1
|
Jiang C, Qian Y, Bai X, Li S, Zhang L, Xie Y, Lu Y, Lu Z, Liu B, Jiang BH. SLC7A5/E2F1/PTBP1/PKM2 axis mediates progression and therapy effect of triple-negative breast cancer through the crosstalk of amino acid metabolism and glycolysis pathway. Cancer Lett 2025; 617:217612. [PMID: 40054655 DOI: 10.1016/j.canlet.2025.217612] [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: 01/20/2025] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Triple-negative breast cancer (TNBC) is one of the most challenging malignancies with the highest mortality rates among women. TNBC relies on both amino acid metabolism and glycolysis to fuel its bioenergetic and biosynthetic demands. However, the potential crosstalk between these two metabolic pathways and its impact on TNBC progression remain largely unexplored. In this study, we observed that SLC7A5, a key amino acid transporter, was upregulated in TNBC and strongly associated with poor patient prognosis. We demonstrated that the elevated SLC7A5 expression activated the amino acid pathway and promoted cell proliferation, tumor growth, and therapeutic resistance by inducing the switch from PKM1 to PKM2 expression, thereby mediating the crosstalk between amino acid metabolism and glycolysis. We further identified that the upregulation of SLC7A5 resulted from miR-152 suppression, which regulates TNBC cellular function and tumor growth. In addition, the miR-152/SLC7A5 axis mediated the expression of PTBP1, which maintains the balance between PKM1 and PKM2, linking amino acid signaling with the glycolysis pathway. To further understand the mechanism of PTBP1 upregulation, we identified that E2F1 transcriptionally activated PTBP1 expression through direct binding at the seed site, while E2F1 expression was also induced by SLC7A5 in TNBC. This novel SLC7A5/E2F1/PTBP1 axis plays a crucial role in regulating the crosstalk between amino acid signaling and glycolysis in TNBC and is essential for TNBC progression and therapeutic effectiveness. Our findings offer valuable insights into the molecular mechanisms underlying TNBC metabolic reprogramming and highlight potential targets for future therapeutic interventions.
Collapse
Affiliation(s)
- Chengfei Jiang
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China; Department of Pathology, Nanjing Medical University, Nanjing, 210029, China
| | - Yingchen Qian
- Department of Pathology, The Affiliated Jiangning Hospital of Nanjing Medical University, Nanjing, 211100, China
| | - Xiaoming Bai
- Department of Pathology, Nanjing Medical University, Nanjing, 210029, China
| | - Shuangya Li
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Liyuan Zhang
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Yunxia Xie
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China
| | - Yifan Lu
- Department of Pathology, Nanjing Medical University, Nanjing, 210029, China
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310029, China
| | - Bingjie Liu
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China.
| | - Bing-Hua Jiang
- The Third Affiliated Hospital of Zhengzhou University, Tianjian Laboratory of Advanced Biomedical Sciences, Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
2
|
Aine M, Nacer DF, Arbajian E, Veerla S, Karlsson A, Häkkinen J, Johansson HJ, Rosengren F, Vallon-Christersson J, Borg Å, Staaf J. The DNA methylation landscape of primary triple-negative breast cancer. Nat Commun 2025; 16:3041. [PMID: 40155623 PMCID: PMC11953470 DOI: 10.1038/s41467-025-58158-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 03/10/2025] [Indexed: 04/01/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is a clinically challenging and molecularly heterogenous breast cancer subgroup. Here, we investigate the DNA methylation landscape of TNBC. By analyzing tumor methylome profiles and accounting for the genomic context of CpG methylation, we divide TNBC into two epigenetic subtypes corresponding to a Basal and a non-Basal group, in which characteristic transcriptional patterns are correlated with DNA methylation of distal regulatory elements and epigenetic regulation of key steroid response genes and developmental transcription factors. Further subdivision of the Basal and non-Basal subtypes identifies subgroups transcending genetic and proposed TNBC mRNA subtypes, demonstrating widely differing immunological microenvironments, putative epigenetically-mediated immune evasion strategies, and a specific metabolic gene network in older patients that may be epigenetically regulated. Our study attempts to target the epigenetic backbone of TNBC, an approach that may inform future studies regarding tumor origins and the role of the microenvironment in shaping the cancer epigenome.
Collapse
Affiliation(s)
- Mattias Aine
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Deborah F Nacer
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Elsa Arbajian
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Srinivas Veerla
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Anna Karlsson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Jari Häkkinen
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Henrik J Johansson
- Department of Oncology-Pathology, Science for Life Laboratory, Karolinska Institutet, Solna, Sweden
| | - Frida Rosengren
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Johan Vallon-Christersson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE 22381, Lund, Sweden.
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, Medicon Village, SE 22381, Lund, Sweden.
| |
Collapse
|
3
|
Maggisano V, Gargano A, Maiuolo J, Ortuso F, De Amicis F, Alcaro S, Bulotta S. Rational Identification of Ritonavir as IL-20 Receptor A Ligand Endowed with Antiproliferative Properties in Breast Cancer Cells. Int J Mol Sci 2025; 26:1285. [PMID: 39941053 PMCID: PMC11818535 DOI: 10.3390/ijms26031285] [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: 12/18/2024] [Revised: 01/22/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Targeting the tumor microenvironment (TME) is an attractive strategy for developing new drugs with anticancer activity against triple-negative breast cancer (TNBC). Interleukins (ILs) are key players in the TME cytokine network promoting cancer progression. Recent studies have highlighted the involvement of IL-20 receptor subunit alpha (IL-20RA) signalling in several cancers, including BC, in which IL-20RA is highly expressed, correlating with poor prognosis and influencing tumoral characteristics such as proliferation, cell death, invasiveness, and TME activity. Therefore, elucidating the role of the IL-20RA signalling pathway could form the basis for developing new therapeutic strategies. This study aimed to identify selective bioactive ligands able to affect IL-20RA activity. Virtual screening of over 310,000 compounds from both the DrugBank and ZINC15 databases identified four potential hit compounds tested for their anticancer activity against TNBC in vitro cell lines. Notably, Ritonavir, a well-known Human Immunodeficiency Virus Type 1 (HIV-1) protease inhibitor, significantly inhibited cell proliferation (about 40% at 50 µM, p < 0.001). IL-20 preincubation counteracted Ritonavir's cytostatic effect while IL-20RA knockdown restored proliferation in Ritonavir-treated TNBC cells. In conclusion, these findings demonstrated that Ritonavir reduced TNBC cell proliferation through IL-20RA activity modulation, suggesting its potential repurposing as a therapeutic agent for TNBC management.
Collapse
Affiliation(s)
- Valentina Maggisano
- Dipartimento di Scienze della Salute, Università degli Studi “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (V.M.); (A.G.); (J.M.); (F.O.); (S.B.)
| | - Adriana Gargano
- Dipartimento di Scienze della Salute, Università degli Studi “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (V.M.); (A.G.); (J.M.); (F.O.); (S.B.)
- Associazione CRISEA—Centro di Ricerca e Servizi Avanzati per l’Innovazione Rurale, Località Condoleo, 88055 Belcastro, Italy
| | - Jessica Maiuolo
- Dipartimento di Scienze della Salute, Università degli Studi “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (V.M.); (A.G.); (J.M.); (F.O.); (S.B.)
| | - Francesco Ortuso
- Dipartimento di Scienze della Salute, Università degli Studi “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (V.M.); (A.G.); (J.M.); (F.O.); (S.B.)
- Net4Science Academic Spinoff, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Francesca De Amicis
- Dipartimento di Farmacia e Scienze della Salute e della Nutrizione, Università della Calabria, 87036 Rende, Italy;
- Centro Sanitario, Università della Calabria, 87036 Rende, Italy
| | - Stefano Alcaro
- Dipartimento di Scienze della Salute, Università degli Studi “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (V.M.); (A.G.); (J.M.); (F.O.); (S.B.)
- Associazione CRISEA—Centro di Ricerca e Servizi Avanzati per l’Innovazione Rurale, Località Condoleo, 88055 Belcastro, Italy
- Net4Science Academic Spinoff, Università “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy
| | - Stefania Bulotta
- Dipartimento di Scienze della Salute, Università degli Studi “Magna Græcia” di Catanzaro, Campus Universitario “S. Venuta”, Viale Europa, 88100 Catanzaro, Italy; (V.M.); (A.G.); (J.M.); (F.O.); (S.B.)
| |
Collapse
|
4
|
Zhang J, Lu L, Zhang W, Miao Y, Du H, Xia H, Tao Z, Du Z, Tang Y, Fang Q. Gadolinium ion-loaded mesoporous organosilica nanoplatform for enhanced radiotherapy in breast tumor treatment. Colloids Surf B Biointerfaces 2025; 246:114374. [PMID: 39541910 DOI: 10.1016/j.colsurfb.2024.114374] [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: 08/19/2024] [Revised: 10/22/2024] [Accepted: 11/10/2024] [Indexed: 11/17/2024]
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive subtype with limited therapeutic options, often exhibiting resistance to standard radiotherapy (RT) and chemotherapy. Recent advancements in nanomedicine provide an opportunity to enhance treatment efficacy through innovative drug delivery systems and radiosensitizers. In this study, we present a novel nanotheranostic platform, MOs-G@DOX, engineered to enhance the therapeutic efficacy of RT in the treatment of TNBC. This platform consists of gadolinium-containing mesoporous organosilica nanoparticles (MOs-G) that serve a dual function as a drug carrier and a radiosensitizer. The MOs-G were synthesized via a surfactant-mediated sol-gel process, followed by gadolinium incorporation through nanoprecipitation. The antitumor drug doxorubicin (DOX) was subsequently loaded into the mesoporous structure, forming the MOs-G@DOX nanoplatform. Comprehensive in vitro and in vivo studies demonstrated that MOs-G@DOX exhibits excellent biocompatibility and significantly enhances the radiosensitivity of TNBC cells, leading to superior tumor growth inhibition compared to conventional treatments. The stability of MOs-G, with minimal gadolinium ion leakage, further underscores its potential as a safe and effective nanomedicine. Additionally, the combination of MOs-G@DOX with RT showed a marked increase in reactive oxygen species (ROS) generation and tumor cell apoptosis, which were confirmed through histological analyses. These findings suggest that MOs-G@DOX is a promising candidate for advancing cancer therapy, particularly in the context of RT for TNBC.
Collapse
Affiliation(s)
- Junjie Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China.
| | - Li Lu
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Wenqing Zhang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Yuchen Miao
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Hengda Du
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Hui Xia
- Department of Microbiology and Parasitology, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Zhiyong Tao
- Department of Microbiology and Parasitology, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Zhaofeng Du
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Yulong Tang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China
| | - Qiang Fang
- School of Fundamental Sciences, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Department of Microbiology and Parasitology, Bengbu Medical University, Bengbu, Anhui Province 233030, China; Anhui Key Laboratory of Infection and Immunity, Bengbu Medical University, Bengbu, Anhui Province 233030, China.
| |
Collapse
|
5
|
Zhang Z, Huang W, Wang L, Li G, Xu F, Wu P, Luo C, Huang Q, Kuang W, Liu Z, Jiang Y, Zhao X, Zhang Y, Ye W, Li J, Ma N, Wang J. Ailanthone induces triple-negative breast cancer cells death involving the inhibition of OTUB1-mediated ERRα deubiquitylation. J Adv Res 2025:S2090-1232(25)00054-2. [PMID: 39864589 DOI: 10.1016/j.jare.2025.01.035] [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: 10/28/2024] [Revised: 01/04/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025] Open
Abstract
INTRODUCTION Triple-negative breast cancer (TNBC) remains the most aggressive subtype of breast cancer, and effective therapeutic strategies are needed. Estrogen-related receptor alpha (ERRα) is considered a promising target for managing TNBC. OBJECTIVES Here, we aimed to screen natural products to find downregulator of ERRα and elucidate its mechanism of action. METHODS TNBC cells (MDA-MB-231, MDA-MB-468, MDA-MB-453, and BT-549) were used for in vitro studies, and a subcutaneous MDA-MB-231 tumor model was created for in vivo studies. Immunofluorescence assessed protein distribution, while competitive activity-based protein profiling identified potential target proteins. Co-immunoprecipitation detected protein interactions and modifications, and a luciferase reporter assay evaluated ERRα transcriptional activity. RESULTS The natural product Ailanthone (AIL) effectively induced cell death in TNBC cells by reducing the protein level of ERRα. The mechanism of action involved AIL promoting the degradation of ERRα through the ubiquitin-proteasome system, consequently reducing its transcriptional activity. The competitive-ABPP method mapped the profile of target proteins for AIL, and OTU domain-containing ubiquitin aldehyde-binding protein 1 (OTUB1) was identified as a pivotal target of AIL in regulating ERRα protein levels. OTUB1 was validated as a novel deubiquitinating enzyme for ERRα, with its C91 residue being crucial for this deubiquitination process. AIL was found to inhibit the enzyme activity of OTUB1 by interacting with the C91 residue and disrupt the interaction between OTUB1 and ERRα, ultimately leading to the inhibition of ERRα. CONCLUSION AIL is a promising downregulator of ERRα, and the mechanism of this downregulation has been elucidated. Additionally, a new regulatory relationship between ERRα and OTUB1 is identified. The research presented in this article is anticipated to yield potential lead compounds for ERRα regulatory agents and to stimulate the development of novel therapeutic strategies designed to modulate ERRα activity for the treatment of TNBC.
Collapse
Affiliation(s)
- Ziyue Zhang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China; State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM & New Drugs Research, Center for Bioactive Natural Molecules and Innovative Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Wei Huang
- Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, Department of Pharmacy, Hubei University of Chinese Medicine, Hubei Shizhen Laboratory, Wuhan 430061, China; Institute for Safflower Industry Research of Shihezi University, Pharmacy College of Shihezi University, KeyLaboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, Shihezi 832003, China
| | - Li Wang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China; Department of Breast Surgery, Xingtai Peoples' Hospital, Xingtai 054000, China
| | - Guanjun Li
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| | - Fang Xu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM & New Drugs Research, Center for Bioactive Natural Molecules and Innovative Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Pengfei Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM & New Drugs Research, Center for Bioactive Natural Molecules and Innovative Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Chuqiao Luo
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| | - Qian Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM & New Drugs Research, Center for Bioactive Natural Molecules and Innovative Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Wenhua Kuang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| | - Zhengyong Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM & New Drugs Research, Center for Bioactive Natural Molecules and Innovative Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China
| | - Ying Jiang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| | - Xiaoling Zhao
- Department of Breast Surgery, Xingtai Peoples' Hospital, Xingtai 054000, China
| | - Yayuan Zhang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China.
| | - Wencai Ye
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM & New Drugs Research, Center for Bioactive Natural Molecules and Innovative Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Juan Li
- Hubei Province Key Laboratory of Traditional Chinese Medicine Resource and Chemistry, Department of Pharmacy, Hubei University of Chinese Medicine, Hubei Shizhen Laboratory, Wuhan 430061, China.
| | - Nan Ma
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic Constituents of TCM & New Drugs Research, Center for Bioactive Natural Molecules and Innovative Drugs Research, College of Pharmacy, Jinan University, Guangzhou 510632, China.
| | - Jigang Wang
- Department of Pulmonary and Critical Care Medicine, Shenzhen Institute of Respiratory Diseases, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatrics, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China; State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| |
Collapse
|
6
|
Piva R, Gharari N, Labrador M, Mader S. IDH2 Inhibitors Gain a Wildcard Status in the Cancer Therapeutics Competition. Cancers (Basel) 2024; 16:3280. [PMID: 39409901 PMCID: PMC11476114 DOI: 10.3390/cancers16193280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
The metabolic reprogramming characteristic of cancer cells, including the Warburg effect, has long been recognized as a hallmark of malignancy. This commentary explores three recent investigations focusing on the role of wild-type IDH2 in cancer and immune cell function. The first publication identifies wild-type IDH2 as a crucial factor in the survival of triple-negative breast cancer (TNBC) cells, with its inhibition leading to disrupted energy metabolism, reduced tumor growth, and enhanced apoptosis. The second analysis examines the role of IDH2 in CD8+ T cells, revealing that its inhibition promotes the differentiation of memory T cells, thereby enhancing the efficacy of cell-based immunotherapies like CAR T cells. A third investigation supports these findings, demonstrating that IDH2 inhibition in CAR T cells reduces exhaustion, enhances memory T cell formation, and improves anti-tumor efficacy. Collectively, these reports highlight wild-type IDH2 as a promising therapeutic target, with potential applications as a two-edged sword in both cancer treatment and immunotherapy. The development of specific wild-type IDH2 inhibitors could offer new avenues for therapy, particularly in tumors reliant on IDH2 activity as well as in enhancing the effectiveness of CAR T cell therapies.
