1
|
Chan HW, Kuo DY, Shueng PW, Chuang HY. Visualizing the Tumor Microenvironment: Molecular Imaging Probes Target Extracellular Matrix, Vascular Networks, and Immunosuppressive Cells. Pharmaceuticals (Basel) 2024; 17:1663. [PMID: 39770505 PMCID: PMC11676442 DOI: 10.3390/ph17121663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
The tumor microenvironment (TME) is a critical factor in cancer progression, driving tumor growth, immune evasion, therapeutic resistance, and metastasis. Understanding the dynamic interactions within the TME is essential for advancing cancer management. Molecular imaging provides a non-invasive, real-time, and longitudinal approach to studying the TME, with techniques such as positron emission tomography (PET), magnetic resonance imaging (MRI), and fluorescence imaging offering complementary strengths, including high sensitivity, spatial resolution, and intraoperative precision. Recent advances in imaging probe development have enhanced the ability to target and monitor specific components of the TME, facilitating early cancer diagnosis, therapeutic monitoring, and deeper insights into tumor biology. By integrating these innovations, molecular imaging offers transformative potential for precision oncology, improving diagnostic accuracy and treatment outcomes through a comprehensive assessment of TME dynamics.
Collapse
Affiliation(s)
- Hui-Wen Chan
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei City 112, Taiwan;
| | - Deng-Yu Kuo
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
| | - Pei-Wei Shueng
- Division of Radiation Oncology, Department of Radiology, Far Eastern Memorial Hospital, New Taipei City 220, Taiwan
- Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei City 112, Taiwan
| | - Hui-Yen Chuang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong St., Beitou Dist., Taipei City 112, Taiwan;
| |
Collapse
|
2
|
Wang Y, Zhao H, Fu P, Tian L, Su Y, Lyu Z, Gu W, Wang Y, Liu S, Wang X, Zheng H, Du J, Zhang R. Preoperative prediction of lymph node metastasis in colorectal cancer using 18F-FDG PET/CT peritumoral radiomics analysis. Med Phys 2024; 51:5214-5225. [PMID: 38801340 DOI: 10.1002/mp.17193] [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: 11/09/2023] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Radiomics has been used in the diagnosis of tumor lymph node metastasis (LNM). However, to date, most studies have been based on intratumoral radiomics. Few studies have focused on the use of 18F-fluorodeoxyglucose positron emission computed tomography (18F-FDG PET/CT) peritumoral radiomics for the diagnosis of LNM in colorectal cancer (CRC). PURPOSE Determining the value of radiomics features extracted from 18F-FDG PET/CT images of the peritumoral region in predicting LNM in patients with CRC. METHODS The clinical data and preoperative 18F-FDG PET/CT images of 244 CRC patients were retrospectively analyzed. Intratumoral and peritumoral radiomics features were screened using the mutual information method, and least absolute shrinkage and selection operator regression. Based on the selected radiomics features, a radiomics score (Rad-score) was calculated, and independent risk factors obtained from univariate and multivariate logistic regression analyses were used to construct clinical and combined (Radiomics + Clinical) models. The performance of these models was evaluated using the DeLong test, while their clinical utility was assessed by decision curve analysis. Finally, a nomogram was constructed to visualize the predictive model. RESULTS The most optimal set of features retained by the feature filtering process were all peritumoral radiomic features. Carcinoembryonic antigen levels, PET/CT-reported lymph node status and Rad-score were found to be independent risk factors for LNM. All three LNM risk assessment models exhibited good predictive performance, with the combined model showing the best classification results, with areas under the curve of 0.85 and 0.76 in the training and validation groups, respectively. The DeLong test revealed that the performance of the combined model was superior to that of the clinical and radiomics models in both the training and validation groups, although this difference was only statistically significant in the training group. DCA indicated that the combined model displayed better clinical utility. CONCLUSIONS 18F-FDG PET/CT peritumoral radiomics is uniquely suited to predict the presence of LNM in patients with CRC. In particular, the predictive efficacy of LNM for precision therapy and individualized patient management can be improved by using a combination of clinical risk factors.
