like factor 5 (KLF5) is an oncogenic transcription factor that is highly expressed in BLBC. The deubiquitinase (DUB) BRCA1-associated protein 1 (BAP1) stabilizes KLF5 and promotes BLBC growth and metastasis. Therefore, pharmacological inhibition of the BAP1‒KLF5 axis is an effective therapeutic strategy for BLBC. Here, through screening, we identified a series of tetrahydro-β-carboline derivatives that effectively reduced the protein expression of KLF5 and exhibited strong antitumour activity. Among the investigated compounds, the lead compound LN-439A presented the strongest antitumour activity and inhibitory effect on KLF5 expression. LN-439A suppressed the proliferation and migration of BLBC cells, induced G2/M arrest, and induced apoptosis. Mechanistically, LN-439A functions as a small molecule catalytic inhibitor of BAP1 by binding to the catalytic pocket of BAP1, leading to the ubiquitination and degradation of KLF5. Consistent with this finding, the overexpression of KLF5 suppressed the antitumour effects of LN-439A. In summary, LN-439A is a promising therapeutic agent for BLBC that functions by targeting the BAP1‒KLF5 axis.
Collapse
Affiliation(s)
- Tian-Tian Wang
- School of Life Science, University of Science and Technology of China, Hefei, 230027, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Long-Long Zhang
- Academy of Biomedical Engineering, Kunming Medical University, Kunming, 650500, China
| | - Fu-Bing Li
- Academy of Biomedical Engineering, Kunming Medical University, Kunming, 650500, China
| | - Jie Zhang
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Zhi-Bi Zhang
- Academy of Biomedical Engineering, Kunming Medical University, Kunming, 650500, China
| | - Da-Zhao Mi
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Jian Sun
- The Third Affiliated Hospital, Kunming Medical University, Kunming, 650118, China
| | - Hong-Yan Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Faculty of Life science and Technology, Kunming University of Science and Technology, Kunming, 650500, China
| | - Chun-Yan Wang
- Department of the Pathology, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
| | - Yi-Hua Chen
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
- School of Pharmaceutical Sciences and Yunnan Key Laboratory of Pharmacology for Natural Products, Kunming Medical University, Kunming, 650500, China.
- Yunnan College of Modern Biomedical Industry, Kunming Medical University, Kunming, 650500, China.
| | - Ce-Shi Chen
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences and Yunnan Province, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Academy of Biomedical Engineering, Kunming Medical University, Kunming, 650500, China.
- The Third Affiliated Hospital, Kunming Medical University, Kunming, 650118, China.
| |
Collapse
|
12
|
Hashmi AA, Ajaz Y, Sajjad M, Zia F, Irfan M, Abu Bakar SM, Khan EY, Faridi N. Predictive Value of Excision Repair Cross Complementation Group 1 (ERCC1) by Immunohistochemistry for Determining Neoadjuvant Chemotherapy Response in Triple-Negative Breast Cancers. Breast J 2025; 2025:8410670. [PMID: 40008380 PMCID: PMC11858828 DOI: 10.1155/tbj/8410670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 01/30/2025] [Indexed: 02/27/2025]
Abstract
Introduction: Triple-negative breast cancers (TNBCs) constitute a significant proportion of breast cancers in Pakistan. Owing to the lack of expression of hormone (estrogen/progesterone) receptor and human epidermal growth factor receptor 2 (HER2neu), treatment options for TNBCs are limited. Therefore, it is important to identify markers that predict response to chemotherapy in these patients. Previous studies have demonstrated that the excision repair cross complementation group 1 (ERCC1) protein can successfully augur the response to chemotherapy in cancer; however, data related to TNBCs, particularly in Pakistan, are limited. Therefore, in this study, we evaluated the role of ERCC1 in predicting the response to neoadjuvant chemotherapy in patients with TNBCs. Methods: This cross-sectional study was conducted at the Liaquat National Hospital, Histopathology Department, between January 2019 and June 2023. A total of 132 biopsy-proven cases of breast cancer that were negative for estrogen receptor (ER), progesterone receptor (PR), and HER/2neu and were administered neoadjuvant chemotherapy before surgery were included in the study. ERCC1 immunohistochemical (IHC) staining was performed on prechemotherapy needle biopsies. The results were scored semiquantitatively by assessing the average intensity on a scale of 0-3 (0, no staining; 1, weak nuclear staining; 2, intermediate nuclear staining; and 3, strong nuclear staining) and the proportion of tumor cells showing positive nuclear staining. The intensity and proportion scores were then multiplied to give a score that was divided by 100 to give an overall score, and scores equal to or higher than 1.0 were considered positive. Neoadjuvant chemotherapy response was categorized as pathological complete response (pCR) when no residual invasive breast carcinoma was found on the postneoadjuvant chemotherapy excision specimen and as pathological partial response (pPR) when residual cancer cells were present in admixed chemotherapy-related changes. The residual cancer burden (RCB) was calculated using the MD Anderson RCB calculator. The association between ERCC1 expression and the chemotherapy response/RCB class was determined. Results: We found that 90.9% (n = 120) of TNBC cases expressed ERCC1, whereas pCR was noted in 24 (18.2%) cases. A significant association was observed between ERCC1 expression and pCR. Cases with negative ERCC1 expression had a significantly higher frequency of pCR (66.7%) than those with positive ERCC1 expression (13.3%). Additionally, the ERCC1-positive group showed a higher frequency of RCB classes II (36.7%) and III (43.3%) than the ERCC1-negative group (RCB II: 25%; RCB III: 0%). Moreover, positive ERCC1 expression was associated with higher nodal (N) stage. Conclusion: In this study, we established the role of negative ERCC1 expression in predicting the response to chemotherapy in neoadjuvant TNBC. Therefore, ERCC1 can be used as a predictive marker to stratify patients who will benefit from neoadjuvant therapy. Moreover, we also noted an association between ERCC1 expression and nodal metastasis; however, more large-scale studies are needed to establish its role as a prognostic biomarker in TNBC.
Collapse
Affiliation(s)
- Atif Ali Hashmi
- Department of Histopathology, Liaquat National Hospital and Medical College, Karachi, Pakistan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York City, New York, USA
| | - Yumna Ajaz
- Department of Histopathology, Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Muhsana Sajjad
- Department of Pathology, Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Fazail Zia
- Department of Pathology, Jinnah Sindh Medical University, Karachi, Pakistan
| | - Muhammad Irfan
- Department of Statistics, Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Syed Muhammad Abu Bakar
- Department of Histopathology, Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Erum Yousuf Khan
- Department of Histopathology, Liaquat National Hospital and Medical College, Karachi, Pakistan
| | - Naveen Faridi
- Department of Histopathology, Liaquat National Hospital and Medical College, Karachi, Pakistan
| |
Collapse
|
13
|
Nedeljković M, Vuletić A, Mirjačić Martinović K. Divide and Conquer-Targeted Therapy for Triple-Negative Breast Cancer. Int J Mol Sci 2025; 26:1396. [PMID: 40003864 PMCID: PMC11855393 DOI: 10.3390/ijms26041396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive and malignant type of breast cancer with limited treatment options and poor prognosis. One of the most significant impediments in TNBC treatment is the high heterogeneity of this disease, as highlighted by the detection of several molecular subtypes of TNBC. Each subtype is driven by distinct mutations and pathway aberrations, giving rise to specific molecular characteristics closely connected to clinical behavior, outcomes, and drug sensitivity. This review summarizes the knowledge regarding TNBC molecular subtypes and how it can be harnessed to devise tailored treatment strategies instead of blindly using targeted drugs. We provide an overview of novel targeted agents and key insights about new treatment modalities with an emphasis on the androgen receptor signaling pathway, cancer stem cell-associated pathways, phosphatidylinositol 3-kinase (PI3K)/AKT pathway, growth factor signaling, and immunotherapy.
Collapse
Affiliation(s)
- Milica Nedeljković
- Department of Experimental Oncology, Institute for Oncology and Radiology of Serbia, 11000 Belgrade, Serbia; (A.V.); (K.M.M.)
| | | | | |
Collapse
|
14
|
Lo C, Chang DY, Lu YS, Wang MY, Tsai LW, Huang CS, Tang CH, Lin CH. Benefit of adjuvant chemotherapy for T1cN0M0 and selected T1bN0M0 triple-negative breast cancer: a nationwide cancer registry-based study. Oncologist 2025; 30:oyae346. [PMID: 40042201 PMCID: PMC11880992 DOI: 10.1093/oncolo/oyae346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 11/13/2024] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND The efficacy of adjuvant chemotherapy for T1N0M0 triple-negative breast cancer (TNBC) has not been clearly elucidated. Thus, we aimed to evaluate the efficacy of adjuvant chemotherapy for patients with T1a-cN0M0 TNBC. PATIENTS AND METHODS Patients newly diagnosed with TNBC between 2011 and 2015 were identified and followed up until the end of 2020 using the Taiwan Cancer Registry. Univariate and multivariate Cox proportional hazards regression analyses were performed to compare the recurrence-free survival (RFS) and OS between patients who received and those who did not receive adjuvant chemotherapy. RESULTS Of the 62 483 patients registered during 2011-2015, 1074 patients with T1N0M0 TNBC (T1a, n = 103; T1b, n = 167; and T1c, n = 804) who underwent definitive breast surgery were included. Overall, 850 (79%) patients received adjuvant chemotherapy; these comprised 24.3%, 67.7%, and 88.6% of the patients with T1a, T1b, and T1c disease, respectively. Over a median follow-up of 7.18 years, a significant RFS and OS benefit from adjuvant chemotherapy was observed in the T1c subgroup but not in the T1a and T1b subgroups. However, subgroup analysis of T1b disease indicated that adjuvant chemotherapy yielded an OS benefit to patients with histological grade III disease (adjusted hazard ratio = 0.08, 95% CI, 0.01-0.77; P = .03). CONCLUSIONS Adjuvant chemotherapy improved the RFS and OS in patients with T1cN0M0 TNBC and improved the OS in patients with histological grade III T1bN0M0 disease. Our study advocates for the utilization of adjuvant chemotherapy in patients diagnosed with T1cN0M0 and histological grade III T1bN0M0 TNBC.
Collapse
Affiliation(s)
- Chiao Lo
- Department of Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100229, Taiwan
| | - Dwan-Ying Chang
- Department of Oncology, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Medical Oncology, National Taiwan University Hospital, Cancer Center Branch, Taipei 106037, Taiwan
| | - Yen-Shen Lu
- Department of Oncology, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Medical Oncology, National Taiwan University Hospital, Cancer Center Branch, Taipei 106037, Taiwan
| | - Ming-Yang Wang
- Department of Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Surgical Oncology, National Taiwan University Hospital, Cancer Center Branch, Taipei 106037, Taiwan
| | - Li-Wei Tsai
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100229, Taiwan
- Department of Surgical Oncology, National Taiwan University Hospital, Cancer Center Branch, Taipei 106037, Taiwan
| | - Chiun-Sheng Huang
- Department of Surgery, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Surgery, College of Medicine, National Taiwan University, Taipei 100233, Taiwan
| | - Chao-Hsiun Tang
- School of Health Care Administration, College of Management, Taipei Medical University, Taipei 235603, Taiwan
| | - Ching-Hung Lin
- Department of Oncology, National Taiwan University Hospital, Taipei 100225, Taiwan
- Department of Medical Oncology, National Taiwan University Hospital, Cancer Center Branch, Taipei 106037, Taiwan
| |
Collapse
|
15
|
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
|
16
|
Than PP, Yao SJ, Althagafi E, Kaur K. A Conjugate of an EGFR-Binding Peptide and Doxorubicin Shows Selective Toxicity to Triple-Negative Breast Cancer Cells. ACS Med Chem Lett 2025; 16:109-115. [PMID: 39811122 PMCID: PMC11726362 DOI: 10.1021/acsmedchemlett.4c00480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/04/2024] [Accepted: 12/06/2024] [Indexed: 01/16/2025] Open
Abstract
Selective targeting of cancer cells via overexpressed cell-surface receptors is a promising strategy to enhance chemotherapy efficacy and minimize off-target side effects. In this study, we designed peptide 31 (YHWYGYTPERVI) to target the overexpressed epidermal growth factor receptor (EGFR) in triple-negative breast cancer (TNBC) cells. Peptide 31 is internalized by TNBC cells through EGFR-mediated endocytosis and shares sequence and structural similarities with human EGF (hEGF), a natural EGFR ligand. Unlike hEGF, peptide 31 does not induce cell migration in TNBC cells. A novel conjugate of peptide 31 with doxorubicin (Dox) retains selectivity for TNBC cells and exhibits significant toxicity comparable to that of unconjugated Dox. Importantly, this conjugate shows no toxicity toward normal breast epithelial cells up to a high concentration (25 μM). Thus, peptide 31 serves as a versatile targeting ligand for developing novel conjugates with high selectivity for EGFR-positive cancers.
