1
|
Chatatikun M, Pattaranggoon NC, Sama-Ae I, Ranteh O, Poolpirom M, Pantanakong O, Chumworadet P, Kawakami F, Imai M, Tedasen A. Mechanistic exploration of bioactive constituents in Gnetum gnemon for GPCR-related cancer treatment through network pharmacology and molecular docking. Sci Rep 2024; 14:25738. [PMID: 39468096 PMCID: PMC11519448 DOI: 10.1038/s41598-024-75240-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 10/03/2024] [Indexed: 10/30/2024] Open
Abstract
G Protein-Coupled Receptors (GPCRs) are integral membrane proteins that have gained considerable attention as drug targets, particularly in cancer treatment. In this study, we explored the capacity of bioactive compounds derived from Gnetum gnemon (GG) for the development of of pharmaceuticals targeting GPCRs within the context of cancer therapy. Integrated approach combined network pharmacology and molecular docking to identify and validate the underlying pharmacological mechanisms. We retrieved targets for GG-derived compounds and GPCRs-related cancer from databases. Subsequently, we established a protein-protein interaction (PPI) network by mapping the shared targets. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses were employed to predict the mechanism of action of these targets. Molecular docking was conducted to validate our findings. We identified a total of 265 targets associated with GG-derived bioactive compounds for the treatment of GPCRs-related cancer. Functional enrichment analysis revealed the promising therapeutic effects of these targets on GPCRs-related cancer pathways. The PPI network analysis identified hub targets, including MAPK3, SRC, EGFR, STAT3, ESR1, MTOR, CCND1, and PPARG, which demonstrate as treatment targets for GPCRs-related cancer using GG-derived compounds. Additionally, molecular docking experiments demonstrated the strong binding affinity of gnetin A, gnetin C, (-)-viniferin, and resveratrol dimer, thus inhibiting MAPK3, SRC, EGFR, and MTOR. Survival analysis established the clinical prognostic relevance of identified hub genes in cancer. This study presents a novel approach for comprehending the therapeutic mechanisms of GG-derived active compounds and thereby paving the way for their prospective clinical applications in the field of cancer treatment.
Collapse
Affiliation(s)
- Moragot Chatatikun
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
- Research Excellence Center for Innovation and Health Products, Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Nawanwat C Pattaranggoon
- Faculty of Medical Technology, Rangsit University, Muang Pathumthani, Pathumthani, 12000, Thailand
- Program in Bioinformatics and Computational Biology, Graduate School, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Imran Sama-Ae
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
- Center of Excellence Research for Melioidosis and Microorganisms (CERMM), Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Onggan Ranteh
- Department of Community Public Health, School of Public Health, Walailak University, Nakhon Si Thammarat, 80161, Thailand
- Excellent Center for Dengue and Community Public Health (EC for DACH), Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Manlika Poolpirom
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Oranan Pantanakong
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Pitchaporn Chumworadet
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand
| | - Fumitaka Kawakami
- Research Facility of Regenerative Medicine and Cell Design, School of Allied Health Sciences, Kitasato University, Sagamihara, 252-0373, Japan
- Department of Regulatory Biochemistry, Kitasato University Graduate School of Medical Sciences, Sagamihara, 252-0373, Japan
| | - Motoki Imai
- Research Facility of Regenerative Medicine and Cell Design, School of Allied Health Sciences, Kitasato University, Sagamihara, 252-0373, Japan
- Department of Molecular Diagnostics, School of Allied Health Sciences, Kitasato University, Sagamihara, 252-0373, Japan
| | - Aman Tedasen
- School of Allied Health Sciences, Walailak University, Nakhon Si Thammarat, 80161, Thailand.
- Research Excellence Center for Innovation and Health Products, Walailak University, Nakhon Si Thammarat, 80161, Thailand.
| |
Collapse
|
2
|
Chen X, Zhao W, Huang Y, Luo S, Tang X, Yi Q. Association of GATA3 expression in triple-positive breast cancer with overall survival and immune cell infiltration. Sci Rep 2024; 14:17795. [PMID: 39090342 PMCID: PMC11294334 DOI: 10.1038/s41598-024-68788-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024] Open
Abstract
Breast cancer remains a leading cause of cancer-related mortality among women, with triple-positive breast cancer (TPBC) being a particularly aggressive subtype. GATA binding protein 3 (GATA3) plays a crucial role in the luminal differentiation of breast epithelium and T-cell differentiation. However, the relationship between GATA3 and immune infiltration in TPBC remains unclear. This study collected and analyzed TPBC data from The Cancer Genome Atlas (TCGA), METABRIC, and GSE123845 databases. Univariate and multivariate Cox regression analyses, along with Kaplan-Meier survival analyses, were employed to assess the prognostic value of GATA3 and other clinical features. Subsequently, Gene Set Enrichment Analysis (GSEA) was conducted to explore the potential biological functions and regulatory mechanisms of GATA3 in TPBC. Additionally, ssGSEA analysis revealed the connection between GATA3 and immune infiltration. And the effects of neoadjuvant chemotherapy and immunotherapy on GATA3 expression were also explored. Finally, clinical samples were used to detect the relationship between GATA3 expression and tumor infiltrating lymphocyte (TIL) levels. Our results demonstrated that GATA3 was significantly overexpressed in TPBC tissues compared to normal tissues (P < 0.05). A positive correlation between GATA3 mRNA and protein levels was observed (R = 0.55, P < 0.05). Notably, high GATA3 expression was associated with poor overall survival (HR = 1.24, 95% confidence interval (CI) 1.25-11.76, P < 0.05). GSEA indicated significant enrichment of immune-related gene sets in low GATA3 expression groups. Furthermore, pathologic complete response (pCR) patients exhibited significantly lower GATA3 expression compared to residual disease (RD) patients. Mutation analysis revealed higher PIK3CA and TP53 mutation rates in high GATA3 expression groups. Finally, clinical validation data showed that the degree of TILs was significantly higher in the low GATA3 expression group. In conclusion, this study suggests that high GATA3 expression may be associated with poor prognosis and may reduce immune infiltration in TPBC.
Collapse
Affiliation(s)
- Xiuwen Chen
- Department of Pathology and Department of Hematology, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Weilin Zhao
- Department of Pathology and Department of Hematology, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Yugang Huang
- Department of Pathology and Department of Hematology, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Senyuan Luo
- Department of Pathology and Department of Hematology, Taihe Hospital, Hubei University of Medicine, Hubei, China
| | - Xianbin Tang
- Department of Pathology and Department of Hematology, Taihe Hospital, Hubei University of Medicine, Hubei, China.
| | - Qiong Yi
- Department of Pathology and Department of Hematology, Taihe Hospital, Hubei University of Medicine, Hubei, China.
| |
Collapse
|
3
|
Wang X, Yang F, Sun Z, Zhao G, Pu Q, Geng C, Dong K, Zhang X, Liu Z, Song H. NKAIN1, as an oncogene, promotes the proliferation and metastasis of breast cancer, affecting its prognosis. Mol Carcinog 2024; 63:1392-1405. [PMID: 38651944 DOI: 10.1002/mc.23732] [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: 01/10/2024] [Revised: 03/31/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024]
Abstract
Na, K-ATPase interaction (NKAIN) is a transmembrane protein family, which can interact with Na, K-ATPase β1 subunit. NKAIN1 plays an important role in alcohol-dependent diseases such as endometrial and prostate cancers. However, the relationship between NKAIN1 and human breast cancer has not been studied. Hence, this study aimed to explore the relationship between NKAIN1 expression and breast cancer. Data used in this study were mainly from the Cancer Genome Atlas, including differential expression analysis, Kaplan-Meier survival analysis, receiver operating characteristic curve analysis, multiple Cox regression analysis, co-expression gene analysis, and gene set enrichment analysis. Analyses were performed using reverse transcription-quantitative polymerase chain reaction, western blot analysis, and immunohistochemistry on 46 collected samples. The knockdown or overexpression of NKAIN1 in vitro in MCF-7 and MDA-MB-231 cell lines altered the proliferation and migration abilities of tumor cells. In vivo experiments further confirmed that NKAIN1 knockdown effectively inhibited the proliferation and migration of cancer cells. Therefore, our study identified NKAIN1 as an oncogene that is highly expressed in breast cancer tissues. The findings highlight the potential of NKAIN1 as a molecular biomarker of breast cancer.
