1
|
Li Y, Li H, Du L, Shi Q, Li G, Jia C, Jin L, Liang H, Li F. Construction of a multi-parametric ultrasonographic nomogram for precise assessment of papillary breast lesions. Br J Radiol 2025; 98:422-431. [PMID: 39656793 DOI: 10.1093/bjr/tqae242] [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: 02/01/2024] [Revised: 09/22/2024] [Accepted: 11/26/2024] [Indexed: 12/17/2024] Open
Abstract
OBJECTIVES To analyze the multi-parametric ultrasonographic (MpUS) features of atypical/malignant papillary lesions of the breast with clinical information, identify independent risk factors, and construct a nomogram to improve the diagnostic accuracy. METHODS This retrospective study analyzed consecutively hospitalized patients diagnosed with pathologically confirmed papillary breast lesions from January 2017 to June 2023. Preoperative sonographic examinations, including gray-scale ultrasound (G-US), color Doppler flow imaging (CDFI), and contrast-enhanced ultrasound (CEUS), were conducted. Sonographic scans were retrospectively reviewed alongside clinicopathological data. Binary logistic regression identified independent risk factors for screening atypical/malignant papillary lesions. The receiver operating characteristic curve evaluated the predictive accuracy of these lesions, resulting in the development of a nomogram for assessing risk. RESULTS The study involved 176 female patients with breast papillary lesions, identifying key predictors for atypical or malignant outcomes: age 57 or order, US diameter ≥13.95 mm, resistive index ≥0.70, enlarged enhancement on CEUS, and contrast agent retention, with respective odds ratios of 6.279, 8.078, 9.246, 9.401, and 5.047. The integrated use of G-US, CDFI, and CEUS in the MpUS approach offered higher diagnostic accuracy (area under the curve [AUC]: 0.966) than G-US or CDFI alone (0.869/0.918). CEUS particularly enhanced prediction for non-mass-like lesions, with a positive predictive value of 83.3%. A nomogram incorporating MpUS and patient age achieved an AUC of 0.956 for predicting atypical or malignant papillary lesions. CONCLUSIONS MpUS imaging is highly effective for predicting malignant breast papillary lesions, especially considering patient age. The nomogram offers an intuitive framework for assessing malignant risk in these lesions. ADVANCES IN KNOWLEDGE Ultrasound excels in identifying papillary lesions, and integrating diverse data and multi-parametric imaging enhances malignant risk evaluation. This study establishes a predictive risk model using the nomogram method, demonstrating heightened diagnostic efficacy in breast papillary lesions.
Collapse
Affiliation(s)
- Yunhua Li
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Hui Li
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Lianfang Du
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Qiusheng Shi
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Gang Li
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Chao Jia
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Lifang Jin
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Hongmei Liang
- Department of Nursing, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| | - Fan Li
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China
| |
Collapse
|
2
|
Alqahtani NF, Alfaifi MY, Shati A A, Elbehairi SEI, Saleh AM, Kotb ES, Serag WM, Elshaarawy RFM, Alhamdi HW, Hassan YA. Molecular docking and in vivo/ in vitro studies of a novel thiadiazole Schiff base as a hepatoprotective drug against angiogenesis induced by breast cancer. RSC Adv 2024; 14:39027-39039. [PMID: 39659603 PMCID: PMC11629753 DOI: 10.1039/d4ra06398h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/17/2024] [Indexed: 12/12/2024] Open
Abstract
Two new thiadiazole imidazolium salicylidene Schiff bases (TISSBs) were successfully synthesized, and their structures were analyzed comprehensively using spectroscopic techniques. The results of the MTT assay showed that TISSB2 was the safest and most effective anti-breast cancer agent. The anti-angiogenic activity of TISSB2 was evaluated using in vivo tests in Ehrlich ascites carcinoma (EAC)-bearing Swiss albino mice. The degree of angiogenesis was assessed by measuring the levels of vascular endothelial growth factor (VEGF), tumor necrosis factor-α (TNF-α), and transforming growth factor-β1 (TGF-β1). The results of biochemical, immunohistochemical, and histopathological examinations indicated that TISSB2 could restore the normal functional indices of the injured liver, as evident from the downregulated TGF-β1, TNF-α, and VEGF levels reverting to normal values. Moreover, in the molecular docking study, TISSB2 exhibited stronger interactions with VEGFR-2 and NF-κB proteins, with binding affinity scores of -11.79 and -9.25 kcal mol-1, respectively. These stronger interactions involved H-bonding, ionic bonds, and hydrophobic π-interactions. Overall, TISSB2 can be a promising therapeutic option for the treatment of EAC-induced tumour angiogenesis.
Collapse
Affiliation(s)
- Norah F Alqahtani
- Department of Chemistry, College of Science, University of Jeddah Jeddah 21589 Saudi Arabia
| | - Mohammad Y Alfaifi
- King Khalid University, Faculty of Science, Biology Department Abha 9004 Saudi Arabia
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University Abha 9004 Saudi Arabia
| | - Ali Shati A
- King Khalid University, Faculty of Science, Biology Department Abha 9004 Saudi Arabia
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University Abha 9004 Saudi Arabia
| | - Serag Eldin I Elbehairi
- King Khalid University, Faculty of Science, Biology Department Abha 9004 Saudi Arabia
- Tissue Culture and Cancer Biology Research Laboratory, King Khalid University Abha 9004 Saudi Arabia
- Cell Culture Lab, Egyptian Organization for Biological Products and Vaccines (VACSERA Holding Company) 51 Wezaret El-Zeraa St., Agouza Giza Egypt
| | - Abdulrahman M Saleh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University Kasr El-Aini Street Cairo 11562 Egypt
- Aweash El-Hagar Family Medicine Center, Epidemiological Surveillance Unit, MOHP Mansoura 35711 Egypt
| | - Ebtesam S Kotb
- Department of Chemistry, Faculty of Science, Suez University 43533 Suez Egypt
| | - Waleed M Serag
- Department of Chemistry, Faculty of Science, Suez University 43533 Suez Egypt
| | - Reda F M Elshaarawy
- Department of Chemistry, Faculty of Science, Suez University 43533 Suez Egypt
- Institut für Anorganische Chemie und Strukturchemie, Heinrich-Heine Universität Düsseldorf Düsseldorf Germany
| | - Heba W Alhamdi
- College of Sciences, Biology Department, King Khalid University Abha 61413 Saudi Arabia
| | - Yasser A Hassan
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology Gamasa Egypt
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Al-Kitab University Kirkuk Iraq
| |
Collapse
|
3
|
Eldehna WM, Habib YA, Mahmoud AE, Barghash MF, Elsayed ZM, Elsawi AE, Maklad RM, Rashed M, Khalil A, Hammad SF, Ali MM, El Kerdawy AM. Design, synthesis, and in silico insights of novel N'-(2-oxoindolin-3-ylidene)piperidine-4-carbohydrazide derivatives as VEGFR-2 inhibitors. Bioorg Chem 2024; 153:107829. [PMID: 39317037 DOI: 10.1016/j.bioorg.2024.107829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/09/2024] [Accepted: 09/16/2024] [Indexed: 09/26/2024]
Abstract
Vascular endothelial growth factor (VEGF) is a crucial key factor in breast tumorigenesis. VEGF plays an important role in angiogenesis, tumor proliferation, and metastasis. Herein, we report the design and synthesis of twenty-one novel piperidine/oxindole derivatives as potential VEGFR-2 inhibitors. The designed compound library aimed to occupy the binding site of VEGFR-2 in a similar binding pattern to that of the reference VEGFR-2 inhibitor Sorafenib. The synthesized compounds were biologically evaluated for their cytotoxic effects against two breast cancer cell lines (MCF-7 and MDA-MB-468). Compounds 12e and 6n were the most potent cytotoxic derivatives against the former and the latter cell lines, showing IC50 values of 8.00 and 0.60 µM, respectively. Furthermore, all the synthesized compounds were evaluated for their inhibitory activities towards VEGFR-2, with compound 12e showing the most potent activity with an IC50 value of 45.9 nM, surpassing the reference standard Sorafenib (IC50 = 48.6 nM). Additionally, compound 6n emerged as the top performer when tested with the other most promising compounds for their cytotoxic effects on HUVEC (IC50 = 28.77 nM). The designed library of compounds was subjected to molecular docking and molecular dynamic simulations, which revealed key binding interactions within the VEGFR-2 active site, including hydrogen bonding with Cys919, Glu885, and Asp1046 residues. Moreover, in silico predictions of physicochemical and pharmacokinetic properties for the target compounds indicated favorable drug-like characteristics.
Collapse
Affiliation(s)
- Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh P.O. Box 33516, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia St., Alexandria 21648, Egypt.
| | - Youmna A Habib
- Scientific Research and Innovation Support Unit, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Abeer E Mahmoud
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Dokki 12622, Giza, Egypt
| | - Mohamed F Barghash
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Dokki 12622, Giza, Egypt
| | - Zainab M Elsayed
- Scientific Research and Innovation Support Unit, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh 33516, Egypt
| | - Ahmed E Elsawi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh P.O. Box 33516, Egypt
| | - Raed M Maklad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh P.O. Box 33516, Egypt
| | - Mahmoud Rashed
- Pharmaceutical Medicinal Chemistry & Drug Design Department, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo 11884, Egypt
| | - Amira Khalil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt
| | - Sherif F Hammad
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Ain Helwan, Egypt; PharmD Program, Egypt-Japan University of Science and Technology, New Borg El-Arab, Alexandria, Egypt
| | - Mamdouh M Ali
- Biochemistry Department, Biotechnology Research Institute, National Research Centre, Dokki 12622, Giza, Egypt
| | - Ahmed M El Kerdawy
- School of Pharmacy, College of Health and Science, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln, Lincolnshire, United Kingdom; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, P.O. Box 11562 Cairo, Egypt
| |
Collapse
|
4
|
Yuan T, Fu X, Hu R, Zheng X, Jiang D, Jing L, Kuang X, Guo Z, Luo X, Liu Y, Zou X, Luker GD, Mi S, Liu C, Sun W. Bioprinted, spatially defined breast tumor microenvironment models of intratumoral heterogeneity and drug resistance. Trends Biotechnol 2024; 42:1523-1550. [PMID: 39112274 DOI: 10.1016/j.tibtech.2024.06.007] [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: 03/20/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 11/17/2024]
Abstract
Cellular, extracellular matrix (ECM), and spatial heterogeneity of tumor microenvironments (TMEs) regulate disease progression and treatment efficacy. Developing in vitro models that recapitulate the TME promises to accelerate studies of tumor biology and identify new targets for therapy. Here, we used extrusion-based, multi-nozzle 3D bioprinting to spatially pattern triple-negative MDA-MB-231 breast cancer cells, endothelial cells (ECs), and human mammary cancer-associated fibroblasts (HMCAFs) with biomimetic ECM inks. Bioprinted models captured key features of the spatial architecture of human breast tumors, including varying-sized dense regions of cancer cells and surrounding microvessel-rich stroma. Angiogenesis and ECM stiffening occurred in the stromal area but not the cancer cell-rich (CCR) regions, mimicking pathological changes in patient samples. Transcriptomic analyses revealed upregulation of angiogenesis-related and ECM remodeling-related signatures in the stroma region and identified potential ligand-receptor (LR) mediators of these processes. Breast cancer cells in distinct parts of the bioprinted TME showed differing sensitivities to chemotherapy, highlighting environmentally mediated drug resistance. In summary, our 3D-bioprinted tumor model will act as a platform to discover integrated functions of the TME in cancer biology and therapy.
Collapse
Affiliation(s)
- Tianying Yuan
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, 518055, Shenzhen, China; Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Xihong Fu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Rongcheng Hu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Xiaochun Zheng
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Dong Jiang
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Lanyu Jing
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Xiaying Kuang
- Department of Breast Surgery, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Zhongwei Guo
- School of Mechanics and Safety Engineering, Zhengzhou University, 450001, Zhengzhou, China
| | - Xu Luo
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Yixin Liu
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China
| | - Gary D Luker
- Department of Radiology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| | - Shengli Mi
- Tsinghua Shenzhen International Graduate School, Tsinghua University, 518055, Shenzhen, China.
| | - Chun Liu
- Institute of Precision Medicine, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, The First Affiliated Hospital of Sun Yat-sen University, 510080, Guangzhou, China.
| | - Wei Sun
- Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, 518055, Shenzhen, China; Department of Mechanical Engineering, Biomanufacturing Center, Tsinghua University, 100084, Beijing, China; Department of Mechanical Engineering, Drexel University, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Hamed SF, Hassan NA, Shouman SA, Tohamy TA, Fakhry H, Radwan E. Down regulation of C1q tumor necrosis factor-related protein 6 is associated with increased risk of breast cancer. Arch Biochem Biophys 2024; 757:110039. [PMID: 38750921 DOI: 10.1016/j.abb.2024.110039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 04/26/2024] [Accepted: 05/11/2024] [Indexed: 05/27/2024]
Abstract
C1q tumor necrosis factor-related protein 6 (CTRP6), a member of the C1q tumor necrosis factor-related protein (CTRP) family, is reported to be associated with the progression of different malignancies, however, its expression levels and role in breast cancer (BC) are yet unknown. In this study, we investigated the levels of circulating CTRP6 in BC patients and evaluated its role as a potential diagnostic biomarker in BC patients. Then we investigated the effect of recombinant CTRP6 on cellular viability in MCF-7 cells along with its effects on the expression of inflammatory cytokines, interleukin 6 (IL-6), tumor necrosis factor alpha (TNF-α) in addition to the expression of vascular endothelial growth factor (VEGF) as a marker of angiogenesis. Our results showed decreased expression of circulating CTRP6 in BC patients with an inverse correlation between CTRP6 and IL-6, TNF-α and VEGF levels. Besides, Receiver operating characteristic (ROC) curve showed that the assessment of CTRP6 levels could be used to predict BC. Moreover, treatment of MCF-7 cells with recombinant CTRP6 protein reduced cellular viability and decreased IL-6, TNF-α and VEGF expression. In conclusion, these results provide new insights into the role of CTRP6 in BC pathogenesis and suggest its potential use as a novel diagnostic biomarker of BC.
Collapse
Affiliation(s)
- Samia F Hamed
- Cancer Biology Department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Nivin A Hassan
- Cancer Biology Department (Pharmacology and Experimental Oncology), South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Samia A Shouman
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| | | | - Hussein Fakhry
- Surgical Oncology Department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Eman Radwan
- Medical Biochemistry Department, Faculty of Medicine, Assiut University, Assiut, Egypt; Biochemistry Department, Sphinx University, New Assiut City, Assiut, Egypt.
| |
Collapse
|
6
|
Sindhura N, Kaumudi K. Vascular Endothelial Growth Factor Expression by Immunohistochemistry as a Possible Indicator of Prognosis in Invasive Breast Carcinoma of No Special Type. Int J Appl Basic Med Res 2024; 14:124-130. [PMID: 38912361 PMCID: PMC11189271 DOI: 10.4103/ijabmr.ijabmr_17_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/20/2024] [Accepted: 03/22/2024] [Indexed: 06/25/2024] Open
Abstract
Context Angiogenesis, the formation of new blood vessels from preexisting vascular network, is essential for tumor growth and spread. Vascular endothelial growth factor (VEGF) is a potent angiogenic growth factor. Aims To assess the expression of VEGF in invasive carcinoma of no special type and its correlation with all the known prognostic factors of breast carcinoma. Settings and Design Descriptive. Materials and Methods Mastectomy specimens were studied noting the clinical details. The formalin-fixed tissues were subjected to routine processing and hematoxylin and eosin sections and studied extensively for all the histological prognostic factors. Representative sections from each case with the tumor were subjected to immunohistochemistry (IHC) staining with VEGF, estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2/neu) antibodies. Statistical Analysis Used Descriptive statistics, Chi-square tests, contingency table analysis using SPSS for Windows. Results One hundred and twelve cases of invasive carcinoma of special type were studied to evaluate various clinicopathological parameters. The association of VEGF with clinicopathological parameters and all the known prognostic factors was studied to note its significance. VEGF overexpression was observed in 69% of the cases. It was noted that larger tumor size, higher histological grade, lymphovascular invasion, nodal involvement, tumor necrosis, high microvessel density, ER negativity, PR negativity, and HER2/neu positivity had a significant statistical association with VEGF overexpression. Conclusions We conclude that incorporating VEGF as a biomarker along with the known factors into a prognostic index will not only help predict clinical outcome more accurately, but also determines the patient who can be benefited with combinational therapy including anti-VEGF factors.
