1
|
Motta L, Puglisi M, Pavone G, Motta G, Martorana F, Bambaci M, Aricò D, Vigneri P. Unveiling Prostate-Specific Membrane Antigen's Potential in Breast Cancer Management. Cancers (Basel) 2025; 17:456. [PMID: 39941824 PMCID: PMC11816243 DOI: 10.3390/cancers17030456] [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: 12/17/2024] [Revised: 01/23/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND In recent years, the role of prostate-specific membrane antigen (PSMA) in the imaging and treatment of prostate cancer (PCa) has been extensively investigated. However, despite its name, PSMA is not exclusively specific to PCa. It has been found to be expressed in the neo-vasculature of various solid tumors, including breast cancer (BC), in which it is associated with tumor angiogenesis. METHODS This review aims to assess the potential of PSMA-based radiopharmaceuticals for BC diagnosis and treatment. It explores the current landscape by analyzing preclinical and clinical studies, as well as ongoing clinical trials, to provide insights into the PSMA-targeted approaches in BC management. RESULTS Early studies suggest PSMA-based imaging could improve BC lesion detection, especially in TNBC. The available data remains too preliminary to conclusively assess whether PSMA-based imaging or therapy will offer a significant advantage in BC. However, some preclinical findings suggest that this approach may hold promise as a novel strategy for managing this widespread malignancy. CONCLUSIONS PSMA-based strategies show potential for BC diagnosis and treatment, but further research is needed. Ongoing and future clinical trials are expected to provide deeper insights into the potential utility of this approach.
Collapse
Affiliation(s)
- Lucia Motta
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (L.M.); (G.M.); (F.M.); (P.V.)
- Medical Oncology Unit, Humanitas Istituto Clinico Catanese, Misterbianco, 95045 Catania, Italy
| | - Marialuisa Puglisi
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98124 Messina, Italy;
| | - Giuliana Pavone
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (L.M.); (G.M.); (F.M.); (P.V.)
- Medical Oncology Unit, Humanitas Istituto Clinico Catanese, Misterbianco, 95045 Catania, Italy
| | - Gianmarco Motta
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (L.M.); (G.M.); (F.M.); (P.V.)
- Medical Oncology Unit, Humanitas Istituto Clinico Catanese, Misterbianco, 95045 Catania, Italy
| | - Federica Martorana
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (L.M.); (G.M.); (F.M.); (P.V.)
| | - Michelangelo Bambaci
- Nuclear Medicine Unit, Humanitas Istituto Clinico Catanese, Misterbianco, 95045 Catania, Italy; (M.B.); (D.A.)
| | - Demetrio Aricò
- Nuclear Medicine Unit, Humanitas Istituto Clinico Catanese, Misterbianco, 95045 Catania, Italy; (M.B.); (D.A.)
| | - Paolo Vigneri
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy; (L.M.); (G.M.); (F.M.); (P.V.)
- Medical Oncology Unit, Humanitas Istituto Clinico Catanese, Misterbianco, 95045 Catania, Italy
| |
Collapse
|
2
|
Elavarasu SM, Vasudevan K, Sasikumar K, Doss C GP. The role of ABI2 in modulating nuclear proteins: Therapeutic implications for NUP54 and NUP153 in TNBC. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2025; 143:97-115. [PMID: 39843146 DOI: 10.1016/bs.apcsb.2024.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype of breast cancer that lacks hormone receptors, which makes it more likely to metastasize and have a poor prognosis. Despite some effectiveness of chemotherapy, TNBC remains challenging to manage, with high relapse and mortality rates. Recent findings have highlighted the role of the ubiquitin-protease system in TNBC, with ABI2 identified as a significant regulator. Reduced ABI2 expression is associated with aggressive disease and poor outcomes, whereas ABI2 overexpression (OE-ABI2) inhibits TNBC cell proliferation by modulating the PI3K/Akt signaling pathway. Although ABI2 is not a nuclear protein, it influences critical nuclear functions such as DNA repair and gene expression. Nuclear proteins, particularly those in the nuclear pore complex and nuclear matrix, are essential for cellular functions and have been linked to various diseases, including cancer. This study used RNA sequencing (RNA-seq) to examine the gene expression in MDA-MB-231 cell line and ABI2-overexpressing cells. Differentially expressed genes were annotated, and a protein-protein interaction network was constructed. Network and enrichment analysis identified the nucleoporins NUP54 and NUP153 as potential novel targets for TNBC. This study emphasizes the impact of ABI2 on nuclear proteins and suggests that targeting NUP54 and NUP153 could offer new therapeutic options for TNBC.
Collapse
Affiliation(s)
- Santhosh Mudipalli Elavarasu
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - Karthick Vasudevan
- Manipal Academy of Higher Education (MAHE), Manipal, India; Institute of Bioinformatics, International Technology Park, Bangalore, India
| | - K Sasikumar
- Department of Sensor and Biomedical Technology, School of Electronics Engineering, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India
| | - George Priya Doss C
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu, India.
| |
Collapse
|
3
|
Wang Y, Wang X, Sun H, Zhang Z, Gu J. LncRNA MCM3AP-AS1 promotes chemoresistance in triple-negative breast cancer through the miR-524-5p/RBM39 axis. Mol Cell Biochem 2025; 480:371-384. [PMID: 38472681 DOI: 10.1007/s11010-023-04908-8] [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: 06/08/2023] [Accepted: 11/25/2023] [Indexed: 03/14/2024]
Abstract
Triple-negative breast cancer (TNBC) is the most lethal subtype of BC, with unfavorable treatment outcomes. Evidence suggests the engagement of lncRNA MCM3AP-AS1 in BC development. This study investigated the action of MCM3AP-AS1 in chemoresistance of TNBC cells. Drug-resistant TNBC cell lines SUM159PTR and MDA-MB-231R were constructed by exposure to increasing concentrations of doxorubicin/docetaxel (DOX/DXL). MCM3AP-AS1 and miR-524-5p expression levels were determined by RT-qPCR. RNA binding motif 39 (RBM39) level was measured using Western blot. Cell viability and apoptosis were assessed by CCK-8 assay and flow cytometry. The targeted binding of miR-524-5p with MCM3AP-AS1 or RBM39 was predicted by ECORI database and validated by dual-luciferase assays. The gain-and-loss of function assays were conducted in cells to investigate the interactions among MCM3AP-AS1, miR-524-5p, and RBM39. TNBC xenograft mouse models were established through subcutaneous injection of MCM3AP-AS1-silencing MDA-MB-231R cells and intraperitoneally administrated with DOX/DXL to verify the role of MCM3AP-AS1 in vivo. MCM3AP-AS1 was upregulated in drug-resistant TNBC cells, and MCM3AP-AS1 silencing could sensitize drug-resistant TNBC cells to chemotherapeutic drugs by promoting apoptosis. MCM3AP-AS1 targeted miR-524-5p. After DOX/DXL treatment, miR-524-5p inhibition partially reversed the effect of MCM3AP-AS1 silencing on inhibiting chemoresistance and promoting apoptosis of drug-resistant TNBC cells. miR-524-5p targeted RBM39. Silencing MCM3AP-AS1 promoted apoptosis via the miR-524-5p/RBM39 axis, thereby enhancing chemosensitivity of drug-resistant TNBC cells. MCM3AP-AS1 knockdown upregulated miR-524-5p, downregulated RBM39, and restrained tumor development in vivo. MCM3AP-AS1 silencing potentiates apoptosis of drug-resistant TNBC cells by upregulating miR-524-5p and downregulating RBM39, thereby suppressing chemoresistance in TNBC.
Collapse
Affiliation(s)
- Yueping Wang
- Department of Medical Laboratory Science, Anhui No. 2 Provincial People's Hospital, 1868 #Dangshan Road, North 2nd Ring, Hefei, 230041, Anhui, China
- Department of Molecular and Cellular Biology, University of Connecticut, Storrs, CT, 06269, USA
| | - Xuedong Wang
- Department of Medical Laboratory Science, Anhui No. 2 Provincial People's Hospital, 1868 #Dangshan Road, North 2nd Ring, Hefei, 230041, Anhui, China.
| | - Haiyi Sun
- School of Clinical Medicine, Anhui Medical University, Hefei, 230032, Anhui, China
| | - Ziyun Zhang
- Department of Medical Laboratory Science, Anhui No. 2 Provincial People's Hospital, 1868 #Dangshan Road, North 2nd Ring, Hefei, 230041, Anhui, China
| | - Juan Gu
- Department of Medical Laboratory Science, Anhui No. 2 Provincial People's Hospital, 1868 #Dangshan Road, North 2nd Ring, Hefei, 230041, Anhui, China
| |
Collapse
|
4
|
Zhang X, Dai S, Li L, Wang P, Dong M. UL16‑binding protein 1 is a significant prognostic and diagnostic marker for breast cancer. Oncol Lett 2025; 29:15. [PMID: 39492940 PMCID: PMC11526324 DOI: 10.3892/ol.2024.14761] [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: 05/17/2024] [Accepted: 09/05/2024] [Indexed: 11/05/2024] Open
Abstract
The aim of the present study was to investigate the association between UL16 binding protein 1 (ULBP1) and the prognosis of patients with and immune cell infiltration in breast cancer (BRCA). The mRNA data of BRCA and immune-related genes were extracted from The Cancer Genome Atlas and were analyzed using bioinformatics tools. Subsequently, the results obtained by bioinformatics were validated through the collection of clinical patient data at the Zibo Central hospital (Zibo, China). The difference in the expression of the ULBP1 gene between BRCA tissues and normal precancerous tissues was analyzed, followed by validation using immunohistochemistry. By combining clinical data from patients with BRCA, the prognostic and diagnostic significance of the ULBP1 gene in patients with BRCA was analyzed. Gene enrichment analysis was conducted to gain insight into the molecular mechanisms underlying the regulatory role of ULBP1 in BRCA by analyzing its related functions and signaling pathways. Furthermore, single sample gene set enrichment analysis (ssGSEA) and Spearman's correlation analysis were performed to explore the correlation between ULBP1 as a target gene related with tumor immune cell infiltration. The data revealed that ULBP1 is a target gene associated with immunity and the prognosis of patients with BRCA. Patients with BRCA with a high expression of ULBP1 had a poorer prognosis. ULBP1 expression correlated with progesterone receptor expression, estrogen receptor expression and histological type in patients with BRCA; thus, it may serve as an independent predictor for the overall survival rate of patients. Functional enrichment analysis revealed a significant co-expression between ULBP1 and ULBP2, ULBP3, retinoic acid early transcript 1K, as well as a significant enrichment of pathways associated with carcinogenesis and immune suppression. ssGSEA and Spearman's correlation analysis demonstrated significant correlations between ULBP1 expression and tumor immune cells, as well as immune checkpoints. Overall, the present study demonstrated that ULBP1 was associated with BRCA immunity and might serve as a prognostic and diagnostic biomarker for patients with BRCA. In addition, it might also be a potential target for the immunotherapy of BRCA.
Collapse
Affiliation(s)
- Xiaowei Zhang
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao, Shandong 266023, P.R. China
- Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Shuhong Dai
- Department of Cardiology, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Liang Li
- Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Pengyun Wang
- Department of Orthopedics, Zibo Central Hospital, Zibo, Shandong 255036, P.R. China
| | - Mingxin Dong
- Department of Medicinal Chemistry, School of Pharmacy, Qingdao University, Qingdao, Shandong 266023, P.R. China
| |
Collapse
|
5
|
Shaikh M, Doshi G. Unraveling non-coding RNAs in breast cancer: mechanistic insights and therapeutic potential. Med Oncol 2024; 42:37. [PMID: 39730979 DOI: 10.1007/s12032-024-02589-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 12/16/2024] [Indexed: 12/29/2024]
Abstract
Breast cancer remains a leading global health challenge requiring innovative, therapeutic strategies to improve patient outcomes. This review explores the pivotal roles of non-coding RNAs (ncRNAs), including long non-coding RNA, micro RNA, and circular RNA, in breast cancer biology. We highlight how these molecules regulate critical signaling pathways, influence tumor microenvironments, and contribute to treatment resistance. Our findings underscore the potential of ncRNAs as biomarkers for early diagnosis and as treatment targets for personalized treatment strategies. To pave the way for innovative cancer management approaches, we investigate the complex interactions of ncRNAs and their impact on tumor progression. This comprehensive review enhances our understanding of breast cancer biology while emphasizing the translational significance of ncRNA research in developing effective treatment strategies. Additional research and clinical studies are required to confirm the diagnostic and medicinal value of ncRNAs in breast cancer. Investigating the complex networks of ncRNA interactions and their links to other biological pathways can lead to the discovery of new treatment targets. Furthermore, leveraging advanced technologies, such as machine learning and multi-omics methods, will be critical in improving our understanding of ncRNAs biomarkers and translating these insights into impactful clinical applications.
Collapse
Affiliation(s)
- Muqtada Shaikh
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, 400 056, India
| | - Gaurav Doshi
- Department of Pharmacology, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, Mumbai, 400 056, India.
| |
Collapse
|
6
|
Chen L, Zhou H, Wu H, Lu Q, Huang J, Wang S. Effect of immunotherapy or anti-angiogenesis therapy combined with chemotherapy for advanced triple-negative breast cancer: A real-world retrospective study. Int Immunopharmacol 2024; 143:113516. [PMID: 39515042 DOI: 10.1016/j.intimp.2024.113516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 10/09/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors combined with chemotherapy (ICI-chemo) and anti-angiogenesis therapy combined with chemotherapy (anti-angio-chemo) have demonstrated superiority over traditional chemotherapy in patients with advanced triple-negative breast cancer (TNBC). However, due to the absence of a direct comparison between ICI-chemo and anti-angio-chemo, it remains unclear which treatment is superior. OBJECTIVE This study aimed to investigate the efficacy and safety of ICI-chemo or anti-angio-chemo for advanced TNBC at Sun Yat-sen University Cancer Center. METHODS A retrospective analysis was conducted on the medical records of advanced TNBC patients who received ICI-chemo or anti-angio-chemo treatment between January 2017 and March 2023. Survival outcomes and safety profiles were evaluated. RESULTS A total of 178 patients were enrolled, including 101 who received ICI-chemo and 77 who received anti-angio-chemo. The median follow-up time was 19.93 months [95 % confidence interval (CI): 17.05-22.81]. There was no significant difference in patient outcomes, including progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and disease control rate (DCR) between the two treatment regimens in the overall population. However, ICI-chemo demonstrated clinical survival benefits, with significant improvements in PFS and OS [hazard ratio (HR) = 0. 546, P = 0.048; HR = 0.313, P = 0.032] in patients receiving first-line treatment. The median PFS (mPFS) for the ICI-chemo and anti-angio-chemo cohorts was 9.37 and 6.03 months, respectively. Univariate and multivariate analyses showed that ICI-chemo independently achieved favorable PFS. No statistically significant difference was observed in PFS or OS between patients who received second-line or later ICI-chemo or anti-angio-chemo. The mPFS was 4.83 and 5.03 months, respectively. The toxicity profiles of adverse events were similar across two cohorts. CONCLUSION Among patients with advanced TNBC, ICI-chemo is associated with potentially longer survival compared to anti-angio-chemo as first-line treatment. Given their efficacy and better cost-effectiveness, these two treatment regimens may be considered potentially effective options for second-line therapy and beyond.
