51
|
Park HM, Park JY, Kim NY, Kim J, Pham TH, Hong JT, Yoon DY. Modulatory effects of point-mutated IL-32θ (A94V) on tumor progression in triple-negative breast cancer cells. Biofactors 2024; 50:294-310. [PMID: 37658685 DOI: 10.1002/biof.2005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 08/01/2023] [Indexed: 09/03/2023]
Abstract
Breast cancer is a frequently diagnosed cancer and the leading cause of death among women worldwide. Tumor-associated macrophages stimulate cytokines and chemokines, which induce angiogenesis, metastasis, proliferation, and tumor-infiltrating immune cells. Although interleukin-32 (IL-32) has been implicated in the development and modulation of several cancers, its function in breast cancer remains elusive. Mutation of interleukin-32θ (IL-32θ) in the tissues of patients with breast cancer was detected by Sanger sequencing. RT-qPCR was used to detect the mRNA levels of inflammatory cytokines, chemokines, and mediators. The secreted proteins were detected using respective enzyme-linked immunosorbent assays. Evaluation of the inhibitory effect of mutant IL-32θ on proliferation, migration, epithelial-mesenchymal transition (EMT), and cell cycle arrest in breast cancer cells was conducted using MTS assays, migration assays, and Western blotting. A point mutation (281C>T, Ala94Val) was detected in IL-32θ in both breast tumors and adjacent normal tissues, which suppressed the expression of pro-inflammatory factors, EMT factors, and cell cycle related factors. Mutated IL-32θ inhibited the expression of inflammatory factors by regulating the NF-κB pathway. Furthermore, mutated IL-32θ suppressed EMT markers and cell cycle related factors through the FAK/PI3K/AKT pathway. It was inferred that mutated IL-32θ modulates breast cancer progression. Mutated IL-32θ (A94V) inhibited inflammation, EMT, and proliferation in breast cancer by regulating the NF-κB (p65/p50) and FAK-PI3K-GSK3 pathways.
Collapse
Affiliation(s)
- Hyo-Min Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Jae-Young Park
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Na-Yeon Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Jinju Kim
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Thu-Huyen Pham
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy and Medical Research Center, Chungbuk National University, Chungbuk, Republic of Korea
| | - Do-Young Yoon
- Department of Bioscience and Biotechnology, Konkuk University, Seoul, Republic of Korea
| |
Collapse
|
52
|
Wu H, Jiang Y, Tian H, Ye X, Cui C, Shi S, Chen M, Ding Z, Li S, Huang Z, Luo Y, Peng Q, Xu J, Dong F. Sonography-based multimodal information platform for identifying the surgical pathology of ductal carcinoma in situ. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 245:108039. [PMID: 38266556 DOI: 10.1016/j.cmpb.2024.108039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 01/26/2024]
Abstract
BACKGROUND The risk of ductal carcinoma in situ (DCIS) identified by biopsy often increases during surgery. Therefore, confirming the DCIS grade preoperatively is necessary for clinical decision-making. PURPOSE To train a three-classification deep learning (DL) model based on ultrasound (US), combining clinical data, mammography (MG), US, and core needle biopsy (CNB) pathology to predict low-grade DCIS, intermediate-to-high-grade DCIS, and upstaged DCIS. MATERIALS AND METHODS Data of 733 patients with 754 DCIS cases confirmed by biopsy were retrospectively collected from May 2013 to June 2022 (N1), and other data (N2) were confirmed by biopsy as low-grade DCIS. The lesions were randomly divided into training (n=471), validation (n=142), and test (n = 141) sets to establish the DCIS-Net. Information on the DCIS-Net, clinical (age and sign), US (size, calcifications, type, breast imaging reporting and data system [BI-RADS]), MG (microcalcifications, BI-RADS), and CNB pathology (nuclear grade, architectural features, and immunohistochemistry) were collected. Logistic regression and random forest analyses were conducted to develop Multimodal DCIS-Net to calculate the specificity, sensitivity, accuracy, receiver operating characteristic curve, and area under the curve (AUC). RESULTS In the test set of N1, the accuracy and AUC of the multimodal DCIS-Net were 0.752-0.766 and 0.859-0.907 in the three-classification task, respectively. The accuracy and AUC for discriminating DCIS from upstaged DCIS were 0.751-0.780 and 0.829-0.861, respectively. In the test set of N2, the accuracy and AUC of discriminating low-grade DCIS from upstaged low-grade DCIS were 0.769-0.987 and 0.818-0.939, respectively. DL was ranked from one to five in the importance of features in the multimodal-DCIS-Net. CONCLUSION By developing the DCIS-Net and integrating it with multimodal information, diagnosing low-grade DCIS, intermediate-to high-grade DCIS, and upstaged DCIS is possible. It can also be used to distinguish DCIS from upstaged DCIS and low-grade DCIS from upstaged low-grade DCIS, which could pave the way for the DCIS clinical workflow.
Collapse
Affiliation(s)
- Huaiyu Wu
- Department of Ultrasound, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China
| | - Yitao Jiang
- Department of Ultrasound, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China; Research and Development Department, Microport Prophecy, Shanghai 201203, China
| | - Hongtian Tian
- Department of Ultrasound, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China
| | - Xiuqin Ye
- Department of Ultrasound, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China
| | - Chen Cui
- Research and Development Department, Illuminate, LLC, Shenzhen, Guangdong 518000, China
| | - Siyuan Shi
- Research and Development Department, Illuminate, LLC, Shenzhen, Guangdong 518000, China
| | - Ming Chen
- Department of Ultrasound, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China
| | - Zhimin Ding
- Department of Ultrasound, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China
| | - Shiyu Li
- Department of Ultrasound, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China
| | - Zhibin Huang
- Department of Ultrasound, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China
| | - Yuwei Luo
- Department of Breast Surgery, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China; Department of General Surgery, Shenzhen People's Hospital, Shenzhen 518020, Guangdong, China
| | - Quanzhou Peng
- Department of Pathology, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China
| | - Jinfeng Xu
- Department of Ultrasound, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China
| | - Fajin Dong
- Department of Ultrasound, The Second Clinical Medical College, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen People's Hospital, Jinan University, Shenzhen 518020, Guangdong, China.
| |
Collapse
|
53
|
Wang Y, Wang DY, Bu KN, Gao JD, Zhang BL. Prognosis prediction and risk stratification of breast cancer patients based on a mitochondria-related gene signature. Sci Rep 2024; 14:2859. [PMID: 38310106 PMCID: PMC10838276 DOI: 10.1038/s41598-024-52981-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 01/25/2024] [Indexed: 02/05/2024] Open
Abstract
As the malignancy with the highest global incidence, breast cancer represents a significant threat to women's health. Recent advances have shed light on the importance of mitochondrial function in cancer, particularly in metabolic reprogramming within tumors. Recognizing this, we developed a novel risk signature based on mitochondrial-related genes to improve prognosis prediction and risk stratification in breast cancer patients. In this study, transcriptome data and clinical features of breast cancer samples were extracted from two sources: the TCGA, serving as the training set, and the METABRIC, used as the independent validation set. We developed the signature using LASSO-Cox regression and assessed its prognostic efficacy via ROC curves. Furthermore, the signature was integrated with clinical features to create a Nomogram model, whose accuracy was validated through clinical calibration curves and decision curve analysis. To further elucidate prognostic variations between high and low-risk groups, we conducted functional enrichment and immune infiltration analyses. Additionally, the study encompassed a comparison of mutation landscapes and drug sensitivity, providing a comprehensive understanding of the differing characteristics in these groups. Conclusively, we established a risk signature comprising 8 mitochondrial-related genes-ACSL1, ALDH2, MTHFD2, MRPL13, TP53AIP1, SLC1A1, ME3, and BCL2A1. This signature was identified as an independent risk predictor for breast cancer patient survival, exhibiting a significant high hazard ratio (HR = 3.028, 95%CI 2.038-4.499, P < 0.001). Patients in the low-risk group showed a more favorable prognosis, with enhanced immune infiltration, distinct mutation landscapes, and greater sensitivity to anti-tumor drugs. In contrast, the high-risk group exhibited an adverse trend in these aspects. This risk signature represents a novel and effective prognostic indicator, suggesting valuable insights for patient stratification in breast cancer.
Collapse
Affiliation(s)
- Yang Wang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ding-Yuan Wang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Ke-Na Bu
- Xingyuan Hospital of Yulin City, Yulin City, 719051, Shanxi Province, China
| | - Ji-Dong Gao
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union College, Shenzhen, 518116, China.
| | - Bai-Lin Zhang
- Department of Breast Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
54
|
Chen W, Kang Y, Sheng W, Huang Q, Cheng J, Pei S, Meng Y. A new 4-gene-based prognostic model accurately predicts breast cancer prognosis and immunotherapy response by integrating WGCNA and bioinformatics analysis. Front Immunol 2024; 15:1331841. [PMID: 38370403 PMCID: PMC10869553 DOI: 10.3389/fimmu.2024.1331841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Background Breast cancer (BRCA) is a common malignancy in women, and its resistance to immunotherapy is a major challenge. Abnormal expression of genes is important in the occurrence and development of BRCA and may also affect the prognosis of patients. Although many BRCA prognosis model scores have been developed, they are only applicable to a limited number of disease subtypes. Our goal is to develop a new prognostic score that is more accurate and applicable to a wider range of BRCA patients. Methods BRCA patient data from The Cancer Genome Atlas database was used to identify breast cancer-related genes (BRGs). Differential expression analysis of BRGs was performed using the 'limma' package in R. Prognostic BRGs were identified using co-expression and univariate Cox analysis. A predictive model of four BRGs was established using Cox regression and the LASSO algorithm. Model performance was evaluated using K-M survival and receiver operating characteristic curve analysis. The predictive ability of the signature in immune microenvironment and immunotherapy was investigated. In vitro experiments validated POLQ function. Results Our study identified a four-BRG prognostic signature that outperformed conventional clinicopathological characteristics in predicting survival outcomes in BRCA patients. The signature effectively stratified BRCA patients into high- and low-risk groups and showed potential in predicting the response to immunotherapy. Notably, significant differences were observed in immune cell abundance between the two groups. In vitro experiments demonstrated that POLQ knockdown significantly reduced the viability, proliferation, and invasion capacity of MDA-MB-231 or HCC1806 cells. Conclusion Our 4-BRG signature has the potential as an independent biomarker for predicting prognosis and treatment response in BRCA patients, complementing existing clinicopathological characteristics.
Collapse
Affiliation(s)
- Wenlong Chen
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Yakun Kang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wenyi Sheng
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Qiyan Huang
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Jiale Cheng
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Shengbin Pei
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- Department of Breast Surgical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - You Meng
- Department of Thyroid and Breast Surgery, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| |
Collapse
|
55
|
Moura MLV, de Menezes AAPM, de Oliveira Filho JWG, do Nascimento MLLB, dos Reis AC, Ribeiro AB, da Silva FCC, Nunes AMV, Rolim HML, de Carvalho Melo Cavalcante AA, Sousa JMDCE. Advances in Antitumor Effects Using Liposomal Citrinin in Induced Breast Cancer Model. Pharmaceutics 2024; 16:174. [PMID: 38399235 PMCID: PMC10892831 DOI: 10.3390/pharmaceutics16020174] [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: 08/09/2023] [Revised: 12/19/2023] [Accepted: 12/27/2023] [Indexed: 02/25/2024] Open
Abstract
The study aimed to evaluate the antitumor and toxicogenetic effects of liposomal nanoformulations containing citrinin in animal breast carcinoma induced by 7,12-dimethylbenzanthracene (DMBA). Mus musculus virgin females were divided into six groups treated with (1) olive oil (10 mL/kg); (2) 7,12-DMBA (6 mg/kg); (3) citrinin, CIT (2 mg/kg), (4) cyclophosphamide, CPA (25 mg/kg), (5) liposomal citrinin, LP-CIT (2 μg/kg), and (6) LP-CIT (6 µg/kg). Metabolic, behavioral, hematological, biochemical, histopathological, and toxicogenetic tests were performed. DMBA and cyclophosphamide induced behavioral changes, not observed for free and liposomal citrinin. No hematological or biochemical changes were observed for LP-CIT. However, free citrinin reduced monocytes and caused hepatotoxicity. During treatment, significant differences were observed regarding the weight of the right and left breasts treated with DMBA compared to negative controls. Treatment with CPA, CIT, and LP-CIT reduced the weight of both breasts, with better results for liposomal citrinin. Furthermore, CPA, CIT, and LP-CIT presented genotoxic effects for tumor, blood, bone marrow, and liver cells, although less DNA damage was observed for LP-CIT compared to CIT and CPA. Healthy cell damage induced by LP-CIT was repaired during treatment, unlike CPA, which caused clastogenic effects. Thus, LP-CIT showed advantages for its use as a model of nanosystems for antitumor studies.
Collapse
Affiliation(s)
- Michely Laiany Vieira Moura
- Laboratory of Toxicological Genetics—LAPGENIC, Graduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina 64049-550, Brazil; (M.L.V.M.); (A.-A.P.M.d.M.); (J.W.G.d.O.F.); (M.L.L.B.d.N.); (A.C.d.R.); (F.C.C.d.S.); (A.A.d.C.M.C.); (J.M.d.C.e.S.)
| | - Ag-Anne Pereira Melo de Menezes
- Laboratory of Toxicological Genetics—LAPGENIC, Graduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina 64049-550, Brazil; (M.L.V.M.); (A.-A.P.M.d.M.); (J.W.G.d.O.F.); (M.L.L.B.d.N.); (A.C.d.R.); (F.C.C.d.S.); (A.A.d.C.M.C.); (J.M.d.C.e.S.)
| | - José Williams Gomes de Oliveira Filho
- Laboratory of Toxicological Genetics—LAPGENIC, Graduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina 64049-550, Brazil; (M.L.V.M.); (A.-A.P.M.d.M.); (J.W.G.d.O.F.); (M.L.L.B.d.N.); (A.C.d.R.); (F.C.C.d.S.); (A.A.d.C.M.C.); (J.M.d.C.e.S.)
| | - Maria Luiza Lima Barreto do Nascimento
- Laboratory of Toxicological Genetics—LAPGENIC, Graduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina 64049-550, Brazil; (M.L.V.M.); (A.-A.P.M.d.M.); (J.W.G.d.O.F.); (M.L.L.B.d.N.); (A.C.d.R.); (F.C.C.d.S.); (A.A.d.C.M.C.); (J.M.d.C.e.S.)
| | - Antonielly Campinho dos Reis
- Laboratory of Toxicological Genetics—LAPGENIC, Graduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina 64049-550, Brazil; (M.L.V.M.); (A.-A.P.M.d.M.); (J.W.G.d.O.F.); (M.L.L.B.d.N.); (A.C.d.R.); (F.C.C.d.S.); (A.A.d.C.M.C.); (J.M.d.C.e.S.)
| | - Alessandra Braga Ribeiro
- CBQF—Centro de Biotecnologia e Química Fina—Laboratório Associado, Escola Superior de Biotecnologia, Universidade Católica Portuguesa, Rua Diogo Botelho 1327, 4169-005 Porto, Portugal;
| | - Felipe Cavalcanti Carneiro da Silva
- Laboratory of Toxicological Genetics—LAPGENIC, Graduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina 64049-550, Brazil; (M.L.V.M.); (A.-A.P.M.d.M.); (J.W.G.d.O.F.); (M.L.L.B.d.N.); (A.C.d.R.); (F.C.C.d.S.); (A.A.d.C.M.C.); (J.M.d.C.e.S.)
