1
|
Lin S, Fu B, Khan M. Identifying subgroups deriving the most benefit from PD-1 checkpoint inhibition plus chemotherapy in advanced metastatic triple-negative breast cancer: a systematic review and meta-analysis. World J Surg Oncol 2024; 22:346. [PMID: 39709499 DOI: 10.1186/s12957-024-03424-3] [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: 03/22/2024] [Accepted: 05/21/2024] [Indexed: 12/23/2024] Open
Abstract
BACKGROUND The combination of immunotherapy and chemotherapy has demonstrated an enhancement in progression-free survival (PFS) for individuals with advanced and metastatic triple-negative breast cancer (TNBC) when compared to the use of chemotherapy alone. Nevertheless, the extent to which different subgroups of metastatic TNBC patients experience this benefit remains uncertain. OBJECTIVES Our objective was to conduct subgroup analyses to more precisely identify the factors influencing these outcomes. MATERIALS AND METHODS The PubMed database was searched until Dec 2023 for studies that compared PD-1 checkpoint inhibitors plus chemotherapy (ICT) with chemotherapy (CT) alone. The primary outcome of interest was progression-free survival (PFS). Review Manager (RevMan) version 5.4. was used for the data analysis. RESULTS Four randomized controlled trials (RCTs) comprising 2468 advanced and metastatic TNBC were included in this systematic review and meta-analysis. PFS surge with combined therapy was observed in White (HR 0.80 [0.70, 0.91], p = 0.0007) and Asian ethnicities (HR 0.73 [0.58, 0.93], p = 0.01) but not in Blacks (HR 0.72 [0.42, 1.24], p = 0.24). Overall, patients with distant metastasis demonstrated to derive the PFS benefit from additional immunotherapy (HR 0.87 [0.77, 0.99], p = 0.03); however, metastasis to individual distant site was associated with failure to achieve any treatment difference (Bone: HR 0.79 [0.41, 1.52], p = 0.49; Lung: HR 0.85 [0.70, 1.04], p = 0.11; Liver: HR 0.80 [0.64, 1.01], p = 0.06). While number of metastases > 3 also showed to impact the PFS advantage (HR 0.89 [0.69, 1.16], p = 0.39). While patients, regardless of prior chemotherapy, experienced a notable enhancement in PFS with ICT (Overall: HR 0.79 [0.71, 0.88], p < 0.0001; Yes: HR 0.87 [0.76, 1.00], p = 0.05; No: HR 0.67 [0.56, 0.80], p < 0.00001), those previously exposed to chemotherapy exhibited a significantly smaller PFS advantage compared to those without prior chemotherapy, as evidenced by a significant subgroup difference (Test for subgroup difference: P = 0.02, I2 = 82.2%). Patients lacking PD-L1 expression also failed to achieve any additional benefit from immunotherapy (PD-L1-: HR 0.95 [0.81, 1.12]; p = 0.54; PD-L1+: HR 0.73 [0.64, 0.85], p < 0.0001). Age, ECOG status, and presentation with de novo metastasis/recurrent shown no impact on IT-associated PFS advantage. CONCLUSIONS Patient- and treatment- related factors such as ethnicity, distant metastases, number of metastases (> 3), previous exposure to chemotherapy and PD-L1 expression, seem to influence or restrict the advantage in progression-free survival associated with the addition of immunotherapy to chemotherapy, as opposed to chemotherapy alone, in patients with advanced and metastatic TNBC. Larger studies are warranted to validate these outcomes.
Collapse
Affiliation(s)
- Shengfa Lin
- Department of Oncology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510168, People's Republic of China
| | - Bihe Fu
- Department of Oncology, Jinshazhou Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510168, People's Republic of China
| | - Muhammad Khan
- Department of Radiation Oncology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou, Guangdong, 510095, People's Republic of China.
| |
Collapse
|
2
|
Hsiao CH, Huang HL, Liu HL, Huang CC, Su JC, Chen YH, Lin YH. Combining ultrasound technology with targeted fucoidan/arginine-gelatin nanoparticles loaded with doxorubicin to enhance therapeutic efficacy and modulate bioeffects in drug-resistant triple-negative breast cancer. Int J Biol Macromol 2024; 283:137764. [PMID: 39557262 DOI: 10.1016/j.ijbiomac.2024.137764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/04/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
Triple-negative breast cancer (TNBC) presents formidable challenges due to its aggressive nature and high recurrence rates, compounded by the involvement of epithelial-mesenchymal transition (EMT) in its progression and metastasis. Standard chemotherapy, which typically employs doxorubicin (DOX), remains a primary treatment approach. However, multidrug resistance (MDR) mechanisms, which include ATP-binding cassette transporters and EMT, contribute to treatment failures. Ultrasound has emerged as a promising modality among the various strategies explored to address MDR in TNBC. It serves as a diagnostic tool and holds therapeutic potential by inducing various biological effects depending on the exposure level. Targeted nanoparticles offer a means to enhance drug delivery efficiency. Our study aims to advance ultrasound technology combined with biocompatible nanoparticles using simplified preparation methods to improve treatment outcomes for drug-resistant TNBC. In particular, employing DOX-loaded fucoidan/arginine-gelatin nanoparticles facilitated the targeted delivery of chemotherapy drugs to tumors by effectively interacting with P-selectin, resulting in tumor growth inhibition. Furthermore, these nanoparticles mitigated MDR and EMT, particularly when combined with ultrasound treatment. This integrated approach of nanoparticle delivery with ultrasonography opens up a promising and innovative avenue for clinical cancer research.
Collapse
Affiliation(s)
- Chi-Huang Hsiao
- Division of Medical Oncology and Hematology, Department of Internal Medicine, Far Eastern Memorial Hospital, New Taipei 220216, Taiwan
| | - Hau-Lun Huang
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, Department of Biomedical Engineering, National Taiwan University, Taipei 106216, Taiwan
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300044, Taiwan
| | - Jung-Chen Su
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yi-Hsuan Chen
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Yu-Hsin Lin
- Department of Pharmacy, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Medical Device Innovation and Translation Center, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404327, Taiwan.
| |
Collapse
|
3
|
Tuo Y, Ye Y. Sterile Alpha Motif Domain-Containing 5 Suppresses Malignant Phenotypes and Tumor Growth in Breast Cancer: Regulation of Polo-Like Kinase 1 and c-Myc Signaling in a Xenograft Model. Cureus 2024; 16:e73259. [PMID: 39524172 PMCID: PMC11550111 DOI: 10.7759/cureus.73259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2024] [Indexed: 11/16/2024] Open
Abstract
Background Breast cancer, particularly the triple-negative breast cancer (TNBC) subtype, remains a significant clinical challenge due to its resistance to standard chemotherapy and high recurrence rate. In this study, we explored the role of Sterile Alpha Motif Domain-Containing 5 (SAMD5) as a potential regulatory partner with the c-Myc oncogenic signaling pathway in breast cancer. Materials and methods Functional assays were conducted to investigate the effects of SAMD5 overexpression on cell viability, colony formation, and invasive behavior in TNBC cell lines. This study further assessed the expression levels of proliferation and invasion markers, including Ki67 (a marker for cell proliferation), Matrix Metalloproteinase-2 (MMP2), and Matrix Metalloproteinase-9 (MMP9). Mechanistic analyses identified a negative correlation between SAMD5 and Polo-like Kinase 1 (PLK1), a gene frequently overexpressed in breast cancer, particularly in TNBC. The effects of PLK1 knockdown on cell viability, colony formation, and invasion were observed, along with the impact of PLK1 overexpression on SAMD5's inhibitory activity. In vivo studies were performed using a xenograft tumor model in nude mice to evaluate the impact of SAMD5 overexpression on tumor weight and volume. Results SAMD5 overexpression significantly reduced cell viability, colony formation, and invasion in TNBC cells, and downregulated key proteins in the c-Myc signaling pathway, including c-Myc itself, β-catenin, Cyclin-Dependent Kinase 4 (CDK4), Cyclin-Dependent Kinase 6 (CDK6), and Cyclin D1. PLK1 overexpression was found to counteract SAMD5's inhibitory effects. In vivo experiments demonstrated that SAMD5 overexpression led to a marked reduction in tumor weight and volume, effects that were partially reversed by PLK1 overexpression. Conclusions SAMD5 acts as a tumor suppressor in breast cancer, particularly in TNBC, by inhibiting critical cellular processes and downregulating the c-Myc signaling pathway. This effect appears to be mediated, in part, through its negative association with PLK1. Targeting the SAMD5/PLK1 axis offers a promising therapeutic strategy for addressing aggressive breast cancers.
Collapse
Affiliation(s)
- YouLin Tuo
- Department of Breast Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, CHN
| | - YiFeng Ye
- Department of Breast Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, CHN
| |
Collapse
|
4
|
Kim HS, Youn YH, Kim HJ, Koo YH, Lee J, Kwon IK, Do SH. Enhanced Antitumor Efficacy of Oncolytic Vaccinia Virus Therapy Through Keratin-Mediated Delivery in Triple-Negative Breast Cancer. Int J Mol Sci 2024; 25:11470. [PMID: 39519023 PMCID: PMC11546765 DOI: 10.3390/ijms252111470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 07/12/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Triple-negative breast cancer (TNBC) represents an aggressive subtype characterized by high rates of recurrence and metastasis, necessitating the exploration of alternative treatment strategies. Oncolytic vaccinia virus (OVV) therapy has emerged as a promising approach, selectively infecting and destroying tumor cells. However, its efficacy is often hampered by inadequate viral distribution within the tumor microenvironment. Here, we investigate the potential of keratin (KTN) as a carrier for OVV delivery to enhance viral distribution and antitumor efficacy. In vitro assays revealed that KTN significantly improves OVV stability, leading to increased tumor cell apoptosis and necrosis. Furthermore, KTN effectively inhibits cancer cell migration by suppressing the epithelial-mesenchymal transition (EMT) process and downregulating metastasis-related proteins. These findings are corroborated in a syngeneic TNBC mouse model, where KTN-mediated OVV delivery enhances cytotoxic T cell-mediated antitumor immune responses without compromising the anti-angiogenic effects of the virus. Notably, KTN alone exhibits antitumor effects by suppressing tumor growth and metastasis, underscoring its potential as a standalone therapeutic agent. In conclusion, our study underscores the promise of KTN-mediated OVV delivery as a promising therapeutic strategy for TNBC. By improving viral distribution, suppressing EMT, and enhancing antitumor immunity, this approach holds significant potential for enhancing patient outcomes in TNBC treatment. Further investigation is warranted to explore the broader utility of KTN in various cancer therapy approaches.
Collapse
Affiliation(s)
- Hyo-Sung Kim
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Yun Hee Youn
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Han-Jun Kim
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong 30019, Republic of Korea
| | - Young-Hyun Koo
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Junho Lee
- Department of Veterinary Surgery, College of Veterinary Medicine, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Il Keun Kwon
- Department of Dental Materials, School of Dentistry, Kyung Hee University, 26 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
- Medical Device Research Center, Medical Science Research Institute, Kyung Hee University Medical Center, 23 Kyung Hee Dae-ro, Dongdaemun-gu, Seoul 02447, Republic of Korea
| | - Sun Hee Do
- Department of Veterinary Clinical Pathology, College of Veterinary Medicine, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
5
|
Sijisha KS, Anusha R, Priya S. Synergistic effects of epoxyazadiradione (EAD) and paclitaxel against triple-negative breast cancer cells. Fundam Clin Pharmacol 2024; 38:758-766. [PMID: 38482560 DOI: 10.1111/fcp.13000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 07/09/2024]
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is the most aggressive and chemo-resistant form of breast cancer subtype, and chemotherapy is a vital treatment option for that. Paclitaxel is an effective chemo drug for TNBC. However, in clinical settings, paclitaxel has adverse side effects. The synergistic combination is the most promising method for overcoming undesirable toxicity and achieving a beneficial therapeutic outcome. Previous reports, including our study, showed certain anticancer potential of epoxyazadiradione (EAD), the neem limonoid, in different types of cancer cells, including TNBC. OBJECTIVE This study was designed to investigate the possible synergistic effects of EAD and paclitaxel against TNBC cells. METHODS We examined the effects of EAD and paclitaxel alone and in combination in MDA-MB 231 cells, and the percentage cytotoxicity was used to calculate synergism. Characteristic apoptotic changes were observed by visualizing cellular morphology, nuclear fragmentation and membrane integrity. We further estimated anti-migratory potential of experimental compounds by wound healing assay. The reduction in inflammation during combinatorial treatment was evaluated by observing NF-κB translocation. RESULTS The combined treatment with EAD (5 μM) and paclitaxel (5 nM), which were used at doses lower than their individual IC50 concentrations, showed a synergistic effect in MDA-MB-231 cells. This combination effectively induced apoptosis and antimigration and reduced the inflammatory reactions induced by the higher dose of paclitaxel. CONCLUSION To conclude, EAD could be the drug of choice for combined treatment with paclitaxel in a chemotherapy regimen.
Collapse
Affiliation(s)
- Kunnathully Sudhan Sijisha
- Biochemistry Section, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, Kerala, India
| | - Rajitha Anusha
- Biochemistry Section, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, Kerala, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Sulochana Priya
- Biochemistry Section, Agro-Processing and Technology Division, CSIR-National Institute for Interdisciplinary Science and Technology (CSIR-NIIST), Thiruvananthapuram, Kerala, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
6
|
Yang C, Zhang W, Xiang S, Chen L, Chun J, Chen H. Navel orange peel essential oil inhibits the growth and progression of triple negative breast cancer. BMC Complement Med Ther 2024; 24:233. [PMID: 38877505 PMCID: PMC11177363 DOI: 10.1186/s12906-024-04525-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 05/28/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND Triple Negative Breast Cancer (TNBC) is a particular type of breast cancer with the highest mortality rate. Essential oils are concerned more and more as potential anti-cancer drugs. METHODS TNBC cells were treated with different concentrations of navel orange peel essential oil (NOPEO), and then a variety of experiments were performed to investigate the changes in the growth and progression of TNBC cells. MTT assay was performed to detect the proliferation of TNBC cells. The changes of cell cycle and apoptosis were analyzed by FACS. In order to explored the migration of TNBC cells, scratch wound assay was carried out. Western blotting and qPCR were used to examine the expression of proteins and mRNA of related genes. Furthermore, RNA-seq was used to analyze the altered genes and explored the possible signal pathway. RESULTS NOPEO demonstrated dose- and time-dependent suppression of TNBC cell growth. TNBC cells showed an increased percentage of G2/M-phase cells and the protein levels of CyclinB1 and CyclinD1 were decreased after NOPEO treatment. The apoptotic cells were increased in the NOPEO treated TNBC cells. The migration mobility was significantly inhibited by NOPEO. In total, 1376 genes were found to be up-regulated and 1335 genes were down-regulated after NOPEO treatment. According to KEGG and GO pathways, the differentially expressed genes were related to MAPK, Jak/stat and FoxQ signaling pathways. CONCLUSION This investigation explored the bio-activity and molecular mechanisms of NOPEO against TNBC cells. These results indicated that NOPEO could suppress TNBC growth and migration perhaps via the MAPK and Jak/stat signaling pathways, which may provide theoretical reference for anticancer drug development. NOPEO may be a potential natural product for the chemotherapeutic of TNBC.