Collapse
Affiliation(s)
- Nugala Sindhura
- Department of Pathology, Guntur Medical College, Guntur, Andhra Pradesh, India
| | - Konkay Kaumudi
- Department of Pathology, Siddhartha Medical College, Vijayawada, Andhra Pradesh, India
| |
Collapse
|
7
|
Lyu Y, Wu C, Sun W, Li Z. Regional analysis to delineate intrasample heterogeneity with RegionalST. Bioinformatics 2024; 40:btae186. [PMID: 38579257 PMCID: PMC11026142 DOI: 10.1093/bioinformatics/btae186] [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: 07/10/2023] [Revised: 03/06/2024] [Accepted: 04/03/2024] [Indexed: 04/07/2024] Open
Abstract
MOTIVATION Spatial transcriptomics has greatly contributed to our understanding of spatial and intra-sample heterogeneity, which could be crucial for deciphering the molecular basis of human diseases. Intra-tumor heterogeneity, e.g. may be associated with cancer treatment responses. However, the lack of computational tools for exploiting cross-regional information and the limited spatial resolution of current technologies present major obstacles to elucidating tissue heterogeneity. RESULTS To address these challenges, we introduce RegionalST, an efficient computational method that enables users to quantify cell type mixture and interactions, identify sub-regions of interest, and perform cross-region cell type-specific differential analysis for the first time. Our simulations and real data applications demonstrate that RegionalST is an efficient tool for visualizing and analyzing diverse spatial transcriptomics data, thereby enabling accurate and flexible exploration of tissue heterogeneity. Overall, RegionalST provides a one-stop destination for researchers seeking to delve deeper into the intricacies of spatial transcriptomics data. AVAILABILITY AND IMPLEMENTATION The implementation of our method is available as an open-source R/Bioconductor package with a user-friendly manual available at https://bioconductor.org/packages/release/bioc/html/RegionalST.html.
Collapse
Affiliation(s)
- Yue Lyu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
- Department of Biostatistics and Data Science, The University of Texas Health Science Center at Houston, Houston, TX 77030, United States
| | - Chong Wu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| | - Wei Sun
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Center, Seattle, WA 98109, United States
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27516, United States
- Department of Biostatistics, University of Washington, Seattle, WA 98195, United States
| | - Ziyi Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, United States
| |
Collapse
|
8
|
Shakartalla SB, Ashmawy NS, Semreen MH, Fayed B, Al Shareef ZM, Jayakumar MN, Ibrahim S, Rahmani M, Hamdy R, Soliman SSM. 1H-NMR metabolomics analysis identifies hypoxanthine as a novel metastasis-associated metabolite in breast cancer. Sci Rep 2024; 14:253. [PMID: 38167685 PMCID: PMC10762038 DOI: 10.1038/s41598-023-50866-y] [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: 09/23/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024] Open
Abstract
Breast cancer is one of the leading causes of death in females, mainly because of metastasis. Oncometabolites, produced via metabolic reprogramming, can influence metastatic signaling cascades. Accordingly, and based on our previous results, we propose that metabolites from highly metastatic breast cancer cells behave differently from less-metastatic cells and may play a significant role in metastasis. For instance, we aim to identify these metabolites and their role in breast cancer metastasis. Less metastatic cells (MCF-7) were treated with metabolites secreted from highly metastatic cells (MDA-MB-231) and the gene expression of three epithelial-to-mesenchymal transition (EMT) markers including E-cadherin, N-cadherin and vimentin were examined. Some metabolites secreted from MDA-MB-231 cells significantly induced EMT activity. Specifically, hypoxanthine demonstrated a significant EMT effect and increased the migration and invasion effects of MCF-7 cells through a hypoxia-associated mechanism. Hypoxanthine exhibited pro-angiogenic effects via increasing the VEGF and PDGF gene expression and affected lipid metabolism by increasing the gene expression of PCSK-9. Notably, knockdown of purine nucleoside phosphorylase, a gene encoding for an important enzyme in the biosynthesis of hypoxanthine, and inhibition of hypoxanthine uptake caused a significant decrease in hypoxanthine-associated EMT effects. Collectively for the first time, hypoxanthine was identified as a novel metastasis-associated metabolite in breast cancer cells and represents a promising target for diagnosis and therapy.
Collapse
Affiliation(s)
- Sarra B Shakartalla
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
- College of Medicine, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
- Faculty of Pharmacy, University of Gezira, P.O. Box. 21111, Wadmedani, Sudan
| | - Naglaa S Ashmawy
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
- Department of Pharmaceutical Sciences, College of Pharmacy, Gulf Medical University, P.O. Box 4184, Ajman, United Arab Emirates
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Abbassia, P.O. Box 11566, Cairo, Egypt
| | - Mohammad H Semreen
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Bahgat Fayed
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
- Chemistry of Natural and Microbial Product Department, National Research Centre, P.O. Box 12622, Cairo, Egypt
| | - Zainab M Al Shareef
- College of Medicine, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Manju N Jayakumar
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
| | - Saleh Ibrahim
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
| | - Mohamed Rahmani
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
- Center for Biotechnology, Khalifa University, Abu Dhabi, United Arab Emirates
- College of Medicine and Health Sciences, Khalifa University, P.O. Box 127788, Abu Dhabi, United Arab Emirates
| | - Rania Hamdy
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates
- Faculty of Pharmacy, Zagazig University, P.O. Box 44519, Zagazig, Egypt
| | - Sameh S M Soliman
- Research Institute for Medical and Health Sciences, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, P.O. Box 27272, Sharjah, United Arab Emirates.
| |
Collapse
|
9
|
Zhang X, Cheng F, Song X, Wang P, Tian S, Zhao X, Wang Q, Zhang M. Superb microvascular imaging for evaluation of microvascularity in breast nodules compared with conventional Doppler imaging. Quant Imaging Med Surg 2023; 13:7029-7040. [PMID: 37869333 PMCID: PMC10585493 DOI: 10.21037/qims-23-136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 09/04/2023] [Indexed: 10/24/2023]
Abstract
Background Neovascularity visualization in breast nodules is challenging due to the limitations of conventional Doppler imaging methods. This study aims to assess the performance of superb microvascular imaging (SMI) in evaluating the microvascularity of breast nodules (diameter ≤2 cm). The comparison of performances of SMI with color Doppler flow imaging (CDFI) and power Doppler imaging (PDI) was made by using a three-factor scoring system of vascularity. This study also investigated the common features of microvascularity in small malignant nodules on SMI for early differentiating from benign nodules. Methods Ninety-one female patients (with 125 breast nodules) were enrolled in this retrospective study. All the breast nodules were examined by grayscale ultrasonography (US), CDFI, PDI, and SMI. The number, morphologic features, and distribution of blood vessels were scored to evaluate the nodular vascularity in light of the three-factor scoring system. The diagnostic value of SMI for microvascularity in breast nodules was analyzed and compared with CDFI and PDI. Results Histological analysis showed 53 malignant and 72 benign nodules. The vascularity grades detected by SMI were significantly different from those of CDFI and PDI (P<0.05). SMI detected 47 grade-IV nodules of the total 125 nodules (37.6%), which was more than those detected by CDFI (10.4%, 13/125) and PDI (12.8%, 16/125), while more grade-I nodules were detected by CDFI (42.4%, 53/125) and PDI (36.8%, 46/125) compared with SMI (21.6%, 27/125). Differences in the vessel number, morphologic features, and distribution between benign and malignant breast nodules were significant on SMI (P<0.05). The vessel number ≥6, penetrating vessels, and a mixed distribution of vessels in peripheral and central nodular tissues were the common features of microvascularity in the grade-IV malignant nodules on SMI, whereas the blood vessels in the benign nodules were straight and branching and peripherally distributed. Conclusions In comparison with CDFI and PDI, SMI enhances microvascularity detection, depicts the microvascular architecture in breast nodules and has potential in the differential diagnosis of malignant nodules from benign nodules.
Collapse
Affiliation(s)
- Xiuwen Zhang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
| | - Fangyuan Cheng
- Department of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, China
- Department of Function Test, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xingjun Song
- Department of Imaging, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Peng Wang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
| | - Shuangyan Tian
- Department of Imaging, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Xiaopei Zhao
- Department of Imaging, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Qing Wang
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, China
- Guangdong Province Engineering Laboratory for Medical Imaging and Diagnostic Technology, Southern Medical University, Guangzhou, China
| | - Mei Zhang
- Department of Radiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Tai’an, China
| |
Collapse
|
10
|
Qi J, Wang C, Ma Y, Wang J, Yang G, Wu Y, Wang H, Mi C. The potential role of combined shear wave elastography and superb microvascular imaging for early prediction the pathological response to neoadjuvant chemotherapy in breast cancer. Front Oncol 2023; 13:1176141. [PMID: 37746288 PMCID: PMC10515084 DOI: 10.3389/fonc.2023.1176141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 08/22/2023] [Indexed: 09/26/2023] Open
Abstract
Objectives The potential role of shear wave elastography (SWE) and superb microvascular imaging (SMI) for early assessment of treatment response to neoadjuvant chemotherapy (NAC) in breast cancer remains unexplored. This study aimed to identify potential factors associated with the pathological response to NAC using these advanced ultrasound techniques. Methods Between August 2021 and October 2022, 68 patients with breast cancer undergoing NAC were recruited. Patients underwent conventional ultrasonography, SMI, and SWE examinations at baseline and post-2nd cycle of NAC. Maximum tumor diameter (Dmax), maximum elastic value (Emax), peak systolic velocity (PSV), and resistance index (RI) at baseline and the rate of change of these parameters post-2nd cycle were recorded. After chemotherapy, all patients underwent surgery. Using the Miller-Payne's grade, patients were categorized into response (grades 3, 4, or 5) and non-response (grades 1 or 2) group. Parameters were compared using t-tests at baseline and post-2nd cycle. Binary logistic regression analysis was used to identify variables and their odds ratios (ORs) related to responses and a prediction model was established. ROC curves were drawn to analyze the efficacy of each parameter and their combined model for early NAC response prediction. Results Among the 68 patients, 15(22.06%) were categorized into the non-response group, whereas 53(77.94%) were categorized into the response group. At baseline, no significant differences were observed between the two groups (p>0.05). Post-2nd cycle of NAC, rates of change of Emax, PSV and RI (ΔEmax, ΔPSV and ΔRI) were higher in responders than non-responders (p<0.05). Binary logistic regression analysis revealed that ΔEmax (OR 0.797 95% CI, 0.683-0.929), ΔPSV (OR 0.926, 95%CI, 0.860-0.998), and ΔRI (OR 0.841, 95%CI, 0.736-0.960) were independently associated with the pathological response of breast cancer after NAC. The combined prediction model exhibited higher accuracy in the early evaluation of the response to NAC (AUC 0.945, 95%CI, 0.873-1.000). Conclusion SWE and SMI techniques enable early identification of tumor characteristics associated with the pathological response to NAC and may be potentially indicative of an effective response. These factors may eventually be used for the early assessment of NAC treatment for clinical management.
Collapse
Affiliation(s)
- Jiaojiao Qi
- Department of Obstetrics and Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Chenyu Wang
- Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yongxin Ma
- Ningxia Medical University, Yinchuan, Ningxia, China
| | - Jiaxing Wang
- Department of Obstetrics and Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Guangfei Yang
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Yating Wu
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Haiyan Wang
- Department of Obstetrics and Gynecology, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Chengrong Mi
- Department of Ultrasound, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| |
Collapse
|
11
|
Adusei S, Ternifi R, Fatemi M, Alizad A. Custom-made flow phantoms for quantitative ultrasound microvessel imaging. ULTRASONICS 2023; 134:107092. [PMID: 37364357 PMCID: PMC10530522 DOI: 10.1016/j.ultras.2023.107092] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 06/28/2023]
Abstract
Morphologically realistic flow phantoms are essential experimental tools for quantitative ultrasound-based microvessel imaging. As new quantitative flow imaging tools are developed, the need for more complex vessel-mimicking phantoms is indisputable. In this article, we propose a method for fabricating phantoms with sub-millimeter channels consisting of branches and curvatures in various shapes and sizes suitable for quantifying vessel morphological features. We used different tissue-mimicking materials (TMMs) compatible with ultrasound imaging as the base and metal wires of different diameters (0.15-1.25 mm) to create wall-less channels. The TMMs used are silicone rubber, plastisol, conventional gelatin, and medical gelatin. Mother channels in these phantoms were made in diameters of 1.25 mm or 0.3 mm and the daughter channels in diameters 0.3 mm or 0.15 mm. Bifurcations were created by soldering wires together at branch points. Quantitative parameters were assessed, and accuracy of measurements from the ground truth were determined. Channel diameters were seen to have increased (76-270%) compared to the initial state in the power Doppler images, partly due to blood mimicking fluid pressure. Amongst the microflow phantoms made from the different TMMs, the medical gelatin phantom was selected as the best option for microflow imaging, fulfilling the objective of being easy to fabricate with high transmittance while having a speed of sound and acoustic attenuation close to human tissue. A flow velocity of 0.85 ± 0.01 mm/s, comparable to physiological flow velocity was observed in the smallest diameter phantom (medical gelatin branch) presented here. We successfully constructed more complex geometries, including tortuous and multibranch channels using the medical gelatin as the TMM. We anticipate this will create new avenues for validating quantitative ultrasound microvessel imaging techniques.
Collapse
Affiliation(s)
- Shaheeda Adusei
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Redouane Ternifi
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Mostafa Fatemi
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA
| | - Azra Alizad
- Department of Radiology, Mayo Clinic College of Medicine and Science, Rochester, MN 55905, USA.
| |
Collapse
|
12
|
Ferroni G, Sabeti S, Abdus-Shakur T, Scalise L, Carter JM, Fazzio RT, Larson NB, Fatemi M, Alizad A. Noninvasive prediction of axillary lymph node breast cancer metastasis using morphometric analysis of nodal tumor microvessels in a contrast-free ultrasound approach. Breast Cancer Res 2023; 25:65. [PMID: 37296471 PMCID: PMC10257266 DOI: 10.1186/s13058-023-01670-z] [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: 02/19/2023] [Accepted: 06/02/2023] [Indexed: 06/12/2023] Open
Abstract
PURPOSE Changes in microcirculation of axillary lymph nodes (ALNs) may indicate metastasis. Reliable noninvasive imaging technique to quantify such variations is lacking. We aim to develop and investigate a contrast-free ultrasound quantitative microvasculature imaging technique for detection of metastatic ALN in vivo. EXPERIMENTAL DESIGN The proposed ultrasound-based technique, high-definition microvasculature imaging (HDMI) provides superb images of tumor microvasculature at sub-millimeter size scales and enables quantitative analysis of microvessels structures. We evaluated the new HDMI technique on 68 breast cancer patients with ultrasound-identified suspicious ipsilateral axillary lymph nodes recommended for fine needle aspiration biopsy (FNAB). HDMI was conducted before the FNAB and vessel morphological features were extracted, analyzed, and the results were correlated with the histopathology. RESULTS Out of 15 evaluated quantitative HDMI biomarkers, 11 were significantly different in metastatic and reactive ALNs (10 with P << 0.01 and one with 0.01 < P < 0.05). We further showed that through analysis of these biomarkers, a predictive model trained on HDMI biomarkers combined with clinical information (i.e., age, node size, cortical thickness, and BI-RADS score) could identify metastatic lymph nodes with an area under the curve of 0.9 (95% CI [0.82,0.98]), sensitivity of 90%, and specificity of 88%. CONCLUSIONS The promising results of our morphometric analysis of HDMI on ALNs offer a new means of detecting lymph node metastasis when used as a complementary imaging tool to conventional ultrasound. The fact that it does not require injection of contrast agents simplifies its use in routine clinical practice.