| | | | - Hercília Maria Lins Rolim
- Laboratory of Pharmaceutical Nanosystems—NANOSFAR, Graduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina 64049-550, Brazil
| | - Ana Amélia de Carvalho Melo Cavalcante
- Laboratory of Toxicological Genetics—LAPGENIC, Graduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina 64049-550, Brazil; (M.L.V.M.); (A.-A.P.M.d.M.); (J.W.G.d.O.F.); (M.L.L.B.d.N.); (A.C.d.R.); (F.C.C.d.S.); (A.A.d.C.M.C.); (J.M.d.C.e.S.)
| | - João Marcelo de Castro e Sousa
- Laboratory of Toxicological Genetics—LAPGENIC, Graduate Program in Pharmaceutical Sciences, Federal University of Piauí, Teresina 64049-550, Brazil; (M.L.V.M.); (A.-A.P.M.d.M.); (J.W.G.d.O.F.); (M.L.L.B.d.N.); (A.C.d.R.); (F.C.C.d.S.); (A.A.d.C.M.C.); (J.M.d.C.e.S.)
| |
Collapse
|
56
|
Cheng S, Zhang C, Hu X, Zhu Y, Shi H, Tan W, Luo X, Xian Y. Ultrasensitive determination of surface proteins on tumor-derived small extracellular vesicles for breast cancer identification based on lanthanide-activated signal amplification strategy. Talanta 2024; 267:125189. [PMID: 37714039 DOI: 10.1016/j.talanta.2023.125189] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/04/2023] [Accepted: 09/08/2023] [Indexed: 09/17/2023]
Abstract
Small extracellular vesicles (sEVs) carrying multiple tumor-associated proteins inherited from parental cells play crucial roles in noninvasive breast cancer (BC) diagnosis. However, it is challenging to assess the subtle variations of surface proteins on sEV membranes due to the highly heterogeneous BC. Therefore, a simple and ultrasensitive assay based on lanthanide (Ln3+)-activated luminescence signal amplification was developed to detect multiple surface proteins on BC-derived sEVs. Multiple protein biomarkers on sEVs can be well identified with high sensitivity and specificity through dissolution-amplified luminescence of the NaEuF4 nanoparticle-based nanoprobe. We employ linear discriminant analysis to successfully discriminate triple negative BC cell (MDA-MB-231 cell) derived sEVs from other breast cell lines (MCF-7, SK-BR-3, BT474 and MCF-10A cell). Furthermore, the strategy enables high accuracy for districting the progression stages of BC patients and healthy donors. The simple and sensitive signal amplification strategy exhibits great potential for early clinic diagnosis by precise protein profiling of sEVs.
Collapse
Affiliation(s)
- Shasha Cheng
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Cuiling Zhang
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China.
| | - Xinyu Hu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Yingxin Zhu
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Hui Shi
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, Institute of Stem Cell, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wenqiao Tan
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Xianzhu Luo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Yuezhong Xian
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, Department of Chemistry, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China.
| |
Collapse
|
57
|
Yu H, Liu J. Identification of breast cancer subgroups and immune characterization based on glutamine metabolism-related genes. BMC Med Genomics 2024; 17:17. [PMID: 38200578 PMCID: PMC10782609 DOI: 10.1186/s12920-023-01792-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Immunotherapy is a promising treatment for breast cancer (BC). However, due to individual differences and tumor heterogeneity, immunotherapy is only applicable to some BC patients. Glutamine metabolism plays a role in inhibiting immunotherapy, but its role in BC is limitedly studied. Therefore, we aimed to identify different BC subgroups based on glutamine metabolism and characterize the features of different subgroups to provide guidance for personalized immunotherapy for BC patients. Using unsupervised clustering analysis, we classified BC patients in The Cancer Genome Atlas (TCGA) with glutamine metabolism-related genes and obtained low-risk (LR) and high-risk (HR) subgroups. Survival analysis revealed that prognosis of LR subgroup was notably better than HR subgroup. Through ssGSEA and CIBERSORT methods, we disclosed that infiltration levels of B cells, Mast cells, T helper cells, and Th2 cells, and Type II IFN Response immune function were notably higher in LR subgroup than in HR subgroup. The Wilcox algorithm comparison denoted that DEPTH of LR subgroup was significantly lower than HR subgroup. The TIDE of LR subgroup was significantly higher than HR subgroup. Functional annotation of differentially expressed genes revealed that channel activity and the Estrogen signaling pathway may be related to BC prognosis. Ten hub genes were selected between the subgroups through the STRING database and Cytoscape, and their correlation with drugs was predicted on the CellMiner website. This study analyzed the immune characteristics of BC subgroups based on glutamine metabolism and provided reference for prognosis prediction and personalized immunotherapy.
Collapse
Affiliation(s)
- Hongjing Yu
- Department of Oncology, Jiande Branch, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Junchen Liu
- Department of Pharmacy, Jiande Branch, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
58
|
Wang L, Li P, Zhang X, Gu Z, Pan X, Wu Y, Li H. The role of basic leucine zipper transcription factor E4BP4 in cancer: a review and update. Mol Biol Rep 2024; 51:91. [PMID: 38193973 DOI: 10.1007/s11033-023-09079-9] [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: 07/21/2023] [Accepted: 10/25/2023] [Indexed: 01/10/2024]
Abstract
Mutations in the genes of tumor cells and the disorder of immune microenvironment are the main factors of tumor development. The sensitivity of tumor cells to chemotherapy drugs affect the treatment of tumor. Nuclear transcription factor E4BP4 is dysregulated in a variety of malignant tumors. It can suppress the transcription of NFKBIA, RASSF8, SOSTDC1, FOXO-induced genes (TRAIL, FAS, GADD45a and GADD45b) and Hepcidin, up-regulate RCAN1-1 and PRNP, activate mTOR and p38 in cancer cells. Also, E4BP4 can regulate tumor immune microenvironment. TGFb1/Smad3/E4BP4/ IFNγ axis in NK cells plays an important role in antitumor immunotherapy. Over expression of E4BP4 inhibited the development of Th17 cells by directly binding to the RORγt promoter. Moreover, recent studies have shown that E4BP4 inhibited the expression of multidrug resistance genes. In this review, we summarize the molecular mechanism of E4BP4 in cancer cellular process, the effects of E4BP4 in cancer immunotherapy and antitumor drug resistance, to provide theoretical basis for tumor treatment strategies targeting E4BP4.
Collapse
Affiliation(s)
- Liang Wang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, School of Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Peifen Li
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, School of Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Xueying Zhang
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, School of Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Zhenwu Gu
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, School of Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Xinyu Pan
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, School of Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Yihao Wu
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, School of Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China
| | - Huanan Li
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, School of Medicine, Yangzhou University, Yangzhou, 225009, Jiangsu, China.
| |
Collapse
|
59
|
Sharma P, Gupta K, Khandai SK, Malik S, Thareja S. Phytometabolites as modulators of breast cancer: a comprehensive review of mechanistic insights. Med Oncol 2024; 41:45. [PMID: 38172452 DOI: 10.1007/s12032-023-02269-2] [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: 10/12/2023] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
Breast cancer (BC) is a highly debilitating malignancy affecting females globally and imposing a substantial burden on healthcare systems in both developed and developing nations. Despite the application of conventional therapeutic modalities such as chemotherapy, radiation therapy, and hormonal intervention, BC frequently exhibits resistance, necessitating the urgent development of novel, cost-effective, and accessible treatment strategies. In this context, there is a growing scientific interest in exploring the pharmacological potential of chemical compounds derived from botanical sources, which often exhibit notable biological activity. Extensive in vitro and in vivo investigations have revealed the capacity of these compounds, referred to as phytochemicals, to attenuate the metastatic cascade and reduce the risk of cancer dissemination. These phytochemicals exert their effects through modulation of key molecular and metabolic processes, including regulation of the cell cycle, induction of apoptotic cell death, inhibition of angiogenesis, and suppression of metastatic progression. To shed light on the latest advancements in this field, a comprehensive review of the scientific literature has been conducted, focusing on secondary metabolite agents that have recently been investigated and have demonstrated promising anticancer properties. This review aims to delineate their underlying mechanisms of action and elucidate the associated signaling pathways, thereby contributing to a deeper understanding of their therapeutic potential in the context of BC management.
Collapse
Affiliation(s)
- Priyanka Sharma
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Khushi Gupta
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Sumit Kumar Khandai
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India
| | - Sonia Malik
- Laboratory of Woody Plants and Crops Biology, University of Orleans, Orleans, France
| | - Suresh Thareja
- Department of Pharmaceutical Sciences and Natural Products, School of Pharmaceutical Sciences, Central University of Punjab, Bathinda, Punjab, 151401, India.
| |
Collapse
|
60
|
Lan HR, Chen M, Yao SY, Chen JX, Jin KT. Bispecific antibodies revolutionizing breast cancer treatment: a comprehensive overview. Front Immunol 2023; 14:1266450. [PMID: 38111570 PMCID: PMC10725925 DOI: 10.3389/fimmu.2023.1266450] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 11/16/2023] [Indexed: 12/20/2023] Open
Abstract
Breast cancer (BCa) is known as a complex and prevalent disease requiring the development of novel anticancer therapeutic approaches. Bispecific antibodies (BsAbs) have emerged as a favorable strategy for BCa treatment due to their unique ability to target two different antigens simultaneously. By targeting tumor-associated antigens (TAAs) on cancer cells, engaging immune effector cells, or blocking critical signaling pathways, BsAbs offer enhanced tumor specificity and immune system involvement, improving anti-cancer activity. Preclinical and clinical studies have demonstrated the potential of BsAbs in BCa. For example, BsAbs targeting human epidermal growth factor receptor 2 (HER2) have shown the ability to redirect immune cells to HER2-positive BCa cells, resulting in effective tumor cell killing. Moreover, targeting the PD-1/PD-L1 pathway by BsAbs has demonstrated promising outcomes in overcoming immunosuppression and enhancing immune-mediated tumor clearance. Combining BsAbs with existing therapeutic approaches, such as chemotherapy, targeted therapies, or immune checkpoint inhibitors (ICIs), has also revealed synergistic effects in preclinical models and early clinical trials, emphasizing the usefulness and potential of BsAbs in BCa treatment. This review summarizes the latest evidence about BsAbs in treating BCa and the challenges and opportunities of their use in BCa.
Collapse
Affiliation(s)
- Huan-Rong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, China
| | - Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Shi-Ya Yao
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| | - Jun-Xia Chen
- Department of Gynecology, Shaoxing People’s Hospital, Shaoxing, Zhejiang, China
| | - Ke-Tao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, Zhejiang, China
| |
Collapse
|
61
|
Wu J, Yang Q, Zhu Y, Xia T, Yi L, Wang J, Ren X. DNAJA1 promotes proliferation and metastasis of breast cancer by activating mutant P53/NF-κB pathway. Pathol Res Pract 2023; 252:154921. [PMID: 37977037 DOI: 10.1016/j.prp.2023.154921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/25/2023] [Accepted: 11/02/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE Breast cancer is one of the most common tumors with high malignancy and metastatic rate. DNAJA1 is closely related to tumor progress in several tumors. However, the role and mechanisms of DNAJA1 in the metastasis and proliferation of breast cancer are unknown. METHODS Immunohistochemistry and western blot were used to detect the protein expression genes. In vivo and vitro experiments were performed to evaluate the proliferation, invasive and metastatic abilities of breast cancer cells. RESULTS DNAJA1 was high expressed in 234 cases of breast cancer tissues and associated with metastasis, p53 expression and poor survival for patients. Knock down of DNAJA1 decreased the number of plate clone formation and the OD value of CCK8 assays in breast cancer cells. Depletion of DNAJA1 also in decreased the invasive abilities of breast cancer cells. In vivo, knock down DNAJA1 decreased the growth of subcutaneous tumor and lung metastatic nodes. Mechanically, DNAJA1 could bind with P53-R175H and reduced its degradation. Up regulation of DNAJA1 in mutant P53-R175H breast cancer cell promoted the nuclear translocation of p65, activated NF-κB pathway and enhanced the transcription of its downstream genes such as MMP9, CXCL10 et al. Blockade of NF-κB pathway effectively rescued the effects of DNAJA1 on proliferation and metastasis in breast cancer. CONCLUSION Our study reveals that DNAJA1 is up regulated in breast cancer and promotes breast cancer cells proliferation and metastasis via P53-R175H/NF-κB pathway. It might be a potential prognosis marker for the breast cancer patients.
Collapse
Affiliation(s)
- Jiao Wu
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Department of Pathology, Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China
| | - Qiao Yang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Department of Pathology, Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China
| | - Ye Zhu
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Department of gastroenterology, The People' Hospital of Leshan, Leshan 644000, Sichuan Province, People's Republic of China; Precision Pathology Diagnosis for Serious Diseases Key Laboratory of Luzhou, Luzhou 646000, Sichuan Province, People's Republic of China
| | - Tian Xia
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Precision Pathology Diagnosis for Serious Diseases Key Laboratory of Luzhou, Luzhou 646000, Sichuan Province, People's Republic of China
| | - Lizhi Yi
- Department of gastroenterology, The People' Hospital of Leshan, Leshan 644000, Sichuan Province, People's Republic of China
| | - Jianmei Wang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Department of Pathology, Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Precision Pathology Diagnosis for Serious Diseases Key Laboratory of Luzhou, Luzhou 646000, Sichuan Province, People's Republic of China.
| | - Xiaoli Ren
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Department of Pathology, Southwest Medical University, Luzhou 646000, Sichuan Province, People's Republic of China; Precision Pathology Diagnosis for Serious Diseases Key Laboratory of Luzhou, Luzhou 646000, Sichuan Province, People's Republic of China.
| |
Collapse
|
62
|
Wang S, Wu W, Lin X, Zhang KM, Wu Q, Luo M, Zhou J. Predictive and prognostic biomarkers of bone metastasis in breast cancer: current status and future directions. Cell Biosci 2023; 13:224. [PMID: 38041134 PMCID: PMC10693103 DOI: 10.1186/s13578-023-01171-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 11/10/2023] [Indexed: 12/03/2023] Open
Abstract
The most common site of metastasis in breast cancer is the bone, where the balance between osteoclast-mediated bone resorption and osteoblast-mediated bone formation is disrupted. This imbalance causes osteolytic bone metastasis in breast cancer, which leads to bone pain, pathological fractures, spinal cord compression, and other skeletal-related events (SREs). These complications reduce patients' quality of life significantly and have a profound impact on prognosis. In this review, we begin by providing a brief overview of the epidemiology of bone metastasis in breast cancer, including current diagnostic tools, treatment approaches, and existing challenges. Then, we will introduce the pathophysiology of breast cancer bone metastasis (BCBM) and the animal models involved in the study of BCBM. We then come to the focus of this paper: a discussion of several biomarkers that have the potential to provide predictive and prognostic value in the context of BCBM-some of which may be particularly compatible with more comprehensive liquid biopsies. Beyond that, we briefly explore the potential of new technologies such as single-cell sequencing and organoid models, which will improve our understanding of tumor heterogeneity and aid in the development of improved biomarkers. The emerging biomarkers discussed hold promise for future clinical application, aiding in the prevention of BCBM, improving the prognosis of patients, and guiding the implementation of personalized medicine.
Collapse
Affiliation(s)
- Shenkangle Wang
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Wenxin Wu
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | - Xixi Lin
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China
| | | | - QingLiang Wu
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China
- Hangzhou Ninth People's Hospital, Hangzhou, 310014, China
| | - Mingpeng Luo
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
- The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310014, China.
| | - Jichun Zhou
- Department of Surgical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, Zhejiang, China.