Collapse
Affiliation(s)
- Limin Chen
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou 510060, Guangdong, China
| | - Hanxing Zhou
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou 510060, Guangdong, China
| | - Huailiang Wu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou 510060, Guangdong, China
| | - Qianyi Lu
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou 510060, Guangdong, China
| | - Jiajia Huang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou 510060, Guangdong, China
| | - Shusen Wang
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Provincial Clinical Research Center for Cancer, Guangzhou 510060, Guangdong, China.
| |
Collapse
|
7
|
Hajimolaali M, Dorkoosh FA, Antimisiaris SG. Review of recent preclinical and clinical research on ligand-targeted liposomes as delivery systems in triple negative breast cancer therapy. J Liposome Res 2024; 34:671-696. [PMID: 38520185 DOI: 10.1080/08982104.2024.2325963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 02/06/2024] [Accepted: 02/27/2024] [Indexed: 03/25/2024]
Abstract
Triple-negative breast Cancer (TNBC) is one of the deadliest types, making up about 20% of all breast cancers. Chemotherapy is the traditional manner of progressed TNBC treatment; however, it has a short-term result with a high reversibility pace. The lack of targeted treatment limited and person-dependent treatment options for those suffering from TNBC cautions to be the worst type of cancer among breast cancer patients. Consequently, appropriate treatment for this disease is considered a major clinical challenge. Therefore, various treatment methods have been developed to treat TNBC, among which chemotherapy is the most common and well-known approach recently studied. Although effective methods are chemotherapies, they are often accompanied by critical limitations, especially the lack of specific functionality. These methods lead to systematic toxicity and, ultimately, the expansion of multidrug-resistant (MDR) cancer cells. Therefore, finding novel and efficient techniques to enhance the targeting of TNBC treatment is an essential requirement. Liposomes have demonstrated that they are an effective method for drug delivery; however, among a large number of liposome-based drug delivery systems annually developed, a small number have just received authorization for clinical application. The new approaches to using liposomes target their structure with various ligands to increase therapeutic efficiency and diminish undesired side effects on various body tissues. The current study describes the most recent strategies and research associated with functionalizing the liposomes' structure with different ligands as targeted drug carriers in treating TNBCs in preclinical and clinical stages.
Collapse
Affiliation(s)
- Mohammad Hajimolaali
- Department of Pharmacy, Laboratory of Pharmaceutical Technology, University of Patras, Patras, Greece
| | - Farid Abedin Dorkoosh
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Medical Biomaterial Research Center (MBRC), Tehran University of Medical Sciences, Tehran, Iran
| | - Sophia G Antimisiaris
- Department of Pharmacy, Laboratory of Pharmaceutical Technology, University of Patras, Patras, Greece
- Institute of Chemical Engineering, Foundation for Research and Technology Hellas, FORTH/ICEHT, Patras, Greece
| |
Collapse
|
8
|
Pellegrino B, David K, Rabani S, Lampert B, Tran T, Doherty E, Piecychna M, Meza-Romero R, Leng L, Hershkovitz D, Vandenbark AA, Bucala R, Becker-Herman S, Shachar I. CD74 promotes the formation of an immunosuppressive tumor microenvironment in triple-negative breast cancer in mice by inducing the expansion of tolerogenic dendritic cells and regulatory B cells. PLoS Biol 2024; 22:e3002905. [PMID: 39576827 PMCID: PMC11623796 DOI: 10.1371/journal.pbio.3002905] [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: 12/21/2023] [Revised: 12/06/2024] [Accepted: 10/18/2024] [Indexed: 11/24/2024] Open
Abstract
CD74 is a cell-surface receptor for the cytokine macrophage migration inhibitory factor (MIF). MIF binding to CD74 induces a signaling cascade resulting in the release of its cytosolic intracellular domain (CD74-ICD), which regulates transcription in naïve B and chronic lymphocytic leukemia (CLL) cells. In the current study, we investigated the role of CD74 in the regulation of the immunosuppressive tumor microenvironment (TME) in triple-negative breast cancer (TNBC). TNBC is the most aggressive breast cancer subtype and is characterized by massive infiltration of immune cells to the tumor microenvironment, making this tumor a good candidate for immunotherapy. The tumor and immune cells in TNBC express high levels of CD74; however, the function of this receptor in the tumor environment has not been extensively characterized. Regulatory B cells (Bregs) and tolerogenic dendritic cells (tol-DCs) were previously shown to attenuate the antitumor immune response in TNBC. Here, we demonstrate that CD74 enhances tumor growth by inducing the expansion of tumor-infiltrating tol-DCs and Bregs. Utilizing CD74-KO mice, Cre-flox mice lacking CD74 in CD23+ mature B cells, mice lacking CD74 in the CD11c+ population, and a CD74 inhibitor (DRQ), we elucidate the mechanism by which CD74 inhibits antitumor immunity. MIF secreted from the tumor cells activates CD74 expressed on DCs. This activation induces the binding of CD74-ICD to the SP1 promotor, resulting in the up-regulation of SP1 expression. SP1 binds the IL-1β promotor, leading to the down-regulation of its transcription. The reduced levels of IL-1β lead to decreased antitumor activity by allowing expansion of the tol-DC, which induces the expansion of the Breg population, supporting the cross-talk between these 2 populations. Taken together, these results suggest that CD74+ CD11c+ DCs are the dominant cell type involved in the regulation of TNBC progression. These findings indicate that CD74 might serve as a novel therapeutic target in TNBC.
Collapse
Affiliation(s)
- Bianca Pellegrino
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Keren David
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Stav Rabani
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Bar Lampert
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Thuy Tran
- Yale Cancer Center and School of Medicine, New Haven, Connecticut, United States of America
| | - Edward Doherty
- Yale Cancer Center and School of Medicine, New Haven, Connecticut, United States of America
| | - Marta Piecychna
- Yale Cancer Center and School of Medicine, New Haven, Connecticut, United States of America
| | - Roberto Meza-Romero
- Neuroimmunology Research, VA Portland Health Care System, Portland, Oregon, United States of America
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Lin Leng
- Yale Cancer Center and School of Medicine, New Haven, Connecticut, United States of America
| | - Dov Hershkovitz
- Insitute of Pathology, Sourasky Medical Center, Tel Aviv, Israel
| | - Arthur A. Vandenbark
- Neuroimmunology Research, VA Portland Health Care System, Portland, Oregon, United States of America
- Department of Neurology, Oregon Health & Science University, Portland, Oregon, United States of America
- Department of Molecular Microbiology and Immunology, Oregon Health & Science University, Portland, Oregon, United States of America
| | - Richard Bucala
- Yale Cancer Center and School of Medicine, New Haven, Connecticut, United States of America
| | - Shirly Becker-Herman
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Idit Shachar
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
9
|
Sen K, Kumar Das S, Ghosh N, Sinha K, Sil PC. Lupeol: A dietary and medicinal triterpene with therapeutic potential. Biochem Pharmacol 2024; 229:116545. [PMID: 39293501 DOI: 10.1016/j.bcp.2024.116545] [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: 06/03/2024] [Revised: 09/04/2024] [Accepted: 09/13/2024] [Indexed: 09/20/2024]
Abstract
Lupeol, a triterpene derived from various plants, has emerged as a potent dietary supplement with extensive therapeutic potential. This review offers a comprehensive examination of lupeol's applications across diverse health conditions. By meticulously analyzing current scientific literature, we have synthesized findings that underscore lupeol's impact on cancer, diabetes, gastrointestinal disorders, neurological diseases, dermatological conditions, nephrological issues, and cardiovascular health. The review delves into molecular studies that reveal lupeol's ability to modulate disease pathways and alleviate symptoms, positioning it as a promising therapeutic agent. Moreover, we discuss the potential role of lupeol in clinical practice and public health strategies, emphasizing its substantial benefits as a natural compound. This thorough analysis serves as a critical resource for researchers, providing insights into the multifaceted therapeutic properties of lupeol and its potential to significantly enhance health outcomes.
Collapse
Affiliation(s)
- Koushik Sen
- Jhargram Raj College, Jhargram 721507, India
| | | | | | | | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Kolkata 700054, India.
| |
Collapse
|
10
|
Sinanian MM, Rahman A, Elshazly AM, Neely V, Nagarajan B, Kellogg GE, Risinger AL, Gewirtz DA. A BPTF Inhibitor That Interferes with the Multidrug Resistance Pump to Sensitize Murine Triple-Negative Breast Cancer Cells to Chemotherapy. Int J Mol Sci 2024; 25:11346. [PMID: 39518898 PMCID: PMC11545213 DOI: 10.3390/ijms252111346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 07/31/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is associated with a generally poor prognosis due to its highly aggressive and metastatic nature, lack of targetable receptors, as well as the frequent development of resistance to chemotherapy. We previously reported that AU1, a small molecule developed as an inhibitor of BPTF (bromodomain PHD finger-containing transcription factor), was capable of sensitizing preclinical models of TNBC to chemotherapy in part via the promotion of autophagy. In studies reported here, we identify an additional property of this compound, specifically that sensitization is associated with the inhibition of the P-glycoprotein (P-gp) efflux pump. In silico molecular docking studies indicate that AU1 binds to active regions of the efflux pump in a manner consistent with the inhibition of the pump function. This work identifies a novel chemical structure that can influence multidrug efflux, an established mechanism of drug resistance in TNBC, that has not yet been successfully addressed by clinical efforts.
Collapse
Affiliation(s)
- Melanie M. Sinanian
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA; (M.M.S.); (A.R.); (A.M.E.)
| | - Afshan Rahman
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA; (M.M.S.); (A.R.); (A.M.E.)
| | - Ahmed M. Elshazly
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA; (M.M.S.); (A.R.); (A.M.E.)
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Victoria Neely
- Philips Institute for Oral Health Research, School of Dentistry, Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Balaji Nagarajan
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA; (B.N.); (G.E.K.)
| | - Glen E. Kellogg
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA; (B.N.); (G.E.K.)
| | - April L. Risinger
- Department of Pharmacology, University of Texas Health Science Center, San Antonio, TX 78229, USA;
| | - David A. Gewirtz
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, Richmond, VA 23298, USA; (M.M.S.); (A.R.); (A.M.E.)
| |
Collapse
|
11
|
Ahmadi-Hadad A, de Queiroz PCC, Schettini F, Giuliano M. Reawakening the master switches in triple-negative breast cancer: A strategic blueprint for confronting metastasis and chemoresistance via microRNA-200/205: A systematic review. Crit Rev Oncol Hematol 2024; 204:104516. [PMID: 39306311 DOI: 10.1016/j.critrevonc.2024.104516] [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: 12/28/2023] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Triple-negative breast cancer (TNBC) exhibits a proclivity for early recurrence and development of metastasis. Moreover, drug resistance tends to arise few months following chemotherapeutic regimen with agents such as Doxorubicin, Paclitaxel, Docetaxel, and Cisplatin. miR-200 family and miR-205 are considered key regulators of metastasis by regulating the Epithelial-to-mesenchymal transition (EMT) via inhibiting ZEB1. Therefore, these microRNAs may offer therapeutic applications. Moreover, they hold potential for inhibiting chemoresistance and increasing chemosensitivity. These microRNAs are suppressed in TNBC cells. Increasing their levels, however, can inhibit EMT and improve progression-free survival (PFS). Besides using direct miRNA therapy via viral vectors, some drugs like Acetaminophen, or Tamoxifen are deemed useful for TNBC due to their ability to upregulate these miRNAs. In this review, by conducting an advanced search on PubMed, Embase, and Medline and selecting pertinent studies, we aimed to explore the potential applications of these microRNAs in controlling EMT and overcoming chemoresistance.
Collapse
Affiliation(s)
- Armia Ahmadi-Hadad
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| | | | - Francesco Schettini
- Faculty of Medicine, University of Barcelona, Barcelona, Spain; Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain; Translational Genomics and Targeted Therapies in Solid Tumors, August Pi I Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain, University of Barcelona, Barcelona, Spain.
| | - Mario Giuliano
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
12
|
Metwali E, Pennington S. Mass Spectrometry-Based Proteomics for Classification and Treatment Optimisation of Triple Negative Breast Cancer. J Pers Med 2024; 14:944. [PMID: 39338198 PMCID: PMC11432759 DOI: 10.3390/jpm14090944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/30/2024] Open
Abstract
Triple-negative breast cancer (TNBC) presents a significant medical challenge due to its highly invasive nature, high rate of metastasis, and lack of drug-targetable receptors, which together lead to poor prognosis and limited treatment options. The traditional treatment guidelines for early TNBC are based on a multimodal approach integrating chemotherapy, surgery, and radiation and are associated with low overall survival and high relapse rates. Therefore, the approach to treating early TNBC has shifted towards neoadjuvant treatment (NAC), given to the patient before surgery and which aims to reduce tumour size, reduce the risk of recurrence, and improve the pathological complete response (pCR) rate. However, recent studies have shown that NAC is associated with only 30% of patients achieving pCR. Thus, novel predictive biomarkers are essential if treatment decisions are to be optimised and chemotherapy toxicities minimised. Given the heterogeneity of TNBC, mass spectrometry-based proteomics technologies offer valuable tools for the discovery of targetable biomarkers for prognosis and prediction of toxicity. These biomarkers can serve as critical targets for therapeutic intervention. This review aims to provide a comprehensive overview of TNBC diagnosis and treatment, highlighting the need for a new approach. Specifically, it highlights how mass spectrometry-based can address key unmet clinical needs by identifying novel protein biomarkers to distinguish and early prognostication between TNBC patient groups who are being treated with NAC. By integrating proteomic insights, we anticipate enhanced treatment personalisation, improved clinical outcomes, and ultimately, increased survival rates for TNBC patients.
Collapse
Affiliation(s)
- Essraa Metwali
- School of Medicine, UCD Conway Institute for Biomolecular Research, University College Dublin, D04 C1P1 Dublin, Ireland;
- King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard, Jeddah-Makka Expressway, Jeddah 22384, Saudi Arabia
| | - Stephen Pennington
- School of Medicine, UCD Conway Institute for Biomolecular Research, University College Dublin, D04 C1P1 Dublin, Ireland;
| |
Collapse
|
13
|
Zárate-Pérez A, Cruz-Cázares AP, Ordaz-Rosado D, García-Quiroz J, León-Del-Rio A, Avila E, Milo-Rocha E, Díaz L, García-Becerra R. The vitamin D analog EB1089 sensitizes triple-negative breast cancer cells to the antiproliferative effects of antiestrogens. Adv Med Sci 2024; 69:398-406. [PMID: 39233278 DOI: 10.1016/j.advms.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 05/12/2024] [Accepted: 08/31/2024] [Indexed: 09/06/2024]
Abstract
PURPOSE Patients bearing estrogen receptor (ER)α-negative breast cancer tumors confront poor prognosis and are typically unresponsive to hormone therapy. Previous studies have shown that calcitriol, the active vitamin D metabolite, can induce ERα expression in ERα-negative cells. EB1089, a calcitriol analog with reduced calcemic effects, exhibits greater potency than calcitriol in inhibiting cancer cell growth. However, the impact of EB1089 on ERα expression in triple-negative breast cancer (TNBC) cells remains unexplored. This study aims to investigate whether EB1089 could induce functional ERα expression in TNBC cell lines, potentially enabling the antiproliferative effects of antiestrogens. MATERIALS AND METHODS TNBC cell lines HCC1806 and HCC1937 were treated with EB1089, and ERα expression was analyzed using real-time PCR and Western blots. The transcriptional activity of induced ERα was evaluated through a luciferase reporter assay. The antiproliferative effects of tamoxifen and fulvestrant antiestrogens were assessed using the sulforhodamine B assay in the EB1089-treated cells. RESULTS Our findings indicated that EB1089 significantly induced ERα mRNA and protein expression in TNBC cells. Moreover, EB1089-induced ERα exhibited transcriptional activity and effectively restored the inhibitory effects of antiestrogens, thereby suppressing cell proliferation in TNBC cells. CONCLUSION EB1089 induced the expression of functional ERα in TNBC cells, restoring the antiproliferative effects of antiestrogens. These results highlight the potential of using EB1089 as a promising strategy for re-establishment of the antiproliferative effect of antiestrogens as a possible management for TNBC. This research lays the foundation for potential advancements in TNBC treatment, offering new avenues for targeted and effective interventions.