Collapse
Affiliation(s)
- Giulia Ferroni
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Soroosh Sabeti
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Tasneem Abdus-Shakur
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 1st. St. SW, Rochester, MN, 55905, USA
| | - Lorenzo Scalise
- Department of Industrial Engineering and Mathematical Science, Marche Polytechnic University, 60131, Ancona, Italy
| | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, AB, Canada
| | - Robert T Fazzio
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 1st. St. SW, Rochester, MN, 55905, USA
| | - Nicholas B Larson
- Department of Quantitative Health Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Mostafa Fatemi
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA
| | - Azra Alizad
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, 55905, USA.
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 1st. St. SW, Rochester, MN, 55905, USA.
| |
Collapse
|
13
|
Lin Y, Zhao Y, Chen M, Li Z, Liu Q, Chen J, Ding Y, Ding C, Ding Y, Qi C, Zheng L, Li J, Zhang R, Zhou J, Wang L, Zhang QQ. CYD0281, a Bcl-2 BH4 domain antagonist, inhibits tumor angiogenesis and breast cancer tumor growth. BMC Cancer 2023; 23:479. [PMID: 37237269 DOI: 10.1186/s12885-023-10974-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND B-cell lymphoma 2 (Bcl-2) family proteins are key regulators of apoptosis, which possess four conserved Bcl-2 homologies (BH) domains. Among the BH domains, the BH3 domain is considered as a potent 'death domain' while the BH4 domain is required for anti-apoptotic activity. Bcl-2 can be converted to a pro-apoptotic molecule through the removal or mutation of the BH4 domain. Bcl-2 is considered as an inducer of angiogenesis, which can promote tumor vascular network formation and further afford nutrients and oxygen to promote tumor progression. However, whether disrupting the function of the BH4 domain to convert Bcl-2 into a pro-apoptotic molecule could make Bcl-2 possess the potential for anti-angiogenic therapy remains to be defined. METHODS CYD0281 was designed and synthesized according to the lead structure of BDA-366, and its function on inducing a conformational change of Bcl-2 was further evaluated via immunoprecipitation (IP) and immunofluorescence (IF) assays. Moreover, the function of CYD0281 on apoptosis of endothelial cells was analyzed via cell viability, flow cytometry, and western blotting assays. Additionally, the role of CYD0281 on angiogenesis in vitro was determined via endothelial cell migration and tube formation assays and rat aortic ring assay. Chick embryo chorioallantoic membrane (CAM) and yolk sac membrane (YSM) models, breast cancer cell xenograft tumor on CAM and in mouse models as well as the Matrigel plug angiogenesis assay were used to explore the effects of CYD0281 on angiogenesis in vivo. RESULTS We identified a novel potent small-molecule Bcl-2-BH4 domain antagonist, CYD0281, which exhibited significant anti-angiogenic effects both in vitro and in vivo, and further inhibited breast cancer tumor growth. CYD0281 was found to induce conformational changes in Bcl-2 through the exposure of the BH3 domain and convert Bcl-2 from an anti-apoptotic molecule into a cell death inducer, thereby resulting in the apoptosis of vascular endothelial cells. CONCLUSIONS This study has revealed CYD0281 as a novel Bcl-2-BH4 antagonist that induces conformational changes of Bcl-2 to convert to a pro-apoptotic molecule. Our findings indicate that CYD0281 plays a crucial role in anti-angiogenesis and may be further developed as a potential anti-tumor drug candidate for breast cancer. This work also provides a potential anti-angiogenic strategy for breast cancer treatment.
Collapse
Affiliation(s)
- Yihua Lin
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yiling Zhao
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Minggui Chen
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Zishuo Li
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Qiao Liu
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jian Chen
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Yi Ding
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Chunyong Ding
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Ye Ding
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Cuiling Qi
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Lingyun Zheng
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jiangchao Li
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Rongxin Zhang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Jia Zhou
- Chemical Biology Program, Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| | - Lijing Wang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Qian-Qian Zhang
- School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| |
Collapse
|
14
|
Shibabaw T, Teferi B, Ayelign B. The role of Th-17 cells and IL-17 in the metastatic spread of breast cancer: As a means of prognosis and therapeutic target. Front Immunol 2023; 14:1094823. [PMID: 36993955 PMCID: PMC10040566 DOI: 10.3389/fimmu.2023.1094823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/21/2023] [Indexed: 03/14/2023] Open
Abstract
Metastatic breast cancer is one of the most common and well-known causes of death for women worldwide. The inflammatory tumor cell and other cancer hallmarks dictate the metastatic form and dissemination of breast cancer. Taking these into account, from various components of the tumor microenvironment, a pro-inflammatory infiltrative cell known as Th-17 plays an immense role in breast cancer proliferation, invasiveness, and metastasis. It has been demonstrated that IL-17, a pleiotropic pro-inflammatory cytokine generated by Th-17, is upregulated in a metastatic form of breast cancer. Recent research updates stated that chronic inflammation and mediators like cytokines and chemokines are causative hallmarks in many human cancers, including breast cancer. Therefore, IL-17 and its multiple downward signaling molecules are the centers of research attention to develop potent treatment options for cancer. They provide information on the role of IL-17-activated MAPK, which results in tumor cell proliferation and metastasis via NF-kB-mediated expression of MMP signaling. Overall, this review article emphasizes IL-17A and its intermediate signaling molecules, such as ERK1/2, NF-kB, MMPs, and VEGF, as potential molecular targets for the prevention and treatment of breast cancer.
Collapse
Affiliation(s)
- Tewodros Shibabaw
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Banchamlak Teferi
- Department of Clinical Pharmacy, School of Pharmacy, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Birhanu Ayelign
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Science, College of Medicine and Health Science, University of Gondar, Gondar, Ethiopia
- Research School of Biology, College of Science, Australian National University, Canberra, ACT, Australia
- *Correspondence: Birhanu Ayelign,
| |
Collapse
|
15
|
Ternifi R, Wang Y, Gu J, Polley EC, Carter JM, Pruthi S, Boughey JC, Fazzio RT, Fatemi M, Alizad A. Ultrasound high-definition microvasculature imaging with novel quantitative biomarkers improves breast cancer detection accuracy. Eur Radiol 2022; 32:7448-7462. [PMID: 35486168 PMCID: PMC9616967 DOI: 10.1007/s00330-022-08815-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/24/2022] [Accepted: 04/12/2022] [Indexed: 01/03/2023]
Abstract
OBJECTIVES To overcome the limitations of power Doppler in imaging angiogenesis, we sought to develop and investigate new quantitative biomarkers of a contrast-free ultrasound microvasculature imaging technique for differentiation of benign from malignant pathologies of breast lesion. METHODS In this prospective study, a new high-definition microvasculature imaging (HDMI) was tested on 521 patients with 527 ultrasound-identified suspicious breast masses indicated for biopsy. Four new morphological features of tumor microvessels, microvessel fractal dimension (mvFD), Murray's deviation (MD), bifurcation angle (BA), and spatial vascularity pattern (SVP) as well as initial biomarkers were extracted and analyzed, and the results correlated with pathology. Multivariable logistic regression analysis was used to study the performance of different prediction models, initial biomarkers, new biomarkers, and combined new and initial biomarkers in differentiating benign from malignant lesions. RESULTS The new HDMI biomarkers, mvFD, BA, MD, and SVP, were statistically significantly different in malignant and benign lesions, regardless of tumor size. Sensitivity and specificity of the new biomarkers in lesions > 20 mm were 95.6% and 100%, respectively. Combining the new and initial biomarkers together showed an AUC, sensitivity, and specificity of 97% (95% CI: 95-98%), 93.8%, and 89.2%, respectively, for all lesions regardless of mass size. The classification was further improved by adding the Breast Imaging Reporting and Data System (BI-RADS) score to the prediction model, showing an AUC, sensitivity, and specificity of 97% (95% CI: 95-98%), 93.8%, and 89.2%, respectively. CONCLUSION The addition of new quantitative HDMI biomarkers significantly improved the accuracy in breast lesion characterization when used as a complementary imaging tool to the conventional ultrasound. KEY POINTS • Novel quantitative biomarkers extracted from tumor microvessel images increase the sensitivity and specificity in discriminating malignant from benign breast masses. • New HDMI biomarkers Murray's deviation, bifurcation angles, microvessel fractal dimension, and spatial vascularity pattern outperformed the initial biomarkers. • The addition of BI-RADS scores based on US descriptors to the multivariable analysis using all biomarkers remarkably increased the sensitivity, specificity, and AUC in all size groups.
Collapse
Affiliation(s)
- Redouane Ternifi
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Yinong Wang
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Juanjuan Gu
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Eric C Polley
- Department of Health Science, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Jodi M Carter
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Sandhya Pruthi
- Department of Medicine, Mayo Clinic College of Medicine, Rochester, MN, USA
| | - Judy C Boughey
- Department of Surgery, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Robert T Fazzio
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 1st Street SW, Rochester, MN, 55905, USA
| | - Mostafa Fatemi
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Azra Alizad
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine and Science, Rochester, MN, USA.
- Department of Radiology, Mayo Clinic College of Medicine and Science, 200 1st Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
16
|
Manjunath M, Swaroop S, Pradhan SS, Rao K R, Mahadeva R, Sivaramakrishnan V, Choudhary B. Integrated Transcriptome and Metabolomic Analysis Reveal Anti-Angiogenic Properties of Disarib, a Novel Bcl2-Specific Inhibitor. Genes (Basel) 2022; 13:genes13071208. [PMID: 35885991 PMCID: PMC9316176 DOI: 10.3390/genes13071208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 06/25/2022] [Accepted: 06/29/2022] [Indexed: 12/04/2022] Open
Abstract
Transcriptomic profiling of several drugs in cancer cell lines has been utilised to obtain drug-specific signatures and guided combination therapy to combat drug resistance and toxicity. Global metabolomics reflects changes due to altered activity of enzymes, environmental factors, etc. Integrating transcriptomics and metabolomics can provide genotype-phenotype correlation, providing meaningful insights into alterations in gene expression and its outcome to understand differential metabolism and guide therapy. This study uses a multi-omics approach to understand the global gene expression and metabolite changes induced by Disarib, a novel Bcl2-specific inhibitor in the Ehrlich adenocarcinoma (EAC) breast cancer mouse model. RNAseq analysis was performed on EAC mouse tumours treated with Disarib and compared to the controls. The expression of 6 oncogenes and 101 tumour suppressor genes interacting with Bcl2 and Bak were modulated upon Disarib treatment. Cancer hallmark pathways like DNA repair, Cell cycle, angiogenesis, and mitochondrial metabolism were downregulated, and programmed cell death platelet-related pathways were upregulated. Global metabolomic profiling using LC-MS revealed that Oncometabolites like carnitine, oleic acid, glycine, and arginine were elevated in tumour mice compared to normal and were downregulated upon Disarib treatment. Integrated transcriptomic and metabolomic profiles identified arginine metabolism, histidine, and purine metabolism to be altered upon Disarib treatment. Pro-angiogenic metabolites, arginine, palmitic acid, oleic acid, and myristoleic acid were downregulated in Disarib-treated mice. We further validated the effect of Disarib on angiogenesis by qRT-PCR analysis of genes in the VEGF pathway. Disarib treatment led to the downregulation of pro-angiogenic markers. Furthermore, the chorioallantoic membrane assay displayed a reduction in the formation of the number of secondary blood vessels upon Disarib treatment. Disarib reduces tumours by reducing oncometabolite and activating apoptosis and downregulating angiogenesis.
Collapse
Affiliation(s)
- Meghana Manjunath
- Institute of Bioinformatics and Applied Biotechnology, Bengaluru 560100, Karnataka, India; (M.M.); (R.R.K.); (R.M.)
- Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Sai Swaroop
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur 515001, Andhra Pradesh, India; (S.S.); (S.S.P.); (V.S.)
| | - Sai Sanwid Pradhan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur 515001, Andhra Pradesh, India; (S.S.); (S.S.P.); (V.S.)
| | - Raksha Rao K
- Institute of Bioinformatics and Applied Biotechnology, Bengaluru 560100, Karnataka, India; (M.M.); (R.R.K.); (R.M.)
| | - Raghunandan Mahadeva
- Institute of Bioinformatics and Applied Biotechnology, Bengaluru 560100, Karnataka, India; (M.M.); (R.R.K.); (R.M.)
| | - Venketesh Sivaramakrishnan
- Disease Biology Lab, Department of Biosciences, Sri Sathya Sai Institute of Higher Learning, Anantapur 515001, Andhra Pradesh, India; (S.S.); (S.S.P.); (V.S.)
| | - Bibha Choudhary
- Institute of Bioinformatics and Applied Biotechnology, Bengaluru 560100, Karnataka, India; (M.M.); (R.R.K.); (R.M.)
- Correspondence:
| |
Collapse
|
17
|
Shi P, Liu Y, Yang H, Hu B. Breast cancer derived exosomes promoted angiogenesis of endothelial cells in microenvironment via circHIPK3/miR-124-3p/MTDH axis. Cell Signal 2022; 95:110338. [PMID: 35460835 DOI: 10.1016/j.cellsig.2022.110338] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 04/12/2022] [Accepted: 04/17/2022] [Indexed: 11/03/2022]
Abstract
Circular RNAs (circRNAs) are important contents in exosomes, which can regulate peripheral cell functions, thus influencing the tumor microenvironment. This work investigated the mechanisms underlying the angiogenesis in peripheral human endothelial cells (ECs) mediated by the breast cancer (BC) cells derived exosomal circRNAs and aimed to explore the biomarkers for the anti-angiogenesis therapy for BC.The BC cell derived exosomes were extracted and the expression level and the circular formation of HIPK3 enclosed was determined. To examine the impact of this exosomal circRNA on ECs, cell viability and tube formation were determined in recipient cells co-cultured with exosomes or transfected with circHIPK3 and the related controls. Target microRNAs (miRNAs) for circHIPK3 and target genes for miRNAs were predicted and confirmed by multiple assays like dual luciferase reporter assay, western blot, and qPCR assays. The existence of the circHIPK3/miR-124-3p/MTDH axis were further confirmed with rescue experiment in mice xenograft model.HIPK3s were mainly in forms of circRNAs and were highly expressed in the BC cell derived exosomes, which could be absorbed by the recipient ECs. The cell viability and angiogenesis in ECs were enhanced when treated with circHIPK3s and decreased when treated with circHIPK3-si. Furthermore, MTDH was proved to be the responsible gene in this process which was regulated by miR-124-3p, the local miRNA sponged by the exosomal circHIPK3.circHIPK3 enclosed in the BC cell-derived exosomes enhanced MTDH expression in the endothelial cell by sponging miR-124-3p, favoring the tube formation in ECs, which might serve as a therapeutic target for anti-angiogenesis therapy for breast cancer.