Collapse
Affiliation(s)
- Roberto Piva
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; (N.G.); (M.L.)
- Città della Salute e della Scienza Hospital, 10126 Turin, Italy
| | - Nariman Gharari
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; (N.G.); (M.L.)
| | - Maria Labrador
- Department of Molecular Biotechnology and Health Sciences, University of Turin, 10126 Turin, Italy; (N.G.); (M.L.)
| | - Sylvie Mader
- Department of Biochemistry and Molecular Medicine, Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC H3T 1J4, Canada;
| |
Collapse
|
7
|
Fu Z, Feng M, Wu J, Liu B, Fu J, Song W. Photodynamic Therapy Synergizes CD47 Blockade Strategy for Enhanced Antitumor Therapy. Mol Pharm 2024; 21:3897-3908. [PMID: 38959154 DOI: 10.1021/acs.molpharmaceut.4c00254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The antitumor strategies based on innate immunity activation have become favored by researchers in recent years. In particular, strategies targeting antiphagocytic signaling blockade to enhance phagocytosis have been widely reported. For example, the addition of prophagocytic signals such as calreticulin could make the strategy significantly more effective. In this study, an antitumor strategy that combines photodynamic therapy (PDT) with CD47 blockade has been reported. This approach promotes the maturation of dendritic cells and the presentation of tumor antigens by PDT-mediated tumor immunogenic cell death, as well as the enhancement of cytotoxic T lymphocyte infiltration in tumor areas and the phagocytic activity of phagocytes. Furthermore, the downregulation and blockage of CD47 protein could further promote phagocytic activity, strengthen the innate immune system, and ultimately elevate the antitumor efficacy and inhibit tumor metastasis.
Collapse
Affiliation(s)
- Zhaoming Fu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Minghui Feng
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Jinxian Wu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Bo Liu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Jiajia Fu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| | - Wen Song
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Sanya 572025, China
- Key Laboratory of Biomedical Engineering of Hainan Province, One Health Institute, Hainan University, Sanya 572025, China
| |
Collapse
|
8
|
Huang H, Guo H, Liu J, Ni C, Xia L, Cao X, Xia J, Shi X, Guo R. Dendrimer/metal-phenolic nanocomplexes encapsulating CuO 2 for targeted magnetic resonance imaging and enhanced ferroptosis/cuproptosis/chemodynamic therapy by regulating the tumor microenvironment. Acta Biomater 2024; 183:252-263. [PMID: 38801869 DOI: 10.1016/j.actbio.2024.05.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 05/15/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
The combination of ferroptosis, cuproptosis, and chemodynamic therapy (CDT) would be a potential strategy for tumor diagnosis and enhanced treatment. However, the therapeutic effect was severely limited by the lack of specific delivery of catalytic ions and the low Fenton reaction efficiency in tumor microenvironment (TME) with excess glutathione, limited acidity and insufficient endogenous hydrogen peroxide. In this work, p-carboxybenzenesulfonamide (BS), a carbonic anhydrase IX (CA IX) inhibitor, was modified on the surface of generation-5 poly(amidoamine) dendrimer to load copper peroxide nanoparticles, which were complexed with iron (Fe)-tannic acid (TF) networks for targeted magnetic resonance (MR) imaging and enhanced ferroptosis/cuproptosis/CDT by regulating TME. The formed CuO2@G5-BS/TF nanocomplexes with an average size of 39.4 nm could be specifically accumulated at tumor site and effectively internalized by metastatic 4T1 cells via the specific interaction between BS and CA IX over-expressed on tumor cells. Meanwhile, the inhibition of CA IX activity could not only decrease the intracellular pH to accelerate Fe3+/Cu2+ release, H2O2 self-supply and Fenton reaction, but also suppress tumor metastasis by alleviating the extracellular acidity in TME. Moreover, the reduction of Fe3+/Cu2+ by intracellular glutathione (GSH) could further amplify ROS generation and enhance CDT efficacy, and the GSH depletion could in turn inhibit GPX-4 mediated antioxidant reaction to induce ferroptosis, resulting in effective therapeutic efficacy. In vivo experimental results demonstrated that CuO2@G5-BS/TF could provide better tumor MR imaging, effectively inhibit the growth and metastasis of 4T1 breast tumors, and be metabolized without significant systemic toxicity. Thus, CuO2@G5-BS/TF nanocomplexes provided a new approach for targeted MR imaging and enhanced ferroptosis/cuproptosis/CDT of triple-negative breast cancer. STATEMENT OF SIGNIFICANCE: Taking the advantage of dendrimer and metal-phenolic system, stable CuO2@G5-BS/TF nanocomplexes with an average size of 39.4 nm were synthesized to efficiently load Fe3+ and CuO2 nanoparticles for TNBC treatment and MR imaging. CuO2@G5-BS/TF nanocomplexes could target tumor cells overexpressing CAIX via the specific binding with BS, and the inhibition of CAIX activity could not only decrease the intracellular pH to accelerate Fe3+/Cu2+ release, H2O2 self-supply and Fenton reaction, but also suppress tumor metastasis by alleviating the extracellular acidity. The reduction of Fe3+/Cu2+ by intracellular GSH could further amplify ·OH generation, and the GSH depletion could in turn inhibit GPX-4 mediated antioxidant reaction to induce ferroptosis, resulting in effective therapeutic efficacy by enhanced ferroptosis/cuproptosis/CDT via tumor microenvironment regulation.
Collapse
Affiliation(s)
- Haoyu Huang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China; College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, PR China
| | - Honghua Guo
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201620, PR China
| | - Junjie Liu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China; College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, PR China
| | - Cheng Ni
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China
| | - Li Xia
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China; College of Chemistry and Chemical Engineering, Donghua University, Shanghai 201620, PR China
| | - Xueyan Cao
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China
| | - Jindong Xia
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai 201620, PR China
| | - Xiangyang Shi
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China.
| | - Rui Guo
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Shanghai Engineering Research Center of Nano-Biomaterials and Regenerative Medicine, College of Biological Science and Medical Engineering, Donghua University, Shanghai 201620, PR China.
| |
Collapse
|
9
|
Li JJ, Yu T, Zeng P, Tian J, Liu P, Qiao S, Wen S, Hu Y, Liu Q, Lu W, Zhang H, Huang P. Wild-type IDH2 is a therapeutic target for triple-negative breast cancer. Nat Commun 2024; 15:3445. [PMID: 38658533 PMCID: PMC11043430 DOI: 10.1038/s41467-024-47536-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/26/2024] [Indexed: 04/26/2024] Open
Abstract
Mutations in isocitrate dehydrogenases (IDH) are oncogenic events due to the generation of oncogenic metabolite 2-hydroxyglutarate. However, the role of wild-type IDH in cancer development remains elusive. Here we show that wild-type IDH2 is highly expressed in triple negative breast cancer (TNBC) cells and promotes their proliferation in vitro and tumor growth in vivo. Genetic silencing or pharmacological inhibition of wt-IDH2 causes a significant increase in α-ketoglutarate (α-KG), indicating a suppression of reductive tricarboxylic acid (TCA) cycle. The aberrant accumulation of α-KG due to IDH2 abrogation inhibits mitochondrial ATP synthesis and promotes HIF-1α degradation, leading to suppression of glycolysis. Such metabolic double-hit results in ATP depletion and suppression of tumor growth, and renders TNBC cells more sensitive to doxorubicin treatment. Our study reveals a metabolic property of TNBC cells with active utilization of glutamine via reductive TCA metabolism, and suggests that wild-type IDH2 plays an important role in this metabolic process and could be a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Jiang-Jiang Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Tiantian Yu
- Metabolic Innovation Center, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, 510080, China
| | - Peiting Zeng
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Jingyu Tian
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Panpan Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Medical Oncology, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Shuang Qiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Shijun Wen
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Yumin Hu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Qiao Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Wenhua Lu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Hui Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Metabolic Innovation Center, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, 510080, China
| | - Peng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
- Metabolic Innovation Center, Sun Yat-sen University Zhongshan School of Medicine, Guangzhou, 510080, China.
| |
Collapse
|
10
|
Liao H, Wang Y, Zou L, Fan Y, Wang X, Tu X, Zhu Q, Wang J, Liu X, Dong C. Relationship of mTORC1 and ferroptosis in tumors. Discov Oncol 2024; 15:107. [PMID: 38583115 PMCID: PMC10999401 DOI: 10.1007/s12672-024-00954-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 03/28/2024] [Indexed: 04/08/2024] Open
Abstract
Ferroptosis is a novel form of programmed death, dependent on iron ions and oxidative stress, with a predominant intracellular form of lipid peroxidation. In recent years, ferroptosis has gained more and more interest of people in the treatment mechanism of targeted tumors. mTOR, always overexpressed in the tumor, and controlling cell growth and metabolic activities, has an important role in both autophagy and ferroptosis. Interestingly, the selective types of autophay plays an important role in promoting ferroptosis, which is related to mTOR and some metabolic pathways (especially in iron and amino acids). In this paper, we list the main mechanisms linking ferroptosis with mTOR signaling pathway and further summarize the current compounds targeting ferroptosis in these ways. There are growing experimental evidences that targeting mTOR and ferroptosis may have effective impact in many tumors, and understanding the mechanisms linking mTOR to ferroptosis could provide a potential therapeutic approach for tumor treatment.
Collapse
Affiliation(s)
- Huilin Liao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Yueqing Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Lili Zou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Yanmei Fan
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Xinyue Wang
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Xiancong Tu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Qiaobai Zhu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China, 443002
| | - Jun Wang
- The People's Hospital of China Three Gorges University and The First People's Hospital of Yichang, Yichang, Hubei, China, 443002
| | - Xiaowen Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang, Hubei, China, 443002.
- The Institute of Infection and Inflammation, College of Basic Medical Sciences, China Three Gorges University, Yichang, Hubei, China, 443002.
| | - Chuanjiang Dong
- Department of Urology, The First Dongguan Affiliated Hospital of Guangdong Medical University, Dongguan, Guangdong, China, 523000.
| |
Collapse
|
11
|
Cao T, Huang M, Huang X, Tang T. Research and experimental verification on the mechanisms of cellular senescence in triple-negative breast cancer. PeerJ 2024; 12:e16935. [PMID: 38435998 PMCID: PMC10909353 DOI: 10.7717/peerj.16935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/22/2024] [Indexed: 03/05/2024] Open
Abstract
Background Triple-negative breast cancer (TNBC) is an aggressive breast cancer subtype with high heterogeneity, poor prognosis, and a low 10-year survival rate of less than 50%. Although cellular senescence displays extensive effects on cancer, the comprehensions of cellular senescence-related characteristics in TNBC patients remains obscure. Method Single-cell RNA sequencing (scRNA-seq) data were analyzed by Seurat package. Scores for cellular senescence-related pathways were computed by single-sample gene set enrichment analysis (ssGSEA). Subsequently, unsupervised consensus clustering was performed for molecular cluster identification. Immune scores of patients in The Cancer Genome Atlas (TCGA) dataset and associated immune cell scores were calculated using Estimation of STromal and Immune cells in MAlignantTumours using Expression data (ESTIMATE) and Microenvironment Cell Populations-counter (MCP-counter), Tumor Immune Estimation Resource (TIMER) and Estimating the Proportion of Immune and Cancer cells (EPIC) methods, respectively. Immunotherapy scores were assessed using TIDE. Furthermore, feature genes were identified by univariate Cox and Least Absolute Shrinkage and Selection Operator (LASSO) regression analyses; these were used to construct a risk model. Additionally, quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and transwell assay were conducted for in vitro validation of hub genes. Result TNBC was classified into three subtypes based on cellular senescence-related pathways as clusters 1, 2, and 3. Specifically, cluster 1 showed the best prognosis, followed by cluster 2 and cluster 3. The levels of gene expression in cluster 2 were the lowest, whereas these were the highest in cluster 3. Moreover, clusters 1 and 3 showed a high degree of immune infiltration. TIDE scores were higher for cluster 3, suggesting that immune escape was more likely in patients with the cluster 3 subtype who were less likely to benefit from immunotherapy. Next, the TNBC risk model was constructed and validated. RT-qPCR revealed that prognostic risk genes (MMP28, ACP5 and KRT6A) were up-regulated while protective genes (CT83) were down-regulated in TNBC cell lines, validating the results of the bioinformatics analysis. Meanwhile, cellular experiments revealed that ACP5 could promote the migration and invasion abilities in two TNBC cell lines. Finally, we evaluated the validity of prognostic models for assessing TME characteristics and TNBC chemotherapy response. Conclusion In conclusion, these findings help to assess the efficacy of targeted therapies in patients with different molecular subtypes, have practical applications for subtype-specific treatment of TNBC patients, and provide information on prognostic factors, as well as guidance for the revelation of the molecular mechanisms by which senescence-associated genes influence TNBC progression.
Collapse
Affiliation(s)
- Tengfei Cao
- Department of Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Mengjie Huang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xinyue Huang
- Department of Breast Surgery, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Tian Tang
- Department of Pathology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
12
|
Lin LH, Tran I, Yang Y, Shen G, Miah P, Cotzia P, Roses D, Schnabel F, Darvishian F, Snuderl M. DNA Methylation Identifies Epigenetic Subtypes of Triple-Negative Breast Cancers With Distinct Clinicopathologic and Molecular Features. Mod Pathol 2023; 36:100306. [PMID: 37595637 DOI: 10.1016/j.modpat.2023.100306] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/25/2023] [Accepted: 08/02/2023] [Indexed: 08/20/2023]
Abstract
Triple-negative breast cancers (TNBC) include diverse carcinomas with heterogeneous clinical behavior. DNA methylation is a useful tool in classifying a variety of cancers. In this study, we analyzed TNBC using DNA methylation profiling and compared the results to those of mutational analysis. DNA methylation profiling (Infinium MethylationEPIC array, Illumina) and 50-gene panel-targeted DNA sequencing were performed in 44 treatment-naïve TNBC. We identified 3 distinct DNA methylation clusters with specific clinicopathologic and molecular features. Cluster 1 (phosphoinositide 3-kinase/protein kinase B-enriched cluster; n = 9) patients were significantly older (mean age, 71 years; P = .008) with tumors that were more likely to exhibit apocrine differentiation (78%; P < .001), a lower grade (44% were grade 2), a lower proliferation index (median Ki-67, 15%; P = .002), and lower tumor-infiltrating lymphocyte fractions (median, 15%; P = .0142). Tumors carried recurrent PIK3CA and AKT1 mutations and a higher percentage of low HER-2 expression (89%; P = .033). Cluster 3 (chromosomal instability cluster; n = 28) patients were significantly younger (median age, 57 years). Tumors were of higher grade (grade 3, 93%), had a higher proliferation index (median Ki-67, 75%), and were with a high fraction of tumor-infiltrating lymphocytes (median, 30%). Ninety-one percent of the germline BRCA1/2 mutation carriers were in cluster 3, and these tumors showed the highest level of copy number alterations. Cluster 2 represented cases with intermediate clinicopathologic characteristics and no specific molecular profile (no specific molecular profile cluster; n = 7). There were no differences in relation to stage, recurrence, and survival. In conclusion, DNA methylation profiling is a promising tool to classify patients with TNBC into biologically relevant groups, which may result in better disease characterization and reveal potential targets for emerging therapies.