Collapse
Affiliation(s)
- XiMei Wang
- Department of General Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - FangZheng Yang
- Department of General Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Zhi Sun
- Department of General Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Department of Breast Disease(II), Shandong Second Provincial General Hospital, Jinan, China
| | - GuangHui Zhao
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Qingdao Key Lab of Mitochondrial Medicine, Qingdao, China
| | - Qian Pu
- Department of General Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - ChenChen Geng
- Department of General Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - Ke Dong
- Department of General Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| | - XiaoDong Zhang
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Qingdao Key Lab of Mitochondrial Medicine, Qingdao, China
| | - ZiQian Liu
- Department of Medical Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
- Qingdao Key Lab of Mitochondrial Medicine, Qingdao, China
| | - HaiYun Song
- Department of Pathology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, China
| |
Collapse
|
4
|
Ding W, Ye D, Chen H, Lin Y, Li Z, Tu C. Clinicopathological differences and survival benefit in ER+/PR+/HER2+ vs ER+/PR-/HER2+ breast cancer subtypes. Breast Cancer 2024; 31:295-304. [PMID: 38231460 DOI: 10.1007/s12282-023-01538-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 12/13/2023] [Indexed: 01/18/2024]
Abstract
INTRODUCTION Breast cancer subtypes based on estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression have significant implications for prognosis. HER2-positive tumors historically demonstrated poorer survival, but anti-HER2 targeted therapy improved outcomes. However, hormone receptor (HR)-positive patients may experience reduced benefit due to HER2-HR signaling crosstalk. METHODS Data from two databases, the Shanghai Jiao Tong University Breast Cancer Data Base (SJTUBCDB) and the National Cancer Institute's Surveillance, Epidemiology, and End Results (SEER) database, were analyzed. Propensity score adjustments were used to balance patient characteristics between ER+/PR+/HER2+ and ER+/PR-/HER2+ subtypes. Kaplan-Meier survival curves estimated disease-free survival (DFS), breast cancer-specific survival (BCSS), overall survival (OS) for these subtypes in the SJTUBCDB, while subgroup analyses using multivariable models were performed based on menstruation, pN stage, HER2-targeted therapy, and endocrinotherapy. RESULTS The ER+/PR+/HER2+ group showed significantly better DFS and BCSS than the ER+/PR-/HER2+ group, particularly in postmenopausal and pN0 stage patients. Survival outcomes were similar after anti-HER2 therapy or endocrine aromatase inhibitor (AI) therapy in both groups. However, among patients receiving selective estrogen receptor modulator (SERM) treatment, those in the ER+/PR-/HER2+ group had a significantly worse prognosis compared to ER+/PR+/HER2+ patients. CONCLUSIONS HER2-positive breast cancers with different HR statuses exhibit distinct clinicopathological features and survival outcomes. Patients in the ER+/PR+/HER2+ group generally experience better survival, particularly in postmenopausal and pN0 stage patients. Treatment strategies should consider HR status and specific modalities for better personalized management.
Collapse
Affiliation(s)
- Wu Ding
- Department of Oncological Surgery, Shaoxing Second Hospital, Shaoxing, 312000, China
- Department of Clinical Medicine, Shaoxing University School of Medicine, Shaoxing, China
| | - Dengfeng Ye
- Department of Oncological Surgery, Shaoxing Second Hospital, Shaoxing, 312000, China
| | - Haifeng Chen
- Department of Oncological Surgery, Shaoxing Second Hospital, Shaoxing, 312000, China
| | - Yingli Lin
- Department of Early Childhood Education, Shaoxing Vocational and Technical College, Shaoxing, China
| | - Zhian Li
- Department of Oncological Surgery, Shaoxing Second Hospital, Shaoxing, 312000, China.
| | - Chuanjian Tu
- Department of Surgery, Shaoxing Second Hospital, Shaoxing, 312000, China.
| |
Collapse
|
5
|
Si G, Hapuarachchige S, Lesniak W, Artemov D. PET-MR Guided, Pre-targeted delivery to HER2(+) Breast Cancer Model. RESEARCH SQUARE 2024:rs.3.rs-3974001. [PMID: 38464126 PMCID: PMC10925432 DOI: 10.21203/rs.3.rs-3974001/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Purpose: HER2(+) metastatic breast cancer (mBC) is one of the most aggressive and lethal cancer types among females. While initially effective, targeted therapeutic approaches with trastuzumab and pertuzumab antibodies and antibody-drug conjugates (ADC) lack long-term efficacy against HER2(+) mBC and can cause severe systemic toxicity due to off-target effects. Therefore, the development of novel targeted delivery platforms that minimize toxicity and increase therapeutic efficacy is critical to the treatment of HER2(+) breast cancer (BC). A pretargeting delivery platform can minimize the non-specific accumulation and off-target toxicity caused by traditional one-step delivery method by separating the single delivery step into a pre-targeting step with high-affinity biomarker binding ligand followed by the subsequent delivery step of therapeutic component with fast clearance. Each delivery component is functionalized with bioorthogonal reactive groups that quickly react in situ , forming cross-linked clusters on the cell surface, which facilitates rapid internalization and intracellular delivery of therapeutics. Procedures: We have successfully developed a click chemistry-based pretargeting platform for HER2(+) BC enabling PET-MR image guidance for reduced radiation dose, high sensitivity, and good soft tissue contrast. Radiolabeled trastuzumab and superparamagnetic iron-oxide carriers (uSPIO) were selected as pretargeting and delivery components, respectively. HER2(+) BT-474 cell line and corresponding xenografts were used for in vitro and in vivo studies. Results: An enhanced tumor accumulation as well as tumor- to-organ accumulation ratio was observed in pretargeted mice up to 24 h post uSPIO injection. A 40% local T 1 decrease in the pretargeted mice tumor was observed within 4 h, and an overall 15% T 1 drop was retained for 24 h post uSPIO injection. Conclusions: Prolonged tumor retention and increased tumor-to-organ accumulation ratio provided a solid foundation for pretargeted image-guided delivery approach for in vivo applications.
Collapse
|
6
|
Cao LQ, Sun H, Xie Y, Patel H, Bo L, Lin H, Chen ZS. Therapeutic evolution in HR+/HER2- breast cancer: from targeted therapy to endocrine therapy. Front Pharmacol 2024; 15:1340764. [PMID: 38327984 PMCID: PMC10847323 DOI: 10.3389/fphar.2024.1340764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 01/08/2024] [Indexed: 02/09/2024] Open
Abstract
Breast cancer, a complex and varied disease, has four distinct subtypes based on estrogen receptor and human epidermal growth factor receptor 2 (HER2) levels, among which a significant subtype known as HR+/HER2-breast cancer that has spurred numerous research. The prevalence of breast cancer and breast cancer-related death are the most serious threats to women's health worldwide. Current progress in treatment strategies for HR+/HER2-breast cancer encompasses targeted therapy, endocrine therapy, genomic immunotherapy, and supplementing traditional methods like surgical resection and radiotherapy. This review article summarizes the current epidemiology of HR+/HER2-breast cancer, introduces the classification of HR+/HER2-breast cancer and the commonly used treatment methods. The mechanisms of action of various drugs, including targeted therapy drugs and endocrine hormone therapy drugs, and their potential synergistic effects are deeply discussed. In addition, clinical trials of these drugs that have been completed or are still in progress are included.