Collapse
Affiliation(s)
- Pengfei Shi
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430013, People's Republic of China
| | - Yongjun Liu
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430013, People's Republic of China
| | - Hua Yang
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430013, People's Republic of China.
| | - Bo Hu
- Department of Thyroid and Breast Surgery, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430013, People's Republic of China.
| |
Collapse
|
18
|
Sadiq M. Modeling survival response using a parametric approach in the presence of multicollinearity. COMMUN STAT-SIMUL C 2022. [DOI: 10.1080/03610918.2022.2060509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- Maryam Sadiq
- Department of Statistics, University of Azad Jammu and Kashmir, Muzaffarabad, Pakistan
| |
Collapse
|
19
|
Ayoub NM, Jaradat SK, Al-Shami KM, Alkhalifa AE. Targeting Angiogenesis in Breast Cancer: Current Evidence and Future Perspectives of Novel Anti-Angiogenic Approaches. Front Pharmacol 2022; 13:838133. [PMID: 35281942 PMCID: PMC8913593 DOI: 10.3389/fphar.2022.838133] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Angiogenesis is a vital process for the growth and dissemination of solid cancers. Numerous molecular pathways are known to drive angiogenic switch in cancer cells promoting the growth of new blood vessels and increased incidence of distant metastasis. Several angiogenesis inhibitors are clinically available for the treatment of different types of advanced solid cancers. These inhibitors mostly belong to monoclonal antibodies or small-molecule tyrosine kinase inhibitors targeting the classical vascular endothelial growth factor (VEGF) and its receptors. Nevertheless, breast cancer is one example of solid tumors that had constantly failed to respond to angiogenesis inhibitors in terms of improved survival outcomes of patients. Accordingly, it is of paramount importance to assess the molecular mechanisms driving angiogenic signaling in breast cancer to explore suitable drug targets that can be further investigated in preclinical and clinical settings. This review summarizes the current evidence for the effect of clinically available anti-angiogenic drugs in breast cancer treatment. Further, major mechanisms associated with intrinsic or acquired resistance to anti-VEGF therapy are discussed. The review also describes evidence from preclinical and clinical studies on targeting novel non-VEGF angiogenic pathways in breast cancer and several approaches to the normalization of tumor vasculature by targeting pericytes, utilization of microRNAs and extracellular tumor-associate vesicles, using immunotherapeutic drugs, and nanotechnology.
Collapse
Affiliation(s)
- Nehad M. Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
- *Correspondence: Nehad M. Ayoub,
| | - Sara K. Jaradat
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| | - Kamal M. Al-Shami
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Amer E. Alkhalifa
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology (JUST), Irbid, Jordan
| |
Collapse
|
20
|
Khoshbakht S, Mokhtari M, Moravveji SS, Azimzadeh Jamalkandi S, Masoudi-Nejad A. Re-wiring and gene expression changes of AC025034.1 and ATP2B1 play complex roles in early-to-late breast cancer progression. BMC Genom Data 2022; 23:6. [PMID: 35031021 PMCID: PMC8759272 DOI: 10.1186/s12863-021-01015-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/17/2021] [Indexed: 12/24/2022] Open
Abstract
Background Elucidating the dynamic topological changes across different stages of breast cancer, called stage re-wiring, could lead to identifying key latent regulatory signatures involved in cancer progression. Such dynamic regulators and their functions are mostly unknown. Here, we reconstructed differential co-expression networks for four stages of breast cancer to assess the dynamic patterns of cancer progression. A new computational approach was applied to identify stage-specific subnetworks for each stage. Next, prognostic traits of genes and the efficiency of stage-related groups were evaluated and validated, using the Log-Rank test, SVM classifier, and sample clustering. Furthermore, by conducting the stepwise VIF-feature selection method, a Cox-PH model was developed to predict patients’ risk. Finally, the re-wiring network for prognostic signatures was reconstructed and assessed across stages to detect gain/loss, positive/negative interactions as well as rewired-hub nodes contributing to dynamic cancer progression. Results After having implemented our new approach, we could identify four stage-specific core biological pathways. We could also detect an essential non-coding RNA, AC025034.1, which is not the only antisense to ATP2B1 (cell proliferation regulator), but also revealed a statistically significant stage-descending pattern; Moreover, AC025034.1 revealed both a dynamic topological pattern across stages and prognostic trait. We also identified a high-performance Overall-Survival-Risk model, including 12 re-wired genes to predict patients’ risk (c-index = 0.89). Finally, breast cancer-specific prognostic biomarkers of LINC01612, AC092142.1, and AC008969.1 were identified. Conclusions In summary new scoring method highlighted stage-specific core pathways for early-to-late progressions. Moreover, detecting the significant re-wired hub nodes indicated stage-associated traits, which reflects the importance of such regulators from different perspectives. Supplementary Information The online version contains supplementary material available at 10.1186/s12863-021-01015-9.
Collapse
Affiliation(s)
- Samane Khoshbakht
- Laboratory of Systems Biology and Bioinformatics (LBB), Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran
| | - Majid Mokhtari
- Laboratory of Systems Biology and Bioinformatics (LBB), Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran
| | - Sayyed Sajjad Moravveji
- Laboratory of Systems Biology and Bioinformatics (LBB), Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran
| | | | - Ali Masoudi-Nejad
- Laboratory of Systems Biology and Bioinformatics (LBB), Department of Bioinformatics, Kish International Campus, University of Tehran, Kish Island, Iran. .,Laboratory of Systems Biology and Bioinformatics (LBB), Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran.
| |
Collapse
|
21
|
Ab Mumin N, Ramli Hamid MT, Wong JHD, Rahmat K, Ng KH. Magnetic Resonance Imaging Phenotypes of Breast Cancer Molecular Subtypes: A Systematic Review. Acad Radiol 2022; 29 Suppl 1:S89-S106. [PMID: 34481705 DOI: 10.1016/j.acra.2021.07.017] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/14/2021] [Accepted: 07/20/2021] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Magnetic resonance imaging (MRI) is the most sensitive imaging modality in detecting breast cancer. The purpose of this systematic review is to investigate the role of human extracted MRI phenotypes in classifying molecular subtypes of breast cancer. METHODS We performed a literature search of published articles on the application of MRI phenotypic features in invasive breast cancer molecular subtype classifications by radiologists' interpretation on Medline Complete, Pubmed, and Google scholar from 1st January 2000 to 31st March 2021. Of the 1453 literature identified, 42 fulfilled the inclusion criteria. RESULTS All studies were case-controlled, retrospective study and research-based. The majority of the studies assessed the MRI features using American College of Radiology- Breast Imaging Reporting and Data System (ACR-BIRADS) classification and using dynamic contrast-enhanced (DCE) kinetic features, Apparent Diffusion Coefficient (ADC) values, and T2 sequence. Most studies divided invasive breast cancer into 4 main subtypes, luminal A, luminal B, HER2, and triple-negative (TN) cancers, and used 2 readers. We present a summary of the radiologists' extracted breast MRI phenotypical features and their correlating breast cancer subtypes classifications. The characteristic features are morphology, enhancement kinetics, and T2 signal intensity. We found that the TN subtype has the most distinctive MRI features compared to the other subtypes and luminal A and B have many similar features. CONCLUSION The MRI features which are predictive of each subtype are the morphology, internal enhancement features, and T2 signal intensity, predominantly between TN and the rest. Radiologists' visual interpretation of some of MRI features may offer insight into the respective invasive breast cancer molecular subtype. However, current evidence are still limited to "suggestive" features instead of a diagnostic standard. Further research is recommended to explore this potential application, for example, by augmentation of radiologists' visual interpretation by artificial intelligence.
Collapse
|
22
|
Golhani V, Ray SK, Mukherjee S. Role of MicroRNAs and Long Non-Coding RNAs in Regulating Angiogenesis in Human Breast Cancer- A Molecular Medicine Perspective. Curr Mol Med 2021; 22:882-893. [PMID: 34923940 DOI: 10.2174/1566524022666211217114527] [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: 04/07/2021] [Revised: 10/26/2021] [Accepted: 11/05/2021] [Indexed: 11/22/2022]
Abstract
MicroRNAs (miRNAs) and long non-coding RNAs (lncRNAs) are proficient in regulating gene expression post-transcriptionally. Considering the recent trend in exploiting non-coding RNAs (ncRNAs) as cancer therapeutics, the potential use of miRNAs and lncRNAs as biomarkers and novel therapeutic agents against angiogenesis is an important scientific aspect. An estimated 70% of the genome is actively transcribed, only 2% of which codes for known protein-coding genes. Long noncoding RNAs (lncRNAs) are a large and diverse class of RNAs > 200 nucleotides in length, and not translated into protein, and are of utmost importance and it governs the expression of genes in a temporal, spatial, and cell context-dependent manner. Angiogenesis is an essential process for organ morphogenesis and growth during development, and it is relevant during the repair of wounded tissue in adults. It is coordinated by an equilibrium of pro-and anti-angiogenic factors; nevertheless, when affected, it promotes several diseases, including breast cancer. Signaling pathways involved here are tightly controlled systems that regulate the appropriate timing of gene expression required for the differentiation of cells down a particular lineage essential for proper tissue development. Lately, scientific reports are indicating that ncRNAs, such as miRNAs, and lncRNAs, play critical roles in angiogenesis related to breast cancer. The specific roles of various miRNAs and lncRNAs in regulating angiogenesis in breast cancer, with particular focus on the downstream targets and signaling pathways regulated by these ncRNAs with molecular medicine perspective, are highlighted in this write-up.
Collapse
Affiliation(s)
- Vandana Golhani
- Department of Biochemistry. All India Institute of Medical Sciences. Bhopal, Madhya Pradesh-462020, India
| | | | - Sukhes Mukherjee
- Department of Biochemistry. All India Institute of Medical Sciences. Bhopal, Madhya Pradesh-462020, India
| |
Collapse
|
23
|
Ternifi R, Wang Y, Polley EC, Fazzio RT, Fatemi M, Alizad A. Quantitative Biomarkers for Cancer Detection Using Contrast-Free Ultrasound High-Definition Microvessel Imaging: Fractal Dimension, Murray's Deviation, Bifurcation Angle & Spatial Vascularity Pattern. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:3891-3900. [PMID: 34329160 PMCID: PMC8668387 DOI: 10.1109/tmi.2021.3101669] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
A growing body of evidence indicates that there is a strong correlation between microvascular morphological features and malignant tumors. Therefore, quantification of these features might allow more accurate differentiation of benign and malignant tumors. The main objective of this research project is to improve the quantification of microvascular networks depicted in contrast-free ultrasound microvessel images. To achieve this goal, a new series of quantitative microvessel morphological parameters are introduced for differentiation of breast masses using contrast-free ultrasound-based high-definition microvessel imaging (HDMI). Using HDMI, we quantified and analyzed four new parameters: 1) microvessel fractal dimension (mvFD), a marker of tumor microvascular complexity; 2) Murray's deviation (MD), the diameter mismatch, defined as the deviation from Murray's law; 3) bifurcation angle (BA), abnormally decreased angle; and 4) spatial vascular pattern (SVP), indicating tumor vascular distribution pattern, either intratumoral or peritumoral. The new biomarkers have been tested on 60 patients with breast masses. Validation of the feature's extraction algorithm was performed using a synthetic data set. All the proposed parameters had the power to discriminate the breast lesion malignancy (p < 0.05), displaying BA as the most sensitive test, with a sensitivity of 90.6%, and mvFD as the most specific test, with a specificity of 92%. The results of all four new biomarkers showed an AUC = 0.889, sensitivity of 80% and specificity of 91.4% In conclusion, the added value of the proposed quantitative morphological parameters, as new biomarkers of angiogenesis within breast masses, paves the way for more accurate breast cancer detection with higher specificity.
Collapse
|
24
|
Multi-Aspect Optoacoustic Imaging of Breast Tumors under Chemotherapy with Exogenous and Endogenous Contrasts: Focus on Apoptosis and Hypoxia. Biomedicines 2021; 9:biomedicines9111696. [PMID: 34829925 PMCID: PMC8615838 DOI: 10.3390/biomedicines9111696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 12/01/2022] Open
Abstract
Breast cancer is a complex tumor type involving many biological processes. Most chemotherapeutic agents exert their antitumoral effects by rapid induction of apoptosis. Another main feature of breast cancer is hypoxia, which may drive malignant progression and confer resistance to various forms of therapy. Thus, multi-aspect imaging of both tumor apoptosis and oxygenation in vivo would be of enormous value for the effective evaluation of therapy response. Herein, we demonstrate the capability of a hybrid imaging modality known as multispectral optoacoustic tomography (MSOT) to provide high-resolution, simultaneous imaging of tumor apoptosis and oxygenation, based on both the exogenous contrast of an apoptosis-targeting dye and the endogenous contrast of hemoglobin. MSOT imaging was applied on mice bearing orthotopic 4T1 breast tumors before and following treatment with doxorubicin. Apoptosis was monitored over time by imaging the distribution of xPLORE-APOFL750©, a highly sensitive poly-caspase binding apoptotic probe, within the tumors. Oxygenation was monitored by tracking the distribution of oxy- and deoxygenated hemoglobin within the same tumor areas. Doxorubicin treatment induced an increase in apoptosis-depending optoacoustic signal of xPLORE-APOFL750© at 24 h after treatment. Furthermore, our results showed spatial correspondence between xPLORE-APO750© and deoxygenated hemoglobin. In vivo apoptotic status of the tumor tissue was independently verified by ex vivo fluorescence analysis. Overall, our results provide a rationale for the use of MSOT as an effective tool for simultaneously investigating various aspects of tumor pathophysiology and potential effects of therapeutic regimes based on both endogenous and exogenous molecular contrasts.
Collapse
|
25
|
Herrera-Vargas AK, García-Rodríguez E, Olea-Flores M, Mendoza-Catalán MA, Flores-Alfaro E, Navarro-Tito N. Pro-angiogenic activity and vasculogenic mimicry in the tumor microenvironment by leptin in cancer. Cytokine Growth Factor Rev 2021; 62:23-41. [PMID: 34736827 DOI: 10.1016/j.cytogfr.2021.10.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/18/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022]
Abstract
The acquired ability to induce the formation of a functional vasculature is a hallmark of cancer. Blood vessels in tumors are formed through various mechanisms, among the most important in cancer biology, angiogenesis, and vasculogenic mimicry have been described. Leptin is one of the main adipokines secreted by adipocytes in normal breast tissue and the tumor microenvironment. Here, we provide information on the relationship between leptin and the development of angiogenesis and vasculogenic mimicry in different types of cancer. Here, we report that leptin activates different pathways such as JAK-STAT3, MAPK/ERK, PKC, JNK, p38, and PI3K-Akt to induce the expression of various angiogenic factors and vasculogenic mimicry. In vivo models, leptin induces blood vessel formation through the PI3K-Akt-mTOR pathway. Interestingly, the relationship between leptin and vasculogenic mimicry was more significant in breast cancer. The information obtained suggests that leptin could be playing an essential role in tumor survival and metastasis through the induction of vascular mechanisms such as angiogenesis and vasculogenic mimicry; thus, leptin-induced pathways could be suggested as a promising therapeutic target.
Collapse
Affiliation(s)
- Ana K Herrera-Vargas
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| | - Eduardo García-Rodríguez
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| | - Monserrat Olea-Flores
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| | - Miguel A Mendoza-Catalán
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, GRO, 39090, Mexico.
| | - Eugenia Flores-Alfaro
- Laboratorio de Epidemiología Clínica y Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, GRO 39087, Mexico.