- Biomedical Research Center and Key Laboratory of Biotherapy of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
63
|
Zhao Y, Sheldon M, Sun Y, Ma L. New Insights into YAP/TAZ-TEAD-Mediated Gene Regulation and Biological Processes in Cancer. Cancers (Basel) 2023; 15:5497. [PMID: 38067201 PMCID: PMC10705714 DOI: 10.3390/cancers15235497] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/07/2023] [Accepted: 11/17/2023] [Indexed: 02/12/2024] Open
Abstract
The Hippo pathway is conserved across species. Key mammalian Hippo pathway kinases, including MST1/2 and LATS1/2, inhibit cellular growth by inactivating the TEAD coactivators, YAP, and TAZ. Extensive research has illuminated the roles of Hippo signaling in cancer, development, and regeneration. Notably, dysregulation of Hippo pathway components not only contributes to tumor growth and metastasis, but also renders tumors resistant to therapies. This review delves into recent research on YAP/TAZ-TEAD-mediated gene regulation and biological processes in cancer. We focus on several key areas: newly identified molecular patterns of YAP/TAZ activation, emerging mechanisms that contribute to metastasis and cancer therapy resistance, unexpected roles in tumor suppression, and advances in therapeutic strategies targeting this pathway. Moreover, we provide an updated view of YAP/TAZ's biological functions, discuss ongoing controversies, and offer perspectives on specific debated topics in this rapidly evolving field.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
| | - Marisela Sheldon
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
| | - Yutong Sun
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (Y.Z.); (M.S.)
- The University of Texas MD Anderson Cancer Center UTHealth Houston Graduate School of Biomedical Sciences, Houston, TX 77030, USA
| |
Collapse
|
64
|
Qi X, Shi Q, Xuhong J, Zhang Y, Jiang J. Pyrotinib-based therapeutic approaches for HER2-positive breast cancer: the time is now. Breast Cancer Res 2023; 25:113. [PMID: 37789330 PMCID: PMC10546716 DOI: 10.1186/s13058-023-01694-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 08/05/2023] [Indexed: 10/05/2023] Open
Abstract
Human epidermal growth factor receptor 2 (HER2)-positive breast cancer (BC) is a highly aggressive subtype associated with poor prognosis. The advent of HER2-targeted drugs, including monoclonal antibodies, tyrosine-kinase inhibitors (TKIs) and antibody-drug conjugates, has yielded improved prognosis for patients. Compared with widely used monoclonal antibodies, small-molecule TKIs have unique advantages including oral administration and favorable penetration of blood-brain barrier for brain metastatic BC, and reduced cardiotoxicity. Pyrotinib is an irreversible TKI of the pan-ErbB receptor, and has recently been shown to be clinically effective for the treatment of HER2-positive BC in metastatic and neoadjuvant settings. This review highlights the development on the application of pyrotinib-based therapeutic approaches in the clinical settings of HER2-positive BC.
Collapse
Affiliation(s)
- Xiaowei Qi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
| | - Qiyun Shi
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- The Eighth Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Juncheng Xuhong
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China
- Shigatse Branch, Xinqiao Hospital, Army Medical University, Shigatse, 857000, China
| | - Yi Zhang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| | - Jun Jiang
- Department of Breast and Thyroid Surgery, Southwest Hospital, Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
65
|
Zambelli A, Cazzaniga M, La Verde N, Munzone E, Antonazzo IC, Mantovani LG, Di Cosimo S, Mancuso A, Generali D, Cortesi PA. A cost-consequence analysis of adding pertuzumab to the neoadjuvant combination therapy in HER2-positive high-risk early breast cancer in Italy. Breast 2023; 71:113-121. [PMID: 37573652 PMCID: PMC10428118 DOI: 10.1016/j.breast.2023.08.005] [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: 04/29/2023] [Revised: 08/03/2023] [Accepted: 08/05/2023] [Indexed: 08/15/2023] Open
Abstract
INTRODUCTION Clinical trials confirmed the beneficial effects of adding pertuzumab (P) to the combination of trastuzumab-chemotherapy (TC) in the (neo)adjuvant setting of high-risk HER2-positive early breast cancer (HER2+BC). We evaluated the clinical, economic and societal impact of adding pertuzumab to neoadjuvant TC combination (TPC) in Italy. METHODS A cost-consequence analysis comparing TPC vs. TC was performed developing a cohort-based multi-state Markov model to estimate the clinical, societal and economic impact of the neoadjuvant therapy of TPC versus TC in HER2+BC at high-risk of recurrence. The model works on a cycle length of 1 month and 5-years-time horizon. Literature review-based data were used to populate the model. The following clinical and economic outcomes were estimated: cumulative incidence of loco-regional/distant recurrences, life of years and QALY and both direct and indirect costs (€). Finally, sensitivity analyses were performed. RESULTS TPC was associated with a 75,630 € saved of direct costs. Specifically, it was associated with an initial increase of treatment costs (+4.8%) followed by reduction of recurrence management cost (-20.4%). TPC was also associated with an indirect cost reduction of 1.40%, as well as decreased incidence of distant recurrence (-20.14%), days of work lost (-1.53%) and days lived with disability (-0.50%). Furthermore, TPC reported 10,47 QALY gained (+2.77%) compared to TC. The probability to achieve the pathological complete response (pCR) was the parameter that mostly affected the results in the sensitivity analysis. CONCLUSION Our findings suggested that TPC combination could be a cost-saving option in patients with HER2+BC at high-risk of recurrence.
Collapse
Affiliation(s)
- Alberto Zambelli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Marina Cazzaniga
- Phase 1 Research Centre, ASST-Monza (MB), 20900, Monza, Italy; School of Medicine and Surgery, University of Milano-Bicocca, 20900, Monza, Italy
| | - Nicla La Verde
- Department of Oncology, Sacco Hospital, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Elisabetta Munzone
- Division of Medical Senology, European Institute of Oncology, IRCCS, Milan, Italy
| | | | | | - Serena Di Cosimo
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Daniele Generali
- Department of Medicine, Surgery and Health Sciences, University of Trieste, 34127, Trieste, Italy; Multidisciplinary Unit of Breast Pathology and Translational Research, ASST of Cremona Hospital, 26100, Cremona, Italy
| | - Paolo Angelo Cortesi
- Research Centre on Public Health (CESP), University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
66
|
Muñoz JP, Pérez-Moreno P, Pérez Y, Calaf GM. The Role of MicroRNAs in Breast Cancer and the Challenges of Their Clinical Application. Diagnostics (Basel) 2023; 13:3072. [PMID: 37835815 PMCID: PMC10572677 DOI: 10.3390/diagnostics13193072] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/14/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
MicroRNAs (miRNAs) constitute a subclass of non-coding RNAs that exert substantial influence on gene-expression regulation. Their tightly controlled expression plays a pivotal role in various cellular processes, while their dysregulation has been implicated in numerous pathological conditions, including cancer. Among cancers affecting women, breast cancer (BC) is the most prevalent malignant tumor. Extensive investigations have demonstrated distinct expression patterns of miRNAs in normal and malignant breast cells. Consequently, these findings have prompted research efforts towards leveraging miRNAs as diagnostic tools and the development of therapeutic strategies. The aim of this review is to describe the role of miRNAs in BC. We discuss the identification of oncogenic, tumor suppressor and metastatic miRNAs among BC cells, and their impact on tumor progression. We describe the potential of miRNAs as diagnostic and prognostic biomarkers for BC, as well as their role as promising therapeutic targets. Finally, we evaluate the current use of artificial intelligence tools for miRNA analysis and the challenges faced by these new biomedical approaches in its clinical application. The insights presented in this review underscore the promising prospects of utilizing miRNAs as innovative diagnostic, prognostic, and therapeutic tools for the management of BC.
Collapse
Affiliation(s)
- Juan P. Muñoz
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| | - Pablo Pérez-Moreno
- Programa de Comunicación Celular en Cáncer, Facultad de Medicina Clínica Alemana, Universidad del Desarrollo, Santiago 7780272, Chile
| | - Yasmín Pérez
- Laboratorio de Bioquímica, Departamento de Química, Facultad de Ciencias, Universidad de Tarapacá, Arica 1000007, Chile
| | - Gloria M. Calaf
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica 1000000, Chile
| |
Collapse
|
67
|
Guijarro LG, Justo Bermejo FJ, Boaru DL, De Castro-Martinez P, De Leon-Oliva D, Fraile-Martínez O, Garcia-Montero C, Alvarez-Mon M, Toledo-Lobo MDV, Ortega MA. Is Insulin Receptor Substrate4 (IRS4) a Platform Involved in the Activation of Several Oncogenes? Cancers (Basel) 2023; 15:4651. [PMID: 37760618 PMCID: PMC10526421 DOI: 10.3390/cancers15184651] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 09/06/2023] [Accepted: 09/13/2023] [Indexed: 09/29/2023] Open
Abstract
The IRS (insulin receptor substrate) family of scaffold proteins includes insulin receptor substrate-4 (IRS4), which is expressed only in a few cell lines, including human kidney, brain, liver, and thymus and some cell lines. Its N-terminus carries a phosphotyrosine-binding (PTB) domain and a pleckstrin homology domain (PH), which distinguishes it as a member of this family. In this paper, we collected data about the molecular mechanisms that explain the relevance of IRS4 in the development of cancer and identify IRS4 differences that distinguish it from IRS1 and IRS2. Search engines and different databases, such as PubMed, UniProt, ENSEMBL and SCANSITE 4.0, were used. We used the name of the protein that it encodes "(IRS-4 or IRS4)", or the combination of these terms with the word "(cancer)" or "(human)", for searches. Terms related to specific tumor pathologies ("breast", "ovary", "colon", "lung", "lymphoma", etc.) were also used. Despite the lack of knowledge on IRS4, it has been reported that some cancers and benign tumors are characterized by high levels of IRS-4 expression. Specifically, the role of IRS-4 in different types of digestive tract neoplasms, gynecological tumors, lung cancers, melanomas, hematological tumors, and other less common types of cancers has been shown. IRS4 differs from IRS1 and IRS2 in that can activate several oncogenes that regulate the PI3K/Akt cascade, such as BRK and FER, which are characterized by tyrosine kinase-like activity without regulation via extracellular ligands. In addition, IRS4 can activate the CRKL oncogene, which is an adapter protein that regulates the MAP kinase cascade. Knowledge of the role played by IRS4 in cancers at the molecular level, specifically as a platform for oncogenes, may enable the identification and validation of new therapeutic targets.
Collapse
Affiliation(s)
- Luis G. Guijarro
- Unit of Biochemistry and Molecular Biology, Department of System Biology (CIBEREHD), University of Alcalá, 28801 Alcala de Henares, Spain
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (D.L.B.); (D.D.L.-O.); (O.F.-M.); (C.G.-M.); (M.A.-M.); (M.A.O.)
| | | | - Diego Liviu Boaru
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (D.L.B.); (D.D.L.-O.); (O.F.-M.); (C.G.-M.); (M.A.-M.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Patricia De Castro-Martinez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Diego De Leon-Oliva
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (D.L.B.); (D.D.L.-O.); (O.F.-M.); (C.G.-M.); (M.A.-M.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Oscar Fraile-Martínez
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (D.L.B.); (D.D.L.-O.); (O.F.-M.); (C.G.-M.); (M.A.-M.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Cielo Garcia-Montero
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (D.L.B.); (D.D.L.-O.); (O.F.-M.); (C.G.-M.); (M.A.-M.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Melchor Alvarez-Mon
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (D.L.B.); (D.D.L.-O.); (O.F.-M.); (C.G.-M.); (M.A.-M.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
- Immune System Diseases-Rheumatology, Oncology Service and Internal Medicine (CIBEREHD), University Hospital Príncipe de Asturias, 28806 Alcala de Henares, Spain
| | - María del Val Toledo-Lobo
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (D.L.B.); (D.D.L.-O.); (O.F.-M.); (C.G.-M.); (M.A.-M.); (M.A.O.)
- Department of Biomedicine and Biotechnology, University of Alcalá, 28801 Alcala de Henares, Spain;
| | - Miguel A. Ortega
- Ramón y Cajal Institute of Sanitary Research (IRYCIS), 28034 Madrid, Spain; (D.L.B.); (D.D.L.-O.); (O.F.-M.); (C.G.-M.); (M.A.-M.); (M.A.O.)
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, 28801 Alcala de Henares, Spain;
- Cancer Registry and Pathology Department, Principe de Asturias University Hospital, 28806 Alcala de Henares, Spain
| |
Collapse
|
68
|
Al-Asadi S, Mansour H, Ataimish AJ, Al-Kahachi R, Rampurawala J. MicroRNAs Regulate Tumorigenesis by Downregulating SOCS3 Expression: An In silico Approach. Bioinform Biol Insights 2023; 17:11779322231193535. [PMID: 37701630 PMCID: PMC10493049 DOI: 10.1177/11779322231193535] [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: 03/04/2023] [Accepted: 07/24/2023] [Indexed: 09/14/2023] Open
Abstract
Tumor microenvironment is characterized by the occurrence of significant changes due to disrupted signaling pathways that affect a broad spectrum of cellular activities such as proliferation, differentiation, signaling, invasiveness, migration, and apoptosis. Similarly, a downregulated suppressor of cytokine signaling 3 (SOCS3) promotes increased JAK/STAT function due to aberrant cytokine signaling, which results in increased cell proliferation, differentiation, and migration. Multiple carcinomas including breast cancer, prostate cancer, hepatocellular carcinoma, pancreatic cancer, and colorectal cancer involve the disruption of SOCS3 expression due to microRNA overexpression. MicroRNAs are small, conserved, and non-coding RNA molecules that regulate gene expression through post-transcriptional inhibition and mRNA destabilization. The aim of this study was to identify putative microRNAs that interact with SOCS3 and downregulate its expression. In this study, miRWalk, TargetScan, and miRDB were used to identify microRNAs that interact with SOCS3, whereas RNA22 was utilized to identify the binding sites of 238 significant microRNAs. The tertiary structures of shortlisted microRNAs and SOCS3 regions were predicted through MC Sym and RNAComposer, respectively. For molecular docking, HDOCK was used, which predicted 80 microRNA-messengerRNA complexes and the interactions of the top 5 shortlisted complexes were assessed. The complexes were shortlisted on the basis of least binding affinity score and maximum confidence score. This study identifies the interactions of known (miR-203a-5p) and novel (miR-6756-5p, miR-6732-5p, miR-1203, miR-6887-5p) microRNAs with SOCS3 regions due to their maximum interactions. Identifying the interactions of these microRNAs with SOCS3 will significantly advance the understanding of oncomiRs (miRNAs that are associated with cancer development) in tumor development due to their influence on SOCS3 expression. These insights will assist in future studies to understand the significance of miRNA-SOCS3-associated tumor development and progression.