Collapse
Affiliation(s)
- Phi-Phung Than
- Chapman
University School of Pharmacy, Irvine, California 92618, United States
| | - Shih-Jing Yao
- Chapman
University School of Pharmacy, Irvine, California 92618, United States
| | - Emad Althagafi
- Chapman
University School of Pharmacy, Irvine, California 92618, United States
| | - Kamaljit Kaur
- Chapman
University School of Pharmacy, Irvine, California 92618, United States
| |
Collapse
|
17
|
Däster K, Hench J, Diepenbruck M, Volkmann K, Rouchon A, Palafox M, Miragaya JG, Preca BT, Kurzeder C, Weber WP, Bentires-Alj M, Soysal SD, Muenst S. BRCA promoter methylation in triple-negative breast cancer is preserved in xenograft models and represents a potential therapeutic marker for PARP inhibitors. Breast Cancer Res Treat 2025; 209:389-396. [PMID: 39392573 PMCID: PMC11785619 DOI: 10.1007/s10549-024-07502-8] [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: 11/30/2023] [Accepted: 09/23/2024] [Indexed: 10/12/2024]
Abstract
PURPOSE Most triple-negative breast cancers (TNBC) are sporadic in nature and often associated with dysfunction of the BRCA1 or BRCA2 genes. Since somatic BRCA mutations are rare in breast cancer (BC), this dysfunction frequently is the result of BRCA promoter methylation. Despite the phenotypic similarities of these tumors to those with germline or somatic BRCA mutation, the evidence of response to PARP inhibitors is unclear. METHODS We analyzed the prevalence of BRCA promoter methylation in 29 BC metastases through the well-established Illumina Infinium EPIC Human Methylation Bead Chip. In cases with BRCA methylation, the xenograft of the same tumor was tested. Additionally, we compared BC xenografts with an identified BRCA methylation to their matched primary tumors and subsequently investigated the efficacy of PARP inhibitors on tumor organoids from a BRCA2 promoter-methylated BC. RESULTS BRCA2 promotor hypermethylation was identified in one pleural metastasis of a young patient as well as in the xenograft tissue. We also identified five more xenograft models with BRCA2 promotor hypermethylation. Analysis of one matched primary tumor confirmed the same BRCA2 methylation. PARP inhibitor treatment of tumor organoids derived from the BRCA2 methylated xenograft tumor tissue of the young patient showed a significant decline in cell viability, similar to organoids with somatic BRCA1 mutation, while having no effect on organoids with BRCA1 wildtype. CONCLUSION BRCA promotor hypermethylation seems to be a rare event in metastatic BC but is preserved in subsequent xenograft models and might represent an attractive therapeutic marker for PARP inhibitors.
Collapse
Affiliation(s)
- Kavitha Däster
- Breast Center Zurich, Zurich, Switzerland.
- University of Basel, Basel, Switzerland.
| | - Jürgen Hench
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Maren Diepenbruck
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Katrin Volkmann
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Adelin Rouchon
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Marta Palafox
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Jorge Gomez Miragaya
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Bogdan Tiberius Preca
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Christian Kurzeder
- University of Basel, Basel, Switzerland
- Breast Center, University Hospital Basel, Basel, Switzerland
| | - Walter Paul Weber
- University of Basel, Basel, Switzerland
- Breast Center, University Hospital Basel, Basel, Switzerland
| | - Mohamed Bentires-Alj
- Tumor Heterogeneity Metastasis and Resistance, Department of Biomedicine, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Savas Deniz Soysal
- University of Basel, Basel, Switzerland
- Praxis Chirurgie Im Zentrum, Basel, Switzerland
| | - Simone Muenst
- University of Basel, Basel, Switzerland
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
18
|
Vashisth P, Smith CL, Amarasekara DL, Dasanyake GS, Singh G, Chism CM, Hamadani CM, Shaikh T, Grovich N, Gamboa B, Fitzkee NC, Hammer NI, Tanner EEL. Choline Carboxylic Acid Ionic Liquid-stabilized Anisotropic Gold Nanoparticles for Photothermal Therapy. ACS APPLIED NANO MATERIALS 2024; 7:26332-26343. [PMID: 39935958 PMCID: PMC11810124 DOI: 10.1021/acsanm.3c04645] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/13/2025]
Abstract
Gold nanoparticles (AuNPs) are commonly used in cancer research due to their unique physical and optical properties. However, current AuNP synthesis methods often involve cytotoxic cationic surfactants like cetyltrimethyl ammonium bromide (CTAB). Tedious CTAB replacement methodologies have been used to increase the biocompatibility, further increasing the complexity of synthesizing biocompatible AuNPs limiting their biomedical applications. To address this issue, we explore cholinium decanoate (CADA) ionic liquid (IL) as a biocompatible stabilizing agent by replacing CTAB using a simple modified seeded method for synthesizing anisotropic AuNPs for photothermal therapy of triple-negative breast cancer cells (MDA-MB-231). The prepared CADA AuNPs showed quasi-spherical morphology, confirmed by transmission electron microscopy (TEM) and a broad plasmonic absorption band using Vis-NIR spectroscopy. CADA AuNPs exhibited excellent in-vitro biocompatibility with both MCF-10A (healthy human mammary cells) and MDA-MB-231 cells. We evaluate their in-vitro photothermal efficacy against MDA-MB-231 cancer cells, demonstrating significant cell death even at low AuNP concentration (20 μg/mL), low laser power density (0.6 W/cm2, 808 nm continuous laser), and short irradiation time of 5 minutes, primarily through apoptosis. Overall, this work represents the first effort in using a modified seeded method for synthesizing biocompatible IL-based anisotropic AuNPs for photothermal therapy, offering a promising avenue for future cancer treatment research using ILs.
Collapse
Affiliation(s)
- Priyavrat Vashisth
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Cameron L. Smith
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Dhanush L. Amarasekara
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Gaya S. Dasanyake
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Gagandeep Singh
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Claylee M. Chism
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Christine M. Hamadani
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Tanveer Shaikh
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Noah Grovich
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Briana Gamboa
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Nicholas C. Fitzkee
- Department of Chemistry, Mississippi State University, Mississippi State, Mississippi 39762, United States
| | - Nathan I. Hammer
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| | - Eden E. L. Tanner
- Department of Chemistry & Biochemistry, The University of Mississippi, University, MS 38677, United States
| |
Collapse
|
19
|
Jin Y, Lee Y. Proteolysis Targeting Chimeras (PROTACs) in Breast Cancer Therapy. ChemMedChem 2024; 19:e202400267. [PMID: 39136599 PMCID: PMC11617661 DOI: 10.1002/cmdc.202400267] [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: 04/14/2024] [Revised: 07/28/2024] [Indexed: 10/16/2024]
Abstract
Breast cancer (BC) accounts for 30 % of cancer cases among women cancer patients globally, indicating the urgent need for the development of selective therapies targeting BCs. Recently, proteolysis-targeting chimera (PROTAC) has emerged as a promising strategy to target breast cancer. PROTAC is a chimeric molecule consisting of a target protein ligand, an E3 ligase ligand, and conjugating linkers, enabling it to facilitate the degradation of desired target proteins by recruiting E3 ligase in close proximity. Due to the catalytic behavior and direct degradation of BC-causing proteins, PROTAC could achieve high drug efficacy with low doses, drawing great attention for its potential as therapeutics. This review provides cases of the currently developed PROTACs targeting BCs depending on the type of BCs, limitations, and future perspectives of PROTAC in targeting BCs.
Collapse
Affiliation(s)
- Yerim Jin
- Department of ChemistryPusan National UniversityBusan46241Korea
| | - Yeongju Lee
- Department of ChemistryPusan National UniversityBusan46241Korea
| |
Collapse
|
20
|
Shetty SR, Debnath S, Majumdar K, Rajagopalan M, Ramaswamy A, Das A. Virtual screening, molecular dynamics simulations, and in vitro validation of EGFR inhibitors as breast cancer therapeutics. Bioorg Chem 2024; 153:107849. [PMID: 39368144 DOI: 10.1016/j.bioorg.2024.107849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
A high abundance of Epidermal Growth Factor Receptor (EGFR) in malignant cells makes them a prospective therapeutic target for basal breast tumors. Although EGFR inhibitors are in development as anticancer therapeutics, there exists limitations due to the dose-limiting cytotoxicity that limits their clinical utilization, thereby necessitating the advancement of effective inhibitors. In the present study, we have developed common pharmacophore hypotheses using 30 known EGFR inhibitors. The best pharmacophore hypothesis DHRRR_1 was utilized for virtual screening (VS) of the Phase database containing 4.3 × 106 fully prepared compounds. The top 1000 hits were further subjected to ADME filtration followed by structure-based VS and Molecular Dynamics (MD) simulation investigations. Based on pharmacophore hypothesis matching, XP glide score, interactions between ligands and active site residues, ADME properties, and MD simulations, the five best hits (SN-01 through SN-05) were preferred for in-vitro cytotoxicity studies. All the molecules except SN-02 exhibited cytotoxicity in Triple Negative Breast Cancer (TNBC) cells. These potential EGFR inhibitors effectively downregulated the EGF-induced proliferation, migration, in-vitro tumorigenic capability, and EGFR activation (pEGFR) in the TNBCs. Additionally, in combination with doxorubicin, the identified EGFR inhibitors significantly decreased the EGF-induced proliferation. SN-04, and SN-05 in the presence of a lower concentration of doxorubicin markedly increased the apoptotic markers expression in the TNBCs, an effect which was comparable to a higher concentration of doxorubicin treatment, alone. These observations suggest that both SN-04 and/or SN-05 can improve the efficacy of chemotherapeutic drug, doxorubicin at a lower concentration to avert the higher dose of chemotherapeutic-induced side effects during breast cancer treatment.
Collapse
Affiliation(s)
- Swathi R Shetty
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad 500007 TS, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sudhan Debnath
- Maharaja Bir Bikram College, Agartala, Tripura, India; Department of Chemistry, Netaji Subhash Mahavidyalaya, Udaipur, Tripura, India
| | | | - Muthukumaran Rajagopalan
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry 605014, INDIA
| | - Amutha Ramaswamy
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Puducherry 605014, INDIA
| | - Amitava Das
- Department of Applied Biology, Council of Scientific & Industrial Research-Indian Institute of Chemical Technology (CSIR-IICT), Uppal Road, Tarnaka, Hyderabad 500007 TS, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
21
|
Bao W, Ma X, Xue Y, Zou X, Guo Y. Pregnancy-associated triple-negative breast cancer: A case report and literature review. Medicine (Baltimore) 2024; 103:e40059. [PMID: 39465823 PMCID: PMC11479494 DOI: 10.1097/md.0000000000040059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/24/2024] [Indexed: 10/29/2024] Open
Abstract
RATIONALE The incidence of pregnancy-associated breast cancer (PABC) is relatively low, but it has been increasing in recent years. The onset of PABC causes serious harm to the fetus and the mother due to the unique physiological characteristics of pregnancy, which poses a particular challenge to clinicians. This article reports a case of pregnancy-associated triple-negative breast cancer and describes the patient characteristics and systematic treatment of this type of breast cancer. PATIENT CONCERNS A 33-year-old woman was admitted to hospital with a left breast mass that had appeared more than a year earlier. She was a second-time pregnant woman with a single live intrauterine fetus at 23 + 4 weeks of gestation. During the examination of the left breast, a 6 by 8 cm sized mass can be observed on the upper outer quadrant. DIAGNOSIS Pregnancy-associated triple-negative breast cancer. INTERVENTION The patient underwent a breast ultrasound which showed a left breast mass and the diagnosis was confirmed by a puncture biopsy of the left breast mass. The pregnancy was terminated after multidisciplinary discussion, taking into account the wishes of the patient and her family. After termination of the pregnancy, all treatments were given according to the standard triple-negative breast cancer (TNBC) treatment protocol. The patient was treated with neoadjuvant chemotherapy with epirubicin in combination with docetaxel (TE) in cycles of 21 days. After 3 cycles of TE, a modified radical mastectomy for left breast cancer was performed, and the appropriate radiotherapy and chemotherapy treatments were carried out in sequence. OUTCOMES After the surgery, the disease-free survival for the patient was 3 months until local metastases were diagnosed. Thus the radiotherapy and chemotherapy were carried out, and then the patient was in good general condition with no recurrence or metastases. LESSONS Clinicians need more research into the diagnosis, treatment and prognosis of PABC. Improving the rate of early diagnosis and using standardized and individualized comprehensive treatment plans will minimize fetal damage and improve survival and quality of life for patients.