Collapse
Affiliation(s)
- Lawrence Hsu Lin
- Department of Pathology, New York University Langone Health and Grossman School of Medicine, New York, New York
| | - Ivy Tran
- Department of Pathology, New York University Langone Health and Grossman School of Medicine, New York, New York
| | - Yiying Yang
- Department of Pathology, New York University Langone Health and Grossman School of Medicine, New York, New York
| | - Guomiao Shen
- Department of Pathology, New York University Langone Health and Grossman School of Medicine, New York, New York
| | - Pabel Miah
- Department of Surgery, New York University Langone Health and Grossman School of Medicine, New York, New York
| | - Paolo Cotzia
- Department of Pathology, New York University Langone Health and Grossman School of Medicine, New York, New York
| | - Daniel Roses
- Department of Surgery, New York University Langone Health and Grossman School of Medicine, New York, New York
| | - Freya Schnabel
- Department of Surgery, New York University Langone Health and Grossman School of Medicine, New York, New York
| | - Farbod Darvishian
- Department of Pathology, New York University Langone Health and Grossman School of Medicine, New York, New York
| | - Matija Snuderl
- Department of Pathology, New York University Langone Health and Grossman School of Medicine, New York, New York.
| |
Collapse
|
13
|
Lei J, Pan Y, Gao R, He B, Wang Z, Lei X, Zhang Z, Yang N, Yan M. Rutaecarpine induces the differentiation of triple-negative breast cancer cells through inhibiting fumarate hydratase. J Transl Med 2023; 21:553. [PMID: 37592347 PMCID: PMC10436383 DOI: 10.1186/s12967-023-04396-w] [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: 05/10/2023] [Accepted: 07/29/2023] [Indexed: 08/19/2023] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is one of the most aggressive human cancers and has poor prognosis. Approximately 80% of TNBC cases belong to the molecular basal-like subtype, which can be exploited therapeutically by inducing differentiation. However, the strategies for inducing the differentiation of TNBC remain underexplored. METHODS A three-dimensional (3D) morphological screening model based on a natural compound library was used to identify possible candidate compounds that can induce TNBC cell differentiation. The efficacy of rutaecarpine was verified using assays: RT-qPCR, RNA-seq, flow cytometry, immunofluorescence, SCENITH and label-free LC-MS/MS. The direct targets of rutaecarpine were identified through drug affinity responsive target stability (DARTS) assay. A xenograft mice model was also constructed to confirm the effect of rutaecarpine in vivo. RESULTS We identified that rutaecarpine, an indolopyridoquinazolinone, induces luminal differentiation of basal TNBC cells in both 3D spheroids and in vivo mice models. Mechanistically, rutaecarpine treatment leads to global metabolic stress and elevated ROS in 3D cultured TNBC cells. Moreover, NAC, a scavenger of ROS, impedes rutaecarpine-induced differentiation of TNBC cells in 3D culture. Finally, we identified fumarate hydratase (FH) as the direct interacting target of rutaecarpine. The inhibition of FH and the knockdown of FH consistently induced the differentiation of TNBC cells in 3D culture. CONCLUSIONS Our results provide a platform for differentiation therapy drug discovery using 3D culture models and identify rutaecarpine as a potential compound for TNBC treatment.
Collapse
Affiliation(s)
- Jie Lei
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yujia Pan
- Institute of Cancer Stem Cell, Cancer Center, Dalian Medical University, Dalian, 116023, China
| | - Rui Gao
- Department of Medical Oncology, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 510275, China
| | - Bin He
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Zifeng Wang
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Xinxing Lei
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Zijian Zhang
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Na Yang
- Department of Laboratory Medicine, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, 510180, China.
| | - Min Yan
- State Key Laboratory of Oncology in South China, Cancer Center, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
14
|
Ferraro MG, Bocchetti M, Riccardi C, Trifuoggi M, Paduano L, Montesarchio D, Misso G, Santamaria R, Piccolo M, Irace C. Triple Negative Breast Cancer Preclinical Therapeutic Management by a Cationic Ruthenium-Based Nucleolipid Nanosystem. Int J Mol Sci 2023; 24:ijms24076473. [PMID: 37047448 PMCID: PMC10094725 DOI: 10.3390/ijms24076473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 03/22/2023] [Accepted: 03/28/2023] [Indexed: 03/31/2023] Open
Abstract
Based on compelling preclinical evidence concerning the progress of our novel ruthenium-based metallotherapeutics, we are focusing research efforts on challenging indications for the treatment of invasive neoplasms such as the triple-negative breast cancer (TNBC). This malignancy mainly afflicts younger women, who are black, or who have a BRCA1 mutation. Because of faster growing and spreading, TNBC differs from other invasive breast cancers having fewer treatment options and worse prognosis, where existing therapies are mostly ineffective, resulting in a large unmet biomedical need. In this context, we benefited from an experimental model of TNBC both in vitro and in vivo to explore the effects of a biocompatible cationic liposomal nanoformulation, named HoThyRu/DOTAP, able to effectively deliver the antiproliferative ruthenium(III) complex AziRu, thus resulting in a prospective candidate drug. As part of the multitargeting mechanisms featuring metal-based therapeutics other than platinum-containing agents, we herein validate the potential of HoThyRu/DOTAP liposomes to act as a multimodal anticancer agent through inhibition of TNBC cell growth and proliferation, as well as migration and invasion. The here-obtained preclinical findings suggest a potential targeting of the complex pathways network controlling invasive and migratory cancer phenotypes. Overall, in the field of alternative chemotherapy to platinum-based drugs, these outcomes suggest prospective brand-new settings for the nanostructured AziRu complex to get promising goals for the treatment of metastatic TNBC.
Collapse
|
15
|
Yang JZ, Bao YF, Ci YJ, Zhu JJ. Efficacy of neoadjuvant chemotherapy combined with low-dose Apatinib in the treatment of triple negative breast cancer. Asian J Surg 2023:S1015-9584(23)00375-5. [PMID: 36990815 DOI: 10.1016/j.asjsur.2023.03.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
|
16
|
Wang K, Zheng C, Xue L, Deng D, Zeng L, Li M, Deng X. A bibliometric analysis of 16,826 triple-negative breast cancer publications using multiple machine learning algorithms: Progress in the past 17 years. Front Med (Lausanne) 2023; 10:999312. [PMID: 36844225 PMCID: PMC9945529 DOI: 10.3389/fmed.2023.999312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 01/16/2023] [Indexed: 02/11/2023] Open
Abstract
Background Triple-negative breast cancer (TNBC) is proposed at the beginning of this century, which is still the most challenging breast cancer subtype due to its aggressive behavior, including early relapse, metastatic spread, and poor survival. This study uses machine learning methods to explore the current research status and deficiencies from a macro perspective on TNBC publications. Methods PubMed publications under "triple-negative breast cancer" were searched and downloaded between January 2005 and 2022. R and Python extracted MeSH terms, geographic information, and other abstracts from metadata. The Latent Dirichlet Allocation (LDA) algorithm was applied to identify specific research topics. The Louvain algorithm established a topic network, identifying the topic's relationship. Results A total of 16,826 publications were identified, with an average annual growth rate of 74.7%. Ninety-eight countries and regions in the world participated in TNBC research. Molecular pathogenesis and medication are most studied in TNBC research. The publications mainly focused on three aspects: Therapeutic target research, Prognostic research, and Mechanism research. The algorithm and citation suggested that TNBC research is based on technology that advances TNBC subtyping, new drug development, and clinical trials. Conclusion This study quantitatively analyzes the current status of TNBC research from a macro perspective and will aid in redirecting basic and clinical research toward a better outcome for TNBC. Therapeutic target research and Nanoparticle research are the present research focus. There may be a lack of research on TNBC from a patient perspective, health economics, and end-of-life care perspectives. The research direction of TNBC may require the intervention of new technologies.
Collapse
Affiliation(s)
- Kangtao Wang
- Department of General Surgery, The Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chanjuan Zheng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan, Department of Pathophysiology, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Lian Xue
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan, Department of Pathophysiology, School of Medicine, Hunan Normal University, Changsha, Hunan, China
| | - Dexin Deng
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Liang Zeng
- Department of Pathology, Guangzhou Women and Children’s Medical Center, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China,*Correspondence: Liang Zeng,
| | - Ming Li
- Department of Immunology, College of Basic Medical Sciences, Central South University, Changsha, Hunan, China,Ming Li,
| | - Xiyun Deng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan, Department of Pathophysiology, School of Medicine, Hunan Normal University, Changsha, Hunan, China,Xiyun Deng,
| |
Collapse
|
17
|
Triple-Negative Apocrine Carcinomas: Toward a Unified Group With Shared Molecular Features and Clinical Behavior. Mod Pathol 2023; 36:100125. [PMID: 36870308 DOI: 10.1016/j.modpat.2023.100125] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/27/2023] [Indexed: 02/09/2023]
Abstract
Triple-negative apocrine carcinomas (TNACs) are rare breast tumors with limited studies evaluating their molecular characteristics and clinical behavior. We performed a histologic, immunohistochemical, genetic, and clinicopathologic assessment of 42 invasive TNACs (1 with a focal spindle cell component) from 41 patients, 2 pure apocrine ductal carcinomas in situ (A-DCIS), and 1 A-DCIS associated with spindle cell metaplastic carcinoma (SCMBC). All TNACs had characteristic apocrine morphology and expressed androgen receptor (42/42), gross cystic disease fluid protein 15 (24/24), and CK5/6 (16/16). GATA3 was positive in most cases (16/18, 89%), and SOX10 was negative (0/22). TRPS1 was weakly expressed in a minority of tumors (3/14, 21%). Most TNACs had low Ki67 proliferation (≤10% in 67%, 26/39), with a median index of 10%. Levels of tumor infiltrating lymphocytes were low (≤10% in 93%, 39/42, and 15% in 7%, 3/42). Eighteen percent of TNACs presented with axillary nodal metastasis (7/38). No patients treated with neoadjuvant chemotherapy achieved pathologic complete response (0%, 0/10). Nearly all patients with TNAC (97%, n = 32) were without evidence of disease at the time of study (mean follow-up of 62 months). Seventeen invasive TNACs and 10 A-DCIS (7 with paired invasive TNAC) were profiled by targeted capture-based next-generation DNA sequencing. Pathogenic mutations in phosphatidylinositol 3-kinase pathway genes PIK3CA (53%) and/or PIK3R1 (53%) were identified in all TNACs (100%), including 4 (24%) with comutated PTEN. Ras-MAPK pathway genes, including NF1 (24%), and TP53 were mutated in 6 tumors each (35%). All A-DCIS shared mutations, such as phosphatidylinositol 3-kinase aberrations and copy number alterations with paired invasive TNACs or SCMBC, and a subset of invasive carcinomas showed additional mutations in tumor suppressors (NF1, TP53, ARID2, and CDKN2A). Divergent genetic profiles between A-DCIS and invasive carcinoma were identified in 1 case. In summary, our findings support TNAC as a morphologically, immunohistochemically, and genetically homogeneous subgroup of triple-negative breast carcinomas and suggest overall favorable clinical behavior.
Collapse
|
18
|
Wu C, Sun C, Han X, Ye Y, Qin Y, Liu S. Sanyin Formula Enhances the Therapeutic Efficacy of Paclitaxel in Triple-Negative Breast Cancer Metastases through the JAK/STAT3 Pathway in Mice. Pharmaceuticals (Basel) 2022; 16:9. [PMID: 36678509 PMCID: PMC9867389 DOI: 10.3390/ph16010009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/15/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
Sanyin formula (SYF) is used as a complementary treatment for triple-negative breast cancer (TNBC). The purpose of this study was to identify the potential functional components and clarify the underlying molecular mechanisms of SYF in TNBC. High-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) was used to identify the main components of SYF extracts. Network pharmacology and bioinformatic analyses were carried out to identify potential candidate targets of SYF in TNBC. Cell proliferation was determined with a Celigo imaging cytometer. Wound-healing and Transwell assays were adopted to evaluate cell migration. A Transwell cell-invasion assay was performed with Matrigel-coated membranes. In vivo bioluminescence imaging (BLI) and pathological analyses illustrated the effect of SYF on cancer cell metastasis in tumour-bearing mice. The inhibitory mechanism of SYF was investigated via quantitative PCR (qPCR) and Western blotting. We found that 3,4-dihydroxyphenyllactic acid, kaempferol, p-coumaric acid, and vanillic acid may be the active components of SYF. Molecular docking confirmed that kaempferol, p-coumaric acid, vanillic acid, and 3,4-dihydroxyphenyllactic acid bound stably to proteins such as AKR1C3, MMPs, and STAT3. SYF extract suppressed TNBC cell proliferation, migration, invasion, and metastasis by inhibiting JAK/STAT3 signalling and then regulating downstream genes, such as MMP-2/MMP-9. SYF regulates the expression of genes involved in cell proliferation, migration, and invasion by regulating the JAK/STAT3 signalling pathway and finally inhibits tumour cell metastasis in TNBC. The present study clarifies the mechanism by which SYF inhibits TNBC metastasis and lays an experimental foundation for the continued clinical development of SYF targeting the JAK/STAT3 pathway.
Collapse
Affiliation(s)
- Chunyu Wu
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China
| | - Chenping Sun
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China
| | - Xianghui Han
- Longhua Hospital, Institute of Chinese Traditional Surgery, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China
| | - Yiyi Ye
- Longhua Hospital, Institute of Chinese Traditional Surgery, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China
| | - Yuenong Qin
- Department of Breast Surgery (Integrated Traditional and Western Medicine), Longhua Hospital, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China
| | - Sheng Liu
- Longhua Hospital, Institute of Chinese Traditional Surgery, Shanghai University of Traditional Chinese Medicine, 725 South Wanping Road, Shanghai 200032, China
| |
Collapse
|
19
|
Zeng Z, Wang Z, Chen S, Xiao C, Liu M, Zhang J, Fan J, Zhao Y, Liu B. Bio-nanocomplexes with autonomous O 2 generation efficiently inhibit triple negative breast cancer through enhanced chemo-PDT. J Nanobiotechnology 2022; 20:500. [PMID: 36424589 PMCID: PMC9694858 DOI: 10.1186/s12951-022-01706-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 11/11/2022] [Indexed: 11/25/2022] Open
Abstract
As one kind of aggressive cancer, triple-negative breast cancer (TNBC) has become one of the major causes of women mortality worldwide. Recently, combinational chemo-PDT therapy based on nanomaterials has been adopted for the treatment of malignant tumor. However, the efficacy of PDT was partly compromised under tumor hypoxia environment due to the lack of sustainable O2 supply. In this study, CeO2-loaded nanoparticles (CeNPs) with peroxidase activity were synthesized to autonomously generate O2 by decomposing H2O2 within tumor region and reprogramming the hypoxia microenvironment as well. Meanwhile, the compound cinobufagin (CS-1) was loaded for inhibiting TNBC growth and metastasis. Moreover, the hybrid membrane camouflage was adopted to improve the biocompatibility and targeting ability of nanocomplexes. In vitro assay demonstrated that decomposition of H2O2 by CeO2 achieved sustainable O2 supply, which accordingly improved the efficacy of PDT. In turn, the generated O2 improved the cytotoxicity and anti-tumor migration effect of CS-1 by downregulating HIF-1α and MMP-9 levels. In vivo assay demonstrated that the combination of CS-1 and PDT significantly inhibited the growth and distance metastasis of tumor in MDA-MB-231 bearing mice. Thus, this chemo-PDT strategy achieved satisfactory therapeutic effects by smartly utilizing the enzyme activity of nanodrugs and special micro-environment of tumor.