Collapse
Affiliation(s)
- Sura Al-Asadi
- College of Health and Medical Techniques, Middle Technical University, Baghdad, Iraq
| | - Hiba Mansour
- College of Health and Medical Techniques, Middle Technical University, Baghdad, Iraq
| | | | - Rusul Al-Kahachi
- Department of Scholarships and Cultural Relationship, Republic of Iraq Ministry of Higher Education and Scientific Research, Baghdad, Iraq
| | | |
Collapse
|
69
|
Shakiba E, Bazi A, Ghasemi H, Eshaghi‐Gorji R, Mehdipour SA, Nikfar B, Rashidi M, Mirzaei S. Hesperidin suppressed metastasis, angiogenesis and tumour growth in Balb/c mice model of breast cancer. J Cell Mol Med 2023; 27:2756-2769. [PMID: 37581480 PMCID: PMC10494297 DOI: 10.1111/jcmm.17902] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/16/2023] Open
Abstract
Considering the unfavourable response of breast cancer (BC) to treatment, we assessed the therapeutic potential hesperidin in mice bearing 4T1 BC tumours. Anti-tumour effects were assessed by measuring pathologic complete response (pCR), survival analysis, immunohistochemistry for E-cadherin, VEGF, MMP9, MMP2 and Ki-67, serum measurement of IFNγ and IL-4, and gene expression analysis of CD105, VEGFa, VEGFR2 and COX2. Survival of tumour-bearing mice was the highest in mice receiving a combination of hesperidin and doxorubicin (Dox) (80%) compared to the normal saline (43%), hesperidin 5 (54%), 10 (55.5%), 10 (60.5%) and 40 (66%) mg/kg, and 10 mg/kg Dox-treated (73%) groups (p < 0.0001 for all). Compared to the normal saline group, there was a significant elevation in IFNγ level in the animals receiving 20 (p = 0.0026) and 40 (p < 0.001) mg/kg hesperidin, 10 mg/kg Dox (p < 0.001), and combined hesperidin (20 mg/kg) and Dox (10 mg/kg) (p < 0.001). A significant reduction in the gene expression of CD 105 (p = 0.0106), VEGFa (p < 0.0001), VEGFR2 (p < 0.0001), and Cox2 (p = 0.034) and a significant higher pCR score (p = 0.006) were noticed in mice treated with 10 mg/kg Dox + 20 mg/kg hesperidin compared to those treated with 10 mg/kg Dox alone. Immunohistochemical staining showed significant reductions in Ki-67 (p < 0.001) and VEGF (p < 0.001) and a significant elevation in E-cadherin (p = 0.005) in the 10 mg/kg Dox + 20 mg/kg treatment group than in 10 mg/kg Dox alone group. Hesperidin can be considered as a potentially suitable anti-cancer agent for BC that can synergize with other chemotherapeutics.
Collapse
Affiliation(s)
- Elham Shakiba
- Department of Biochemistry, Faculty of Biological Sciences, North Tehran BranchIslamic Azad UniversityTehranIran
| | - Ali Bazi
- Department of HematologyKerman University of Medical SciencesKermanIran
- Faculty of Allied Medical SciencesZabol University of Medical SciencesZabolIran
| | - Hamed Ghasemi
- Student Research CommitteeMazandaran University of Medical SciencesSariIran
| | - Reza Eshaghi‐Gorji
- Student Research CommitteeMazandaran University of Medical SciencesSariIran
| | | | - Banafsheh Nikfar
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars HospitalIran University of Medical SciencesTehranIran
| | - Mohsen Rashidi
- Department of PharmacologyMazandaran University of Medical SciencesSariIran
- The Health of Plant and Livestock Products Research CenterMazandaran University of Medical SciencesSariIran
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Science and Research BranchIslamic Azad UniversityTehranIran
| |
Collapse
|
70
|
Qi X, Qiao B, Song T, Huang D, Zhang H, Liu Y, Jin Q, Yang M, Liu D. Clinical utility of the pan-immune-inflammation value in breast cancer patients. Front Oncol 2023; 13:1223786. [PMID: 37711203 PMCID: PMC10499041 DOI: 10.3389/fonc.2023.1223786] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Background The newly discovered pan-immune-inflammation value (PIV) has been illustrated to have good prognostic value for cancer patient prognosis. However, the prognostic usefulness of PIV in breast cancer patients is unknown. As a result, to aid the clinic in providing a distinctive and trustworthy biomarker to better assess breast cancer patient's prognosis, we conducted this meta-analysis to investigate the relationship between PIV and the survival of breast cancer patients. Methods We conducted a systematic search of Pubmed, Embase, the Cochrane Library, and the CNKI databases to screen for eligible studies published up to April 2023. Outcomes included overall survival (OS), progression-free survival (PFS), and pathological complete response (pCR). The hazard ratio (HR) and the corresponding 95% confidence interval (CI) were used as the indicators. STATA 15.0 software was used to perform meta-analysis, sensitivity analysis, and publication bias analysis. Results A total of eight articles, involving 2953 patients, met the inclusion criteria and were included in this meta-analysis. The results showed that patients with higher PIV levels had a significantly shorter OS (HR: 2.045, 95% CI: 1.355-3.086, P = 0.001) and PFS (HR: 1.466, 95% CI: 1.163-1.848, P = 0.001). Besides, the PIV value was negatively correlated with the efficacy of neoadjuvant chemotherapy. Sensitivity analysis showed that the results of this study were reliable and stable. Conclusions PIV has a good prognostic value in breast cancer patients and is expected to be a prognostic biomarker for breast cancer.
Collapse
Affiliation(s)
- Xiaoyan Qi
- Department of Breast Surgery, Liaoning Cancer Hospital & Institution, Shenyang, Liaoning, China
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Boyang Qiao
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | - Tingting Song
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Dan Huang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Hui Zhang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yang Liu
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Qi Jin
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Ming Yang
- Department of Breast Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Delong Liu
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institution, Shenyang, Liaoning, China
| |
Collapse
|
71
|
Dai Y, Zhang X, Ou Y, Zou L, Zhang D, Yang Q, Qin Y, Du X, Li W, Yuan Z, Xiao Z, Wen Q. Anoikis resistance--protagonists of breast cancer cells survive and metastasize after ECM detachment. Cell Commun Signal 2023; 21:190. [PMID: 37537585 PMCID: PMC10399053 DOI: 10.1186/s12964-023-01183-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/04/2023] [Indexed: 08/05/2023] Open
Abstract
Breast cancer exhibits the highest global incidence among all tumor types. Regardless of the type of breast cancer, metastasis is a crucial cause of poor prognosis. Anoikis, a form of apoptosis initiated by cell detachment from the native environment, is an outside-in process commencing with the disruption of cytosolic connectors such as integrin-ECM and cadherin-cell. This disruption subsequently leads to intracellular cytoskeletal and signaling pathway alterations, ultimately activating caspases and initiating programmed cell death. Development of an anoikis-resistant phenotype is a critical initial step in tumor metastasis. Breast cancer employs a series of stromal alterations to suppress anoikis in cancer cells. Comprehensive investigation of anoikis resistance mechanisms can inform strategies for preventing and regressing metastatic breast cancer. The present review first outlines the physiological mechanisms of anoikis, elucidating the alterations in signaling pathways, cytoskeleton, and protein targets that transpire from the outside in upon adhesion loss in normal breast cells. The specific anoikis resistance mechanisms induced by pathological changes in various spatial structures during breast cancer development are also discussed. Additionally, the genetic loci of targets altered in the development of anoikis resistance in breast cancer, are summarized. Finally, the micro-RNAs and targeted drugs reported in the literature concerning anoikis are compiled, with keratocin being the most functionally comprehensive. Video Abstract.
Collapse
Affiliation(s)
- Yalan Dai
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Department of Oncology, Garze Tibetan Autonomous Prefecture People's Hospital, Kangding, China
| | - Xinyi Zhang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shenzhen, China
| | - Yingjun Ou
- Clinical Medicine School, Southwest Medicial Univercity, Luzhou, China
- Orthopaedics, Garze Tibetan Autonomous Prefecture People's Hospital, Kangding, China
| | - Linglin Zou
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Duoli Zhang
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Qingfan Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yi Qin
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiuju Du
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Li
- Southwest Medical University, Luzhou, China
| | | | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China.
| | - Qinglian Wen
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
72
|
Girithar HN, Staats Pires A, Ahn SB, Guillemin GJ, Gluch L, Heng B. Involvement of the kynurenine pathway in breast cancer: updates on clinical research and trials. Br J Cancer 2023; 129:185-203. [PMID: 37041200 PMCID: PMC10338682 DOI: 10.1038/s41416-023-02245-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 03/04/2023] [Accepted: 03/17/2023] [Indexed: 04/13/2023] Open
Abstract
Breast cancer (BrCa) is the leading cause of cancer incidence and mortality in women worldwide. While BrCa treatment has been shown to be highly successful if detected at an early stage, there are few effective strategies to treat metastatic tumours. Hence, metastasis remains the main cause in most of BrCa deaths, highlighting the need for new approaches in this group of patients. Immunotherapy has been gaining attention as a new treatment for BrCa metastasis and the kynurenine pathway (KP) has been suggested as one of the potential targets. The KP is the major biochemical pathway in tryptophan (TRP) metabolism, catabolising TRP to nicotinamide adenine dinucleotide (NAD+). The KP has been reported to be elevated under inflammatory conditions such as cancers and that its activity suppresses immune surveillance. Dysregulation of the KP has previously been reported implicated in BrCa. This review aims to discuss and provide an update on the current mechanisms involved in KP-mediated immune suppression and cancer growth. Furthermore, we also provide a summary on 58 studies about the involvement of the KP and BrCa and five clinical trials targeting KP enzymes and their outcome.
Collapse
Affiliation(s)
- Hemaasri-Neya Girithar
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Ananda Staats Pires
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Seong Beom Ahn
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Gilles J Guillemin
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia
| | - Laurence Gluch
- The Strathfield Breast Centre, Strathfield, NSW, Australia
| | - Benjamin Heng
- Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, Australia.
| |
Collapse
|
73
|
Shin WS, Oh SW, Park HN, Kim JH, Lee ST. Knockdown of PTK7 Reduces the Oncogenic Potential of Breast Cancer Cells by Impeding Receptor Tyrosine Kinase Signaling. Int J Mol Sci 2023; 24:12173. [PMID: 37569547 PMCID: PMC10418930 DOI: 10.3390/ijms241512173] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 07/26/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
Protein tyrosine kinase 7 (PTK7), a catalytically defective receptor tyrosine kinase (RTK), is often upregulated in various cancers. This study aimed to validate PTK7 as a target for breast cancer (BC) and investigate its oncogenic signaling mechanism. BC tissue analysis showed significantly elevated PTK7 mRNA levels, especially in refractory triple-negative breast cancer (TNBC) tissues, compared with normal controls. Similarly, BC cell lines exhibited increased PTK7 expression. Knockdown of PTK7 inhibited the proliferation of T-47D and MCF-7 hormone-receptor-positive BC cell-lines and of HCC1187, MDA-MB-231, MDA-MB-436, and MDA-MB-453 TNBC cells. PTK7 knockdown also inhibited the adhesion, migration, and invasion of MDA-MB-231, MDA-MB-436, and MDA-MB-453 cells, and reduced the phosphorylation levels of crucial oncogenic regulators including extracellular signal-regulated kinase (ERK), Akt, and focal adhesion kinase (FAK). Furthermore, PTK7 interacts with fibroblast growth factor receptor 1 (FGFR1) and epidermal growth factor receptor (EGFR) expressed in MDA-MB-231 cells. Knockdown of PTK7 decreased the growth-factor-induced phosphorylation of FGFR1 and EGFR in MDA-MB-231 cells, indicating its association with RTK activation. In conclusion, PTK7 plays a significant role in oncogenic signal transduction by enhancing FGFR1 and EGFR activation, influencing BC tumorigenesis and metastasis. Hence, PTK7 represents a potential candidate for targeted BC therapy, including TNBC.
Collapse
Affiliation(s)
| | | | | | | | - Seung-Taek Lee
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (W.-S.S.); (S.W.O.); (H.N.P.); (J.H.K.)
| |
Collapse
|
74
|
Tian Y, Dong J, Li L. Bridging Pyroptosis and Immunity: A Comprehensive Study of the Pyroptosis-Related Long Non-Coding RNA Signature in Breast Cancer. Life (Basel) 2023; 13:1599. [PMID: 37511974 PMCID: PMC10381440 DOI: 10.3390/life13071599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/14/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Breast cancer continuously poses serious clinical challenges to human health due to its intrinsic heterogenicity and evolving drug resistance. Recently, increasing evidence has shown that pyroptosis, known as a programmed and inflammatory form of cell death, participates in tumorigenesis, progression, and remodeling of the tumor immune microenvironment (TIME). However, a comprehensive insight into pyroptosis-related signatures for breast cancer remains elusive. The current study established a pyroptosis-related lncRNA signature using transcriptome data and corresponding clinical information from The Cancer Genome Atlas (TCGA). Pyroptosis-related gene clusters, the associated differential expression in breast cancer patients' subtypes, and the potential mechanisms were all discussed. This integrative analysis revealed a unique signature underpinning the dichotomy of breast cancer progression and survival outcomes. Interestingly, the pyroptosis-related lncRNA signature was revealed as closely intertwined with the TIME. A correlation was established between the pyroptosis-related LncRNA signature and the TIME, underlying the mutual effect between pyroptosis and the immune responses implicated in breast cancer. The findings in this work underline the critical role exerted by pyroptosis in breast cancer, providing new insights into disease progression, prognosis, and therapeutic potential. This work has been poised to provide new avenues for personalized, immune-based cancer therapeutics by enhancing our understanding of pyroptosis in breast cancer.
Collapse
Affiliation(s)
- Ye Tian
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang 110866, China
| | - Jing Dong
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang 110866, China
| | - Lin Li
- College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang 110866, China
- Key Laboratory of Livestock Infectious Diseases, Ministry of Education, Shenyang Agricultural University, Shenyang 110866, China
| |
Collapse
|
75
|
Jia T, Lv Q, Zhang B, Yu C, Sang S, Deng S. Assessment of androgen receptor expression in breast cancer patients using 18 F-FDG PET/CT radiomics and clinicopathological characteristics. BMC Med Imaging 2023; 23:93. [PMID: 37460990 PMCID: PMC10353086 DOI: 10.1186/s12880-023-01052-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/30/2023] [Indexed: 07/20/2023] Open
Abstract
OBJECTIVE In the present study, we mainly aimed to predict the expression of androgen receptor (AR) in breast cancer (BC) patients by combing radiomic features and clinicopathological factors in a non-invasive machine learning way. MATERIALS AND METHODS A total of 48 BC patients, who were initially diagnosed by 18F-FDG PET/CT, were retrospectively enrolled in this study. LIFEx software was used to extract radiomic features based on PET and CT data. The most useful predictive features were selected by the LASSO (least absolute shrinkage and selection operator) regression and t-test. Radiomic signatures and clinicopathologic characteristics were incorporated to develop a prediction model using multivariable logistic regression analysis. The receiver operating characteristic (ROC) curve, Hosmer-Lemeshow (H-L) test, and decision curve analysis (DCA) were conducted to assess the predictive efficiency of the model. RESULTS In the univariate analysis, the metabolic tumor volume (MTV) was significantly correlated with the expression of AR in BC patients (p < 0.05). However, there only existed feeble correlations between estrogen receptor (ER), progesterone receptor (PR), and AR status (p = 0.127, p = 0.061, respectively). Based on the binary logistic regression method, MTV, SHAPE_SphericityCT (CT Sphericity from SHAPE), and GLCM_ContrastCT (CT Contrast from grey-level co-occurrence matrix) were included in the prediction model for AR expression. Among them, GLCM_ContrastCT was an independent predictor of AR status (OR = 9.00, p = 0.018). The area under the curve (AUC) of ROC in this model was 0.832. The p-value of the H-L test was beyond 0.05. CONCLUSIONS A prediction model combining radiomic features and clinicopathological characteristics could be a promising approach to predict the expression of AR and noninvasively screen the BC patients who could benefit from anti-AR regimens.