Collapse
Affiliation(s)
- Lu-Qi Cao
- Institute for Biotechnology, St. John’s University, Queens, NY, United States
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Haidong Sun
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Yuhao Xie
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Harsh Patel
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| | - Hanli Lin
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Zhe-Sheng Chen
- Institute for Biotechnology, St. John’s University, Queens, NY, United States
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, NY, United States
| |
Collapse
|
7
|
Wang J, Yu Y, Lin Q, Zhang J, Song C. Efficacy and safety of first-line therapy in patients with HER2-positive advanced breast cancer: a network meta-analysis of randomized controlled trials. J Cancer Res Clin Oncol 2024; 150:21. [PMID: 38244085 PMCID: PMC10799814 DOI: 10.1007/s00432-023-05530-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 11/03/2023] [Indexed: 01/22/2024]
Abstract
PURPOSE The numerous first-line treatment regimens for human epidermal growth factor receptor 2 (HER2)-positive advanced breast cancer (ABC) necessitate a comprehensive evaluation to inform clinical decision-making. We conducted a Bayesian network meta-analysis (NMA) to compare the efficacy and safety of different interventions. METHODS We systematically searched for relevant randomized controlled trials (RCTs) in Pubmed, Embase, Cochrane Library and online abstracts from inception to June 1, 2023. NMA was performed to calculate and analyze progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and adverse events of grade 3 or higher (≥ 3 AEs). RESULTS Out of the 10,313 manuscripts retrieved, we included 28 RCTs involving 11,680 patients. Regarding PFS and ORR, the combination of trastuzumab with tyrosine kinase inhibitors (TKIs) was more favorable than dual-targeted therapy. If only using trastuzumab, combination chemotherapy is superior to monochemotherapy in terms of PFS. It is important to note that the addition of anthracycline did not result in improved PFS. For patients with hormone receptor-positive HER2-positive diseases, dual-targeted combined with endocrine therapy showed better benefit in terms of PFS compared to dual-targeted alone, but it did not reach statistical significance. The comprehensive analysis of PFS and ≥ 3 AEs indicates that monochemotherapy combined with dual-targeted therapy still has the optimal balance between efficacy and safety. CONCLUSION Monochemotherapy (Docetaxel) plus dual-target (Trastuzumab and Pertuzumab) therapy remains the optimal choice among all first-line treatment options for ABC. The combination of trastuzumab with TKIs (Pyrotinib) demonstrated a significant improvement in PFS and ORR, but further data are warranted to confirm the survival benefit.
Collapse
Affiliation(s)
- Junxiao Wang
- Department of Breast Surgery, College of Clinical Medicine for Oncology, Fujian Medical University, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian Province, China
- Department of Thyroid and Breast Surgery, The Second Hospital of Sanming, Sanming City, Fujian Province, China
| | - Yushuai Yu
- Department of Breast Surgery, College of Clinical Medicine for Oncology, Fujian Medical University, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian Province, China
- Breast Surgery Institute, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, Fujian Province, China
| | - Qisheng Lin
- Department of Thyroid and Breast Surgery, The Second Hospital of Sanming, Sanming City, Fujian Province, China
| | - Jie Zhang
- Department of Breast Surgery, Fujian Medical University Union Hospital, No.29, Xin Quan Road, Gulou District, Fuzhou, 350001, Fujian Province, China.
- Breast Surgery Institute, Fujian Medical University, Fuzhou, Fujian Province, China.
| | - Chuangui Song
- Department of Breast Surgery, College of Clinical Medicine for Oncology, Fujian Medical University, No.91, Fuma Road, Jin'an District, Fuzhou, Fujian Province, China.
- Breast Surgery Institute, College of Clinical Medicine for Oncology, Fujian Medical University, Fuzhou, Fujian Province, China.
| |
Collapse
|
8
|
Dhanushkumar T, M E S, Selvam PK, Rambabu M, Dasegowda KR, Vasudevan K, George Priya Doss C. Advancements and hurdles in the development of a vaccine for triple-negative breast cancer: A comprehensive review of multi-omics and immunomics strategies. Life Sci 2024; 337:122360. [PMID: 38135117 DOI: 10.1016/j.lfs.2023.122360] [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: 10/12/2023] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023]
Abstract
Triple-Negative Breast Cancer (TNBC) presents a significant challenge in oncology due to its aggressive behavior and limited therapeutic options. This review explores the potential of immunotherapy, particularly vaccine-based approaches, in addressing TNBC. It delves into the role of immunoinformatics in creating effective vaccines against TNBC. The review first underscores the distinct attributes of TNBC and the importance of tumor antigens in vaccine development. It then elaborates on antigen detection techniques such as exome sequencing, HLA typing, and RNA sequencing, which are instrumental in identifying TNBC-specific antigens and selecting vaccine candidates. The discussion then shifts to the in-silico vaccine development process, encompassing antigen selection, epitope prediction, and rational vaccine design. This process merges computational simulations with immunological insights. The role of Artificial Intelligence (AI) in expediting the prediction of antigens and epitopes is also emphasized. The review concludes by encapsulating how Immunoinformatics can augment the design of TNBC vaccines, integrating tumor antigens, advanced detection methods, in-silico strategies, and AI-driven insights to advance TNBC immunotherapy. This could potentially pave the way for more targeted and efficacious treatments.
Collapse
Affiliation(s)
- T Dhanushkumar
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - Santhosh M E
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - Prasanna Kumar Selvam
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - Majji Rambabu
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - K R Dasegowda
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India
| | - Karthick Vasudevan
- Department of Biotechnology, School of Applied Sciences, REVA University, Bengaluru 560064, India.
| | - C George Priya Doss
- Laboratory of Integrative Genomics, Department of Integrative Biology, School of BioSciences and Technology, Vellore Institute of Technology (VIT), Vellore, India.
| |
Collapse
|
9
|
Gamrani S, Akhouayri L, Boukansa S, Karkouri M, El Fatemi H. The Clinicopathological Features and Prognostic Significance of HER2-Low in Early Breast Tumors Patients Prognostic Comparison of HER-Low and HER2-Negative Breast Cancer Stratified by Hormone Receptor Status. Breast J 2023; 2023:6621409. [PMID: 38075551 PMCID: PMC10703525 DOI: 10.1155/2023/6621409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 12/18/2023]
Abstract
Introduction There has been increased interest in HER2-low breast tumors recently, as these tumors may have distinct clinical and molecular characteristics compared to HER2-negative and HER2-positive tumors. A new nomenclature has been proposed for HER2 1+ and HER2 2+ tumors that are confirmed negative according to fluorescence in situ hybridization (FISH). These tumors are now referred to as HER2-low, and it is thought that they may represent a distinct subtype of breast cancer that warrants further investigation. In this study, we aimed to evaluate the clinicopathological characteristics and prognostic impact of this particular subtype in a North-African context where HER2-low breast cancer is a relatively understudied subtype, particularly in non-Western populations. Methods We conducted a retrospective cohort study on 1955 breast tumors in Moroccan patients over 10 years, collected at the Pathology Department of Ibn Rochd University Hospital in Casablanca and at the pathology department of Hassan II University Hospital in Fes. We elaborated on their complete immunohistochemical profile based on the main breast cancer biomarkers: Ki-67, HER2, estrogen, and progesterone receptors. Their overall survival and disease free survival data were also retrieved from their respective records. Results Out of 1955 BC patients, 49.3% were classified as HER2-low; of which 80.7% and 19.2% were hormone receptors positive and negative, respectively. The clinicopathologic features indicate that HER2-low subtype tumors behave much more like HER2-positive than HER2-negative tumors. The survival analysis showed that the HER2-low subtype-belonging patients present significantly the poorest prognosis in disease-free survival (p = 0.003) in comparison with HER2-negative ones. When considering the hormonal status, hormonal-dependent tumors show a significant difference according to HER2 subtypes in disease-free survival (p < 0.001). Yet no significant difference was shown among hormonal negative tumors. Moreover, patients with hormonal positive tumors and simultaneously belonging to the HER2-low subgroup present a significantly good prognosis in overall survival compared to the ones with hormonal negative tumors (p = 0.008). Conclusion Our study has shown that the HER2-low phenotype is common among hormone-positive patients. The clinicopathological features and prognostic data indicate that the hormonal receptors effect and HER2 heterogeneity are crucial factors to consider. It is important to note that this particular subgroup is different from the HER2-negative one and should not be treated in the same way. Therefore, this study offers a new perspective in the management of HER2-low patients and can serve as a basis for future prospective analyses.