Collapse
Affiliation(s)
- Chao Yang
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
- College of Life Sciences, National Navel Orange Engineering Research Center, Gannan Normal University, Ganzhou, 341000, China
| | - Wenwen Zhang
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Shi Xiang
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Lai Chen
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China
| | - Jiong Chun
- College of Life Sciences, National Navel Orange Engineering Research Center, Gannan Normal University, Ganzhou, 341000, China.
| | - Hui Chen
- Oncology Research Center, Jiangxi Provincial Key Laboratory of Traditional Chinese Medicine Diagnosis and Rehabilitation of Malignant Tumors, Jiangxi University of Chinese Medicine, Nanchang, 330004, China.
- College of Life Sciences, National Navel Orange Engineering Research Center, Gannan Normal University, Ganzhou, 341000, China.
| |
Collapse
|
7
|
Shen HY, Xu JL, Zhang W, Chen QN, Zhu Z, Mao Y. Exosomal circRHCG promotes breast cancer metastasis via facilitating M2 polarization through TFEB ubiquitination and degradation. NPJ Precis Oncol 2024; 8:22. [PMID: 38287113 PMCID: PMC10825185 DOI: 10.1038/s41698-024-00507-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 12/06/2023] [Indexed: 01/31/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a highly aggressive cancer with distant metastasis. Accumulated evidence has demonstrated that exosomes are involved in TNBC metastasis. Elucidating the mechanism underlying TNBC metastasis has important clinical significance. In the present study, exosomes were isolated from clinical specimens and TNBC cell lines. Colony formation, EdU incorporation, wound healing, and transwell assays were performed to examine TNBC cell proliferation, migration, and metastasis. Macrophage polarization was evaluated by flow cytometry and RT-qPCR analysis of polarization markers. A mouse model of subcutaneous tumor was established for assessment of tumor growth and metastasis. RNA pull-down, RIP and Co-IP assays were used for analyzing molecular interactions. Here, we proved that high abundance of circRHCG was observed in exosomes derived from TNBC patients, and increased exosomal circRHCG indicated poor prognosis. Silencing of circRHCG suppressed TNBC cell proliferation, migration, and metastasis. TNBC cell-derived exosomes promoted M2 polarization via delivering circRHCG. Exosomal circRHCG stabilized BTRC mRNA via binding FUS and naturally enhanced BTRC expression, thus promoting the ubiquitination and degradation of TFEB in THP-1 cells. In addition, knockdown of BTRC or overexpression of TFEB counteracted exosomal circRHCG-mediated facilitation of M2 polarization. Furthermore, exosomal circRHCG promoted TNBC cell proliferation and metastasis by facilitating M2 polarization. Knockdown of circRHCG reduced tumor growth, metastasis, and M2 polarization through the BTRC/TFEB axis in vivo. In summary, exosomal circRHCG promotes M2 polarization by stabilizing BTRC and promoting TFEB degradation, thereby accelerating TNBC metastasis and growth. Our study provides promising therapeutic strategies against TNBC.
Collapse
Affiliation(s)
- Hong-Yu Shen
- Gusu School, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jia-Lin Xu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Zhang
- Division of Gastrointestinal Surgery, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, China
| | - Qin-Nan Chen
- Department of Clinical Medicine, Jiangsu Health Vocational College, Nanjing, China.
| | - Zhen Zhu
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Yuan Mao
- Department of Oncology, The Fourth Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
8
|
Akash S, Aovi FI, Azad MAK, Kumer A, Chakma U, Islam MR, Mukerjee N, Rahman MM, Bayıl I, Rashid S, Sharma R. A drug design strategy based on molecular docking and molecular dynamics simulations applied to development of inhibitor against triple-negative breast cancer by Scutellarein derivatives. PLoS One 2023; 18:e0283271. [PMID: 37824496 PMCID: PMC10569544 DOI: 10.1371/journal.pone.0283271] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Accepted: 05/07/2023] [Indexed: 10/14/2023] Open
Abstract
Triple-negative breast cancer (TNBC), accounting for 10-15% of all breast malignancies, is more prevalent in women under 40, particularly in those of African descent or carrying the BRCA1 mutation. TNBC is characterized by the absence of estrogen and progesterone receptors (ER, PR) and low or elevated HER2 expression. It represents a particularly aggressive form of breast cancer with limited therapeutic options and a poorer prognosis. In our study, we utilized the protein of TNBC collected from the Protein Data Bank (PDB) with the most stable configuration. We selected Scutellarein, a bioactive molecule renowned for its anti-cancer properties, and used its derivatives to design potential anti-cancer drugs employing computational tools. We applied and modified structural activity relationship methods to these derivatives and evaluated the probability of active (Pa) and inactive (Pi) outcomes using pass prediction scores. Furthermore, we employed in-silico approaches such as the assessment of absorption, distribution, metabolism, excretion, and toxicity (ADMET) parameters, and quantum calculations through density functional theory (DFT). Within the DFT calculations, we analyzed Frontier Molecular Orbitals, specifically the Highest Occupied Molecular Orbital (HOMO) and Lowest Unoccupied Molecular Orbital (LUMO). We then conducted molecular docking and dynamics against TNBC to ascertain binding affinity and stability. Our findings indicated that Scutellarein derivatives, specifically DM03 with a binding energy of -10.7 kcal/mol and DM04 with -11.0 kcal/mol, exhibited the maximum binding tendency against Human CK2 alpha kinase (PDB ID 7L1X). Molecular dynamic simulations were performed for 100 ns, and stability was assessed using root-mean-square deviation (RMSD) and root-mean-square fluctuation (RMSF) parameters, suggesting significant stability for our chosen compounds. Furthermore, these molecules met the pharmacokinetics requirements for potential therapeutic candidates, displaying non-carcinogenicity, minimal aquatic and non-aquatic toxicity, and greater aqueous solubility. Collectively, our computational data suggest that Scutellarein derivatives may serve as potential therapeutic agents for TNBC. However, further experimental investigations are needed to validate these findings.
Collapse
Affiliation(s)
- Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Farjana Islam Aovi
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Md. A. K. Azad
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Ajoy Kumer
- Laboratory of Computational Research for Drug Design and Material Science, Department of Chemistry, European University of Bangladesh, Dhaka, Bangladesh
| | - Unesco Chakma
- School of Electronic Science and Engineering, Southeast University, Nanjing, China
| | - Md. Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Nobendu Mukerjee
- Center for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, India
- Department of Microbiology, West Bengal State University, West Bengal, Kolkata, India
| | - Md. Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| | - Imren Bayıl
- Department of Bioinformatics and Computational Biology, Gaziantep University, Gaziantep, Turkey
| | - Summya Rashid
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Rohit Sharma
- Department of Rasa Shastra and Bhaishajya Kalpana, Faculty of Ayurveda, Institute of Medical Science, Banaras Hindu University, Varanasi, India
| |
Collapse
|
9
|
El Hejjioui B, Lamrabet S, Amrani Joutei S, Senhaji N, Bouhafa T, Malhouf MA, Bennis S, Bouguenouch L. New Biomarkers and Treatment Advances in Triple-Negative Breast Cancer. Diagnostics (Basel) 2023; 13:diagnostics13111949. [PMID: 37296801 DOI: 10.3390/diagnostics13111949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 06/12/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is a specific subtype of breast cancer lacking hormone receptor expression and HER2 gene amplification. TNBC represents a heterogeneous subtype of breast cancer, characterized by poor prognosis, high invasiveness, high metastatic potential, and a tendency to relapse. In this review, the specific molecular subtypes and pathological aspects of triple-negative breast cancer are illustrated, with particular attention to the biomarker characteristics of TNBC, namely: regulators of cell proliferation and migration and angiogenesis, apoptosis-regulating proteins, regulators of DNA damage response, immune checkpoints, and epigenetic modifications. This paper also focuses on omics approaches to exploring TNBC, such as genomics to identify cancer-specific mutations, epigenomics to identify altered epigenetic landscapes in cancer cells, and transcriptomics to explore differential mRNA and protein expression. Moreover, updated neoadjuvant treatments for TNBC are also mentioned, underlining the role of immunotherapy and novel and targeted agents in the treatment of TNBC.
Collapse
Affiliation(s)
- Brahim El Hejjioui
- Biomedical and Translational Research Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
- Department of Medical Genetics and Oncogenetics, HASSAN II University Hospital, Fez 30050, Morocco
| | - Salma Lamrabet
- Biomedical and Translational Research Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Sarah Amrani Joutei
- Department of Radiotherapy, HASSAN II University Hospital, Fez 30050, Morocco
| | - Nadia Senhaji
- Faculty of Sciences, Moulay Ismail University, Meknès 50000, Morocco
| | - Touria Bouhafa
- Department of Radiotherapy, HASSAN II University Hospital, Fez 30050, Morocco
| | | | - Sanae Bennis
- Biomedical and Translational Research Laboratory, Faculty of Medicine and Pharmacy, Sidi Mohamed Ben Abdellah University, Fez 30050, Morocco
| | - Laila Bouguenouch
- Department of Medical Genetics and Oncogenetics, HASSAN II University Hospital, Fez 30050, Morocco
| |
Collapse
|
10
|
Khan M, Du K, Ai M, Wang B, Lin J, Ren A, Chen C, Huang Z, Qiu W, Yuan Y, Tian Y. PD-L1 expression as biomarker of efficacy of PD-1/PD-L1 checkpoint inhibitors in metastatic triple negative breast cancer: A systematic review and meta-analysis. Front Immunol 2023; 14:1060308. [PMID: 36949944 PMCID: PMC10027008 DOI: 10.3389/fimmu.2023.1060308] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Background Inhibitors of programmed cell death 1 (PD-1)/programmed cell death ligand 1(PD-L1) checkpoint have been approved for metastatic triple negative breast cancer (mTNBC) in patients positive for PD-L1 expression. Negative results from the recent phase III trials (IMPassion131 and IMPassion132) have raises questions on the efficacy of PD-1/PD-L1 checkpoint inhibitors and the predictive value of PD-L1 expression. Here we attempt to systematically analyze the biomarker value of PD-L1 expression for predicting the response of PD-1/PD-L1 checkpoint inhibitors in mTNBC. Materials and methods PubMed database was searched until Dec 2021 for studies evaluating PD-1/PD-L1 checkpoint inhibitors plus/minus chemotherapy in mTNBC. Outcome of interest included objective response rate (ORR), progression-free survival (PFS), and overall survival (OS). Review Manager (RevMan) version 5.4. was used for data-analysis. Results In total, 20 clinical trials comprising 3962 mTNBC patients (ICT: 2665 (67%); CT: 1297 (33%) were included in this study. Overall ORR was 22% (95%CI, 14-30%) and significant improvement was observed for PD-L1+ patients (ORR 1.78 [95%CI, 1.45-2.19], p<0.00001) as compared to PD-L1- cohort. Pooled outcome also indicated a significant 1-year PFS and 2-year OS advantage for patients with PD-L1 expression (1-year PFS: ORR 1.39 [95%CI, 1.04-1.85], p=0.02; I2 = 0%; 2-year OS: (ORR 2.47 [95%CI, 1.30-4.69], p=0.006; I2 = 63%). Subgroup analysis indicated that PD-L1 expression can successfully predict tumor response and 2-year OS benefit in mTNBC patients regardless of the type of investigating agent, line of treatment administration, and to some extent the type of treatment. Biomarker ability of PD-L1 expression to predict 1-year PFS was slightly better with pembrolizumab (p=0.09) than atezolizumab (p=0.18), and significantly better when treatment was administered in the first-line setting (OR 1.38 [95%CI, 1.02-1.87], p=0.04) and chemotherapy was added (OR 1.38 [95%CI, 1.02-1.86], p=0.03). Immune-related toxicity of any grade and grade≥3 was 39% (95%CI, 26%-52%) and 10% (95%CI, 8%-13%), respectively. Conclusions PD-L1 expression can predict objective response rate and 2-year OS in mTNBC patients receiving PD-1/PD-L1 checkpoint inhibitors. One-year PFS is also predicted in selected patients. PD-L1 expression can be a useful biomarker of efficacy of PD-1/PD-L1 checkpoint inhibitors in mTNBC.
Collapse
Affiliation(s)
- Muhammad Khan
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Kunpeng Du
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Meiling Ai
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Baiyao Wang
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Jie Lin
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Anbang Ren
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Chengcong Chen
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Zhong Huang
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Wenze Qiu
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Yawei Yuan
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Yunhong Tian
- Department of Radiation Oncology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- State Key Laboratory of Respiratory Diseases, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
11
|
Hirano R, Okamoto K, Shinke M, Sato M, Watanabe S, Watanabe H, Kondoh G, Kadonosono T, Kizaka-Kondoh S. Tissue-resident macrophages are major tumor-associated macrophage resources, contributing to early TNBC development, recurrence, and metastases. Commun Biol 2023; 6:144. [PMID: 36737474 PMCID: PMC9898263 DOI: 10.1038/s42003-023-04525-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/24/2023] [Indexed: 02/05/2023] Open
Abstract
Triple-negative breast cancer (TNBC) is an aggressive and highly heterogenous disease with no well-defined therapeutic targets. Treatment options are thus limited and mortality is significantly higher compared with other breast cancer subtypes. Mammary gland tissue-resident macrophages (MGTRMs) are found to be the most abundant stromal cells in early TNBC before angiogenesis. We therefore aimed to explore novel therapeutic approaches for TNBC by focusing on MGTRMs. Local depletion of MGTRMs in mammary gland fat pads the day before TNBC cell transplantation significantly reduced tumor growth and tumor-associated macrophage (TAM) infiltration in mice. Furthermore, local depletion of MGTRMs at the site of TNBC resection markedly reduced recurrence and distant metastases, and improved chemotherapy outcomes. This study demonstrates that MGTRMs are a major TAM resource and play pivotal roles in the growth and malignant progression of TNBC. The results highlight a possible novel anti-cancer approach targeting tissue-resident macrophages.
Collapse
Affiliation(s)
- Ryuichiro Hirano
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Koki Okamoto
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Miyu Shinke
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Marika Sato
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Shigeaki Watanabe
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Hitomi Watanabe
- grid.258799.80000 0004 0372 2033Institute for Life and Medical Sciences, Kyoto University, Sakyo, Kyoto, 606-8507 Japan
| | - Gen Kondoh
- grid.258799.80000 0004 0372 2033Institute for Life and Medical Sciences, Kyoto University, Sakyo, Kyoto, 606-8507 Japan
| | - Tetsuya Kadonosono
- grid.32197.3e0000 0001 2179 2105School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501 Japan
| | - Shinae Kizaka-Kondoh
- School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, 226-8501, Japan.
| |
Collapse
|
12
|
Rajput S, Sharma PK, Malviya R. Biomarkers and Treatment Strategies for Breast Cancer Recurrence. Curr Drug Targets 2023; 24:1209-1220. [PMID: 38164731 DOI: 10.2174/0113894501258059231103072025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 07/14/2023] [Accepted: 10/03/2023] [Indexed: 01/03/2024]
Abstract
Despite recent treatment advancements, breast cancer remains a life-threatening disease. Although treatment is successful in the early stages, a significant proportion of individuals with breast cancer eventually experience a recurrence of the disease. Breast tumour recurrence poses a significant medical issue. Despite tumours being a primary cause of mortality, there remains a limited understanding of the fundamental mechanisms underlying tumour recurrence. The majority of the time, after surgery or medical treatment, this metastatic disease manifests itself after the disease is undiagnosed for a considerable amount of time. This phenomenon is commonly referred to as a relapse or recurrence. Metastatic breast cancer has the potential to recur at varying intervals, ranging from a few months to several decades following the initial diagnosis and treatment. This article aimed to summarise the primary causes of breast cancer recurrence and highlight the key issues that need to be addressed in order to effectively decrease the mortality rate among breast cancer patients. This article discusses various therapeutic approaches currently employed and emerging treatment strategies that hold the potential for the complete cure of cancer.