Collapse
Affiliation(s)
- Yan Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyue Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Peng Fu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lin Tian
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yexin Su
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhehao Lyu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenchao Gu
- Department of Diagnostic and Interventional Radiology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yang Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shan Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xi Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Han Zheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jingjing Du
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Rui Zhang
- Department of Magnetic Resonance, The First Hospital of Qiqihar, Qiqihar, Heilongjiang, China
| |
Collapse
|
3
|
Padinharayil H, George A. Small extracellular vesicles: Multi-functional aspects in non-small cell lung carcinoma. Crit Rev Oncol Hematol 2024; 198:104341. [PMID: 38575042 DOI: 10.1016/j.critrevonc.2024.104341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 03/13/2024] [Accepted: 03/28/2024] [Indexed: 04/06/2024] Open
Abstract
Extracellular vesicles (EVs) impact normal and pathological cellular signaling through bidirectional trafficking. Exosomes, a subset of EVs possess biomolecules including proteins, lipids, DNA fragments and various RNA species reflecting a speculum of their parent cells. The involvement of exosomes in bidirectional communication and their biological constituents substantiate its role in regulating both physiology and pathology, including multiple cancers. Non-small cell lung cancer (NSCLC) is the most common lung cancers (85%) with high incidence, mortality and reduced overall survival. Lack of efficient early diagnostic and therapeutic tools hurdles the management of NSCLC. Interestingly, the exosomes from body fluids similarity with parent cells or tissue offers a potential future multicomponent tool for the early diagnosis of NSCLC. The structural twinning of exosomes with a cell/tissue and the competitive tumor derived exosomes in tumor microenvironment (TME) promotes the unpinning horizons of exosomes as a drug delivery, vaccine, and therapeutic agent. Exosomes in clinical point of view assist to trace: acquired resistance caused by various therapeutic agents, early diagnosis, progression, and surveillance. In an integrated approach, EV biomarkers offer potential cutting-edge techniques for the detection and diagnosis of cancer, though the purification, characterization, and biomarker identification processes for the translational research regarding EVs need further optimization.
Collapse
Affiliation(s)
- Hafiza Padinharayil
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur-05, Kerala, India
| | - Alex George
- Jubilee Centre for Medical Research, Jubilee Mission Medical College and Research Institute, Thrissur-05, Kerala, India.
| |
Collapse
|
4
|
Wang Y, Lyu D, Hu S, Ma Y, Duan S, Geng Y, Zhou T, Tu W, Xiao Y, Fan L, Liu S. Nomogram using intratumoral and peritumoral radiomics for the preoperative prediction of visceral pleural invasion in clinical stage IA lung adenocarcinoma. J Cardiothorac Surg 2024; 19:307. [PMID: 38822379 PMCID: PMC11141037 DOI: 10.1186/s13019-024-02807-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Accurate prediction of visceral pleural invasion (VPI) in lung adenocarcinoma before operation can provide guidance and help for surgical operation and postoperative treatment. We investigate the value of intratumoral and peritumoral radiomics nomograms for preoperatively predicting the status of VPI in patients diagnosed with clinical stage IA lung adenocarcinoma. METHODS A total of 404 patients from our hospital were randomly assigned to a training set (n = 283) and an internal validation set (n = 121) using a 7:3 ratio, while 81 patients from two other hospitals constituted the external validation set. We extracted 1218 CT-based radiomics features from the gross tumor volume (GTV) as well as the gross peritumoral tumor volume (GPTV5, 10, 15), respectively, and constructed radiomic models. Additionally, we developed a nomogram based on relevant CT features and the radscore derived from the optimal radiomics model. RESULTS The GPTV10 radiomics model exhibited superior predictive performance compared to GTV, GPTV5, and GPTV15, with area under the curve (AUC) values of 0.855, 0.842, and 0.842 in the three respective sets. In the clinical model, the solid component size, pleural indentation, solid attachment, and vascular convergence sign were identified as independent risk factors among the CT features. The predictive performance of the nomogram, which incorporated relevant CT features and the GPTV10-radscore, outperformed both the radiomics model and clinical model alone, with AUC values of 0.894, 0.828, and 0.876 in the three respective sets. CONCLUSIONS The nomogram, integrating radiomics features and CT morphological features, exhibits good performance in predicting VPI status in lung adenocarcinoma.