Collapse
Affiliation(s)
- Sanaa Gamrani
- Medical Center of Biomedical and Translational Research, Hassan II University Hospital, Faculty of Medicine and Pharmacy, University Sidi Mohamed Ben Abdellah, Fez, Morocco
- Laboratory of Anatomic Pathology and Molecular Pathology, University Hospital Hassan II, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Laila Akhouayri
- Dipartimento di Scienze Cliniche e Biologiche, Università Degli Studi Di Torino, Via Giuseppe Verdi, Torino, Italy
- Department of Biomedical Sciences, Genetics and Molecular Biology Laboratory, Faculty of Medicine and Pharmacy, Hassan II-Casablanca University, Rue Tariq Ibn Ziad, Casablanca, Morocco
| | - Sara Boukansa
- Medical Center of Biomedical and Translational Research, Hassan II University Hospital, Faculty of Medicine and Pharmacy, University Sidi Mohamed Ben Abdellah, Fez, Morocco
- Laboratory of Anatomic Pathology and Molecular Pathology, University Hospital Hassan II, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| | - Mehdi Karkouri
- Department of Biomedical Sciences, Genetics and Molecular Biology Laboratory, Faculty of Medicine and Pharmacy, Hassan II-Casablanca University, Rue Tariq Ibn Ziad, Casablanca, Morocco
- Department of Pathology, Ibn Rochd University Hospital, Rue des Hôpitaux, Casablanca, Morocco
| | - Hinde El Fatemi
- Medical Center of Biomedical and Translational Research, Hassan II University Hospital, Faculty of Medicine and Pharmacy, University Sidi Mohamed Ben Abdellah, Fez, Morocco
- Laboratory of Anatomic Pathology and Molecular Pathology, University Hospital Hassan II, Sidi Mohamed Ben Abdellah University, Fez, Morocco
| |
Collapse
|
10
|
Chekhun V, Martynyuk О, Lukianova Y, Mushii O, Zadvornyi T, Lukianova N. FEATURES OF BREAST CANCER IN PATIENTS OF YOUNG AGE: SEARCH FOR DIAGNOSIS OPTIMIZATION AND PERSONALIZED TREATMENT. Exp Oncol 2023; 45:139-150. [PMID: 37824778 DOI: 10.15407/exp-oncology.2023.02.139] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Indexed: 10/14/2023]
Abstract
The statistical data of the recent decades demonstrate a rapid growth of breast cancer (BCa) incidence and a tendency toward its increase especially in young women. In the structure of morbidity of women in the age group of 18-29 years, BCa ranks first and in the age range of 15-39 years, BCa is one of the leading causes of mortality. According to the data of the epidemiological and clinical studies, the young age is an independent unfavorable prognostic factor of BCa that is associated with an unfavorable prognosis and low survival rates and is considered an important predictor of the disease aggressiveness, a high risk of metastasis and recurrence. The variability of clinicopathological and molecular-biological features of BCa in patients of different age groups as well as the varying course of the disease and different responses to the therapy are mediated by many factors. The analysis of the literature data on the factors and mechanisms of BCa initiation in patients of different age groups demonstrates that the pathogen- esis of BCa depends not only on the molecular-genetic alterations but also on the metabolic disorders caused by the current social and household rhythm of life and nutrition peculiarities. All these factors affect both the general con- dition of the body and the formation of an aggressive microenvironment of the tumor lesion. The identified features of transcriptome and the differential gene expression give evidence of different regulations of the immune response and the metabolic processes in BCa patients of different age groups. Association between the high expression of the components of the stromal microenvironment and the inflammatory immune infiltrate as well as the increased vascu- larization of the tumor lesion has been found in BCa tissue of young patients. Proving the nature of the formation of the landscape comprising molecular-genetic, cytokine, and immune factors of the tumor microenvironment will undoubtedly contribute to our understanding of the mechanisms of tumor growth allowing for the development of algorithms for delineating the groups at high risk of tumor progression, which requires more careful monitoring and personalized treatment approach. Th s will be helpful in the development of innovative technologies for complex BCa treatment.
Collapse
Affiliation(s)
- V Chekhun
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine.
| | - О Martynyuk
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | - Ye Lukianova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | - O Mushii
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | - T Zadvornyi
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| | - N Lukianova
- R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology, NAS of Ukraine, 03022 Kyiv, Ukraine
| |
Collapse
|
11
|
Wu XM, Qian YK, Chen HL, Hu CH, Chen BW. Efficacy and Safety of Anti-HER2 Targeted Therapy for Metastatic HR-Positive and HER2-Positive Breast Cancer: A Bayesian Network Meta-Analysis. Curr Oncol 2023; 30:8444-8463. [PMID: 37754530 PMCID: PMC10528081 DOI: 10.3390/curroncol30090615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 09/09/2023] [Accepted: 09/12/2023] [Indexed: 09/28/2023] Open
Abstract
Despite the development of HER2-targeted drugs, achieving favorable outcomes for patients with HR+/HER2+MBC remains challenging. This study utilized Bayesian Network Meta-analysis to compare the efficacy and safety of anti-HER2 combination regimens. The primary analysis focused on progression-free survival (PFS), while secondary analyses included objective response rate, overall survival (OS) and the incidence rate of grade 3/4 adverse events (AEs). A comprehensive search across seven databases identified 25 randomized controlled trials for inclusion in this meta-analysis. For patients eligible for endocrinotherapy, our findings revealed that dual-target combined endocrine therapy, such as Her2-mAb+Her2-mAb+Endo (HR = 0.38; 95%CrI: 0.16-0.88) and Her2-mAb+Her2-tki+Endo (HR = 0.45; 95%CrI: 0.23-0.89), significantly improved PFS compared to endocrine therapy alone. According to the surface under the cumulative ranking curves (SUCRAs), Her2-mAb+Her2-mAb+Endo and Her2-mAb+Her2-tki+Endo ranked highest in terms of PFS and OS, respectively. For patients unsuitable for endocrine therapy, anti-HER2 dual-target combined chemotherapy, such as Her2-mAb+Her2-mAb+Chem (HR = 0.76; 95%CrI: 0.6-0.96) and Her2-mAb+Her2-tki+Chem (HR = 0.48; 95%CrI: 0.29-0.81), demonstrated significant improvements in PFS compared to Her2-mAb+Chem. The results were the same when compared with Her2-tki+Chem. According to the SUCRAs, Her2-mAb+Her2-tki+Chem and Her2-mAb+Her2-mAb+Chem ranked highest for PFS and OS, respectively. Subgroup analyses consistently supported these overall findings, indicating that dual-target therapy was the optimal approach irrespective of treatment line.
Collapse
Affiliation(s)
| | | | | | | | - Bing-Wei Chen
- Department of Epidemiology and Health Statistics, School of Public Health, Southeast University, Nanjing 210009, China; (X.-M.W.); (Y.-K.Q.); (H.-L.C.); (C.-H.H.)
| |
Collapse
|
12
|
Zhang M, Zhang X, Ma T, Wang C, Zhao J, Gu Y, Zhang Y. Precise subtyping reveals immune heterogeneity for hormone receptor-positive breast cancer. Comput Biol Med 2023; 163:107222. [PMID: 37413851 DOI: 10.1016/j.compbiomed.2023.107222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/18/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
A significant proportion of breast cancer cases are characterized by hormone receptor positivity (HR+). Clinically, the heterogeneity of HR+ breast cancer leads to different therapeutic effects on endocrine. Therefore, definition of subgroups in HR+ breast cancer is important for effective treatment. Here, we have developed a CMBR method utilizing computational functional networks based on DNA methylation to identify conserved subgroups in HR+ breast cancer. Calculated by CMBR, HR+ breast cancer was divided into five subgroups, of which HR+/negative epidermal growth factor receptor-2 (Her2-) was divided into two subgroups, and HR+/positive epidermal growth factor receptor-2 (Her2+) was divided into three subgroups. These subgroups had heterogeneity in the immune microenvironment, tumor infiltrating lymphocyte patterns, somatic mutation patterns and drug sensitivity. Specifically, CMBR identified two subgroups with the "Hot" tumor phenotype. In addition, these conserved subgroups were broadly validated on external validation datasets. CMBR identified the molecular signature of HR+ breast cancer subgroups, providing valuable insights into personalized treatment strategies and management options.