Collapse
Affiliation(s)
- Zhihong Zeng
- College of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China
| | - Zhou Wang
- College of Biology, Hunan University, Changsha, 410082, China
| | - Simin Chen
- College of Biology, Hunan University, Changsha, 410082, China
| | - Chang Xiao
- College of Biology, Hunan University, Changsha, 410082, China
| | - Minzhuo Liu
- College of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China
| | - Jie Zhang
- College of Biological and Chemical Engineering, Changsha University, Changsha, 410022, China
| | - Jialong Fan
- College of Biology, Hunan University, Changsha, 410082, China
| | - Yanzhong Zhao
- Department of Health Management, The Third Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, 410082, China.
| |
Collapse
|
20
|
Celano M, Gagliardi A, Maggisano V, Ambrosio N, Bulotta S, Fresta M, Russo D, Cosco D. Co-Encapsulation of Paclitaxel and JQ1 in Zein Nanoparticles as Potential Innovative Nanomedicine. MICROMACHINES 2022; 13:1580. [PMID: 36295933 PMCID: PMC9609127 DOI: 10.3390/mi13101580] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 09/12/2022] [Accepted: 09/19/2022] [Indexed: 06/16/2023]
Abstract
The manuscript describes the development of zein nanoparticles containing paclitaxel (PTX) and the bromo-and extra-terminal domain inhibitor (S)-tertbutyl2-(4-(4-chlorophenyl)-2,3,9-trimethyl-6H-thieno(3,2-f)(1,2,4)triazolo(4,3-a)(1,4)diazepin-6-yl)acetate (JQ1) together with their cytotoxicity on triple-negative breast cancer cells. The rationale of this association is that of exploiting different types of cancer cells as targets in order to obtain increased pharmacological activity with respect to that exerted by the single agents. Zein, a protein found in the endosperm of corn, was used as a biomaterial to obtain multidrug carriers characterized by mean sizes of ˂200 nm, a low polydispersity index (0.1-0.2) and a negative surface charge. An entrapment efficiency of ~35% of both the drugs was obtained when 0.3 mg/mL of the active compounds were used during the nanoprecipitation procedure. No adverse phenomena such as sedimentation, macro-aggregation or flocculation occurred when the nanosystems were heated to 37 °C. The multidrug nanoformulation demonstrated significant in vitro cytototoxic activity against MDA-MB-157 and MDA-MB-231 cancer cells by MTT-test and adhesion assay which was stronger than that of the compounds encapsulated as single agents. The results evidence the potential application of zein nanoparticles containing PTX and JQ1 as a novel nanomedicine.
Collapse
Affiliation(s)
- Marilena Celano
- Correspondence: (M.C.); (D.C.); Tel.: +39-0961-369-4099 (M.C.); +39-0961-369-4119 (D.C.)
| | | | | | | | | | | | | | - Donato Cosco
- Correspondence: (M.C.); (D.C.); Tel.: +39-0961-369-4099 (M.C.); +39-0961-369-4119 (D.C.)
| |
Collapse
|
21
|
LincRNAs and snoRNAs in Breast Cancer Cell Metastasis: The Unknown Players. Cancers (Basel) 2022; 14:cancers14184528. [PMID: 36139687 PMCID: PMC9496948 DOI: 10.3390/cancers14184528] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 09/10/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Recent advances in research have led to earlier diagnosis and targeted therapies against breast cancer, which has resulted in reduced breast cancer-related mortality. However, the majority of breast cancer-related deaths are due to metastasis of cancer cells to other organs, a process that has not been fully elucidated. Among the factors and genes implicated in the metastatic process regulation, non-coding RNAs have emerged as crucial players. This review focuses on the role of long intergenic noncoding RNAs (lincRNAs) and small nucleolar RNAs (snoRNAs) in breast cancer cell metastasis. LincRNAs are transcribed between two protein-coding genes and are longer than 200 nucleotides, they do not code for a specific protein but function as regulatory molecules in processes such as cell proliferation, apoptosis, epithelial-to-mesenchymal transition, migration, and invasion while most of them are highly elevated in breast cancer tissues and seem to function as competing endogenous RNAs (ceRNAs) inhibiting relevant miRNAs that specifically target vital metastasis-related genes. Similarly, snoRNAs are 60-300 nucleotides long and are found in the nucleolus being responsible for the post-transcriptional modification of ribosomal and spliceosomal RNAs. Most snoRNAs are hosted inside intron sequences of protein-coding and non-protein-coding genes, and they also regulate metastasis-related genes affecting related cellular properties.
Collapse
|
22
|
Yang J, Chang Y, Tien JCY, Wang Z, Zhou Y, Zhang P, Huang W, Vo J, Apel IJ, Wang C, Zeng VZ, Cheng Y, Li S, Wang GX, Chinnaiyan AM, Ding K. Discovery of a Highly Potent and Selective Dual PROTAC Degrader of CDK12 and CDK13. J Med Chem 2022; 65:11066-11083. [PMID: 35938508 PMCID: PMC9876424 DOI: 10.1021/acs.jmedchem.2c00384] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Indexed: 01/28/2023]
Abstract
Selective degradation of the cyclin-dependent kinases 12 and 13 (CDK12/13) presents a novel therapeutic opportunity for triple-negative breast cancer (TNBC), but there is still a lack of dual CDK12/13 degraders. Here, we report the discovery of the first series of highly potent and selective dual CDK12/13 degraders by employing the proteolysis-targeting chimera (PROTAC) technology. The optimal compound 7f effectively degraded CDK12 and CDK13 with DC50 values of 2.2 and 2.1 nM, respectively, in MDA-MB-231 breast cancer cells. Global proteomic profiling demonstrated the target selectivity of 7f. In vitro, 7f suppressed expression of core DNA damage response (DDR) genes in a time- and dose-dependent manner. Further, 7f markedly inhibited proliferation of multiple TNBC cell lines including MFM223, with an IC50 value of 47 nM. Importantly, 7f displayed a significantly improved antiproliferative activity compared to the structurally similar inhibitor 4, suggesting the potential advantage of a CDK12/13 degrader for TNBC targeted therapy.
Collapse
Affiliation(s)
- Jianzhang Yang
- International
Cooperative Laboratory of Traditional Chinese Medicine Modernization
and Innovative Drug Discovery of Chinese Ministry of Education (MOE),
Guangzhou City Key Laboratory of Precision Chemical Drug Development,
College of Pharmacy, Jinan University, 855 Xingye Avenue East, Guangzhou 511400, People’s Republic of China
| | - Yu Chang
- International
Cooperative Laboratory of Traditional Chinese Medicine Modernization
and Innovative Drug Discovery of Chinese Ministry of Education (MOE),
Guangzhou City Key Laboratory of Precision Chemical Drug Development,
College of Pharmacy, Jinan University, 855 Xingye Avenue East, Guangzhou 511400, People’s Republic of China
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - Jean Ching-Yi Tien
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Zhen Wang
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy
of Sciences, #345 Ling Ling Road, Shanghai 200032, People’s Republic of China
| | - Yang Zhou
- International
Cooperative Laboratory of Traditional Chinese Medicine Modernization
and Innovative Drug Discovery of Chinese Ministry of Education (MOE),
Guangzhou City Key Laboratory of Precision Chemical Drug Development,
College of Pharmacy, Jinan University, 855 Xingye Avenue East, Guangzhou 511400, People’s Republic of China
| | - Pujuan Zhang
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy
of Sciences, #345 Ling Ling Road, Shanghai 200032, People’s Republic of China
| | - Weixue Huang
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy
of Sciences, #345 Ling Ling Road, Shanghai 200032, People’s Republic of China
| | - Josh Vo
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ingrid J. Apel
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - Cynthia Wang
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - Victoria Zhixuan Zeng
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - Yunhui Cheng
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - Shuqin Li
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
| | - George Xiaoju Wang
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Arul M. Chinnaiyan
- Michigan
Center for Translational Pathology, University
of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Pathology, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, Michigan 48109, United States
- Howard Hughes
Medical Institute, University of Michigan, Ann Arbor, Michigan 48109, United States
- Department
of Urology, University of Michigan, Ann Arbor, Michigan 48109, United States
| | - Ke Ding
- International
Cooperative Laboratory of Traditional Chinese Medicine Modernization
and Innovative Drug Discovery of Chinese Ministry of Education (MOE),
Guangzhou City Key Laboratory of Precision Chemical Drug Development,
College of Pharmacy, Jinan University, 855 Xingye Avenue East, Guangzhou 511400, People’s Republic of China
- State
Key Laboratory of Bioorganic and Natural Products Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy
of Sciences, #345 Ling Ling Road, Shanghai 200032, People’s Republic of China
- Institute
of Basic Medicine and Cancer (IBMC), Chinese
Academy of Sciences, Hangzhou, Zhejiang 310022, People’s Republic of China
- The
First Affiliated Hospital (Huaqiao Hospital), Jinan University, 601
Huangpu Avenue West, Guangzhou 510632, China
| |
Collapse
|
23
|
Zhang K, Chen L, Zheng H, Zeng Y. Cytokines secreted from adipose tissues mediate tumor proliferation and metastasis in triple negative breast cancer. BMC Cancer 2022; 22:886. [PMID: 35964108 PMCID: PMC9375239 DOI: 10.1186/s12885-022-09959-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 07/29/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Obesity is a high-risk factor for development and poor prognosis of triple-negative breast cancer (TNBC), which was considered as a high malignant and poor clinical outcome breast cancer subtype. TNBC proliferation and migration regulated by obesity is complex. Here, we studied effects of cytokines secreted from adipose tissue on development of TNBC. METHODS Forty postmenopausal cases by Yuebei People's Hospital of Shaoguan with stage I/IIA TNBC were enrolled. Cytokine concentrations were examined using ELISA analysis. Proliferation and migration of TNBC cell lines were performed using CCK8 assays and Transwell tests. The Log-rank (Mantel-Cox) test, two-tailed Mann-Whitney U test and two-tailed unpaired t test were performed using GraphPad Prism 8.4.2. RESULTS Survival analysis indicated that obese patients with TNBC had worse disease free survival (DFS) as compared with normal weight group (Hazard Ratio 4.393, 95% confidence interval (CI) of ratio 1.071-18.02, p < 0.05). Obese patients with TNBC had severe insulin resistance and high plasma triglycerides. However, plasma adiponectin concentration was decreased and interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) concentration was increased in obese TNBC patients as compared with the nonobese group. The similar results were found in the cytokine secretion from adipose tissues and insulin-resistant adipocytes. The secretion of adipose tissue from obese TNBC patients could promote proliferation and migration of TNBC cell lines, including MDA-MB-157, MDA-MB-231, MDA-MB-453 and HCC38 cells. These TNBC cell lines co-incubated with insulin-resistant 3T3-L1 adipocytes or supplementing these cytokines medium also exhibited increase of proliferative and migratory capacity. CONCLUSION TNBC patients with obesity had worse prognosis compared with the normal weight groups. Alteration of cytokines secreted from adipose tissues mediated proliferation and migration of TNBC, leading to tumor progression in TNBC patients with obesity.
Collapse
Affiliation(s)
- Kai Zhang
- Head and Neck Breast Surgery, The Yuebei People's Hospital of Shaoguan, Guangdong Province, 512025, Shaoguan, China
| | - Lin Chen
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Hubei Province, 430070, Wuhan, China
| | - Hongbo Zheng
- Department of Medicine, Genecast Biotechnology Co., Ltd, Jiangsu Province, 214000, Wuxi, China
| | - Yi Zeng
- Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 420 Fuma Road, Fuzhou, Fujian Province, 350014, China.
| |
Collapse
|
24
|
Llera AS, Abdelhay ESFW, Artagaveytia N, Daneri-Navarro A, Müller B, Velazquez C, Alcoba EB, Alonso I, Alves da Quinta DB, Binato R, Bravo AI, Camejo N, Carraro DM, Castro M, Castro-Cervantes JM, Cataldi S, Cayota A, Cerda M, Colombo A, Crocamo S, Del Toro-Arreola A, Delgadillo-Cisterna R, Delgado L, Dreyer-Breitenbach M, Fejerman L, Fernández EA, Fernández J, Fernández W, Franco-Topete RA, Gabay C, Gaete F, Garibay-Escobar A, Gómez J, Greif G, Gross TG, Guerrero M, Henderson MK, Lopez-Muñoz ME, Lopez-Vazquez A, Maldonado S, Morán-Mendoza AJ, Nagai MA, Oceguera-Villanueva A, Ortiz-Martínez MA, Quintero J, Quintero-Ramos A, Reis RM, Retamales J, Rivera-Claisse E, Rocha D, Rodríguez R, Rosales C, Salas-González E, Sanchotena V, Segovia L, Sendoya JM, Silva-García AA, Trinchero A, Valenzuela O, Vedham V, Zagame L, Podhajcer OL. The Transcriptomic Portrait of Locally Advanced Breast Cancer and Its Prognostic Value in a Multi-Country Cohort of Latin American Patients. Front Oncol 2022; 12:835626. [PMID: 35433488 PMCID: PMC9007037 DOI: 10.3389/fonc.2022.835626] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/14/2022] [Indexed: 11/13/2022] Open
Abstract
PURPOSES Most molecular-based published studies on breast cancer do not adequately represent the unique and diverse genetic admixture of the Latin American population. Searching for similarities and differences in molecular pathways associated with these tumors and evaluating its impact on prognosis may help to select better therapeutic approaches. PATIENTS AND METHODS We collected clinical, pathological, and transcriptomic data of a multi-country Latin American cohort of 1,071 stage II-III breast cancer patients of the Molecular Profile of Breast Cancer Study (MPBCS) cohort. The 5-year prognostic ability of intrinsic (transcriptomic-based) PAM50 and immunohistochemical classifications, both at the cancer-specific (OSC) and disease-free survival (DFS) stages, was compared. Pathway analyses (GSEA, GSVA and MetaCore) were performed to explore differences among intrinsic subtypes. RESULTS PAM50 classification of the MPBCS cohort defined 42·6% of tumors as LumA, 21·3% as LumB, 13·3% as HER2E and 16·6% as Basal. Both OSC and DFS for LumA tumors were significantly better than for other subtypes, while Basal tumors had the worst prognosis. While the prognostic power of traditional subtypes calculated with hormone receptors (HR), HER2 and Ki67 determinations showed an acceptable performance, PAM50-derived risk of recurrence best discriminated low, intermediate and high-risk groups. Transcriptomic pathway analysis showed high proliferation (i.e. cell cycle control and DNA damage repair) associated with LumB, HER2E and Basal tumors, and a strong dependency on the estrogen pathway for LumA. Terms related to both innate and adaptive immune responses were seen predominantly upregulated in Basal tumors, and, to a lesser extent, in HER2E, with respect to LumA and B tumors. CONCLUSIONS This is the first study that assesses molecular features at the transcriptomic level in a multicountry Latin American breast cancer patient cohort. Hormone-related and proliferation pathways that predominate in PAM50 and other breast cancer molecular classifications are also the main tumor-driving mechanisms in this cohort and have prognostic power. The immune-related features seen in the most aggressive subtypes may pave the way for therapeutic approaches not yet disseminated in Latin America. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov (Identifier: NCT02326857).