Collapse
Affiliation(s)
- Weichao Bao
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Xiaolin Ma
- Department of Oncology, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, China
| | - Yuan Xue
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Xin Zou
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong Province, China
| | - Ying Guo
- Department of Endocrinology and Metabolic Diseases, Affiliated Hospital of Shandong Second Medical University, Weifang, Shandong Province, China
| |
Collapse
|
22
|
Yam C, Patel M, Hill HA, Sun R, Bassett RL, Kong E, Damodaran S, Koenig KB, Abouharb S, Saleem S, Bisen AK, Murthy RK, Ramirez DL, Rauch GM, Adrada BE, Candelaria RP, Wang X, Mittendorf EA, Thompson AM, White JB, Ravenberg EE, Clayborn AR, Ding QQ, Booser DJ, Oke O, Brewster AM, Hortobagyi GN, Ibrahim NK, Litton JK, Valero V, Arun BK, Tripathy D, Chang JT, Chen K, Korkut A, Moulder SL, Huo L, Lim B, Ueno NT. Targeting the Epidermal Growth Factor Receptor Pathway in Chemotherapy-Resistant Triple-Negative Breast Cancer: A Phase II Study. CANCER RESEARCH COMMUNICATIONS 2024; 4:2823-2834. [PMID: 39356138 PMCID: PMC11520071 DOI: 10.1158/2767-9764.crc-24-0255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/27/2024] [Accepted: 09/27/2024] [Indexed: 10/03/2024]
Abstract
PURPOSE Epidermal growth factor receptor (EGFR) pathway activation causes chemotherapy resistance, and inhibition of the EGFR pathway sensitizes triple-negative breast cancer (TNBC) cells to chemotherapy in preclinical models. Given the high prevalence of EGFR overexpression in TNBC, we conducted a single-arm phase II study of panitumumab (anti-EGFR monoclonal antibody), carboplatin, and paclitaxel as the second phase of neoadjuvant therapy (NAT) in patients with doxorubicin and cyclophosphamide (AC)-resistant TNBC (NCT02593175). PATIENTS AND METHODS Patients with early-stage, AC-resistant TNBC, defined as disease progression or ≤80% reduction in tumor volume after four cycles of AC, were eligible for this study and received panitumumab (2.5 mg/kg i.v., every week × 13), paclitaxel (80 mg/m2 i.v. every week × 12), and carboplatin (AUC = 4 i.v., every 3 weeks × 4) as the second phase of NAT. A two-stage Gehan-type design was used to detect an improvement in the pathological complete response (pCR)/residual cancer burden class I (RCB-I) rate from 5% to 20%. Whole-exome sequencing was performed on diagnostic tumor biospecimens, where available. RESULTS From November 3, 2016, through August 23, 2021, 43 patients with AC-resistant TNBC were enrolled. The combined pCR/RCB-I rate was 30.2%. The most common treatment-related adverse events were neutropenia (72%) and anemia (61%), with 7 (16%), 16 (37%), and 8 (19%) patients experiencing grade 4 neutropenia, grade 3 neutropenia, and grade 3 anemia, respectively. No new safety signals were observed. CONCLUSIONS This study met its primary endpoint (pCR/RCB-I = 30.2% vs. 5% in historical controls), suggesting that panitumumab should be evaluated as a component of NAT in patients with chemotherapy-resistant TNBC in a larger, randomized clinical trial. SIGNIFICANCE The epidermal growth factor receptor (EGFR) pathway has been implicated as a driver of chemotherapy resistance in triple-negative breast cancer (TNBC). Here, we evaluate the combination of panitumumab, carboplatin, and paclitaxel as the second phase of neoadjuvant therapy (NAT) in patients with AC-resistant TNBC. This study met its primary efficacy endpoint, and molecular alterations in EGFR pathway genes did not seem to influence response to the study regimen.
Collapse
Affiliation(s)
- Clinton Yam
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Miral Patel
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Holly A. Hill
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ryan Sun
- Division of Basic Sciences, Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roland L. Bassett
- Division of Basic Sciences, Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elisabeth Kong
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Senthil Damodaran
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kimberly B. Koenig
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sausan Abouharb
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sadia Saleem
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Ajit K. Bisen
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rashmi K. Murthy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - David L. Ramirez
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gaiane M. Rauch
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Beatriz E. Adrada
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Rosalind P. Candelaria
- Department of Breast Imaging, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaoping Wang
- Cancer Biology and Therapeutic Program, University of Hawai’i Cancer Center, Honolulu, Hawaii
| | - Elizabeth A. Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women’s Hospital, Boston, Massachusetts
- Breast Oncology Program, Dana-Farber/Brigham Cancer Center, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Alastair M. Thompson
- Division of Surgical Oncology, Section of Breast Surgery, Baylor College of Medicine, Houston, Texas
| | - Jason B. White
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elizabeth E. Ravenberg
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alyson R. Clayborn
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qing-Qing Ding
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel J. Booser
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Oluchi Oke
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Abenaa M. Brewster
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Gabriel N. Hortobagyi
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nuhad K. Ibrahim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jennifer K. Litton
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Vicente Valero
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Banu K. Arun
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Debu Tripathy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jeffrey T. Chang
- Department of Integrative Biology and Pharmacology, McGovern Medical School, Houston, Texas
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anil Korkut
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Stacy L. Moulder
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lei Huo
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Bora Lim
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Naoto T. Ueno
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
23
|
Yang M, Wang C, Ouyang L, Zhang H, Lin J. Establishment of prognostic model for invasive ductal carcinoma with distant metastasis within the triple-negative breast cancer: a SEER population-based study. Eur J Cancer Prev 2024:00008469-990000000-00176. [PMID: 39724567 DOI: 10.1097/cej.0000000000000925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Triple-negative breast cancer (TNBC) is a complex and diverse group of malignancies. Invasive ductal carcinoma (IDC) is the predominant pathological subtype and is closely linked to the ominous potential for distant metastasis, a pivotal factor that significantly influences patient outcomes. In light of these considerations, the present study was conceived with the objective of developing a nomogram model. This model was designed to predict the prognosis observed in IDC with distant metastasis in TNBC. This was a retrospective study based on the SEER database. Data of 9739 IDC-TNBC patients diagnosed from 2010 to 2020 were included in our study. Independent risk factors were screened by univariate and multivariate Cox regression analyses successively, which were used to develop a nomogram model predicting for prognosis. Cox multivariable analysis showed statistical significance in bone metastasis, liver metastasis, surgery, and chemotherapy. Incorporating statistically significant variables, as well as clinically significant age, lung metastasis, and brain metastasis into the construction of the prediction model, the C-indexes of the training group and validation group were 0.702 (0.663-0.741) and 0.667 (0.600-0.734), respectively, while the calibration curves were all close to the ideal 45° reference line, and decision curve analysis curves show excellent net benefit in the predictive model. The prognostic prediction model developed in this study demonstrated enhanced predictive accuracy, enabling a more precise evaluation of mortality risks associated with IDC with distant metastasis in TNBC.
Collapse
Affiliation(s)
- Minghao Yang
- General Surgery Department, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan
| | - Chunxi Wang
- General Surgery Department, Chinese PLA General Hospital, Beijing
| | - Lu Ouyang
- General Surgery Department, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Haowen Zhang
- General Surgery Department, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan
| | - Junlong Lin
- General Surgery Department, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan
| |
Collapse
|
24
|
Altman JE, Olex AL, Zboril EK, Walker CJ, Boyd DC, Myrick RK, Hairr NS, Koblinski JE, Puchalapalli M, Hu B, Dozmorov MG, Chen XS, Chen Y, Perou CM, Lehmann BD, Visvader JE, Harrell JC. Single-cell transcriptional atlas of human breast cancers and model systems. Clin Transl Med 2024; 14:e70044. [PMID: 39417215 PMCID: PMC11483560 DOI: 10.1002/ctm2.70044] [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: 06/04/2024] [Revised: 09/12/2024] [Accepted: 09/21/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Breast cancer's complex transcriptional landscape requires an improved understanding of cellular diversity to identify effective treatments. The study of genetic variations among breast cancer subtypes at single-cell resolution has potential to deepen our insights into cancer progression. METHODS In this study, we amalgamate single-cell RNA sequencing data from patient tumours and matched lymph metastasis, reduction mammoplasties, breast cancer patient-derived xenografts (PDXs), PDX-derived organoids (PDXOs), and cell lines resulting in a diverse dataset of 117 samples with 506 719 total cells. These samples encompass hormone receptor positive (HR+), human epidermal growth factor receptor 2 positive (HER2+), and triple-negative breast cancer (TNBC) subtypes, including isogenic model pairs. Herein, we delineated similarities and distinctions across models and patient samples and explore therapeutic drug efficacy based on subtype proportions. RESULTS PDX models more closely resemble patient samples in terms of tumour heterogeneity and cell cycle characteristics when compared with TNBC cell lines. Acquired drug resistance was associated with an increase in basal-like cell proportions within TNBC PDX tumours as defined with SCSubtype and TNBCtype cell typing predictors. All patient samples contained a mixture of subtypes; compared to primary tumours HR+ lymph node metastases had lower proportions of HER2-Enriched cells. PDXOs exhibited differences in metabolic-related transcripts compared to PDX tumours. Correlative analyses of cytotoxic drugs on PDX cells identified therapeutic efficacy was based on subtype proportion. CONCLUSIONS We present a substantial multimodel dataset, a dynamic approach to cell-wise sample annotation, and a comprehensive interrogation of models within systems of human breast cancer. This analysis and reference will facilitate informed decision-making in preclinical research and therapeutic development through its elucidation of model limitations, subtype-specific insights and novel targetable pathways. KEY POINTS Patient-derived xenografts models more closely resemble patient samples in tumour heterogeneity and cell cycle characteristics when compared with cell lines. 3D organoid models exhibit differences in metabolic profiles compared to their in vivo counterparts. A valuable multimodel reference dataset that can be useful in elucidating model differences and novel targetable pathways.
Collapse
Affiliation(s)
- Julia E. Altman
- Department of Human and Molecular GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Amy L. Olex
- C. Kenneth and Diane Wright Center for Clinical and Translational ResearchVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Emily K. Zboril
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of BiochemistryVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Carson J. Walker
- Department of Human and Molecular GeneticsVirginia Commonwealth UniversityRichmondVirginiaUSA
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - David C. Boyd
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Rachel K. Myrick
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Nicole S. Hairr
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Jennifer E. Koblinski
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Madhavi Puchalapalli
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Bin Hu
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Mikhail G. Dozmorov
- Department of BiostatisticsVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - X. Steven Chen
- Department of Public Health SciencesUniversity of Miami Miller School of MedicineMiamiFloridaUSA
- Sylvester Comprehensive Cancer CenterUniversity of Miami Miller School of MedicineMiamiFloridaUSA
| | - Yunshun Chen
- Walter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - Charles M. Perou
- Lineberger Comprehensive Cancer CenterUniversity of North CarolinaChapel HillNorth CarolinaUSA
| | - Brian D. Lehmann
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Jane E. Visvader
- Walter and Eliza Hall Institute of Medical ResearchMelbourneVictoriaAustralia
- Department of Medical BiologyUniversity of MelbourneParkvilleVictoriaAustralia
| | - J. Chuck Harrell
- Department of PathologyVirginia Commonwealth UniversityRichmondVirginiaUSA
- Massey Comprehensive Cancer CenterVirginia Commonwealth UniversityRichmondVirginiaUSA
- Center for Pharmaceutical EngineeringVirginia Commonwealth UniversityRichmondVirginiaUSA
| |
Collapse
|
25
|
Ge Y, Wei X, Liu JN, Sun PL, Gao H. New insights into acinic cell carcinoma of the breast: clinicopathology, origin of histology, molecular features, prognosis, and treatment. Front Oncol 2024; 14:1438179. [PMID: 39286022 PMCID: PMC11402605 DOI: 10.3389/fonc.2024.1438179] [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: 05/25/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Acinic cell carcinoma (AciCC) of the breast is a rare malignant epithelial neoplasm, with approximately 60 cases reported in the literature. It predominantly affects women and exhibits significant histological heterogeneity. The diagnosis of breast AciCC is primarily based on the presence of eosinophilic and/or basophilic granular cytoplasm and markers of serous acinar differentiation. Despite being considered a low-grade variant of conventional triple-negative breast cancer (TNBC), over 25% of patients with breast AciCC have adverse clinical outcomes. Additionally, in early research, microglandular adenosis (MGA) and atypical MGA were considered potential precursors for various breast cancers, including intraductal carcinoma, invasive ductal carcinoma, adenoid cystic carcinoma, metaplastic carcinoma, and AciCC. Similarly, some studies have proposed that breast AciCC should be considered a type of carcinoma developing in MGA with acinic cell differentiation rather than a distinct entity. Therefore, the pathogenesis of breast AciCC has not yet been clarified. Moreover, to the best of our knowledge, the literature has not summarized the latest prognosis and treatment of breast AciCC. In this review, we synthesized the current literature and the latest developments, aiming at exploring the clinicopathology, histological origin, molecular features, prognosis, and treatment of breast AciCC from a novel perspective.