Collapse
Affiliation(s)
- Adriana Zárate-Pérez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Alitzin Pamela Cruz-Cázares
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - David Ordaz-Rosado
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico
| | - Janice García-Quiroz
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico
| | - Alfonso León-Del-Rio
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico; Programa de Investigación de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Euclides Avila
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico
| | - Edgar Milo-Rocha
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico
| | - Lorenza Díaz
- Departamento de Biología de La Reproducción Dr. Carlos Gual Castro, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, 14080, Mexico.
| | - Rocío García-Becerra
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico; Programa de Investigación de Cáncer de Mama, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, 04510, Mexico.
| |
Collapse
|
14
|
Han D, Spehar JM, Richardson DS, Leelananda S, Chakravarthy P, Grecco S, Reardon J, Stover DG, Bennett C, Sizemore GM, Li Z, Lindert S, Sizemore ST. The RAL Small G Proteins Are Clinically Relevant Targets in Triple Negative Breast Cancer. Cancers (Basel) 2024; 16:3043. [PMID: 39272901 PMCID: PMC11394424 DOI: 10.3390/cancers16173043] [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: 01/17/2024] [Revised: 02/16/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Breast cancer (BC) is the most frequent cancer and second-leading cause of cancer deaths in women in the United States. While RAS mutations are infrequent in BC, triple-negative (TN) and HER2-positive (HER2+) BC both exhibit increased RAS activity. Here, we tested the RAS effectors RALA and RALB, which are overexpressed in BC, as tractable molecular targets in these subtypes. While analysis of the breast cancer patient sample data suggests that the RALs are associated with poor outcome in both TNBC and HER2+ BC, our in vivo and in vitro experimental findings revealed the RALs to be essential in only the TNBC cell lines. While testing the response of the BC cell lines to the RAL inhibitors RBC8 and BQU57, we observed no correlation between drug efficacy and cell line dependency on RAL expression for survival, suggesting that these compounds kill via off-target effects. Finally, we report the discovery of a new small molecule inhibitor, OSURALi, which exhibits strong RAL binding, effectively inhibits RAL activation, and is significantly more toxic to RAL-dependent TNBC cells than RAL-independent HER2+ and normal cell lines. These results support the RALs as viable molecular targets in TNBC and the further investigation of OSURALi as a therapeutic agent.
Collapse
Affiliation(s)
- David Han
- Department of Radiation Oncology, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jonathan M Spehar
- Department of Radiation Oncology, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Dillon S Richardson
- Department of Radiation Oncology, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | | | - Prathik Chakravarthy
- Department of Radiation Oncology, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Samantha Grecco
- Department of Radiation Oncology, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Jesse Reardon
- Department of Radiation Oncology, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel G Stover
- Department of Internal Medicine, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Chad Bennett
- Drug Development Institute, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Gina M Sizemore
- Department of Radiation Oncology, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Zaibo Li
- Department of Pathology, Wexner Medical Center, The Ohio State University, Columbus, OH 43210, USA
| | - Steffen Lindert
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH 43210, USA
| | - Steven T Sizemore
- Department of Radiation Oncology, Arthur G. James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
15
|
Kunachowicz D, Kłosowska K, Sobczak N, Kepinska M. Applicability of Quantum Dots in Breast Cancer Diagnostic and Therapeutic Modalities-A State-of-the-Art Review. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1424. [PMID: 39269086 PMCID: PMC11396817 DOI: 10.3390/nano14171424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/15/2024]
Abstract
The increasing incidence of breast cancers (BCs) in the world population and their complexity and high metastatic ability are serious concerns for healthcare systems. Despite the significant progress in medicine made in recent decades, the efficient treatment of invasive cancers still remains challenging. Chemotherapy, a fundamental systemic treatment method, is burdened with severe adverse effects, with efficacy limited by resistance development and risk of disease recurrence. Also, current diagnostic methods have certain drawbacks, attracting attention to the idea of developing novel, more sensitive detection and therapeutic modalities. It seems the solution for these issues can be provided by nanotechnology. Particularly, quantum dots (QDs) have been extensively evaluated as potential targeted drug delivery vehicles and, simultaneously, sensing and bioimaging probes. These fluorescent nanoparticles offer unlimited possibilities of surface modifications, allowing for the attachment of biomolecules, such as antibodies or proteins, and drug molecules, among others. In this work, we discuss the potential applicability of QDs in breast cancer diagnostics and treatment in light of the current knowledge. We begin with introducing the molecular and histopathological features of BCs, standard therapeutic regimens, and current diagnostic methods. Further, the features of QDs, along with their uptake, biodistribution patterns, and cytotoxicity, are described. Based on the reports published in recent years, we present the progress in research on possible QD use in improving BC diagnostics and treatment efficacy as chemotherapeutic delivery vehicles and photosensitizing agents, along with the stages of their development. We also address limitations and open questions regarding this topic.
Collapse
Affiliation(s)
- Dominika Kunachowicz
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Karolina Kłosowska
- Students' Scientific Association at the Department of Pharmaceutical Biochemistry (SKN No. 214), Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Natalia Sobczak
- Students' Scientific Association of Biomedical and Environmental Analyses (SKN No. 85), Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| | - Marta Kepinska
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
16
|
Rezazadeh F, Saadat W, Smith R, Pattyn A, Malik M, Yazdani F, Saliganan AD, Mehrmohammadi M, Viola NT. Mild Hyperthermia Enhanced Liposomal Doxorubicin Delivery and CD8 + T cell Infiltration in Triple Negative Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.25.591226. [PMID: 38712049 PMCID: PMC11071532 DOI: 10.1101/2024.04.25.591226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Mild hyperthermia (MHTh) is often used in combination with chemotherapy and radiotherapy for cancer treatment. In the current study, the effect of MHTh on the enhanced uptake of the FDA-approved chemotherapy drug, liposomal doxorubicin (dox) in syngeneic 4T1 tumors was investigated. Doxorubicin has inherent fluorescence properties having an emission signal at 590 nm upon excitation with a 480 nm laser. A group of mice administered with doxorubicin (dox) were exposed to MHTh (42 °C) for 30 minutes whereas control group given dox did not receive MHTh. Ex vivo optical imaging of harvested tumors confirmed higher uptake of dox in treated versus the control untreated tumors. Confocal microscopy of tumor sections indicates higher fluorescent intensity due to increased accumulation of dox in MHTh-treated compared to untreated tumors. We examined the effect of MHTh to enhance CD8 tumor infiltration, production of interferon-γ (IFN-γ) and expression of programmed death ligand-1 (PD-L1). mRNA in situ hybridization was performed to test for transcripts of CD8, IFN-γ and PD-L1. Results showed that higher expression of CD8 mRNA was observed in MHTh-administered tumors versus untreated cohorts. The signal for IFN-γ and PD-L1 in both groups were not significantly different. Taken together, our findings imply that MHTh can improve tumor uptake of dox. Importantly, our data suggests that MHTh can boost CD8+ T cell infiltration.
Collapse
Affiliation(s)
- Farzaneh Rezazadeh
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Wajfa Saadat
- Department of Biological Science, Wayne State University, Detroit, MI 48201
| | - Ryan Smith
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA
| | - Alexander Pattyn
- Department of Biomedical Engineering, Wayne State University, Detroit, MI 48202, USA
| | - Mohammad Malik
- Department of Kinesiology, Wayne State University, Detroit, MI 48201
| | - Fuad Yazdani
- Department of Public Health, Wayne State University, Detroit, MI 48201
| | - Allen-Dexter Saliganan
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| | - Mohammad Mehrmohammadi
- Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY
- Department of Biomedical Engineering, University of Rochester, Rochester, NY
- Wilmot Cancer Center, Rochester, NY
| | - Nerissa T. Viola
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201
| |
Collapse
|
17
|
Li Z, Liu Q, Cai Y, Ye N, He Z, Yao Y, Ding Y, Wang P, Qi C, Zheng L, Wang L, Zhou J, Zhang QQ. EPAC inhibitor suppresses angiogenesis and tumor growth of triple-negative breast cancer. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167114. [PMID: 38447883 DOI: 10.1016/j.bbadis.2024.167114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/29/2024] [Accepted: 03/01/2024] [Indexed: 03/08/2024]
Abstract
AIMS Exchange protein directly activated by cAMP 1 (EPAC1), a major isoform of guanine nucleotide exchange factors, is highly expressed in vascular endothelia cells and regulates angiogenesis in the retina. High intratumor microvascular densities (MVD) resulting from angiogenesis is responsible for breast cancer development. Downregulation of EPAC1 in tumor cell reduces triple-negative breast cancer (TNBC)-induced angiogenesis. However, whether Epac1 expressed in vascular endothelial cells contributes to angiogenesis and tumor development of TNBC remains elusive. MAIN METHODS We employed NY0123, a previously identified potent EPAC inhibitor, to explore the anti-angiogenic biological role of EPAC1 in vitro and in vivo through vascular endothelial cells, rat aortic ring, Matrigel plug, and chick embryo chorioallantoic membrane (CAM) and yolk sac membrane (YSM) assays, as well as the in vivo xenograft tumor models of TNBC in both chick embryo and mice. KEY FINDINGS Inhibiting EPAC1 in vascular endothelial cells by NY0123 significantly suppresses angiogenesis and tumor growth of TNBC. In addition, NY0123 possesses a better inhibitory efficacy than ESI-09, a reported specific EPAC inhibitor tool compound. Importantly, inhibiting EPAC1 in vascular endothelia cells regulates the typical angiogenic signaling network, which is associated with not only vascular endothelial growth factor (VEGF)/vascular endothelial growth factor receptor-2 (VEGFR2) signaling, but also PI3K/AKT, MEK/ERK and Notch pathway. CONCLUSIONS Our findings support that EPAC1 may serve as an effective anti-angiogenic therapeutic target of TNBC, and EPAC inhibitor NY0123 has the therapeutic potential to be developed for the treatment of TNBC.
Collapse
Affiliation(s)
- Zishuo Li
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Qiao Liu
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuhao Cai
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Na Ye
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Zinan He
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuying Yao
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yi Ding
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Pingyuan Wang
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States
| | - Cuiling Qi
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lingyun Zheng
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lijing Wang
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX 77555, United States.
| | - Qian-Qian Zhang
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
18
|
Han G, Bai X, Li F, Huang L, Hao Y, Li W, Bu P, Zhang H, Liu X, Xie J. Long non-coding RNA HANR modulates the glucose metabolism of triple negative breast cancer via stabilizing hexokinase 2. Heliyon 2024; 10:e23827. [PMID: 38192790 PMCID: PMC10772629 DOI: 10.1016/j.heliyon.2023.e23827] [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: 03/06/2023] [Revised: 12/05/2023] [Accepted: 12/13/2023] [Indexed: 01/10/2024] Open
Abstract
Increasing evidence has demonstrated the oncogenic roles of long non-coding RNA (lncRNA) hepatocellular carcinoma (HCC)-associated long non-coding RNA (HANR) in the development of HCC and lung cancer; however, the involvement of HANR in triple-negative breast cancer (TNBC) remains largely unknown. Our results demonstrated the significant overexpression of HANR in TNBC tissues and cells. Higher HANR levels significantly correlated with the poorer phenotypes in patients with TNBC. HANR down-regulation inhibited the proliferation and cell cycle progression and increased the apoptosis of TNBC cells. Mechanistically, immunoprecipitation-mass spectrometry revealed hexokinase II (HK2) as a direct binding target of HANR. HANR binds to and stabilizes HK2 through the proteasomal pathway. Consistent with the important role of HK2 in cancer cells, HANR depletion represses the glucose absorbance and lactate secretion, thus reprogramming the metabolism of TNBC cells. An in vivo xenograft model also demonstrated that HANR promoted tumor growth and aerobic glycolysis. This study reveals the role of HANR in modulating the glycolysis in TNBC cells by regulating HK2 stability, suggesting that HANR is a potential drug target for TNBC.
Collapse
Affiliation(s)
- Guohui Han
- Department of Breast Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiangdong Bai
- Department of Breast Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Feng Li
- Department of Biochemistry and Molecular Biology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Li Huang
- Department of Breast Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yating Hao
- Department of Breast Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Weina Li
- Department of radiotherapy, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Peng Bu
- Department of Pathology, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huanhu Zhang
- Gastroenterology Department, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinxin Liu
- Department of Breast Surgery, Shanxi Province Cancer Hospital/Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
19
|
Ilie SM, Briot N, Constatin G, Ilie A, Beltjens F, Ladoire S, Desmoulins I, Hennequin A, Bertaut A, Coutant C, Causeret S, Ghozali N, Coudert B, Arnould L. Pathologic and immunohistochemical prognostic markers in residual triple-negative breast cancer after neoadjuvant chemotherapy. Front Oncol 2024; 13:1309890. [PMID: 38273853 PMCID: PMC10809386 DOI: 10.3389/fonc.2023.1309890] [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: 10/08/2023] [Accepted: 12/04/2023] [Indexed: 01/27/2024] Open
Abstract
Background The persistence of residual tumour after neoadjuvant chemotherapy (NAC) in localised triple-negative breast cancer (TNBC) is known to have a negative prognostic value. However, different degrees of expression of some immunohistochemical markers may correlate with different prognoses. Methods The expression of biomarkers with a known prognostic value, i.e., cytokeratin 5/6 (CK5/6), androgen receptor (AR), epidermal growth factor receptor (EGFR) proliferation-related nuclear antigen Ki-67, human epidermal growth factor receptor 2 (HER2), protein 53 (p53), forkhead box protein 3 (FOXP3), and cluster differentiation 8 (CD8), was analysed by immunohistochemistry in 111 samples after NAC in non-metastatic TNBC patients addressed to Georges-François Leclerc Cancer Centre Dijon, France. Clinical and pathological variables were retrospectively collected. Cox regression was used to identify immunohistochemical (IHC) and clinicopathological predictors of event-free survival (EFS) (relapse or death). Results Median age was 50.4 years (range 25.6-88.3), 55.9% (n = 62) were non-menopausal, 70 (63.1%) had stage IIA-IIB disease. NAC was mostly sequential anthracycline-taxanes (72.1%), and surgical intervention was principally conservative (51.3%). We found 65.7% ypT1, 47.2% lymph node involvement (ypN+), and 29.4% lymphovascular invasion (LVI). Most residual tumours were EGFR >110 (H-score) (60.5%, n = 66), AR ≥4% (53.2%, n = 58), p53-positive mutated (52.7%, n = 58), CD8 ≥26 (58.1%, n = 61), FOXP3 ≥7 (51.4%, n = 54), more than half in the stroma, and 52.3% (n = 58) HER2 score 0. After a median follow-up of 80.8 months, 48.6% had relapsed. Median EFS was 62.3 months (95% CI, 37.2-not reached (NR)). Factors independently associated with poor EFS were AR-low (p = 0.002), ypN+ (p < 0.001), and LVI (p = 0.001). Factors associated with lower overall survival (OS) were EGFR-low (p = 0.041), Ki-67 high (p = 0.024), and ypN+ (p < 0.001). Conclusion Post-NAC residual disease in TNBC showed biomarkers specific to a basal-like subtype and markers of lymphocyte infiltration mostly present in the stroma. Prognostic markers for EFS were AR, LVI, and ypN and warrant further validation in a prognostic model.