Collapse
Affiliation(s)
- Tongtong Jia
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Qingfu Lv
- Department of General Surgery, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Bin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Chunjing Yu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University, Wuxi, 214122, China.
| | - Shibiao Sang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Shengming Deng
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| |
Collapse
|
76
|
Li J, Zhang W, Ma X, Wei Y, Zhou F, Li J, Zhang C, Yang Z. Cuproptosis/ferroptosis-related gene signature is correlated with immune infiltration and predict the prognosis for patients with breast cancer. Front Pharmacol 2023; 14:1192434. [PMID: 37521466 PMCID: PMC10374203 DOI: 10.3389/fphar.2023.1192434] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/19/2023] [Indexed: 08/01/2023] Open
Abstract
Background: Breast invasive carcinoma (BRCA) is a malignant tumor with high morbidity and mortality, and the prognosis is still unsatisfactory. Both ferroptosis and cuproptosis are apoptosis-independent cell deaths caused by the imbalance of corresponding metal components in cells and can affect the proliferation rate of cancer cells. The aim in this study was to develop a prognostic model of cuproptosis/ferroptosis-related genes (CFRGs) to predict survival in BRCA patients. Methods: Transcriptomic and clinical data for breast cancer patients were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Cuproptosis and ferroptosis scores were determined for the BRCA samples from the TCGA cohort using Gene Set Variation Analysis (GSVA), followed by weighted gene coexpression network analysis (WGCNA) to screen out the CFRGs. The intersection of the differentially expressed genes grouped by high and low was determined using X-tile. Univariate Cox regression and least absolute shrinkage and selection operator (LASSO) were used in the TGCA cohort to identify the CFRG-related signature. In addition, the relationship between risk scores and immune infiltration levels was investigated using various algorithms, and model genes were analyzed in terms of single-cell sequencing. Finally, the expression of the signature genes was validated with quantitative real-time PCR (qRT‒PCR) and immunohistochemistry (IHC). Results: A total of 5 CFRGs (ANKRD52, HOXC10, KNOP1, SGPP1, TRIM45) were identified and were used to construct proportional hazards regression models. The high-risk groups in the training and validation sets had significantly worse survival rates. Tumor mutational burden (TMB) was positively correlated with the risk score. Conversely, Tumor Immune Dysfunction and Exclusion (TIDE) and tumor purity were inversely associated with risk scores. In addition, the infiltration degree of antitumor immune cells and the expression of immune checkpoints were lower in the high-risk group. In addition, risk scores and mTOR, Hif-1, ErbB, MAPK, PI3K/AKT, TGF-β and other pathway signals were correlated with progression. Conclusion: We can accurately predict the survival of patients through the constructed CFRG-related prognostic model. In addition, we can also predict patient immunotherapy and immune cell infiltration.
Collapse
Affiliation(s)
- Jixian Li
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Wentao Zhang
- Shandong Provincial Hospital, Shandong First Medical University, Jinan, Shandong, China
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaoqing Ma
- Radiotherapy and Minimally Invasive Group I, The Second Affiliated Hospital of Shandong First Medical University, Taian, Shandong, China
| | - Yanjun Wei
- Department of Radiation Oncology, Weifang People’s Hospital, Weifang, China
| | - Fengge Zhou
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jianan Li
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Chenggui Zhang
- Department of Orthopedics, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Zhe Yang
- Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
77
|
Lu X, Yuan Q, Zhang C, Wang S, Wei W. Predicting the immune microenvironment and prognosis with a anoikis - related signature in breast cancer. Front Oncol 2023; 13:1149193. [PMID: 37469408 PMCID: PMC10353543 DOI: 10.3389/fonc.2023.1149193] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/15/2023] [Indexed: 07/21/2023] Open
Abstract
Background Tumor heterogeneity is widely recognized as a crucial factor impacting the prognosis of breast cancer (BC) patients. However, there remains an insufficient understanding of the underlying impact of anoikis on the prognosis of BC patients. Methods The researchers utilized the TCGA-BRCA dataset to screen and analyze the differentially expressed genes of anoikis-related genes (ARGs) in BC and normal breast tissue. Prognostic gene signatures were established through univariate, LASSO, and multivariate Cox regression analyses. These signatures were evaluated using Kaplan-Meier curve and receiver operating characteristic (ROC) analyses, resulting in the development of an anoikis-related index (ACI). The training dataset was TCGA-BRCA, while METABRIC and GSE96058 were used for external validation. Additionally, nomograms were developed by combining risk scores and clinical parameters, enabling gene set enrichment analysis (GSEA) and tumor immunoassay. Furthermore, an exploration of small molecule compounds was conducted to identify potential therapeutic benefits. Results A six-gene anoikis-related signature was constructed, which divided BC patients into high- and low-ACI groups based on median ACI scores. The ACI accurately predicted prognosis and acted as an independent prognostic factor for BC patients. Patients in the high-ACI group exhibited poorer overall survival (OS) across all cohorts and showed more severe clinical manifestations compared to the low-ACI group. The study also explored the potential impacts of anoikis on immune cells infiltrating tumors, immune checkpoints, growth factors, and cytokine levels. Additionally, the potential implications of anoikis in BC immunotherapy were discussed, along with highlighting small molecule compounds that could offer therapeutic benefits. Conclusions Anoikis was found to hold significant prognostic value in breast cancer, providing a novel approach for managing patients with different prognoses and implementing more precise immunotherapy strategies.
Collapse
Affiliation(s)
- Xiuqing Lu
- Department of Breast Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qi Yuan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Chao Zhang
- Department of Breast Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Sifen Wang
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Weidong Wei
- Department of Breast Surgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
78
|
Kadi MS, Alhebshi AH, Shabkah AA, Alzahrani WA, Enani GN, Samkari AA, Iskanderani O, Saleem AM, Farsi AH, Trabulsi NH. Histopathological Patterns and Outcomes of Triple-Positive Versus Triple-Negative Breast Cancer: A Retrospective Study at a Tertiary Cancer Center. Cureus 2023; 15:e42389. [PMID: 37621828 PMCID: PMC10446888 DOI: 10.7759/cureus.42389] [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: 07/24/2023] [Indexed: 08/26/2023] Open
Abstract
Background One of the leading causes of cancer-related deaths in females under 45 years old is breast cancer (BC). The definition of triple-negative breast cancer (TNBC) is the lack of expression of estrogen receptors (ERs) as well as progesterone receptors (PRs) and Erb-B2 receptor tyrosine kinase 2 (HER2) gene amplification. Triple-positive breast cancer (TPBC), on the other hand, is defined as tumors expressing a high level of ER, PR, and HER2 receptors. This study aims to assess the phenotypes of TNBC and TPBC by comparing their individual clinical behavior patterns and prognosis throughout the course of the disease in a tertiary cancer center in the Kingdom of Saudi Arabia (KSA). Methods Our study is a retrospective study using electronic medical records (EMRs) to identify all female patients diagnosed with BC using the International Classification of Diseases-10 (ICD-10) codes (between C50 and C50.9). About 1209 cases with primary BC female patients were recognized based on histopathology reports. Further subclassification into TPBC and TNBC was performed. Statistical analysis was performed using Rv3.6.2 (R Studio, version 3.5.2, Boston, MA, USA). The descriptive data were presented as means and standard deviations (SD). Survival curves were approximated using the Kaplan-Meier method. The comparison between survival curves between both groups was achieved using the log-rank test. The multivariate model was constructed based on the identified predictors using univariate analysis. Results Univariate analysis of overall survival (OS) showed that mortality was higher in TNBC compared to TPBC (HR = 2.82, P-value <0.05). However, in a multivariate analysis, molecular subtypes did not show a significant effect on OS with a P-value of 0.94. We found that age at diagnosis has been associated with a 4% increase in mortality risk with a yearly rise in age. Conclusion In this limited retrospective cohort study, we found that TNBC may not be associated with a higher risk of death than TPBC. However, other factors, including age at diagnosis, surgical intervention, and lymphovascular invasion (LVI), have been observed to increase the risk of mortality. On the other hand, patients with TNBC were found to have a worse prognosis in terms of local recurrence. This information cannot be generalized to all patients with BC given the limitations of this study. Further, larger cohorts are needed to explore biological and treatment-related outcomes in patients with TNBC and TPBC.
Collapse
Affiliation(s)
- Mai S Kadi
- Department of Community Medicine, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | | | - Alaa A Shabkah
- Department of Surgery, International Medical Center, Jeddah, SAU
| | - Walaa A Alzahrani
- Department of Pediatrics, King Faisal Specialist Hospital and Research Center, Jeddah, SAU
| | - Ghada N Enani
- Department of Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Ali A Samkari
- Department of Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Omar Iskanderani
- Department of Radiology, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Abdulaziz M Saleem
- Department of Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Ali H Farsi
- Department of Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| | - Nora H Trabulsi
- Department of Surgery, Faculty of Medicine, King Abdulaziz University, Jeddah, SAU
| |
Collapse
|
79
|
Cai GX, Kong WY, Liu Y, Zhong SY, Liu Q, Deng YF, Ye GL. Nuclear transport maintenance of USP22-AR by Importin-7 promotes breast cancer progression. Cell Death Discov 2023; 9:211. [PMID: 37391429 DOI: 10.1038/s41420-023-01525-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 06/11/2023] [Accepted: 06/22/2023] [Indexed: 07/02/2023] Open
Abstract
The translocation of biological macromolecules between cytoplasm and nucleus is of great significance to maintain various life processes in both normal and cancer cells. Disturbance of transport function likely leads to an unbalanced state between tumor suppressors and tumor-promoting factors. In this study, based on the unbiased analysis of protein expression differences with a mass spectrometer between human breast malignant tumors and benign hyperplastic tissues, we identified that Importin-7, a nuclear transport factor, is highly expressed in breast cancer (BC) and predicts poor outcomes. Further studies showed that Importin-7 promotes cell cycle progression and proliferation. Mechanistically, through co-immunoprecipitation, immunofluorescence, and nuclear-cytoplasmic protein separation experiments, we discovered that AR and USP22 can bind to Importin-7 as cargoes to promote BC progression. In addition, this study provides a rationale for a therapeutic strategy to restream the malignant progression of AR-positive BC by inhibiting the high expression state of Importin-7. Moreover, the knockdown of Importin-7 increased the responsiveness of BC cells to the AR signaling inhibitor, enzalutamide, suggesting that targeting Importin-7 may be a potential therapeutic strategy.
Collapse
Affiliation(s)
- Geng-Xi Cai
- Department of Breast Surgery, The First People's Hospital of Foshan, 528000, Foshan, Guangdong, China
| | - Wei-Yao Kong
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, 511436, Guangzhou, Guangdong, China
| | - Yuan Liu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Medical University, 511436, Guangzhou, Guangdong, China
| | - Shu-Yi Zhong
- Guangzhou Medical University-Guangzhou Institute of Biomedicine and Health (GMU-GIBH) Joint School of Life Sciences, Guangzhou Medical University, 511436, Guangzhou, Guangdong, China
| | - Qing Liu
- Department of Pathology, The First People's Hospital of Foshan, 528000, Foshan, Guangdong, China
| | - Yuan-Fei Deng
- Department of Pathology, The First People's Hospital of Foshan, 528000, Foshan, Guangdong, China.
| | - Guo-Lin Ye
- Department of Breast Surgery, The First People's Hospital of Foshan, 528000, Foshan, Guangdong, China.
| |
Collapse
|
80
|
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
|
81
|
Dave A, Charytonowicz D, Francoeur NJ, Beaumont M, Beaumont K, Schmidt H, Zeleke T, Silva J, Sebra R. The Breast Cancer Single-Cell Atlas: Defining cellular heterogeneity within model cell lines and primary tumors to inform disease subtype, stemness, and treatment options. Cell Oncol (Dordr) 2023; 46:603-628. [PMID: 36598637 PMCID: PMC10205851 DOI: 10.1007/s13402-022-00765-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/13/2022] [Indexed: 01/05/2023] Open
Abstract
PURPOSE Breast Cancer (BC) is the most diagnosed cancer in women; however, through significant research, relative survival rates have significantly improved. Despite progress, there remains a gap in our understanding of BC subtypes and personalized treatments. This manuscript characterized cellular heterogeneity in BC cell lines through scRNAseq to resolve variability in subtyping, disease modeling potential, and therapeutic targeting predictions. METHODS We generated a Breast Cancer Single-Cell Cell Line Atlas (BSCLA) to help inform future BC research. We sequenced over 36,195 cells composed of 13 cell lines spanning the spectrum of clinical BC subtypes and leveraged publicly available data comprising 39,214 cells from 26 primary tumors. RESULTS Unsupervised clustering identified 49 subpopulations within the cell line dataset. We resolve ambiguity in subtype annotation comparing expression of Estrogen Receptor, Progesterone Receptor, and Human Epidermal Growth Factor Receptor 2 genes. Gene correlations with disease subtype highlighted S100A7 and MUCL1 overexpression in HER2 + cells as possible cell motility and localization drivers. We also present genes driving populational drifts to generate novel gene vectors characterizing each subpopulation. A global Cancer Stem Cell (CSC) scoring vector was used to identify stemness potential for subpopulations and model multi-potency. Finally, we overlay the BSCLA dataset with FDA-approved targets to identify to predict the efficacy of subpopulation-specific therapies. CONCLUSION The BSCLA defines the heterogeneity within BC cell lines, enhancing our overall understanding of BC cellular diversity to guide future BC research, including model cell line selection, unintended sample source effects, stemness factors between cell lines, and cell type-specific treatment response.
Collapse
Affiliation(s)
- Arpit Dave
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave - Icahn (East) Building, Floor 14, Room 14-20E, New York, NY 10029 USA
| | - Daniel Charytonowicz
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave - Icahn (East) Building, Floor 14, Room 14-20E, New York, NY 10029 USA
| | - Nancy J. Francoeur
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave - Icahn (East) Building, Floor 14, Room 14-20E, New York, NY 10029 USA
- Pacific Biosciences, CA Menlo Park, USA
| | - Michael Beaumont
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave - Icahn (East) Building, Floor 14, Room 14-20E, New York, NY 10029 USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - Kristin Beaumont
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave - Icahn (East) Building, Floor 14, Room 14-20E, New York, NY 10029 USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | | | - Tizita Zeleke
- Department of Pathology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY 10029 USA
| | - Jose Silva
- Department of Pathology, Icahn School of Medicine at Mount Sinai Hospital, New York, NY 10029 USA
| | - Robert Sebra
- Department of Genetics & Genomic Sciences, Icahn School of Medicine at Mount Sinai, 1425 Madison Ave - Icahn (East) Building, Floor 14, Room 14-20E, New York, NY 10029 USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
- Center for Advanced Genomics Technology, Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| |
Collapse
|
82
|
Tang L, Lei X, Hu H, Li Z, Zhu H, Zhan W, Zhang T. Investigation of fatty acid metabolism-related genes in breast cancer: Implications for Immunotherapy and clinical significance. Transl Oncol 2023; 34:101700. [PMID: 37247503 DOI: 10.1016/j.tranon.2023.101700] [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: 05/09/2023] [Accepted: 05/21/2023] [Indexed: 05/31/2023] Open
Abstract
Breast cancer (BRCA) is a major global health issue, characterized by high mortality and low early diagnosis rates. The tumor immune microenvironment (TME) of BRCA is closely linked to fatty acid metabolism (FAM). This study aimed to identify FAM-related subtypes in BRCA based on gene expression and clinical data from the Cancer Genome Atlas (TCGA) database. The study found two distinct FAM-related subtypes, each with unique immune characteristics and prognostic implications. A FAM-related risk score prognostic model was developed and validated using TCGA and International Cancer Genome Consortium (GEO) cohorts, showing potential clinical applications for chemotherapy and immunotherapy. Additionally, a nomogram was established to facilitate clinical use of the risk score. These results highlight the significant correlation between FAM genes and TME in BRCA, and demonstrate the potential clinical utility of the FAM-related risk score in informing treatment decisions for BRCA patients.