Collapse
Affiliation(s)
- Yunjie Ge
- Department of Pathology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xianping Wei
- Department of Clinical Research, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jing-Nan Liu
- Department of Respiratory Medicine, The First Affiliated Hospital of Jilin University, Changchun, Jilin, China
| | - Ping-Li Sun
- Department of Pathology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Hongwen Gao
- Department of Pathology, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
26
|
Agrawal S, Vagha S. Exploring the Usefulness of Tumor Budding as a Histopathological Marker Compared to Tumor-Node-Metastasis (TNM) Staging in Breast Cancer. Cureus 2024; 16:e70315. [PMID: 39469410 PMCID: PMC11513207 DOI: 10.7759/cureus.70315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 09/27/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Tumor budding, defined as small clusters of tumor cells at the invasive front of carcinomas, has gained attention as a potential prognostic marker in various cancers. This study aimed to evaluate the utility of tumor budding as a histopathological marker in breast cancer and compare it to traditional prognostic markers such as histological grading, tumor-node-metastasis (TNM) staging, and molecular subtypes. METHODS A prospective, cross-sectional study was conducted over two years (June 2022 to May 2024) in the Department of Pathology at Jawaharlal Nehru Medical College, Wardha. Seventy-two female patients diagnosed with breast carcinoma who underwent modified radical mastectomy were included. Tumor budding was assessed through histopathological analysis and categorized as high or low. Statistical correlations were established between tumor budding and tumor size, histological grade, lymph node involvement, vascular invasion, and molecular subtypes (estrogen receptor, progesterone receptor, human epidermal growth factor receptor 2, and triple-negative breast cancer, TNBC). The chi-square test and multiple regression analyses were applied to assess significance. RESULTS High tumor budding was observed in 68.1% of patients and was significantly associated with larger tumor size (p = 0.040), higher histological grade (p = 0.009), lymph node metastasis (p = 0.002), and vascular invasion (p = 0.003). The postmenopausal age group (>55 years) demonstrated a higher prevalence of high budding (p = 0.010). No significant correlation was found between tumor budding and molecular subtypes (p = 0.39), although high budding was more frequent in TNBC cases. CONCLUSION Tumor budding is significantly associated with more aggressive tumor characteristics in breast cancer. Incorporating tumor budding into routine pathological assessments alongside TNM staging and histological grading may enhance the ability to identify high-risk patients and guide treatment strategies. Further large-scale, multicenter studies are warranted to confirm its prognostic value.
Collapse
Affiliation(s)
- Soumya Agrawal
- Department of Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sunita Vagha
- Department of Pathology, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
27
|
Pracharova J, Cyrikova T, Berecka M, Biersack B, Kasparkova J, Brabec V. Antimetastatic activity of (arene)ruthenium(II) complex of 4-aryl-4H-naphthopyran. Chem Biol Interact 2024; 400:111180. [PMID: 39089413 DOI: 10.1016/j.cbi.2024.111180] [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: 06/06/2024] [Revised: 07/21/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024]
Abstract
Metastatic cancer remains a formidable challenge in anticancer therapy. Despite efforts to develop effective antimetastasis drugs over the past half-century, currently approved treatments fall short of expectations. This report highlights the promising antiproliferative activity of a ruthenium-based therapeutic agent, namely dichlorido(p-cymene)[2-amino-4-(pyridin-3-yl)-4H-benzo[h]-chromene-3-carbonitrile]ruthenium(II) (complex 1) against metastatic cell lines. Complex 1 shows significant efficacy in metastatic LoVo and Du-145 cell lines at nanomolar concentrations, being markedly more active than clinically used anticancer cisplatin. Studies on the MDA-MB-231 cell line, which displays invasive characteristics, demonstrated that 1 significantly reduces cell invasion. This efficacy was confirmed by its impact on matrix metalloproteinase production in MDA-MB-231 cells. Given that cell migration drives cancer invasion and metastasis, complex 1's effect on MDA-MB-231 cell migration was evaluated via wound healing assay and vimentin network analysis. Results indicated a strong reduction in migration. A re-adhesion assay further demonstrated that 1 significantly lowers the re-adhesion ability of MDA-MB-231 cells compared to cisplatin. To better simulate the human body environment, a 3D spheroid invasion assay was used. This method showed that 1 effectively inhibits tumor spheroids from infiltrating the surrounding extracellular matrix. This study underscores the potential of (arene)ruthenium(II) complexes with naphthopyran ligands as potent antimetastatic agents for chemotherapy.
Collapse
Affiliation(s)
- Jitka Pracharova
- Department of Biophysics, Faculty of Science, Palacky University, CZ-77900, Olomouc, Czech Republic
| | - Tereza Cyrikova
- Department of Biophysics, Faculty of Science, Palacky University, CZ-77900, Olomouc, Czech Republic
| | - Michal Berecka
- Department of Biophysics, Faculty of Science, Palacky University, CZ-77900, Olomouc, Czech Republic
| | - Bernhard Biersack
- Organic Chemistry Laboratory, University Bayreuth, 95440, Bayreuth, Germany
| | - Jana Kasparkova
- Department of Biophysics, Faculty of Science, Palacky University, CZ-77900, Olomouc, Czech Republic; Czech Academy of Sciences, Institute of Biophysics, CZ-61200, Brno, Czech Republic
| | - Viktor Brabec
- Department of Biophysics, Faculty of Science, Palacky University, CZ-77900, Olomouc, Czech Republic; Czech Academy of Sciences, Institute of Biophysics, CZ-61200, Brno, Czech Republic.
| |
Collapse
|
28
|
Giffoni de Mello Morais Mata D, Rush MB, Smith-Uffen M, Younus J, Lohmann AE, Trudeau M, Morgan RL. The Omission of Anthracycline Chemotherapy in Women with Early HER2-Negative Breast Cancer-A Systematic Review and Meta-Analysis. Curr Oncol 2024; 31:4486-4506. [PMID: 39195318 PMCID: PMC11352883 DOI: 10.3390/curroncol31080335] [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: 07/02/2024] [Revised: 08/02/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Anthracycline-taxane is the standard chemotherapy strategy for treating high-risk early breast cancer despite the potentially life-threatening adverse events caused by anthracyclines. Commonly, the combination of docetaxel and cyclophosphamide (TC) is considered an alternative option. However, the efficacy of TC compared to anthracycline-taxane chemotherapy is unclear. This study compares disease-free survival (DFS), overall survival (OS) and cardiotoxicity between adjuvant TC and anthracycline-taxane for stages I-III, HER2-negative breast cancer. METHODS A systematic search on MEDLINE, Embase and Cochrane CENTRAL for randomized-controlled trials published until 11 March 2024, yielded 203 studies with 11,803 patients, and seven trials were included. RESULTS TC results in little to no difference in DFS (HR 1.09, 95% CI 0.98-1.20; moderate-certainty of evidence); OS (1.02, 95% CI 0.89-1.16; high-certainty of evidence); and cardiotoxicity (RR 0.54, 95% CI 0.16-1.76; high-certainty of evidence), compared to anthracycline-taxane. In the subgroup analysis, patients with ≥4 lymph nodes had improved DFS from anthracycline-taxane over TC. CONCLUSIONS Overall, there was no difference between TC and anthracycline-taxane in DFS, OS and cardiotoxicity. In women with ≥4 nodes, anthracycline-taxane was associated with a substantial reduction in relapse events, compared to TC. Our study supports the current standard of practice, which is to use anthracycline-taxane and TC chemotherapy as a reasonable option in select cases.
Collapse
Affiliation(s)
- Danilo Giffoni de Mello Morais Mata
- Division of Medical Oncology, Verspeeten Family Cancer Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada; (J.Y.); (A.E.L.)
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Mary-Beth Rush
- Department of Health Research Methods, Evidence and Impact (HEI), Faculty of Health Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada;
| | - Megan Smith-Uffen
- Department of Medicine, Faculty of Health Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada;
| | - Jawaid Younus
- Division of Medical Oncology, Verspeeten Family Cancer Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada; (J.Y.); (A.E.L.)
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Ana Elisa Lohmann
- Division of Medical Oncology, Verspeeten Family Cancer Centre, London Health Sciences Centre, London, ON N6A 5W9, Canada; (J.Y.); (A.E.L.)
- Department of Medicine, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Maureen Trudeau
- Division of Medical Oncology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Rebecca L. Morgan
- Department of Health Research Methods, Evidence and Impact (HEI), Faculty of Health Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada;
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
29
|
Wan X, Chen C, Zhan J, Ye S, Li R, Shen M. Dendritic polylysine co-delivery of paclitaxel and siAXL enhances the sensitivity of triple-negative breast cancer chemotherapy. Front Bioeng Biotechnol 2024; 12:1415191. [PMID: 39148942 PMCID: PMC11324506 DOI: 10.3389/fbioe.2024.1415191] [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: 04/10/2024] [Accepted: 07/12/2024] [Indexed: 08/17/2024] Open
Abstract
Background: Drug resistance is common in triple-negative breast cancer (TNBC) therapy. To identify a method to overcome chemotherapy resistance in TNBC cells, an siRNA targeting the AXL gene (siAXL), which can overcome drug resistance, was used in this study. A nanodelivery system was constructed to co-deliver siAXL and paclitaxel (PTX). Methods: A biodegradable and tumor microenvironment (TME)-sensitive mPEG-coated dendritic polylysine material (PDPLL) was synthesized. This material was used to construct single-molecule nanoparticles to co-deliver PTX and siAXL. The drug encapsulation and morphological properties of the nanoparticles (NPs) were characterized. The sensitivity of the NPs to the TME was evaluated in vitro with a dialysis method. The tumor-targeting effect of the PDPLL NPs was evaluated by fluorescence imaging and drug distribution evaluation in vivo. The ability to overcome drug resistance was evaluated using PTX-resistant 4T1 cells (4T1/PTX cells) in both in vitro and in vivo models. Results: PDPLL NPs had a particle size of 49.6 ± 5.9 nm and a zeta potential of 7.87 ± 0.68 mV. The PTX drug loading (DL)% was 2.59%. The siAXL DL was 2.5 mg PDPLL: 10 nmol siAXL. The release of PTX showed sustained release performance. The release of siAXL showed sensitivity for the TME. The NPs were stable in the plasma. The NPs promoted cell uptake by PTX-resistant 4T1 cells (4T1/PTX) and promoted tumor targeting and permeability in vivo. siAXL enhanced the toxicity and apoptosis efficiency of PTX in 4T1/PTX cells, as well as the cycle arrest efficiency caused by PTX. The NPs improved the above effects. In mouse 4T1/PTX orthotopic tumors, the NPs enhanced the sensitization of PTX to siAXL. Conclusion: The PDPLL NP co-delivery system possesses good encapsulating potential not only for PTX but also for siRNA. It can enhance the tumor-targeting effect and overcome the drug resistance of 4T1/PTX both in vitro and in vivo. This system is a potential delivery system for RNAs.
Collapse
Affiliation(s)
- Xiaofeng Wan
- National Health Commission (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Shanghai, China
| | - Chuanrong Chen
- Department of Oncology, Yijishan Hospital of Wannan Medical College, Wuhu, China
| | - Jianmin Zhan
- National Health Commission (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Shanghai, China
| | - Shuke Ye
- National Health Commission (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Shanghai, China
| | - Runsheng Li
- National Health Commission (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Shanghai, China
| | - Ming Shen
- National Health Commission (NHC) Key Lab of Reproduction Regulation (Shanghai Institute for Biomedical and Pharmaceutical Technologies), Shanghai, China
| |
Collapse
|
30
|
Shukla S, Karbhari A, Rastogi S, Agarwal U, Rai P, Mahajan A. Bench-to-bedside imaging in brain metastases: a road to precision oncology. Clin Radiol 2024; 79:485-500. [PMID: 38637186 DOI: 10.1016/j.crad.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/08/2024] [Accepted: 02/12/2024] [Indexed: 04/20/2024]
Abstract
Radiology has seen tremendous evolution in the last few decades. At the same time, oncology has made great strides in diagnosing and treating cancer. Distant metastases of neoplasms are being encountered more often in light of longer patient survival due to better therapeutic strategies and diagnostic methods. Brain metastasis (BM) is a dismal manifestation of systemic cancer. In the present scenario, magnetic resonance imaging (MRI), computed tomography (CT) and positron emission tomography (PET) are playing a big role in providing molecular information about cancer. Lately, molecular imaging has emerged as a stirring arena of dynamic imaging techniques that have enabled clinicians and scientists to noninvasively visualize and understand biological processes at the cellular and molecular levels. This knowledge has impacted etiopathogenesis, detection, personalized treatment, drug development, and our understanding of carcinogenesis. This article offers insight into the molecular biology underlying brain metastasis, its pathogenesis, imaging protocols, and algorithms. It also discusses disease-specific molecular imaging features, focusing on common tumors that spread to the brain, such as lung, breast, colorectal cancer, melanoma, and renal cell carcinoma. Additionally, it covers various targeted treatment options, criteria for assessing treatment response, and the role of artificial intelligence in diagnosing, managing, and predicting prognosis for patients with brain metastases.