Collapse
Affiliation(s)
- Shivam Rajput
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Pramod Kumar Sharma
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
13
|
Wu K, Zhang H, Zhou L, Chen L, Mo C, Xu S, Lin J, Kong L, Chen X. Histone deacetylase inhibitor panobinostat in combination with rapamycin confers enhanced efficacy against triple-negative breast cancer. Exp Cell Res 2022; 421:113362. [PMID: 36152730 DOI: 10.1016/j.yexcr.2022.113362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 09/14/2022] [Accepted: 09/17/2022] [Indexed: 12/29/2022]
Abstract
Triple-negative breast cancer (TNBC) accounts for about 15% of diagnosed breast cancer patients, which has a poor survival outcome owing to a lack of effective therapies. This study aimed to explore the in vitro and in vivo efficiency of histone deacetylase (HDAC) inhibitor panobinostat (PANO) in combination with mTOR inhibitor rapamycin (RAPA) against TNBC. TNBC cells were treated with PANO, RAPA alone or the combination of drugs, then cell growth and apoptosis were evaluated by CCK-8, colony formation and flow cytometry. Cell migration and invasion were detected by wound healing assay and transwell assay, respectively. ROS production was detected by DCFH-DA staining. Western blotting was performed to detect protein levels. In vivo tumor growth was assessed in nude mice. The expression of cleaved caspase-3 and Ki-67 in tumor tissues was detected by immunofluorescence staining. H&E staining was conducted to observe the pathological changes in heart, liver, and kidney tissues. The combination of PANO and RAPA exerted a stronger role in repressing growth, migration, invasion, and inducing apoptosis of TNBC cells compared with monotherapy. Furthermore, this combination presented a more effective anti-cancer efficacy than a single treatment in the xenograft model without apparent toxic side effects. Importantly, mechanistic studies indicated that PANO and RAPA combination led to ROS overproduction, which subsequently activated endoplasmic reticulum stress. Conclusion: PANO in combination with RAPA exhibits enhanced efficacy against TNBC, which may be considered a promising therapeutic candidate.
Collapse
Affiliation(s)
- Kunlin Wu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350004, China.
| | - Huihao Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350004, China
| | - Linlin Zhou
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, Fujian, 350122, China
| | - Ling Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350004, China
| | - Caiqin Mo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350004, China
| | - Sunwang Xu
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350004, China
| | - Junyu Lin
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350004, China
| | - Lingjun Kong
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350004, China
| | - Xiangjin Chen
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, 350004, China.
| |
Collapse
|
14
|
Gómez Bergna SM, Marchesini A, Amorós Morales LC, Arrías PN, Farina HG, Romanowski V, Gottardo MF, Pidre ML. Exploring the Role of the Inhibitor of Apoptosis BIRC6 in Breast Cancer: A Database Analysis. JCO Clin Cancer Inform 2022; 6:e2200093. [PMID: 36455174 DOI: 10.1200/cci.22.00093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
PURPOSE The aim of the present work was to investigate the role of apoptosis inhibitor BIRC6 (baculoviral IAP repeat-containing protein 6) in breast cancer (BC), focusing particularly on its involvement in the metastatic cascade. METHODS We analyzed BIRC6 mRNA expression levels and copy number variations in three BC databases from The Cancer Genome Atlas comparing clinical and molecular attributes. Genomic analysis was performed using the cBioPortal platform, whereas transcriptomic studies (mRNA expression levels, correlation heatmaps, survival plots, and gene ontology) were performed using USC Xena and R. Statistical significance was set at P < .05. RESULTS Our bioinformatic analyses showed that there was a differential expression of BIRC6 in cancer samples when compared with normal samples. Copy number variations that involve amplification and gain of BIRC6 gene were correlated with negative hormone receptor tumors, higher prognostic indexes, younger age at diagnosis, and both chemotherapy and radiotherapy administration. Transcriptomic and gene ontology analyses showed that, under conditions of high BIRC6 mRNA levels, there are differential expression patterns in apoptotic, proliferation, and metastatic pathways. CONCLUSION In summary, our in silico data suggest that BIRC6 plays an antiapoptotic, pro-proliferative, and apparent prometastatic role and could be a relevant molecular target for treatment of BC tumors.
Collapse
Affiliation(s)
- Santiago M Gómez Bergna
- Facultad de Ciencias Exactas, Instituto de Biotecnología y Biología Molecular (IBBM-CONICET-UNLP), Universidad Nacional de La Plata, La Plata, Argentina
| | - Abril Marchesini
- Facultad de Ciencias Exactas, Instituto de Biotecnología y Biología Molecular (IBBM-CONICET-UNLP), Universidad Nacional de La Plata, La Plata, Argentina
| | - Leslie C Amorós Morales
- Facultad de Ciencias Exactas, Instituto de Biotecnología y Biología Molecular (IBBM-CONICET-UNLP), Universidad Nacional de La Plata, La Plata, Argentina
| | - Paula N Arrías
- Facultad de Ciencias Exactas, Instituto de Biotecnología y Biología Molecular (IBBM-CONICET-UNLP), Universidad Nacional de La Plata, La Plata, Argentina
| | - Hernán G Farina
- Departamento de Ciencia y Tecnología, Centro de Oncología Molecular y Traslacional, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Víctor Romanowski
- Facultad de Ciencias Exactas, Instituto de Biotecnología y Biología Molecular (IBBM-CONICET-UNLP), Universidad Nacional de La Plata, La Plata, Argentina
| | - M Florencia Gottardo
- Departamento de Ciencia y Tecnología, Centro de Oncología Molecular y Traslacional, Universidad Nacional de Quilmes, Buenos Aires, Argentina
| | - Matias L Pidre
- Facultad de Ciencias Exactas, Instituto de Biotecnología y Biología Molecular (IBBM-CONICET-UNLP), Universidad Nacional de La Plata, La Plata, Argentina
| |
Collapse
|
15
|
Hsiao CH, Huang HL, Chen YH, Chen ML, Lin YH. Enhanced antitumor effect of doxorubicin through active-targeted nanoparticles in doxorubicin-resistant triple-negative breast cancer. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
16
|
Gao L, Zhou W, Xie N, Qiu J, Huang J, Zhang Z, Hong M, Xia J, Xu J, Zhao P, Fu L, Luo Y, Jiang J, Gong H, Wang J, Dai Y, Luo D, Zou C. Yin Yang 1 promotes aggressive cell growth in high-grade breast cancer by directly transactivating kinectin 1. MedComm (Beijing) 2022; 3:e133. [PMID: 35811688 PMCID: PMC9253731 DOI: 10.1002/mco2.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/15/2022] [Accepted: 03/17/2022] [Indexed: 11/05/2022] Open
Abstract
Invasive cancer growth and metastasis account for the poor prognosis of high-grade breast cancer. Recently, we reported that kinectin 1 (KTN1), a member of the kinesin-binding protein family, promotes cell invasion of triple-negative breast cancer and high-grade breast cancer cells by augmenting the NF-κB signaling pathway. However, the upstream mechanism regulating KTN1 is unknown. Therefore, this functional study was performed to decipher the regulatory cohort of KTN1 in high-grade breast cancer. Bioinformatic analysis indicated that transcription factor Yin Yang 1 (YY1) was a potential transactivator of KTN1. High YY1 expression correlated positively with pathological progression and poor prognosis of high-grade breast cancer. Additionally, YY1 promoted cell invasive growth both in vitro and in vivo, in a KTN1-dependent manner. Mechanistically, YY1 could transactivate the KTN1 gene promoter. Alternatively, YY1 could directly interact with a co-factor, DEAD-box helicase 3 X-linked (DDX3X), which significantly co-activated YY1-mediated transcriptional expression of KTN1. Moreover, DDX3X augmented YY1-KTN1 signaling-promoted invasive cell growth of breast cancer. Importantly, overexpression of YY1 enhanced tumor aggressive growth in a mouse breast cancer model. Our findings established a novel DDX3X-assisted YY1-KTN1 regulatory axis in breast cancer progression, which could lead to the development novel therapeutic targets for breast cancer.
Collapse
Affiliation(s)
- Lin Gao
- Department of Clinical Medical Research CenterThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
| | - Wenbin Zhou
- Department of Thyroid and Breast SurgeryDepartment of General SurgeryThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
| | - Ni Xie
- BiobankShenzhen Second People’ s HospitalShenzhen, Health Science CenterFirst Affiliated Hospital of Shenzhen UniversityShenzhenGuangdongChina
| | - Junying Qiu
- Medical Laboratory of Shenzhen Luohu People's HospitalShenzhenGuangdongChina
| | - Jingyi Huang
- Department of Clinical Medical Research CenterThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
| | - Zhe Zhang
- Department of Clinical Medical Research CenterThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
| | - Malin Hong
- Department of Clinical Medical Research CenterThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
- Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosisthe Second Clinical Medical CollegeJinan UniversityShenzhenGuangdongPR China
| | - Jinquan Xia
- Department of Clinical Medical Research CenterThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
| | - Jing Xu
- Department of Clinical Medical Research CenterThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
| | - Pan Zhao
- Department of Clinical Medical Research CenterThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
- Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosisthe Second Clinical Medical CollegeJinan UniversityShenzhenGuangdongPR China
| | - Li Fu
- Guangdong Provincial Key Laboratory of Regional Immunity and DiseasesDepartment of Pharmacology and International Cancer CenterShenzhen University Health Science CenterShenzhenGuangdongChina
| | - Yuwei Luo
- Department of Thyroid and Breast SurgeryDepartment of General SurgeryThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
| | - Jing Jiang
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)ShenzhenGuangdongChina
| | - Hui Gong
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)ShenzhenGuangdongChina
| | - Jigang Wang
- Department of Clinical Medical Research CenterThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
- Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosisthe Second Clinical Medical CollegeJinan UniversityShenzhenGuangdongPR China
| | - Yong Dai
- Department of Clinical Medical Research CenterThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
| | - Dixian Luo
- Department of Laboratory MedicineHuazhong University of Science and Technology Union Shenzhen Hospital (Nanshan Hospital)ShenzhenGuangdongChina
| | - Chang Zou
- Department of Clinical Medical Research CenterThe Second Clinical Medical CollegeJinan University (Shenzhen People's Hospital)The First Affiliated Hospital of Southern University of Science and TechnologyShenzhenGuangdongChina
- Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosisthe Second Clinical Medical CollegeJinan UniversityShenzhenGuangdongPR China
- School of Life and Health SciencesThe Chinese University of Kong HongShenzhenGuangdongChina
| |
Collapse
|
17
|
Guha L, Bhat IA, Bashir A, Rahman JU, Pottoo FH. Nanotechnological Approaches for the Treatment of Triple-Negative Breast Cancer: A Comprehensive Review. Curr Drug Metab 2022; 23:781-799. [PMID: 35676850 DOI: 10.2174/1389200223666220608144551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 02/01/2022] [Accepted: 03/10/2022] [Indexed: 01/05/2023]
Abstract
Breast cancer is the most prevalent cancer in women around the world, having a sudden spread nowadays because of the poor sedentary lifestyle of people. Comprising several subtypes, one of the most dangerous and aggressive ones is triple-negative breast cancer or TNBC. Even though conventional surgical approaches like single and double mastectomy and preventive chemotherapeutic approaches are available, they are not selective to cancer cells and are only for symptomatic treatment. A new branch called nanotechnology has emerged in the last few decades that offers various novel characteristics, such as size in nanometric scale, enhanced adherence to multiple targeting moieties, active and passive targeting, controlled release, and site-specific targeting. Among various nanotherapeutic approaches like dendrimers, lipid-structured nanocarriers, carbon nanotubes, etc., nanoparticle targeted therapeutics can be termed the best among all for their specific cytotoxicity to cancer cells and increased bioavailability to a target site. This review focuses on the types and molecular pathways involving TNBC, existing treatment strategies, various nanotechnological approaches like exosomes, carbon nanotubes, dendrimers, lipid, and carbon-based nanocarriers, and especially various nanoparticles (NPs) like polymeric, photodynamic, peptide conjugated, antibody-conjugated, metallic, inorganic, natural product capped, and CRISPR based nanoparticles already approved for treatment or are under clinical and pre-clinical trials for TNBC.
Collapse
Affiliation(s)
- Lahanya Guha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research Mohali, S.A.S Nagar, Punjab 160062, India
| | - Ishfaq Ahmad Bhat
- Northern Railway Hospital, Sri Mata Vaishno Devi, Katra, Reasi 182320, India
| | - Aasiya Bashir
- Department of Pharmaceutical Sciences, Faculty of Applied Sciences and Technology, University of Kashmir, Hazratbal, Srinagar-190006, J&K, India
| | - Jawad Ur Rahman
- Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam 31441, Saudi Arabia
| | - Faheem Hyder Pottoo
- Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P.O.BOX 1982, Dammam 31441, Saudi Arabia
| |
Collapse
|
18
|
Ramos JP, Abdel-Salam MAL, Nobre DAB, Glanzmann N, de Souza CP, Leite EA, de Abreu Teles PP, Barbosa AS, Barcelos LS, Dos Reis DC, Cassali GD, de Lima ME, de Castro QJT, Grabe-Guimarães A, da Silva AD, de Souza-Fagundes EM. Acute toxicity and antitumor potential of 1,3,4-trisubstituted-1,2,3-triazole dhmtAc-loaded liposomes on a triple-negative breast cancer model. Arch Pharm (Weinheim) 2022; 355:e2200004. [PMID: 35621705 DOI: 10.1002/ardp.202200004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 04/28/2022] [Accepted: 05/02/2022] [Indexed: 11/05/2022]
Abstract
For the first time, compounds developed from the 1,2,3-triazole scaffold were evaluated as novel drugs to treat triple-negative breast cancer (TNBC). Four organic salts were idealized as nonclassical bioisosteres of miltefosine, which is used in the topical treatment for skin metastasizing breast carcinoma. Among them, derivative dhmtAc displayed better solubility and higher cytotoxicity against the human breast adenocarcinoma cell line and mouse 4T1 cell lines, which are representatives of TNBC. In vitro assays revealed that dhmtAc interferes with cell integrity, confirmed by lactate dehydogenase leakage. Due to its human peripheral blood mononuclear cell (PBMC) toxicity, dhmtAc in vivo studies were carried out with the drug incorporated in a long-circulating and pH-sensitive liposome (SpHL-dhmtAc), and the acute toxicity in BALB/c mice was determined. Free dhmtAc displayed cardiac and pulmonary toxicity after the systemic administration of 5 mg/kg doses. On the other hand, SpHL-dhmtAc displayed no toxicity at 20 mg/kg. The in vivo antitumor effect of SpHL-dhmtAc was investigated using the 4T1 heterotopic murine model. Intravenous administration of SpHL-dhmtAc reduced the tumor volume and weight, without interfering with the body weight, compared with the control group and the dhmtAc free form. The incorporation of the triazole compound in the liposome allowed the demonstration of its anticancer potential. These findings evidenced 1,3,4-trisubstituted-1,2,3-triazole as a promising scaffold for the development of novel drugs with applicability for the treatment of patients with TNBC.