Collapse
Affiliation(s)
- Liyang Tang
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, China; The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Chinese Traditional Medicine(TCM) research platform of major Epidemic Treatment base, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Xiaoyong Lei
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, China
| | - Haihong Hu
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, China; The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Chinese Traditional Medicine(TCM) research platform of major Epidemic Treatment base, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Zhuo Li
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, China; The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Chinese Traditional Medicine(TCM) research platform of major Epidemic Treatment base, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Hongxia Zhu
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, China; The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Chinese Traditional Medicine(TCM) research platform of major Epidemic Treatment base, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Wendi Zhan
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, China; The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Chinese Traditional Medicine(TCM) research platform of major Epidemic Treatment base, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China
| | - Taolan Zhang
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, Hunan 421001, China; The First Affiliated Hospital, Department of Pharmacy, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China; The First Affiliated Hospital, Chinese Traditional Medicine(TCM) research platform of major Epidemic Treatment base, Hengyang Medical School, University of South China, 69 Chuanshan Road, Hengyang, Hunan, 421001, China.
| |
Collapse
|
83
|
Huang Y, Peng H, Zeng A, Song L. The role of peptides in reversing chemoresistance of breast cancer: current facts and future prospects. Front Pharmacol 2023; 14:1188477. [PMID: 37284316 PMCID: PMC10239817 DOI: 10.3389/fphar.2023.1188477] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/10/2023] [Indexed: 06/08/2023] Open
Abstract
Breast cancer is the first malignant tumor in women, and its incidence is also increasing year by year. Chemotherapy is one of the standard therapies for breast cancer, but the resistance of breast cancer cells to chemotherapy drugs is a huge challenge for the effective treatment of breast cancer. At present, in the study of reversing the drug resistance of solid tumors such as breast cancer, peptides have the advantages of high selectivity, high tissue penetration, and good biocompatibility. Some of the peptides that have been studied can overcome the resistance of tumor cells to chemotherapeutic drugs in the experiment, and effectively control the growth and metastasis of breast cancer cells. Here, we describe the mechanism of different peptides in reversing breast cancer resistance, including promoting cancer cell apoptosis; promoting non-apoptotic regulatory cell death of cancer cells; inhibiting the DNA repair mechanism of cancer cells; improving the tumor microenvironment; inhibiting drug efflux mechanism; and enhancing drug uptake. This review focuses on the different mechanisms of peptides in reversing breast cancer drug resistance, and these peptides are also expected to create clinical breakthroughs in promoting the therapeutic effect of chemotherapy drugs in breast cancer patients and improving the survival rate of patients.
Collapse
Affiliation(s)
- Yongxiu Huang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongyao Peng
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Anqi Zeng
- Institute of Translational Pharmacology and Clinical Application, Sichuan Academy of Chinese Medical Science, Chengdu, Sichuan, China
| | - Linjiang Song
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
84
|
Yang C, Wang M, Huang R, Ou L, Li M, Wu W, Lei R. Circ_0108942 Regulates the Progression of Breast Cancer by Regulating the MiR-1178-3p/TMED3 Axis. Clin Breast Cancer 2023; 23:291-301. [PMID: 36764873 DOI: 10.1016/j.clbc.2022.12.014] [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: 04/23/2022] [Revised: 09/21/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND Breast cancer (BC) has posed a fatal threat to women's lives and the search for new methods of diagnosis and treatment is an important way to break the bottleneck of high mortality in BC. Circular RNAs (circRNAs) have been confirmed to be aberrantly expressed in several types of cancers, and this study is intended to elucidate the role and mechanism of circ_0108942 in BC. MATERIALS AND METHODS The levels of circ_0108942, microRNA-1178-3p (miR-1178-3p), and transmembrane p24 trafficking protein 3 (TMED3) were measured using real-time quantitative polymerase chain reaction (RT-qPCR) or western blot. Meanwhile, the cell proliferation, migration, invasion, angiopoiesis, and apoptosis were analyzed using 5-ethynyl-2'-deoxyuridine (EdU), transwell, tubule formation, and flow cytometry assays. Protein levels were determined by western blot. In addition, we used dual-luciferase reporter and RNA pull-down assays to identify the interplay between miR-1178-3p and circ_0108942 or TMED3. Lastly, the impact of circ_0108942 on the growth of BC tumors in vivo was analyzed by xenograft models. RESULTS Circ_0108942 and TMED3 were notably upregulated in BC, and the miR-1178-3p was downregulated. Functionally, silencing circ_0108942 suppressed cell proliferation, migration, invasion and promoted apoptosis in BC cells. In mechanism, circ_0108942 regulated TMED3 expression by sponging miR-1178-3p. Meanwhile, circ_0108942 knockdown also greatly constrained tumor growth in vivo. CONCLUSION Circ_0108942 boosted BC progression by regulating miR-1178-3p and thus upregulating TMED3.
Collapse
Affiliation(s)
- Chuansheng Yang
- Department of Head-Neck and Breast Surgery, Yuebei People's Hospital of Shantou University, Shaoguan, Guangdong, China
| | - Meijiao Wang
- Operation room, Yuebei People's Hospital of Shantou University, Shaoguan, Guangdong, China
| | - Renfeng Huang
- Department of Head-Neck and Breast Surgery, Yuebei People's Hospital of Shantou University, Shaoguan, Guangdong, China
| | - Linyang Ou
- Department of Head-Neck and Breast Surgery, Yuebei People's Hospital of Shantou University, Shaoguan, Guangdong, China
| | - Min Li
- Department of Head-Neck and Breast Surgery, Yuebei People's Hospital of Shantou University, Shaoguan, Guangdong, China
| | - Wanming Wu
- Department of Head-Neck and Breast Surgery, Yuebei People's Hospital of Shantou University, Shaoguan, Guangdong, China
| | - Ruiwen Lei
- Department of Head-Neck and Breast Surgery, Yuebei People's Hospital of Shantou University, Shaoguan, Guangdong, China.
| |
Collapse
|
85
|
Kątnik E, Gomułkiewicz A, Piotrowska A, Grzegrzółka J, Rusak A, Kmiecik A, Ratajczak-Wielgomas K, Dzięgiel P. BCL11A Expression in Breast Cancer. Curr Issues Mol Biol 2023; 45:2681-2698. [PMID: 37185699 PMCID: PMC10137054 DOI: 10.3390/cimb45040175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/20/2023] [Accepted: 03/21/2023] [Indexed: 05/17/2023] Open
Abstract
B-cell leukemia/lymphoma 11A (BCL11A) is a transcription factor that regulates the expression of genes involved in cell division or apoptosis. A link between high BCL11A expression and a worse prognosis has been demonstrated in patients with various cancers. The aim of this study was to investigate the expression pattern of BCL11A in breast cancer (BC) cases and mastopathy samples and to correlate the results with the clinicopathological data. The expression of the BCL11A protein was investigated using immunohistochemistry (IHC) on 200 cases of BC and 13 mastopathy samples. The level of BCL11A mRNA was determined using real-time PCR in 22 cases of BC and 6 mastopathy samples. The expression of BCL11A was also examined at the protein and mRNA levels in BC cell lines. A higher expression level of BCL11A in BC cases was shown compared to mastopathy samples. The expression level of BCL11A in BC cases and in the studied cell lines decreased with the increasing grade of histological malignancy (G). It was also negatively correlated with the primary tumor size. A significantly lower expression of BCL11A was found in BC that did not express estrogen or progesterone receptors and in triple-negative cases. The results of our research suggest that BCL11A may be relevant in the development of BC.
Collapse
Affiliation(s)
- Ewa Kątnik
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Agnieszka Gomułkiewicz
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Aleksandra Piotrowska
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Jędrzej Grzegrzółka
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Agnieszka Rusak
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Alicja Kmiecik
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Katarzyna Ratajczak-Wielgomas
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Piotr Dzięgiel
- Division of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, 50-368 Wroclaw, Poland
| |
Collapse
|
86
|
Albi E, Mandarano M, Cataldi S, Ceccarini MR, Fiorani F, Beccari T, Sidoni A, Codini M. The Effect of Cholesterol in MCF7 Human Breast Cancer Cells. Int J Mol Sci 2023; 24:ijms24065935. [PMID: 36983016 PMCID: PMC10052157 DOI: 10.3390/ijms24065935] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 03/14/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
In the last decade, cholesterol level has been implicated in several types of cancer, including breast cancer. In the current study, we aimed to investigate the condition of lipid depletion, hypocholesterolemia or hypercholesterolemia reproduced in vitro to analyze the response of different human breast cancer cells. Thus, MCF7 as the luminal A model, MB453 as the HER2 model and MB231 as the triple-negative model were used. No effect on cell growth and viability was detected in MB453 and MB231 cells. In MCF7 cells, hypocholesterolemia (1) reduced cell growth, and Ki67 expression; (2) increased ER/PgR expression; (3) stimulated the 3-Hydroxy-3-Methylglutaryl-CoA reductase and neutral sphingomyelinase and; (4) stimulated the expression of CDKN1A gene coding cyclin-dependent kinase inhibitor 1A protein, GADD45A coding growth arrest and DNA-damage-inducible alpha protein and, PTEN gene coding phosphatase and tensin homolog. All these effects were exacerbated by the lipid-depleted condition and reversed by the hypercholesterolemic condition. The relationship between cholesterol level and sphingomyelin metabolism was demonstrated. In summary, our data suggest that cholesterol levels should be controlled in luminal A breast cancer.
Collapse
Affiliation(s)
- Elisabetta Albi
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| | - Martina Mandarano
- Division of Pathological Anatomy and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, 06126 Perugia, Italy
| | - Samuela Cataldi
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| | | | - Federico Fiorani
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| | - Tommaso Beccari
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| | - Angelo Sidoni
- Division of Pathological Anatomy and Histology, Department of Experimental Medicine, School of Medicine and Surgery, University of Perugia, 06126 Perugia, Italy
| | - Michela Codini
- Department of Pharmaceutical Sciences, University of Perugia, 06126 Perugia, Italy
| |
Collapse
|
87
|
CD24-associated ceRNA network reveals prognostic biomarkers in breast carcinoma. Sci Rep 2023; 13:3826. [PMID: 36882451 PMCID: PMC9992383 DOI: 10.1038/s41598-022-25072-x] [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: 05/08/2022] [Accepted: 11/24/2022] [Indexed: 03/09/2023] Open
Abstract
Breast cancer is one of the most common cancer types which is described as the leading cause of cancer death in women. After competitive endogenous RNA (ceRNA) hypothesis was proposed, this triple regulatory network has been observed in various cancers, and increasing evidences reveal that ceRNA network plays a significant role in the migration, invasion, proliferation of cancer cells. In the current study, our target is to construct a CD24-associated ceRNA network, and to further identify key prognostic biomarkers in breast cancer. Using the transcriptom profiles from TCGA database, we performed a comprehensive analysis between CD24high tumor samples and CD24low tumor samples, and identified 132 DElncRNAs, 602 DEmRNAs and 26 DEmiRNAs. Through comprehensive analysis, RP1-228H13.5/miR-135a-5p/BEND3 and SIM2 were identified as key CD24-associated biomarkers, which exhibited highly significance with overall survival, immune microenvironment as well as clinical features. To sum up the above, the current study constructed a CD24-associated ceRNA network, and RP1-228H13.5/miR-135a-5p/BEND3 and SIM2 axis worked as a potential therapeutic target and a predictor for BRCA diagnosis and prognosis.
Collapse
|
88
|
Soghli N, Yousefi H, Naderi T, Fallah A, Moshksar A, Darbeheshti F, Vittori C, Delavar MR, Zare A, Rad HS, Kazemi A, Bitaraf A, Hussen BM, Taheri M, Jamali E. NRF2 signaling pathway: A comprehensive prognostic and gene expression profile analysis in breast cancer. Pathol Res Pract 2023; 243:154341. [PMID: 36739754 DOI: 10.1016/j.prp.2023.154341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 01/23/2023] [Accepted: 01/23/2023] [Indexed: 01/27/2023]
Abstract
Breast cancer is the most frequently diagnosed malignant tumor in women and a major public health concern. NRF2 axis is a cellular protector signaling pathway protecting both normal and cancer cells from oxidative damage. NRF2 is a transcription factor that binds to the gene promoters containing antioxidant response element-like sequences. In this report, differential expression of NRF2 signaling pathway elements, as well as the correlation of NRF2 pathway mRNAs with various clinicopathologic characteristics, including molecular subtypes, tumor grade, tumor stage, and methylation status, has been investigated in breast cancer using METABRIC and TCGA datasets. In the current report, our findings revealed the deregulation of several NRF2 signaling elements in breast cancer patients. Moreover, there were negative correlations between the methylation of NRF2 genes and mRNA expression. The expression of NRF2 genes significantly varied between different breast cancer subtypes. In conclusion, substantial deregulation of NRF2 signaling components suggests an important role of these genes in breast cancer. Because of the clear associations between mRNA expression and methylation status, DNA methylation could be one of the mechanisms that regulate the NRF2 pathway in breast cancer. Differential expression of Hippo genes among various breast cancer molecular subtypes suggests that NRF2 signaling may function differently in different subtypes of breast cancer. Our data also highlights an interesting link between NRF2 components' transcription and tumor grade/stage in breast cancer.
Collapse
Affiliation(s)
- Negin Soghli
- Babol University of Medical Sciences, Faculty of Dentistry, Babol, Iran
| | - Hassan Yousefi
- Louisiana State University Health Science Center (LSUHSC), Biochemistry & Molecular Biology, New Orleans, LA, USA; Stanley S. Scott Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Tohid Naderi
- Department of Laboratory Hematology and Blood Bank, School of Allied Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aysan Fallah
- Department of hematology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amin Moshksar
- University of Texas Medical Branch (UTMB), Interventional Radiology, Galveston, TX, USA
| | - Farzaneh Darbeheshti
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Cecilia Vittori
- Stanley S. Scott Cancer Research Center, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Mahsa Rostamian Delavar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Ali Zare
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Habib Sadeghi Rad
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, Australia
| | - Abtin Kazemi
- Fasa University of Medical Sciences, School of Medicine, Fasa, Iran
| | - Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Kurdistan Region, Erbil, Iraq
| | - Mohammad Taheri
- Men's Health and Reproductive Health Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Institute of Human Genetics, Jena University Hospital, Jena, Germany.
| | - Elena Jamali
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
89
|
Preclinical and Clinical Trials of New Treatment Strategies Targeting Cancer Stem Cells in Subtypes of Breast Cancer. Cells 2023; 12:cells12050720. [PMID: 36899854 PMCID: PMC10001180 DOI: 10.3390/cells12050720] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/01/2023] [Accepted: 02/09/2023] [Indexed: 02/26/2023] Open
Abstract
Breast cancer (BC) can be classified into various histological subtypes, each associated with different prognoses and treatment options, including surgery, radiation, chemotherapy, and endocrine therapy. Despite advances in this area, many patients still face treatment failure, the risk of metastasis, and disease recurrence, which can ultimately lead to death. Mammary tumors, like other solid tumors, contain a population of small cells known as cancer stem-like cells (CSCs) that have high tumorigenic potential and are involved in cancer initiation, progression, metastasis, tumor recurrence, and resistance to therapy. Therefore, designing therapies specifically targeting at CSCs could help to control the growth of this cell population, leading to increased survival rates for BC patients. In this review, we discuss the characteristics of CSCs, their surface biomarkers, and the active signaling pathways associated with the acquisition of stemness in BC. We also cover preclinical and clinical studies that focus on evaluating new therapy systems targeted at CSCs in BC through various combinations of treatments, targeted delivery systems, and potential new drugs that inhibit the properties that allow these cells to survive and proliferate.