Collapse
Affiliation(s)
- Silvia Mihaela Ilie
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Nathalie Briot
- Department of Biostatistics Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Guillaume Constatin
- Department of Biostatistics Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Alis Ilie
- Cancer Biology Research Platform, Centre Georges Francois Leclerc, Dijon, France
| | - Francoise Beltjens
- Department of Bio-pathology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Sylvain Ladoire
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
- Cancer Biology Research Platform, Centre Georges Francois Leclerc, Dijon, France
| | - Isabelle Desmoulins
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Audrey Hennequin
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Aurelie Bertaut
- Department of Biostatistics Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Charles Coutant
- Surgery Department Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Sylvain Causeret
- Surgery Department Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Niama Ghozali
- Department of Medical Oncology, University Hospital Mohammed VI, Tangier, Morocco
| | - Bruno Coudert
- Department of Medical Oncology, Georges Francois Leclerc Cancer Centre, Dijon, France
| | - Laurent Arnould
- Department of Bio-pathology, Georges Francois Leclerc Cancer Centre, Dijon, France
| |
Collapse
|
20
|
Dinakar YH, Rajana N, Kumari NU, Jain V, Mehra NK. Recent Advances of Multifunctional PLGA Nanocarriers in the Management of Triple-Negative Breast Cancer. AAPS PharmSciTech 2023; 24:258. [PMID: 38097825 DOI: 10.1208/s12249-023-02712-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Even though chemotherapy stands as a standard option in the therapy of TNBC, problems associated with it such as anemia, bone marrow suppression, immune suppression, toxic effects on healthy cells, and multi-drug resistance (MDR) can compromise their effects. Nanoparticles gained paramount importance in overcoming the limitations of conventional chemotherapy. Among the various options, nanotechnology has appeared as a promising path in preclinical and clinical studies for early diagnosis of primary tumors and metastases and destroying tumor cells. PLGA has been extensively studied amongst various materials used for the preparation of nanocarriers for anticancer drug delivery and adjuvant therapy because of their capability of higher encapsulation, easy surface functionalization, increased stability, protection of drugs from degradation versatility, biocompatibility, and biodegradability. Furthermore, this review also provides an overview of PLGA-based nanoparticles including hybrid nanoparticles such as the inorganic PLGA nanoparticles, lipid-coated PLGA nanoparticles, cell membrane-coated PLGA nanoparticles, hydrogels, exosomes, and nanofibers. The effects of all these systems in various in vitro and in vivo models of TNBC were explained thus pointing PLGA-based NPs as a strategy for the management of TNBC.
Collapse
Affiliation(s)
- Yirivinti Hayagreeva Dinakar
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India
| | - Naveen Rajana
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India
| | - Nalla Usha Kumari
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, 570015, India
| | - Neelesh Kumar Mehra
- Pharmaceutical Nanotechnology Research Laboratory, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, 500 037, India.
| |
Collapse
|
21
|
Han Y, Wang J, Sun T, Ouyang Q, Li J, Yuan J, Xu B. Predictive biomarkers of response and survival following immunotherapy with a PD-L1 inhibitor benmelstobart (TQB2450) and antiangiogenic therapy with a VEGFR inhibitor anlotinib for pretreated advanced triple negative breast cancer. Signal Transduct Target Ther 2023; 8:429. [PMID: 37973901 PMCID: PMC10654734 DOI: 10.1038/s41392-023-01672-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/17/2023] [Accepted: 10/10/2023] [Indexed: 11/19/2023] Open
Abstract
In our phase Ib trial (ClinialTrials.gov Identifier: NCT03855358), benmelstobart (TQB2450), a novel humanized IgG1 antibody against PD-L1, plus antiangiogenic multikinase inhibitor, anlotinib, demonstrated promising antitumor activities in pretreated triple negative breast cancer (TNBC) patients. We conducted explorative analyses of genomic biomarkers to explore the associations with treatment response and survival outcomes. Targeted next generation sequencing (NGS) was undertaken toward circulating tumor DNA (ctDNA) collected from peripheral blood samples prior to the start of treatment and after disease progression. A total of 31 patients received targeted NGS and functional driver mutations in 29 patients were analyzed. The most frequent mutations were TP53 (72%), MLL3 (28%), and PIK3CA (17%). At a blood-based tumor mutational burden (bTMB) cutoff of 6.7 mutations per megabase, patients with low bTMB showed better response to anlotinib plus TQB2450 (50% vs. 7%, P = 0.015) and gained greater PFS benefits (7.3 vs. 4.1 months, P = 0.012) than those with high bTMB. At a maximum somatic allele frequency (MSAF) cutoff of 10%, a low MSAF indicated a better objective response (43% vs. 20%) as well as a significantly longer median PFS (7.9 vs. 2.7 months, P < 0.001). Patients with both low MSAF and low bTMB showed a notably better objective response to anlotinib plus TQB2450 (70% vs. 11%, P < 0.001) and a significantly longer median PFS (11.0 vs. 2.9 months, P < 0.001) than patients with other scenarios. Our findings support future studes and validation of MSAF and the combined bTMB-MSAF classification as predictive biomarkers of immune checkpoint inhibitor-based regimens in advanced TNBC patients.
Collapse
Affiliation(s)
- Yiqun Han
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jiayu Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Tao Sun
- Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, 110042, China
| | | | - Jianwen Li
- Geneplus-Shenzhen, Shenzhen, 518118, China
| | - Jie Yuan
- Geneplus-Shenzhen, Shenzhen, 518118, China
| | - Binghe Xu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
22
|
Gorenšek R, Kresnik M, Takač I, Rojko T, Sobočan M. Advances in Tumour-Infiltrating Lymphocytes for Triple-Negative Breast Cancer Management. BREAST CANCER (DOVE MEDICAL PRESS) 2023; 15:773-783. [PMID: 37936879 PMCID: PMC10627091 DOI: 10.2147/bctt.s399157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 08/13/2023] [Indexed: 11/09/2023]
Abstract
Triple negative breast cancer (TNBC) is a subtype of breast cancer which does not express or expresses a minimum amount of estrogen receptors (ER), progesterone receptors (PR) and human epidermal growth factor receptor 2 (HER2) protein. TNBCs include a heterogenic group of cancers that are aggressive, grow rapidly and are associated with poor prognosis and overall survival, mainly attributed to a lack of effective therapeutic targets. For a long time, a major issue with predicting the outcome and prognosis of TNBCs was the lack of an accurate biomarker, a molecule that helps us objectively assess a patient's health status. In recent times, defining the presence of tumor-infiltrating lymphocytes (TIL) is becoming an indispensable method of determining a patient's prognosis. TILs are found in tumor tissue and the surrounding stroma and carry a prognostic value. Furthermore, they are known to improve the effect of systemic therapy. With the rise of immunotherapy, the role of TIL in this newer therapeutic option is a topic of increased importance. The goal behind this research article is a comprehensive review of the current literature on the importance of tumor-infiltrating lymphocytes in the prognosis of TNBC.
Collapse
Affiliation(s)
- Rok Gorenšek
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Oncology, University Medical Centre Maribor, Maribor, Slovenia
| | - Martin Kresnik
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Emergency Medicine, University Medical Centre Maribor, Maribor, Slovenia
| | - Iztok Takač
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Division of Gynecology and Perinatology, University Medical Centre Maribor, Maribor, Slovenia
| | - Tomaž Rojko
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Department of Pathology, University Medical Centre Maribor, Maribor, Slovenia
| | - Monika Sobočan
- Faculty of Medicine, University of Maribor, Maribor, Slovenia
- Division of Gynecology and Perinatology, University Medical Centre Maribor, Maribor, Slovenia
| |
Collapse
|
23
|
Wu Q, Ma X, Jin Z, Ni R, Pan Y, Yang G. Zhuidu Formula suppresses the migratory and invasive properties of triple-negative breast cancer cells via dual signaling pathways of RhoA/ROCK and CDC42/MRCK. JOURNAL OF ETHNOPHARMACOLOGY 2023; 315:116644. [PMID: 37196814 DOI: 10.1016/j.jep.2023.116644] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 05/04/2023] [Accepted: 05/15/2023] [Indexed: 05/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Zhuidu Formula (ZDF) is composed of triptolide, cinobufagin and paclitaxel, which are the active ingredients of Tripterygium wilfordii Hook. F, dried toad skin and Taxus wallichiana var. chinensis (Pilg) Florin, respectively. Modern pharmacological studies show that triptolide, cinobufagin, and paclitaxel are well-known natural compounds that exert anti-tumor effects by interfering with DNA synthesis, inducing tumor cell apoptosis, and inhibiting the dynamic balance of the tubulin. However, the mechanism by which the three compounds inhibit triple-negative breast cancer (TNBC) metastasis is unknown. OBJECTIVE The objective of this investigation was to examine the inhibitory essences of ZDF on the metastasis of TNBC and elucidate its potential mechanism. MATERIALS AND METHODS Cell viability of triptolide (TPL), cinobufagin (CBF), and paclitaxel (PTX) on MDA-MB-231 cells was assessed employing a CCK-8 assay. The drug interactions of the three drugs on MDA-MB-231 cells were determined in vitro utilizing the Chou-Talalay method. MDA-MB-231 cells were identified for migration, invasion and adhesion in vitro through the implementation of the scratch assay, transwell assay and adhesion assay, respectively. The formation of cytoskeleton protein F-actin was detected by immunofluorescence assay. The expressions of MMP-2 and MMP-9 in the supernatant of the cells were determined by ELISA analysis. The Western blot and RT-qPCR were employed to explore the protein expressions associated with the dual signaling pathways of RhoA/ROCK and CDC42/MRCK. The anti-tumor efficacy of ZDF in vivo and its preliminary mechanism were investigated in the mouse 4T1 TNBC model. RESULTS The results demonstrated that ZDF could significantly reduce the viability of the MDA-MB-231 cell, and the combination index (CI) values of actual compatibility experimental points were all less than 1, demonstrating a favorable synergistic compatibility relationship. It was found that ZDF reduces RhoA/ROCK and CDC42/MRCK dual signaling pathways, which are responsible for MDA-MB-231cell migration, invasion, and adhesion. Additionally, there has been a significant reduction in the manifestation of cytoskeleton-related proteins. Furthermore, the expression levels of RhoA, CDC42, ROCK2, and MRCKβ mRNA and protein were down-regulated. ZDF significantly decreased the protein expressions of vimentin, cytokeratin-8, Arp2 and N-WASP, and inhibited actin polymerization and actomyosin contraction. Furthermore, MMP-2 and MMP-9 levels in the high-dose ZDF group were decreased by 30% and 26%, respectively. ZDF significantly reduced the tumor volume and protein expressions of ROCK2 and MRCKβ in tumor tissues without eliciting any perceptible alterations in the physical mass of the mice, and the reduction was more pronounced than that of the BDP5290 treated group. CONCLUSION The current investigation demonstrates that ZDF exhibits a proficient inhibitory impact on TNBC metastasis by regulating cytoskeletal proteins through the dual signaling pathways of RhoA/ROCK and CDC42/MRCK. Furthermore, the findings indicate that ZDF has significant anti-tumorigenic and anti-metastatic characteristics in breast cancer animal models.
Collapse
Affiliation(s)
- Qinhang Wu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Xuelin Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Zhuolin Jin
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Ruijun Ni
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Yang Pan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China.
| | - Guangming Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China.
| |
Collapse
|
24
|
Hu X, Zhao W, Li R, Chai K, Shang F, Shi S, Dong C. A cascade nanoplatform for the regulation of the tumor microenvironment and combined cancer therapy. NANOSCALE 2023; 15:16314-16322. [PMID: 37786260 DOI: 10.1039/d3nr03199c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/04/2023]
Abstract
Recently, disulfiram (DSF), an anti-alcoholism drug, has attracted increasing biomedical interest due to its anticancer effects. However, the anticancer activity of DSF is Cu(II)-dependent and it is extremely unstable, which severely hinders its clinical translation. Herein, we report the fabrication of a multifunctional nanoplatform (MCDGF) that can improve the stability of diethyldithiocarbamate (DTC), a main metabolite of DSF, by modifying the aryl boronic ester group to form a prodrug (DQ), and also realize the in situ generation of Cu(DTC)2, which relies on a cascade reaction. The delivered Cu/DQ induces immunogenic cell death (ICD) and powerfully enhances immune responses of cytotoxic T lymphocytes (CTLs) and the infiltration of dendritic cells as well as T cells. Furthermore, the grafted glucose oxidase (GOx) decomposes glucose, thus "starving" the cancer cells and providing H2O2 for the production of Cu(DTC)2. More importantly, H2O2 significantly promotes the polarization of macrophages to the anti-tumor subtype. The nano-carrier "mesoporous polydopamine (MPDA)" also displays a good photothermal therapeutic effect. The nanoplatform-integrated chemotherapy, starvation therapy, photothermal therapy, and immunotherapy synergistically stimulated CTL activation and M1 macrophage polarization. Taken together, the as-prepared nanoplatform could regulate the tumor immune microenvironment and eliminate cancer with combined cancer therapy, which will offer a promising strategy for cancer treatment and promote the clinical application of DSF in breast cancer.
Collapse
Affiliation(s)
- Xiaochun Hu
- Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
- School of Medicine, Shanghai University, Shanghai 200444, China
| | - Wenrong Zhao
- Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Ruihao Li
- Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Keke Chai
- Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Fangjian Shang
- College of Aeronautical Engineering, Binzhou University, Binzhou 256603, China
| | - Shuo Shi
- Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| | - Chunyan Dong
- Department of Comprehensive Cancer Therapy, Shanghai East Hospital, School of Medicine, Shanghai Key Laboratory of Chemical Assessment and Sustainability, School of Chemical Science and Engineering, Tongji University, Shanghai 200092, China.
| |
Collapse
|
25
|
Giacomini A, Turati M, Grillo E, Rezzola S, Ghedini GC, Schuind AC, Foglio E, Maccarinelli F, Faletti J, Filiberti S, Chambery A, Valletta M, Melocchi L, Gofflot S, Chiavarina B, Turtoi A, Presta M, Ronca R. The PTX3/TLR4 autocrine loop as a novel therapeutic target in triple negative breast cancer. Exp Hematol Oncol 2023; 12:82. [PMID: 37749607 PMCID: PMC10519006 DOI: 10.1186/s40164-023-00441-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 09/04/2023] [Indexed: 09/27/2023] Open
Abstract
BACKGROUND The pattern recognition receptor long pentraxin-3 (PTX3) plays conflicting roles in cancer by acting as an oncosuppressor or as a pro-tumor mediator depending on tumor context. Triple negative breast cancer (TNBC) represents the most aggressive histotype of breast cancer, characterized by the lack of efficacious therapeutic targets/approaches and poor prognosis. Thus, the characterization of new molecular pathways and/or alternative druggable targets is of great interest in TNBC. METHODS The expression of PTX3 in BC tumor samples and in BC cell lines has been analyzed using the Gene Expression-Based Outcome for Breast Cancer Online (GOBO), qPCR, Western blot and ELISA assay. The contribution of tumor and stromal cells to PTX3 production in TNBC was assessed by analyzing single cell RNA sequencing data and RNAscope performed on TNBC tumor samples. In order to investigate the effects of PTX3 in TNBC, different cell lines were engineered to knock-down (MDA-MB-231 and BT549 cells) or overexpress (MDA-MB-468 and E0771 cells) PTX3. Finally, using these engineered cells, in vitro (including gene expression profiling and gene set enrichment analyses) and in vivo (orthotopic tumor models in immune-compromised and immune competent mice) analyses were performed to assess the role and the molecular mechanism(s) exerted by PTX3 in TNBC. RESULTS In silico and experimental data indicate that PTX3 is mainly produced by tumor cells in TNBC and that its expression levels correlate with tumor stage. Accordingly, gene expression and in vitro results demonstrate that PTX3 overexpression confers a high aggressive/proliferative phenotype and fosters stem-like features in TNBC cells. Also, PTX3 expression induces a more tumorigenic potential when TNBC cells are grafted orthotopically in vivo. Conversely, PTX3 downregulation results in a less aggressive behavior of TNBC cells. Mechanistically, our data reveal that PTX3 drives the activation of the pro-tumorigenic Toll-like receptor 4 (TLR4) signaling pathway in TNBC, demonstrating for the first time that the PTX3/TLR4 autocrine stimulation loop contributes to TNBC aggressiveness and that TLR4 inhibition significantly impacts the growth of PTX3-producing TNBC cells. CONCLUSION Altogether, these data shed light on the role of tumor-produced PTX3 in TNBC and uncover the importance of the PTX3/TLR4 axis for therapeutic and prognostic exploitation in TNBC.