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Guerrero, Av. Lázaro Cárdenas s/n, Chilpancingo, GRO 39090, Mexico.
| |
Collapse
|
26
|
Li JJ, Tsang JY, Tse GM. Tumor Microenvironment in Breast Cancer-Updates on Therapeutic Implications and Pathologic Assessment. Cancers (Basel) 2021; 13:cancers13164233. [PMID: 34439387 PMCID: PMC8394502 DOI: 10.3390/cancers13164233] [Citation(s) in RCA: 99] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/15/2021] [Accepted: 08/16/2021] [Indexed: 12/16/2022] Open
Abstract
The tumor microenvironment (TME) in breast cancer comprises local factors, cancer cells, immune cells and stromal cells of the local and distant tissues. The interaction between cancer cells and their microenvironment plays important roles in tumor proliferation, propagation and response to therapies. There is increasing research in exploring and manipulating the non-cancerous components of the TME for breast cancer treatment. As the TME is now increasingly recognized as a treatment target, its pathologic assessment has become a critical component of breast cancer management. The latest WHO classification of tumors of the breast listed stromal response pattern/fibrotic focus as a prognostic factor and includes recommendations on the assessment of tumor infiltrating lymphocytes and PD-1/PD-L1 expression, with therapeutic implications. This review dissects the TME of breast cancer, describes pathologic assessment relevant for prognostication and treatment decision, and details therapeutic options that interacts with and/or exploits the TME in breast cancer.
Collapse
Affiliation(s)
| | | | - Gary M. Tse
- Correspondence: ; Tel.: 852-3505-2359; Fax: 852-2637-4858
| |
Collapse
|
27
|
Hsu MY, Hsieh CH, Huang YT, Chu SY, Chen CM, Lee WJ, Liu SJ. Enhanced Paclitaxel Efficacy to Suppress Triple-Negative Breast Cancer Progression Using Metronomic Chemotherapy with a Controlled Release System of Electrospun Poly-d-l-Lactide-Co-Glycolide (PLGA) Nanofibers. Cancers (Basel) 2021; 13:cancers13133350. [PMID: 34283075 PMCID: PMC8268060 DOI: 10.3390/cancers13133350] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/24/2022] Open
Abstract
Simple Summary Treatment of metastatic triple-negative breast cancer (TNBC) relies on chemotherapy. To improve the efficacy of chemotherapy and avoid systemic toxicity, metronomic chemotherapy using continuous administration of low-dose chemotherapy could be a solution. The paclitaxel-loaded PLGA nanofibers allow for continuous and prolonged drug release, which is compatible with the concept of metronomic chemotherapy. The animal study revealed that the strategy successfully inhibited the growth of the primary tumor and distant metastasis without sarcopenia. These data offer new insights into the role of drug-loaded nanofibers in the treatment of metastatic TNBC. Abstract Triple-negative breast cancer (TNBC) is highly aggressive and responds poorly to conventional chemotherapy. The challenge of TNBC therapy is to maximize the efficacies of conventional chemotherapeutic agents and reduce their toxicities. Metronomic chemotherapy using continuous low-dose chemotherapy has been proposed as a new treatment option, but this approach is limited by the selection of drugs. To improve antitumor therapeutic effects, we developed electrospun paclitaxel-loaded poly-d-l-lactide-co-glycolide (PLGA) nanofibers as a topical implantable delivery device for controlled drug release and site-specific treatment. The subcutaneously implanted paclitaxel-loaded nanofibrous membrane in mice was compatible with the concept of metronomic chemotherapy; it significantly enhanced antitumor activity, inhibited local tumor growth, constrained distant metastasis, and prolonged survival compared with intraperitoneal paclitaxel injection. Furthermore, under paclitaxel-loaded nanofiber treatment, systemic toxicity was low with a persistent increase in lean body weight in mice; in contrast, body weight decreased in other groups. The paclitaxel-loaded nanofibrous membranes provided sustained drug release and site-specific treatment by directly targeting and changing the tumor microenvironment, resulting in low systemic toxicity and a significant improvement in the therapeutic effect and safety compared with conventional chemotherapy. Thus, metronomic chemotherapy with paclitaxel-loaded nanofibrous membranes offers a promising strategy for the treatment of TNBC.
Collapse
Affiliation(s)
- Ming-Yi Hsu
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Cheng-Hsien Hsieh
- Department of Emergency Medicine, En-Chu-Kong Hospital, New Taipei City 23741, Taiwan;
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Yu-Ting Huang
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital, Keelung 20401, Taiwan
| | - Sung-Yu Chu
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
| | - Chien-Ming Chen
- Department of Medical Imaging and Intervention, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan; (M.-Y.H.); (Y.-T.H.); (S.-Y.C.); (C.-M.C.)
| | - Wei-Jiunn Lee
- Department of Medical Education and Research, Wan Fang Hospital, Taipei Medical University, Taipei 11695, Taiwan
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Correspondence: (W.-J.L.); (S.-J.L.); Tel.: +886-2-2930-7930 (ext. 2551/2547) (W.-J.L.); +886-3-2118166 (S.-J.L.)
| | - Shih-Jung Liu
- Department of Mechanical Engineering, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Orthopedic Surgery, Bone and Joint Research Center, Chang Gung Memorial Hospital, Linkou, Taoyuan 33305, Taiwan
- Correspondence: (W.-J.L.); (S.-J.L.); Tel.: +886-2-2930-7930 (ext. 2551/2547) (W.-J.L.); +886-3-2118166 (S.-J.L.)
| |
Collapse
|
28
|
El-Benhawy SA, Ebeid SA, Abd El Moneim NA, Arab ARR, Ramadan R. Repression of protocadherin 17 is correlated with elevated angiogenesis and hypoxia markers in female patients with breast cancer. Cancer Biomark 2021; 31:139-148. [PMID: 33896826 DOI: 10.3233/cbm-201593] [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: 11/15/2022]
Abstract
BACKGROUND Altered cadherin expression plays a vital role in tumorigenesis, angiogenesis and tumor progression. However, the function of protocadherin 17 (PCDH17) in breast cancer remains unclear. OBJECTIVE Our target is to explore PCDH17 gene expression in breast carcinoma tissues and its relation to serum angiopoietin-2 (Ang-2), carbonic anhydrase IX (CAIX) and % of circulating CD34+ cells in breast cancer patients (BCPs). METHODS This study included Fifty female BCPs and 50 healthy females as control group. Cancerous and neighboring normal breast tissues were collected from BCPs as well as blood samples at diagnosis. PCDH17 gene expression was evaluated by RT-PCR. Serum Ang-2, CAIX levels were measured by ELISA and % CD34+ cells were assessed by flow cytometry. RESULTS PCDH17 was downregulated in cancerous breast tissues and its repression was significantly correlated with advanced stage and larger tumor size. Low PCDH17 was significantly correlated with serum Ang-2, % CD34+ cells and serum CAIX levels. Serum CAIX, Ang-2 and % CD34+ cells levels were highly elevated in BCPs and significantly correlated with clinical stage. CONCLUSIONS PCDH17 downregulation correlated significantly with increased angiogenic and hypoxia biomarkers. These results explore the role of PCDH17 as a tumor suppressor gene inhibiting tumor growth and proliferation.
Collapse
Affiliation(s)
- Sanaa A El-Benhawy
- Radiation Sciences Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Samia A Ebeid
- Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Nadia A Abd El Moneim
- Cancer Management and Research Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Amal R R Arab
- Applied Medical Chemistry Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | - Rabie Ramadan
- Experimental and Clinical Surgery Department, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
29
|
Xiao J, Rahbar H, Hippe DS, Rendi MH, Parker EU, Shekar N, Hirano M, Cheung KJ, Partridge SC. Dynamic contrast-enhanced breast MRI features correlate with invasive breast cancer angiogenesis. NPJ Breast Cancer 2021; 7:42. [PMID: 33863924 PMCID: PMC8052427 DOI: 10.1038/s41523-021-00247-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 03/15/2021] [Indexed: 02/02/2023] Open
Abstract
Angiogenesis is a critical component of breast cancer development, and identification of imaging-based angiogenesis assays has prognostic and treatment implications. We evaluated the association of semi-quantitative kinetic and radiomic breast cancer features on dynamic contrast-enhanced (DCE)-MRI with microvessel density (MVD), a marker for angiogenesis. Invasive breast cancer kinetic features (initial peak percent enhancement [PE], signal enhancement ratio [SER], functional tumor volume [FTV], and washout fraction [WF]), radiomics features (108 total features reflecting tumor morphology, signal intensity, and texture), and MVD (by histologic CD31 immunostaining) were measured in 27 patients (1/2016-7/2017). Lesions with high MVD levels demonstrated higher peak SER than lesions with low MVD (mean: 1.94 vs. 1.61, area under the receiver operating characteristic curve [AUC] = 0.79, p = 0.009) and higher WF (mean: 50.6% vs. 22.5%, AUC = 0.87, p = 0.001). Several radiomics texture features were also promising for predicting increased MVD (maximum AUC = 0.84, p = 0.002). Our study suggests DCE-MRI can non-invasively assess breast cancer angiogenesis, which could stratify biology and optimize treatments.
Collapse
Affiliation(s)
- Jennifer Xiao
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Habib Rahbar
- Department of Radiology, University of Washington, Seattle, WA, USA
- Breast Imaging, Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Daniel S Hippe
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Mara H Rendi
- Department of Pathology, University of Washington, Seattle, WA, USA
| | | | - Neal Shekar
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Michael Hirano
- Department of Radiology, University of Washington, Seattle, WA, USA
| | - Kevin J Cheung
- Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA, USA
- Translational Research Program, Public Health Sciences and Human Biology Divisions, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Savannah C Partridge
- Department of Radiology, University of Washington, Seattle, WA, USA.
- Breast Imaging, Seattle Cancer Care Alliance, Seattle, WA, USA.
| |
Collapse
|
30
|
SPAER: Sparse Deep Convolutional Autoencoder Model to Extract Low Dimensional Imaging Biomarkers for Early Detection of Breast Cancer Using Dynamic Thermography. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11073248] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Early diagnosis of breast cancer unequivocally improves the survival rate of patients and is crucial for disease treatment. With the current developments in infrared imaging, breast screening using dynamic thermography seems to be a great complementary method for clinical breast examination (CBE) prior to mammography. In this study, we propose a sparse deep convolutional autoencoder model named SPAER to extract low-dimensional deep thermomics to aid breast cancer diagnosis. The model receives multichannel, low-rank, approximated thermal bases as input images. SPAER provides a solution for high-dimensional deep learning features and selects the predominant basis matrix using matrix factorization techniques. The model has been evaluated using five state-of-the-art matrix factorization methods and 208 thermal breast cancer screening cases. The best accuracy was for non-negative matrix factorization (NMF)-SPAER + Clinical and NMF-SPAER for maintaining thermal heterogeneity, leading to finding symptomatic cases with accuracies of 78.2% (74.3–82.5%) and 77.7% (70.9–82.1%), respectively. SPAER showed significant robustness when tested for additive Gaussian noise cases (3–20% noise), evaluated by the signal-to-noise ratio (SNR). The results suggest high performance of SPAER for preserveing thermal heterogeneity, and it can be used as a noninvasive in vivo tool aiding CBE in the early detection of breast cancer.
Collapse
|
31
|
Grape Seed Procyanidins Inhibit the Growth of Breast Cancer MCF-7 Cells by Down-Regulating the EGFR/VEGF/MMP9 Pathway. Nat Prod Commun 2021. [DOI: 10.1177/1934578x21991691] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Breast cancer is the most common invasive cancer in women and the second leading cause of cancer death in women. However, it is not clear about its effective treatments. As a potential anticancer agent, grape seed procyanidins (GSPs) have been shown to inhibit the proliferation of various cancer cells in vitro and in vivo. In this study, it was shown that GSPs significantly inhibit MCF-7 cell proliferation in a concentration/time-dependent manner. The flow cytometric data clearly demonstrated that GSPs cause cell cycle arrest in the G2/M phase, followed by cell apoptosis. Moreover, it also confirmed that growth inhibition mediated by treatment with GSPs is related to the induction of apoptosis due to p53 elevation, purportedly by inhibition of the epidermal growth factor receptor (EGFR)/vascular endothelial growth factor (VEGF)/matrix metalloproteinase 9 (MMP9) pathway. Taken together, these findings suggest that GSPs inhibit MCF-7 cells proliferation and induce cell apoptosis by suppressing EGFR/VEGF/MMP9 pathway.
Collapse
|
32
|
S VS, Royea R, Buckman KJ, Benardis M, Holmes J, Fletcher RL, Eyk N, Rajendra Acharya U, Ellenhorn JDI. An introduction to the Cyrcadia Breast Monitor: A wearable breast health monitoring device. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 197:105758. [PMID: 33007593 DOI: 10.1016/j.cmpb.2020.105758] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/10/2020] [Indexed: 05/08/2023]
Abstract
BACKGROUND The most common breast cancer detection modalities are generally limited by radiation exposure, discomfort, high costs, inter-observer variabilities in image interpretation, and low sensitivity in detecting cancer in dense breast tissue. Therefore, there is a clear need for an affordable and effective adjunct modality that can address these limitations. The Cyrcadia Breast Monitor (CBM) is a non-invasive, non-compressive, and non-radiogenic wearable device developed as an adjunct to current modalities to assist in the detection of breast tissue abnormalities in any type of breast tissue. METHODS The CBM records thermodynamic metabolic data from the breast skin surface over a period of time using two wearable biometric patches consisting of eight sensors each and a data recording device. The acquired multi-dimensional temperature time series data are analyzed to determine the presence of breast tissue abnormalities. The objective of this paper is to present the scientific background of CBM and also to describe the history around the design and development of the technology. RESULTS The results of using the CBM device in the initial clinical studies are also presented. Twenty four-hour long breast skin temperature circadian rhythm data was collected from 93 benign and 108 malignant female study subjects in the initial clinical studies. The predictive model developed using these datasets could differentiate benign and malignant lesions with 78% accuracy, 83.6% sensitivity and 71.5% specificity. A pilot study of 173 female study subjects is underway, in order to validate this predictive model in an independent test population. CONCLUSIONS The results from the initial studies indicate that the CBM may be valuable for breast health monitoring under physician supervision for confirmation of any abnormal changes, potentially prior to other methods, such as, biopsies. Studies are being conducted and planned to validate the technology and also to evaluate its ability as an adjunct breast health monitoring device for identifying abnormalities in difficult-to-diagnose dense breast tissue.
Collapse
Affiliation(s)
- Vinitha Sree S
- Cyrcadia Health, 1325 Airmotive Way, Ste. 175-L, Reno, NV 89502, United States; Cyrcadia Asia, Ltd., Hong Kong.
| | | | - Kevin J Buckman
- Cyrcadia Health, 1325 Airmotive Way, Ste. 175-L, Reno, NV 89502, United States; Adventist Health Lodi Memorial Hospital, Lodi, CA 95240, United States
| | - Matt Benardis
- Cyrcadia Health, 1325 Airmotive Way, Ste. 175-L, Reno, NV 89502, United States
| | - Jim Holmes
- Cyrcadia Health, 1325 Airmotive Way, Ste. 175-L, Reno, NV 89502, United States
| | - Ronald L Fletcher
- Cyrcadia Health, 1325 Airmotive Way, Ste. 175-L, Reno, NV 89502, United States
| | - Ng Eyk
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798
| | - U Rajendra Acharya
- School of Engineering, Division of ECE, Ngee Ann Polytechnic, Singapore 599489; Department of Biomedical Engineering, School of Science and Technology, Singapore University of Social Sciences, Singapore; Department of Biomedical Informatics and Medical Engineering, Asia University, Taiwan
| | - Joshua D I Ellenhorn
- Cyrcadia Health, 1325 Airmotive Way, Ste. 175-L, Reno, NV 89502, United States; Cyrcadia Asia, Ltd., Hong Kong; Surgery Group LA, Cedars-Sinai Medical Towers, Los Angeles, CA 90048, United States; John Wayne Cancer Clinics, Santa Monica, CA 90404, United States
| |
Collapse
|
33
|
Luo CW, Hou MF, Huang CW, Wu CC, Ou-Yang F, Li QL, Wu CC, Pan MR. The CDK6-c-Jun-Sp1-MMP-2 axis as a biomarker and therapeutic target for triple-negative breast cancer. Am J Cancer Res 2020; 10:4325-4341. [PMID: 33415002 PMCID: PMC7783751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/16/2020] [Indexed: 06/12/2023] Open
Abstract
Triple-negative breast cancer (TNBC) has high metastatic, drug-resistance, and recurrence rates, and is characterized by an angiogenic and fibrotic microenvironment that favors cancer malignancy. However, details of the mechanisms underlying malignancy are still largely unknown. Our mouse model indicated that knockdown of CDK6 inhibited lung metastasis significantly compared to parental cells. Immunohistochemical analyses revealed that the levels of collagen and the angiogenic marker matrix metalloproteinase (MMP)-2 were much lower in CDK6-deficient cells. To examine mechanisms in the CDK6-mediated phenotype of cancer cells, we studied its role in MMP-2 expression. CDK6 mediated the recruitment of transcription factors including c-Jun and Sp1 to the MMP2 promoter. Knockdown of CDK6 significantly suppressed the expression of MMP2 mRNA. Consistent with the in vitro data, the expression of CDK6 was positively correlated with the angiogenic and fibrotic tumor microenvironment in TNBC patient tissues as shown by MMP-2 and fibronectin staining, respectively. More importantly, after screening a small molecule library of 31 protein kinase inhibitors, we found that the Raf inhibitor sorafenib displayed the highest cytotoxicity in CDK6-depleted cells. These data indicate that CDK6 serves as an anti-microenvironment target and affects the drug response in retinoblastoma-proficient TNBC, suggesting that combining a CDK6 inhibitor and sorafenib leads to a synthetic effect that may represent a personalized therapeutic approach for patients with TNBC.