Collapse
|
90
|
Mao H, Cao Y, Zou Z, Xia J, Zhao J. An enzyme-powered microRNA discriminator for the subtype-specific diagnosis of breast cancer. Chem Sci 2023; 14:2097-2106. [PMID: 36845930 PMCID: PMC9944337 DOI: 10.1039/d3sc00090g] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Breast cancer, a disease with highly heterogeneous features, is the most common malignancy diagnosed in people worldwide. Early diagnosis of breast cancer is crucial for improving its cure rate, and accurate classification of the subtype-specific features is essential to precisely treat the disease. An enzyme-powered microRNA (miRNA, RNA = ribonucleic acid) discriminator was developed to selectively distinguish breast cancer cells from normal cells and further identify subtype-specific features. Specifically, miR-21 was used as a universal biomarker to discriminate between breast cancer cells and normal cells, and miR-210 was used to identify triple-negative subtype features. The experimental results demonstrated that the enzyme-powered miRNA discriminator displayed low limits of detection at fM levels for both miR-21 and miR-210. Moreover, the miRNA discriminator enabled the discrimination and quantitative determination of breast cancer cells derived from different subtypes based on their miR-21 levels, and the further identification of the triple-negative subtype in combination with the miR-210 levels. Therefore, it is hoped that this study will provide insight into subtype-specific miRNA profiling, which may have potential use in the clinical management of breast tumours based on their subtype characteristics.
Collapse
Affiliation(s)
- Huiru Mao
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University Shanghai 200444 P. R. China
| | - Ya Cao
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing UniversityNanjing 210023P. R. China
| | - Zihan Zou
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University Shanghai 200444 P. R. China
| | - Jianan Xia
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University Shanghai 200444 P. R. China
| | - Jing Zhao
- Center for Molecular Recognition and Biosensing, Shanghai Engineering Research Center of Organ Repair, School of Life Sciences, Shanghai University Shanghai 200444 P. R. China
| |
Collapse
|
91
|
Patkulkar P, Subbalakshmi AR, Jolly MK, Sinharay S. Mapping Spatiotemporal Heterogeneity in Tumor Profiles by Integrating High-Throughput Imaging and Omics Analysis. ACS OMEGA 2023; 8:6126-6138. [PMID: 36844580 PMCID: PMC9948167 DOI: 10.1021/acsomega.2c06659] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 01/05/2023] [Indexed: 05/14/2023]
Abstract
Intratumoral heterogeneity associates with more aggressive disease progression and worse patient outcomes. Understanding the reasons enabling the emergence of such heterogeneity remains incomplete, which restricts our ability to manage it from a therapeutic perspective. Technological advancements such as high-throughput molecular imaging, single-cell omics, and spatial transcriptomics allow recording of patterns of spatiotemporal heterogeneity in a longitudinal manner, thus offering insights into the multiscale dynamics of its evolution. Here, we review the latest technological trends and biological insights from molecular diagnostics as well as spatial transcriptomics, both of which have witnessed burgeoning growth in the recent past in terms of mapping heterogeneity within tumor cell types as well as the stromal constitution. We also discuss ongoing challenges, indicating possible ways to integrate insights across these methods to have a systems-level spatiotemporal map of heterogeneity in each tumor and a more systematic investigation of the implications of heterogeneity for patient outcomes.
Collapse
|
92
|
(Stămat) LRB, Dinescu S, Costache M. Regulation of Inflammasome by microRNAs in Triple-Negative Breast Cancer: New Opportunities for Therapy. Int J Mol Sci 2023; 24:ijms24043245. [PMID: 36834660 PMCID: PMC9963301 DOI: 10.3390/ijms24043245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
During the past decade, researchers have investigated the molecular mechanisms of breast cancer initiation and progression, especially triple-negative breast cancer (TNBC), in order to identify specific biomarkers that could serve as feasible targets for innovative therapeutic strategies development. TNBC is characterized by a dynamic and aggressive nature, due to the absence of estrogen, progesterone and human epidermal growth factor 2 receptors. TNBC progression is associated with the dysregulation of nucleotide-binding oligomerization domain-like receptor and pyrin domain-containing protein 3 (NLRP3) inflammasome, followed by the release of pro-inflammatory cytokines and caspase-1 dependent cell death, termed pyroptosis. The heterogeneity of the breast tumor microenvironment triggers the interest of non-coding RNAs' involvement in NLRP3 inflammasome assembly, TNBC progression and metastasis. Non-coding RNAs are paramount regulators of carcinogenesis and inflammasome pathways, which could help in the development of efficient treatments. This review aims to highlight the contribution of non-coding RNAs that support inflammasome activation and TNBC progression, pointing up their potential for clinical applications as biomarkers for diagnosis and therapy.
Collapse
Affiliation(s)
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania
- Research Institute of the University of Bucharest, 050663 Bucharest, Romania
- Correspondence:
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania
- Research Institute of the University of Bucharest, 050663 Bucharest, Romania
| |
Collapse
|
93
|
Zheng J, Huang J, Xia J, Zhou W, Dai L, Lin S, Gao L, Zou C. Transcription factor E2F8 is a therapeutic target in the basal-like subtype of breast cancer. Front Oncol 2023; 13:1038787. [PMID: 36814821 PMCID: PMC9939474 DOI: 10.3389/fonc.2023.1038787] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 01/17/2023] [Indexed: 02/09/2023] Open
Abstract
Introduction Tumorigenesis in breast cancers usually accompanied by the dysregulation of transcription factors (TFs). Abnormal amplification of TFs leads aberrant expression of its downstream target genes. However, breast cancers are heterogeneous disease with different subtypes that have distinguished clinical behaviours, and the identification of prognostic TFs may enable to provide diagnosis and treatment of breast cancer based on subtypes, especially in Basal-like breast cancer. Methods The RNA-sequencing was performed to screen differential TFs in breast cancer subtypes. The GEPIA dataset analysis was used to analyze the genes expression in invasive breast carcinoma. The expression of MYBL2, HOXC13, and E2F8 was verified by qRT-PCR assay in breast cancers. The depiction analysis of co-expressed proteins was revealed using the STRING datasets. The cellular infiltration level analysis by the TISIDB and TIMER databases. The transwell assay was performed to analyze cellular migration and invasion. CCK-8 assay was used to evaluate cellular drug susceptibility for docetaxel treatment. Predicted targeted drugs in breast cancers by GSCA Lite database online. Results Kaplan-Meier plotter suggested that high expression of both E2F8 and MYBL2 in Basal-like subtype had a poor relapse-free survival. Functional enrichment results identified that apoptosis, cell cycle, and hormone ER pathway were represented the crucial regulation pathways by both E2F8 and MYBL2. In the meantime, database analysis indicated that high expression of E2F8 responded to chemotherapy, while those patients of high expression of MYBL2 responded to endocrinotherapy, and a positive correlation between the expression of E2F8 and PD-L1/CTLA4. Our cell line experiments confirmed the importance of E2F8 and MYBL2 in proliferation and chemotherapy sensitivity, possibly, the relationship with PD-L1. Additionally, we also observed that the up-regulation of E2F8 was accompanied with higher enrichments of CD4+ T cells and CD8+ T cells in breast cancers. Conclusion Taken together, our findings elucidated a prospective target in Basal-like breast cancer, providing underlying molecular biomarkers for the development of breast cancer treatment.
Collapse
Affiliation(s)
- Jing Zheng
- Department of Ultrasound, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jingyi Huang
- Department of Clinical Medical Research Center, The First Affiliated Hospital, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Jinquan Xia
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Wenbin Zhou
- Department of Thyroid and Breast Surgery, Department of General Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Lingyun Dai
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Sihang Lin
- Department of Thyroid and Breast Surgery, Department of General Surgery, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Lin Gao
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China,*Correspondence: Lin Gao, ; Chang Zou,
| | - Chang Zou
- Department of Clinical Medical Research Center, The Second Clinical Medical College, Jinan University (Shenzhen People’s Hospital), The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, Guangdong, China,Shenzhen Public Service Platform on Tumor Precision Medicine and Molecular Diagnosis, Shenzhen, Guangdong, China,School of Life and Health Sciences, The Chinese University of Kong Hong, Shenzhen, Guangdong, China,*Correspondence: Lin Gao, ; Chang Zou,
| |
Collapse
|
94
|
Zhou Y, Yu S, Chen D, Li H, Xu P, Yuan C, Jiang L, Huang M. Nafamostat Mesylate in Combination with the Mouse Amino-Terminal Fragment of Urokinase-Human Serum Albumin Improves the Treatment Outcome of Triple-Negative Breast Cancer Therapy. Mol Pharm 2023; 20:905-917. [PMID: 36463525 DOI: 10.1021/acs.molpharmaceut.2c00297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive and causes a higher proportion of metastatic cases. However, therapies directed to specific molecular targets have rarely achieved clinically meaningful improvements in the outcome of TNBC therapy. A urokinase-type plasminogen activator (uPA), one of the best-validated biomarkers of breast cancer, is an extracellular proteolytic serine protease involved in many pathological and physiological processes, including tumor cell invasion and metastasis. Nafamostat mesylate (NM) is a synthetic compound that inhibits various serine proteases and has been used as a therapeutic agent for the treatment of TNBC. Nevertheless, NM has poor specificity for serine proteases and is easy be hydrolyzed; moreover, the inhibitory mechanism of TNBC therapy is unclear. In this study, we combine NM with a macromolecular drug delivery vehicle, mouse amino-terminal fragment of urokinase-human serum albumin (mATF-HSA), to form a complex (mATF-HSA:NM) using the dilution-incubation-purification method. mATF specifically targets uPAR overexpressed on the surface of TNBC cells; moreover, HSA prevents NM from being hydrolyzed by numerous serine proteases. mATF-HSA:NM showed stronger inhibitory effects on the proliferation and metastasis of TNBC in vitro and in vivo without significant cytotoxicity on normal cells and tissues. In addition, we demonstrated that NM mediates metastasis of TNBC cells through inhibition of uPA using a stable uPA knockdown cell line (MDA-MB231 shuPA). Overall, we have developed a macromolecular complex targeted to treat high uPAR-expressing tumor types, and mATF-HSA can potentially be used to load other types of drugs with tumor-targeting specificity for mouse tumor models and is a promising tool to study tumor biology in mouse tumor models.
Collapse
Affiliation(s)
- Yang Zhou
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Shujuan Yu
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Dan Chen
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Hanlin Li
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Peng Xu
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Cai Yuan
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Longguang Jiang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China.,Fujian Key Lab Moratory of Marine Enzyme Engineering, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| | - Mingdong Huang
- College of Chemistry, Fuzhou University, Fuzhou, Fujian350116, P.R. China
| |
Collapse
|
95
|
Gonçalves JPL, Bollwein C, Noske A, Jacob A, Jank P, Loibl S, Nekljudova V, Fasching PA, Karn T, Marmé F, Müller V, Schem C, Sinn BV, Stickeler E, van Mackelenbergh M, Schmitt WD, Denkert C, Weichert W, Schwamborn K. Characterization of Hormone Receptor and HER2 Status in Breast Cancer Using Mass Spectrometry Imaging. Int J Mol Sci 2023; 24:ijms24032860. [PMID: 36769215 PMCID: PMC9918176 DOI: 10.3390/ijms24032860] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/27/2023] [Accepted: 01/31/2023] [Indexed: 02/05/2023] Open
Abstract
Immunohistochemical evaluation of estrogen receptor, progesterone receptor, and human epidermal growth factor receptor-2 status stratify the different subtypes of breast cancer and define the treatment course. Triple-negative breast cancer (TNBC), which does not register receptor overexpression, is often associated with worse patient prognosis. Mass spectrometry imaging transcribes the molecular content of tissue specimens without requiring additional tags or preliminary analysis of the samples, being therefore an excellent methodology for an unbiased determination of tissue constituents, in particular tumor markers. In this study, the proteomic content of 1191 human breast cancer samples was characterized by mass spectrometry imaging and the epithelial regions were employed to train and test machine-learning models to characterize the individual receptor status and to classify TNBC. The classification models presented yielded high accuracies for estrogen and progesterone receptors and over 95% accuracy for classification of TNBC. Analysis of the molecular features revealed that vimentin overexpression is associated with TNBC, supported by immunohistochemistry validation, revealing a new potential target for diagnosis and treatment.
Collapse
Affiliation(s)
- Juliana Pereira Lopes Gonçalves
- Institute of Pathology, School of Medicine, Technical University of Munich, Trogerstraße 18, 81675 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Christine Bollwein
- Institute of Pathology, School of Medicine, Technical University of Munich, Trogerstraße 18, 81675 Munich, Germany
| | - Aurelia Noske
- Institute of Pathology, School of Medicine, Technical University of Munich, Trogerstraße 18, 81675 Munich, Germany
| | - Anne Jacob
- Institute of Pathology, School of Medicine, Technical University of Munich, Trogerstraße 18, 81675 Munich, Germany
| | - Paul Jank
- Institute of Pathology, Philipps-University Marburg and University Hospital Marburg (UKGM), 35043 Marburg, Germany
| | - Sibylle Loibl
- German Breast Group (GBG), 63263 Neu-Isenburg, Germany
| | | | - Peter A. Fasching
- Department of Gynecology and Obstetrics, Comprehensive Cancer Center Erlangen-EMN, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054 Erlangen, Germany
| | - Thomas Karn
- Department of Gynecology and Obstetrics, Goethe-University Frankfurt, 60590 Frankfurt, Germany
| | - Frederik Marmé
- Department of Obstetrics and Gynecology, University Hospital Mannheim, Medical Faculty Mannheim, Heidelberg University, 68167 Mannheim, Germany
| | - Volkmar Müller
- Department of Gynecology, Universitätsklinikum Hamburg-Eppendorf, 20251 Hamburg, Germany
| | | | | | - Elmar Stickeler
- Department of Obstetrics and Gynecology, University Hospital Aachen, 52074 Aachen, Germany
| | - Marion van Mackelenbergh
- Klinik für Gynäkologie und Geburtshilfe, Universitätsklinikum Schleswig-Holstein, 24105 Kiel, Germany
| | | | - Carsten Denkert
- Institute of Pathology, Philipps-University Marburg and University Hospital Marburg (UKGM), 35043 Marburg, Germany
| | - Wilko Weichert
- Institute of Pathology, School of Medicine, Technical University of Munich, Trogerstraße 18, 81675 Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, 80336 Munich, Germany
| | - Kristina Schwamborn
- Institute of Pathology, School of Medicine, Technical University of Munich, Trogerstraße 18, 81675 Munich, Germany
- Correspondence:
| |
Collapse
|
96
|
Radiosensitivity is associated with antitumor immunity in estrogen receptor-negative breast cancer. Breast Cancer Res Treat 2023; 197:479-488. [PMID: 36515748 DOI: 10.1007/s10549-022-06818-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 11/11/2022] [Indexed: 12/15/2022]
Abstract
PURPOSE This study evaluated radiosensitivity and the tumor microenvironment (TME) to identify characteristics of breast cancer patients who would benefit most from radiation therapy. METHODS We analyzed 1903 records from the Molecular Taxonomy of Breast Cancer International Consortium cohort using the radiosensitivity index and gene expression deconvolution algorithms, CIBERSORT and xCell, that estimates the TME composition of tumor samples. In this study, patients were stratified according to TME and radiosensitivity. We performed integrative analyses of clinical and immuno-genomic data to characterize molecular features associated with radiosensitivity. RESULTS Radiosensitivity was significantly associated with activation of antitumor immunity. In contrast, radioresistance was associated with a reactive stromal microenvironment. The immuno-genomic analysis revealed that estrogen receptor (ER) pathway activity was correlated with suppression of antitumor immunity. In ER-negative disease, the best prognosis was shown in the immune-high and radiosensitive group patients, and the lowest was in the immune-low and radioresistant group patients. In ER-positive disease, immune signature and radiosensitivity had no prognostic significance. CONCLUSION Taken together, these results suggest that tumor radiosensitivity is associated with activation of antitumor immunity and a better prognosis, particularly in patients with ER-negative breast cancer.