Collapse
Affiliation(s)
- Jonas P Ramos
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Mostafa A L Abdel-Salam
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Daniel A B Nobre
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Nicolas Glanzmann
- Departamento de Química, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Camila P de Souza
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Elaine A Leite
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Pedro P de Abreu Teles
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alan S Barbosa
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luciola S Barcelos
- Departamento de Fisiologia e Biofísica, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Diego C Dos Reis
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Geovanni D Cassali
- Departamento de Patologia Geral, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Maria E de Lima
- Programa de Pós-Graduação em Medicina-Biomedicina, Faculdade Santa Casa de Belo Horizonte, Belo Horizonte, Brazil
| | - Quênia J T de Castro
- Departamento de Farmácia, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Andrea Grabe-Guimarães
- Departamento de Farmácia, Escola de Farmácia, Universidade Federal de Ouro Preto, Ouro Preto, Brazil
| | - Adilson D da Silva
- Departamento de Química, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | | |
Collapse
|
19
|
Abou-Fadel J, Bhalli M, Grajeda B, Zhang J. CmP Signaling Network Leads to Identification of Prognostic Biomarkers for Triple-Negative Breast Cancer in Caucasian Women. Genet Test Mol Biomarkers 2022; 26:198-219. [PMID: 35481969 DOI: 10.1089/gtmb.2021.0221] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Objective: Triple-negative breast cancer (TNBC) constitutes ∼15% of all diagnosed invasive breast cancer cases with limited options for treatment since immunotherapies that target ER, PR, and HER2 receptors are ineffective. Progesterone (PRG) can induce its effects through either classic, nonclassic, or combined responses by binding to classic nuclear PRG receptors (nPRs) or nonclassic membrane PRG receptors (mPRs). Under PRG-induced actions, we previously demonstrated that the CCM signaling complex (CSC) can couple both nPRs and mPRs into a CmPn signaling network, which plays an important role during nPR(+) breast cancer tumorigenesis. We recently defined the novel CmP signaling network in African American women (AAW)-derived TNBC cells, which overlapped with our previously defined CmPn network in nPR(+) breast cancer cells. Methods: Under mPR-specific steroid actions, we measured alterations to key tumorigenic pathways in Caucasian American women (CAW)- derived TNBC cells, with RNAseq/proteomic and systems biology approaches. Exemption from ethics approval from IRB: This study only utilized cultured NBC cell lines with publicly available TNBC clinical data sets. Results: Our results demonstrated that TNBCs in CAW share similar altered signaling pathways, as TNBCs in AAW, under mPR-specific steroid actions, demonstrating the overall aggressive nature of TNBCs, regardless of racial differences. Furthermore, in this report, we have deconvoluted the CmP signalosome, using systems biology approaches and CAW-TNBC clinical data, to identify 21 new CAW-TNBC-specific prognostic biomarkers that reinforce the definitive role of CSC and mPR signaling during CAW-TNBC tumorigenesis. Conclusion: This new set of potential prognostic biomarkers may revolutionize molecular mechanisms and currently known concepts of tumorigenesis in CAW-TNBCs, leading to hopeful new therapeutic strategies.
Collapse
Affiliation(s)
- Johnathan Abou-Fadel
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, Texas, USA
| | - Muaz Bhalli
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, Texas, USA
| | - Brian Grajeda
- Department of Biological Sciences, University of Texas at El Paso, El Paso, Texas, USA
| | - Jun Zhang
- Department of Molecular and Translational Medicine (MTM), Texas Tech University Health Science Center El Paso, El Paso, Texas, USA
| |
Collapse
|
20
|
Wu Y, Ma Z, Mai X, Liu X, Li P, Qi X, Li G, Li J. Identification of a Novel Inhibitor of TfR1 from Designed and Synthesized Muriceidine A Derivatives. Antioxidants (Basel) 2022; 11:834. [PMID: 35624697 PMCID: PMC9137542 DOI: 10.3390/antiox11050834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Revised: 04/12/2022] [Accepted: 04/22/2022] [Indexed: 02/06/2023] Open
Abstract
The transferrin receptor 1 (TfR1) plays a key role in cellular iron uptake through its interaction with iron-bound Tf. TfR1 is often reported to be overexpressed in malignant cells, and this increase may be associated with poor prognosis in different types of cancer, which makes it an attractive target for antitumor therapy. The marine natural product Muriceidine A is a potent anticancer agent reported in our previous work. In this study, we designed and synthesized a series of Muriceidine A derivatives and described the systematic investigation into their cytotoxic activities against four tumor cells. Most of the derivatives showed stronger antitumor activity and we found that the introduction of electron-donating groups at position C-2 of unsaturated piperidine was beneficial to anticancer activity and unsaturated piperidine was responsible for the antiproliferative activity. Among these compounds, 12b (methyl at position C-2 of unsaturated piperidine) exhibited the strongest cytotoxicity against MDA-MB-231 cells. Further pharmacological research showed that 12b bound to Transferrin receptor 1 (TfR1) directly caused iron deprivation and ROS imbalance along with the degradations of several oncoproteins, especially FGFR1, through the proteasome pathway; thus, inducing cell cycle arrest and apoptosis in MDA-MB-231 breast cancer cells. Our findings indicate that 12b is a promising lead compound targeting TfR1 for triple negative breast cancer.
Collapse
Affiliation(s)
- Yu Wu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.W.); (Z.M.); (X.M.); (X.L.); (P.L.); (X.Q.)
| | - Zongchen Ma
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.W.); (Z.M.); (X.M.); (X.L.); (P.L.); (X.Q.)
| | - Xiaoyuan Mai
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.W.); (Z.M.); (X.M.); (X.L.); (P.L.); (X.Q.)
| | - Xiaoling Liu
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.W.); (Z.M.); (X.M.); (X.L.); (P.L.); (X.Q.)
| | - Pinglin Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.W.); (Z.M.); (X.M.); (X.L.); (P.L.); (X.Q.)
| | - Xin Qi
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.W.); (Z.M.); (X.M.); (X.L.); (P.L.); (X.Q.)
| | - Guoqiang Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.W.); (Z.M.); (X.M.); (X.L.); (P.L.); (X.Q.)
- Laboratory for Marine Drugs and Bioproducts, Open Studio for Druggability Research of Marine Natural Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| | - Jing Li
- Key Laboratory of Marine Drugs, Chinese Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (Y.W.); (Z.M.); (X.M.); (X.L.); (P.L.); (X.Q.)
- Laboratory for Marine Drugs and Bioproducts, Open Studio for Druggability Research of Marine Natural Products, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266237, China
| |
Collapse
|
21
|
Jafari SH, Jahanmir A, Bahramvand Y, Tahmasebi S, Dallaki M, Nasrollahi E. Association of Estrogen Receptor, Progesterone Receptor, and Human Epidermal Growth Factor Receptor 2 Expression with Breast Cancer Metastasis in Iran. IRANIAN JOURNAL OF MEDICAL SCIENCES 2022; 47:40-47. [PMID: 35017776 PMCID: PMC8743368 DOI: 10.30476/ijms.2021.88366.1906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/27/2021] [Accepted: 02/16/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND Metastasis is an important factor in the survival estimate of patients with breast cancer. The present study aimed to examine the frequency of epidermal growth factor receptor 2 (HER2), estrogen receptor (ER), and progesterone receptor (PR) expression in relation to the metastatic site, pattern, and tumor size in patients with metastatic breast cancer (MBC). METHODS In this retrospective study, the medical records of patients diagnosed with MBC at Motahari Clinic (Shiraz, Iran) during 2017-2019 were examined. Metastasis was confirmed using computed tomography, and a total of 276 patients were included in the study. Based on the expression of receptors, the patients were categorized into luminal A, luminal B, HER2, and TNBC groups. The frequency and percentage of receptors in relation to the metastatic site, size, and pattern were compared using the Chi square test. P<0.05 was considered statistically significant. RESULTS The frequency of receptor positivity in the 276 selected medical records were of the subtype HER2-enriched (n=48), luminal A (n=43), luminal B (n=146), and TNBC (n=39). The most common metastatic sites were the bones (47.1%), lungs (34.4%), liver (27.9%), brain (20.3%), and other organs (12.7%). The first site of metastasis occurred in the bones (36.6%), lungs (17.4%), liver (15.6%), brain (10.5%), and other organs (7.6%). The frequency of receptor expression was different in relation to the first metastatic site (P=0.024). There was a statistically significant difference between the frequency of receptor expression in patients with bone (P=0.036), brain (P=0.031), and lung (P=0.020) metastases. The frequency of receptor expression was also significantly different in relation to the size of liver metastasis (P=0.009). Luminal A and B subtypes showed higher rates of bone metastasis as the first metastatic site. CONCLUSION The difference in the frequency of receptor expression in relation to the metastatic site and tumor size can be used as predictive and prognostic factors in patients with breast cancer.
Collapse
Affiliation(s)
- Seyed Hamed Jafari
- Medical Imaging Research Center, Shiraz University of Medical Sciences, Shiraz, Iran,
Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Armaghan Jahanmir
- Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Yaser Bahramvand
- Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sedigheh Tahmasebi
- Breast Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Manoochehr Dallaki
- Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Elham Nasrollahi
- Department of Radiology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
22
|
Chen Z, Wang M, De Wilde RL, Feng R, Su M, Torres-de la Roche LA, Shi W. A Machine Learning Model to Predict the Triple Negative Breast Cancer Immune Subtype. Front Immunol 2021; 12:749459. [PMID: 34603338 PMCID: PMC8484710 DOI: 10.3389/fimmu.2021.749459] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/30/2021] [Indexed: 12/29/2022] Open
Abstract
Background Immune checkpoint blockade (ICB) has been approved for the treatment of triple-negative breast cancer (TNBC), since it significantly improved the progression-free survival (PFS). However, only about 10% of TNBC patients could achieve the complete response (CR) to ICB because of the low response rate and potential adverse reactions to ICB. Methods Open datasets from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were downloaded to perform an unsupervised clustering analysis to identify the immune subtype according to the expression profiles. The prognosis, enriched pathways, and the ICB indicators were compared between immune subtypes. Afterward, samples from the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset were used to validate the correlation of immune subtype with prognosis. Data from patients who received ICB were selected to validate the correlation of the immune subtype with ICB response. Machine learning models were used to build a visual web server to predict the immune subtype of TNBC patients requiring ICB. Results A total of eight open datasets including 931 TNBC samples were used for the unsupervised clustering. Two novel immune subtypes (referred to as S1 and S2) were identified among TNBC patients. Compared with S2, S1 was associated with higher immune scores, higher levels of immune cells, and a better prognosis for immunotherapy. In the validation dataset, subtype 1 samples had a better prognosis than sub type 2 samples, no matter in overall survival (OS) (p = 0.00036) or relapse-free survival (RFS) (p = 0.0022). Bioinformatics analysis identified 11 hub genes (LCK, IL2RG, CD3G, STAT1, CD247, IL2RB, CD3D, IRF1, OAS2, IRF4, and IFNG) related to the immune subtype. A robust machine learning model based on random forest algorithm was established by 11 hub genes, and it performed reasonably well with area Under the Curve of the receiver operating characteristic (AUC) values = 0.76. An open and free web server based on the random forest model, named as triple-negative breast cancer immune subtype (TNBCIS), was developed and is available from https://immunotypes.shinyapps.io/TNBCIS/. Conclusion TNBC open datasets allowed us to stratify samples into distinct immunotherapy response subgroups according to gene expression profiles. Based on two novel subtypes, candidates for ICB with a higher response rate and better prognosis could be selected by using the free visual online web server that we designed.
Collapse
Affiliation(s)
- Zihao Chen
- Department of Urology, University of Freiburg, Freiburg, Germany
| | - Maoli Wang
- Department of Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Rudy Leon De Wilde
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
| | - Ruifa Feng
- Breast Center of The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Mingqiang Su
- Department of Urology, Zigong Hospital, Affiliated to Southwest Medical University, Zigong, China
| | | | - Wenjie Shi
- University Hospital for Gynecology, Pius-Hospital, University Medicine Oldenburg, Oldenburg, Germany
| |
Collapse
|
23
|
Niţă I, Niţipir C, Toma ŞA, Limbău AM, Pîrvu E, Bădărău IA. The importance of androgen receptors in breast cancer. Med Pharm Rep 2021; 94:273-281. [PMID: 34430848 DOI: 10.15386/mpr-1842] [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: 08/05/2020] [Revised: 11/05/2020] [Accepted: 02/03/2021] [Indexed: 11/23/2022] Open
Abstract
Background and aim Breast cancer (BC) is the most common malignancy among women worldwide, and one of the leading causes of cancer-related deaths in females. For the breast malignant tumors there are numerous targeted therapies, depending on the receptors expressed. Regulating the process of epithelial-mesenchyme transcription, the steroid nuclear receptors are important in invasion and progression of BC cells. Till now, it is known that androgen receptor (AR) is present in about 60-80% of BC cells but, unfortunately, there is no targeted therapy available yet. Methods We revised the recent literature that included the AR mechanism of action in patients diagnosed with breast cancer, the preclinical, retrospective and clinical studies and the aspects related to the prognosis of these patients, depending on the molecular subtype. Results A total of 12 articles were eligible for this review. AR positivity was assessed using immunohistochemistry. Herein, neither 1 nor 10% cut-points were robustly prognostic. AR was an independent prognostic marker of BC outcome, especially in triple negative BC group. Conclusion AR is a potential targeted pathway which can improve the prognostic of AR positive patients with BC. Further preclinical and clinical studies are necessary to clarify the mechanism of action and to establish the drugs which can be used, either alone or in combination.