Collapse
Affiliation(s)
- Chi-Wen Luo
- Department of Surgery, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- Division of Breast Surgery, Department of Surgery, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Ming-Feng Hou
- Department of Surgery, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- Division of Breast Surgery, Department of Surgery, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical UniversityKaohsiung, Taiwan, R.O.C.
| | - Chia-Wei Huang
- Graduate Institute of Clinical Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Chun-Chieh Wu
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Fu Ou-Yang
- Department of Surgery, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- Division of Breast Surgery, Department of Surgery, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Qiao-Lin Li
- Graduate Institute of Clinical Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
| | - Cheng-Che Wu
- Department of Surgery, Kaohsiung Medical University HospitalKaohsiung, Taiwan
- Division of Breast Surgery, Department of Surgery, Kaohsiung Medical University HospitalKaohsiung, Taiwan
| | - Mei-Ren Pan
- Graduate Institute of Clinical Medicine, Kaohsiung Medical UniversityKaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical UniversityKaohsiung, Taiwan
| |
Collapse
|
34
|
Zduniak K, Gdesz-Birula K, Woźniak M, Duś-Szachniewicz K, Ziółkowski P. The Assessment of the Combined Treatment of 5-ALA Mediated Photodynamic Therapy and Thalidomide on 4T1 Breast Carcinoma and 2H11 Endothelial Cell Line. Molecules 2020; 25:molecules25215184. [PMID: 33171718 PMCID: PMC7664331 DOI: 10.3390/molecules25215184] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/29/2020] [Accepted: 11/02/2020] [Indexed: 12/31/2022] Open
Abstract
Photodynamic therapy (PDT) is a low-invasive method of treatment of various diseases, mainly neoplastic conditions. PDT has been experimentally combined with multiple treatment methods. In this study, we tested a combination of 5-aminolevulinic acid (5-ALA) mediated PDT with thalidomide (TMD), which is a drug presently used in the treatment of plasma cell myeloma. TMD and PDT share similar modes of action in neoplastic conditions. Using 4T1 murine breast carcinoma and 2H11 murine endothelial cells lines as an experimental tumor model, we tested 5-ALA-PDT and TMD combination in terms of cytotoxicity, apoptosis, Vascular Endothelial Growth Factor (VEGF) expression, and, in 2H11 cells, migration capabilities by wound healing assay. We have found an enhancement of cytotoxicity in 4T1 cells, whereas, in normal 2H11 cells, this effect was not statistically significant. The addition of TMD decreased the production of VEGF after PDT in 2H11 cell line. Our results reveal enhanced effectiveness of 5-ALA-PDT with TMD treatment compared to 5-ALA-PDT or TMD treatment alone. The addition of TMD may be a promising proceeding of the anti-tumor effect of PDT by decreasing the VEGF concentration in the culture medium. Further studies, including testing on different cell lines, are needed to confirm this assumption.
Collapse
|
35
|
Burton J, Umu SU, Langseth H, Grotmol T, Grimsrud TK, Haugen TB, Rounge TB. Serum RNA Profiling in the 10-Years Period Prior to Diagnosis of Testicular Germ Cell Tumor. Front Oncol 2020; 10:574977. [PMID: 33251139 PMCID: PMC7673397 DOI: 10.3389/fonc.2020.574977] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022] Open
Abstract
Although testicular germ cell tumor (TGCT) overall is highly curable, patients may experience late effects after treatment. An increased understanding of the mechanisms behind the development of TGCT may pave the way for better outcome for patients. To elucidate molecular changes prior to TGCT diagnosis we sequenced small RNAs in serum from 69 patients who were later diagnosed with TGCT and 111 matched controls. The deep RNA profiles, with on average 18 million sequences per sample, comprised of nine classes of RNA, including microRNA. We found that circulating RNA signals differed significantly between cases and controls regardless of time to diagnosis. Different levels of TSIX related to X-chromosome inactivation and TEX101 involved in spermatozoa production are among the interesting findings. The RNA signals differed between seminoma and non-seminoma TGCT subtypes, with seminoma cases showing lower levels of RNAs and non-seminoma cases showing higher levels of RNAs, compared with controls. The differentially expressed RNAs were typically associated with cancer related pathways. Our results indicate that circulating RNA profiles change during TGCT development according to histology and may be useful for early detection of this tumor type.
Collapse
Affiliation(s)
- Joshua Burton
- Department of Lifesciences and Health, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Sinan U. Umu
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Hilde Langseth
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Tom Grotmol
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Tom K. Grimsrud
- Department of Research, Cancer Registry of Norway, Oslo, Norway
| | - Trine B. Haugen
- Department of Lifesciences and Health, OsloMet - Oslo Metropolitan University, Oslo, Norway
| | - Trine B. Rounge
- Department of Research, Cancer Registry of Norway, Oslo, Norway
- Department of Informatics, University of Oslo, Oslo, Norway
| |
Collapse
|
36
|
Avagliano A, Fiume G, Ruocco MR, Martucci N, Vecchio E, Insabato L, Russo D, Accurso A, Masone S, Montagnani S, Arcucci A. Influence of Fibroblasts on Mammary Gland Development, Breast Cancer Microenvironment Remodeling, and Cancer Cell Dissemination. Cancers (Basel) 2020; 12:E1697. [PMID: 32604738 PMCID: PMC7352995 DOI: 10.3390/cancers12061697] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/20/2020] [Accepted: 06/23/2020] [Indexed: 12/20/2022] Open
Abstract
The stromal microenvironment regulates mammary gland development and tumorigenesis. In normal mammary glands, the stromal microenvironment encompasses the ducts and contains fibroblasts, the main regulators of branching morphogenesis. Understanding the way fibroblast signaling pathways regulate mammary gland development may offer insights into the mechanisms of breast cancer (BC) biology. In fact, the unregulated mammary fibroblast signaling pathways, associated with alterations in extracellular matrix (ECM) remodeling and branching morphogenesis, drive breast cancer microenvironment (BCM) remodeling and cancer growth. The BCM comprises a very heterogeneous tissue containing non-cancer stromal cells, namely, breast cancer-associated fibroblasts (BCAFs), which represent most of the tumor mass. Moreover, the different components of the BCM highly interact with cancer cells, thereby generating a tightly intertwined network. In particular, BC cells activate recruited normal fibroblasts in BCAFs, which, in turn, promote BCM remodeling and metastasis. Thus, comparing the roles of normal fibroblasts and BCAFs in the physiological and metastatic processes, could provide a deeper understanding of the signaling pathways regulating BC dissemination. Here, we review the latest literature describing the structure of the mammary gland and the BCM and summarize the influence of epithelial-mesenchymal transition (EpMT) and autophagy in BC dissemination. Finally, we discuss the roles of fibroblasts and BCAFs in mammary gland development and BCM remodeling, respectively.
Collapse
Affiliation(s)
- Angelica Avagliano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.F.); (E.V.)
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, 80131 Naples, Italy;
| | - Nunzia Martucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Eleonora Vecchio
- Department of Experimental and Clinical Medicine, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (G.F.); (E.V.)
| | - Luigi Insabato
- Anatomic Pathology Unit, Department of Advanced Biomedical Sciences, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (D.R.)
| | - Daniela Russo
- Anatomic Pathology Unit, Department of Advanced Biomedical Sciences, School of Medicine, University of Naples Federico II, 80131 Naples, Italy; (L.I.); (D.R.)
| | - Antonello Accurso
- Department of General, Oncological, Bariatric and Endocrine-Metabolic Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Stefania Masone
- Department of Clinical Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy;
| | - Stefania Montagnani
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| | - Alessandro Arcucci
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (N.M.); (S.M.)
| |
Collapse
|
37
|
Sinha S, Sharma S, Vora J, Shrivastava N. Emerging role of sirtuins in breast cancer metastasis and multidrug resistance: Implication for novel therapeutic strategies targeting sirtuins. Pharmacol Res 2020; 158:104880. [PMID: 32442721 DOI: 10.1016/j.phrs.2020.104880] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/25/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022]
Abstract
Sirtuins (SIRTs), a class III histone deacetylases (HDACs) that require NAD+ as a cofactor and include SIRT1-7 proteins in mammals. Accumulative evidence has established that every sirtuin possesses exclusive and poised biology, implicating their role in the regulation of multifaceted biological functions leading to breast cancer initiation, progression, and metastasis. This article provides an outline of recent developments in the role of sirtuins in breast cancer metastasis and development of multidrug resistance (MDR). In addition, we have also highlighted the impending prospects of targeting SIRTs to overcome MDR to bring advancement in breast cancer management. Further, this review will focus on strategies for improving the activity and efficacy of existing cancer therapeutics by combining (adjuvant treatment/therapy) them with sirtuin inhibitors/modulators. All available as well as newly discovered synthetic and dietary sirtuin inhibitors, activators/modulators have been extensively reviewed and compiled to provide a rationale for targeting sirtuins. Further, we discuss their potential in developing future therapeutics against sirtuins proposing their use along with conventional chemotherapeutics to overcome the problem of breast cancer metastasis and MDR.
Collapse
Affiliation(s)
- Sonam Sinha
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India; School of Science, Gujarat University, Ahmedabad, Gujarat, India
| | - Sonal Sharma
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India
| | - Jaykant Vora
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India; School of Science, Gujarat University, Ahmedabad, Gujarat, India
| | - Neeta Shrivastava
- Department of Pharmacognosy and Phytochemistry, B. V. Patel Pharmaceutical Education and Research Development (PERD) Centre, Ahmedabad, 380054, Gujarat, India.
| |
Collapse
|
38
|
Hulin JA, Gubareva EA, Jarzebska N, Rodionov RN, Mangoni AA, Tommasi S. Inhibition of Dimethylarginine Dimethylaminohydrolase (DDAH) Enzymes as an Emerging Therapeutic Strategy to Target Angiogenesis and Vasculogenic Mimicry in Cancer. Front Oncol 2020; 9:1455. [PMID: 31993367 PMCID: PMC6962312 DOI: 10.3389/fonc.2019.01455] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/05/2019] [Indexed: 01/01/2023] Open
Abstract
The small free radical gas nitric oxide (NO) plays a key role in various physiological and pathological processes through enhancement of endothelial cell survival and proliferation. In particular, NO has emerged as a molecule of interest in carcinogenesis and tumor progression due to its crucial role in various cancer-related events including cell invasion, metastasis, and angiogenesis. The dimethylarginine dimethylaminohydrolase (DDAH) family of enzymes metabolize the endogenous nitric oxide synthase (NOS) inhibitors, asymmetric dimethylarginine (ADMA) and monomethyl arginine (L-NMMA), and are thus key for maintaining homeostatic control of NO. Dysregulation of the DDAH/ADMA/NO pathway resulting in increased local NO availability often promotes tumor growth, angiogenesis, and vasculogenic mimicry. Recent literature has demonstrated increased DDAH expression in tumors of different origins and has also suggested a potential ADMA-independent role for DDAH enzymes in addition to their well-studied ADMA-mediated influence on NO. Inhibition of DDAH expression and/or activity in cell culture models and in vivo studies has indicated the potential therapeutic benefit of this pathway through inhibition of both angiogenesis and vasculogenic mimicry, and strategies for manipulating DDAH function in cancer are currently being actively pursued by several research groups. This review will thus provide a timely discussion on the expression, regulation, and function of DDAH enzymes in regard to angiogenesis and vasculogenic mimicry, and will offer insight into the therapeutic potential of DDAH inhibition in cancer based on preclinical studies.
Collapse
Affiliation(s)
- Julie-Ann Hulin
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Ekaterina A Gubareva
- N.N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia
| | - Natalia Jarzebska
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,Department of Anesthesiology and Intensive Care Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Roman N Rodionov
- Division of Angiology, Department of Internal Medicine III, University Center for Vascular Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Arduino A Mangoni
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Sara Tommasi
- Clinical Pharmacology, College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| |
Collapse
|
39
|
Attia ABE, Balasundaram G, Moothanchery M, Dinish U, Bi R, Ntziachristos V, Olivo M. A review of clinical photoacoustic imaging: Current and future trends. PHOTOACOUSTICS 2019; 16:100144. [PMID: 31871888 PMCID: PMC6911900 DOI: 10.1016/j.pacs.2019.100144] [Citation(s) in RCA: 437] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 07/05/2019] [Accepted: 08/21/2019] [Indexed: 05/02/2023]
Abstract
Photoacoustic imaging (or optoacoustic imaging) is an upcoming biomedical imaging modality availing the benefits of optical resolution and acoustic depth of penetration. With its capacity to offer structural, functional, molecular and kinetic information making use of either endogenous contrast agents like hemoglobin, lipid, melanin and water or a variety of exogenous contrast agents or both, PAI has demonstrated promising potential in a wide range of preclinical and clinical applications. This review provides an overview of the rapidly expanding clinical applications of photoacoustic imaging including breast imaging, dermatologic imaging, vascular imaging, carotid artery imaging, musculoskeletal imaging, gastrointestinal imaging and adipose tissue imaging and the future directives utilizing different configurations of photoacoustic imaging. Particular emphasis is placed on investigations performed on human or human specimens.