Collapse
Affiliation(s)
- Andrea Sabina Llera
- Molecular and Cellular Therapy Laboratory, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
| | | | - Nora Artagaveytia
- Hospital de Clínicas Manuel Quintela, Universidad de la República, Montevideo, Uruguay
| | | | | | | | - Elsa B. Alcoba
- Hospital Municipal de Oncología María Curie, Buenos Aires, Argentina
| | - Isabel Alonso
- Centro Hospitalario Pereira Rossell, Montevideo, Uruguay
| | - Daniela B. Alves da Quinta
- Molecular and Cellular Therapy Laboratory, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
- Universidad Argentina de la Empresa (UADE), Instituto de Tecnología (INTEC), Buenos Aires, Argentina
| | - Renata Binato
- Bone Marrow Transplantation Unit, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | - Natalia Camejo
- Hospital de Clínicas Manuel Quintela, Universidad de la República, Montevideo, Uruguay
| | - Dirce Maria Carraro
- Laboratory of Genomics and Molecular Biology/Centro Internacional de Pesquisa (CIPE), AC Camargo Cancer Center, Sao Paulo, Brazil
| | - Mónica Castro
- Instituto de Oncología Angel Roffo, Buenos Aires, Argentina
| | | | | | | | - Mauricio Cerda
- Integrative Biology Program, Instituto de Ciencias Biomédicas (ICBM), Centro de Informática Médica y Telemedicina, Facultad de Medicina, Instituto de Neurociencias Biomédicas, Universidad de Chile, Santiago, Chile
| | - Alicia Colombo
- Department of Pathology, Facultad de Medicina y Hospital Clínico, Universidad de Chile, Santiago, Chile
| | - Susanne Crocamo
- Oncology Department, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | | | | | - Lucía Delgado
- Hospital de Clínicas Manuel Quintela, Universidad de la República, Montevideo, Uruguay
| | - Marisa Dreyer-Breitenbach
- Instituto de Biologia Roberto Alcantara Gomes, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Laura Fejerman
- Department of Public Health Sciences and Comprehensive Cancer Center, University of California Davis, Davis, CA, United States
| | - Elmer A. Fernández
- Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas [Centro de Investigación y Desarrollo en Inmunología y Enfermedades Infecciosas (CIDIE) CONICET/Universidad Católica de Córdoba], Córdoba, Argentina
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | | | - Ramón A. Franco-Topete
- Organismo Público Descentralizado (OPD), Hospital Civil de Guadalajara, Universidad de Guadalajara, Guadalajara, Mexico
| | - Carolina Gabay
- Instituto de Oncología Angel Roffo, Buenos Aires, Argentina
| | | | | | - Jorge Gómez
- Texas A&M University, Houston, TX, United States
| | | | - Thomas G. Gross
- Center for Global Health, National Cancer Institute, Rockville, MD, United States
| | | | | | | | | | | | | | - Maria Aparecida Nagai
- Center for Translational Research in Oncology, Cancer Institute of São Paulo (ICESP), Sao Paulo University Medical School, Sao Paulo, Brazil
| | | | | | | | | | - Rui M. Reis
- Molecular Oncology Research Center, Hospital de Câncer de Barretos, Barretos, Brazil
| | - Javier Retamales
- Grupo Oncológico Cooperativo Chileno de Investigación, Santiago, Chile
| | | | - Darío Rocha
- Facultad de Ciencias Exactas, Físicas y Naturales, Universidad Nacional de Córdoba, Córdoba, Argentina
| | | | - Cristina Rosales
- Hospital Municipal de Oncología María Curie, Buenos Aires, Argentina
| | | | | | | | - Juan Martín Sendoya
- Molecular and Cellular Therapy Laboratory, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
| | - Aida A. Silva-García
- Organismo Público Descentralizado (OPD), Hospital Civil de Guadalajara, Universidad de Guadalajara, Guadalajara, Mexico
| | | | | | - Vidya Vedham
- Center for Global Health, National Cancer Institute, Rockville, MD, United States
| | - Livia Zagame
- Instituto Jalisciense de Cancerologia, Guadalajara, Mexico
| | - Osvaldo L. Podhajcer
- Molecular and Cellular Therapy Laboratory, Fundación Instituto Leloir-CONICET, Buenos Aires, Argentina
| |
Collapse
|
25
|
Wu F, Chen Y, Li D, Wang Z, Yu M. Synthesis and Evaluation of Radioiodine-Labeled pH (Low) Insertion Peptide Variant 7-Like Peptide as a Noninvasive Tumor Microenvironment Imaging Agent in a Mouse MDA-MB-231 Triple-Negative Breast Cancer Model. Mol Imaging Biol 2022; 24:570-579. [PMID: 35006491 DOI: 10.1007/s11307-021-01702-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 12/27/2021] [Accepted: 12/28/2021] [Indexed: 10/19/2022]
Abstract
PURPOSE The pH (low) insertion peptide (pHLIP) family can target the tumor microenvironment (TME). If pHLIP can be labeled with radioiodine, the imaging and treatment of tumors can be considered. However, tyrosine and tryptophan can bind with iodine in the insertion region of pHLIP, and radioiodine labeling may affect the formation of α-helix structures in acidic environments; therefore, it is necessary to adjust the structure of pHLIP. This study aims to develop an 125I-labeled pH (low) insertion peptide variant 7-like peptide (pHLIP (Var7) LP) for imaging the TME in MDA-MB-231 triple-negative breast cancer (TNBC) xenograft tumor models. PROCEDURES Based on pHLIP (Var7), a new peptide sequence, pHLIP (Var7) LP, was obtained by the sequence modification method and then characterized. The binding of pHLIP (Var7) LP to MDA-MB-231 cells was analyzed. pHLIP (Var7) LP was labeled with 125I by the iodogen iodination method. Serial biodistribution studies and small-animal single photon emission computed tomography (SPECT)/computed tomography (CT) imaging in subcutaneous MDA-MB-231 TNBC-bearing mice were performed using [125I] I-pHLIP (Var7) LP. RESULTS A novel peptide, pHLIP (Var7) LP, has the characteristics of an α-helix structure, electronegativity, and amphiphilicity. Circular dichroism (CD) spectroscopy showed that the peptide presented a typical pH-dependent transition from an unstructured conformation to an α-helix structure when the pH was reduced from 8.0 to 4.0. The relative fluorescence intensities of 5-carboxytetramethylrhodamine (5-TAMRA)-pHLIP(var7) LP at pH = 6.0, 6.6, and 7.4 were 100.00 ± 5.98%, 72.10 ± 4.65%, and 13.72 ± 1.41%, respectively. The distribution of [125I] I-pHLIP (Var7) LP in tumors reached the highest level (8.7 ± 1.6% ID/g) at 2 h after injection, and the tumor-to-muscle ratios and tumor-to-blood ratios increased with time. Of the measured off-target organs, the stomach, kidney, and bladder showed higher uptake levels. SPECT imaging revealed rapid and sustained tumor uptake of [125I] I-pHLIP (Var7) LP in breast cancer-bearing mice. CONCLUSIONS This study showed that [125I]I-pHLIP (Var7)LP had rapid and sustained tumor uptake in MDA-MB-231 TNBC and provided a new method for TNBC imaging and further treatment.
Collapse
Affiliation(s)
- FengYu Wu
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No.59, Haier St., Laoshan District, Qingdao, 266100, China
| | - YueHua Chen
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No.59, Haier St., Laoshan District, Qingdao, 266100, China
| | - DaCheng Li
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No.59, Haier St., Laoshan District, Qingdao, 266100, China
| | - ZhenGuang Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No.59, Haier St., Laoshan District, Qingdao, 266100, China
| | - MingMing Yu
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No.59, Haier St., Laoshan District, Qingdao, 266100, China.
| |
Collapse
|
26
|
Jian Y, Kong L, Xu H, Shi Y, Huang X, Zhong W, Huang S, Li Y, Shi D, Xiao Y, Yang M, Li S, Chen X, Ouyang Y, Hu Y, Chen X, Song L, Ye R, Wei W. Protein phosphatase 1 regulatory inhibitor subunit 14C promotes triple-negative breast cancer progression via sustaining inactive glycogen synthase kinase 3 beta. Clin Transl Med 2022; 12:e725. [PMID: 35090098 PMCID: PMC8797469 DOI: 10.1002/ctm2.725] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/28/2021] [Accepted: 01/17/2022] [Indexed: 11/21/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is fast-growing and highly metastatic with the poorest prognosis among the breast cancer subtypes. Inactivation of glycogen synthase kinase 3 beta (GSK3β) plays a vital role in the aggressiveness of TNBC; however, the underlying mechanism for sustained GSK3β inhibition remains largely unknown. Here, we find that protein phosphatase 1 regulatory inhibitor subunit 14C (PPP1R14C) is upregulated in TNBC and relevant to poor prognosis in patients. Overexpression of PPP1R14C facilitates cell proliferation and the aggressive phenotype of TNBC cells, whereas the depletion of PPP1R14C elicits opposite effects. Moreover, PPP1R14C is phosphorylated and activated by protein kinase C iota (PRKCI) at Thr73. p-PPP1R14C then represses Ser/Thr protein phosphatase type 1 (PP1) to retain GSK3β phosphorylation at high levels. Furthermore, p-PPP1R14C recruits E3 ligase, TRIM25, toward the ubiquitylation and degradation of non-phosphorylated GSK3β. Importantly, the blockade of PPP1R14C phosphorylation inhibits xenograft tumorigenesis and lung metastasis of TNBC cells. These findings provide a novel mechanism for sustained GSK3β inactivation in TNBC and suggest that PPP1R14C might be a potential therapeutic target.
Collapse
Affiliation(s)
- Yunting Jian
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Pathology, Key Laboratory of Reproduction and Genetics of Guangdong Higher Education Institutes, Key Laboratory for Major Obstetric Diseases of Guangdong ProvinceThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouChina
| | - Lingzhi Kong
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Hongyi Xu
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Yawei Shi
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Xinjian Huang
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Wenjing Zhong
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Shumei Huang
- Department of Biochemistry, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Yue Li
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Dongni Shi
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Yunyun Xiao
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Muwen Yang
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Siqi Li
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| | - Xiangfu Chen
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Ying Ouyang
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Yameng Hu
- Department of Biochemistry, Zhongshan School of MedicineSun Yat‐sen UniversityGuangzhouChina
| | - Xin Chen
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences; Guangzhou Institute of OncologyTumor Hospital, Guangzhou Medical UniversityGuangzhouChina
| | - Libing Song
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
| | - Runyi Ye
- Department of Thyroid and Breast SurgeryThe First Affiliated Hospital of Sun Yat‐sen UniversityGuangzhouChina
| | - Weidong Wei
- Department of Experimental Research, Sun Yat‐sen University Cancer Center, State Key Laboratory of Oncology in South ChinaCollaborative Innovation Center for Cancer MedicineGuangzhouChina
- Department of Breast SurgerySun Yat‐sen University Cancer CenterGuangzhouChina
| |
Collapse
|
27
|
Di Spazio L, Rivano M, Cancanelli L, Chiumente M, Mengato D, Messori A. The Degree of Programmed Death-Ligand 1 (PD-L1) Positivity as a Determinant of Outcomes in Metastatic Triple-Negative Breast Cancer Treated With First-Line Immune Checkpoint Inhibitors. Cureus 2022; 14:e21065. [PMID: 35028245 PMCID: PMC8743568 DOI: 10.7759/cureus.21065] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2022] [Indexed: 12/31/2022] Open
Abstract
In metastatic triple-negative breast cancer (TNBC), the efficacy of immune checkpoint inhibitors (ICIs) in combination with chemotherapy has been demonstrated in randomized clinical trials (RCTs). Despite this, an indirect comparison is not yet available. Reconstruction of individual patient data from Kaplan-Meier curves allows the indirect comparison of different treatments. We analyzed six overall survival (OS) curves from three RCTs. In patients with ≥1% positivity, atezolizumab was found to determine a significantly better OS than pembrolizumab. As regards pembrolizumab, adopting a threshold of PD-L1 positivity ≥10% (as opposed to ≥1%) improved median survival to a remarkable extent (23.0 vs 15.5 months).
Collapse
Affiliation(s)
| | - Melania Rivano
- Clinical Oncology Pharmacy, A. Businco Hospital, Cagliari, ITA
| | - Luca Cancanelli
- Hospital Pharmacy, Azienda Ulss 2 Marca Trevigiana, Treviso, ITA
| | - Marco Chiumente
- Scientific Direction, Italian Society for Clinical Pharmacy and Therapeutics, Milano, ITA
| | - Daniele Mengato
- Hospital Pharmacy, Azienda Ospedale Università di Padova, Padova, ITA
| | - Andrea Messori
- Health Technology Assessment (HTA) Unit, Regione Toscana, Firenze, ITA
| |
Collapse
|
28
|
Zhao W, Liu J, Li Y, Chen Z, Qi D, Zhang Z. Immune Effect of Active Components of Traditional Chinese Medicine on Triple-Negative Breast Cancer. Front Pharmacol 2021; 12:731741. [PMID: 34925002 PMCID: PMC8678494 DOI: 10.3389/fphar.2021.731741] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/18/2021] [Indexed: 12/31/2022] Open
Abstract
Triple-negative breast cancers are heterogeneous, poorly prognostic, and metastatic malignancies that result in a high risk of death for patients. Targeted therapy for triple-negative breast cancer has been extremely challenging due to the lack of expression of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2. Clinical treatment regimens for triple-negative breast cancer are often based on paclitaxel and platinum drugs, but drug resistance and side effects from the drugs frequently lead to treatment failure, thus requiring the development of new therapeutic platforms. In recent years, research on traditional Chinese medicine in modulating the immune function of the body has shown that it has the potential to be an effective treatment option against triple-negative breast cancer. Active components of herbal medicines such as alkaloids, flavonoids, polyphenols, saponins, and polysaccharides have been shown to inhibit cancer cell proliferation and metastasis by activating inflammatory immune responses and can modulate tumor-related signaling pathways to further inhibit the invasion of triple-negative breast cancer. This paper reviews the immunomodulatory mechanisms of different herbal active ingredients against triple-negative breast cancer and provides an outlook on the challenges and directions of development for the treatment of triple-negative breast cancer with herbal active ingredients.
Collapse
Affiliation(s)
- Wenjie Zhao
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- College of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jinhua Liu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yaqun Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zichao Chen
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dongmei Qi
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhen Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
29
|
Vietri MT, D'Elia G, Benincasa G, Ferraro G, Caliendo G, Nicoletti GF, Napoli C. DNA methylation and breast cancer: A way forward (Review). Int J Oncol 2021; 59:98. [PMID: 34726251 DOI: 10.3892/ijo.2021.5278] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 10/01/2021] [Indexed: 11/05/2022] Open
Abstract
The current management of breast cancer (BC) lacks specific non‑invasive biomarkers able to provide an early diagnosis of the disease. Epigenetic‑sensitive signatures are influenced by environmental exposures and are mediated by direct molecular mechanisms, mainly guided by DNA methylation, which regulate the interplay between genetic and non‑genetic risk factors during cancerogenesis. The inactivation of tumor suppressor genes due to promoter hypermethylation is an early event in carcinogenesis. Of note, targeted tumor suppressor genes are frequently hypermethylated in patient‑derived BC tissues and peripheral blood biospecimens. In addition, epigenetic alterations in triple‑negative BC, as the most aggressive subtype, have been identified. Thus, detecting both targeted and genome‑wide DNA methylation changes through liquid‑based assays appears to be a useful clinical strategy for early detection, more accurate risk stratification and a personalized prediction of therapeutic response in patients with BC. Of note, the DNA methylation profile may be mapped by isolating the circulating tumor DNA from the plasma as a more accessible biospecimen. Furthermore, the sensitivity to treatment with chemotherapy, hormones and immunotherapy may be altered by gene‑specific DNA methylation, suggesting novel potential drug targets. Recently, the use of epigenetic drugs administered alone and/or with anticancer therapies has led to remarkable results, particularly in patients with BC resistant to anticancer treatment. The aim of the present review was to provide an update on DNA methylation changes that are potentially involved in BC development and their putative clinical utility in the fields of diagnosis, prognosis and therapy.