Collapse
|
97
|
Wang C, Wang G, Zhang Y, Dai Y, Yang D, Wang C, Li J. Differentiation of benign and malignant breast lesions using diffusion-weighted imaging with a fractional-order calculus model. Eur J Radiol 2023; 159:110646. [PMID: 36577184 DOI: 10.1016/j.ejrad.2022.110646] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 11/25/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
PURPOSE To assess the feasibility of using three diffusion parameters (D, β, and μ) derived from fractional-order calculus (FROC) diffusion model for improving the differentiation between benign and malignant breast lesions. METHOD In this prospective study, 103 patients with breast lesions were enrolled. All subjects underwent diffusion-weighted imaging (DWI) with 12b values. Inter-observer agreement with respect to quantification of parameters by two radiologists was assessed using intraclass coefficient. Conventional apparent diffusion coefficient (ADC) and three FROC model parameters D, β, and μ were compared between the benign lesion and malignant lesion groups using the Mann-Whitney U test. Then, a comprehensive prediction model was created by using binary logistic regression. Receiver operating characteristic (ROC) curve analysis was performed to evaluate the diagnostic performance of the parameters using histopathological diagnosis as the reference standard. RESULTS The FROC parameters and ADC all exhibited significant differences between benign lesions and malignant lesions (P<0.001). Among the individual parameters, the sensitivity of μ was higher than ADC (95.92% for μ vs 91.84% for ADC), and the specificity of β was higher than ADC (72.22% for β vs 70.37% for ADC). The combination of ADC and FROC parameters (D and β) generated the largest area under the ROC curve (0.841) when compared with individual parameters, indicating an improved performance for differentiating benign lesions from malignant lesions. CONCLUSIONS This study demonstrated the feasibility of using the FROC diffusion model to improve the accuracy of identifying malignant breast lesions.
Collapse
Affiliation(s)
- Chunhong Wang
- Department of Radiology, Xinyang Central Hospital, No. 01 Xinyang Siyi Road, Xinyang 464000, Henan, China
| | - Guanying Wang
- Department of Radiology, Xinyang Central Hospital, No. 01 Xinyang Siyi Road, Xinyang 464000, Henan, China
| | - Yunfei Zhang
- MR Collaboration, Central Research Institute, United Imaging Healthcare, Shanghai, China
| | - Yongming Dai
- MR Collaboration, Central Research Institute, United Imaging Healthcare, Shanghai, China
| | - Dan Yang
- Department of Radiology, Xinyang Central Hospital, No. 01 Xinyang Siyi Road, Xinyang 464000, Henan, China
| | - Changfu Wang
- Imaging department, Huaihe Hospital, Henan University, Kaifeng, 475000, Henan, China
| | - Jianhong Li
- Department of Radiology, Xinyang Central Hospital, No. 01 Xinyang Siyi Road, Xinyang 464000, Henan, China.
| |
Collapse
|
98
|
Sun Y, Wang Y, Lu F, Zhao X, Nie Z, He B. The prognostic values of FOXP3 + tumor-infiltrating T cells in breast cancer: a systematic review and meta-analysis. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:1830-1843. [PMID: 36692642 DOI: 10.1007/s12094-023-03080-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/09/2023] [Indexed: 01/25/2023]
Abstract
BACKGROUND Tumor microenvironment is infiltrated by many immune cells, of which Regulatory T (Treg) cells are usually considered as negative regulators of the immune responses. However, the effect of FOXP3+ (forkhead box transcription factor 3) Treg cells infiltrated into the tumor areas on the prognosis of breast cancer is controversial. This meta-analysis aimed to dissect the potential values of FOXP3+ tumor-infiltrating lymphocytes (TILs) as a prognosis predictor of breast cancer. METHODS After systematic retrieval of all relevant studies, 28 eligible articles were identified for meta-analysis. Odd ratio (OR), hazard ratio (HR), and 95% confidence interval (CI) were obtained for pooled analyses of pathological complete response (pCR), overall survival (OS), and corresponding forest plots and funnel plots were plotted, respectively. RESULTS Pooled results revealed that patients with higher levels of FOXP3+ TILs experienced better pCR (OR: 1.24, 95% CI 1.09-1.41) and OS (HR: 0.79, 95% CI 0.64-0.97). Subgroup analysis revealed that elevated FOXP3+ TILs were significantly associated with improved pCR (OR: 1.20, 95% CI 1.02-1.40) and OS (HR: 0.22, 95% CI 0.06-0.88) in human epidermal growth factor receptor 2 positive (HER2+) breast cancer patients. Furthermore, FOXP3+ TILs in the stromal area were statistically correlated with the favorable pCR (OR: 1.22, 95% CI 1.08-1.38) and OS (HR: 0.68, 95% CI 0.49-0.96). CONCLUSIONS The predictive role of FOXP3+ TILs in the prognosis of breast cancer is influenced by various factors such as molecular subtype of breast cancer and the location of Treg. In HER2+ breast cancer and triple-negative breast cancer, FOXP3+ TILs are associated with better pCR and OS. Additionally, FOXP3+ TILs in stromal represent a favourable prognosis.
Collapse
Affiliation(s)
- Yalan Sun
- School of Basic-Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Ying Wang
- School of Basic-Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Fang Lu
- School of Basic-Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Xianghong Zhao
- School of Basic-Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.,Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China
| | - Zhenlin Nie
- Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China.
| | - Bangshun He
- School of Basic-Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China. .,Department of Laboratory Medicine, Nanjing First Hospital, Nanjing Medical University, Nanjing, 210006, Jiangsu, China.
| |
Collapse
|
99
|
Alsaleh K, Al Zahwahry H, Bounedjar A, Oukkal M, Saadeddine A, Mahfouf H, Bouzid K, Bensalem A, Filali T, Abdel-Razeq H, Larbaoui B, Kandil A, Abulkhair O, Al Foheidi M, Ghosn M, Rasool H, Boussen H, Mezlini A, Haddaoui A, Ayari J, Al Ghamdi M, Errihani H, Abdel-Aziz N, Arafah M, Dabouz F, Bahadoor M, Kullab S, Nabholtz JM. Neoadjuvant endocrine therapy with or without palbociclib in low-risk patients: a phase III randomized double-blind SAFIA trial. J Cancer Res Clin Oncol 2023:10.1007/s00432-023-04588-3. [PMID: 36680581 PMCID: PMC9864499 DOI: 10.1007/s00432-023-04588-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 01/11/2023] [Indexed: 01/22/2023]
Abstract
BACKGROUND The most prevalent subtype of breast cancer (BC) is luminal hormonal-positive breast cancer. The neoadjuvant chemotherapy regimens have side effects, emphasizing the need to identify new startegies. OBJECTIVE Analyze the complete pathologic response (pCR) rate and overall response in a low-risk hormone-positive subset of patients receiving neoadjuvant hormone treatment (NAHT) with or without Palbociclib (a CDK4/CDK6 inhibitor) to boost NAHT effectiveness. MATERIALS AND METHODS Based on the upfront 21-gene Oncotype DX or low-risk Breast Recurrence Score assay (RS™), the SAFIA trial is designed as a prospective multicenter international, double-blind neoadjuvant phase-III trial that selects operable with luminal BC patients that are HER2-negative for the induction hormonal therapy with Fulvestrant 500 mg ± Goserelin (F/G) followed by randomization of responding patients to palbociclib versus placebo. The pCR rate served as the study's main outcome, while the secondary endpoint was a clinical benefit. RESULTS Of the 354 patients enrolled, 253 initially responded and were randomized to either F/G fulvestrant with palbociclib or placebo. Two hundred twenty-nine were eligible for the evaluation of the pathologic response. No statistically significant changes were observed in the pCR rates for the patients treated with the F/G therapy with placebo or palbociclib (7% versus 2%, respectively) per the Chevallier classification (Class1 + Class2) (p = 0.1464) and 3% versus 10% assessed per Sataloff Classification (TA, NA/NB) (p = 0.3108). Palbociclib did not increase the rate of complete pathological response. CONCLUSION Neoadjuvant hormonal therapy is feasible in a selected population with a low RS score of < 31 CLINICAL TRIAL: NCT03447132.
Collapse
Affiliation(s)
- K. Alsaleh
- grid.56302.320000 0004 1773 5396College of Medicine, King Saud University, King Saud University Medical City (KSUMC), Oncology Center, Riyadh, 12372 Saudi Arabia
| | - H. Al Zahwahry
- grid.7776.10000 0004 0639 9286National Cancer Institute (NCI), Cairo University, Cairo, Egypt
| | | | - M. Oukkal
- Medical Oncology Department/Beni-Messous University Hospital, Algiers, Algeria
| | - A. Saadeddine
- grid.415254.30000 0004 1790 7311Oncology Center, King Abdulaziz Medical City (KAMC), Riyadh, Saudi Arabia
| | | | - K. Bouzid
- Oncology Center of Pierre Et Marie Curie, Algiers, Algeria
| | - A. Bensalem
- Dr. Benbadis University Hospital, Constantine, Algeria
| | - T. Filali
- Constantine University Hospital, Constantine, Algeria
| | - H. Abdel-Razeq
- grid.419782.10000 0001 1847 1773King Hussein Cancer Center (KHCC), Amman, Jordan
| | - B. Larbaoui
- Oncology Center of Emir Abdelkader, Oran, Algeria
| | - A. Kandil
- grid.7155.60000 0001 2260 6941Alexandria University, Alexandria, Egypt
| | - O. Abulkhair
- Specialized Medical Center (SMC), Riyadh, Saudi Arabia
| | - M. Al Foheidi
- grid.415254.30000 0004 1790 7311Oncology Center of Princess Noorah, King Abdulaziz Medical City (KAMC), Jeddah, Saudi Arabia
| | - M. Ghosn
- grid.413559.f0000 0004 0571 2680Hematology-Oncology Department/Hotel Dieu de France/University Saint Joseph, Beirut, Lebanon
| | - H. Rasool
- grid.415310.20000 0001 2191 4301King Faisal Specialist Hospital and Research Center (KFSHRC), Jeddah, Saudi Arabia
| | - H. Boussen
- grid.12574.350000000122959819Faculty of Medicine, University Tunis El Manar, Abderrahmen Mami Hospital, Tunis, Ariana Tunisia
| | - A. Mezlini
- Medical Oncology Department, Tunis, Tunisia
| | | | - J. Ayari
- Faculty of Medicine Tunis, Oncology Department, University Tunis El Manar, Military Hospital of Tunis, Tunis, Tunisia
| | - M. Al Ghamdi
- grid.56302.320000 0004 1773 5396Oncology center, King Saud University Medical City (KSUMC), King Saud University, Riyadh, 12372 Saudi Arabia
| | - H. Errihani
- Medical Oncology, National Institute of Oncology, University Mohammed V, Rabat, Morocco
| | - N. Abdel-Aziz
- grid.56302.320000 0004 1773 5396Oncology Center, King Saud University Medical City (KSUMC), King Saud University, Riyadh, Saudi Arabia
| | - M. Arafah
- grid.56302.320000 0004 1773 5396Department of Pathology, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - F. Dabouz
- International Cancer Research Group (ICRG), Sharjah, UAE
| | - M. Bahadoor
- International Cancer Research Group (ICRG), Sharjah, UAE
| | - S. Kullab
- grid.56302.320000 0004 1773 5396Oncology center, King Saud University Medical City (KSUMC), King Saud University, Riyadh, 12372 Saudi Arabia
| | - J. M. Nabholtz
- grid.56302.320000 0004 1773 5396Oncology center, King Saud University Medical City (KSUMC), King Saud University, Riyadh, 12372 Saudi Arabia
| | | |
Collapse
|
100
|
Eid RA, Alaa Edeen M, Shedid EM, Kamal ASS, Warda MM, Mamdouh F, Khedr SA, Soltan MA, Jeon HW, Zaki MSA, Kim B. Targeting Cancer Stem Cells as the Key Driver of Carcinogenesis and Therapeutic Resistance. Int J Mol Sci 2023; 24:ijms24021786. [PMID: 36675306 PMCID: PMC9861138 DOI: 10.3390/ijms24021786] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 01/18/2023] Open
Abstract
The emerging concept of cancer stem cells (CSCs) as the key driver behind carcinogenesis, progression, and diversity has displaced the prior model of a tumor composed of cells with similar subsequently acquired mutations and an equivalent capacity for renewal, invasion, and metastasis. This significant change has shifted the research focus toward targeting CSCs to eradicate cancer. CSCs may be characterized using cell surface markers. They are defined by their capacity to self-renew and differentiate, resist conventional therapies, and generate new tumors following repeated transplantation in xenografted mice. CSCs' functional capabilities are governed by various intracellular and extracellular variables such as pluripotency-related transcription factors, internal signaling pathways, and external stimuli. Numerous natural compounds and synthetic chemicals have been investigated for their ability to disrupt these regulatory components and inhibit stemness and terminal differentiation in CSCs, hence achieving clinical implications. However, no cancer treatment focuses on the biological consequences of these drugs on CSCs, and their functions have been established. This article provides a biomedical discussion of cancer at the time along with an overview of CSCs and their origin, features, characterization, isolation techniques, signaling pathways, and novel targeted therapeutic approaches. Additionally, we highlighted the factors endorsed as controlling or helping to promote stemness in CSCs. Our objective was to encourage future studies on these prospective treatments to develop a framework for their application as single or combined therapeutics to eradicate various forms of cancer.
Collapse
Affiliation(s)
- Refaat A. Eid
- Pathology Department, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia
| | - Muhammad Alaa Edeen
- Cell Biology, Histology & Genetics Division, Biology Department, Faculty of Science, Zagazig University, Zagazig 44519, Egypt
- Correspondence: (M.A.E.); (B.K.)
| | - Eslam M. Shedid
- Biotechnology Division, Zoology Department, Faculty of Science, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Al Shaimaa S. Kamal
- Biotechnology Department, Faculty of Agriculture, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Mona M. Warda
- Biotechnology Division, Zoology Department, Faculty of Science, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Farag Mamdouh
- Biotechnology Division, Zoology Department, Faculty of Science, Benha University, Al Qalyubia Governorate, Banha 13511, Egypt
| | - Sohila A. Khedr
- Industrial Biotechnology Department, Faculty of Science, Tanta University, Tanta 31733, Egypt
| | - Mohamed A. Soltan
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, Ismailia 41611, Egypt
| | - Hee Won Jeon
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Mohamed Samir A. Zaki
- Anatomy Department, College of Medicine, King Khalid University, Abha P.O. Box 62529, Saudi Arabia
- Department of Histology and Cell Biology, College of Medicine, Zagazig University, Zagazig 31527, Egypt
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Correspondence: (M.A.E.); (B.K.)
| |
Collapse
|