Collapse
Affiliation(s)
- Mengyan Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Xingda Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, 150 Haping Road, Harbin, 150081, China
| | - Te Ma
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Cong Wang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Jiyun Zhao
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Yue Gu
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China
| | - Yan Zhang
- School of Life Science and Technology, Harbin Institute of Technology, Harbin, 150001, China; College of Pathology, Qiqihar Medical University, Qiqihar, 161042, China.
| |
Collapse
|
13
|
Pegram M, Pietras R, Dang CT, Murthy R, Bachelot T, Janni W, Sharma P, Hamilton E, Saura C. Evolving perspectives on the treatment of HR+/HER2+ metastatic breast cancer. Ther Adv Med Oncol 2023; 15:17588359231187201. [PMID: 37576607 PMCID: PMC10422890 DOI: 10.1177/17588359231187201] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 06/21/2023] [Indexed: 08/15/2023] Open
Abstract
Breast cancer (BC) with expression of the estrogen receptor (ER) and/or progesterone receptor (PR) protein and with overexpression/amplification of the human epidermal growth factor receptor 2 (HER2), termed hormone receptor-positive (HR+)/HER2+ BC, represents ∼10% of all BCs in the United States. HR+/HER2+ BC includes HER2+ BCs that are ER+, PR+, or both ER+ and PR+ (triple-positive BC). Although the current guideline-recommended treatment combination of anti-HER2 monoclonal antibodies plus chemotherapy is an effective first-line therapy for many patients with HER2+ advanced disease, intratumoral heterogeneity within the HR+/HER2+ subtype and differences between the HR+/HER2+ subtype and the HR-/HER2+ subtype suggest that other targeted combinations could be investigated in randomized clinical trials for patients with HR+/HER2+ BC. In addition, published data indicate that crosstalk between HRs and HER2 can lead to treatment resistance. Dual HR and HER2 pathway targeting has been shown to be a rational approach to effective and well-tolerated therapy for patients with tumors driven by HER2 and HR, as it may prevent development of resistance by blocking receptor pathway crosstalk. However, clinical trial data for such approaches are limited. Treatments to attenuate other signaling pathways involved in receptor crosstalk are also under investigation for inclusion in dual receptor targeting regimens. These include cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors, based on the rationale that association of CDK4/6 with cyclin D1 may play a role in resistance to HER2-directed therapies, and others such as phosphatidylinositol-3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway inhibitors. Herein, we will review the scientific and clinical rationale for combined receptor blockade targeting HER2 and ER for patients with advanced-stage HR+/HER2+ disease.
Collapse
Affiliation(s)
- Mark Pegram
- Stanford Comprehensive Cancer Institute, Stanford University School of Medicine, Lorry Lokey Building/SIM 1, 265 Campus Drive, Ste G2103, Stanford, CA 94305-5456, USA
| | - Richard Pietras
- Division of Hematology-Oncology, Department of Medicine, UCLA David Geffen School of Medicine, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA
| | - Chau T. Dang
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, New York, NY, USA
| | - Rashmi Murthy
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Thomas Bachelot
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, University Hospital Ulm, University of Ulm, Ulm, Germany
| | - Priyanka Sharma
- Department of Internal Medicine, Division of Medical Oncology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Erika Hamilton
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA
| | - Cristina Saura
- Vall d’Hebron University Hospital and Vall d’Hebron Institute of Oncology (VHIO), Medical Oncology Service, Barcelona, Spain
| |
Collapse
|
14
|
Darvishi A, Abdi Dezfouli R, Fazaeli A, Daroudi R, Zandieh N. Is Palbociclib a cost-effective strategy in the second-line treatment of metastatic breast cancer in Iran? Expert Rev Pharmacoecon Outcomes Res 2023; 23:1201-1210. [PMID: 37768209 DOI: 10.1080/14737167.2023.2263167] [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: 06/10/2023] [Accepted: 09/21/2023] [Indexed: 09/29/2023]
Abstract
BACKGROUND & AIMS This study evaluates the cost-effectiveness of Palbociclib in the second-line treatment of hormone receptor-positive (HR+) human epidermal growth factor receptor 2-negative (HER2-) metastatic breast cancer (MBC) in Iran. METHODS The present economic evaluation used a partitioned survival model (PSM). This model compares lifetime costs and disease outcomes among groups receiving different medication combinations containing Palbociclib, Fulvestrant, Everolimus, Ribociclib, and Abemaciclib as the second-line therapy for HR+/HER2- MBC. The model was conducted from Iran's healthcare perspective, structured with 1-month cycles, and the evaluation time horizon in the base analysis was set to 180 cycles (15 years). Transition probabilities were extracted using the survival curves. The cost information was extracted based on the year 2020. The Quality Adjusted Life Years (QALY) was considered the final outcome unit, and the cost-effectiveness of different combinations is calculated as cost per QALY. The annual discount rate of 5% was considered for costs and QALYs. Two times Iran's GDP per capita (800,000,000 IRR = US$5934) was used as the threshold. Finally, due to the uncertainty of some parameters, deterministic and probabilistic sensitivity analyses were carried out. RESULTS The base case results showed that the highest cost was for the 'Ribociclib+ Fulvestrant' combination (US$89,629.56), and the lowest price was for the 'Iranian Everolimus + Fulvestrant' combination (US$10,740.09). 'Palbociclib + Fulvestrant' brings about the highest value of 1.456 incremental QALYs compared to other strategies. Finally, the 'Iranian Palbociclib + Fulvestrant' was the cost-effective combination, with an incremental cost-effectiveness ratio (ICER) of US$4,201 compared to other strategies. The base case results were supported by the probabilistic sensitivity analysis. Deterministic sensitivity analysis showed that the cost of Iranian Palbociclib has a threshold of US$582.99. CONCLUSIONS The 'Iranian Palbociclib + Fulvestrant' combination was cost-effective in second-line therapy for HR+ HER2- MBC in Iran.
Collapse
Affiliation(s)
- Ali Darvishi
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Abdi Dezfouli
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Aliakbar Fazaeli
- Department of Health Management and Economics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Rajabali Daroudi
- Department of Health Management and Economics, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
- National Center for Health Insurance Research, Tehran, Iran
| | - Narges Zandieh
- Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
15
|
Zeng N, Han J, Liu Z, He J, Tian K, Chen N. CDK4/6 Inhibitors in the First-Line Treatment of Postmenopausal Women with HR+/HER2- Advanced or Metastatic Breast Cancer: An Updated Network Meta-Analysis and Cost-Effectiveness Analysis. Cancers (Basel) 2023; 15:3386. [PMID: 37444496 DOI: 10.3390/cancers15133386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/08/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
(1) Background: This study aimed to conduct a NMA and CEA combined study to compare the effectiveness and cost-effectiveness of different CDK4/6 inhibitors (Abem, Palbo, and Ribo) plus NSAI with placebo plus NSAI in the first-line treatment of postmenopausal women with HR+/HER2- ABC from the perspective of payers in China. (2) Methods: Studies which evaluated CDK4/6 inhibitors plus NSAI for HR+/HER2- ABC were searched. A Bayesian NMA was carried out and the main outcomes were the hazard ratios (HRs) of overall survival (OS) and progression-free survival (PFS). The costs and efficacy of first-line therapies for HR+/HER2- ABC were evaluated using the Markov model. The main outcomes in the CEA were incremental cost-utility ratios (ICURs), incremental monetary benefit (INMB), and incremental net-health benefit (INHB). The robustness of the model was assessed by one-way, three-way, and probabilistic sensitivity analyses. Then, we further simulated the impact of different prices of CDK4/6 inhibitors on the results. (3) Results: Seven studies involving 5347 patients were included in the NMA. The three first-line CDK4/6 inhibitors plus NSAI groups provided significant PFS and OS superiority to NSAI alone. Abem + NSAI represented a significant statistical advantage onPFS (HR 0.74, 95% CI 0.61-0.90, p = 0.009) and indicated a trend of being the best OS benefit compared to the placebo + NSAI group (HR 0.89, 95% CI 0.72-1.08). The Abem + NSAI, Palbo + NSAI, and Ribo + NSAI groups resulted in additional costs of $12,602, $20,391, and $81,258, with additional effects of 0.38, 0.31, and 0.30 QALYs, respectively, leading to an ICUR of $33,163/QALY, $65,777/QALY, and $270,860/QALY. Additional pairwise comparisons showed that Abem + NSAI was the only cost-effective option in three CDK4/6 inhibitors plus NSAI groups at a willingness-to-pay (WTP) of $38,029/QALY. The sensitivity analyses showed that the proportion of receiving subsequent CDK4/6 inhibitors and the cost of Abem significantly influenced the results of Abem + NSAI compared with placebo + NSAI. (4) Conclusion: From the perspective of Chinese payers, Abem + NSAI was a cost-effective treatment option compared with placebo + NSAI at the WTP of $38,029/QALY, since only the ICUR of $33,163/QALY of Abem + NSAI was lower than the WTP of $38,029/QALY in China (2022). The Palbo + NSAI and Ribo + NSAI groups were not cost-effective unless drug prices were adjusted to 50% or 10% of current prices ($320.67 per cycle or $264.60 per cycle). (5) Others: We have prospectively registered the study with the PROSPERO, and the PROSPERO registration number is CRD42023399342.