Collapse
Key Words
- AR-PAM, acoustic resolution-photoacoustic microscopy
- Clinical applications
- DAQ, data acquisition
- FOV, field-of-view
- Hb, deoxy-hemoglobin
- HbO2, oxy-hemoglobin
- LED, light emitting diode
- MAP, maximum amplitude projection
- MEMS, microelectromechanical systems
- MRI, magnetic resonance imaging
- MSOT, multispectral optoacoustic tomography
- OCT, optical coherence tomography
- OR-PAM, optical resolution-photoacoustic microscopy
- Optoacoustic mesoscopy
- Optoacoustic tomography
- PA, photoacoustic
- PAI, photoacoustic imaging
- PAM, photoacoustic microscopy
- PAT, photoacoustic tomography
- Photoacoustic imaging
- Photoacoustic microscopy
- RSOM, raster-scanning optoacoustic mesoscopy
- SBH-PACT, single breath hold photoacoustic computed tomography system
- US, ultrasound
- sO2, saturation
Collapse
Affiliation(s)
| | | | - Mohesh Moothanchery
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, A*STAR, Singapore
| | - U.S. Dinish
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, A*STAR, Singapore
| | - Renzhe Bi
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, A*STAR, Singapore
| | - Vasilis Ntziachristos
- Institute for Biological and Medical Imaging, Technische Universität München and Helmholtz Zentrum München, Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Malini Olivo
- Laboratory of Bio-optical Imaging, Singapore Bioimaging Consortium, A*STAR, Singapore
| |
Collapse
|
40
|
Maity G, Chakraborty J, Ghosh A, Haque I, Banerjee S, Banerjee SK. Aspirin suppresses tumor cell-induced angiogenesis and their incongruity. J Cell Commun Signal 2019; 13:491-502. [PMID: 30610526 PMCID: PMC6946772 DOI: 10.1007/s12079-018-00499-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Accepted: 11/26/2018] [Indexed: 01/10/2023] Open
Abstract
Tumor neovascularization/tumor angiogenesis is a pathophysiological process in which new blood vessels are formed from existing blood vessels in the primary tumors to supply adequate oxygen and nutrition to cancer cells for their proliferation and metastatic growth to the distant organs. Therefore, controlling tumor angiogenesis is an attractive target for cancer therapy. Structural abnormalities of the vasculature (i.e., leakiness due to the abnormal lining of pericytes on the microvessels) are one of the critical features of tumor angiogenesis that sensitizes vascular cells to cytokines and helps circulating tumor cells to metastasize to distant organs. Our goal is to repurpose the drugs that may prevent tumor angiogenesis or normalize the vessels by repairing leakiness via recruiting pericytes or both. In this study, we tested whether aspirin (ASA), which could block primary tumor growth, regulates tumor angiogenesis. We investigated the effects of low (1 mM) and high (2.5 mM) doses of ASA (direct effect), and ASA-treated or untreated triple negative breast cancer (TNBC) cells' conditioned media (indirect effect) on endothelial cell physiology. These include in vitro migration using modified Boyden chamber assay, in vitro capillary-like structure formation on Matrigel, interactions of pericytes-endothelial cells and cell permeability using in vitro endothelial permeability assay. We also examined the effect of ASA on various molecular factors associated with tumor angiogenesis. Finally, we found the outcome of ASA treatment on in vivo tumor angiogenesis. We found that ASA-treatment (direct or indirect) significantly blocks in vitro migration and capillary-like structure formation by endothelial cells. Besides, we found that ASA recruits pericytes from multipotent stem cells and helps in binding with endothelial cells, which is a hallmark of normalization of blood vessels, and decreases in vitro permeability through endothelial cell layer. The antiangiogenic effect of ASA was also documented in vivo assays. Mechanistically, ASA treatment blocks several angiogenic factors that are associated with tumor angiogenesis, and suggesting ASA blocks paracrine-autocrine signaling network between tumor cells and endothelial cells. Collectively, these studies implicate aspirin with proper dose may provide potential therapeutic for breast cancer via blocking as well as normalizing tumor angiogenesis.
Collapse
Affiliation(s)
- Gargi Maity
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jinia Chakraborty
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA
- Blue Valley High School, 16200 Antioch Rd, Overland Park, KS, 66085, USA
| | - Arnab Ghosh
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Inamul Haque
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Snigdha Banerjee
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - Sushanta K Banerjee
- Cancer Research Unit, Research Division (151), VA Medical Center, 4801 E Linwood Boulevard, Kansas City, MO, 64128, USA.
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA.
| |
Collapse
|
41
|
Yang Y, Tang J, Zhang M, Gu Z, Song H, Yang Y, Yu C. Responsively Aggregatable Sub-6 nm Nanochelators Induce Simultaneous Antiangiogenesis and Vascular Obstruction for Enhanced Tumor Vasculature Targeted Therapy. NANO LETTERS 2019; 19:7750-7759. [PMID: 31657578 DOI: 10.1021/acs.nanolett.9b02691] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Inhibiting the formation of new tumor blood vessels (so-called antiangiogenesis) and obstructing the established ones are two primary strategies in tumor vasculature targeted therapy. However, the therapeutic outcome of conventional methodologies relying on only one mechanism is rather limited. Herein, the first example of ultrasmall responsively aggregatable nanochelators that can intrinsically fulfill both antivasculature functions as well as high renal clearable efficiency is introduced. The nanochelators with sub-6 nm sizes exhibit not only systemic copper depletion activity for tumor antiangiogenesis but also, more surprisingly, the capability to transform from a "dispersed" state to an "aggregated" state to form large secondary particles in response to tumor microenvironment with elevated copper and phosphate levels for blood vessel obstruction. Compared to a benchmark antiangiogenic agent that can only inhibit the formation of tumor blood vessels, the nanochelators with unprecedented synergistic functions demonstrate significantly enhanced tumor inhibition activity in both breast cancer and colon cancer tumor models. Moreover, these ultrasmall nanochelators are noncytotoxic and renal clearable, ensuring superior biocompatibility. It is envisaged that the design of nanomaterials with ground-breaking properties and the synergistic antivasculature functions would offer a substantial conceptual advance for tumor vasculature targeted therapy and may provide vast opportunities for developing advanced nanomedicines.
Collapse
Affiliation(s)
- Yannan Yang
- Australian Institute for Bioengineering and Nanotechnology , The University of Queensland , St. Lucia , Brisbane , Queensland 4072 , Australia
| | - Jie Tang
- Australian Institute for Bioengineering and Nanotechnology , The University of Queensland , St. Lucia , Brisbane , Queensland 4072 , Australia
| | - Min Zhang
- Australian Institute for Bioengineering and Nanotechnology , The University of Queensland , St. Lucia , Brisbane , Queensland 4072 , Australia
- School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , People's Republic of China
| | - Zhengying Gu
- Australian Institute for Bioengineering and Nanotechnology , The University of Queensland , St. Lucia , Brisbane , Queensland 4072 , Australia
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology , The University of Queensland , St. Lucia , Brisbane , Queensland 4072 , Australia
| | - Yang Yang
- Australian Institute for Bioengineering and Nanotechnology , The University of Queensland , St. Lucia , Brisbane , Queensland 4072 , Australia
| | - Chengzhong Yu
- Australian Institute for Bioengineering and Nanotechnology , The University of Queensland , St. Lucia , Brisbane , Queensland 4072 , Australia
- School of Chemistry and Molecular Engineering , East China Normal University , Shanghai 200241 , People's Republic of China
| |
Collapse
|
42
|
Mohamad NE, Abu N, Yeap SK, Lim KL, Romli MF, Sharifuddin SA, Long K, Alitheen NB. Apoptosis and metastasis inhibitory potential of pineapple vinegar against mouse mammary gland cells in vitro and in vivo. Nutr Metab (Lond) 2019; 16:49. [PMID: 31372176 PMCID: PMC6660685 DOI: 10.1186/s12986-019-0380-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/23/2019] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Plant-based food medicine and functional foods have been consumed extensively due to their bioactive substances and health-beneficial effects. Vinegar is one of them due to its bioactivities, which confers benefits on human body. Our previous study has produced pineapple vinegar that is rich in gallic acid and caffeic acid via 2 steps fermentation. There are many evidences that show the effectiveness of these resources in inhibiting the proliferation and metastasis of the cancer cells through several mechanisms. METHODS Freeze-dried pineapple vinegar was evaluated for its in vitro apoptosis and metastasis inhibitory potential using MTT, cell cycle, Annexin V and scratch assays. The in vivo test using BALB/c mice challenged with 4 T1 cells was further investigated by pre-treating the mice with 0.08 or 2 ml/kg body weight of freshly-prepared pineapple vinegar for 28 days. The tumor weight, apoptotic state of cells in tumor, metastasis and immune response of the untreated and pineapple vinegar treatment group were evaluated and compared. RESULTS From the in vitro study, an IC50 value of 0.25 mg/mL after 48 h of treatment was established. Annexin V/PI and scratch closure assays showed that pineapple vinegar induced 70% of cell population to undergo apoptosis and inhibited 30% of wound closure of 4 T1 cells. High concentration of pineapple vinegar (2 ml/kg body weight) led to the reduction of tumor weight and volume by 45%as compared to the untreated 4 T1-challenged mice. This effect might have been contributed by the increase of T cell and NK cells population associated with the overexpression of IL-2 andIFN-γ cytokines and splenocyte cytotoxicity. Furthermore, fewer instances of metastasis events were recorded in the pineapple vinegar treatment group and this could be explained by the downregulation of inflammation related genes (iNOS, NF-kB and COX2), metastasis related genes (iCAM, VEGF and MMP9) and angeogenesis related genes (CD26, TIMP1, HGF, MMP3, IGFBP-1 and IGFBP-2). CONCLUSION The ability of pineapple vinegar to delay cancer progression portrayed its potential as chemopreventive dietry intervention for cancer therapy.
Collapse
Affiliation(s)
- Nurul Elyani Mohamad
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia (UPM), 43400 Serdang, Selangor Malaysia
| | - Nadiah Abu
- UKM Molecular Biology Institute (UMBI), UKM Medical Centre, Jalan Yaa’cob Latiff, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur Malaysia
| | - Swee Keong Yeap
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Jalan Sunsuria, Bandar Sunsuria, 43900 Sepang, Selangor Malaysia
| | - Kian Lam Lim
- Faculty of Medicine and Health Sciences, Universiti Tunku Abdul Rahman, Sungai Long Campus, Jalan Sungai Long, Bandar Sungai Long, Cheras, 43000 Kajang, Selangor Malaysia
| | - Muhammad Firdaus Romli
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia (UPM), 43400 Serdang, Selangor Malaysia
| | - Shaiful Adzni Sharifuddin
- Biotechnology Research Centre, Malaysian Agricultural Research and Development Institute (MARDI), 43400 Serdang, Selangor Malaysia
| | - Kamariah Long
- Biotechnology Research Centre, Malaysian Agricultural Research and Development Institute (MARDI), 43400 Serdang, Selangor Malaysia
| | - Noorjahan Banu Alitheen
- Department of Cell and Molecular Biology, Faculty of Biotechnology and Biomolecular Science, Universiti Putra Malaysia (UPM), 43400 Serdang, Selangor Malaysia
- Institute of Bioscience, Universiti Putra Malaysia (UPM), 43400 Serdang, Selangor Malaysia
| |
Collapse
|
43
|
Optoacoustic imaging of the breast: correlation with histopathology and histopathologic biomarkers. Eur Radiol 2019; 29:6728-6740. [PMID: 31134367 PMCID: PMC6828639 DOI: 10.1007/s00330-019-06262-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Revised: 04/10/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023]
Abstract
Aim This study was conducted in order to investigate the role of gray-scale ultrasound (US) and optoacoustic imaging combined with gray-scale ultrasound (OA/US) to better differentiate between breast cancer molecular subtypes. Materials and methods All 67 malignant masses included in the Maestro trial were retrospectively reviewed to compare US and OA/US feature scores and histopathological findings. Kruskal–Wallis tests were used to analyze the relationship between US and OA/US features and molecular subtypes of breast cancer. If a significant relationship was found, additional Wilcoxon–Mann–Whitney tests were used to identify the differences between molecular subtype groups. Results US sound transmission helped to differentiate between LUMA and LUMB, LUMB and TNBC, and LUMB and all other molecular subtypes combined (p values < 0.05). Regarding OA/US features, the sum of internal features helped to differentiate between TNBC and HER2-enriched subtypes (p = 0.049). Internal vessels (p = 0.025), sum of all internal features (p = 0.019), and sum of internal and external features (p = 0.028) helped to differentiate between LUMA and LUMB. All internal features, the sum of all internal features, the sum of all internal and external features, and the ratio of internal and external features helped to differentiate between LUMA and TNBC. The same features also helped to differentiate between LUMA and TNBC from other molecular subtypes (p values < 0.05). Conclusions The use of OA/US might help radiologists to better differentiate between breast cancer molecular subtypes. Further studies need to be carried out in order to validate these results. Key Points • The combination of functional and morphologic information provided by optoacoustic imaging (OA) combined with gray-scale US helped to differentiate between breast cancer molecular subtypes. Electronic supplementary material The online version of this article (10.1007/s00330-019-06262-0) contains supplementary material, which is available to authorized users.
Collapse
|
44
|
Alique M, Bodega G, Giannarelli C, Carracedo J, Ramírez R. MicroRNA-126 regulates Hypoxia-Inducible Factor-1α which inhibited migration, proliferation, and angiogenesis in replicative endothelial senescence. Sci Rep 2019; 9:7381. [PMID: 31089163 PMCID: PMC6517399 DOI: 10.1038/s41598-019-43689-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 02/01/2019] [Indexed: 12/15/2022] Open
Abstract
Whereas a healthy endothelium maintains physiological vascular functions, endothelial damage contributes to the development of cardiovascular diseases. Endothelial senescence is the main determinant of endothelial dysfunction and thus of age-related cardiovascular disease. The objective of this study is to test the involvement of microRNA-126 and HIF-1α in a model of replicative endothelial senescence and the interrelationship between both molecules in this in vitro model. We demonstrated that senescent endothelial cells experience impaired tube formation and delayed wound healing. Senescent endothelial cells failed to express HIF-1α, and the microvesicles released by these cells failed to carry HIF-1α. Of note, HIF-1α protein levels were restored in HIF-1α stabilizer-treated senescent endothelial cells. Finally, we show that microRNA-126 was downregulated in senescent endothelial cells and microvesicles. With regard to the interplay between microRNA-126 and HIF-1α, transfection with a microRNA-126 inhibitor downregulated HIF-1α expression in early passage endothelial cells. Moreover, while HIF-1α inhibition reduced tube formation and wound healing closure, microRNA-126 levels remained unchanged. These data indicate that HIF-1α is a target of miRNA-126 in protective and reparative functions, and suggest that their therapeutic modulation could benefit age-related vascular disease.
Collapse
Affiliation(s)
- Matilde Alique
- Departamento Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá (IRYCIS), Alcalá de Henares, Madrid, Spain.
| | - Guillermo Bodega
- Departamento de Biomedicina y Biotecnología, Facultad de Biología, Química y Ciencias Ambientales, Universidad de Alcalá. Alcalá de Henares, Madrid, Spain
| | - Chiara Giannarelli
- Cardiovascular Research Center, One Gustave L. Levy Place, New York, NY, USA.,Institute for Genomics and Multiscale Biology, One Gustave L. Levy Place, New York, NY, USA.,Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, USA
| | - Julia Carracedo
- Departamento de Genética, Fisiología y Microbiología, Facultad de Biología, Universidad Complutense de Madrid, Madrid, Spain.,Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Rafael Ramírez
- Departamento Biología de Sistemas, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá (IRYCIS), Alcalá de Henares, Madrid, Spain
| |
Collapse
|
45
|
Kumar N, Prasad P, Jash E, Jayasundar S, Singh I, Alam N, Murmu N, Somashekhar SP, Goldman A, Sehrawat S. cAMP regulated EPAC1 supports microvascular density, angiogenic and metastatic properties in a model of triple negative breast cancer. Carcinogenesis 2019; 39:1245-1253. [PMID: 29982410 DOI: 10.1093/carcin/bgy090] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 07/04/2018] [Indexed: 12/18/2022] Open
Abstract
Breast cancer is a leading cause of cancer-related mortality in women. Triple-negative breast cancer (TNBC; HER2-, ER-/PR-) is an aggressive subtype prone to drug resistance and metastasis, which is characterized by high intratumor microvascular density (iMVD) resulting from angiogenesis. However, the mechanisms contributing to the aggressive phenotypes of TNBC remain elusive. We recently reported that down-regulation of exchange factor directly activated by cyclic AMP (cAMP), also known as EPAC1, leads to a reduction in metastatic properties including proliferation and cell migration in TNBC cell lines. Here, we report that EPAC1 supports TNBC-induced angiogenesis, tumor cell migration and invasiveness as well as pro-metastatic phenotypes in endothelial cells induced through the tumor secretome. Using an approach that integrates proteomics with bioinformatics and gene ontologies, we elucidate that EPAC1 supports a tumor-secreted network of angiogenic, cell adhesion and cell migratory pathways. Using confocal microscopy, we show that signaling molecules involved in focal adhesion, including Paxillin and MENA, are down-regulated in the absence of EPAC1, and electric cell substrate impedance sensing technique confirmed a role for EPAC1 on TNBC-induced endothelial cell permeability. Finally, to provide a translational bridge, we studied iMVD and therapy response using a primary human tumor explant assay, CANscriptTM, which suggests a link between therapy-modulated neovascularization and drug sensitivity. These data provide mechanistic insight into the role of EPAC1 in regulating the tumor microenvironment, iMVD and cancer cell-induced angiogenesis, a dynamic mechanism under drug pressure that may associate to treatment failure.