Collapse
Affiliation(s)
- Arianna Giacomini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| | - Marta Turati
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Elisabetta Grillo
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gaia Cristina Ghedini
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Ander Churruca Schuind
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Eleonora Foglio
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Federica Maccarinelli
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Jessica Faletti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Serena Filiberti
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Angela Chambery
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), University of Campania 'Luigi Vanvitelli', Caserta, Italy
| | - Mariangela Valletta
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies (DiSTABiF), University of Campania 'Luigi Vanvitelli', Caserta, Italy
| | - Laura Melocchi
- Pathology Unit, Fondazione Poliambulanza Hospital Institute, Brescia, 25121, Italy
| | | | - Barbara Chiavarina
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, University of Montpellier, Montpellier, France
| | - Andrei Turtoi
- Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, University of Montpellier, Montpellier, France
| | - Marco Presta
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy.
| |
Collapse
|
26
|
Caetano S, Garcia AR, Figueira I, Brito MA. MEF2C and miR-194-5p: New Players in Triple Negative Breast Cancer Tumorigenesis. Int J Mol Sci 2023; 24:14297. [PMID: 37762600 PMCID: PMC10531597 DOI: 10.3390/ijms241814297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 09/08/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
Among breast cancer (BC) subtypes, the most aggressive is triple negative BC (TNBC), which is prone to metastasis. We previously found that microRNA (miR)-194-5p is downregulated at the early stages of TNBC brain metastasis development. Additionally, the transcription factor myocyte enhancer factor 2 (MEF2)C, a bioinformatically predicted miR-194-5p target, was increasingly expressed throughout TNBC brain metastasis formation and disease severity. However, the contributions of these two players to malignant cells' features remain undetermined. This study aimed at disclosing the role of miR-194-5p and MEF2C in TNBC tumorigenesis. The transfection of 4T1 cells with a silencer for MEF2C or with a pre-miRNA for miR-194-5p was employed to study TNBC cells' phenotypic alterations regarding epithelial and mesenchymal markers, as well as migratory capability alterations. MEF2C-silenced cells presented a decline in both vimentin and cytokeratin expression, whereas the overexpression of miR-194-5p promoted an increase in cytokeratin and a reduction in vimentin, reflecting the acquisition of an epithelial phenotype. Both treatments reduced TNBC cells' migration. These results suggest that MEF2C may determine TNBC cells' invasive properties by partially determining the occurrence of epithelial-mesenchymal transition, while the overexpression of miR-194-5p promotes a decline in TNBC cells' aggressive behavior and reinforces this miRNA's role as a tumor suppressor in TNBC.
Collapse
Affiliation(s)
- Sara Caetano
- iMed—Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (S.C.); (A.R.G.); (I.F.)
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Ana Rita Garcia
- iMed—Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (S.C.); (A.R.G.); (I.F.)
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Inês Figueira
- iMed—Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (S.C.); (A.R.G.); (I.F.)
- Farm-ID—Faculty of Pharmacy Research and Development Association, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| | - Maria Alexandra Brito
- iMed—Research Institute for Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal; (S.C.); (A.R.G.); (I.F.)
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisbon, Portugal
| |
Collapse
|
27
|
Lyu H, Shen F, Ruan S, Tan C, Zhou J, Thor AD, Liu B. HER3 functions as an effective therapeutic target in triple negative breast cancer to potentiate the antitumor activity of gefitinib and paclitaxel. Cancer Cell Int 2023; 23:204. [PMID: 37716943 PMCID: PMC10504712 DOI: 10.1186/s12935-023-03055-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 09/03/2023] [Indexed: 09/18/2023] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) represents a significant clinical challenge. Chemotherapy remains the mainstay for a large part of TNBC patients, whereas drug resistance and tumor recurrence frequently occur. It is in urgent need to identify novel molecular targets for TNBC and develop effective therapy against the aggressive disease. METHODS Immunohistochemistry was performed to examine the expression of HER3 in TNBC samples. Western blots were used to assess protein expression and activation. Cell proliferation and viability were determined by cell growth (MTS) assays. TCGA databases were analyzed to correlate HER3 mRNA expression with the clinical outcomes of TNBC patients. Specific shRNA was used to knockdown HER3 expression. IncuCyte system was utilized to monitor cell growth and migration. LIVE/DEAD Cell Imaging was to detect live and dead cells. HER3 recognition by our anti-HER3 monoclonal antibody (mAb) 4A7 was verified by ELISA, flow cytometry, and co-immunoprecipitation assays. Orthotopic tumor models were established in nude mice to determine the capability of TNBC cells forming tumors and to test if our mAb 4A7 could potentiate the antitumor activity of paclitaxel in vivo. RESULTS Elevated expression of HER3 was observed in approximately half of the TNBC specimens and cell lines tested. Analyses of TCGA databases found that the TNBC patients with high HER3 mRNA expression in the tumors showed significantly worse overall survival (OS) and relapse-free survival (RFS) than those with low HER3 expression. Specific knockdown of HER3 markedly inhibited TNBC cell proliferation and mammosphere formation in vitro and tumor growth in vivo. Our mAb 4A7 abrogated heregulin (a ligand for HER3), but not SDF-1 (a ligand for CXCR4)-induced enhancement of TNBC cell migration. Combinations of 4A7 and the EGFR-tyrosine kinase inhibitor (TKI) gefitinib dramatically decreased the levels of phosphorylated HER3, EGFR, Akt, and ERK1/2 in TNBC cells and potently induced growth inhibition and cell death. Moreover, 4A7 in combination with paclitaxel exerted significant antitumor activity against TNBC in vitro and in vivo. CONCLUSIONS Our data demonstrate that increased HER3 is an effective therapeutic target for TNBC and our anti-HER3 mAb (4A7) may enhance the efficacy of gefitinib or paclitaxel in TNBC.
Collapse
Affiliation(s)
- Hui Lyu
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave, New Orleans, LA, 70112, USA
| | - Fei Shen
- Jiangsu Institute of Hematology, NHC Key Laboratory of Thrombosis and Hemostasis, National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Sanbao Ruan
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave, New Orleans, LA, 70112, USA
| | - Congcong Tan
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave, New Orleans, LA, 70112, USA
| | - Jundong Zhou
- Suzhou Cancer Center Core Laboratory, Nanjing Medical University Affiliated Suzhou Hospital, Suzhou, Jiangsu, China
| | - Ann D Thor
- Department of Pathology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Bolin Liu
- Departments of Interdisciplinary Oncology and Genetics, Stanley S. Scott Cancer Center, School of Medicine, Louisiana State University (LSU) Health Sciences Center, 1700 Tulane Ave, New Orleans, LA, 70112, USA.
| |
Collapse
|
28
|
Zhang W, He Y, Tang Y, Dai W, Si Y, Mao F, Xu J, Yu C, Sun X. A meta-analysis of application of PD-1/PD-L1 inhibitor-based immunotherapy in unresectable locally advanced triple-negative breast cancer. Immunotherapy 2023; 15:1073-1088. [PMID: 37337734 DOI: 10.2217/imt-2023-0023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/21/2023] Open
Abstract
Aims: The purpose of this study was to explore the efficacy of immunotherapy for patients with triple-negative breast cancer (TNBC). Materials & methods: Randomized clinical trials comparing immunotherapy with chemotherapy for advanced TNBC patients were included. Results: A total of six articles (3183 patients) were eligible for this meta-analysis. PD-1/PD-L1 inhibitor-based immunotherapy combined with chemotherapy can significantly increase the progression-free survival (hazard ratio [HR] = 0.82; 95% CI = 0.76-1.14; p < 0.001) of unresectable locally advanced or metastatic TNBC patients without effect on overall survival, compared with chemotherapy. Conclusion: PD-1/PD-L1 inhibitors-based immunotherapy can safely improve progression-free survival in patients with unresectable locally advanced or metastatic TNBC, but has no effect on overall survival.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Breast Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, 315010, China
| | - Yujing He
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yuning Tang
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Wei Dai
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Yuexiu Si
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Feiyan Mao
- Department of General Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, 315010, China
| | - Jiaxuan Xu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Chiyuan Yu
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, China
| | - Xing Sun
- Department of General Surgery, Ningbo No. 2 Hospital, Ningbo, Zhejiang, 315010, China
| |
Collapse
|
29
|
Liu Y, Dietl MC, Heckershoff R, Han C, Shi H, Rudolph M, Rominger F, Caligiuri I, Asif K, Adeel M, Scattolin T, Hashmi ASK. Gold-Catalyzed Formal [4+2] Cycloaddition as Access to Antitumor-Active Spirocyclic Oxindoles from Alkynes and Isatin-Derived Ketimines. Angew Chem Int Ed Engl 2023; 62:e202304672. [PMID: 37204285 DOI: 10.1002/anie.202304672] [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: 04/02/2023] [Revised: 05/17/2023] [Accepted: 05/17/2023] [Indexed: 05/20/2023]
Abstract
Due to its excellent bioactivity profile, which is increasingly utilized in pharmaceutical and synthetic chemistry, spirooxindole is an important core scaffold. We herein describe an efficient method for the construction of highly functionalized new spirooxindolocarbamates via a gold-catalyzed cycloaddition reaction of terminal alkynes or ynamides with isatin-derived ketimines. This protocol has a good functional group compatibility, uses readily available starting materials, mild reaction conditions, low catalyst loadings and no additives. It enables the transformation of various functionalized alkyne groups into cyclic carbamates. Gram-scale synthesis was achieved and DFT calculations verify the feasibility of the mechanistic proposal. Some of the target products exhibit good to excellent antiproliferative activity on human tumor cell lines. In addition, one of the most active compounds displayed a remarkable selectivity towards tumor cells over normal ones.
Collapse
Affiliation(s)
- Yaowen Liu
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Martin C Dietl
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Robin Heckershoff
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Chunyu Han
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Hongwei Shi
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Matthias Rudolph
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Frank Rominger
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
| | - Isabella Caligiuri
- Pathology Unit, Department of Molecular Biology and Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
| | - Kanwal Asif
- Pathology Unit, Department of Molecular Biology and Translational Research, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, via Franco Gallini 2, 33081, Aviano, Italy
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174, Venezia-Mestre, Italy
| | - Muhammad Adeel
- Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari, Campus Scientifico Via Torino 155, 30174, Venezia-Mestre, Italy
| | - Thomas Scattolin
- Dipartimento di Scienze Chimiche, Università degli studi di Padova, via Marzolo 1, 35131, Padova, Italy
| | - A Stephen K Hashmi
- Organisch-Chemisches Institut, Heidelberg University, Im Neuenheimer Feld 270, 69120, Heidelberg, Germany
- Chemistry Department, Faculty of Science, King Abdulaziz University (KAU), 21589, Jeddah, Saudi Arabia
| |
Collapse
|
30
|
Mondal SK, Haas D, Han J, Whiteside TL. Small EV in plasma of triple negative breast cancer patients induce intrinsic apoptosis in activated T cells. Commun Biol 2023; 6:815. [PMID: 37542121 PMCID: PMC10403597 DOI: 10.1038/s42003-023-05169-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 07/24/2023] [Indexed: 08/06/2023] Open
Abstract
Small extracellular vesicles (sEV) in TNBC patients' plasma promote T cell dysfunction and tumor progression. Here we show that tumor cell-derived exosomes (TEX) carrying surface PDL-1, PD-1, Fas, FasL, TRAIL, CTLA-4 and TGF-β1 induce apoptosis of CD8+T and CD4+T cells but spare B and NK cells. Inhibitors blocking TEX-induce receptor/ligand signals and TEX pretreatments with proteinase K or heat fail to prevent T cell apoptosis. Cytochalasin D, Dynosore or Pit Stop 2, partly inhibit TEX uptake but do not prevent T cell apoptosis. TEX entry into T cells induces cytochrome C and Smac release from mitochondria and caspase-3 and PARP cleavage in the cytosol. Expression of survival proteins is reduced in T cells undergoing apoptosis. Independently of external death receptor signaling, TEX entry into T cells induces mitochondrial stress, initiating relentless intrinsic apoptosis, which is responsible for death of activated T cells in the tumor-bearing hosts. The abundance of TEX in cancer plasma represents a danger for adoptively transferred T cells, limiting their therapeutic potential.
Collapse
Affiliation(s)
- Sujan Kumar Mondal
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Derick Haas
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Jie Han
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA
| | - Theresa L Whiteside
- Department of Pathology, University of Pittsburgh School of Medicine and UPMC Hillman Cancer Center, Pittsburgh, PA, 15213, USA.
- Departments of Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
31
|
Lejeune M, Reverté L, Gallardo N, Sauras E, Bosch R, Mata D, Roso A, Petit A, Peg V, Riu F, García-Fontgivell J, Relea F, Vieites B, de la Cruz-Merino L, Arenas M, Rodriguez V, Galera J, Korzynska A, Plancoulaine B, Álvaro T, López C. Matrix Metalloproteinase-9 Expression Is Associated with the Absence of Response to Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer Patients. Int J Mol Sci 2023; 24:11297. [PMID: 37511057 PMCID: PMC10378773 DOI: 10.3390/ijms241411297] [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: 06/20/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is particularly challenging due to the weak or absent response to therapeutics and its poor prognosis. The effectiveness of neoadjuvant chemotherapy (NAC) response is strongly influenced by changes in elements of the tumor microenvironment (TME). This work aimed to characterize the residual TME composition in 96 TNBC patients using immunohistochemistry and in situ hybridization techniques and evaluate its prognostic implications for partial responders vs. non-responders. Compared with non-responders, partial responders containing higher levels of CD83+ mature dendritic cells, FOXP3+ regulatory T cells, and IL-15 expression but lower CD138+ cell concentration exhibited better OS and RFS. However, along with tumor diameter and positive nodal status at diagnosis, matrix metalloproteinase-9 (MMP-9) expression in the residual TME was identified as an independent factor associated with the impaired response to NAC. This study yields new insights into the key components of the residual tumor bed, such as MMP-9, which is strictly associated with the lack of a pathological response to NAC. This knowledge might help early identification of TNBC patients less likely to respond to NAC and allow the establishment of new therapeutic targets.