Collapse
Affiliation(s)
- S Shukla
- Department of Radiodiagnosis and Imaging, Mahamana Pandit Madan Mohan Malaviya Cancer Centre and Homi Bhabha Cancer Hospital, Tata Memorial Hospital, Varanasi, 221 005, Maharashtra, India; Department of Radiodiagnosis and Imaging, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, 400 012, Maharashtra, India
| | - A Karbhari
- Department of Radiodiagnosis and Imaging, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, 400 012, Maharashtra, India
| | - S Rastogi
- Department of Radiodiagnosis and Imaging, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, 400 012, Maharashtra, India
| | - U Agarwal
- Department of Radiodiagnosis and Imaging, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, 400 012, Maharashtra, India
| | - P Rai
- Department of Radiodiagnosis and Imaging, Homi Bhabha National Institute, Tata Memorial Hospital, Mumbai, 400 012, Maharashtra, India
| | - A Mahajan
- Department of Imaging, The Clatterbridge Cancer Centre NHS Foundation Trust, L7 8YA Liverpool, UK; Faculty of Health and Life Sciences, University of Liverpool, L7 8TX, Liverpool, UK.
| |
Collapse
|
31
|
Duduyemi BM, Ayibor WG, Agyemang-Yeboah F. Tissue Microarray Immunohistochemical Staining for Androgen Receptor in Breast Cancer in a Ghanaian Cohort. Ann Afr Med 2024; 23:452-458. [PMID: 39034572 PMCID: PMC11364299 DOI: 10.4103/aam.aam_83_23] [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: 06/01/2023] [Revised: 10/20/2023] [Accepted: 11/27/2023] [Indexed: 07/23/2024] Open
Abstract
BACKGROUND Despite the advancement in therapy, breast cancer still remains the most common malignancy in women globally due in part to its heterogeneity. Triple-negative breast cancer (TNBC) represents up to 20% of all breast cancer variants, an aggressive disease with poorer outcomes compared to other breast cancer subtypes. No targeted therapies are currently approved for TNBC, and newer treatment approaches are seriously needed. Androgen receptor (AR), another hormonal receptor, is often expressed in breast cancer, and its role depends on the relative levels of circulating estrogens and androgens. This study aimed to assess the expression of AR in breast cancer in a tertiary hospital in Ghana. METHODOLOGY Immunohistochemical staining for AR was performed on tissue microarray (TMA) blocks, of which estrogen receptor, progesterone receptor, and Her-2/neu had already been done. 197 cases were suitable for the study. Results from the immunostaining were analyzed using the SPSS version 23 for descriptive statistics and correlations (χ2 and Pearson tests). RESULTS 197 TMA cases were used. TNBCs constitute 61.9% of the cancers. The majority of these tumors were grade III, ductal carcinoma NST. The mean age was 49.86 ± 14.09, and the modal age group was 40-49 years. Our cases showed 23% AR expression in triple-negative cancers. The study also established that AR is more frequently expressed in low-grade tumors compared to high-grade ones. CONCLUSION There is an appreciable level of AR expression in our cases; however, most are quadruple negative. However, AR is more frequently expressed in low-grade tumors than high-grade ones.
Collapse
Affiliation(s)
- Babatunde M. Duduyemi
- Department of Pathology, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Department of Pathology, University of Sierra Leone Teaching Hospitals Complex, Freetown, Sierra Leone
| | - William G. Ayibor
- Department of Pathology, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Francis Agyemang-Yeboah
- Department of Molecular Medicine, School of Medicine and Dentistry, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
32
|
Marey MA, Abozahra R, El-Nikhely NA, Kamal MF, Abdelhamid SM, El-Kholy MA. Transforming microbial pigment into therapeutic revelation: extraction and characterization of pyocyanin from Pseudomonas aeruginosa and its therapeutic potential as an antibacterial and anticancer agent. Microb Cell Fact 2024; 23:174. [PMID: 38867319 PMCID: PMC11170807 DOI: 10.1186/s12934-024-02438-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/23/2024] [Indexed: 06/14/2024] Open
Abstract
BACKGROUND The objectives of the current study were to extract pyocyanin from Pseudomonas aeruginosa clinical isolates, characterize its chemical nature, and assess its biological activity against different bacteria and cancer cells. Due to its diverse bioactive properties, pyocyanin, being one of the virulence factors of P. aeruginosa, holds a promising, safe, and available therapeutic potential. METHODS 30 clinical P. aeruginosa isolates were collected from different sources of infections and identified by routine methods, the VITEK 2 compact system, and 16 S rRNA. The phenazine-modifying genes (phzM, phzS) were identified using polymerase chain reaction (PCR). Pyocyanin chemical characterization included UV-Vis spectrophotometry, Fourier Transform Infra-Red spectroscopy (FTIR), Gas Chromatography-Mass Spectrometry (GC-MS), and Liquid Chromatography-Mass Spectrometry (LC-MS). The biological activity of pyocyanin was explored by determining the MIC values against different clinical bacterial strains and assessing its anticancer activity against A549, MDA-MB-231, and Caco-2 cancer cell lines using cytotoxicity, wound healing and colony forming assays. RESULTS All identified isolates harboured at least one of the phzM or phzS genes. The co-presence of both genes was demonstrated in 13 isolates. The UV-VIS absorbance peaks were maxima at 215, 265, 385, and 520 nm. FTIR could identify the characteristic pyocyanin functional groups, whereas both GC-MS and LC-MS elucidated the chemical formula C11H18N2O2, with a molecular weight 210. The quadri-technical analytical approaches confirmed the chemical nature of the extracted pyocyanin. The extract showed broad-spectrum antibacterial activity, with the greatest activity against Bacillus, Staphylococcus, and Streptococcus species (MICs 31.25-125 µg/mL), followed by E. coli isolates (MICs 250-1000 µg/mL). Regarding the anticancer activity, the pyocyanin extract showed IC50 values against A549, MDA-MB-231, and Caco-2 cancer cell lines of 130, 105, and 187.9 µg/mL, respectively. Furthermore, pyocyanin has markedly suppressed colony formation and migratory abilities in these cells. CONCLUSIONS The extracted pyocyanin has demonstrated to be a potentially effective candidate against various bacterial infections and cancers. Hence, the current findings could contribute to producing this natural compound easily through an affordable method. Nonetheless, future studies are required to investigate pyocyanin's effects in vivo and analyse the results of combining it with other traditional antibiotics or anticancer drugs.
Collapse
Affiliation(s)
- Moustafa A Marey
- Department of Microbiology and Biotechnology, Division of Clinical and Biological Sciences, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), Abu Kir Campus, P.O. Box 1029, Alexandria, Egypt
| | - Rania Abozahra
- Microbiology and Immunology Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Nefertiti A El-Nikhely
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Miranda F Kamal
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Damanhour University, Beheira, Egypt
| | - Sarah M Abdelhamid
- Microbiology and Immunology Department, Faculty of Pharmacy, Damanhour University, Damanhour, Egypt
| | - Mohammed A El-Kholy
- Department of Microbiology and Biotechnology, Division of Clinical and Biological Sciences, College of Pharmacy, Arab Academy for Science, Technology and Maritime Transport (AASTMT), Abu Kir Campus, P.O. Box 1029, Alexandria, Egypt.
| |
Collapse
|
33
|
McAndrew H, Rigler J, Yeguvapalli S, Chitrala KN. Analysis of gene expression profiles to elucidate racial differences in African American and White patients with Triple-negative breast cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596478. [PMID: 38853921 PMCID: PMC11160710 DOI: 10.1101/2024.05.29.596478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Triple-negative breast cancer (TNBC) is the second most diagnosed subtype of breast cancer. It is known to be the most aggressive one that lacks known targetable receptors. One of the concerns in TNBC is the disparities in its prevalence and tumor pathogenesis among women with non-Hispanic African American backgrounds. Despite extensive research, the genetic underpinnings that lead to these disparities remain elusive. The current study aims to provide initiative for further clinical research in the development of targeted therapy for TNBC. Gene expression profiles from African American (AA) and European American (EA) patients with TNBC were collected from Gene Expression Omnibus and performed differential gene expression (DEG)analysis. Candidate genes for a significant correlation between expression and survival rates for breast invasive carcinoma were analyzed using UALCAN. The DAVID annotation tool, Enrichr web server, KEGG database, and Gene Ontology (GO) database were used for functional enrichment analysis of target genes. The Network Analyst server was used to identify ligands with strong affinities, SeamDock server for molecular docking between the biomarkers/associated ligands and examined protein-protein interactions (PPI) from the STRING server. Data from public breast cancer cohorts was utilized to identify expression patterns associated with poor survival outcomes of AA patients with TNBC. Our results showed three genes of interest ( CCT3 , LSM2 , and MRPS16 ) and potential ligands for molecular docking. Molecular docking was performed for the ICG001 ligand to CCT3 (binding affinities of -9.3 kcal/mol and -8.9 kcal/mol) and other interacting proteins ( CDC20 and PPP2CA ) with high degrees of connectivity. The results determined molecular docking of ICG001 to the CDC20 protein resulted in the highest binding affinity. Our results demonstrated that CCT3 and its interacting partners could serve as potential biomarkers due to their association with the survival outcome of AA patients with TNBC and ICG 001 could be the therapeutic lead for these biomarkers.
Collapse
|
34
|
Mishra A, Mishra SK, Sharanappa V, Krishnani N, Kumari N, Agarwal G. Incidence and Prognostic Significance of Androgen Receptors (AR) in Indian Triple-Negative Breast Cancer (TNBC). Indian J Surg Oncol 2024; 15:250-257. [PMID: 38741650 PMCID: PMC11088609 DOI: 10.1007/s13193-024-01877-2] [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: 02/13/2023] [Accepted: 01/07/2024] [Indexed: 05/16/2024] Open
Abstract
Molecular sub-characterization of triple-negative breast cancer (TNBC) has great therapeutic and possibly prognostic implications. The primary aim of this study was to investigate the incidence of luminal androgen receptor (LAR) subtype of TNBC and secondary aims were sub-categorization and clinico-pathologic correlation of LAR breast cancers. Retrospective study (January 2008 and 31st of December 2018) consisting of 157 TNBC patients. Androgen receptor (AR) expression was measured by immunohistochemical analysis. One percent cutoff was set as a positive expression. Sub-categorization was done on the basis of EGFR (> 15% of tumor cells) and Ki-67 expression (low- < 11%, intermediate- 11-20%, and high- > 21%). AR expression was correlated with various clinico-pathologic features and outcomes of the patients. The incidence of AR expression in TNBC was 24.8%. Considering different thresholds of > 5%, > 10%, and > 20% immunostaining, the incidence of AR positivity was 18.4, 15.2, and 11.5% respectively. The incidence of Ki-67 (p = 0.89) and EGFR (p = 0.643) expression did not differ significantly in AR-positive and -negative TNBC. Based on EGFR expression 19, 67 and 14% patients were categorized as low, intermediate, and high risk respectively. Low-risk (p ≤ 0.001) and low-grade (p = 0.014) tumors were more likely to have > 10% AR expression. Clinico-pathological profile, response to neoadjuvant chemotherapy, disease-free survival (p = 0.458), and overall survival (p = 0.806) did not significantly differ between AR expressing and negative TNBC. On multivariate analysis, only tumor staging was a significant predictor of survival (p = 0.012) and AR expression of > 10% revealed a trend towards improved survival (p = 0.07). When considering only AR-positive TNBC, AR expression of > 10% (p = 0.038), distant metastases (p = 0.003), and EGFR status (p = 0.024) were significantly associated with survival. AR expression does not seem to very strongly correlate with prognosis in TNBC and further studies could focus more on its predictive role in deciding anti-androgen therapy.
Collapse
Affiliation(s)
- Anjali Mishra
- Department of Endocrine Surgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Shravan Kumar Mishra
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Vikram Sharanappa
- Department of Endocrine Surgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Narendra Krishnani
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Niraj Kumari
- Department of Pathology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| | - Gaurav Agarwal
- Department of Endocrine Surgery, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Raebareli Road, Lucknow, 226014 India
| |
Collapse
|
35
|
Folz J, Jo J, Gonzalez ME, Eido A, Zhai T, Caruso R, Kleer CG, Wang X, Kopelman R. Photoacoustic lifetime oxygen imaging of radiotherapy-induced tumor reoxygenation In Vivo. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2024; 21:100241. [PMID: 39005728 PMCID: PMC11243757 DOI: 10.1016/j.jpap.2024.100241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
Purpose Early detection and diagnosis of cancer is critical for achieving positive therapeutic outcomes. Biomarkers that can provide clinicians with clues to the outcome of a given therapeutic course are highly desired. Oxygen is a small molecule that is nearly universally present in biological tissues and plays a critical role in the effectiveness of radiotherapies by reacting with DNA radicals and subsequently impairing cellular repair of double strand breaks.Techniques for measuring oxygen in biological tissues often use blood oxygen saturation to approximate the oxygen partial pressure in surrounding tissues despite the complex, nonlinear, and dynamic relationship between these two separate oxygen populations. Methods and materials We combined a directly oxygen-sensitive, tumor-targeted, chemical contrast nanoelement with the photoacoustic lifetime-based (PALT) oxygen imaging technique to obtain image maps of oxygen in breast cancer tumors in vivo. The oxygen levels of patient-derived xenografts in a mouse model were characterized before and after a course of radiotherapy. Results We show that, independent of tumor size, radiotherapy induced an increase in the overall oxygenation levels of the tumor. Further, this increase in the oxygenation of the tumor significantly correlated with a positive response to radiotherapy, as demonstrated by a reduction in tumor volume over the twenty-day monitoring period following therapy and histological staining. Conclusion Our PALT imaging presented here is simple, fast, and non-invasive. Facilized by the PALT approach, imaging of tumor reoxygenation may be utilized as a simple, early indicator for evaluating cancer response to radiotherapy. Further characterization of the reoxygenation degree, temporal onset, and possible theragnostic implications are warranted.