Collapse
Affiliation(s)
- Irina Niţă
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,Medical Oncology Department, Elias University Emergency Hospital, Bucharest, Romania
| | - Cornelia Niţipir
- Carol Davila University of Medicine and Pharmacy, Bucharest, Romania.,Medical Oncology Department, Elias University Emergency Hospital, Bucharest, Romania
| | | | | | - Edvina Pîrvu
- Medical Oncology Department, "Colţea" Clinical Hospital, Bucharest, Romania
| | | |
Collapse
|
24
|
Sahu R, Pattanayak SP. Strategic Developments & Future Perspective on Gene Therapy for Breast Cancer: Role of mTOR and Brk/ PTK6 as Molecular Targets. Curr Gene Ther 2021; 20:237-258. [PMID: 32807051 DOI: 10.2174/1566523220999200731002408] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 07/16/2020] [Accepted: 07/24/2020] [Indexed: 12/24/2022]
Abstract
Breast cancer is a serious health issue and a major concern in biomedical research. Alteration in major signaling (viz. PI3K-AKT-mTOR, Ras-Raf-MEK-Erk, NF-kB, cyclin D1, JAK-STAT, Wnt, Notch, Hedgehog signaling and apoptotic pathway) contributes to the development of major subtypes of mammary carcinoma such as HER2 positive, TNBC, luminal A and B and normal-like breast cancer. Further, mutation and expression parameters of different genes involved in the growth and development of cells play an important role in the progress of different types of carcinoma, making gene therapy an emerging new therapeutic approach for the management of life-threatening diseases like cancer. The genetic targets (oncogenes and tumor suppressor genes) play a major role in the formation of a tumor. Brk/PTK6 and mTOR are two central molecules that are involved in the regulation of numerous signaling related to cell growth, proliferation, angiogenesis, survival, invasion, metastasis, apoptosis, and autophagy. Since these two proteins are highly upregulated in mammary carcinogenesis, this can be used as targeted genes for the treatment of breast cancer. However, not much work has been done on them. This review highlights the therapeutic significance of Brk and mTOR and their associated signaling in mammary carcinogenesis, which may provide a strategy to develop gene therapy for breast cancer management.
Collapse
Affiliation(s)
- Roja Sahu
- Division of Advanced Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand- 835 215, India
| | - Shakti P Pattanayak
- Division of Advanced Pharmacology, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand- 835 215, India,Department of Pharmacy, Central University of South Bihar (Gaya), Bihar-824 236, India
| |
Collapse
|
25
|
Demeule M, Charfi C, Currie JC, Larocque A, Zgheib A, Kozelko S, Béliveau R, Marsolais C, Annabi B. TH1902, a new docetaxel-peptide conjugate for the treatment of sortilin-positive triple-negative breast cancer. Cancer Sci 2021; 112:4317-4334. [PMID: 34314556 PMCID: PMC8486219 DOI: 10.1111/cas.15086] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 07/23/2021] [Accepted: 07/24/2021] [Indexed: 01/01/2023] Open
Abstract
Triple‐negative breast cancer (TNBC) is a heterogeneous subgroup of cancers which lacks the expression and/or amplification of targetable biomarkers (ie, estrogen receptor, progestrogen receptor, and human epidermal growth factor receptor 2), and is often associated with the worse disease‐specific outcomes than other breast cancer subtypes. Here, we report that high expression of the sortilin (SORT1) receptor correlates with the decreased survival in TNBC patients, and more importantly in those bearing lymph node metastases. By exploiting SORT1 function in ligand internalization, a new anticancer treatment strategy was designed to target SORT1‐positive TNBC‐derived cells both in vitro and in two in vivo tumor xenografts models. A peptide (TH19P01), which requires SORT1 for internalization and to which many anticancer drugs could be conjugated, was developed. In vitro, while the TH19P01 peptide itself did not exert any antiproliferative or apoptotic effects, the docetaxel‐TH19P01 conjugate (TH1902) exerted potent antiproliferative and antimigratory activities when tested on TNBC‐derived MDA‐MB‐231 cells. TH1902 triggered faster and more potent apoptotic cell death than did unconjugated docetaxel. The apoptotic and antimigratory effects of TH1902 were both reversed by two SORT1 ligands, neurotensin and progranulin, and on siRNA‐mediated silencing of SORT1. TH1902 also altered microtubule polymerization and triggered the downregulation of the anti‐apoptotic Bcl‐xL biomarker. In vivo, both i.p. and i.v. administrations of TH1902 led to greater tumor regression in two MDA‐MB‐231 and HCC‐70 murine xenograft models than did docetaxel, without inducing neutropenia. Altogether, the data demonstrates the high in vivo efficacy and safety of TH1902 against TNBC through a SORT1 receptor‐mediated mechanism. This property allows for selective treatment of SORT1‐positive TNBC and makes TH1902 a promising avenue for personalized therapy with the potential of improving the therapeutic window of cytotoxic anticancer drugs such as docetaxel.
Collapse
Affiliation(s)
| | | | | | | | - Alain Zgheib
- Laboratoire d'Oncologie Moléculaire, Université du Québec à Montréal, Montréal, QC, Canada
| | - Sophie Kozelko
- Laboratoire d'Oncologie Moléculaire, Université du Québec à Montréal, Montréal, QC, Canada
| | - Richard Béliveau
- Laboratoire d'Oncologie Moléculaire, Université du Québec à Montréal, Montréal, QC, Canada
| | | | - Borhane Annabi
- Laboratoire d'Oncologie Moléculaire, Université du Québec à Montréal, Montréal, QC, Canada
| |
Collapse
|
26
|
Chakrabarty A, Chakraborty S, Bhattacharya R, Chowdhury G. Senescence-Induced Chemoresistance in Triple Negative Breast Cancer and Evolution-Based Treatment Strategies. Front Oncol 2021; 11:674354. [PMID: 34249714 PMCID: PMC8264500 DOI: 10.3389/fonc.2021.674354] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 06/01/2021] [Indexed: 01/10/2023] Open
Abstract
Triple negative breast cancer (TNBC) is classically treated with combination chemotherapies. Although, initially responsive to chemotherapies, TNBC patients frequently develop drug-resistant, metastatic disease. Chemotherapy resistance can develop through many mechanisms, including induction of a transient growth-arrested state, known as the therapy-induced senescence (TIS). In this paper, we will focus on chemoresistance in TNBC due to TIS. One of the key characteristics of senescent cells is a complex secretory phenotype, known as the senescence-associated secretory proteome (SASP), which by prompting immune-mediated clearance of senescent cells maintains tissue homeostasis and suppresses tumorigenesis. However, in cancer, particularly with TIS, senescent cells themselves as well as SASP promote cellular reprograming into a stem-like state responsible for the emergence of drug-resistant, aggressive clones. In addition to chemotherapies, outcomes of recently approved immune and DNA damage-response (DDR)-directed therapies are also affected by TIS, implying that this a common strategy used by cancer cells for evading treatment. Although there has been an explosion of scientific research for manipulating TIS for prevention of drug resistance, much of it is still at the pre-clinical stage. From an evolutionary perspective, cancer is driven by natural selection, wherein the fittest tumor cells survive and proliferate while the tumor microenvironment influences tumor cell fitness. As TIS seems to be preferred for increasing the fitness of drug-challenged cancer cells, we will propose a few tactics to control it by using the principles of evolutionary biology. We hope that with appropriate therapeutic intervention, this detrimental cellular fate could be diverted in favor of TNBC patients.
Collapse
|
27
|
Paik HJ, Jung YJ, Kim DI, Lee S, Jung CS, Kang SK, Kim JJ, Oh SY, Joo JH, Kim HY. Clinicopathological Features of BRCA1/2 Mutation-Positive Breast Cancer. Oncology 2021; 99:499-506. [PMID: 34098565 DOI: 10.1159/000515790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 03/10/2021] [Indexed: 12/24/2022]
Abstract
PURPOSE The BRCA1/2 gene is the most well-known and studied gene associated with hereditary breast cancer. BRCA1/2 genetic testing is widely performed in high-risk patients of hereditary breast cancer in Korea. This study aimed to investigate the clinicopathological characteristics of BRCA1/2 mutation-positive breast cancer patients. METHODS The clinical data of 188 Korean breast cancer patients who underwent genetic testing of BRCA1/2 mutation between March 2015 and February 2020 at Pusan National University Yangsan Hospital were retrospectively reviewed. The characteristics of breast cancer according to the expression of BRCA1 and BRCA2 mutations were analyzed using the Health Insurance Review and Assessment Service guideline criteria and other clinicopathological factors. RESULTS The factor associated with BRCA1/2 gene expression was cancer stage, and mutation expression was significantly decreased in stage I compared to stage 0 (p = 0.033; odds ratio [OR], 0.169; 95% confidence interval [CI], 0.033-0.867), and there was a tendency to increase in stage II (p = 0.780; OR, 1.150; 95% CI, 0.432-3.064). BRCA1 was significantly associated with triple-negative breast cancer (TNBC) (p = 0.004; OR, 5.887; 95% CI, 1.778-19.498). Gene expression of BRCA2 was significantly reduced under 40 years of age (p = 0.040; OR, 0.198; 95% CI, 0.042-0.930). There was no difference in disease-free survival (p = 0.900) and overall survival (p = 0.733) between the BRCA1/2 mutation-positive and -negative groups. CONCLUSION In this study, the clinicopathological characteristics of breast cancer patients with BRCA1/2 gene mutations were identified. BRCA1 gene expression was highly correlated with TNBC. BRCA1/2 mutation did not have a poor prognosis regarding recurrence and death.
Collapse
Affiliation(s)
- Hyun-June Paik
- Department of Surgery, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Youn Joo Jung
- Department of Surgery, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Dong Il Kim
- Department of Surgery, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Seungju Lee
- Department of Surgery, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Chang Shin Jung
- Department of Surgery, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Seok Kyung Kang
- Department of Surgery, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Jae-Joon Kim
- Hemato-Oncology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - So Yeon Oh
- Hemato-Oncology, Department of Internal Medicine, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Ji Hyeon Joo
- Department of Radiation Oncology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| | - Hyun Yul Kim
- Department of Surgery, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| |
Collapse
|
28
|
Reisenauer KN, Tao Y, Das P, Song S, Svatek H, Patel SD, Mikhail S, Ingros A, Sheesley P, Masi M, Boari A, Evidente A, Kornienko A, Romo D, Taube J. Epithelial-mesenchymal transition sensitizes breast cancer cells to cell death via the fungus-derived sesterterpenoid ophiobolin A. Sci Rep 2021; 11:10652. [PMID: 34017048 PMCID: PMC8137940 DOI: 10.1038/s41598-021-89923-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Accepted: 04/21/2021] [Indexed: 12/30/2022] Open
Abstract
The epithelial-mesenchymal transition (EMT) imparts properties of cancer stem-like cells, including resistance to frequently used chemotherapies, necessitating the identification of molecules that induce cell death specifically in stem-like cells with EMT properties. Herein, we demonstrate that breast cancer cells enriched for EMT features are more sensitive to cytotoxicity induced by ophiobolin A (OpA), a sesterterpenoid natural product. Using a model of experimentally induced EMT in human mammary epithelial (HMLE) cells, we show that EMT is both necessary and sufficient for OpA sensitivity. Moreover prolonged, sub-cytotoxic exposure to OpA is sufficient to suppress EMT-imparted CSC features including sphere formation and resistance to doxorubicin. In vivo growth of CSC-rich mammary cell tumors, is suppressed by OpA treatment. These data identify a driver of EMT-driven cytotoxicity with significant potential for use either in combination with standard chemotherapy or for tumors enriched for EMT features.
Collapse
Affiliation(s)
| | - Yongfeng Tao
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Provas Das
- Department of Biology, Baylor University, Waco, TX, USA
| | - Shuxuan Song
- Department of Biology, Baylor University, Waco, TX, USA
| | | | | | | | - Alec Ingros
- Department of Biology, Baylor University, Waco, TX, USA
| | | | - Marco Masi
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Naples, Italy
| | - Angela Boari
- Institute of Sciences and Food Production, CNR, Bari, Italy
| | - Antonio Evidente
- Department of Chemical Sciences, University of Naples Federico II, Complesso Universitario Monte Sant'Angelo, Naples, Italy
| | - Alexander Kornienko
- Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX, USA
| | - Daniel Romo
- Department of Chemistry and Biochemistry, Baylor University, Waco, TX, USA
| | - Joseph Taube
- Department of Biology, Baylor University, Waco, TX, USA.
| |
Collapse
|
29
|
Yang PT, Wu WS, Wu CC, Shih YN, Hsieh CH, Hsu JL. Breast cancer recurrence prediction with ensemble methods and cost-sensitive learning. Open Med (Wars) 2021; 16:754-768. [PMID: 34027105 PMCID: PMC8122465 DOI: 10.1515/med-2021-0282] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 03/11/2021] [Accepted: 04/03/2021] [Indexed: 11/15/2022] Open
Abstract
Breast cancer is one of the most common cancers in women all over the world. Due to the improvement of medical treatments, most of the breast cancer patients would be in remission. However, the patients have to face the next challenge, the recurrence of breast cancer which may cause more severe effects, and even death. The prediction of breast cancer recurrence is crucial for reducing mortality. This paper proposes a prediction model for the recurrence of breast cancer based on clinical nominal and numeric features. In this study, our data consist of 1,061 patients from Breast Cancer Registry from Shin Kong Wu Ho-Su Memorial Hospital between 2011 and 2016, in which 37 records are denoted as breast cancer recurrence. Each record has 85 features. Our approach consists of three stages. First, we perform data preprocessing and feature selection techniques to consolidate the dataset. Among all features, six features are identified for further processing in the following stages. Next, we apply resampling techniques to resolve the issue of class imbalance. Finally, we construct two classifiers, AdaBoost and cost-sensitive learning, to predict the risk of recurrence and carry out the performance evaluation in three-fold cross-validation. By applying the AdaBoost method, we achieve accuracy of 0.973 and sensitivity of 0.675. By combining the AdaBoost and cost-sensitive method of our model, we achieve a reasonable accuracy of 0.468 and substantially high sensitivity of 0.947 which guarantee almost no false dismissal. Our model can be used as a supporting tool in the setting and evaluation of the follow-up visit for early intervention and more advanced treatments to lower cancer mortality.
Collapse
Affiliation(s)
- Pei-Tse Yang
- Department of Computer Science and Information Engineering, Fu Jen Catholic University, New Taipei City, Taiwan, Republic of China
| | - Wen-Shuo Wu
- Department of Computer Science and Information Engineering, Fu Jen Catholic University, New Taipei City, Taiwan, Republic of China
| | - Chia-Chun Wu
- Department of Computer Science and Information Engineering, Fu Jen Catholic University, New Taipei City, Taiwan, Republic of China
| | - Yi-Nuo Shih
- Department of Occupational Therapy, Fu Jen Catholic University, New Taipei City, Taiwan, Republic of China
| | - Chung-Ho Hsieh
- Department of General Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan, Republic of China
| | - Jia-Lien Hsu
- Department of Computer Science and Information Engineering, Fu Jen Catholic University, New Taipei City, Taiwan, Republic of China
| |
Collapse
|
30
|
Chen Q, Song H, Yu J, Kim K. Current Development and Applications of Super-Resolution Ultrasound Imaging. SENSORS 2021; 21:s21072417. [PMID: 33915779 PMCID: PMC8038018 DOI: 10.3390/s21072417] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023]
Abstract
Abnormal changes of the microvasculature are reported to be key evidence of the development of several critical diseases, including cancer, progressive kidney disease, and atherosclerotic plaque. Super-resolution ultrasound imaging is an emerging technology that can identify the microvasculature noninvasively, with unprecedented spatial resolution beyond the acoustic diffraction limit. Therefore, it is a promising approach for diagnosing and monitoring the development of diseases. In this review, we introduce current super-resolution ultrasound imaging approaches and their preclinical applications on different animals and disease models. Future directions and challenges to overcome for clinical translations are also discussed.