Collapse
Affiliation(s)
- Maria Teresa Vietri
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', I-80138 Naples, Italy
| | - Giovanna D'Elia
- Unit of Clinical and Molecular Pathology, AOU, University of Campania 'Luigi Vanvitelli', I-80138 Naples, Italy
| | - Giuditta Benincasa
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania 'Luigi Vanvitelli', I-80138 Naples, Italy
| | - Giuseppe Ferraro
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, Plastic Surgery Unit, University of Campania 'Luigi Vanvitelli', I-80138 Naples, Italy
| | - Gemma Caliendo
- Unit of Clinical and Molecular Pathology, AOU, University of Campania 'Luigi Vanvitelli', I-80138 Naples, Italy
| | - Giovanni Francesco Nicoletti
- Multidisciplinary Department of Medical, Surgical and Dental Sciences, Plastic Surgery Unit, University of Campania 'Luigi Vanvitelli', I-80138 Naples, Italy
| | - Claudio Napoli
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania 'Luigi Vanvitelli', I-80138 Naples, Italy
| |
Collapse
|
30
|
Ma L, Diao L, Peng Z, Jia Y, Xie H, Li B, Ma J, Zhang M, Cheng L, Ding D, Zhang X, Chen H, Mo F, Jiang H, Xu G, Meng F, Zhong Z, Liu M. Immunotherapy and Prevention of Cancer by Nanovaccines Loaded with Whole-Cell Components of Tumor Tissues or Cells. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2104849. [PMID: 34536044 DOI: 10.1002/adma.202104849] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/12/2021] [Indexed: 06/13/2023]
Abstract
Tumor tissues/cells are the best sources of antigens to prepare cancer vaccines. However, due to the difficulty of solubilization and delivery of water-insoluble antigens in tumor tissues/cells, including water-insoluble antigens into cancer vaccines and delivering such vaccines efficiently to antigen-presenting cells (APCs) remain challenging. To solve these problems, herein, water-insoluble components of tumor tissues/cells are solubilized by 8 m urea and thus whole components of micrometer-sized tumor cells are reasssembled into nanosized nanovaccines. To induce maximized immunization efficacy, various antigens are loaded both inside and on the surface of nanovaccines. By encapsulating both water-insoluble and water-soluble components of tumor tissues/cells into nanovaccines, the nanovaccines are efficiently phagocytosed by APCs and showed better therapeutic efficacy than the nanovaccine loaded with only water-soluble components in melanoma and breast cancer. Anti-PD-1 antibody and metformin can improve the efficacy of nanovaccines. In addition, the nanovaccines can prevent lung cancer (100%) and melanoma (70%) efficiently in mice. T cell analysis and tumor microenvironment analysis indicate that tumor-specific T cells are induced by nanovaccines and both adaptive and innate immune responses against cancer cells are activated by nanovaccines. Overall, this study demonstrates a universal method to make tumor-cell-based nanovaccines for cancer immunotherapy and prevention.
Collapse
Affiliation(s)
- Lin Ma
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Lu Diao
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Zuofu Peng
- Alpha X (Beijing) Biotech Co., Ltd., Beijing, 102600, P. R. China
| | - Yun Jia
- Alpha X (Beijing) Biotech Co., Ltd., Beijing, 102600, P. R. China
| | - Huimin Xie
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Baisong Li
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Jianting Ma
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Meng Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Lifang Cheng
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Dawei Ding
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Xuenong Zhang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Huabing Chen
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Fengfeng Mo
- Department of Naval Nutrition and Food Hygiene, Faculty of Naval Medicine, Naval Medical University, Shanghai, 200433, P. R. China
| | - Honglv Jiang
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Guoqiang Xu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| | - Fenghua Meng
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Zhiyuan Zhong
- Biomedical Polymers Laboratory, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou, 215123, P. R. China
| | - Mi Liu
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, P. R. China
| |
Collapse
|
31
|
A computational approach on studying the regulation of TGF-β1-stimulated Runx2 expression by MicroRNAs in human breast cancer cells. Comput Biol Med 2021; 137:104823. [PMID: 34492519 DOI: 10.1016/j.compbiomed.2021.104823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 08/15/2021] [Accepted: 08/29/2021] [Indexed: 01/20/2023]
Abstract
BACKGROUND Transforming growth factor-beta1 (TGF-β1) acts as a most effective growth inhibitor for normal epithelial cells. Loss of this anti-proliferative factor in breast tissues favors invasion and development of osteolytic metastases, aided by a master transcription factor, runt-related transcription factor 2 (Runx2). Several reports identified Runx2 regulation with the help of non-coding RNAs such as microRNAs (miRNAs) under physiological and pathological conditions. METHODS Using bioinformatics tools such as miRDB, STarMir, Venny, TarBase, a unique list of miRNAs that putatively target the 3' UTR Runx2 was identified. Further, the expression patterns of those miRNAs at the precursor and mature levels were studied by RT-qPCR analyses. Following this, computational analyses using software like TransmiR and bc-GenExMiner v4.6 were done to speculate the miRNA's other target genes that indirectly regulate Runx2 activity in breast cancer. RESULTS There were 13 miRNAs that putatively target Runx2 identified using bioinformatics tools. Among these miRNAs, miR-5703 expression was significantly downregulated at both precursor and mature levels upon TGF-β1-treatment in human breast cancer cells. Computational analyses speculated an indirect targeting of Runx2 by miR-5703 by influencing multiple Runx2 regulatory signaling pathways including Jak/Stat, MAPK, Wnt/β-Catenin, Notch, BMP, and PKA pathways. Furthermore, a correlation of the expression profiles of the speculated genes and Runx2 with miR-5703 was depicted in triple-negative breast cancer patients. CONCLUSION Identification of miR-5703 and its network for Runx2 regulation directly or indirectly in breast cancer cells could significantly advance our understanding of breast cancer-mediated bone metastasis. In addition, it would potentially pave the way for miRNAs to be used as biomarkers and therapeutic agents in cancer research.
Collapse
|
32
|
Ming L, Song L, Xu J, Wang R, Shi J, Chen M, Zhang Y. Smart Manganese Dioxide-Based Lanthanide Nanoprobes for Triple-Negative Breast Cancer Precise Gene Synergistic Chemodynamic Therapy. ACS APPLIED MATERIALS & INTERFACES 2021; 13:35444-35455. [PMID: 34292714 DOI: 10.1021/acsami.1c08927] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Small interfering RNA (siRNA)-based gene therapy has been widely studied as a promising treatment for malignant triple-negative breast cancer (TNBC), but efficient delivery of siRNA still remains a challenge. In this study, a smart manganese dioxide (MnO2)-based lanthanide nanoprobe therapeutic nanoplatform (ErNPs@MnO2-siS100A4-RGD) was developed for tumor imaging and precise stimuli-responsive S100A4 siRNA (siS100A4)-mediated gene therapy in synergism with chemodynamic therapy (CDT) of TNBC. ErNPs@MnO2-siS100A4-RGD has a tumor microenvironment-responsive capability attributed to the presence of MnO2, which can be degraded by glutathione (GSH) in the tumor region while releasing siRNA and generating Mn2+ to achieve precise gene therapy and a Fenton-like reaction-mediated CDT effect on TNBC. Subsequently, the lanthanide nanoprobes (ErNPs) are exposed to the second near-infrared region (NIR-II) fluorescence emission to realize the precise tumor location. Both the in vitro and in vivo results demonstrated that the smart nanoplatform possessed high siRNA delivery efficiency and GSH-responsive precise siRNA releasing ability, and compared with individual gene therapy, the GSH-depletion-enhanced CDT effect further reinforced TNBC inhibition, demonstrating excellent GSH-responsive-enhanced NIR-II precise tumor imaging therapy. These results indicate that the nanoplatform provides a crucial foundation for further research on theranostic systems of TNBC.
Collapse
Affiliation(s)
- Liyan Ming
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, P. R. China
| | - Liang Song
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, P. R. China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Jiangxi 341000, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jixuan Xu
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, P. R. China
| | - Ruoping Wang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Junpeng Shi
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, P. R. China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Jiangxi 341000, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Min Chen
- Clinical Central Research Core, Xiangan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, P. R. China
| | - Yun Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, P. R. China
- Xiamen Key Laboratory of Rare Earth Photoelectric Functional Materials, Xiamen Institute of Rare Earth Materials, Haixi Institute, Chinese Academy of Sciences, Xiamen 361021, P. R. China
- Ganjiang Innovation Academy, Chinese Academy of Sciences, Jiangxi 341000, P. R. China
- University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
33
|
Busheri L, Dixit S, Nare S, Alhat R, Thomas G, Jagtap M, Navgire R, Shinde P, Banale R, Unde R, Reddy R, Shaikh S, Konnur A, Namewar N, Bapat A, Patil A, Johari R, Kushwaha R, Kumari W, Varghese B, Deshpande P, Deshmukh C, Kelkar DA, Shashidhara LS, Koppiker CB, Kulkarni M. Breast cancer biobank from a single institutional cohort in an urban setting in india: Tumor characteristics and survival outcomes. Cancer Treat Res Commun 2021; 28:100409. [PMID: 34098400 DOI: 10.1016/j.ctarc.2021.100409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/20/2021] [Accepted: 05/25/2021] [Indexed: 12/24/2022]
Abstract
BACKGROUND A breast cancer biobank with retrospectively collected patient data and FFPE tissue samples was established in 2018 at Prashanti Cancer Care Mission, Pune, India. It runs a cancer care clinic with support from a single surgeon's breast cancer practice. The clinical data and tissue sample collection is undertaken with appropriate patient consent following ethical approval and guidelines. METHODS The biobank holds clinical history, diagnostic reports, treatment and follow-up information along with FFPE tumor tissue specimens, adjacent normal and, in few cases, contralateral normal breast tissue. Detailed family history and germline mutational profiles of eligible and consenting patients and their relatives are also deposited in the biobank. RESULTS Here, we report the first audit of the biobank. A total number of 994 patients with breast disease have deposited consented clinical records in the biobank. The majority of the records (80%, n = 799) are of patients with infiltrating ductal carcinoma (IDC). Of 799 IDC patients, 434 (55%) have deposited tumor tissue in the biobank with consent. In addition, germline mutation profiles of 84 patients and their family members are deposited. Follow-up information is available for 85% of the 434 IDC patients with an average follow-up of 3 years. CONCLUSION The biobank has aided the initiation of translational research at our center in collaboration with eminent institutes like IISER Pune and SJRI Bangalore to evaluate profiles of breast cancer in an Indian cohort. The biobank will be a valuable resource to the breast cancer research community, especially to understand South Asian profiles of breast cancer.
Collapse
Affiliation(s)
| | - Santosh Dixit
- Prashanti Cancer Care Mission, Pune; Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and IISER Pune
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Ankita Patil
- Prashanti Cancer Care Mission, Pune; Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and IISER Pune
| | | | - Roli Kushwaha
- Prashanti Cancer Care Mission, Pune; Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and IISER Pune
| | | | | | | | | | - Devaki A Kelkar
- Prashanti Cancer Care Mission, Pune; Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and IISER Pune
| | - L S Shashidhara
- Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and IISER Pune; Ashoka University, Sonipat, Delhi
| | - Chaitanyanand B Koppiker
- Prashanti Cancer Care Mission, Pune; Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and IISER Pune
| | - Madhura Kulkarni
- Prashanti Cancer Care Mission, Pune; Center for Translational Cancer Research, a Joint venture between Prashanti Cancer Care Mission and IISER Pune.
| |
Collapse
|
34
|
Mitochondria-targeted high-load sound-sensitive micelles for sonodynamic therapy to treat triple-negative breast cancer and inhibit metastasis. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 124:112054. [PMID: 33947548 DOI: 10.1016/j.msec.2021.112054] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/24/2022]
Abstract
Breast cancer is the most common cancer among women worldwide, of which 10-20% accounts for triple-negative breast cancer (TNBC). TNBC is more aggressive, lacks an effective treatment target, and has a higher metastasis rate compared to other types of breast cancers. These characteristics result in poor therapeutic and prognostic outcomes in patients with TNBC. Sonodynamic therapy (SDT) is an emerging non-invasive procedure with high-tissue penetration properties to treat cancer. Therefore, we designed a new sonosensitizer, PEG-IR780@Ce6 for SDT, which showed excellent performance in inhibiting cancer cells and in simultaneously suppressing the migration and invasion of cancer cells. In vitro and in vivo experiments showed that PEG-IR780@Ce6 as a sonosensitizer could generate higher levels of reactive oxygen species (ROS) than IR780 and free Ce6 alone, thereby resulting in better anti-cancer effects. Besides, PEG-IR780@Ce6 inhibited the migration and invasion of MDA-MB-231 cells, both in vitro and in vivo, which indicated that it could suppress the metastasis of TNBC. Moreover, the long circulation time and the mitochondria-targeting ability of PEG-IR780@Ce6 guaranteed its accumulation in the tumor. In addition, both in vitro and in vivo experiments indicated the biocompatibility and biosafety of PEG-IR780@Ce6. In conclusion, our results collectively suggested that the newly designed sonosensitizer, PEG-IR780@Ce6, is a promising treatment option for TNBC with excellent therapeutic effects and low side effects.
Collapse
|
35
|
Singh S, Numan A, Maddiboyina B, Arora S, Riadi Y, Md S, Alhakamy NA, Kesharwani P. The emerging role of immune checkpoint inhibitors in the treatment of triple-negative breast cancer. Drug Discov Today 2021; 26:1721-1727. [PMID: 33745879 DOI: 10.1016/j.drudis.2021.03.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/16/2021] [Accepted: 03/14/2021] [Indexed: 12/13/2022]
Abstract
Triple-negative breast cancers (TNBCs) form a heterogeneous group of breast carcinomas that lack expression of estrogen receptor, progesterone receptor and epidermal growth factor receptor 2. In the past decade, immune checkpoint inhibitors (ICIs) have revolutionized the arena of cancer immunotherapy. Early results are now accumulating from trials involving the treatment of TNBCs with radical ICIs therapies, including combinational therapies that include ICI technologies. In this review, we provide a broad overview of the progress of immunotherapy-based treatments and discuss future opportunities for their use in TNBC.