Collapse
Affiliation(s)
- Jeff Folz
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
| | - Janggun Jo
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Maria E. Gonzalez
- Department of Pathology, University of Michigan Health System, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Ahmad Eido
- Department of Pathology, University of Michigan Health System, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Tianqu Zhai
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Roberta Caruso
- Department of Pathology, University of Michigan Health System, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Celina G. Kleer
- Department of Pathology, University of Michigan Health System, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Xueding Wang
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA
| | - Raoul Kopelman
- Department of Chemistry, University of Michigan, Ann Arbor, MI 48109, USA
- Rogel Cancer Center, University of Michigan Health System, Ann Arbor, MI 48109, USA
| |
Collapse
|
36
|
Qiu Y, Chen A, Yu R, Llevenes P, Seen M, Ko NY, Monti S, Denis GV. Insulin Resistance Increases TNBC Aggressiveness and Brain Metastasis via Adipocyte-derived Exosomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.01.592097. [PMID: 38746141 PMCID: PMC11092600 DOI: 10.1101/2024.05.01.592097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Patients with triple negative breast cancer (TNBC) and comorbid Type 2 Diabetes (T2D), characterized by insulin resistance of adipose tissue, have higher risk of metastasis and shorter survival. Adipocytes are the main non-malignant cells of the breast tumor microenvironment (TME). However, adipocyte metabolism is usually ignored in oncology and mechanisms that couple T2D to TNBC outcomes are poorly understood. Here we hypothesized that exosomes, small vesicles secreted by TME breast adipocytes, drive epithelial-to-mesenchymal transition (EMT) and metastasis in TNBC via miRNAs. Exosomes were purified from conditioned media of 3T3-L1 mature adipocytes, either insulin-sensitive (IS) or insulin-resistant (IR). Murine 4T1 cells, a TNBC model, were treated with exosomes in vitro (72h). EMT, proliferation and angiogenesis were elevated in IR vs. control and IS. Brain metastases showed more mesenchymal morphology and EMT enrichment in the IR group. MiR-145a-3p is highly differentially expressed between IS and IR, and potentially regulates metastasis. Significance IR adipocyte exosomes modify TME, increase EMT and promote metastasis to distant organs, likely through miRNA pathways. We suggest metabolic diseases such as T2D reshape the TME, promoting metastasis and decreasing survival. Therefore, TNBC patients with T2D should be closely monitored for metastasis, with metabolic medications considered.
Collapse
|
37
|
Sanli AN, Tekcan Sanli DE, Aydogan F, Altundag MK. Should the Breast Cancer Staging System be Revised? Am Surg 2024; 90:1066-1073. [PMID: 38128067 DOI: 10.1177/00031348231223074] [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: 12/23/2023]
Abstract
OBJECTIVE The purpose of this study was to determine whether breast cancer patients at stage T2N0 with tumor size ≥4 cm and <4 cm. METHOD Patients with T2N0 stage breast cancer diagnosed between 2010 and 2019 were analyzed in 2 groups as <4 cm (T2a) and ≥4 cm (T2b) in the study using the SEER 17 Research Plus database. The patients' clinicopathological characteristics and oncological outcomes were included. Group comparisons of prognostic factors, overall survival (OS), and cancer-specific survival (CSS) were made. RESULTS In this study, which involved 70971 patients, the T2a group had higher 5-year OS rate (87.2 ± .2 vs 80.8 ± .5%) and 5-year CSS rate (93.7 ± .1% vs 89.4 ± .4%) than the T2b group (P < .001). Univariate analysis revealed that the overall risk of death was 1.5 times higher in T2b than T2a (HR: 1.533 [95% CI: 1.450-1.622], P < .001), whereas multivariate analysis demonstrated the risk was 1.4 times higher (HR: 1.384 [95% CI: 1.307-1.466], P < .001). The risk of cancer-specific death was 1.7 times higher in univariate analysis (HR: 1.691 [95% CI: 1.561-1.832], P < .001) and 1.4 times higher in multivariate analysis (HR: 1.420 [95% CI: 1.309-1.541], P < .001). CONCLUSION Overall survival and BCSS rates in stage T2b breast cancer patients are significantly lower than in T2a patients. Tumor size ≥4 cm in breast cancer is a negative predictor of prognosis.
Collapse
Affiliation(s)
- Ahmet Necati Sanli
- Department of General Surgery, Abdulkadir Yuksel State Hospital, Gaziantep, Turkey
| | | | - Fatih Aydogan
- Department of General Surgery, Kirklareli University, Faculty of Medicine, Kirklareli, Turkey
- Breast Health Center, Memorial Bahcelievler Hospital, Istanbul, Turkey
| | | |
Collapse
|
38
|
Kim U, Debnath R, Maiz JE, Rico J, Sinha S, Blanco MA, Chakrabarti R. ΔNp63 regulates MDSC survival and metabolism in triple-negative breast cancer. iScience 2024; 27:109366. [PMID: 38510127 PMCID: PMC10951988 DOI: 10.1016/j.isci.2024.109366] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 12/20/2023] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Triple-negative breast cancer (TNBC) contributes greatly to mortality of breast cancer, demanding new targetable options. We have shown that TNBC patients have high ΔNp63 expression in tumors. However, the function of ΔNp63 in established TNBC is yet to be explored. In current studies, targeting ΔNp63 with inducible CRISPR knockout and Histone deacetylase inhibitor Quisinostat showed that ΔNp63 is important for tumor progression and metastasis in established tumors by promoting myeloid-derived suppressor cell (MDSC) survival through tumor necrosis factor alpha. Decreasing ΔNp63 levels are associated with decreased CD4+ and FOXP3+ T-cells but increased CD8+ T-cells. RNA sequencing analysis indicates that loss of ΔNp63 alters multiple MDSC properties such as lipid metabolism, chemotaxis, migration, and neutrophil degranulation besides survival. We further demonstrated that targeting ΔNp63 sensitizes chemotherapy. Overall, we showed that ΔNp63 reprograms the MDSC-mediated immunosuppressive functions in TNBC, highlighting the benefit of targeting ΔNp63 in chemotherapy-resistant TNBC.
Collapse
Affiliation(s)
- Ukjin Kim
- Department of Surgery, Sylvester Comprehensive Cancer, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Rahul Debnath
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Javier E. Maiz
- Department of Surgery, Sylvester Comprehensive Cancer, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Joshua Rico
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Satrajit Sinha
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Mario Andrés Blanco
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rumela Chakrabarti
- Department of Surgery, Sylvester Comprehensive Cancer, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
39
|
Long L, Fei X, Chen L, Yao L, Lei X. Potential therapeutic targets of the JAK2/STAT3 signaling pathway in triple-negative breast cancer. Front Oncol 2024; 14:1381251. [PMID: 38699644 PMCID: PMC11063389 DOI: 10.3389/fonc.2024.1381251] [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: 02/03/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
Triple-negative breast cancer (TNBC) poses a significant clinical challenge due to its propensity for metastasis and poor prognosis. TNBC evades the body's immune system recognition and attack through various mechanisms, including the Janus Kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) signaling pathway. This pathway, characterized by heightened activity in numerous solid tumors, exhibits pronounced activation in specific TNBC subtypes. Consequently, targeting the JAK2/STAT3 signaling pathway emerges as a promising and precise therapeutic strategy for TNBC. The signal transduction cascade of the JAK2/STAT3 pathway predominantly involves receptor tyrosine kinases, the tyrosine kinase JAK2, and the transcription factor STAT3. Ongoing preclinical studies and clinical research are actively investigating this pathway as a potential therapeutic target for TNBC treatment. This article comprehensively reviews preclinical and clinical investigations into TNBC treatment by targeting the JAK2/STAT3 signaling pathway using small molecule compounds. The review explores the role of the JAK2/STAT3 pathway in TNBC therapeutics, evaluating the benefits and limitations of active inhibitors and proteolysis-targeting chimeras in TNBC treatment. The aim is to facilitate the development of novel small-molecule compounds that target TNBC effectively. Ultimately, this work seeks to contribute to enhancing therapeutic efficacy for patients with TNBC.
Collapse
Affiliation(s)
- Lin Long
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| | - Xiangyu Fei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Liucui Chen
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
| | - Liang Yao
- Department of Pharmacy, Central Hospital of Hengyang, Hengyang, China
| | - Xiaoyong Lei
- School of Pharmaceutical Science, Hengyang Medical School, University of South China, Hengyang, China
- The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, China
| |
Collapse
|
40
|
Prutianu I, Giuşcă SE, Gafton B, Chifu MB, Terinte C, Antonescu A, Popovici L, Căruntu ID. Triple-negative breast cancer: from classical clinicopathological features to androgen receptor profile. ROMANIAN JOURNAL OF MORPHOLOGY AND EMBRYOLOGY = REVUE ROUMAINE DE MORPHOLOGIE ET EMBRYOLOGIE 2024; 65:209-216. [PMID: 39020535 PMCID: PMC11384857 DOI: 10.47162/rjme.65.2.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/19/2024]
Abstract
Triple-negative breast cancer (BC) represents an extensively analyzed entity to establish the overall framework of clinicopathological characteristics, with an impact on defining prognostic and predictive factors. The relationship between triple-negative BC and androgen receptor (AR) is far from being clarified. We aimed to evaluate the classical clinicopathological spectrum that characterized a triple-negative BC, focusing on AR expression. The study group comprised 124 cases of triple-negative BC. The main clinicopathological parameters were extracted from medical records. The immunohistochemical (IHC) exam was run using the following antibodies: anti-estrogen receptor (ER), anti-progesterone receptor (PR), anti-human epidermal growth factor receptor (HER2∕neu), anti-Ki67 and anti-AR. AR immunoexpression was assessed as absent (completely negative) or present (unrelated to percentages and intensity). Data were statistically analyzed. AR expression was positive in 78 (63%) cases and negative in 46 (37%) cases. Among the study group, 28 cases exhibited an AR percentage ranging from 1% to 10%, 15 cases showed a percentage between 11% and 50%, while 12 cases had AR values between 51% and 75% and 23 cases fell within the AR range of 76% to 100%. No significant differences between AR immunoexpression (negative versus positive), clinicopathological characteristics and survival parameters were found. Statistically significant differences were registered between histological type, tumor stage, distant metastasis, tumor-infiltrating lymphocytes (TILs), treatment and residual cancer burden (RCB), and survival parameters. Thus, our results sustain that AR does not affect the biological behavior of triple-negative BC.
Collapse
Affiliation(s)
- Iulian Prutianu
- Department of Morpho-Functional Sciences I, Grigore T. Popa University of Medicine and Pharmacy, Iaşi, Romania; ;
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Kim AI, Oh JH, Cho JY. QSOX2 Upregulated in triple-negative breast cancer exacerbates patient prognosis by stabilizing integrin β1. Heliyon 2024; 10:e27148. [PMID: 38500982 PMCID: PMC10945127 DOI: 10.1016/j.heliyon.2024.e27148] [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: 10/31/2023] [Revised: 02/07/2024] [Accepted: 02/25/2024] [Indexed: 03/20/2024] Open
Abstract
Breast cancer (BC) remains a significant global health threat, with triple-negative breast cancer (TNBC) standing out as a particularly aggressive subtype lacking targeted therapies. Addressing this gap, we propose Quiescin Q6 sulfhydryl oxidase 2 (QSOX2) as a potential therapeutic target, a disulfide bond-forming enzyme implicated in cancer progression. Using publicly available datasets, we conducted a comprehensive analysis of QSOX2 expression in BC tumor and non-tumor tissues, assessing its specificity across different molecular subtypes. We further explored correlations between QSOX2 expression and patient outcomes, utilizing datasets like TCGA and METABRIC. In addition, we performed in vitro experiments to evaluate QSOX2 expression in BC cell lines and investigate the effects of QSOX2 knockdown on various TNBC cellular processes, including cell proliferation, apoptosis resistance, migration, and the epithelial-to-mesenchymal transition (EMT). Our results reveal significantly elevated QSOX2 expression in BC tumor tissues, particularly in TNBC, and establish an association between high QSOX2 expression and increased patient mortality, cancer progression, and recurrence across various BC subtypes. Notably, QSOX2 knockdown in TNBC cell lines reduces cell proliferation, enhances apoptosis, and suppresses migration, potentially mediated through its influence on the EMT process. Furthermore, we identify a significant link between QSOX2 and integrin β1 (ITGB1), suggesting that QSOX2 enhances ITGB1 stability, subsequently exacerbating the malignancy of TNBC. In conclusion, elevated QSOX2 expression emerges as a key factor associated with adverse patient outcomes in BC, particularly in TNBC, contributing to disease progression through various mechanisms, including the modulation of ITGB1 stability. Our findings underscore the potential of targeting QSOX2 as a therapeutic strategy for improving patient prognoses not only in TNBC but also in other BC subtypes.