Collapse
Affiliation(s)
- Qiyang Chen
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Hyeju Song
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea;
| | - Jaesok Yu
- Department of Robotics Engineering, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea;
- DGIST Robotics Research Center, Daegu Gyeongbuk Institute of Science & Technology (DGIST), Daegu 42988, Korea
- Correspondence: (J.Y.); (K.K.)
| | - Kang Kim
- Department of Bioengineering, School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA;
- Center for Ultrasound Molecular Imaging and Therapeutics, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Division of Cardiology, Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Mechanical Engineering and Materials Science, School of Engineering, University of Pittsburgh, Pittsburgh, PA 15261, USA
- Correspondence: (J.Y.); (K.K.)
| |
Collapse
|
31
|
Kaul K, Misri S, Ramaswamy B, Ganju RK. Contribution of the tumor and obese microenvironment to triple negative breast cancer. Cancer Lett 2021; 509:115-120. [PMID: 33798632 DOI: 10.1016/j.canlet.2021.03.024] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 03/01/2021] [Accepted: 03/24/2021] [Indexed: 01/01/2023]
Abstract
The growing burden of obesity and incidence of the aggressive triple negative breast cancer (TNBC) is a challenge, especially amongst vulnerable populations with unmet medical needs and higher mortality from breast cancer. While some mechanisms linking obesity and TNBC have been identified, the complex nature of pathogenesis, in both obesity as well as TNBC poses a real challenge in establishing a causative role of obesity in risk of TNBC. In this review article, we discuss pathological mechanisms identified in the tumor microenvironment (TME) as well as the obese microenvironment (OME), such as inflammation, insulin resistance and survival pathways that contribute to the development and progression of TNBC. Insights into the cross-talk between TME and OME, and their contribution to TNBC development and progression, may pave the way for personalized therapies against TNBC progression, relapse and metastasis.
Collapse
Affiliation(s)
- Kirti Kaul
- Comprehensive Cancer Center, USA; Department of Pathology, USA
| | | | | | - Ramesh K Ganju
- Comprehensive Cancer Center, USA; Department of Pathology, USA.
| |
Collapse
|
32
|
Ouyang J, Liu Z, Yuan X, Long C, Chen X, Wang Y, Liu L, Liu S, Liang H. LncRNA PRNCR1 Promotes Breast Cancer Proliferation and Inhibits Apoptosis by Modulating microRNA-377/CCND2/MEK/MAPK Axis. Arch Med Res 2021; 52:471-482. [PMID: 33608112 DOI: 10.1016/j.arcmed.2021.01.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/27/2020] [Accepted: 01/21/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) have recently become the vital gene regulators in diverse cancers. In our study, we purposed to inquiry into the mechanisms of lncRNA PRNCR1 in breast cancer via microRNA-377 (miR-377)/CCND2/MEK/MAPK axis. METHODS PRNCR1 expression in breast cancer tissues was detected, and the correlation between PRNCR1 expression and prognostic survival was analyzed. The expressions of PRNCR1 and miR-377 in breast cancer cell lines were detected. Relationships among PRNCR1, miR-377 and CCND2 were confirmed by luciferase activity, RNA pull-down or RIP assays. Breast cancer cells were introduced with silenced PRNCR1 or restored miR-377 to explore their functions in malignant phenotype of breast cancer cells. The expression of MEK/MAPK pathway-related proteins was determined by western blot analysis. RESULTS PRNCR1 was highly expressed and miR-377 was poorly expressed in patients with breast cancer, and patients with high expression of PRNCR1 had a poor prognosis. PRNCR1 silencing or miR-377 overexpression resulted in suppressed breast cancer cell proliferation ability, blocked cell cycle process and induced apoptosis. PRNCR1 regulated CCND2 expression by competitively binding to miR-377. CCND2 activated the MEK/MAPK pathway, and after treatment with Mirdametinib, the MEK/MAPK pathway was inhibited, which was found to retard breast cancer growth. CONCLUSION Our study highlights that lncRNA PRNCR1 may competitively bind to miR-377, leading to upregulated CCND2, which in turn activated MEK/MAPK pathway to promote breast cancer growth.
Collapse
Affiliation(s)
- Jian Ouyang
- Department of Laboratory of Cancer Research, Pingxiang Health Vocational College, Anyuan District, Pingxiang, Jiangxi, P.R. China
| | - Zilong Liu
- Department of Laboratory of Cancer Research, Pingxiang Health Vocational College, Anyuan District, Pingxiang, Jiangxi, P.R. China
| | - Xiaobing Yuan
- Department of Laboratory of Cancer Research, Pingxiang Health Vocational College, Anyuan District, Pingxiang, Jiangxi, P.R. China
| | - Chunping Long
- Department of Laboratory of Cancer Research, Pingxiang Health Vocational College, Anyuan District, Pingxiang, Jiangxi, P.R. China
| | - Xia Chen
- Department of Laboratory of Cancer Research, Pingxiang Health Vocational College, Anyuan District, Pingxiang, Jiangxi, P.R. China
| | - Yongpeng Wang
- Department of Laboratory of Cancer Research, Pingxiang Health Vocational College, Anyuan District, Pingxiang, Jiangxi, P.R. China
| | - Lu Liu
- Department of Laboratory of Cancer Research, Pingxiang Health Vocational College, Anyuan District, Pingxiang, Jiangxi, P.R. China
| | - Shaohua Liu
- Department of Surgical Oncology, Jiangxi Pingxiang People's Hospital, Pingxiang, Jiangxi, P.R. China
| | - Hui Liang
- Department of Laboratory of Cancer Research, Pingxiang Health Vocational College, Anyuan District, Pingxiang, Jiangxi, P.R. China.
| |
Collapse
|
33
|
Huo X, Li J, Zhao F, Ren D, Ahmad R, Yuan X, Du F, Zhao J. The role of capecitabine-based neoadjuvant and adjuvant chemotherapy in early-stage triple-negative breast cancer: a systematic review and meta-analysis. BMC Cancer 2021; 21:78. [PMID: 33468087 PMCID: PMC7816481 DOI: 10.1186/s12885-021-07791-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 01/05/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND The role of capecitabine in neoadjuvant and adjuvant chemotherapy for early-stage triple-negative breast cancer (TNBC) is highly controversial. Our meta-analysis was designed to further elucidate the effects of capecitabine on survival in early-stage TNBC patients and its safety. METHODS PubMed, Embase, and papers presented at several main conferences were searched up to December 19, 2019, to investigate capecitabine-based versus capecitabine-free neoadjuvant and adjuvant chemotherapy in TNBC patients. Heterogeneity was assessed using I2 test, combined with hazard ratios (HRs) and odds ratios (ORs) with 95% confidence intervals (CI) computed for disease-free survival (DFS), overall survival (OS), and over grade 3 adverse events (AEs). RESULTS A total of 9 randomized clinical trials and 3842 TNBC patients were included. Overall, the combined capecitabine regimens in neoadjuvant and adjuvant chemotherapy showed significantly improved DFS (HR = 0.75; 95% CI, 0.65-0.86; P < 0.001) and OS (HR = 0.63; 95% CI, 0.53-0.77; P < 0.001). In subgroup analysis, there were improvements in DFS in the groups with addition of capecitabine (HR = 0.64; 95% CI, 0.53-0.78; P < 0.001), adjuvant chemotherapy (HR = 0.73; 95% CI, 0.63-0.85; P < 0.001), and lymph node positivity (HR = 0.62; 95% CI, 0.44-0.86; P = 0.005). Capecitabine regimens were related to higher risks of diarrhea (OR = 2.88, 95% CI 2.23-3.74, P < 0.001), stomatitis (OR = 2.01, 95% CI 1.53-2.64, P < 0.001) and hand-foot syndrome (OR = 8.67, 95% CI 6.70-11.22, P < 0.001). CONCLUSION This meta-analysis showed that neoadjuvant and adjuvant chemotherapy combined with capecitabine significantly improved both DFS and OS in early-stage TNBC patients with tolerable AEs. There were benefits to DFS in the groups with the addition of capecitabine, adjuvant chemotherapy, and lymph node positivity.
Collapse
Affiliation(s)
- Xingfa Huo
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000 China
| | - Jinming Li
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000 China
| | - Fuxing Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000 China
| | - Dengfeng Ren
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000 China
| | - Raees Ahmad
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000 China
| | - Xinyue Yuan
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000 China
| | - Feng Du
- Peking University Cancer Hospital and Institute, Beijing, 100142 China
| | - Jiuda Zhao
- Breast Disease Diagnosis and Treatment Center of Affiliated Hospital of Qinghai University & Affiliated Cancer Hospital of Qinghai University, Xining, 810000 China
| |
Collapse
|
34
|
MicroRNA-205-5p targets the HOXD9-Snail1 axis to inhibit triple negative breast cancer cell proliferation and chemoresistance. Aging (Albany NY) 2021; 13:3945-3956. [PMID: 33428601 PMCID: PMC7906129 DOI: 10.18632/aging.202363] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 10/31/2020] [Indexed: 12/11/2022]
Abstract
MicroRNA-205 (miR-205) is believed to be related to the progress of tumors. HOXD9 has been proved to be expressed abnormally in several kinds of cancers. However, the role of miR-205 and HOXD9 in breast cancer remains unclear. The biological role of miR-205 in breast cancer cell proliferation and chemoresistance was investigated. The expression of miR-205 in clinical tissues and breast cancer cell lines were analyzed using quantitative real-time PCR test (qRT-PCR). Overexpression and knockdown models of miR-205 were established to study cell proliferation and chemotherapy-resistant. Moreover, the potential relationships between miR-205 and HOXD9/Snail1 were measured using qRT-PCR, western blot, and chemotherapy-resistant study. miR-205 was lowly expressed in breast cancer tissues and cell lines. Overexpression of miR-205 could inhibit cell proliferation and chemotherapy-resistance. Moreover, we proved that miR-205 could target the HOXD9-Snail1 axis to suppress triple negative breast cancer cell proliferation and chemoresistance. The activation of Snail1 gene by HOXD9 was also proved in this study. The present study may provide a novel insight for the therapeutic strategies of breast cancer through targeting miR-205/HOXD9/Snail1.
Collapse
|
35
|
Trailblazing perspectives on targeting breast cancer stem cells. Pharmacol Ther 2021; 223:107800. [PMID: 33421449 DOI: 10.1016/j.pharmthera.2021.107800] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/30/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Breast cancer (BCa) is one of the most prevalent malignant tumors affecting women's health worldwide. The recurrence and metastasis of BCa have made it a long-standing challenge to achieve remission-persistent or disease-undetectable clinical outcomes. Cancer stem cells (CSCs) possess the ability to self-renew and generate heterogeneous tumor bulk. The existence of CSCs has been found to be vital in the initiation, metastasis, therapy resistance, and recurrence of tumors across cancer types. Because CSCs grow slowly in their dormant state, they are insensitive to conventional chemotherapies; however, when CSCs emerge from their dormant state and become clinically evident, they usually acquire genetic traits that make them resistant to existing therapies. Moreover, CSCs also show evidence of acquired drug resistance in synchrony with tumor relapses. The concept of CSCs provides a new treatment strategy for BCa. In this review, we highlight the recent advances in research on breast CSCs and their association with epithelial-mesenchymal transition (EMT), circulating tumor cells (CTCs), plasticity of tumor cells, tumor microenvironment (TME), T-cell modulatory protein PD-L1, and non-coding RNAs. On the basis that CSCs are associated with multiple dysregulated biological processes, we envisage that increased understanding of disease sub-classification, selected combination of conventional treatment, molecular aberration directed therapy, immunotherapy, and CSC targeting/sensitizing strategy might improve the treatment outcome of patients with advanced BCa. We also discuss novel perspectives on new drugs and therapeutics purposing the potent and selective expunging of CSCs.
Collapse
|
36
|
Jiang J, Feng M, Jacob A, Li LZ, Xu HN. Optical Redox Imaging Differentiates Triple-Negative Breast Cancer Subtypes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1269:253-258. [PMID: 33966226 PMCID: PMC8183433 DOI: 10.1007/978-3-030-48238-1_40] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Triple-negative breast cancer (TNBC) is a highly diverse group of cancers with limited treatment options, responsible for about 15% of all breast cancers. TNBC cells differ from each other in many ways such as gene expression, metabolic activity, tumorigenicity, and invasiveness. Recently, many research and clinical efforts have focused on metabolically targeted therapy for TNBC. Metabolic characterization of TNBC cell lines can facilitate the assessment of therapeutic effects and assist in metabolic drug development. Herein, we used optical redox imaging (ORI) techniques to characterize TNBC subtypes metabolically. We found that various TNBC cell lines had differing redox statuses (levels of reduced nicotinamide adenine dinucleotide (NADH), oxidized flavin adenine dinucleotide (FAD), and the redox ratio (FAD/(NADH+FAD)). We then metabolically perturbed the cells with mitochondrial inhibitors and an uncoupler and performed ORI accordingly. As expected, we observed that these TNBC cell lines had similar response patterns to the metabolic perturbations. However, they exhibited differing redox plasticity. These results suggest that subtypes of TNBC cells are different metabolically and that ORI can serve as a sensitive technique for the metabolic profiling of TNBC cells.