Collapse
Affiliation(s)
- Ni Zeng
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiaqi Han
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zijian Liu
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jinlan He
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kun Tian
- Department of Andrology, Sichuan Human Sperm Bank, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu 610041, China
| | - Nianyong Chen
- Department of Head and Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Radiation Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
16
|
Pegram M, Jackisch C, Johnston SRD. Estrogen/HER2 receptor crosstalk in breast cancer: combination therapies to improve outcomes for patients with hormone receptor-positive/HER2-positive breast cancer. NPJ Breast Cancer 2023; 9:45. [PMID: 37258523 DOI: 10.1038/s41523-023-00533-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/04/2023] [Indexed: 06/02/2023] Open
Abstract
The human epidermal growth factor receptor 2 (HER2) is overexpressed in 13-22% of breast cancers (BC). Approximately 60-70% of HER2+ BC co-express hormone receptors (HRs). HR/HER2 co-expression modulates response to both anti-HER2-directed and endocrine therapy due to "crosstalk" between the estrogen receptor (ER) and HER2 pathways. Combined HER2/ER blockade may be an effective treatment strategy for patients with HR+/HER2+ BC in the appropriate clinical setting(s). In this review, we provide an overview of crosstalk between the ER and HER2 pathways, summarize data from recently published and ongoing clinical trials, and discuss clinical implications for targeted treatment of HR+/HER2+ BC.
Collapse
Affiliation(s)
- Mark Pegram
- Stanford Cancer Institute, Stanford, CA, USA.
| | - Christian Jackisch
- Obstetrics and Gynaecology and Breast Cancer Center, Klinikum Offenbach GmbH, Offenbach, Germany
| | | |
Collapse
|
17
|
Xu Y, Liang Y, Yin G. Detecting the expression of HRs and BCL2 via IHC can help identify luminal A-like subtypes of triple-positive breast cancers. Clin Transl Oncol 2023; 25:1024-1032. [PMID: 36376700 DOI: 10.1007/s12094-022-03007-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022]
Abstract
BACKGROUND Triple-positive breast cancer (TPBC) is a tumor that simultaneously expresses estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2). Luminal A-like TPBC is a special subtype with a favorable prognosis but benefits less from HER2-targeted therapy. However, little is known about how to identify luminal A-like TPBCs. Therefore, our study aims to explore a clinically feasible method to identify luminal A-like TPBCs using immunohistochemical (IHC) markers. METHODS Our cohort enrolled consecutive 190 patients with early-stage TPBCs diagnosed, treated and followed up in our hospital between 2013 and 2019. Patients whose IHC staining displayed ≥ 50% in both ER and PR scores and B-cell lymphoma 2 (BCL2) positivity were classified as cohort A (n = 64), and the rest were enrolled in cohort B (n = 126). Kaplan-Meier plotter and log-rank test were used to compare the survival difference between cohort A and cohort B and the efficacy of trastuzumab therapy in the two cohorts. RESULTS The disease-free survival (DFS) of patients in cohort A was significantly better than in cohort B (p = 0.031). In cohort A, there was no statistically significant difference in DFS between patients treated with trastuzumab and those without trastuzumab (p = 0.663). While in cohort B, patients treated with trastuzumab had significantly better DFS than those without trastuzumab (p = 0.032). Multivariate survival analysis showed that cohort A was associated with better DFS(95%CI 1.046-11.776, p = 0.042). CONCLUSION TPBCs consist of heterogeneous subtypes. Detecting the expression of ER, PR and BCL2 via IHC can help identify luminal A-like TPBCs. This study will enable individualized treatment of TPBCs.
Collapse
Affiliation(s)
- Yingying Xu
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, 130041, China
| | - Yonghao Liang
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, 130041, China
| | - Guanghao Yin
- Department of Breast Surgery, Second Hospital of Jilin University, Changchun, 130041, China.
| |
Collapse
|
18
|
Sánchez-León ML, Jiménez-Cortegana C, Silva Romeiro S, Garnacho C, de la Cruz-Merino L, García-Domínguez DJ, Hontecillas-Prieto L, Sánchez-Margalet V. Defining the Emergence of New Immunotherapy Approaches in Breast Cancer: Role of Myeloid-Derived Suppressor Cells. Int J Mol Sci 2023; 24:5208. [PMID: 36982282 PMCID: PMC10048951 DOI: 10.3390/ijms24065208] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
Breast cancer (BC) continues to be the most diagnosed tumor in women and a very heterogeneous disease both inter- and intratumoral, mainly given by the variety of molecular profiles with different biological and clinical characteristics. Despite the advancements in early detection and therapeutic strategies, the survival rate is low in patients who develop metastatic disease. Therefore, it is mandatory to explore new approaches to achieve better responses. In this regard, immunotherapy arose as a promising alternative to conventional treatments due to its ability to modulate the immune system, which may play a dual role in this disease since the relationship between the immune system and BC cells depends on several factors: the tumor histology and size, as well as the involvement of lymph nodes, immune cells, and molecules that are part of the tumor microenvironment. Particularly, myeloid-derived suppressor cell (MDSC) expansion is one of the major immunosuppressive mechanisms used by breast tumors since it has been associated with worse clinical stage, metastatic burden, and poor efficacy of immunotherapies. This review focuses on the new immunotherapies in BC in the last five years. Additionally, the role of MDSC as a therapeutic target in breast cancer will be described.
Collapse
Affiliation(s)
- María Luisa Sánchez-León
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Carlos Jiménez-Cortegana
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Silvia Silva Romeiro
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Carmen Garnacho
- Department of Normal and Pathological Cytology and Histology, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Luis de la Cruz-Merino
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Daniel J. García-Domínguez
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Lourdes Hontecillas-Prieto
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
- Oncology Service, Virgen Macarena University Hospital, Department of Medicine, School of Medicine, University of Seville, 41009 Seville, Spain
| | - Víctor Sánchez-Margalet
- Laboratory Service, Department of Medical Biochemistry, Molecular Biology and Immunology, School of Medicine, Virgen Macarena University Hospital, University of Seville, 41009 Seville, Spain
| |
Collapse
|
19
|
Brodzki A, Łopuszyński W, Brodzki P, Głodkowska K, Knap B, Gawin P. Pharmacological Treatment of Perianal Gland Tumors in Male Dogs. Animals (Basel) 2023; 13:ani13030463. [PMID: 36766353 PMCID: PMC9913509 DOI: 10.3390/ani13030463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/20/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
The presence of androgen (AR) and estrogen (ER) receptors has been demonstrated both in normal perianal (hepatoid) glands and in perianal tumors. The aim of this study was to demonstrate the relationship between the expression of AR and ER in perianal gland tumors and the effectiveness of antihormonal treatment. The study was performed on 41 male dogs with neoplastic lesions of the anal region. Histopathological evaluation of the lesions revealed 24 adenomas, 12 epitheliomas, and five carcinomas. Treatment was administered orally with tamoxifen at a dose of 1 mg/kg BW and cyproterone acetate at a dose of 5 mg/kg. Tumor diameters were measured regularly with calipers and recorded in millimeters starting with the measurement before treatment, and then after 1, 2, 3, 6, 12, 18, and 24 months of therapy. The results show that hepatoid adenomas that are characterized by high expression of AR and ER receptors respond positively to antihormonal therapy, resulting in complete tumor regression. For locally malignant hepatoid epitheliomas and carcinomas with low expression of AR and ER receptors, antihormonal therapy makes it possible to reduce the size of the tumor, but does not make it possible to cure it completely.