Collapse
Affiliation(s)
- A-In Kim
- Department of Biochemistry, Brain Korea 21 Project and Research Institute for Veterinary Science, Seoul National University College of Veterinary Medicine, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ji Hoon Oh
- Department of Biochemistry, Brain Korea 21 Project and Research Institute for Veterinary Science, Seoul National University College of Veterinary Medicine, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Je-Yoel Cho
- Department of Biochemistry, Brain Korea 21 Project and Research Institute for Veterinary Science, Seoul National University College of Veterinary Medicine, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
42
|
Esmat E, Haidary AM, Saadaat R, Rizvi SN, Aleena S, Haidari M, Hofiani SMS, Hussaini N, Hakimi A, Khairy A, Abdul-Ghafar J. Association of hormone receptors and human epidermal growth factor receptor-2/neu expressions with clinicopathologic factors of breast carcinoma: a cross-sectional study in a tertiary care hospital, Kabul, Afghanistan. BMC Cancer 2024; 24:388. [PMID: 38539179 PMCID: PMC10967195 DOI: 10.1186/s12885-024-12129-5] [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: 05/02/2023] [Accepted: 03/15/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is one of the major causes of death worldwide. It is the most common cause of death before the age of 70 years. The incidence and mortality of BC are rapidly increasing, posing great challenges to the health system and economy of every nation. METHODOLOGY A cross-sectional analytical study was conducted at the Department of Pathology and Clinical Laboratory of the French Medical Institute for Mothers and Children (FMIC) to demonstrate the association of human epidermal growth factor receptor 2 (Her2/Neu) and estrogen receptor (ER)/ progesterone receptor (PR) with clinical as well as pathological parameters among women with BC. A consecutive nonprobability sampling method was used for this study over a span of one and a half years. RESULTS One hundred twenty participants diagnosed with breast cancer were included in the study. The mean age at diagnosis was 44.58 ± 11.16 years. Out of the total patients, 68 (56.7%) were above 40 years old, 108 (90%) were married, 94 (78.3%) were multiparous, and 88 (73.3%) had a history of breastfeeding. 33.3% of cases were within the age range of menopause (40-50 years). The positive expression rates of ER, PR, and Her2/neu were found to be 48.8%, 44.6%, and 44.6%, respectively, and Her2/neu overexpression was found to be higher among ER/PR-negative cases. CONCLUSION In our study, we demonstrated that among Afghan women, grade II invasive ductal carcinoma, not otherwise specified, was the most common type of BC and frequently affected women above the age of 40. We also revealed that the percentage of negative ER (50.4%), negative PR (54.4%), and concordant ER/PR-negative cases were high compared to other possibilities. Additionally, the study revealed that expression of Her2/neu was in contrast with the expression of ER and PR receptors. The findings of our study still support the importance of performing immunohistochemical stains for hormonal receptor classification in terms of better clinical outcomes and prognosis.
Collapse
Affiliation(s)
- Esmatullah Esmat
- Department of Pathology and Clinical Laboratory, French Medical Institute for Mother and Children (FMIC), Kabul, Afghanistan
| | - Ahmed Maseh Haidary
- Department of Pathology and Clinical Laboratory, French Medical Institute for Mother and Children (FMIC), Kabul, Afghanistan
| | - Ramin Saadaat
- Department of Pathology and Clinical Laboratory, French Medical Institute for Mother and Children (FMIC), Kabul, Afghanistan
| | - Syeda Naghma Rizvi
- Aga Khan University School of Nursing and Midwifery (AKU-SoNaM), Karachi, Pakistan
| | - Syeda Aleena
- Aga Khan University School of Nursing and Midwifery (AKU-SoNaM), Karachi, Pakistan
| | - Mujtaba Haidari
- Department of Pathology and Clinical Laboratory, French Medical Institute for Mother and Children (FMIC), Kabul, Afghanistan
| | - Sayed Murtaza Sadat Hofiani
- Department of Academic and Research, Postgraduate Medical Education (PGME), French Medical Institute for Mothers and Children (FMIC), Kabul, Afghanistan
| | - Nasrin Hussaini
- Department of Pathology and Clinical Laboratory, French Medical Institute for Mother and Children (FMIC), Kabul, Afghanistan
| | - Ahmadullah Hakimi
- Department of Pathology and Clinical Laboratory, French Medical Institute for Mother and Children (FMIC), Kabul, Afghanistan
| | - Abdullatif Khairy
- Department of Pathology and Clinical Laboratory, French Medical Institute for Mother and Children (FMIC), Kabul, Afghanistan
| | - Jamshid Abdul-Ghafar
- Department of Pathology and Clinical Laboratory, French Medical Institute for Mother and Children (FMIC), Kabul, Afghanistan.
| |
Collapse
|
43
|
Hoshi N, Uemura T, Tachibana K, Abe S, Murakami-Nishimagi Y, Okano M, Noda M, Saito K, Kono K, Ohtake T, Waguri S. Endosomal protein expression of γ1-adaptin is associated with tumor growth activity and relapse-free survival in breast cancer. Breast Cancer 2024; 31:305-316. [PMID: 38265632 PMCID: PMC10902087 DOI: 10.1007/s12282-023-01539-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 12/20/2023] [Indexed: 01/25/2024]
Abstract
BACKGROUND γ1-Adaptin is a subunit of adaptor protein complex-1 (AP-1), which regulates intracellular transport between the trans-Golgi network (TGN) and endosomes. Since expression levels of AP-1 subunits have been reported to be associated with cell proliferation and cancer malignancy, we investigated the relationships between the immunohistochemical expression of γ1-adaptin and both clinicopathological factors and relapse-free survival (RFS) in breast cancer tissue. MATERIALS AND METHODS SK-BR-3 cell line depleted of γ1-adaptin was used for cell proliferation, migration, and invasion assay. Intracellular localization of γ1-adaptin was examined with immunohistochemistry (IHC) using an antibody against γ1-adaptin, and with double immunohistofluorescence (IHF) microscopy using markers for the TGN and endosome. γ1-Adaptin intensities in IHC samples from 199 primary breast cancer patients were quantified and assessed in relation to clinicopathological factors and RFS. RESULTS Cell growth, migration, and invasion of SK-BR-3 cells were significantly suppressed by the depletion of γ1-adaptin. Although the staining patterns in the cancer tissues varied among cases by IHC, double IHF demonstrated that γ1-adaptin was mainly localized in EEA1-positive endosomes, but not in the TGN. γ1-Adaptin intensity was significantly higher in the tumor regions than in non-tumor regions. It was also higher in patients with Ki-67 (high), ER (-), PgR (-), and HER2 (+). Among subtypes of breast cancer, γ1-adaptin intensity was higher in HER2 than in luminal A or luminal B. The results of the survival analysis indicated that high γ1-adaptin intensity was significantly associated with worse RFS, and this association was also observed in group with ER (+), PgR (+), HER2 (-), Ki-67 (high), or luminal B. In addition, the Cox proportional hazards model showed that high γ1-adaptin intensity was an independent prognostic factor. CONCLUSION These results suggest that the endosomal expression of γ1-adaptin is positively correlated with breast cancer malignancy and could be a novel prognostic marker.
Collapse
Affiliation(s)
- Nobuhiro Hoshi
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Takefumi Uemura
- Department of Anatomy and Histology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kazunoshin Tachibana
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Sadahiko Abe
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Yuko Murakami-Nishimagi
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Maiko Okano
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Masaru Noda
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Katsuharu Saito
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Koji Kono
- Department of Gastrointestinal Tract Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tohru Ohtake
- Department of Breast Surgery, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Satoshi Waguri
- Department of Anatomy and Histology, Fukushima Medical University School of Medicine, Fukushima, Japan.
| |
Collapse
|
44
|
Eldehna WM, Al-Ansary GH, Al-Warhi T, Jaballah MY, Elaasser M, Rashed M. Identification of novel ureido benzothiophenes as dual VEGFR-2/EGFR anticancer agents. Bioorg Chem 2024; 143:107037. [PMID: 38134521 DOI: 10.1016/j.bioorg.2023.107037] [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: 10/20/2023] [Revised: 12/05/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023]
Abstract
Presently, dual-targeting by a single small molecule stands out as a fruitful cancer-fighting strategy. Joining the global effort to fight cancer, a leading cause of death worldwide, we report in this study a novel set for benzothiophene-based aryl urea derivatives as potential anti-proliferative candidates endowed with dual VEGFR-2/EGFR inhibitory activities. The prepared ureido benzothiophenes 6a-r have been evaluated for their anticancer action on a panel of tumor cell lines, namely PanC-1, MCF-7, and HepG2 cells. Most newly synthesized benzo[b]thiophene ureas disclosed effective cytotoxic activities against the examined cancer cell lines. In particular, compound 6q, with an appended 4-trifluoromethoxy group on the terminal phenyl ring, exhibited the most significant cytotoxic activity in MCF-7 with IC50 3.86 ± 0.72 ug/mL; IC50 of 3.65 ± 0.18 ug/ml in PanC-1 cell line and an IC50 of 4.78 ± 0.06 ug/ml in HepG2. After that, derivatives that exhibited the most potent cytotoxic activities (6g, 6j, 6q, and 6r) were further evaluated as VEGFR-2 and EGFR inhibitors. Fortunately, they displayed low nanomolar IC50 values against both enzymes, where compound 6q emerged to possess superior inhibitory effects towards both EGFR and VEGFR-2 with IC50 46.6 nM and 11.3 nM simultaneously compared to the reference medications Erlotinib and Sorafenib, respectively. The docked structure of 6q within the catalytic region of VEGFR-2 and EGFR kinases was acquired and studied so that we could investigate potential binding mechanisms for the target ureido benzothiophenes. Hence, the benzothiophene-based aryl urea scaffold has great potential for advancing the development of highly effective dual inhibitors targeting both EGFR and VEGFR-2, which can serve as effective candidates for anticancer therapy.
Collapse
Affiliation(s)
- Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt.
| | - Ghada H Al-Ansary
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo 11566, Egypt
| | - Tarfah Al-Warhi
- Department of Chemistry, College of Science, Princess Nourah Bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Maiy Y Jaballah
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Ain Shams University, Abassia, Cairo 11566, Egypt
| | - Mahmoud Elaasser
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo 11651, Egypt
| | - Mahmoud Rashed
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| |
Collapse
|
45
|
Pankaj D, Kumar N, Singh A, Kumar M, Imam ZS, Bhushan V, Jha PK. Clinical Profile of Triple-Negative Breast Cancer: A Hospital-Based Study. Cureus 2024; 16:e53373. [PMID: 38435145 PMCID: PMC10907967 DOI: 10.7759/cureus.53373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2024] [Indexed: 03/05/2024] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) is a new concept and an important area of investigation. In Western country's literature, different studies reported on TNBC and all indicated the poor prognostic aspect of this molecular subtype over other types of breast cancer. However, there is a scarcity of comprehensive data from India. Hence, the present study was carried out to look at the epidemiological and clinical characteristics of TNBC in the Indian population. Methods The present study was performed between January 2020 and June 2021 at a tertiary care hospital in Eastern India. A total of 150 patients with TNBC were enrolled in the study. The epidemiological and clinical features of enrolled patients were collected and reviewed. Results The median age of patients at TNBC presentation was 45.53 years (24 to 74 years). The median tumor size was reported to be 5.32 cm. Of 150 patients, 94(62.67%) showed enlarged lymph nodes and 56 (37.33%) patients had no lymph node enlargement. In the present study, 85 (56.67%) patients were in the pre/perimenopausal stage at presentation, whereas 65 (43.33%) patients were in the postmenopausal stage. Upon evaluating the spread of TNBC, it was observed that a maximum of patients 60 (40%) were at the T4 stage and 56 (37.33%) at the N0 condition. The clinical staging of TNBC reported a maximum of 74 (49.33%) patients at the IIA, and IIB stages followed by 53 (35.33%) patients at the IIIA, IIIB, and IIIC stages and a minimum of 11 (7.33%) patients at stage IV. Only five (3.33%) patients were reported with a family history of breast cancer. Of all patients, 126 (84%) had detected early breast cancer thereby applicable for surgery at the time of presentation, whereas 71 (47.33%) patients were eligible for radiation therapy and 138 (92%) patients received chemotherapy. A total of 112 (74.67%) patients were found alive after 24 months of follow-up, 22 (4.67%) patients were observed with remission, and 11 (7.33%) patients died due to TNBC progression. During the course of follow-up, five (3.33%) patients were lost in the study. Conclusion TNBC is an aggressive malignancy that has a high risk of systemic relapses in the first two years after diagnosis. For more mature evidence on TNBC, longer follow-up of patients is necessary.