Collapse
Affiliation(s)
- Jinxia Jiang
- Britton Chance Laboratory of Redox Imaging, Department of Radiology and Johnson Research Foundation, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Min Feng
- Britton Chance Laboratory of Redox Imaging, Department of Radiology and Johnson Research Foundation, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Annemarie Jacob
- Britton Chance Laboratory of Redox Imaging, Department of Radiology and Johnson Research Foundation, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lin Z Li
- Britton Chance Laboratory of Redox Imaging, Department of Radiology and Johnson Research Foundation, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - He N Xu
- Britton Chance Laboratory of Redox Imaging, Department of Radiology and Johnson Research Foundation, Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Huang X, Shao D, Wu H, Zhu C, Guo D, Zhou Y, Chen C, Lin Y, Lu T, Zhao B, Wang C, Sun Q. Genomic Profiling Comparison of Germline BRCA and Non- BRCA Carriers Reveals CCNE1 Amplification as a Risk Factor for Non- BRCA Carriers in Patients With Triple-Negative Breast Cancer. Front Oncol 2020; 10:583314. [PMID: 33194720 PMCID: PMC7662137 DOI: 10.3389/fonc.2020.583314] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/16/2020] [Indexed: 12/11/2022] Open
Abstract
Background: Differences in genomic profiling and immunity-associated parameters between germline BRCA and non-BRCA carriers in TNBC with high tumor burden remain unexplored. This study aimed to compare the differences and explore potential prognostic predictors and therapeutic targets. Methods: The study cohort included 21 consecutive TNBC cases with germline BRCA1/2 mutations and 54 non-BRCA carriers with a tumor size ≥ 2 cm and/or ≥1 affected lymph nodes. Differences in clinicopathological characteristics and genomic profiles were analyzed through next-generation sequencing. Univariate Kaplan-Meier analysis and Cox regression model were applied to survival analysis. Immunohistochemistry was used to confirm the consistency between CCNE1 amplification and cyclin E1 protein overexpression. Results: The cohort included 16 and five patients with germline BRCA1 and BRCA2 mutations, respectively. Patients with germline BRCA1/2 mutations were diagnosed at a significantly younger age and were more likely to have a family history of breast and/or ovarian cancer. Six non-BRCA carriers (11.11%) carried germline mutations in other cancer susceptibility genes, including five mutations in five homologous recombination repair (HRR) pathway genes (9.26%) and one mutation in MSH3 (1.85%). Somatic mutations in HRR pathway genes were found in 22.22 and 14.29% of the non-BRCA and BRCA carriers, respectively. PIK3CA missense mutation (p = 0.046) and CCNE1 amplification (p = 0.2) were found only in the non-BRCA carriers. The median tumor mutation burden (TMB) was 4.1 Muts/Mb, whereas none of the cases had high microsatellite instability (MSI). BRCA status did not affect disease-free survival (DFS, p = 0.15) or overall survival (OS, p = 0.52). CCNE1 amplification was an independent risk factor for DFS in non-BRCA carriers with TNBC (HR 13.07, 95% CI 2.47-69.24, p = 0.003). Consistency between CCNE1 amplification and cyclin E1 protein overexpression was confirmed with an AUC of 0.967 for cyclin E1 signal intensity. Conclusions: We found differences in genetic alterations between germline BRCA and non-BRCA carriers with TNBC and a high tumor burden. TMB and MSI may not be suitable predictors of TNBC for immune checkpoint inhibitors. Notably, CCNE1 amplification is a novel potential prognostic marker and therapeutic target for non-BRCA carriers with TNBC. Cyclin E1 may be used instead of CCNE1 to improve clinical applicability.
Collapse
Affiliation(s)
- Xin Huang
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Di Shao
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - Huanwen Wu
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | | | - Dan Guo
- Clinical Biobank, Medical Science Research Center, Peking Union Medical College Hospital, Beijing, China
| | - Yidong Zhou
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Chang Chen
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Yan Lin
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Tao Lu
- Department of Pathology, Peking Union Medical College Hospital, Beijing, China
| | - Bin Zhao
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Changjun Wang
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, China
| | - Qiang Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
38
|
Downregulation of miR-205 contributes to epithelial-mesenchymal transition and invasion in triple-negative breast cancer by targeting HMGB1-RAGE signaling pathway. Anticancer Drugs 2020; 30:225-232. [PMID: 30334817 PMCID: PMC6410973 DOI: 10.1097/cad.0000000000000705] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Our aim was to study the regulatory molecule networks involved in the epithelial-to-mesenchymal transition and thus promoting the early onset of metastasis in triple-negative breast cancer (TNBC). Forty pairs of human TNBC and their adjacent normal breast tissues were analyzed by real-time PCR and immunochemistry to demonstrate the correlation between the miR-205 expression and clinicopathological characteristics. In vitro, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide assay, cell migration, and invasion assay were used to detect the cell growth and invasive ability of TNBC cells after upregulation or downregulation of miR-205 expression. Luciferase reporter assay was used to confirm the potential target directly influenced by miR-205. Our results showed that miR-205 abnormal expression may be involved and associated with the biological traits of TNBC. Ectopic expression of miR-205 not only inhibited cell growth, but also suppressed migration and invasion of mesenchymal-like TNBC cells. In addition, we found that overexpression of miR-205 significantly suppressed HMGB1 by binding its 3′-untranslated region, and that miR-205 was inversely correlated with the expression of HMGB1 and RAGE in cell lines and clinical samples. Our study illustrated that miR-205 was a tumor suppressor in TNBC, which attenuated the viability and the acquisition of the epithelial-to-mesenchymal transition phenotype TNBC cells at least partially exerted through targeting of HMGB1–RAGE signaling pathway.
Collapse
|
39
|
Zubareva EY, Sen’chukova MA. The modern views of the clinical, morphological and molecular biological predictors of breast cancer sensitivity to chemotherapy. ADVANCES IN MOLECULAR ONCOLOGY 2020. [DOI: 10.17650/2313-805x-2020-7-2-20-28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In the treatment of breast cancer, the neoadjuvant chemotherapy is vitally important and the evaluation of its effectiveness is crucial for determining the further therapy treatment, as well as the prognosis of the disease. This review provides current data of the physical, instrumental, morphological, molecular biology and genetics analysis used for the estimation of the neoadjuvant treatment effectiveness. Thus, review discusses the data concerning association of the disease peculiarities with the efficient therapeutic response to neoadjuvant chemotherapy including characteristics of patients (age, status of regional lymph nodes, presence of the lymphovascular invasion) and tumors (size, histological type, degree of differentiation, severity of the lymphoid tumor infiltration, molecular biological and genetic peculiarities). Particular attention is paid to such a promising predictive marker of the breast cancer response to chemotherapy as the level of tissue hypoxia. This section discusses the currently known mechanisms that might enable the effect of tissue hypoxia on the sensitivity of the tumor to drug treatment. The prospects for the use of a comprehensive analysis of predictive markers of the effectiveness of chemotherapeutic treatment are discussed.
Collapse
Affiliation(s)
- E. Yu. Zubareva
- Orenburg Regional Clinical Oncology Dispensary; Orenburg State Medical University, Ministry of Health of Russia
| | - M. A. Sen’chukova
- Orenburg Regional Clinical Oncology Dispensary; Orenburg State Medical University, Ministry of Health of Russia
| |
Collapse
|
40
|
MGMT promoter methylation in triple negative breast cancer of the GeparSixto trial. PLoS One 2020; 15:e0238021. [PMID: 32841306 PMCID: PMC7446962 DOI: 10.1371/journal.pone.0238021] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 08/06/2020] [Indexed: 12/27/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is typically treated with chemotherapeutic agents, including carboplatin (Cb), an DNA platinating agent. The O6-methylguanine-DNA-methyltransferase gene (MGMT) encodes for the protein O6-alkylguanine-DNA-alkyltransferase (MGMT protein). MGMT protein is involved in DNA repair mechanisms to remove mutagenic and cytotoxic adducts from O6-guanine in DNA. In glioblastoma multiforme, MGMT methylation status is a predictive biomarker for increased response to temozolomide therapy. It has been suggested, that MGMT protein may have relevance for cellular adaptation and could have an influence on resistance to carboplatin therapy. We investigated the influence of MGMT promoter methylation on pathologic complete response and survival of patients with TNBC treated in the neoadjuvant GeparSixto trial. In 174 of 210 available TNBC tumors a valid MGMT promoter methylation status was determined by pyrosequencing of 5 CpG islands. In 21.8%, we detected a mean MGMT promoter methylation >10%. Overall, MGMT promoter methylation was not significantly associated with pathological complete response (pCR) rate. After stratification for the two therapy arms with and without Cb no statistically significant differences in therapy response rates between the two MGMT promoter methylation groups could be observed. Our results show that different MGMT promoter methylation status is not related to different chemotherapy response rates in the TNBC setting in GeparSixto.
Collapse
|
41
|
Gonçalves BÔP, De Andrade WP, Da Conceição Braga L, Fialho SL, Silva LM. Epithelial-to-mesenchymal transition markers are differentially expressed in epithelial cancer cell lines after everolimus treatment. Oncol Lett 2020; 20:158. [PMID: 32934726 PMCID: PMC7471649 DOI: 10.3892/ol.2020.12019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/14/2020] [Indexed: 12/15/2022] Open
Abstract
The epithelial-to-mesenchymal transition (EMT) is a phenomenon during which cancer epithelial cells undergo changes in plasticity and lose cell-cell adhesion with consequent remodeling of the extracellular matrix and development of mesenchymal characteristics. Long non-coding RNAs (lncRNAs) have been described as EMT modulation markers, becoming a promising target in the development of new therapies for cancer. The present study aimed to investigate the role of everolimus at 100 nM as inductor of the EMT phenomenon in cell lines derived from human breast (BT-549), colorectal (RKO-AS45-1) and ovary (TOV-21G) cancer. The integrity of cellular junctions was monitored using an in vitro model of epithelial resistance. The results demonstrated that the EMT genes ZEB1, TWIST1 and TGFB1 were differentially expressed in cells treated with everolimus compared with in untreated cells. lncRNA HOTAIR was upregulated post-treatment only in BT-549 cells compared with in untreated cells. After treatment with everolimus, the intensity of fluorescence of P-cadherin decreased, and that of fibronectin increased in RKO-AS45-1 and TOV-21G cells compared with control cells. The transepithelial electrical resistance at the RKO-AS45-1 monolayer treated with everolimus started to decrease at 48 h. The changes in the gene expression and epithelial resistance may confirm the role of everolimus in EMT.
Collapse
Affiliation(s)
- Bryan Ôrtero Perez Gonçalves
- Cellular Biology, Research and Development Department, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| | - Warne Pedro De Andrade
- Hematology and Oncology Nucleus, Grupo Oncoclinicas, Belo Horizonte, Minas Gerais 30140001, Brazil.,Department of Obstetrics and Gynecology, School of Medicine, São Paulo State University, Botucatu, São Paulo 18618687, Brazil
| | - Letícia Da Conceição Braga
- Cellular Biology, Research and Development Department, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| | - Sílvia Ligório Fialho
- Pharmaceutical Research and Development, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| | - Luciana Maria Silva
- Cellular Biology, Research and Development Department, Ezequiel Dias Foundation, Belo Horizonte, Minas Gerais 30510-010, Brazil
| |
Collapse
|
42
|
Zagorac I, Lončar B, Dmitrović B, Kralik K, Kovačević A. Correlation of folate receptor alpha expression with clinicopathological parameters and outcome in triple negative breast cancer. Ann Diagn Pathol 2020; 48:151596. [PMID: 32829070 DOI: 10.1016/j.anndiagpath.2020.151596] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/02/2020] [Accepted: 06/05/2020] [Indexed: 01/03/2023]
Abstract
Folate receptor alpha (FRα) is a membrane-bound protein with a high affinity for folate, which is necessary for the biosynthesis of amino acids and nucleotide bases. It has been shown to be a potential prognostic and therapeutic target, primarily in lung and ovarian cancer, as well as in breast cancer. The aim of this study was to examine FRα expression in a cohort of patients with triple negative breast cancer (TNBC), in correlation with clinicopathological parameters and prognostic factors. By using polyclonal FRα antibody on archival paraffin blocks immunohistochemistry was performed. To evaluate the expression of FRα, H-score was used, which marks both the proportion of stained cells and the intensity of staining. Statistical analysis correlating FRα expression with clinicopathologic parameters and clinical outcome were performed. FRα was expressed in most of the patients (85%). Significant correlation of expression and histologic grade (Mann Whitney U test, P = 0,03) and type of tumor (P = 0,02), was found. It was noticed that with higher Ki-67 proliferation index values, H-score has lower values (r = -0,284, P = 0,006). Multivariant regression analysis (Cox regression, Stepwise method) showed H-score as a significant predictor for the risk of disease recurrence (OR = 1,005, P = 0,04). No correlation between FRα expression and overall survival (OS) and disease-free survival (DFS) was found. In conclusion, FRα is highly expressed in TNBC, and, given the correlation with clinicopathological parameters, subpopulation of patients could be identified that could be potential targets for new therapeutic perspectives in the treatment of this breast cancer subtype.
Collapse
Affiliation(s)
- Irena Zagorac
- Department of Pathology and Forensic Medicine, Osijek University Hospital, Josipa Huttlera 4, HR-31000 Osijek, Croatia; Faculty of Medicine, University of Osijek, Josipa Huttlera 4, HR-31000 Osijek, Croatia.
| | - Branka Lončar
- Faculty of Medicine, University of Osijek, Josipa Huttlera 4, HR-31000 Osijek, Croatia; Department of Clinical Cytology, Osijek University Hospital, Josipa Huttlera 4, HR-31000 Osijek, Croatia
| | - Branko Dmitrović
- Department of Pathology and Forensic Medicine, Osijek University Hospital, Josipa Huttlera 4, HR-31000 Osijek, Croatia; Faculty of Medicine, University of Osijek, Josipa Huttlera 4, HR-31000 Osijek, Croatia; Faculty of Dental Medicine and Health, University of Osijek, Crkvena 21, HR-31000 Osijek, Croatia
| | - Kristina Kralik
- Faculty of Medicine, University of Osijek, Josipa Huttlera 4, HR-31000 Osijek, Croatia
| | - Andrej Kovačević
- Department of Pathology and Forensic Medicine, Osijek University Hospital, Josipa Huttlera 4, HR-31000 Osijek, Croatia; Faculty of Medicine, University of Osijek, Josipa Huttlera 4, HR-31000 Osijek, Croatia
| |
Collapse
|
43
|
Zecchin D, Moore C, Michailidis F, Horswell S, Rana S, Howell M, Downward J. Combined targeting of G protein-coupled receptor and EGF receptor signaling overcomes resistance to PI3K pathway inhibitors in PTEN-null triple negative breast cancer. EMBO Mol Med 2020; 12:e11987. [PMID: 32672423 PMCID: PMC7411640 DOI: 10.15252/emmm.202011987] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 06/09/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
Triple-negative breast cancer (TNBC) has poorer prognosis compared to other types of breast cancers due to the lack of effective therapies and markers for patient stratification. Loss of PTEN tumor suppressor gene expression is a frequent event in TNBC, resulting in over-activation of the PI 3-kinase (PI3K) pathway and sensitivity to its inhibition. However, PI3K pathway inhibitors show limited efficacy as monotherapies on these tumors. We report a whole-genome screen to identify targets whose inhibition enhanced the effects of different PI3K pathway inhibitors on PTEN-null TNBC. This identified a signaling network that relies on both the G protein-coupled receptor for thrombin (PAR1/F2R) and downstream G protein βγ subunits and also epidermal growth factor receptor (EGFR) for the activation of the PI3K isoform p110β and AKT. Compensation mechanisms involving these two branches of the pathway could bypass PI3K blockade, but combination targeting of both EGFR and PI3Kβ suppressed ribosomal protein S6 phosphorylation and exerted anti-tumor activity both in vitro and in vivo, suggesting a new potential therapeutic strategy for PTEN-null TNBC.