Collapse
Affiliation(s)
- Sima Singh
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab 140413, India
| | - Arshid Numan
- State Key Laboratory of ASIC and System, SIST, Fudan University, 200433 Shanghai, China
| | - Balaji Maddiboyina
- Department of Pharmacy, NRK & KSR Gupta College of Pharmacy, Tenali, Guntur, 522202 AP, India
| | - Saahil Arora
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab 140413, India.
| | - Yassine Riadi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Shadab Md
- Deptartment of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Nabil A Alhakamy
- Deptartment of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
36
|
Methylation-dependent MCM6 repression induced by LINC00472 inhibits triple-negative breast cancer metastasis by disturbing the MEK/ERK signaling pathway. Aging (Albany NY) 2021; 13:4962-4975. [PMID: 33668040 PMCID: PMC7950301 DOI: 10.18632/aging.103568] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 04/17/2020] [Indexed: 12/15/2022]
Abstract
Long noncoding RNAs (lncRNAs) have been identified to be dysregulated in multiple cancer types, which are speculated to be of vital significance in regulating several hallmarks of cancer biology. Triple-negative breast cancer (TNBC) is acknowledged as an aggressive subtype of breast cancer. In this study, we found the lncRNA LINC00472 was poorly expressed in TNBC tissues and cells. Overexpression of LINC00472 could inhibit the proliferation, invasion and migration of MDA-MB-231 cells. On the contrary, minichromosome maintenance complex component 6 (MCM6) was highly expressed in TNBC tissues and MDA-MB-231 cells due to suppressed methylation. LINC00472 induced site-specific DNA methylation and reduced the MCM6 expression by recruiting DNA methyltransferases into the MCM6 promoter. Since the restoration of MCM6 weakened the tumor-suppressive effect of LINC00472 on MDA-MB-231 cells, LINC00472 potentially acted as a tumor suppressor by inhibiting MCM6. In addition, in vivo experiments further substantiated that overexpression of LINC00472 inhibited tumor growth and metastasis to lungs by decreasing the expression of MCM6. Overall, the present study demonstrated that LINC00472-mediated epigenetic silencing of MCM6 contributes to the prevention of tumorigenesis and metastasis in TNBC, providing an exquisite therapeutic target for TNBC.
Collapse
|
37
|
Aine M, Boyaci C, Hartman J, Häkkinen J, Mitra S, Campos AB, Nimeus E, Ehinger A, Vallon-Christersson J, Borg Å, Staaf J. Molecular analyses of triple-negative breast cancer in the young and elderly. Breast Cancer Res 2021; 23:20. [PMID: 33568222 PMCID: PMC7874480 DOI: 10.1186/s13058-021-01392-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/11/2021] [Indexed: 01/09/2023] Open
Abstract
Background Breast cancer in young adults has been implicated with a worse outcome. Analyses of genomic traits associated with age have been heterogenous, likely because of an incomplete accounting for underlying molecular subtypes. We aimed to resolve whether triple-negative breast cancer (TNBC) in younger versus older patients represent similar or different molecular diseases in the context of genetic and transcriptional subtypes and immune cell infiltration. Patients and methods In total, 237 patients from a reported population-based south Swedish TNBC cohort profiled by RNA sequencing and whole-genome sequencing (WGS) were included. Patients were binned in 10-year intervals. Complimentary PD-L1 and CD20 immunohistochemistry and estimation of tumor-infiltrating lymphocytes (TILs) were performed. Cases were analyzed for differences in patient outcome, genomic, transcriptional, and immune landscape features versus age at diagnosis. Additionally, 560 public WGS breast cancer profiles were used for validation. Results Median age at diagnosis was 62 years (range 26–91). Age was not associated with invasive disease-free survival or overall survival after adjuvant chemotherapy. Among the BRCA1-deficient cases (82/237), 90% were diagnosed before the age of 70 and were predominantly of the basal-like subtype. In the full TNBC cohort, reported associations of patient age with changes in Ki67 expression, PIK3CA mutations, and a luminal androgen receptor subtype were confirmed. Within DNA repair deficiency or gene expression defined molecular subgroups, age-related alterations in, e.g., overall gene expression, immune cell marker gene expression, genetic mutational and rearrangement signatures, amount of copy number alterations, and tumor mutational burden did, however, not appear distinct. Similar non-significant associations for genetic alterations with age were obtained for other breast cancer subgroups in public WGS data. Consistent with age-related immunosenescence, TIL counts decreased linearly with patient age across different genetic TNBC subtypes. Conclusions Age-related alterations in TNBC, as well as breast cancer in general, need to be viewed in the context of underlying genomic phenotypes. Based on this notion, age at diagnosis alone does not appear to provide an additional layer of biological complexity above that of proposed genetic and transcriptional phenotypes of TNBC. Consequently, treatment decisions should be less influenced by age and more driven by tumor biology. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-021-01392-0.
Collapse
Affiliation(s)
- Mattias Aine
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Ceren Boyaci
- Department of Clinical Pathology and Cytology, Karolinska University Laboratory, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology and Pathology, Karolinska Institute, Stockholm, Sweden
| | - Jari Häkkinen
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Shamik Mitra
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Ana Bosch Campos
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Emma Nimeus
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden.,Division of Surgery, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Anna Ehinger
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden.,Department of Genetics and Pathology, Laboratory Medicine, Region Skåne, Lund, Sweden
| | - Johan Vallon-Christersson
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Åke Borg
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden
| | - Johan Staaf
- Division of Oncology, Department of Clinical Sciences Lund, Lund University, Medicon Village, SE-22381, Lund, Sweden.
| |
Collapse
|
38
|
Liu J, Wang S, Wang C, Kong X, Sun P. Prognostic value of using glucosylceramide synthase and cytochrome P450 family 1 subfamily A1 expression levels for patients with triple-negative breast cancer following neoadjuvant chemotherapy. Exp Ther Med 2021; 21:247. [PMID: 33603855 DOI: 10.3892/etm.2021.9678] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022] Open
Abstract
Neoadjuvant chemotherapy (NACT) has been considered to be the preferred treatment option for early operable triple-negative breast cancer (TNBC). However, resistance to drugs remains to be the barrier to the efficacy of NACT. Glucosylceramide synthase (GCS) and cytochrome P450 family 1 subfamily A1 (CYP1A1) have been previously associated with drug resistance in breast cancer. The present study aimed to explore whether the expression levels of GCS and/or CYP1A1 are associated with the prognosis of TNBC after NACT. Immunohistochemistry was used to detect and measure GCS and CYP1A1 expression. Associations between GCS or CYP1A1 expression and the clinicopathological characteristics, pathological complete response (pCR), clinical complete response (cCR) and disease-free survival (DFS) were analyzed. GCS expression was found to be associated with tumor size (P=0.021) and TNM staging (P=0.042), whilst CYP1A1 expression was associated with lymph node metastasis (P = 0.026) and TNM staging (P=0.034). The expression levels of GCS (P=0.024) and CYP1A1 (P=0.027) were upregulated after NACT. GCS and CYP1A1 expression were positively correlated (P=0.003; r=0.327). No difference was observed between the GCS+ (P=0.188) or CYP1A1+ group (P=0.073) and the GCS- or CYP1A1- group in terms of pCR. However, compared with that in the GCS+CYP1A1+ group, the pCR was markedly increased in the GCS-CYP1A1- group (P=0.031). The cCR was lower in the GCS+ (P=0.021) and CYP1A1+ groups (P=0.016) compared with in the GCS- or CYP1A1- group. The DFS rate (57.9 vs. 65.4%; P=0.049) was lower in the GCS+CYP1A1+ group compared with that in the GCS-CYP1A1- group. However, there was no statistical significance after P-value was adjusted for multiple comparisons using Bonferroni correction. In conclusion, co-expression of GCS and CYP1A1 was associated with pCR and DFS in TNBC, which may serve a role in the prediction of the prognosis of patients with TNBC following treatment with NACT.
Collapse
Affiliation(s)
- Jiannan Liu
- Department of Oncology, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Shuhua Wang
- Department of Medical Record Information, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Congcong Wang
- Department of Oncology, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Xiangshuo Kong
- Department of Oncology, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| | - Ping Sun
- Department of Oncology, Yantai Yuhuangding Hospital, Yantai, Shandong 264000, P.R. China
| |
Collapse
|
39
|
Chondroitin sulphate and α-tocopheryl succinate tethered multiwalled carbon nanotubes for dual-action therapy of triple-negative breast cancer. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.102080] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
40
|
Shen M, Pan H, Chen Y, Xu YH, Yang W, Wu Z. A review of current progress in triple-negative breast cancer therapy. Open Med (Wars) 2020; 15:1143-1149. [PMID: 33336070 PMCID: PMC7718625 DOI: 10.1515/med-2020-0138] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 09/19/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a particularly aggressive subtype known for its extremely high drug resistance, progression, poor prognosis, and lack of clear therapeutic targets. Researchers are aiming to advance TNBC treatment worldwide. In the past 2–3 years, more positive results have emerged in the clinical research on TNBC treatment. Based on the results, several impressive drugs have been approved to benefit patients with TNBC, including the PARP inhibitors olaparib and talazoparib for germline BRCA mutation-associated breast cancer (gBRCAm-BC) and immunotherapy using the checkpoint inhibitor atezolizumab in combination with nab-paclitaxel for programmed cell death-ligand 1-positive (PD-L1+) advanced TNBC. Although neoadjuvant therapy has focused on combinations of systemic agents to optimize pathologically complete response, metastatic TNBC still has a poor prognosis. Innovative multidrug combination systemic therapies based on neoadjuvants and adjuvants have led to significant improvements in outcomes, particularly over the past decade.
Collapse
Affiliation(s)
- Meiying Shen
- Department of Mammary Gland, Maoming People's Hospital, Maoming, 525000, China
| | - Huawen Pan
- Department of Spinal, Maoming People's Hospital, Maoming, 525000, China
| | - Yuxia Chen
- Department of Mammary Gland, Maoming People's Hospital, Maoming, 525000, China
| | - Yu Hang Xu
- Department of Mammary Gland, Maoming People's Hospital, Maoming, 525000, China
| | - Weixiong Yang
- Department of Mammary Gland, Maoming People's Hospital, Maoming, 525000, China
| | - Zhaojun Wu
- Department of Ultrasound, Maoming People's Hospital, Maoming, 525000, China
| |
Collapse
|
41
|
De Palma FDE, Del Monaco V, Pol JG, Kremer M, D’Argenio V, Stoll G, Montanaro D, Uszczyńska-Ratajczak B, Klein CC, Vlasova A, Botti G, D’Aiuto M, Baldi A, Guigó R, Kroemer G, Maiuri MC, Salvatore F. The abundance of the long intergenic non-coding RNA 01087 differentiates between luminal and triple-negative breast cancers and predicts patient outcome. Pharmacol Res 2020; 161:105249. [DOI: 10.1016/j.phrs.2020.105249] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 10/08/2020] [Accepted: 10/08/2020] [Indexed: 02/07/2023]
|
42
|
Yao H, Zhang X, Zhang N, Li J, Li Y, Wei Q. Wikstromol from Wikstroemia indica induces apoptosis and suppresses migration of MDA-MB-231 cells via inhibiting PI3K/Akt pathway. J Nat Med 2020; 75:178-185. [PMID: 32865667 DOI: 10.1007/s11418-020-01447-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/23/2020] [Indexed: 11/24/2022]
Abstract
Triple negative breast cancer (TNBC) is the most severe type of breast cancer due to the lack of specific targets and rapid metastasis, which result in the poor prognosis. Recently, phosphatidylinositol 3-kinase (PI3K)/Akt pathway has emerged as a potential target for the treatment of TNBC. In our research interest to discover phytochemicals targeting TNBC, we have investigated wikstromol from Wikstroemia indica using the human TNBC MDA-MB-231 cells. The results showed wikstromol at 10 μM inhibited cell growth of MDA-MB-231 cells which was confirmed by MTT assay. Further DAPI staining has revealed wikstromol at 10 μM induced apoptosis of cancer cells, which was associated with the activation of caspase-3 following down-regulation of Bcl-2 as well as up-regulation of Bax, cleaved PARP and phosphorylated p53. Meanwhile, it was observed at 0.1 μM wikstromol suppressed the migration of the cancer cells via decreasing transcription of NF-κB and reducing activity and secretion of downstream MMP-9. In addition, p-PI3K and p-Akt were down-regulated in MDA-MB-231 cells in the presence of wikstromol at 0.1 μM, which indicated inactivation of PI3K/Akt pathway was involved in these inhibitory effects.
Collapse
Affiliation(s)
- Huankai Yao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Xiuli Zhang
- Shanxian Central Hospital, Heze, 274300, Shandong, China
| | - Nan Zhang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Jindong Li
- Department of Pharmacy, The Hospital Affiliated to Nanjing University of Traditional Chinese Medicine (Taizhou People's Hospital), Taizhou, 225300, Jiangsu, China
| | - Yan Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| | - Qunli Wei
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, School of Pharmacy, Xuzhou Medical University, No. 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| |
Collapse
|
43
|
Riaz N, Idress R, Habib S, Lalani EN. Lack of Androgen Receptor Expression Selects for Basal-Like Phenotype and Is a Predictor of Poor Clinical Outcome in Non-Metastatic Triple Negative Breast Cancer. Front Oncol 2020; 10:1083. [PMID: 32850312 PMCID: PMC7399239 DOI: 10.3389/fonc.2020.01083] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Accepted: 05/29/2020] [Indexed: 12/12/2022] Open
Abstract
Background: Androgen receptor (AR) has emerged as a significant favorable prognostic indicator in estrogen receptor expressing (ER+) breast cancer (BCa); however, its clinical and biological relevance in triple negative breast cancer (TNBC) and association with cancer stem cell (CSC) markers remain ambiguous. Methods: We examined the immunohistochemical expression of AR in a cohort of stage I-III TNBC cases (n = 197) with a long-term clinical follow-up data (mean follow-up = 53.6 months). Significance of AR expression was correlated with prognostic biomarkers including cancer stem cell markers (CD44, CD24, and ALDH1), basal markers (CK5, CK14, and nestin), proliferation marker (ki-67), apoptotic marker (Bcl-2), and COX-2. Expression of CK5 and nestin was used for the categorization of TNBC into basal (TN, CK5+, and/or nestin+) and non-basal (TN, CK5-, and/or nestin-) phenotypes, and Kaplan-Meier curves were used for estimation of overall survival and breast cancer-specific survival (BCSS). Results: AR expression was observed in 18.8% of non-metastatic TNBC tumors. Expression of AR correlated with lower grade (P < 0.001) and conferred a favorable prognostic significance in patients with axillary lymph node metastasis (P = 0.005). Lack of AR expression correlated with expression of CSC phenotype (CD44+/CD24-) (P < 0.001), COX-2 (P = 0.02), basal markers (CK5: P = 0.03), and nestin (P = 0.01). Basal-like phenotype (TN, CK5+, and/or nestin+) correlated with quadruple-negative breast cancer (QNBC) and showed a significant association with adverse prognostic markers including high proliferation index (P < 0.001), expression of COX-2 (P = 0.009), and CSC phenotype (CD44+/CD24-: P = 0.01). Expression of AR remained an independent prognostic indicator for improved overall survival (P = 0.003), whereas basal-like phenotype was associated with an adverse BCSS (P = 0.013). Conclusions: Assessment of AR and basal markers identified biologically and clinically distinct subgroups of TNBC. Expression of AR defined a low-risk TNBC subgroup associated with improved overall survival, whereas expression of basal markers (CK5 and nestin) identified a high-risk subgroup associated with adverse BCSS. Integration of immunohistochemical analysis of AR and basal biomarkers to the assessment of TNBC tumors is expected to improve the prognostication of an otherwise heterogeneous disease.