Collapse
Affiliation(s)
- Adam Brodzki
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-612 Lublin, Poland
| | - Wojciech Łopuszyński
- Department of Pathomorphology and Forensic Veterinary Medicine, University of Life Sciences in Lublin, 20-612 Lublin, Poland
- Correspondence: ; Tel.: +48-81-445-6162
| | - Piotr Brodzki
- Department of Andrology and Biotechnology of Reproduction, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-612 Lublin, Poland
| | - Katarzyna Głodkowska
- Department and Clinic of Animal Surgery, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, 20-612 Lublin, Poland
| | - Bartosz Knap
- Doctoral School, Medical University of Lublin, 20-093 Lublin, Poland
- Chair and Department of Experimental and Clinical Pharmacology, Faculty of Medicine, Medical University of Lublin, 20-090 Lublin, Poland
| | | |
Collapse
|
20
|
Shaaban S, Ji Y. Pharmacogenomics and health disparities, are we helping? Front Genet 2023; 14:1099541. [PMID: 36755573 PMCID: PMC9900000 DOI: 10.3389/fgene.2023.1099541] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 01/10/2023] [Indexed: 01/24/2023] Open
Abstract
Pharmacogenomics has been at the forefront of precision medicine during the last few decades. Precision medicine carries the potential of improving health outcomes at both the individual as well as population levels. To harness the benefits of its initiatives, careful dissection of existing health disparities as they relate to precision medicine is of paramount importance. Attempting to address the existing disparities at the early stages of design and implementation of these efforts is the only guarantee of a successful just outcome. In this review, we glance at a few determinants of existing health disparities as they intersect with pharmacogenomics research and implementation. In our opinion, highlighting these disparities is imperative for the purpose of researching meaningful solutions. Failing to identify, and hence address, these disparities in the context of the current and future precision medicine initiatives would leave an already strained health system, even more inundated with inequality.
Collapse
Affiliation(s)
- Sherin Shaaban
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, United States,ARUP Laboratories, Salt Lake City, Utah, United States,*Correspondence: Sherin Shaaban,
| | - Yuan Ji
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah, United States,ARUP Laboratories, Salt Lake City, Utah, United States
| |
Collapse
|
21
|
Guo L, Kong D, Liu J, Zhan L, Luo L, Zheng W, Zheng Q, Chen C, Sun S. Breast cancer heterogeneity and its implication in personalized precision therapy. Exp Hematol Oncol 2023; 12:3. [PMID: 36624542 PMCID: PMC9830930 DOI: 10.1186/s40164-022-00363-1] [Citation(s) in RCA: 70] [Impact Index Per Article: 70.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/29/2022] [Indexed: 01/11/2023] Open
Abstract
Breast cancer heterogeneity determines cancer progression, treatment effects, and prognosis. However, the precise mechanism for this heterogeneity remains unknown owing to its complexity. Here, we summarize the origins of breast cancer heterogeneity and its influence on disease progression, recurrence, and therapeutic resistance. We review the possible mechanisms of heterogeneity and the research methods used to analyze it. We also highlight the importance of cell interactions for the origins of breast cancer heterogeneity, which can be further categorized into cooperative and competitive interactions. Finally, we provide new insights into precise individual treatments based on heterogeneity.
Collapse
Affiliation(s)
- Liantao Guo
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Deguang Kong
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Jianhua Liu
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Ling Zhan
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Lan Luo
- Department of Breast Surgery, The Affiliated Hospital of Guizhou Medical University, No. 28 Guiyi Road, Yunyan District, Guiyang, 550001, Guizhou, China
| | - Weijie Zheng
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Qingyuan Zheng
- Department of Urology, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China
| | - Chuang Chen
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.
| | - Shengrong Sun
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, No. 238 Jiefang Road, Wuchang District, Wuhan, 430060, Hubei, China.
| |
Collapse
|
22
|
Kaneko Y, Yamatsugu K, Yamashita T, Takahashi K, Tanaka T, Aki S, Tatsumi T, Kawamura T, Miura M, Ishii M, Ohkubo K, Osawa T, Kodama T, Ishikawa S, Tsukagoshi M, Chansler M, Sugiyama A, Kanai M, Katoh H. Pathological complete remission of relapsed tumor by photo-activating antibody-mimetic drug conjugate treatment. Cancer Sci 2022; 113:4350-4362. [PMID: 36121618 DOI: 10.1111/cas.15565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/12/2022] [Accepted: 08/23/2022] [Indexed: 02/03/2023] Open
Abstract
Antibody-mimetic drug conjugate is a novel noncovalent conjugate consisting of an antibody-mimetic recognizing a target molecule on the cancer cell surface and low-molecular-weight payloads that kill the cancer cells. In this study, the efficacy of a photo-activating antibody-mimetic drug conjugate targeting HER2-expressing tumors was evaluated in mice, by using the affibody that recognize HER2 (ZHER2:342 ) as a target molecule and an axially substituted silicon phthalocyanine (a novel potent photo-activating compound) as a payload. The first treatment with the photo-activating antibody-mimetic drug conjugates reduced the size of all HER2-expressing KPL-4 xenograft tumors macroscopically. However, during the observation period, relapsed tumors gradually appeared in approximately 50% of the animals. To evaluate the efficacy of repeated antibody-mimetic drug conjugate treatment, animals with relapsed tumors were treated again with the same regimen. After the second observation period, the mouse tissues were examined histopathologically. Unexpectedly, all relapsed tumors were eradicated, and all animals were diagnosed with pathological complete remission. After the second treatment, skin wounds healed rapidly, and no significant side effects were observed in other organs, except for occasional microscopic granulomatous tissues beneath the serosa of the liver in a few mice. Repeated treatments seemed to be well tolerated. These results indicate the promising efficacy of the repeated photo-activating antibody-mimetic drug conjugate treatment against HER2-expressing tumors.
Collapse
Affiliation(s)
- Yudai Kaneko
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.,Medical & Biological Laboratories Co., Ltd, Tokyo, Japan
| | - Kenzo Yamatsugu
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Takefumi Yamashita
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kazuki Takahashi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshiya Tanaka
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Sho Aki
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Toshifumi Tatsumi
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Takeshi Kawamura
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.,Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Mai Miura
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Masazumi Ishii
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Kei Ohkubo
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan.,Institute for Advanced Co-Creation Studies, Osaka University, Osaka, Japan
| | - Tsuyoshi Osawa
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Tatsuhiko Kodama
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan
| | - Shumpei Ishikawa
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | | | | | - Akira Sugiyama
- Research Center for Advanced Science and Technology, The University of Tokyo, Tokyo, Japan.,Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Motomu Kanai
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Hiroto Katoh
- Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
23
|
Ingthorsson S, Traustadottir GA, Gudjonsson T. Cellular Plasticity and Heterotypic Interactions during Breast Morphogenesis and Cancer Initiation. Cancers (Basel) 2022; 14:cancers14215209. [PMID: 36358627 PMCID: PMC9654604 DOI: 10.3390/cancers14215209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/19/2022] [Accepted: 10/20/2022] [Indexed: 12/01/2022] Open
Abstract
Simple Summary This review aims to discuss the structure, function and dynamics of the breast gland and how changes to the function of the breast’s cells can lead to different types of cancer. Abstract The human breast gland is a unique organ as most of its development occurs postnatally between menarche and menopause, a period ranging from 30 to 40 years. During this period, the monthly menstruation cycle drives the mammary gland through phases of cell proliferation, differentiation, and apoptosis, facilitated via a closely choreographed interaction between the epithelial cells and the surrounding stroma preparing the gland for pregnancy. If pregnancy occurs, maximal differentiation is reached to prepare for lactation. After lactation, the mammary gland involutes to a pre-pregnant state. These cycles of proliferation, differentiation, and involution necessitate the presence of epithelial stem cells that give rise to progenitor cells which differentiate further into the luminal and myoepithelial lineages that constitute the epithelial compartment and are responsible for the branching structure of the gland. Maintaining homeostasis and the stem cell niche depends strongly on signaling between the stem and progenitor cells and the surrounding stroma. Breast cancer is a slowly progressing disease whose initiation can take decades to progress into an invasive form. Accumulating evidence indicates that stem cells and/or progenitor cells at different stages, rather than terminally differentiated cells are the main cells of origin for most breast cancer subgroups. Stem cells and cancer cells share several similarities such as increased survival and cellular plasticity which is reflected in their ability to switch fate by receiving intrinsic and extrinsic signals. In this review, we discuss the concept of cellular plasticity in normal breast morphogenesis and cancer, and how the stromal environment plays a vital role in cancer initiation and progression.