Collapse
Affiliation(s)
- Yun Wang
- Department of Radiology, Second Affiliated Hospital of Navy Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Deng Lyu
- Department of Radiology, Second Affiliated Hospital of Navy Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Su Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yanqing Ma
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Shaofeng Duan
- GE Healthcare, Precision Health Institution, Shanghai, China
| | - Yayuan Geng
- Shukun(Beijing) Network Technology Co.,Ltd, Beijing, China
| | - Taohu Zhou
- Department of Radiology, Second Affiliated Hospital of Navy Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Wenting Tu
- Department of Radiology, Second Affiliated Hospital of Navy Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Yi Xiao
- Department of Radiology, Second Affiliated Hospital of Navy Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China
| | - Li Fan
- Department of Radiology, Second Affiliated Hospital of Navy Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China.
| | - Shiyuan Liu
- Department of Radiology, Second Affiliated Hospital of Navy Medical University, 415 Fengyang Road, Huangpu District, Shanghai, 200003, China.
| |
Collapse
|
5
|
Sáinz-Jaspeado M, Ring S, Proulx ST, Richards M, Martinsson P, Li X, Claesson-Welsh L, Ulvmar MH, Jin Y. VE-cadherin junction dynamics in initial lymphatic vessels promotes lymph node metastasis. Life Sci Alliance 2024; 7:e202302168. [PMID: 38148112 PMCID: PMC10751244 DOI: 10.26508/lsa.202302168] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 12/28/2023] Open
Abstract
The endothelial junction component vascular endothelial (VE)-cadherin governs junctional dynamics in the blood and lymphatic vasculature. Here, we explored how lymphatic junction stability is modulated by elevated VEGFA signaling to facilitate metastasis to sentinel lymph nodes. Zippering of VE-cadherin junctions was established in dermal initial lymphatic vessels after VEGFA injection and in tumor-proximal lymphatics in mice. Shape analysis of pan-cellular VE-cadherin fragments revealed that junctional zippering was accompanied by accumulation of small round-shaped VE-cadherin fragments in the lymphatic endothelium. In mice expressing a mutant VEGFR2 lacking the Y949 phosphosite (Vegfr2 Y949F/Y949F ) required for activation of Src family kinases, zippering of lymphatic junctions persisted, whereas accumulation of small VE-cadherin fragments was suppressed. Moreover, tumor cell entry into initial lymphatic vessels and subsequent metastatic spread to lymph nodes was reduced in mutant mice compared with WT, after challenge with B16F10 melanoma or EO771 breast cancer. We conclude that VEGFA mediates zippering of VE-cadherin junctions in initial lymphatics. Zippering is accompanied by increased VE-cadherin fragmentation through VEGFA-induced Src kinase activation, correlating with tumor dissemination to sentinel lymph nodes.
Collapse
Affiliation(s)
- Miguel Sáinz-Jaspeado
- Beijer and Science for Life Laboratories, Department Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Sarah Ring
- Beijer and Science for Life Laboratories, Department Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Steven T Proulx
- ETH Zürich, Institute of Pharmaceutical Sciences, Zürich, Switzerland
- Theodor Kocher Institute, University of Bern, Bern, Switzerland
| | - Mark Richards
- Beijer and Science for Life Laboratories, Department Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Pernilla Martinsson
- Beijer and Science for Life Laboratories, Department Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Xiujuan Li
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Lena Claesson-Welsh
- Beijer and Science for Life Laboratories, Department Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Maria H Ulvmar
- Beijer and Science for Life Laboratories, Department Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Yi Jin
- Beijer and Science for Life Laboratories, Department Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| |
Collapse
|
6
|
Aron A, Zavaleta C. Current and Developing Lymphatic Imaging Approaches for Elucidation of Functional Mechanisms and Disease Progression. Mol Imaging Biol 2024; 26:1-16. [PMID: 37195396 PMCID: PMC10827820 DOI: 10.1007/s11307-023-01827-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 05/05/2023] [Accepted: 05/08/2023] [Indexed: 05/18/2023]
Abstract
Study of the lymphatic system, compared to that of the other body systems, has been historically neglected. While scientists and clinicians have, in recent decades, gained a better appreciation of the functionality of the lymphatics as well as their role in associated diseases (and consequently investigated these topics further in their experimental work), there is still much left to be understood of the lymphatic system. In this review article, we discuss the role lymphatic imaging techniques have played in this recent series of advancements and how new imaging techniques can help bolster this wave of discovery. We specifically highlight the use of lymphatic imaging techniques in understanding the fundamental anatomy and physiology of the lymphatic system; investigating the development of lymphatic vasculature (using techniques such as intravital microscopy); diagnosing, staging, and treating lymphedema and cancer; and its role in other disease states.