Collapse
Affiliation(s)
- Nazia Riaz
- Centre for Regenerative Medicine and Stem Cell Research, Aga Khan University, Karachi, Pakistan.,Section of Breast Diseases, Department of Surgery, Aga Khan University, Karachi, Pakistan
| | - Romana Idress
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, Pakistan
| | - Sadia Habib
- Section of Breast Diseases, Department of Surgery, Aga Khan University, Karachi, Pakistan
| | - El-Nasir Lalani
- Centre for Regenerative Medicine and Stem Cell Research, Aga Khan University, Karachi, Pakistan.,Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, Pakistan
| |
Collapse
|
44
|
Merrill NM, Lachacz EJ, Vandecan NM, Ulintz PJ, Bao L, Lloyd JP, Yates JA, Morikawa A, Merajver SD, Soellner MB. Molecular determinants of drug response in TNBC cell lines. Breast Cancer Res Treat 2020; 179:337-347. [PMID: 31655920 PMCID: PMC7323911 DOI: 10.1007/s10549-019-05473-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 10/10/2019] [Indexed: 12/17/2022]
Abstract
PURPOSE There is a need for biomarkers of drug efficacy for targeted therapies in triple-negative breast cancer (TNBC). As a step toward this, we identify multi-omic molecular determinants of anti-TNBC efficacy in cell lines for a panel of oncology drugs. METHODS Using 23 TNBC cell lines, drug sensitivity scores (DSS3) were determined using a panel of investigational drugs and drugs approved for other indications. Molecular readouts were generated for each cell line using RNA sequencing, RNA targeted panels, DNA sequencing, and functional proteomics. DSS3 values were correlated with molecular readouts using a FDR-corrected significance cutoff of p* < 0.05 and yielded molecular determinant panels that predict anti-TNBC efficacy. RESULTS Six molecular determinant panels were obtained from 12 drugs we prioritized based on their efficacy. Determinant panels were largely devoid of DNA mutations of the targeted pathway. Molecular determinants were obtained by correlating DSS3 with molecular readouts. We found that co-inhibiting molecular correlate pathways leads to robust synergy across many cell lines. CONCLUSIONS These findings demonstrate an integrated method to identify biomarkers of drug efficacy in TNBC where DNA predictions correlate poorly with drug response. Our work outlines a framework for the identification of novel molecular determinants and optimal companion drugs for combination therapy based on these correlates.
Collapse
Affiliation(s)
- Nathan M Merrill
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Eric J Lachacz
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Nathalie M Vandecan
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Peter J Ulintz
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Liwei Bao
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - John P Lloyd
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Joel A Yates
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Aki Morikawa
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA
| | - Sofia D Merajver
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA.
| | - Matthew B Soellner
- Department of Internal Medicine, University of Michigan, 1500 Medical Center Dr, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
45
|
Maggisano V, Celano M, Malivindi R, Barone I, Cosco D, Mio C, Mignogna C, Panza S, Damante G, Fresta M, Andò S, Russo D, Catalano S, Bulotta S. Nanoparticles Loaded with the BET Inhibitor JQ1 Block the Growth of Triple Negative Breast Cancer Cells In Vitro and In Vivo. Cancers (Basel) 2019; 12:cancers12010091. [PMID: 31905936 PMCID: PMC7016573 DOI: 10.3390/cancers12010091] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/20/2019] [Accepted: 12/24/2019] [Indexed: 12/24/2022] Open
Abstract
Inhibition of bromo-and extra-terminal domain (BET) proteins, epigenetic regulators of genes involved in cell viability, has been efficiently tested in preclinical models of triple negative breast cancer (TNBC). However, the use of the selective BET-inhibitor JQ1 on humans is limited by its very short half-life. Herein, we developed, characterized and tested a novel formulation of nanoparticles containing JQ1 (N-JQ1) against TNBC in vitro and in vivo. N-JQ1, prepared using the nanoprecipitation method of preformedpoly-lactid-co-glycolic acid in an aqueous solution containing JQ1 and poloxamer-188 as a stabilizer, presented a high physico-chemical stability. Treatment of MDA-MB 157 and MDA-MB 231 TNBC cells with N-JQ1 determined a significant decrease in cell viability, adhesion and migration. Intra-peritoneal administration (5 days/week for two weeks) of N-JQ1 in nude mice hosting a xenograft TNBC after flank injection of MDA-MB-231 cells determined a great reduction in the growth and vascularity of the neoplasm. Moreover, the treatment resulted in a minimal infiltration of nearby tissues. Finally, the encapsulation of JQ1 in nanoparticles improved the anticancer efficacy of this epigenetic compound against TNBC in vitro and in vivo, opening the way to test it in the treatment of TNBC.
Collapse
Affiliation(s)
- Valentina Maggisano
- Department of Health Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (V.M.); (M.C.); (D.C.); (M.F.); (S.B.)
| | - Marilena Celano
- Department of Health Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (V.M.); (M.C.); (D.C.); (M.F.); (S.B.)
| | - Rocco Malivindi
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Cosenza, Italy; (R.M.); (I.B.); (S.P.); (S.A.)
| | - Ines Barone
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Cosenza, Italy; (R.M.); (I.B.); (S.P.); (S.A.)
| | - Donato Cosco
- Department of Health Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (V.M.); (M.C.); (D.C.); (M.F.); (S.B.)
| | - Catia Mio
- Department of Medical Area, University of Udine, 33100 Udine, Italy; (C.M.); (G.D.)
| | - Chiara Mignogna
- Interdepartmental Service Center, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy;
| | - Salvatore Panza
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Cosenza, Italy; (R.M.); (I.B.); (S.P.); (S.A.)
| | - Giuseppe Damante
- Department of Medical Area, University of Udine, 33100 Udine, Italy; (C.M.); (G.D.)
| | - Massimo Fresta
- Department of Health Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (V.M.); (M.C.); (D.C.); (M.F.); (S.B.)
| | - Sebastiano Andò
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Cosenza, Italy; (R.M.); (I.B.); (S.P.); (S.A.)
| | - Diego Russo
- Department of Health Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (V.M.); (M.C.); (D.C.); (M.F.); (S.B.)
- Correspondence: (D.R.); (S.C.); Tel.: +39-09613694224 (D.R.); +39-0984496207 (S.C.)
| | - Stefania Catalano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Cosenza, Italy; (R.M.); (I.B.); (S.P.); (S.A.)
- Correspondence: (D.R.); (S.C.); Tel.: +39-09613694224 (D.R.); +39-0984496207 (S.C.)
| | - Stefania Bulotta
- Department of Health Sciences, “Magna Graecia” University of Catanzaro, 88100 Catanzaro, Italy; (V.M.); (M.C.); (D.C.); (M.F.); (S.B.)
| |
Collapse
|
46
|
Abstract
Triple-negative breast cancer (TNBC) is a breast cancer subtype renowned for its capacity to affect younger women, metastasise early despite optimal adjuvant treatment and carry a poor prognosis. Neoadjuvant therapy has focused on combinations of systemic agents to optimise pathological complete response. Treatment algorithms now guide the management of patients with or without residual disease, but metastatic TNBC continues to harbour a poor prognosis. Innovative, multi-drug combination systemic therapies in the neoadjuvant and adjuvant settings have led to significant improvements in outcomes, particularly over the past decade. Recently published advances in the treatment of metastatic TNBC have shown impressive results with poly (ADP-ribose) polymerase (PARP) inhibitors and immunotherapy agents. Immunotherapy agents in combination with traditional systemic chemotherapy have been shown to alter the natural history of this devastating condition, particularly in patients whose tumours are positive for programmed cell death ligand 1 (PD-L1).
Collapse
Affiliation(s)
| | - Sherene Loi
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
47
|
Sun ZQ, Hu SD, Shao L, Jin LF, Lv Q, Li YS, Yan G. A pilot study of low-dose CT perfusion imaging (LDCTPI) technology in patients with triple-negative breast cancer. JOURNAL OF X-RAY SCIENCE AND TECHNOLOGY 2019; 27:443-451. [PMID: 30856155 DOI: 10.3233/xst-180465] [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: 06/09/2023]
Abstract
PURPOSE To investigate associations between the clinicopathologic features and CT perfusion parameters of triple-negative breast cancer (TNBC) and non-TNBC using low-dose computed tomography perfusion imaging (LDCTPI), and to find potential clinical applications in the prognosis assessment of TNBC. MATERIALS AND METHODS A total of 60 patients with breast cancer confirmed by pathological examination were studied prospectively using LDCTPI on a 64-slice spiral CT scanner. The acquired volume data were used for calculations, mapping, and analysis by using a tumor perfusion protocol in the CT perfusion software package to measure 2 parameters namely, blood flow (BF), and permeability surface (PS) area product. Patients were grouped into TNBC (n = 27) and non-TNBC (n = 33) subtypes. Associations between these two subtypes and clinicopathologic characteristics were evaluated by both univariate and multivariate logistic regression. CT perfusion parameters values were compared for clinicopathologic characteristics using independent 2-sample t test. RESULTS TNBC displayed higher CT perfusion parameters values (BF: 57.56±10.94 vs 52.70±7.79 mL/100 g/min, p = 0.006; PS: 38.98±9.46 vs 33.39±8.07 mL/100 g/min, p = 0.001) than non-TNBC. In addition, breast cancer with poorly histologic grade or positive Ki-67 expression showed higher BF and PS values than those with well and moderately histologic grade or negative Ki-67 expression (p < 0.05). TNBC had poorer histologic grade (P = 0.032) and higher Ki-67 expression (P = 0.013) than non-TNBC. CONCLUSION LDCTPI is a functional imaging technology from the perspective of hemodynamics with potential of clinical applications. The BF and PS values were higher in TNBC patient group than non-TNBC group. TNBC patients also have poorer clinicopathologic outcome.
Collapse
Affiliation(s)
- Zong-Qiong Sun
- Department of Radiology, Affiliated Hospital of Jiangnan University, The Fourth People's Hospital of Wuxi City, Wuxi, Jiangsu, China
| | - Shu-Dong Hu
- Department of Radiology, Affiliated Hospital of Jiangnan University, The Fourth People's Hospital of Wuxi City, Wuxi, Jiangsu, China
| | - Lin Shao
- Department of Radiology, Affiliated Hospital of Jiangnan University, The Fourth People's Hospital of Wuxi City, Wuxi, Jiangsu, China
| | - Lin-Fang Jin
- Department of Pathology, Affiliated Hospital of Jiangnan University, The Fourth People's Hospital of Wuxi City, Wuxi, Jiangsu, China
| | - Qing Lv
- Department of Breast Surgery, Affiliated Hospital of Jiangnan University, The Fourth People's Hospital of Wuxi City, Wuxi, Jiangsu, China
| | - Yao-Sen Li
- Department of Radiology, Wuxi Huishan Traditional Chinese Medicine Hospital, Wuxi, Jiangsu, China
| | - Gen Yan
- Department of Radiology, Affiliated Hospital of Jiangnan University, The Fourth People's Hospital of Wuxi City, Wuxi, Jiangsu, China
| |
Collapse
|
48
|
Zeng D, Xiao Y, Zhu J, Peng C, Liang W, Lin H. Knockdown of nucleophosmin 1 suppresses proliferation of triple-negative breast cancer cells through activating CDH1/Skp2/p27kip1 pathway. Cancer Manag Res 2018; 11:143-156. [PMID: 30613163 PMCID: PMC6306051 DOI: 10.2147/cmar.s191176] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Background NPM1 is a multifunctional phosphoprotein that commutes between the cytoplasm and nucleus in cell cycle process, which appears to be actively involved in tumorigenesis. Herein, we sought to investigate the possible role and prognostic value of NPM1 in triple-negative breast cancer (TNBC). Methods An array of public databases, including bc-GenExMiner v4.0, GOBO, GEPIA, UAL-CAN, ONCOMINE database and Kaplan-Meier plotter, were used to investigate the expression feature and potential function of NPM1 in TNBC. Immunohistochemistry, immunofluorescence, proliferation and colony formation, flow cytometry and western-blotting assays were used to analyze and verify the function and relevant mechanism of NPM1 in TNBC tissues and cells. Results According to analysis from bc-GenExMiner, the expression level of NPM1 was significantly higher in basal-like subtypes than luminal-A, HER-2 or normal-like subtypes of breast cancer (P<0.0001). GOBO database analysis indicated that the expression of NPM1 in basal-A or basal-B was significantly higher than luminal-like breast cancer cells. Immunohistochemistry assay in 52 TNBC tissue samples showed that positive expression of Ki-67 was 93.5% in the high-NPM1-expression group and 66.7% in the low-NPM1-expression group, respectively (P=0.032). Proliferation and colony formation assays demonstrated that inhibition of NPM1 suppressed cell growth by approximately 2-fold and reduced the number of colonies by 3-4-fold in MDA-MB-231 and BT549 cells. Moreover, inhibition of NPM1 in MDA-MB-231 and BT549 cells increased the percentage of cells at G0/G1 phase and decreased the percentage of cells at both S and G2/M phase, as compared with control counterparts. Western-blotting results showed that down-regulation of NPM1 could elevate CDH1 and p27kip1 expression, while decrease Skp2 expression both in MDA-MB-231 and BT549 cells. In addition, high mRNA expression of NPM1 correlated with shorter RFS (HR=1.64, P=0.00013) and OS (HR=2.45, P=0.00034) in patients with TNBC. Conclusions NPM1 is significantly high expressed basal-like/triple-negative breast cancer and is correlated with shorter RFS and OS in this subset of patients. Knockdown of NPM1 impairs the proliferative capacity of TNBC cells via activation of the CDH1/Skp2/p27kip1 pathway. Targeting NPM1 is a potential therapeutic strategy against TNBC.
Collapse
Affiliation(s)
- De Zeng
- Department of Medical Oncology, Cancer Hospital of Shantou University Medical College, Shantou 515031, China,
| | - Yingsheng Xiao
- Department of Thyroid Surgery, Shantou Central Hospital, Shantou 515000, China
| | - Jianling Zhu
- Department of Pathology, Cancer Hospital of Shantou University Medical College, Shantou 515031, China
| | - Chunyan Peng
- Department of Clinical Laboratory, Taihe Hospital of Hubei University of Medicine, Hubei 442008, China
| | - Weiquan Liang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China,
| | - Haoyu Lin
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, China,
| |
Collapse
|
49
|
Mosly D, Turnbull A, Sims A, Ward C, Langdon S. Predictive markers of endocrine response in breast cancer. World J Exp Med 2018; 8:1-7. [PMID: 30191138 PMCID: PMC6125140 DOI: 10.5493/wjem.v8.i1.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/26/2018] [Accepted: 08/04/2018] [Indexed: 02/07/2023] Open
Abstract
Ongoing clinical and research efforts seek to optimise the use of endocrine therapy in the treatment of breast cancer. Accurate biomarkers are needed that predict response for individual patients. The presence of the estrogen receptor (ER) as the direct (for tamoxifen and fulvestrant) or indirect (for aromatase inhibitors) target molecule for endocrine therapy remains the foremost biomarker and determinant of response. However, ER expression only poorly predicts outcome and further indicators of response or resistance are required. The development and application of molecular signature assays such as Oncotype Dx, Prosigna, Mammaprint and Endopredict have provided valuable information on prognosis and these are being used to support clinical decision making on whether endocrine therapy alone alongside surgery is sufficient for ER-positive early stage breast cancers or whether combination of endocrine with chemotherapy are also warranted. Ki67, the proliferation marker, has been widely used in the neo-adjuvant (pre-operative) setting to help predict response and long term outcome. Gene expression studies within the same setting have allowed monitoring of changes of potential predictive markers. These have identified frequent changes in estrogen-regulated and proliferation genes. Specific molecules such as mutant ER may also prove helpful biomarkers in predicting outcome and monitoring response to treatment.
Collapse
Affiliation(s)
- Duniya Mosly
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh EH4 2XR, United Kingdom
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Arran Turnbull
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh EH4 2XR, United Kingdom
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| | - Andrew Sims
- Applied Bioinformatics of Cancer, University of Edinburgh Cancer Research Centre, MRC Institute of Genetics and Molecular Medicine, Edinburgh EH4 2XR, United Kingdom
| | - Carol Ward
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
- the Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Easter Bush, Roslin, Midlothian, Edinburgh EH25 9RG, United Kingdom
| | - Simon Langdon
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratory, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH4 2XU, United Kingdom
| |
Collapse
|