Collapse
Affiliation(s)
- Deepak Pankaj
- General Surgery, Indira Gandhi Institute of Medical Sciences, Patna, IND
| | - Nitesh Kumar
- General Surgery, Indira Gandhi Institute of Medical Sciences, Patna, IND
| | - Anju Singh
- Pathology, Indira Gandhi Institute of Medical Sciences, Patna, IND
| | - Manish Kumar
- Surgical Oncology, Indira Gandhi Institute of Medical Sciences, Patna, IND
| | - Zeenat S Imam
- Pathology, Indira Gandhi Institute of Medical Sciences, Patna, IND
| | - Vibhuti Bhushan
- General Surgery, Indira Gandhi Institute of Medical Sciences, Patna, IND
| | - Pawan K Jha
- General Surgery, Indira Gandhi Institute of Medical Sciences, Patna, IND
| |
Collapse
|
46
|
Limsakul P, Choochuen P, Jungrungrueang T, Charupanit K. Prognostic Markers in Tyrosine Kinases Specific to Basal-like 2 Subtype of Triple-Negative Breast Cancer. Int J Mol Sci 2024; 25:1405. [PMID: 38338684 PMCID: PMC10855431 DOI: 10.3390/ijms25031405] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/19/2024] [Accepted: 01/21/2024] [Indexed: 02/12/2024] Open
Abstract
Triple-negative breast cancer (TNBC), a heterogeneous and therapeutically challenging subtype, comprises over 50% of patients categorized into basal-like 1 (BL1) and basal-like 2 (BL2) intrinsic molecular subtypes. Despite their shared basal-like classification, BL2 is associated with a poor response to neoadjuvant chemotherapy and reduced relapse-free survival compared to BL1. Here, the study focused on identifying subtype-specific markers for BL2 through transcriptomic analysis of TNBC patients using RNA-seq and clinical integration. Six receptor tyrosine kinase (TK) genes, including EGFR, EPHA4, EPHB2, PDGFRA, PDGFRB, and ROR1, were identified as potential differentiators for BL2. Correlations between TK mRNA expression and TNBC prognosis, particularly EGFR, PDGFRA, and PDGFRB, revealed potential synergistic interactions in pathways related to cell survival and proliferation. Our findings also suggest promising dual markers for predicting disease prognosis. Furthermore, RT-qPCR validation demonstrated that identified BL2-specific TKs were expressed at a higher level in BL2 than in BL1 cell lines, providing insights into unique characteristics. This study advances the understanding of TNBC heterogeneity within the basal-like subtypes, which could lead to novel clinical treatment approaches and the development of targeted therapies.
Collapse
Affiliation(s)
- Praopim Limsakul
- Division of Physical Science, Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand;
- Center of Excellence for Trace Analysis and Biosensor (TAB-CoE), Faculty of Science, Prince of Songkla University, Songkhla 90110, Thailand
| | - Pongsakorn Choochuen
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (T.J.)
| | - Thawirasm Jungrungrueang
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (T.J.)
| | - Krit Charupanit
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla 90110, Thailand; (P.C.); (T.J.)
| |
Collapse
|
47
|
Tong Y, Zhou T, Wang X, Deng S, Qin L. Upregulation of CENPM promotes breast carcinogenesis by altering immune infiltration. BMC Cancer 2024; 24:54. [PMID: 38200449 PMCID: PMC10777552 DOI: 10.1186/s12885-023-11808-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: 10/26/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND The involvement of centromere protein M (CENPM) in various types of cancer has been established, however, its impact on breast cancer and immune infiltration remains unknown. METHODS We examined the expression of CENPM in different cancer types by utilizing the Cancer Genome Atlas (TCGA) and Genotype Tissue Expression Pan-Cancer (GEO) databases. Using data from the TCGA, we examined the correlation between the expression of CENPM, the prognosis, and the clinicopathological features of individuals diagnosed with breast cancer. We conducted an enrichment analysis of CENPM using the clusterProfiler R software tool, utilizing data obtained from breast cancer patients and specimens at our institution. In addition to examining the correlation between CENPM expression and genes associated with immune checkpoints, the TIDE algorithm was employed to explore the potential of CENPM as a biomarker for immunotherapy in breast cancer. The impact of CENPM on the growth of breast cancer cells was evaluated through the utilization of the CCK8 test and the colony formation assay. The effect of CENPM on the migration of breast cancer cells was assessed using scratch and transwell assays. RESULTS Research findings indicate that elevated levels of CENPM are linked to patient outcomes in breast cancer and various clinicopathological features. Furthermore, elevated levels of CENPM expression correlated with decreased levels of CD8 + T cells and mast cells, increased levels of Tregs and Th2, and reduced levels of CD8 + T cells. Additionally, the coexpression of CENPM with the majority of genes related to immune checkpoints indicates its potential to forecast the effectiveness of treatment in breast cancer. Suppression of CENPM hampers the growth and movement of breast tumor cells. CONCLUSIONS In summary, our study findings indicate that CENPM may serve as a cancer-causing gene in breast cancer and also as a biomarker for predicting the efficacy of immunotherapy. The oncogene CENPM is associated with breast cancer and is involved in cell proliferation and immune infiltration.
Collapse
Affiliation(s)
- Yanchu Tong
- Jingzhou Central Hospital, No. 60 Jingzhong Road, Jingzhou District, Jingzhou City, 434020, Hubei Province, China
| | - Tongzhou Zhou
- The HongKong Polytechnic University, 11 Yuk Choi Road, Hung Hom, Kowloon, 999077, HKSAR, China
| | - Xiaokun Wang
- Jingzhou Central Hospital, No. 60 Jingzhong Road, Jingzhou District, Jingzhou City, 434020, Hubei Province, China
| | - Shun Deng
- Jingzhou Central Hospital, No. 60 Jingzhong Road, Jingzhou District, Jingzhou City, 434020, Hubei Province, China
| | - Lu Qin
- Jingzhou Central Hospital, No. 60 Jingzhong Road, Jingzhou District, Jingzhou City, 434020, Hubei Province, China.
| |
Collapse
|
48
|
Mulkearns-Hubert EE, Rhoades EE, Ben-Salem S, Bharti R, Hajdari N, Johnson S, Myers A, Smith IN, Bandyopadhyay S, Eng C, Downs E, Lathia JD, Reizes O. Targeting NANOG and FAK via Cx26-derived Cell-penetrating Peptides in Triple-negative Breast Cancer. Mol Cancer Ther 2024; 23:56-67. [PMID: 37703580 PMCID: PMC10840808 DOI: 10.1158/1535-7163.mct-21-0783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 02/28/2023] [Accepted: 09/11/2023] [Indexed: 09/15/2023]
Abstract
Triple-negative breast cancer (TNBC) represents the most lethal and treatment-resistant breast cancer subtype with limited treatment options. We previously identified a protein complex unique to TNBC composed of the gap junction protein connexin 26 (Cx26), the pluripotency transcription factor NANOG, and focal adhesion kinase (FAK). We sought to determine whether a peptide mimetic of the interaction region of Cx26 attenuated tumor growth in preclinical models. We designed peptides based on Cx26 juxtamembrane domains and performed binding experiments with NANOG and FAK using surface plasmon resonance. Binding studies revealed that the Cx26 C-terminal tail and intracellular loop bound to NANOG and FAK with submicromolar-to-micromolar affinity and that a 5-amino acid sequence in the C-terminal tail of Cx26 (RYCSG) was sufficient for binding. Peptides with high affinity were engineered with a cell-penetrating antennapedia sequence and assessed in functional assays including cell proliferation, tumorsphere formation, and in vivo tumor growth, and downstream signaling changes were measured. The cell-penetrating Cx26 peptide (aCx26-pep) disrupted self-renewal while reducing nuclear FAK and NANOG and inhibiting NANOG target gene expression in TNBC cells but not luminal mammary epithelial cells. In vivo, aCx26-pep reduced tumor growth and proliferation and induced cell death. Here, we provide proof-of-concept that a Cx26 peptide-based strategy inhibits growth and alters NANOG activity specifically in TNBC, indicating the therapeutic potential of this targeting approach.
Collapse
Affiliation(s)
- Erin E. Mulkearns-Hubert
- Department of Cardiovascular and Metabolic Sciences, Cancer Impact Area, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | - Emily Esakov Rhoades
- Department of Cardiovascular and Metabolic Sciences, Cancer Impact Area, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | - Salma Ben-Salem
- Department of Cardiovascular and Metabolic Sciences, Cancer Impact Area, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | - Rashmi Bharti
- Department of Cardiovascular and Metabolic Sciences, Cancer Impact Area, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | - Nicole Hajdari
- Department of Cardiovascular and Metabolic Sciences, Cancer Impact Area, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | - Sadie Johnson
- Department of Cardiovascular and Metabolic Sciences, Cancer Impact Area, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | - Alex Myers
- Department of Cardiovascular and Metabolic Sciences, Cancer Impact Area, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | - Iris Nira Smith
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Smarajit Bandyopadhyay
- Molecular Biotechnology Core, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Erinn Downs
- Department of Pathology, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
| | - Justin D. Lathia
- Department of Cardiovascular and Metabolic Sciences, Cancer Impact Area, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Avenue, Cleveland, Ohio 44195
- Case Comprehensive Cancer Center, 10900 Euclid Ave. Cleveland, OH 44106
| | - Ofer Reizes
- Department of Cardiovascular and Metabolic Sciences, Cancer Impact Area, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, Ohio 44195
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, 9500 Euclid Avenue, Cleveland, Ohio 44195
- Case Comprehensive Cancer Center, 10900 Euclid Ave. Cleveland, OH 44106
| |
Collapse
|
49
|
Guo Z, Han S. Targeting cancer stem cell plasticity in triple-negative breast cancer. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:1165-1181. [PMID: 38213533 PMCID: PMC10776602 DOI: 10.37349/etat.2023.00190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 10/15/2023] [Indexed: 01/13/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive breast cancer subtype with limited treatment options. Cancer stem cells (CSCs) are thought to play a crucial role in TNBC progression and resistance to therapy. CSCs are a small subpopulation of cells within tumors that possess self-renewal and differentiation capabilities and are responsible for tumor initiation, maintenance, and metastasis. CSCs exhibit plasticity, allowing them to switch between states and adapt to changing microenvironments. Targeting CSC plasticity has emerged as a promising strategy for TNBC treatment. This review summarizes recent advances in understanding the molecular mechanisms underlying CSC plasticity in TNBC and discusses potential therapeutic approaches targeting CSC plasticity.
Collapse
Affiliation(s)
- Zhengwang Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Shuyan Han
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Integration of Chinese and Western Medicine, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
50
|
Mai N, Abuhadra N, Jhaveri K. Molecularly Targeted Therapies for Triple Negative Breast Cancer: History, Advances, and Future Directions. Clin Breast Cancer 2023; 23:784-799. [PMID: 37336650 DOI: 10.1016/j.clbc.2023.05.012] [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: 04/10/2023] [Revised: 05/16/2023] [Accepted: 05/21/2023] [Indexed: 06/21/2023]
Abstract
Triple negative breast cancer (TNBC) remains the subtype with poorest prognosis. Despite the subtype's heterogeneity, there is still a paucity in effective targeted therapeutics that offer both good efficacy and tolerability, and chemotherapy remains the backbone of modern TNBC therapy. In the past few years, immunotherapy as well as novel therapeutic modalities like antibody-drug conjugates (ADCs) have shown clinical benefit and have been FDA approved in various clinical stages of unselected TNBC. However, there has not been similar advancement in molecularly targeted therapies, especially when compared to advancements seen in hormone receptor (HR)-positive or HER2-positive breast cancer. PARP inhibitors have been approved for BRCA-mutated TNBC, but responses are short-lived, and resistance remains a barrier for current treatment. PI3K pathway inhibitors approved in HR+ breast cancer has not worked for TNBC and continue to have significant dose-limiting adverse effects. EGFR inhibition has been thoroughly explored in TNBC, but all trials so far have shown minimal efficacy. Nevertheless, despite these setbacks, current research in targeted therapy for TNBC holds great promise in overcoming the barriers of the past and developing novel therapeutic approaches for the future. In this review, we describe molecular targets both identified and validated in the treatment of TNBC, discuss the historical efforts towards development of targeted agents and current areas of improvement, and address promising advances that have the potential to improve outcomes in this heterogenous and aggressive breast cancer subtype. Immunotherapy, ADCs, and AR targeting will be discussed in separate reviews of this edition.
Collapse
Affiliation(s)
- Nicholas Mai
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Nour Abuhadra
- Memorial Sloan Kettering Cancer Center, New York, NY
| | - Komal Jhaveri
- Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|