Collapse
Affiliation(s)
| | | | | | | | - Sareena Rana
- Oncogene BiologyFrancis Crick InstituteLondonUK
- Lung Cancer GroupInstitute of Cancer ResearchLondonUK
| | - Michael Howell
- High Throughput Screening LaboratoriesFrancis Crick InstituteLondonUK
| | - Julian Downward
- Oncogene BiologyFrancis Crick InstituteLondonUK
- Lung Cancer GroupInstitute of Cancer ResearchLondonUK
| |
Collapse
|
44
|
Prognostic Role of Immune Markers in Triple Negative Breast Carcinoma. Pathol Oncol Res 2020; 26:2733-2745. [PMID: 32681436 DOI: 10.1007/s12253-020-00874-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/09/2020] [Indexed: 12/21/2022]
Abstract
Tumor immune microenvironment (TIME) is a significant prognostic parameter for triple negative breast carcinomas (TNBC) due to being a target for immunotherapeutic agents and its essential role during the cancer immunoediting process. In this study, CD8, FOXP3, CD163, PD-L1/SP142 and PD-L1/SP263 antibodies were examined in a sample of 51 TNBC cases. Patients who received neoadjuvant therapy were excluded. CD8, FOXP3 and CD163 antibodies were evaluated separately in intratumoral area (ITA) and tumor stroma (TS). PD-L1 status was also examined in tumor cells (TC) and immune cells (IC) using both SP142 and SP263 antibodies. In multivariate Cox regressions, the only antibody that was found to be significantly associated with survival was SP142. SP142-positivity in TC and IC was related to increased overall survival. Higher CD163 expression in ITA and SP263-positivity in IC were associated with younger age. Lymphatic/angioinvasion was more frequent in cases with negative/low CD8 and FOXP3 expressions. Moreover, metastatic axillary lymph node(s) was associated with negative/low FOXP3 expression in TS. CD8, FOXP3, CD163, SP142 and SP263 expressions were positively correlated with each other, except a mild discordance caused by CD163 in ITA. Although PD-L1 status with both SP142 and SP263 antibodies were concordant in the majority of cases, 33.3% and 13.7% of the cases showed SP142-negative/SP263-positive pattern in TC and IC respectively. In conclusion, we suggest that composition, density and localization of the immune cells and the check point molecules are important prognostic parameters in TNBC. Immunohistochemistry can be used as an accessible and less expensive tool to demonstrate TIME.
Collapse
|
45
|
Suman P, Mishra S, Chander H. High formin binding protein 17 (FBP17) expression indicates poor differentiation and invasiveness of ductal carcinomas. Sci Rep 2020; 10:11543. [PMID: 32665637 PMCID: PMC7360568 DOI: 10.1038/s41598-020-68454-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 05/29/2020] [Indexed: 12/20/2022] Open
Abstract
Formin binding protein 17 (FBP17) belongs to Cdc-42 interacting protein 4 subfamily of F-BAR proteins. Recently, we had reported that FBP17 was overexpressed in invasive breast cancer cells and interacts with the actin regulatory proteins. We also reported that FBP17 promotes invadopodia formation and enhances extracellular matrix degradation. The current study determines FBP17 expression in invasive ductal carcinomas (IDCs) using breast cancer tissue microarrays (TMAs) (82 IDCs with variable receptor status and 8 Normal adjacent tissues) and its correlation with the clinico-pathological features. Immunohistochemistry of human breast cancer TMAs showed the significant elevation in the levels of FBP17 in breast cancer tissues than the normal (p ≤ 0.0001). Interestingly, FBP17 had a higher expression in invasive molecular subtypes HER2 and TNBC (p ≤ 0.05). Similarly, tumors with lymph node positive status showed elevated FBP17 expression in HER2 and TNBC subtypes (p ≤ 0.05). Surprisingly, grade 3 tumors demonstrated higher FBP17 expression (p ≤ 0.01) indicating its role in poorly differentiated tumors. Together, the data demonstrates the overexpression of FBP17 in invasive and poorly differentiated tumors. Understanding the role of FBP17 in poor differentiation and invasion of tumors in molecular subtypes at various level might represent as a potential molecular target against the disease.
Collapse
Affiliation(s)
- Prabhat Suman
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151001, India
| | - Sarthak Mishra
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151001, India.,Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, 48149, Münster, Germany
| | - Harish Chander
- Laboratory of Molecular Medicine, Department of Human Genetics and Molecular Medicine, Central University of Punjab, Bathinda, 151001, India. .,Immuno and Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida, 201309, India.
| |
Collapse
|
46
|
ROS Mediate xCT-Dependent Cell Death in Human Breast Cancer Cells under Glucose Deprivation. Cells 2020; 9:cells9071598. [PMID: 32630312 PMCID: PMC7407809 DOI: 10.3390/cells9071598] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/25/2020] [Accepted: 06/30/2020] [Indexed: 12/19/2022] Open
Abstract
xCT, also known as solute carrier family 7 member 11 (SLC7A11), the light chain of the cystine/glutamate antiporter, is positively correlated with cancer progression due to antioxidant function. During glucose deprivation, the overexpression of xCT does not protect cancer cells but instead promotes cell death. Further understanding the mechanism of glucose deprivation-induced cell death is important for developing anticancer treatments targeting the glucose metabolism. In this study, we found that breast cancer cells with a high expression of xCT demonstrated increased levels of reactive oxygen species (ROS) and were more sensitive to glucose deprivation than the cells with a low expression of xCT. However, AMP-activated protein kinase (AMPK) did not significantly affect glucose-deprivation-induced cell death. The antioxidant N-acetyl-cysteine prevented glucose-deprivation-induced cell death, and the glutathione biosynthesis inhibitor L-buthionine-S, R-sulfoximine enhanced glucose-deprivation-induced cell death. The inhibition of xCT by sulfasalazine or a knockdown of xCT reduced the glucose-deprivation-increased ROS levels and glucose-deprivation-induced cell death. Glucose deprivation reduced the intracellular glutamate, and supplementation with α-ketoglutarate prevented the glucose-deprivation-increased ROS levels and rescued cell death. The knockdown of sirtuin-3 (SIRT3) further enhanced the ROS levels, and promoted xCT-related cell death after glucose deprivation. In conclusion, our results suggested that ROS play a critical role in xCT-dependent cell death in breast cancer cells under glucose deprivation.
Collapse
|
47
|
Ding X, Li Y, Li J, Yin Y. OSW-1 inhibits tumor growth and metastasis by NFATc2 on triple-negative breast cancer. Cancer Med 2020; 9:5558-5569. [PMID: 32515123 PMCID: PMC7402832 DOI: 10.1002/cam4.3196] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 04/21/2020] [Accepted: 05/11/2020] [Indexed: 01/01/2023] Open
Abstract
OSW‐1 is a natural compound extracted from the bulbs of Ornithogalum saundersiae in 1992. It has been shown strong antitumor activities in various cancer cells. However, the effects of OSW‐1 on tumor growth and metastasis in breast cancer are still poorly understood. In our research, we showed that OSW‐1 had a strong anticancer effect on breast cancer cells, but lower toxicity to normal cells. Accordingly, it also revealed significant inhibition of tumor growth by OSW‐1 in xenograft model. In addition, we performed Annexin V/PI‐labeled flow cytometric assay and TUNEL assay and showed that OSW‐1 inhibited tumor growth by inducing apoptosis. Furthermore, we carried out transwell assays and found that OSW‐1 significantly repressed the migratory and invasive capabilities of triple‐negative breast cancer (TNBC) cells via mediating epithelial‐mesenchymal transition. Besides, OSW‐1 also could inhibit metastasis in an orthotopic model and resulted in a longer survival compared with control group. Finally, we performed RNA‐sequencing and cellular functions to investigate the molecular mechanism of how OSW‐1 inhibits TNBC, and identified NFATc2 may as a pivotal factor for OSW‐1‐mediated effects on cell death, tumor growth, invasion, and migration.
Collapse
Affiliation(s)
- Xiaorong Ding
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.,Department of Oncology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Yumei Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jun Li
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yongmei Yin
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
48
|
Bhattacharyya GS, Doval DC, Desai CJ, Chaturvedi H, Sharma S, Somashekhar S. Overview of Breast Cancer and Implications of Overtreatment of Early-Stage Breast Cancer: An Indian Perspective. JCO Glob Oncol 2020; 6:789-798. [PMID: 32511068 PMCID: PMC7328098 DOI: 10.1200/go.20.00033] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2020] [Indexed: 12/15/2022] Open
Abstract
The prevalence and mortality of breast cancer is increasing in Asian countries, including India. With advances in medical technology leading to better detection and characterization of the disease, it has been possible to classify breast cancer into various subtypes using markers, which helps predict the risk of distant recurrence, response to therapy, and prognosis using a combination of molecular and clinical parameters. Breast cancer and its therapy, mainly surgery, systemic therapy (anticancer chemotherapy, hormonal therapy, targeted therapy, and immunotherapy), and radiation therapy, are associated with significant adverse influences on physical and mental health, quality of life, and the economic status of the patient and her family. The fear of recurrence and its devastating effects often leads to overtreatment, with a toxic cost to the patient financially and physically in cases in which this is not required. This article discusses some aspects of a breast cancer diagnosis and its impact on the various facets of the life of the patient and her family. It further elucidates the role of prognostic factors, the currently available biomarkers and prognostic signatures, and the importance of ethnically validating biomarkers and prognostic signatures.
Collapse
Affiliation(s)
| | - Dinesh C. Doval
- Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India
| | - Chirag J. Desai
- Vedanta Institute of Medical Sciences, Ahmedabad, Gujarat, India
| | | | - Sanjay Sharma
- Asian Cancer Institute, Somaiya Ayurvihar, Mumbai, Maharashtra, India
| | - S.P. Somashekhar
- Department of Surgical Oncology, Manipal Comprehensive Cancer Center, Manipal Hospital, Bengaluru, India
| |
Collapse
|
49
|
Zuo Y, Xu H, Chen Z, Xiong F, Zhang B, Chen K, Jiang H, Luo C, Zhang H. 17‑AAG synergizes with Belinostat to exhibit a negative effect on the proliferation and invasion of MDA‑MB‑231 breast cancer cells. Oncol Rep 2020; 43:1928-1944. [PMID: 32236631 PMCID: PMC7160548 DOI: 10.3892/or.2020.7563] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 01/10/2020] [Indexed: 12/15/2022] Open
Abstract
Breast cancer is one of the most common malignancies that threaten the health of women. Although there are a few chemotherapies for the clinical treatment of breast cancer, these therapies are faced with the problems of drug-resistance and metastasis. Drug combination can help to reduce the adverse side effects of chemotherapies using single drugs, and also help to overcome common drug-resistance during clinical treatment of breast cancer. The present study reported the synergistic effect of the heat shock protein 90 inhibitor 17-AAG and the histone deacetylase 6 inhibitor Belinostat in triple-negative breast cancer (TNBC) MDA-MB-231 cells, by detection of proliferation, apoptosis and cell cycle arrest following treatment with this combination. Subsequently, RNA sequencing (RNA-seq) data was collected and analyzed to investigate the synergistic mechanism of this combination. Based on the Kyoto Encyclopedia of Genes and Genomes (KEGG) signaling pathways revealed by RNA-seq data analysis, a wound-healing assay was used to investigate the effect of this combination on the migration of MDA-MB-231 cells. Compared with treatment with 17-AAG or Belinostat alone, both the viability inhibition and apoptosis rate of MDA-MB-231 cells were significantly enhanced in the combination group. The combination index values were <1 in three concentration groups. Revealed by the RNA-seq data analysis, the most significantly enriched KEGG pathways in the combination group were closely associated with cell migration. Based on these findings, the anti-migration effect of this combination was investigated. It was revealed that the migration of MDA-MB-231 cells was significantly suppressed in the combination group compared with in the groups treated with 17-AAG or Belinostat alone. In terms of specific genes, the mRNA expression levels of TEA domain family proteins were significantly decreased in the combination group, whereas the phosphorylation of YY1 associated protein 1 and modulator of VRAC current 1 was significantly enhanced in the combination group. These alterations may help to explain the anti-migration effect of this combination. Belinostat has already been approved as a treatment for T-cell lymphoma and 17-AAG is undergoing clinical trials. These findings could provide a beneficial reference for the clinical treatment of patients with TNBC.
Collapse
Affiliation(s)
- Yu Zuo
- Department of Pharmacy, School of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Heng Xu
- Department of Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Zhifeng Chen
- Department of Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Fengmin Xiong
- Department of Pharmacy, School of Pharmacy, Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Bei Zhang
- Department of Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Kaixian Chen
- Department of Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Hualiang Jiang
- Department of Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Cheng Luo
- Department of Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| | - Hao Zhang
- Department of Drug Discovery and Design Center, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, P.R. China
| |
Collapse
|
50
|
Identification of a stemness-related gene panel associated with BET inhibition in triple negative breast cancer. Cell Oncol (Dordr) 2020; 43:431-444. [PMID: 32166583 PMCID: PMC7214516 DOI: 10.1007/s13402-020-00497-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2020] [Indexed: 12/11/2022] Open
Abstract
Purpose Triple negative breast cancers (TNBCs) are enriched in cells bearing stem-like features, i.e., cancer stem cells (CSCs), which underlie cancer progression. Thus, targeting stemness may be an interesting treatment approach. The epigenetic machinery is crucial for maintaining the stemness phenotype. Bromodomain and extra-terminal domain (BET) epigenetic reader family members are emerging as novel targets for cancer therapy, and have already shown preclinical effects in breast cancer. Here, we aimed to evaluate the effect of the BET inhibitor JQ1 on stemness in TNBC. Methods Transcriptomic, functional annotation and qRT-PCR studies were performed on JQ1-exposed TNBC cells in culture. The results obtained were confirmed in spheroids and spheroid-derived tumours. In addition, limiting dilution, secondary and tertiary tumour sphere formation, matrigel invasion, immunofluorescence and flow cytometry assays were performed to evaluate the effect of JQ1 on CSC features. For clinical outcome analyses, the online tool Kaplan-Meier Plotter and an integrated response database were used. Results We found that JQ1 modified the expression of stemness-related genes in two TNBC-derived cell lines, MDA-MB-231 and BT549. Among these changes, the CD44 Antigen/CD24 Antigen (CD44/CD24) ratio and Aldehyde Dehydrogenase 1 Family Member A1 (ALDH1A1) expression level, i.e., both classical stemness markers, were found to be decreased by JQ1. Using a validated spheroid model to mimic the intrinsic characteristics of CSCs, we found that JQ1 decreased surface CD44 expression, inhibited self-renewal and invasion, and induced cell cycle arrest in G0/G1, thereby altering the stemness phenotype. We also found associations between four of the identified stemness genes, Gap Junction Protein Alpha 1 (GJA1), CD24, Epithelial Adhesion Molecule (EPCAM) and SRY-related HMG-box gene 9 (SOX9), and a worse TNBC patient outcome. The expression of another two of the stemness-related genes was found to be decreased by JQ1, i.e., ATP Binding Cassette Subfamily G Member 2 (ABCG2) and RUNX2, and predicted a low response to chemotherapy in TNBC patients, which supports a role for RUNX2 as a potential predictive marker for chemotherapy response in TNBC. Conclusions We identified a stemness-related gene panel associated with JQ1 and describe how this inhibitor modifies the stemness landscape in TNBC. Therefore, we propose a novel role for JQ1 as a stemness-targeting drug. Loss of the stem cell phenotype via JQ1 treatment could lead to less aggressive and more chemo-sensitive tumours, reflecting a better patient prognosis. Thus, the identified gene panel may be of interest for the clinical management of patients with aggressive TNBC. Electronic supplementary material The online version of this article (10.1007/s13402-020-00497-6) contains supplementary material, which is available to authorized users.
Collapse
|