Collapse
Affiliation(s)
- Marylène Lejeune
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Laia Reverté
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Noèlia Gallardo
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Esther Sauras
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
- Clinical Studies Unit, Hospital de Tortosa Verge de la Cinta, Carretera Esplanetes, 14, 43500 Tortosa, Spain
| | - Ramon Bosch
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Daniel Mata
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Albert Roso
- Institut Universitari d'Investigació en Atenció Primària Jordi Gol (IDIAP Jordi Gol), Gran Via Corts Catalanes, 587, 08007 Barcelona, Spain
| | - Anna Petit
- Pathology Department, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain
| | - Vicente Peg
- Pathology Department, Hospital Universitari de Vall Hebron, 08035 Barcelona, Spain
| | - Francisco Riu
- Pathology Department, Hospital Universitari Sant Joan de Reus, 43204 Reus, Spain
| | - Joan García-Fontgivell
- Pathology Department, Hospital Universitari Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
| | - Fernanda Relea
- Pathology Department, Hospital General de Ciudad Real, 13005 Ciudad Real, Spain
| | - Begoña Vieites
- Pathology Department, Hospital Universitario Virgen del Rocío, 41013 Seville, Spain
| | | | - Meritxell Arenas
- Department of Radiation Oncology, Hospital Universitari Sant Joan de Reus, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili, 43204 Tarragona, Spain
| | - Valeri Rodriguez
- Oncology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), 43500 Tortosa, Spain
| | - Juana Galera
- Gynaecology Department, Hospital Universitari Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), 43005 Tarragona, Spain
| | - Anna Korzynska
- Laboratory of Processing and Analysis of Microscopic Images, Nalecz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, 02-109 Warsaw, Poland
| | - Benoît Plancoulaine
- ANTICIPE, INSERM, François Baclesse Comprehensive Cancer Center, University Caen Normandy, 14000 Caen, France
| | - Tomás Álvaro
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| | - Carlos López
- Oncological Pathology and Bioinformatics Research Group, Molecular Biology and Research Section, Pathology Department, Hospital de Tortosa Verge de la Cinta, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Esplanetes, 14, 43500 Tortosa, Spain
| |
Collapse
|
32
|
Komoto TT, Nishimura FG, Evangelista AF, de Freitas AJA, da Silva G, Silva WA, Peronni K, Marques MMC, Marins M, Fachin AL. Exploring the Therapeutic Potential of trans-Chalcone: Modulation of MicroRNAs Linked to Breast Cancer Progression in MCF-7 Cells. Int J Mol Sci 2023; 24:10785. [PMID: 37445965 DOI: 10.3390/ijms241310785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/10/2023] [Accepted: 06/16/2023] [Indexed: 07/15/2023] Open
Abstract
Breast cancer is responsible for 25% of all cancers that affect women. Due to its high heterogeneity pattern in clinical diagnosis and its molecular profile differences, researchers have been seeking new targets and therapies, with more specificity and fewer side effects. Thus, one compound that has garnered our attention is trans-chalcone, which is naturally occurring in various plants and possesses promising biological properties, including antitumor effects. MiRNA is an extensive class of non-coding small, endogenous, and single-stranded RNAs, and it is involved in post-translational gene regulation. Therefore, the objective of this study was to investigate the effects of TChal on miRNAs expression and its relationship with anticancer activity against MCF-7. Initially, the trans-chalcone IC50 value was established by MTT assay for MCF-7and HaCat (non-cancer cell), in which we found out that it was 53.73 and 44.18 μM, respectively. Subsequently, we treated MCF-7 cells with trans-chalcone at its IC50 concentration and performed Mi-seq analysis, which unveiled 23 differentially expressed miRNAs. From this set, we selected five miRNAs (miR-25-5p, miR-27a-3p, miR-891a, miR-449a, and miR-4485) for further validation using qRT-PCR, guided by in silico analysis and their known association with tumorigenesis. In conclusion, our research provides valuable insights into the potential use of TChal to reveal MicroRNAs molecular targets that can be applied in breast cancer therapy.
Collapse
Affiliation(s)
- Tatiana Takahasi Komoto
- Biotechnology Unit, University of Ribeirão Preto, SP, Av. Costábile Romano, 2201, Ribeirão Preto 14096-900, Brazil
- Molecular Oncology Research Center, Barretos Cancer Hospital, Teaching and Research Institute, Barretos 14784-400, Brazil
| | - Felipe Garcia Nishimura
- Biotechnology Unit, University of Ribeirão Preto, SP, Av. Costábile Romano, 2201, Ribeirão Preto 14096-900, Brazil
| | - Adriane Feijó Evangelista
- Sergio Arouca National School of Public Health, Oswaldo Cruz Foundation, Manguinhos, Rio de Janeiro 21040-361, Brazil
| | - Ana Julia Aguiar de Freitas
- Molecular Oncology Research Center, Barretos Cancer Hospital, Teaching and Research Institute, Barretos 14784-400, Brazil
| | - Gabriel da Silva
- Biotechnology Unit, University of Ribeirão Preto, SP, Av. Costábile Romano, 2201, Ribeirão Preto 14096-900, Brazil
| | - Wilson Araujo Silva
- Center for Medical Genomics at the Clinics Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | - Kamila Peronni
- Center for Medical Genomics at the Clinics Hospital of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto 14049-900, Brazil
| | | | - Mozart Marins
- Biotechnology Unit, University of Ribeirão Preto, SP, Av. Costábile Romano, 2201, Ribeirão Preto 14096-900, Brazil
| | - Ana Lucia Fachin
- Biotechnology Unit, University of Ribeirão Preto, SP, Av. Costábile Romano, 2201, Ribeirão Preto 14096-900, Brazil
| |
Collapse
|
33
|
Obidiro O, Battogtokh G, Akala EO. Triple Negative Breast Cancer Treatment Options and Limitations: Future Outlook. Pharmaceutics 2023; 15:1796. [PMID: 37513983 PMCID: PMC10384267 DOI: 10.3390/pharmaceutics15071796] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Triple negative breast cancer (TNBC) has a negative expression of estrogen receptors (ER), progesterone receptors (PR), and human epidermal growth factor receptors (HER2). The survival rate for TNBC is generally worse than other breast cancer subtypes. TNBC treatment has made significant advances, but certain limitations remain. Treatment for TNBC can be challenging since the disease has various molecular subtypes. A variety of treatment options are available, such as chemotherapy, immunotherapy, radiotherapy, and surgery. Chemotherapy is the most common of these options. TNBC is generally treated with systemic chemotherapy using drugs such as anthracyclines and taxanes in neoadjuvant or adjuvant settings. Developing resistance to anticancer drugs and off-target toxicity are the primary hindrances to chemotherapeutic solutions for cancer. It is imperative that researchers, clinicians, and pharmaceutical companies work together to develop effective treatment options for TNBC. Several studies have suggested nanotechnology as a potential solution to the problem of suboptimal TNBC treatment. In this review, we summarized possible treatment options for TNBC, including chemotherapy, immunotherapy, targeted therapy, combination therapy, and nanoparticle-based therapy, and some solutions for the treatment of TNBC in the future. Moreover, we gave general information about TNBC in terms of its characteristics and aggressiveness.
Collapse
Affiliation(s)
| | | | - Emmanuel O. Akala
- Center for Drug Research and Development, Department of Pharmaceutical Sciences, College of Pharmacy, Howard University, Washington, DC 20059, USA; (O.O.); (G.B.)
| |
Collapse
|
34
|
Zare I, Taheri-Ledari R, Esmailzadeh F, Salehi MM, Mohammadi A, Maleki A, Mostafavi E. DNA hydrogels and nanogels for diagnostics, therapeutics, and theragnostics of various cancers. NANOSCALE 2023. [PMID: 37337663 DOI: 10.1039/d3nr00425b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
As an efficient class of hydrogel-based therapeutic drug delivery systems, deoxyribonucleic acid (DNA) hydrogels (particularly DNA nanogels) have attracted massive attention in the last five years. The main contributor to this is the programmability of these 3-dimensional (3D) scaffolds that creates fundamental effects, especially in treating cancer diseases. Like other active biological ingredients (ABIs), DNA hydrogels can be functionalized with other active agents that play a role in targeting drug delivery and modifying the half-life of the therapeutic cargoes in the body's internal environment. Considering the brilliant advantages of DNA hydrogels, in this survey, we intend to submit an informative collection of feasible methods for the design and preparation of DNA hydrogels and nanogels, and the responsivity of the immune system to these therapeutic cargoes. Moreover, the interactions of DNA hydrogels with cancer biomarkers are discussed in this account. Theragnostic DNA nanogels as an advanced species for both detection and therapeutic purposes are also briefly reviewed.
Collapse
Affiliation(s)
- Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz 7178795844, Iran
| | - Reza Taheri-Ledari
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Farhad Esmailzadeh
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Mohammad Mehdi Salehi
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Adibeh Mohammadi
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Ali Maleki
- Catalysts and Organic Synthesis Research Laboratory, Department of Chemistry, Iran University of Science and Technology, Tehran, 16846-13114, Iran
| | - Ebrahim Mostafavi
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
35
|
Wu L, Bai S, Huang J, Cui G, Li Q, Wang J, Du X, Fu W, Li C, Wei W, Lin H, Luo ML. Nigericin Boosts Anti-Tumor Immune Response via Inducing Pyroptosis in Triple-Negative Breast Cancer. Cancers (Basel) 2023; 15:3221. [PMID: 37370831 DOI: 10.3390/cancers15123221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/04/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Although immune checkpoint inhibitors improved the clinical outcomes of advanced triple negative breast cancer (TBNC) patients, the response rate remains relatively low. Nigericin is an antibiotic derived from Streptomyces hydrophobicus. We found that nigericin caused cell death in TNBC cell lines MDA-MB-231 and 4T1 by inducing concurrent pyroptosis and apoptosis. As nigericin facilitated cellular potassium efflux, we discovered that it caused mitochondrial dysfunction, leading to mitochondrial ROS production, as well as activation of Caspase-1/GSDMD-mediated pyroptosis and Caspase-3-mediated apoptosis in TNBC cells. Notably, nigericin-induced pyroptosis could amplify the anti-tumor immune response by enhancing the infiltration and anti-tumor effect of CD4+ and CD8+ T cells. Moreover, nigericin showed a synergistic therapeutic effect when combined with anti-PD-1 antibody in TNBC treatment. Our study reveals that nigericin may be a promising anti-tumor agent, especially in combination with immune checkpoint inhibitors for advanced TNBC treatment.
Collapse
Affiliation(s)
- Lisha Wu
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Shoumin Bai
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jing Huang
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Guohui Cui
- South China National Bio-Safety Laboratory, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510600, China
| | - Qingjian Li
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Jingshu Wang
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Xin Du
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Wenkui Fu
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Chuping Li
- Department of Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Wei Wei
- Department of Breast and Thyroid Surgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Huan Lin
- Department of Breast Oncology, The Second Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou 510120, China
| | - Man-Li Luo
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| |
Collapse
|
36
|
Bakhsh MR, Rouhi L, Ghaedi K, Hashemi M, Peymani M, Samarghandian S. Therapeutic effects of guanidine hydrochloride on breast cancer through targeting KCNG1 gene. Biomed Pharmacother 2023; 164:114982. [PMID: 37311278 DOI: 10.1016/j.biopha.2023.114982] [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: 03/24/2023] [Revised: 05/28/2023] [Accepted: 06/01/2023] [Indexed: 06/15/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is one of the subtypes of breast cancer (BC) that is associated with poor survival rates and failure to respond to hormonal and targeted therapies. OBJECTIVE The aim of this study was to identify a specific gene at the expression level for TNBC and targeting of this type of breast cancer based on it. Using TCGA database, genes that are particularly high expression in TNBC subtypes compared to other BC subtypes (in terms of receptor status) and normal samples were identified and their sensitivity and specificity were evaluated. Using PharmacoGX and Drug Bank data, drug sensitivity and drug-appropriate genes were identified, respectively. The effects of the identified drug on triple-negative cell lines (MDA-MB-468) were evaluated in comparison with the cell line of other subtypes (MCF7) by apoptosis and MTS tests. RESULTS Data analyzes showed that the expression level of KCNG1 gene in the TNBC subgroup was significantly higher compared to other BC subtypes from the KCN gene family and ROC results showed that this gene had highest sensitivity and specificity in TNBC subtype. The results of drug resistance and sensitivity showed that an increase in the expression level of KCNG1 was associated with sensitivity to Cisplatin and Oxaliplatin. Moreover, Drug Bank results showed that Guanidine hydrochloride (GuHCl) was a suitable inhibitor for KCNG1. In vitro results showed that the expression level of KCNG1 was higher in MDA-MB-468 compared to MCF7. In addition, the rate of apoptosis in response to GuHCl treatment in MDA-MB-468 cell line as TNBC cell model was higher than MCF7 in the same concentration. CONCLUSION This study revealed that GuHCl could be a suitable treatment for TNBC subtype by targeting of KCNG1.
Collapse
Affiliation(s)
- Mehdi Roshanian Bakhsh
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, the Islamic Republic of Iran
| | - Leila Rouhi
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, the Islamic Republic of Iran
| | - Kamran Ghaedi
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, the Islamic Republic of Iran; Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, the Islamic Republic of Iran
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical sciences, Islamic Azad University, Tehran, the Islamic Republic of Iran.
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, the Islamic Republic of Iran.
| | - Saeed Samarghandian
- Healthy Ageing Research Centre, Neyshabur University of Medical Sciences, Neyshabur, the Islamic Republic of Iran.
| |
Collapse
|
37
|
Dong LF, Chen FF, Fan YF, Zhang K, Chen HH. circ-0000512 inhibits PD-L1 ubiquitination through sponging miR-622/CMTM6 axis to promote triple-negative breast cancer and immune escape. J Immunother Cancer 2023; 11:e005461. [PMID: 37349124 PMCID: PMC10314703 DOI: 10.1136/jitc-2022-005461] [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] [Accepted: 05/31/2023] [Indexed: 06/24/2023] Open
Abstract
BACKGROUND This study reported the function and mechanism of circ-0000512 in the progression of triple-negative breast cancer (TNBC). METHODS circ-0000512 expression in TNBC tissues and paired adjacent normal tissues and cells was examined by qRT-PCR. Moreover, circ-0000512 expression in TNBC cells was modulated by transfection. Thereafter, colony formation assay, Transwell assay and flow cytometry were conducted to observe cell proliferation, migration and apoptosis. TNBC cells were treated with cycloheximide and the protease inhibitor MG132. Later, ubiquitination assay was performed to detect programmed cell death ligand 1 (PD-L1) ubiquitination in TNBC cells. The T cell killing ability was assessed by the T cell-mediated tumor cell killing assay. IFNγ and IL-2 levels were detected by ELISA. The percentage of activated T cells was detected with a flow cytometer. In addition, dual luciferase reporter gene assay and RNA immunoprecipitation assay were carried out to evaluate the binding between two genes. In vivo study was conducted on mice. CD8+ T cells in xenograft tumors were detected by immunohistochemistry. RESULTS circ-0000512 was upregulated in patients with TNBC. circ-0000512 knockdown attenuated the proliferation and migration of TNBC cells and enhanced their apoptosis. circ-0000512 overexpression had opposite effects. circ-0000512 knockdown enhanced the PD-L1 protein ubiquitination in TNBC cells by inhibiting CMTM6. Meanwhile, circ-0000512 promoted CMTM6 expression by sponging miR-622. circ-0000512 knockdown increased the ratio of CD8+T cells and the lethality of T cells against TNBC cells. Besides, circ-0000512 knockdown inhibited the growth of TNBC cells in immunodeficient nude mice and normal immune mice and increased the ratio of CD8+T cells in xenograft tumors of normal immune mice. CONCLUSIONS circ-0000512 inhibited PD-L1 ubiquitination by sponging the miR-622/CMTM6 axis, thus promoting TNBC progression and immune escape.