Collapse
Affiliation(s)
- Naveen Kumar
- Brain Metastasis and NeuroVascular Disease Modeling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Dadri, Uttar Pradesh, India
| | - Peeyush Prasad
- Brain Metastasis and NeuroVascular Disease Modeling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Dadri, Uttar Pradesh, India
| | - Eshna Jash
- Brain Metastasis and NeuroVascular Disease Modeling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Dadri, Uttar Pradesh, India
| | - Smruthi Jayasundar
- Brain Metastasis and NeuroVascular Disease Modeling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Dadri, Uttar Pradesh, India
| | - Itender Singh
- Department of Neurology, Washington University School of Medicine, Hope Center Program on Protein Aggregation and Neurodegeneration, Charles F. and Joanne Knight Alzheimer's Disease Research Center, St. Louis, MI, USA
| | - Neyaz Alam
- Department of Surgical Oncology, Chittaranjan National Cancer Institute, Kolkata, India
| | - Nabendu Murmu
- Department of Signal Transduction and Biogenic Amines, Chittaranjan National Cancer Institute, Kolkata, India
| | | | - Aaron Goldman
- Integrative Immuno-Ocology Center, Mitra Biotech, Woburn, MA, USA.,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Seema Sehrawat
- Brain Metastasis and NeuroVascular Disease Modeling Lab, Department of Life Sciences, School of Natural Sciences, Shiv Nadar University, Dadri, Uttar Pradesh, India.,Department of Medicine, Harvard Medical School, Boston, MA, USA.,Division of Engineering in Medicine, Brigham and Women's Hospital, Boston, MA, USA
| |
Collapse
|
46
|
Rampogu S, Baek A, Park C, Son M, Parate S, Parameswaran S, Park Y, Shaik B, Kim JH, Park SJ, Lee KW. Discovery of Small Molecules that Target Vascular Endothelial Growth Factor Receptor-2 Signalling Pathway Employing Molecular Modelling Studies. Cells 2019; 8:E269. [PMID: 30901950 PMCID: PMC6468367 DOI: 10.3390/cells8030269] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/08/2019] [Accepted: 03/16/2019] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis is defined as the formation of new blood vessels and is a key phenomenon manifested in a host of cancers during which tyrosine kinases play a crucial role. Vascular endothelial growth factor receptor-2 (VEGFR-2) is pivotal in cancer angiogenesis, which warrants the urgency of discovering new anti-angiogenic inhibitors that target the signalling pathways. To obtain this objective, a structure-based pharmacophore model was built from the drug target VEGFR-2 (PDB code: 4AG8), complexed with axitinib and was subsequently validated and employed as a 3D query to retrieve the candidate compounds with the key inhibitory features. The model was escalated to molecular docking studies resulting in seven candidate compounds. The molecular docking studies revealed that the seven compounds displayed a higher dock score than the reference-cocrystallised compound. The GROningen MAchine for Chemical Simulations (GROMACS) package guided molecular dynamics (MD) results determined their binding mode and affirmed stable root mean square deviation. Furthermore, these compounds have preserved their key interactions with the residues Glu885, Glu917, Cys919 and Asp1046. The obtained findings deem that the seven compounds could act as novel anti-angiogenic inhibitors and may further assist as the prototype in designing and developing new inhibitors.
Collapse
Affiliation(s)
- Shailima Rampogu
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea; (S.R.); (A.B.); (C.P.); (M.S.); (S.P.); (S.P.)
| | - Ayoung Baek
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea; (S.R.); (A.B.); (C.P.); (M.S.); (S.P.); (S.P.)
| | - Chanin Park
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea; (S.R.); (A.B.); (C.P.); (M.S.); (S.P.); (S.P.)
| | - Minky Son
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea; (S.R.); (A.B.); (C.P.); (M.S.); (S.P.); (S.P.)
| | - Shraddha Parate
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea; (S.R.); (A.B.); (C.P.); (M.S.); (S.P.); (S.P.)
| | - Saravanan Parameswaran
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea; (S.R.); (A.B.); (C.P.); (M.S.); (S.P.); (S.P.)
| | - Yohan Park
- College of Pharmacy, Inje University, 197 Inje-ro, Gimhae, Gyeongnam 50834, Korea;
| | - Baji Shaik
- Department of Chemistry (BK 21 plus), Research Institute of Natural Science (RINS), Gyeongsang National University, Jinju, Gyeongnam 52828, Korea; (B.S.); (J.H.K.)
| | - Ju Hyun Kim
- Department of Chemistry (BK 21 plus), Research Institute of Natural Science (RINS), Gyeongsang National University, Jinju, Gyeongnam 52828, Korea; (B.S.); (J.H.K.)
| | - Seok Ju Park
- Department of Internal Medicine, College of Medicine, Busan Paik Hospital, Inje University, Gyeongnam 47392, Korea
| | - Keun Woo Lee
- Division of Life Sciences, Division of Applied Life Science (BK21 Plus), Plant Molecular Biology and Biotechnology Research Center (PMBBRC), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea; (S.R.); (A.B.); (C.P.); (M.S.); (S.P.); (S.P.)
| |
Collapse
|
47
|
Identification of differentially expressed genes regulated by molecular signature in breast cancer-associated fibroblasts by bioinformatics analysis. Arch Gynecol Obstet 2017; 297:161-183. [PMID: 29063236 DOI: 10.1007/s00404-017-4562-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 09/21/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Breast cancer is a severe risk to public health and has adequately convoluted pathogenesis. Therefore, the description of key molecular markers and pathways is of much importance for clarifying the molecular mechanism of breast cancer-associated fibroblasts initiation and progression. Breast cancer-associated fibroblasts gene expression dataset was downloaded from Gene Expression Omnibus database. METHODS A total of nine samples, including three normal fibroblasts, three granulin-stimulated fibroblasts and three cancer-associated fibroblasts samples, were used to identify differentially expressed genes (DEGs) between normal fibroblasts, granulin-stimulated fibroblasts and cancer-associated fibroblasts samples. The gene ontology (GO) and pathway enrichment analysis was performed, and protein-protein interaction (PPI) network of the DEGs was constructed by NetworkAnalyst software. RESULTS Totally, 190 DEGs were identified, including 66 up-regulated and 124 down-regulated genes. GO analysis results showed that up-regulated DEGs were significantly enriched in biological processes (BP), including cell-cell signalling and negative regulation of cell proliferation; molecular function (MF), including insulin-like growth factor II binding and insulin-like growth factor I binding; cellular component (CC), including insulin-like growth factor binding protein complex and integral component of plasma membrane; the down-regulated DEGs were significantly enriched in BP, including cell adhesion and extracellular matrix organization; MF, including N-acetylgalactosamine 4-sulfate 6-O-sulfotransferase activity and calcium ion binding; CC, including extracellular space and extracellular matrix. WIKIPATHWAYS analysis showed the up-regulated DEGs were enriched in myometrial relaxation and contraction pathways. WIKIPATHWAYS, REACTOME, PID_NCI and KEGG pathway analysis showed the down-regulated DEGs were enriched endochondral ossification, TGF beta signalling pathway, integrin cell surface interactions, beta1 integrin cell surface interactions, malaria and glycosaminoglycan biosynthesis-chondroitin sulfate/dermatan sulphate. The top 5 up-regulated hub genes, CDKN2A, MME, PBX1, IGFBP3, and TFAP2C and top 5 down-regulated hub genes VCAM1, KRT18, TGM2, ACTA2, and STAMBP were identified from the PPI network, and subnetworks revealed these genes were involved in significant pathways, including myometrial relaxation and contraction pathways, integrin cell surface interactions, beta1 integrin cell surface interaction. Besides, the target hsa-mirs for DEGs were identified. hsa-mir-759, hsa-mir-4446-5p, hsa-mir-219a-1-3p and hsa-mir-26a-5p were important miRNAs in this study. CONCLUSIONS We pinpoint important key genes and pathways closely related with breast cancer-associated fibroblasts initiation and progression by a series of bioinformatics analysis on DEGs. These screened genes and pathways provided for a more detailed molecular mechanism underlying breast cancer-associated fibroblasts occurrence and progression, holding promise for acting as molecular markers and probable therapeutic targets.
Collapse
|
48
|
Klahan S, Wong HSC, Tu SH, Chou WH, Zhang YF, Ho TF, Liu CY, Yih SY, Lu HF, Chen SCC, Huang CC, Chang WC. Identification of genes and pathways related to lymphovascular invasion in breast cancer patients: A bioinformatics analysis of gene expression profiles. Tumour Biol 2017. [PMID: 28651487 DOI: 10.1177/1010428317705573] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Surgery is the most effective treatment for breast cancer patients. However, some patients developed recurrence and distant metastasis after surgery. Adjuvant therapy is considered for high-risk patients depending on several prognostic markers, and lymphovascular invasion has become one of such prognostic markers that help physicians to identify the risk for distant metastasis and recurrence. However, the mechanism of lymphovascular invasion in breast cancer remains unknown. This study aims to unveil the genes and pathways that may involve in lymphovascular invasion in breast cancer. In total, 108 breast cancer samples were collected during surgery and microarray analysis was performed. Significance analysis of the microarrays and limma package for R were used to examine differentially expressed genes between lymphovascular invasion-positive and lymphovascular invasion-negative cases. Network and pathway analyses were mapped using the Ingenuity Pathway Analysis and the Database for Annotation, Visualization and Integrated Discovery. In total, 86 differentially expressed genes, including 37 downregulated genes and 49 upregulated genes were identified in lymphovascular invasion-positive patients. Among these genes, TNFSF11, IL6ST, and EPAS1 play important roles in cytokine-receptor interaction, which is the most enriched pathway related to lymphovascular invasion. Moreover, the results also suggested that an imbalance between extracellular matrix components and tumor micro-environment could induce lymphovascular invasion. Our study evaluated the underlying mechanisms of lymphovascular invasion, which may further help to assess the risk of breast cancer progression and identify potential targets of adjuvant treatment.
Collapse
Affiliation(s)
- Sukhontip Klahan
- 1 Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University
| | - Henry Sung-Ching Wong
- 1 Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University.,2 Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Shih-Hsin Tu
- 3 Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,4 Taipei Cancer Center, Taipei Medical University, Taipei, Taiwan
| | - Wan-Hsuan Chou
- 2 Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - Yan-Feng Zhang
- 5 HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Thien-Fiew Ho
- 6 Department of Surgery, Cathay General Hospital Sijhih, New Taipei City, Taiwan
| | - Chih-Yi Liu
- 7 Department of Pathology, Cathay General Hospital Sijhih, New Taipei City, Taiwan
| | - Shih-Ying Yih
- 8 Department of Hematology and Oncology, Cathay General Hospital Sijhih, New Taipei City, Taiwan
| | - Hsing Fang Lu
- 1 Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University
| | - Sean Chun-Chang Chen
- 9 Graduate Institute of Biomedical Informatics, College of Medical Science and Technology, Taipei Medical University
| | - Chi-Cheng Huang
- 3 Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,10 Breast Center, Cathay General Hospital, Taipei, Taiwan.,11 School of Medicine, College of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan
| | - Wei-Chiao Chang
- 1 Department of Clinical Pharmacy, School of Pharmacy, Taipei Medical University.,2 Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, School of Pharmacy, Taipei Medical University, Taipei, Taiwan.,12 Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.,13 Department of Pharmacy, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.,14 Center for Biomarkers and Biotech Drugs, Kaohsiung Medical University, Kaohsiung, Taiwan
| |
Collapse
|
49
|
Endurance Training Attenuates Angiogenesis Following Breast Cancer by Regulation of MiR-126 and MiR-296 in Breast Cancer Bearing Mice. INTERNATIONAL JOURNAL OF CANCER MANAGEMENT 2017. [DOI: 10.5812/ijcm.8067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
Badana A, Chintala M, Varikuti G, Pudi N, Kumari S, Kappala VR, Malla RR. Lipid Raft Integrity Is Required for Survival of Triple Negative Breast Cancer Cells. J Breast Cancer 2016; 19:372-384. [PMID: 28053625 PMCID: PMC5204043 DOI: 10.4048/jbc.2016.19.4.372] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 09/28/2016] [Indexed: 11/30/2022] Open
Abstract
Purpose Lipid rafts are cholesterol enriched microdomains that colocalize signaling pathways involved in cell proliferation, metastasis, and angiogenesis. We examined the effect of methyl-β-cyclodextrin (MβCD)-mediated cholesterol extraction on the proliferation, adhesion, invasion, and angiogenesis of triple negative breast cancer (TNBC) cells. Methods We measured cholesterol and estimated cell toxicity. Detergent resistant membrane (DRM) and non-DRM fractions were separated using the OptiPrep gradient method. Cell cycles stages were analyzed by flow cytometry, apoptosis was assessed using the TdT-mediated dUTP nick end-labeling assay, and metastasis was determined using a Matrigel invasion assay. Neo-vessel pattern and levels of angiogenic modulators were determined using an in vitro angiogenesis assay and an angiogenesis array, respectively. Results The present study found that the cholesterol-depleting agent MβCD, efficiently depleted membrane cholesterol and caused concentration dependent (0.1–0.5 mM) cytotoxicity compared to nystatin and filipin III in TNBC cell lines, MDA-MB 231 and MDA-MB 468. A reduced proportion of caveolin-1 found in DRM fractions indicated a cholesterol extraction-induced disruption of lipid raft integrity. MβCD inhibited 52% of MDA-MB 231 cell adhesion on fibronectin and 56% of MDA-MB 468 cell adhesion on vitronectin, while invasiveness of these cells was decreased by 48% and 52% respectively, following MβCD treatment (48 hours). MβCD also caused cell cycle arrest at the G2M phase and apoptosis in MDA-MB 231 cells (25% and 58% cells, respectively) and in MDA-MB 468 cells (30% and 38% cells, respectively). We found that MβCD treated cells caused a 52% and 58% depletion of neovessel formation in both MDA-MB 231 and MDA-MB 468 cell lines, respectively. This study also demonstrated that MβCD treatment caused a respective 2.6- and 2.5-fold depletion of tyrosine protein kinase receptor (TEK) receptor tyrosine kinase levels in both TNBC cell lines. Conclusion MβCD-induced cholesterol removal enhances alterations in lipid raft integrity, which reduces TNBC cell survival.
Collapse
Affiliation(s)
- Anil Badana
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM University, Visakhapatnam, India
| | - Madhuri Chintala
- Department of Obstetrics & Gynecology, Andhra Medical College, Visakhapatnam, India
| | - Gayathri Varikuti
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM University, Visakhapatnam, India
| | - Nagaseshu Pudi
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM University, Visakhapatnam, India
| | - Seema Kumari
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM University, Visakhapatnam, India
| | - Vijaya Rachel Kappala
- Department of Biochemistry, GITAM Institute of Science, GITAM University, Visakhapatnam, India
| | - Rama Rao Malla
- Cancer Biology Lab, Department of Biochemistry, GIS, GITAM University, Visakhapatnam, India.; Department of Biochemistry, GITAM Institute of Science, GITAM University, Visakhapatnam, India
| |
Collapse
|