Collapse
Affiliation(s)
- Saevar Ingthorsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, 101 Reykjavik, Iceland
- Faculty of nursing and midwifery, School of Health Sciences, University of Iceland, 101 Reykjavik, Iceland
| | - Gunnhildur Asta Traustadottir
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, 101 Reykjavik, Iceland
- Department of Pathology, Landspitali University Hospital, 101 Reykjavik, Iceland
| | - Thorarinn Gudjonsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, 101 Reykjavik, Iceland
- Department of Laboratory Hematology, Landspitali University Hospital, 101 Reykjavik, Iceland
- Correspondence:
| |
Collapse
|
24
|
Grimm M, Radcliff L, Giles M, Nash R, Holley E, Panda S, Brophy L, Williams N, Cherian M, Stover D, Gatti-Mays ME, Wesolowski R, Sardesai S, Sudheendra P, Reinbolt R, Ramaswamy B, Pariser A. Living with Advanced Breast Cancer: A Descriptive Analysis of Survivorship Strategies. J Clin Med 2022; 11:jcm11143992. [PMID: 35887755 PMCID: PMC9319697 DOI: 10.3390/jcm11143992] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/27/2022] [Accepted: 07/05/2022] [Indexed: 02/04/2023] Open
Abstract
Survivors of advanced breast cancer (ABC), also known as metavivors, are often left with fewer treatment options in the landscape of a cure culture. Metavivors have unique psychosocial and physical needs distinct from patients with early-stage breast cancer. This analysis delves into side effects commonly experienced by patients with ABC, such as fatigue, anxiety, and cardiotoxicity; how these side effects impact caregiver support, financial toxicity, emotional strain, and spiritual and emotional distress; as well as current strategies for mitigation, including nutrition, exercise, and participation in clinical research. Overall, this analysis is a mandate for additional research to explore novel treatments and implement strategies to maintain and improve patients’ quality of life.
Collapse
Affiliation(s)
- Michael Grimm
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Lindsey Radcliff
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Mariann Giles
- Family Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Ryan Nash
- Center for Bioethics and Medical Humanities, The Ohio State University, Columbus, OH 43210, USA;
| | - Erin Holley
- Nutrition and Dietetics, The Ohio State University, Columbus, OH 43210, USA;
| | - Shannon Panda
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Lynne Brophy
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Nicole Williams
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Mathew Cherian
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Daniel Stover
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Margaret E. Gatti-Mays
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Robert Wesolowski
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Sagar Sardesai
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Preeti Sudheendra
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Raquel Reinbolt
- Internal Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Bhuvaneswari Ramaswamy
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
| | - Ashley Pariser
- Division of Medical Oncology, Stefanie Spielman Comprehensive Breast Cancer, The Ohio State University, Columbus, OH 43210, USA; (M.G.); (L.R.); (S.P.); (L.B.); (N.W.); (M.C.); (D.S.); (M.E.G.-M.); (R.W.); (S.S.); (P.S.); (B.R.)
- Correspondence: ; Tel.: +1-614-293-6401
| |
Collapse
|
25
|
Wang Y, Yuan X, Li J, Liu Z, Li X, Wang Z, Wei L, Li Y, Wang X. The Synergistic Effects of SHR6390 Combined With Pyrotinib on HER2+/HR+ Breast Cancer. Front Cell Dev Biol 2021; 9:785796. [PMID: 34977029 PMCID: PMC8716872 DOI: 10.3389/fcell.2021.785796] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 10/31/2021] [Indexed: 11/13/2022] Open
Abstract
HER2+/HR+ breast cancer is a special molecular type of breast cancer. Existing treatment methods are prone to resistance; “precision treatment” is necessary. Pyrotinib is a pan-her kinase inhibitor that can be used in HER2-positive tumors, while SHR6390 is a CDK4/6 inhibitor that can inhibit ER+ breast cancer cell cycle progression and cancer cell proliferation. In cancer cells, HER2 and CDK4/6 signaling pathways could be nonredundant; co-inhibition of both pathways by combination of SHR6390 and pyrotinib may have synergistic anticancer activity on HER2+/HR+ breast cancer. In this study, we determined the synergy of the two-drug combination and underlying molecular mechanisms. We showed that the combination of SHR6390 and pyrotinib synergistically inhibited the proliferation, migration, and invasion of HER2+/HR+ breast cancer cells in vitro. The combination of two drugs induced G1/S phase arrest and apoptosis in HER2+/HR+ breast cancer cell lines. The combination of two drugs prolonged the time to tumor recurrence in the xenograft model system. By second-generation RNA sequencing technology and enrichment analysis of the pyrotinib-resistant cell line, we found that FOXM1 was associated with induced resistance to HER2-targeted therapy. In HER2+/HR+ breast cancer cell lines, the combination of the two drugs could further reduce FOXM1 phosphorylation, thereby enhancing the antitumor effect to a certain extent. These findings suggest that SHR6390 combination with pyrotinib suppresses the proliferation, migration, and invasion of HER2+/HR+ breast cancers through regulation of FOXM1.
Collapse
Affiliation(s)
- Yukun Wang
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Xiang Yuan
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Jing Li
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Zhiwei Liu
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Xinyang Li
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Ziming Wang
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Limin Wei
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
| | - Yuanpei Li
- UC Davis Comprehensive Cancer Center, Department of Internal Medicine, University of California, Davis, Davis, CA, United States
| | - Xinshuai Wang
- Henan Key Laboratory of Cancer Epigenetics, Cancer Hospital, The First Affiliated Hospital, College of Clinical Medicine, Medical College of Henan University of Science and Technology, Luoyang, China
- *Correspondence: Xinshuai Wang,
| |
Collapse
|
26
|
Enrico TP, Stallaert W, Wick ET, Ngoi P, Wang X, Rubin SM, Brown NG, Purvis JE, Emanuele MJ. Cyclin F drives proliferation through SCF-dependent degradation of the retinoblastoma-like tumor suppressor p130/RBL2. eLife 2021; 10:70691. [PMID: 34851822 PMCID: PMC8670743 DOI: 10.7554/elife.70691] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 11/19/2021] [Indexed: 12/20/2022] Open
Abstract
Cell cycle gene expression programs fuel proliferation and are universally dysregulated in cancer. The retinoblastoma (RB)-family of proteins, RB1, RBL1/p107, and RBL2/p130, coordinately represses cell cycle gene expression, inhibiting proliferation, and suppressing tumorigenesis. Phosphorylation of RB-family proteins by cyclin-dependent kinases is firmly established. Like phosphorylation, ubiquitination is essential to cell cycle control, and numerous proliferative regulators, tumor suppressors, and oncoproteins are ubiquitinated. However, little is known about the role of ubiquitin signaling in controlling RB-family proteins. A systems genetics analysis of CRISPR/Cas9 screens suggested the potential regulation of the RB-network by cyclin F, a substrate recognition receptor for the SCF family of E3 ligases. We demonstrate that RBL2/p130 is a direct substrate of SCFcyclin F. We map a cyclin F regulatory site to a flexible linker in the p130 pocket domain, and show that this site mediates binding, stability, and ubiquitination. Expression of a mutant version of p130, which cannot be ubiquitinated, severely impaired proliferative capacity and cell cycle progression. Consistently, we observed reduced expression of cell cycle gene transcripts, as well a reduced abundance of cell cycle proteins, analyzed by quantitative, iterative immunofluorescent imaging. These data suggest a key role for SCFcyclin F in the CDK-RB network and raise the possibility that aberrant p130 degradation could dysregulate the cell cycle in human cancers.
Collapse
Affiliation(s)
- Taylor P Enrico
- Department of Pharmacology. The University of North Carolina at Chapel Hill, Chapel Hill, United States.,Lineberger Comprehensive Cancer Center. The University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Wayne Stallaert
- Department of Genetics. The University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Elizaveta T Wick
- Department of Pharmacology. The University of North Carolina at Chapel Hill, Chapel Hill, United States.,Lineberger Comprehensive Cancer Center. The University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Peter Ngoi
- Department of Chemistry and Biochemistry. University of California at Santa Cruz, Santa Cruz, United States
| | - Xianxi Wang
- Lineberger Comprehensive Cancer Center. The University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Seth M Rubin
- Department of Chemistry and Biochemistry. University of California at Santa Cruz, Santa Cruz, United States
| | - Nicholas G Brown
- Department of Pharmacology. The University of North Carolina at Chapel Hill, Chapel Hill, United States.,Lineberger Comprehensive Cancer Center. The University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Jeremy E Purvis
- Lineberger Comprehensive Cancer Center. The University of North Carolina at Chapel Hill, Chapel Hill, United States.,Department of Genetics. The University of North Carolina at Chapel Hill, Chapel Hill, United States
| | - Michael J Emanuele
- Department of Pharmacology. The University of North Carolina at Chapel Hill, Chapel Hill, United States.,Lineberger Comprehensive Cancer Center. The University of North Carolina at Chapel Hill, Chapel Hill, United States
| |
Collapse
|