Collapse
Affiliation(s)
- Li-Feng Dong
- Department of Breast Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Fang-Fang Chen
- Department of Breast Surgery, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Yang-Fan Fan
- Department of Breast Surgery, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Kun Zhang
- Department of Breast Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| | - Hui-Hui Chen
- Department of Breast Surgery and Oncology, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
38
|
Hu J, Dai C, Zhang Y, Chen W, Sun L, Zhang X, Duan M, Fu H, Long T, Kang W, Yin C, Liu X, Yu J. Effect of surgical treatment on patients with stage T3 or T4 triple-negative breast cancer: a SEER-based retrospective observational study. Front Endocrinol (Lausanne) 2023; 14:1184173. [PMID: 37305041 PMCID: PMC10250696 DOI: 10.3389/fendo.2023.1184173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 05/12/2023] [Indexed: 06/13/2023] Open
Abstract
Background The use of surgery is controversial in patients with stage T3 or T4 triple-negative breast cancer (TNBC). We aimed to explore the effect of surgical treatment on overall survival (OS) of these patients. Methods A total of 2,041 patients were selected and divided into the surgical and non-surgical groups based on the Surveillance, Epidemiology, and End Results database from 2010 to 2018. Propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) were applied to balance covariates between different groups. The OS of the two groups were assessed by Kaplan-Meier survival curves and Cox proportional hazards regression models. Results A total of 2,041 patients were included in the study. After PSM and IPTW, baseline characteristics of the matched variables were fully balanced. Kaplan-Meier survival curves showed that the median survival time and OS of TNBC patients with stage T3 or T4 in the surgical group were significantly improved compared with those in the non-surgical group. Multivariate Cox proportional hazards regression analysis showed that surgery was a protective factor for prognosis. Conclusion Our study found that surgery prolonged the median survival and improved OS compared with the non-surgical group of TNBC patients with stage T3 or T4.
Collapse
Affiliation(s)
- Jie Hu
- Department of Tumor Radiotherapy, Taizhou Central Hospital (Affiliated Hospital of Taizhou University), Taizhou, China
| | - Changling Dai
- Department of Hematology, Taizhou Central Hospital (Affiliated Hospital of Taizhou University), Taizhou, China
| | - Yang Zhang
- College of Medical Informatics, Chongqing Medical University, Chongqing, China
| | - Weijun Chen
- Department of Tumor Radiotherapy, Taizhou Central Hospital (Affiliated Hospital of Taizhou University), Taizhou, China
| | - Lihua Sun
- Department of Tumor Radiotherapy, Taizhou Central Hospital (Affiliated Hospital of Taizhou University), Taizhou, China
| | - Xu Zhang
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Hangzhou, China
| | - Minjie Duan
- College of Medical Informatics, Chongqing Medical University, Chongqing, China
| | - Hao Fu
- Department of Infectious Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Teng Long
- Department of Infectious Disease, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Kang
- Department of Mathematics, Physics and Interdisciplinary Studies, Guangzhou Laboratory, Guangzhou, China
| | - Chengliang Yin
- Faculty of Medicine, Macau University of Science and Technology, Macau, China
| | - Xiaozhu Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jie Yu
- Department of Medical Imaging, The Affiliated Taian City Central Hospital of Qingdao University, Taian, China
| |
Collapse
|
39
|
Chaudhuri A, Kumar DN, Dehari D, Patil R, Singh S, Kumar D, Agrawal AK. Endorsement of TNBC Biomarkers in Precision Therapy by Nanotechnology. Cancers (Basel) 2023; 15:cancers15092661. [PMID: 37174125 PMCID: PMC10177107 DOI: 10.3390/cancers15092661] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/15/2023] Open
Abstract
Breast cancer is a heterogeneous disease which accounts globally for approximately 1 million new cases annually, wherein more than 200,000 of these cases turn out to be cases of triple-negative breast cancer (TNBC). TNBC is an aggressive and rare breast cancer subtype that accounts for 10-15% of all breast cancer cases. Chemotherapy remains the only therapy regimen against TNBC. However, the emergence of innate or acquired chemoresistance has hindered the chemotherapy used to treat TNBC. The data obtained from molecular technologies have recognized TNBC with various gene profiling and mutation settings that have helped establish and develop targeted therapies. New therapeutic strategies based on the targeted delivery of therapeutics have relied on the application of biomarkers derived from the molecular profiling of TNBC patients. Several biomarkers have been found that are targets for the precision therapy in TNBC, such as EGFR, VGFR, TP53, interleukins, insulin-like growth factor binding proteins, c-MET, androgen receptor, BRCA1, glucocorticoid, PTEN, ALDH1, etc. This review discusses the various candidate biomarkers identified in the treatment of TNBC along with the evidence supporting their use. It was established that nanoparticles had been considered a multifunctional system for delivering therapeutics to target sites with increased precision. Here, we also discuss the role of biomarkers in nanotechnology translation in TNBC therapy and management.
Collapse
Affiliation(s)
- Aiswarya Chaudhuri
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Dulla Naveen Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Deepa Dehari
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Rohit Patil
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Sanjay Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India
- Department of Pharmaceutics, Babasaheb Bhimrao Ambedkar University (A Central University), Vidya Vihar, Raebareli Road, Lucknow 226025, India
| | - Dinesh Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India
| | - Ashish Kumar Agrawal
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology (BHU), Varanasi 221005, India
| |
Collapse
|
40
|
Daniluk K, Lange A, Wójcik B, Zawadzka K, Bałaban J, Kutwin M, Jaworski S. Effect of Melittin Complexes with Graphene and Graphene Oxide on Triple-Negative Breast Cancer Tumors Grown on Chicken Embryo Chorioallantoic Membrane. Int J Mol Sci 2023; 24:ijms24098388. [PMID: 37176095 PMCID: PMC10179033 DOI: 10.3390/ijms24098388] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
One of the components of bee venom is melittin (M), which has strong lysing properties on membranes. M has high toxicity to cancer cells, but it also affects healthy cells, making it necessary to use methods for targeted delivery to ensure treatment. This research is a continuation of previous studies using graphene nanomaterials as M carriers to breast cancer cells. The studies described below are conducted on a more organized biological structure than what is found in vitro cells, namely, cancerous tumors grown on a chicken embryo chorioallantoic membrane. Caspase 3 and 8 levels are analyzed, and the level of oxidative stress markers and changes in protein expression for cytokines are examined. The results show that M complexes with nanomaterials reduce the level of oxidative stress more than M alone does, but the use of graphene (GN) as a carrier increases the level of DNA damage to a greater extent than the increase caused by M alone. An analysis of cytokine levels shows that the use of the M and GN complex increases the level of proteins responsible for inhibiting tumor progression to a greater extent than the increase occasioned by a complex with graphene oxide (GO). The results suggest that the use of GN as an M carrier may increase the toxic effect of M on structures located inside a cell.
Collapse
Affiliation(s)
- Karolina Daniluk
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Agata Lange
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Barbara Wójcik
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Katarzyna Zawadzka
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Jaśmina Bałaban
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Marta Kutwin
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| | - Sławomir Jaworski
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, 02-786 Warsaw, Poland
| |
Collapse
|
41
|
da Costa NS, Lima LS, Oliveira FAM, Galiciolli MEA, Manzano MI, Garlet QI, Irioda AC, Oliveira CS. Antiproliferative Effect of Inorganic and Organic Selenium Compounds in Breast Cell Lines. Biomedicines 2023; 11:biomedicines11051346. [PMID: 37239017 DOI: 10.3390/biomedicines11051346] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive, fast-growing tumor that is more likely to spread to distant organs. Among women diagnosed with breast cancer, the prevalence of TNBC is 20%, and treatment is currently limited to chemotherapy. Selenium (Se), an essential micronutrient, has been explored as an antiproliferative agent. Therefore, this study aimed to evaluate the effects of exposure to organic (selenomethionine, ebselen, and diphenyl diselenide) and inorganic (sodium selenate and sodium selenite) Se molecules in different breast cell lines. The compounds were tested at 1, 10, 50, and 100 μM for 48 h in the non-tumor breast cell line (MCF-10A) and TNBC derivatives cell lines (BT-549 and MDA-MB-231). The effects of Se on cell viability, apoptotic and necrotic processes, colony formation, and cell migration were analyzed. Exposure to selenomethionine and selenate did not alter the evaluated parameters. However, selenomethionine had the highest selectivity index (SI). The exposure to the highest doses of selenite, ebselen, and diphenyl diselenide resulted in antiproliferative and antimetastatic effects. Selenite had a high SI to the BT cell line; however, the SI of ebselen and diphenyl diselenide was low in both tumoral cell lines. In conclusion, the Se compounds had different effects on the breast cell lines, and additional tests are needed to reveal the antiproliferative effects of Se compounds.
Collapse
Affiliation(s)
- Nayara Souza da Costa
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, Brazil
- Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Luíza Siqueira Lima
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, Brazil
- Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | | | | | - Mariana Inocêncio Manzano
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, Brazil
- Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Quelen Iane Garlet
- Curso de Medicina, Universidade Católica de Pelotas, Pelotas 96010-280, Brazil
| | - Ana Carolina Irioda
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, Brazil
- Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| | - Cláudia Sirlene Oliveira
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba 80250-060, Brazil
- Faculdades Pequeno Príncipe, Curitiba 80230-020, Brazil
| |
Collapse
|
42
|
Bi J, Wu Z, Zhang X, Zeng T, Dai W, Qiu N, Xu M, Qiao Y, Ke L, Zhao J, Cao X, Lin Q, Chen XL, Xie L, Ouyang Z, Guo J, Zheng L, Ma C, Guo S, Chen K, Mo W, Fu G, Zhao TJ, Wang HR. TMEM25 inhibits monomeric EGFR-mediated STAT3 activation in basal state to suppress triple-negative breast cancer progression. Nat Commun 2023; 14:2342. [PMID: 37095176 PMCID: PMC10126118 DOI: 10.1038/s41467-023-38115-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/17/2023] [Indexed: 04/26/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with poor outcome and lacks of approved targeted therapy. Overexpression of epidermal growth factor receptor (EGFR) is found in more than 50% TNBC and is suggested as a driving force in progression of TNBC; however, targeting EGFR using antibodies to prevent its dimerization and activation shows no significant benefits for TNBC patients. Here we report that EGFR monomer may activate signal transducer activator of transcription-3 (STAT3) in the absence of transmembrane protein TMEM25, whose expression is frequently decreased in human TNBC. Deficiency of TMEM25 allows EGFR monomer to phosphorylate STAT3 independent of ligand binding, and thus enhances basal STAT3 activation to promote TNBC progression in female mice. Moreover, supplying TMEM25 by adeno-associated virus strongly suppresses STAT3 activation and TNBC progression. Hence, our study reveals a role of monomeric-EGFR/STAT3 signaling pathway in TNBC progression and points out a potential targeted therapy for TNBC.
Collapse
Affiliation(s)
- Jing Bi
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Zhihui Wu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Xin Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
- State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, 100850, Beijing, China
| | - Taoling Zeng
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Wanjun Dai
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Ningyuan Qiu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Mingfeng Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Yikai Qiao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Lang Ke
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Jiayi Zhao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Xinyu Cao
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Qi Lin
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Xiao Lei Chen
- Cancer Research Center of Xiamen University, 361102, Xiamen, Fujian, China
- School of Medicine, Xiamen University, 361102, Fujian, China
| | - Liping Xie
- School of Medicine, Xiamen University, 361102, Fujian, China
| | - Zhong Ouyang
- Department of Breast Surgery, The First Affiliated Hospital of Xiamen University, 361003, Xiamen, Fujian, China
| | - Jujiang Guo
- Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, 361003, Xiamen, Fujian, China
| | - Liangkai Zheng
- Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, 361003, Xiamen, Fujian, China
| | - Chao Ma
- Medical School of Chinese PLA, 100853, Beijing, China
| | - Shiying Guo
- GemPharmatech Co., Ltd., 210000, Nanjing, Jiangsu, China
| | - Kangmei Chen
- Department of Clinical Laboratory, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510120, Guangzhou, China
| | - Wei Mo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China
| | - Guo Fu
- Cancer Research Center of Xiamen University, 361102, Xiamen, Fujian, China.
- School of Medicine, Xiamen University, 361102, Fujian, China.
- Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, 361003, Xiamen, Fujian, China.
| | - Tong-Jin Zhao
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Zhongshan Hospital, Fudan University, 200438, Shanghai, China.
| | - Hong-Rui Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, 361102, Fujian, China.
- Department of Obstetrics and Gynecology, Women and Children's Hospital, School of Medicine, Xiamen University, 361003, Xiamen, Fujian, China.
| |
Collapse
|
43
|
Chen X, Balko JM, Ling F, Jin Y, Gonzalez A, Zhao Z, Chen J. Convolutional neural network for biomarker discovery for triple negative breast cancer with RNA sequencing data. Heliyon 2023; 9:e14819. [PMID: 37025902 PMCID: PMC10070674 DOI: 10.1016/j.heliyon.2023.e14819] [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: 09/26/2022] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023] Open
Abstract
Triple negative breast cancers (TNBCs) are tumors with a poor treatment response and prognosis. In this study, we propose a new approach, candidate extraction from convolutional neural network (CNN) elements (CECE), for discovery of biomarkers for TNBCs. We used the GSE96058 and GSE81538 datasets to build a CNN model to classify TNBCs and non-TNBCs and used the model to make TNBC predictions for two additional datasets, the cancer genome atlas (TCGA) breast cancer RNA sequencing data and the data from Fudan University Shanghai Cancer Center (FUSCC). Using correctly predicted TNBCs from the GSE96058 and TCGA datasets, we calculated saliency maps for these subjects and extracted the genes that the CNN model used to separate TNBCs from non-TNBCs. Among the TNBC signature patterns that the CNN models learned from the training data, we found a set of 21 genes that can classify TNBCs into two major classes, or CECE subtypes, with distinct overall survival rates (P = 0.0074). We replicated this subtype classification in the FUSCC dataset using the same 21 genes, and the two subtypes had similar differential overall survival rates (P = 0.0490). When all TNBCs were combined from the 3 datasets, the CECE II subtype had a hazard ratio of 1.94 (95% CI, 1.25-3.01; P = 0.0032). The results demonstrate that the spatial patterns learned by the CNN models can be utilized to discover interacting biomarkers otherwise unlikely to be identified by traditional approaches.
Collapse
Affiliation(s)
| | - Justin M. Balko
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2101 W End Ave, Nashville, TN, 37240, USA
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 2101, W End Ave, Nashville, TN, 37240, USA
- Departments of Pathology, Microbiology, and Immunology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fei Ling
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, Guangdong, China
| | - Yabin Jin
- Clinical Research Institute, The First People’s Hospital of Foshan, Foshan, China
| | - Anneliese Gonzalez
- Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, TX77030, USA
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas, Houston, TX, 77030, USA
| | - Jingchun Chen
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, Las Vegas, NV, 89154, USA
| |
Collapse
|
44
|
Amide proton transfer (APT) imaging of breast cancers and its correlation with biological status. Clin Imaging 2023; 96:38-43. [PMID: 36773531 DOI: 10.1016/j.clinimag.2023.02.002] [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: 10/13/2022] [Revised: 01/12/2023] [Accepted: 02/02/2023] [Indexed: 02/11/2023]
Abstract
PURPOSE To assess the usefulness of amide proton transfer (APT) imaging to predict the biological status of breast cancers. METHOD Sixty-six patients (age range 31-85 years, mean 58.9 years) with histopathologically proven invasive ductal carcinomas of 2 cm or larger in diameter were included in this study. 3D APT weighted imaging was conducted on a 3 T scanner. Mean APT signal intensity (SI) was analyzed in relation to biological subtypes, Ki-67 labeling index, and nuclear grades (NGs). RESULTS The triple-negative (TN) cancers (n = 10; 2.75 ± 0.42%) showed significantly higher APT SI than the luminal type cancers (n = 48; 1.74 ± 0.83) and HER2 cancers (n = 8; 1.83 ± 0.21) (P = 0.0007, 0.03). APT SI had weakly positive correlation with the Ki-67 labeling index (r = 0.38, P = 0.002). The mean APT SIs were significantly higher for high-Ki-67 (>30%) (n = 31; 2.25 ± 0.70) than low-Ki-67 (≤30%) cancers (n = 35; 1.60 ± 0.79) (P = 0.0007). There was no significant difference in the APT SIs between NG 1-2 (n = 31; 1.71 ± 0.84) and NG 3 (n = 35; 2.08 ± 0.76%) cancers (P = 0.06). CONCLUSIONS TN and high-Ki-67 breast cancers showed high APT SIs. APT imaging can help to predict the biological status of breast cancers.
Collapse
|
45
|
Lau A, Le N, Nguyen C, Kandpal RP. Signals transduced by Eph receptors and ephrin ligands converge on MAP kinase and AKT pathways in human cancers. Cell Signal 2023; 104:110579. [PMID: 36572189 DOI: 10.1016/j.cellsig.2022.110579] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/24/2022]
Abstract
Eph receptors, the largest known family of receptor tyrosine kinases, and ephrin ligands have been implicated in a variety of human cancers. The novel bidirectional signaling events initiated by binding of Eph receptors to their cognate ephrin ligands modulate many cellular processes such as proliferation, metastasis, angiogenesis, invasion, and apoptosis. The relationships between the abundance of a unique subset of Eph receptors and ephrin ligands with associated cellular processes indicate a key role of these molecules in tumorigenesis. The combinatorial expression of these molecules converges on MAP kinase and/or AKT/mTOR signaling pathways. The intracellular target proteins of the initial signal may, however, vary in some cancers. Furthermore, we have also described the commonality of up- and down-regulation of individual receptors and ligands in various cancers. The current state of research in Eph receptors illustrates MAP kinase and mTOR pathways as plausible targets for therapeutic interventions in various cancers.
Collapse
Affiliation(s)
- Andreas Lau
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Nghia Le
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Claudia Nguyen
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America
| | - Raj P Kandpal
- Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA 91766, United States of America.
| |
Collapse
|
46
|
Zhu X, Bu J, Zhu T, Jiang Y. Targeting KK-LC-1 inhibits malignant biological behaviors of triple-negative breast cancer. J Transl Med 2023; 21:184. [PMID: 36895039 PMCID: PMC9996895 DOI: 10.1186/s12967-023-04030-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 03/01/2023] [Indexed: 03/11/2023] Open
Abstract
BACKGROUND Cancer/testis antigens (CTAs) participate in the regulation of malignant biological behaviors in breast cancer. However, the function and mechanism of KK-LC-1, a member of the CTA family, in breast cancer are still unclear. METHODS Bioinformatic tools, immunohistochemistry, and western blotting were utilized to detect the expression of KK-LC-1 in breast cancer and to explore the prognostic effect of KK-LC-1 expression in breast cancer patients. Cell function assays, animal assays, and next-generation sequencing were utilized to explore the function and mechanism of KK-LC-1 in the malignant biological behaviors of triple-negative breast cancer. Small molecular compounds targeting KK-LC-1 were also screened and drug susceptibility testing was performed. RESULTS KK-LC-1 was significantly highly expressed in triple-negative breast cancer tissues than in normal breast tissues. KK-LC-1 high expression was related to poor survival outcomes in patients with breast cancer. In vitro studies suggested that KK-LC-1 silencing can inhibit triple-negative breast cancer cell proliferation, invasion, migration, and scratch healing ability, increase cell apoptosis ratio, and arrest the cell cycle in the G0-G1 phase. In vivo studies have suggested that KK-LC-1 silencing decreases tumor weight and volume in nude mice. Results showed that KK-CL-1 can regulate the malignant biological behaviors of triple-negative breast cancer via the MAL2/MUC1-C/PI3K/AKT/mTOR pathway. The small-molecule compound Z839878730 had excellent KK-LC-1 targeting ability and cancer cell killing ability. The EC50 value was 9.7 μM for MDA-MB-231 cells and 13.67 µM for MDA-MB-468 cells. Besides, Z839878730 has little tumor-killing effect on human normal mammary epithelial cells MCF10A and can inhibit the malignant biological behaviors of triple-negative breast cancer cells by MAL2/MUC1-C/PI3K/AKT/mTOR pathway. CONCLUSIONS Our findings suggest that KK-LC-1 may serve as a novel therapeutic target for triple-negative breast cancer. Z839878730, which targets KK-LC-1, presents a new path for breast cancer clinical treatment.
Collapse
Affiliation(s)
- Xudong Zhu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, People's Republic of China.
- Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, Liaoning, People's Republic of China.
| | - Jiawen Bu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, People's Republic of China
| | - Tong Zhu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, People's Republic of China
| | - Yi Jiang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, People's Republic of China
| |
Collapse
|
47
|
Zhang H, Chen L, Zhao Y, Luo N, Shi J, Xu S, Ma L, Wang M, Gu M, Mu C, Xiong Y. Relaxin-encapsulated polymeric metformin nanoparticles remodel tumor immune microenvironment by reducing CAFs for efficient triple-negative breast cancer immunotherapy. Asian J Pharm Sci 2023; 18:100796. [PMID: 37008735 PMCID: PMC10064789 DOI: 10.1016/j.ajps.2023.100796] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/14/2023] [Accepted: 02/09/2023] [Indexed: 02/26/2023] Open
Abstract
Cancer-associated fibroblasts (CAFs) are one of the most abundant stromal cells in the tumor microenvironment which mediate desmoplastic response and are the primary driver for an immunosuppressive microenvironment, leading to the failure of triple-negative breast cancer (TNBC) immunotherapy. Therefore, depleting CAFs may enhance the effect of immunotherapy (such as PD-L1 antibody). Relaxin (RLN) has been demonstrated to significantly improve transforming growth factor-β (TGF-β) induced CAFs activation and tumor immunosuppressive microenvironment. However, the short half-life and systemic vasodilation of RLN limit its in vivo efficacy. Here, plasmid encoding relaxin (pRLN) to locally express RLN was delivered with a new positively charged polymer named polymeric metformin (PolyMet), which could increase gene transfer efficiency significantly and have low toxicity that have been certified by our lab before. In order to improve the stability of pRLN in vivo, this complex was further formed lipid poly-γ-glutamic acid (PGA)/PolyMet-pRLN nanoparticle (LPPR). The particle size of LPPR was 205.5 ± 2.9 nm, and the zeta potential was +55.4 ± 1.6 mV. LPPR displayed excellent tumor penetrating efficacy and weaken proliferation of CAFs in 4T1luc/CAFs tumor spheres in vitro. In vivo, it could reverse aberrantly activated CAFs by decreasing the expression of profibrogenic cytokine and remove the physical barrier to reshape the tumor stromal microenvironment, which enabled a 2.2-fold increase in cytotoxic T cell infiltration within the tumor and a decrease in immunosuppressive cells infiltration. Thus, LPPR was observed retarded tumor growth by itself in the 4T1 tumor bearing-mouse, and the reshaped immune microenvironment further led to facilitate antitumor effect when it combined with PD-L1 antibody (aPD-L1). Altogether, this study presented a novel therapeutic approach against tumor stroma using LPPR to achieve a combination regimen with immune checkpoint blockade therapy against the desmoplastic TNBC model.
Collapse
Affiliation(s)
- Hongyan Zhang
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Liying Chen
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yue Zhao
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Ningchao Luo
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Jingbin Shi
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shujun Xu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Lisha Ma
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Menglin Wang
- Division of Pharmacoengineering and Molecular Pharmaceutics, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, NC 27599, United States
| | - Mancang Gu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Chaofeng Mu
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Yang Xiong
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Academy of Chinese Medical Science, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Corresponding author at: School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China.
| |
Collapse
|
48
|
Lopes AD, Galdino NAL, Figueiredo AB, Brianese RC, Morais KLP, De Brot M, Osório CABT, Teixeira-Carvalho A, Calsavara VF, Evangelista GFB, Alves NS, Makdissi FB, Sanches SM, Cordeiro de Lima VC, Carraro DM, Gollob KJ. Systemic immune mediators reflect tumour-infiltrating lymphocyte intensity and predict therapeutic response in triple-negative breast cancer. Immunology 2023; 169:229-241. [PMID: 36703241 DOI: 10.1111/imm.13627] [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: 01/29/2022] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive form of breast cancer (BC). Neoadjuvant chemotherapy has proven efficacy in its treatment, and a pathological complete response (pCR) to therapy is predictive of improved long-term survival. The immune response is key to successful neoadjuvant chemotherapy, as indicated by the relation between the percentage of stromal tumour-infiltrating lymphocytes (TILs) in pre-treated tumour tissue samples and the likelihood of achieving pCR. Here we studied systemic immune mediators from volunteer TNBC patients before undergoing neoadjuvant chemotherapy to determine the systemic response association with TIL intensity, treatment response and survival. Patients were classified into pCR responder or non-responder at time of surgery. We found higher levels of immune mediators before treatment began in patients that went on to be pCR responders versus non-pCR, with area under the curve (AUC) values of 0.64-0.80. We also observed a positive correlation between inflammatory systemic immune mediators and the percentage of TILs in pCR responder patients. Combining TILs and systemic immune mediator levels provided stronger AUC values (range of 0.72-0.82). Last, performing a progression-free survival analysis with several of the systemic cytokines that predict pCR, segregated the patients into long- and short-survival groups based on high and low production of the cytokines, respectively. Our study demonstrates that circulating cytokines, before treatment begins, predict pCR in TNBC patients treated with neoadjuvant chemotherapy. Moreover, they may act as a surrogate marker of high TILs or together with TILs to better predict pCR and survival.
Collapse
Affiliation(s)
- Ananda D Lopes
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Nayane A L Galdino
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Amanda B Figueiredo
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Rafael C Brianese
- Laboratory of Genomics and Molecular Biology, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Katia L P Morais
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Marina De Brot
- Department of Pathology, A.C. Camargo Cancer Center, São Paulo, Brazil
| | | | | | - Vinicius F Calsavara
- Laboratory of Epidemiology and Statistics, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
| | - Guilherme F B Evangelista
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,Translational Immuno-oncology Laboratory, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Natalia S Alves
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil
| | | | - Solange M Sanches
- Department of Mastology, A.C.Camargo Cancer Center, São Paulo, Brazil
| | | | - Dirce M Carraro
- Laboratory of Genomics and Molecular Biology, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,INCT-INCITO, São Paulo, Brazil
| | - Kenneth J Gollob
- Translational Immuno-oncology Group, International Research Center, A.C. Camargo Cancer Center, São Paulo, Brazil.,INCT-INCITO, São Paulo, Brazil.,Center for Research in Immuno-oncology, Hospital Israelita Albert Einstein, São Paulo, Brazil
| |
Collapse
|
49
|
Petrosyan V, Dobrolecki LE, Thistlethwaite L, Lewis AN, Sallas C, Srinivasan RR, Lei JT, Kovacevic V, Obradovic P, Ellis MJ, Osborne CK, Rimawi MF, Pavlick A, Shafaee MN, Dowst H, Jain A, Saltzman AB, Malovannaya A, Marangoni E, Welm AL, Welm BE, Li S, Wulf GM, Sonzogni O, Huang C, Vasaikar S, Hilsenbeck SG, Zhang B, Milosavljevic A, Lewis MT. Identifying biomarkers of differential chemotherapy response in TNBC patient-derived xenografts with a CTD/WGCNA approach. iScience 2023; 26:105799. [PMID: 36619972 PMCID: PMC9813793 DOI: 10.1016/j.isci.2022.105799] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 07/20/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022] Open
Abstract
Although systemic chemotherapy remains the standard of care for TNBC, even combination chemotherapy is often ineffective. The identification of biomarkers for differential chemotherapy response would allow for the selection of responsive patients, thus maximizing efficacy and minimizing toxicities. Here, we leverage TNBC PDXs to identify biomarkers of response. To demonstrate their ability to function as a preclinical cohort, PDXs were characterized using DNA sequencing, transcriptomics, and proteomics to show consistency with clinical samples. We then developed a network-based approach (CTD/WGCNA) to identify biomarkers of response to carboplatin (MSI1, TMSB15A, ARHGDIB, GGT1, SV2A, SEC14L2, SERPINI1, ADAMTS20, DGKQ) and docetaxel (c, MAGED4, CERS1, ST8SIA2, KIF24, PARPBP). CTD/WGCNA multigene biomarkers are predictive in PDX datasets (RNAseq and Affymetrix) for both taxane- (docetaxel or paclitaxel) and platinum-based (carboplatin or cisplatin) response, thereby demonstrating cross-expression platform and cross-drug class robustness. These biomarkers were also predictive in clinical datasets, thus demonstrating translational potential.
Collapse
Affiliation(s)
- Varduhi Petrosyan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lacey E. Dobrolecki
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Lillian Thistlethwaite
- Quantitative and Computational Biosciences Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alaina N. Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Christina Sallas
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | - Jonathan T. Lei
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Vladimir Kovacevic
- School of Electrical Engineering, University of Belgrade, Belgrade, Serbia
| | - Predrag Obradovic
- School of Electrical Engineering, University of Belgrade, Belgrade, Serbia
| | - Matthew J. Ellis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - C. Kent Osborne
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Mothaffar F. Rimawi
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anne Pavlick
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maryam Nemati Shafaee
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Heidi Dowst
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alexander B. Saltzman
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX 77030, USA
| | - Anna Malovannaya
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Verna and Marrs McLean Department of Biochemistry and Molecular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | | | - Alana L. Welm
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT 84112, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Bryan E. Welm
- Department of Surgery, University of Utah, Salt Lake City, UT 84112, USA
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Shunqiang Li
- Division of Oncology, Washington University, St. Louis, MO 63130, USA
| | | | - Olmo Sonzogni
- Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Chen Huang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Suhas Vasaikar
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Susan G. Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Bing Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Quantitative and Computational Biosciences Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Aleksandar Milosavljevic
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Quantitative and Computational Biosciences Program, Baylor College of Medicine, Houston, TX 77030, USA
| | - Michael T. Lewis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Department of Radiology, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
50
|
Prognostic Implications of the Residual Tumor Microenvironment after Neoadjuvant Chemotherapy in Triple-Negative Breast Cancer Patients without Pathological Complete Response. Cancers (Basel) 2023; 15:cancers15030597. [PMID: 36765559 PMCID: PMC9913578 DOI: 10.3390/cancers15030597] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
With a high risk of relapse and death, and a poor or absent response to therapeutics, the triple-negative breast cancer (TNBC) subtype is particularly challenging, especially in patients who cannot achieve a pathological complete response (pCR) after neoadjuvant chemotherapy (NAC). Although the tumor microenvironment (TME) is known to influence disease progression and the effectiveness of therapeutics, its predictive and prognostic potential remains uncertain. This work aimed to define the residual TME profile after NAC of a retrospective cohort with 96 TNBC patients by immunohistochemical staining (cell markers) and chromogenic in situ hybridization (genetic markers). Kaplan-Meier curves were used to estimate the influence of the selected TME markers on five-year overall survival (OS) and relapse-free survival (RFS) probabilities. The risks of each variable being associated with relapse and death were determined through univariate and multivariate Cox analyses. We describe a unique tumor-infiltrating immune profile with high levels of lymphocytes (CD4, FOXP3) and dendritic cells (CD21, CD1a and CD83) that are valuable prognostic factors in post-NAC TNBC patients. Our study also demonstrates the value of considering not only cellular but also genetic TME markers such as MUC-1 and CXCL13 in routine clinical diagnosis to refine prognosis modelling.
Collapse
|