Collapse
Affiliation(s)
- Arjun Aron
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, Los Angeles, CA, 90089, USA
- Michelson Center for Convergent Bioscience, University of Southern California, 1002 Childs Way, Los Angeles, CA, 90089, USA
| | - Cristina Zavaleta
- Department of Biomedical Engineering, University of Southern California, 1042 Downey Way, Los Angeles, CA, 90089, USA.
- Michelson Center for Convergent Bioscience, University of Southern California, 1002 Childs Way, Los Angeles, CA, 90089, USA.
| |
Collapse
|
7
|
Yang H, Wang W, Cheng Z, Zheng T, Cheng C, Cheng M, Wang Z. Radiomic Machine Learning in Invasive Ductal Breast Cancer: Prediction of Ki-67 Expression Level Based on Radiomics of DCE-MRI. Technol Cancer Res Treat 2024; 23:15330338241288751. [PMID: 39431304 PMCID: PMC11504335 DOI: 10.1177/15330338241288751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024] Open
Abstract
PURPOSE Our study aimed to investigate the potential of radiomics with DCE-MRI for predicting Ki-67 expression in invasive ductal breast cancer. METHOD We conducted a retrospective study including 223 patients diagnosed with invasive ductal breast cancer. Radiomics features were extracted from DCE-MRI using 3D-Slicer software. Two Ki-67 expression cutoff values (20% and 29%) were examined. Patients were divided into training (70%) and test (30%) sets. The Elastic Net method selected relevant features, and five machine-learning models were established. Radiomics models were created from intratumoral, peritumoral, and combined regions. Performance was assessed using ROC curves, accuracy, sensitivity, and specificity. RESULT For a Ki-67 cutoff value of 20%, the combined model exhibited the highest performance, with area under the curve (AUC) values of 0.838 (95% confidence interval (CI): 0.774-0.897) for the training set and 0.863 (95% CI: 0.764-0.949) for the test set. The AUC values for the tumor model were 0.816 (95% CI: 0.745-0.880) and 0.830 (95% CI: 0.724-0.916), and for the peritumor model were 0.790 (95% CI: 0.711-0.857) and 0.808 (95% CI: 0.682-0.910). When the Ki-67 cutoff value was set at 29%, the combined model also demonstrated superior predictive ability in both training set (AUC: 0.796; 95% CI: 0.724-0.862) and the test set (AUC: 0.823; 95% CI: 0.723-0.911). The AUC values for the tumor model were 0.785 (95% CI: 0.708-0.861) and 0.784 (95% CI: 0.663-0.882), and for the peritumor model were 0.773 (95% CI: 0.690-0.844) and 0.729 (95% CI: 0.603-0.847). CONCLUSION Radiomics with DCE-MRI can predict Ki-67 expression in invasive ductal breast cancer. Integrating radiomics features from intratumoral and peritumoral regions yields a dependable prognostic model, facilitating pre-surgical detection and treatment decisions. This holds potential for commercial diagnostic tools.
Collapse
Affiliation(s)
- Huan Yang
- Department of Emergency, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Wenxi Wang
- Department of Magnetic Resonance Imaging, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Zhiyong Cheng
- Department of Education, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Tao Zheng
- Department of Magnetic Resonance Imaging, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Cheng Cheng
- Department of Emergency, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Mengyu Cheng
- Department of Magnetic Resonance Imaging, First Hospital of Qinhuangdao, Qinhuangdao, China
| | - Zhanqiu Wang
- Department of Magnetic Resonance Imaging, First Hospital of Qinhuangdao, Qinhuangdao, China
| |
Collapse
|
8
|
Turati M, Mattei G, Boaretto A, Magi A, Calvani M, Ronca R. Molecular Profiling of Lymphatic Endothelial Cell Activation In Vitro. Int J Mol Sci 2023; 24:16587. [PMID: 38068914 PMCID: PMC10706153 DOI: 10.3390/ijms242316587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 11/18/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
The lymphatic vascular system plays a key role in cancer progression. Indeed, the activation of lymphatic endothelial cells (LECs) through the lymphangiogenic process allows for the formation of new lymphatic vessels (LVs) that represent the major route for the dissemination of solid tumors. This process is governed by a plethora of cancer-derived and microevironmental mediators that strictly activate and control specific molecular pathways in LECs. In this work we used an in vitro model of LEC activation to trigger lymphangiogenesis using a mix of recombinant pro-lymphangiogenic factors (VFS) and a co-culture system with human melanoma cells. Both systems efficiently activated LECs, and under these experimental conditions, RNA sequencing was exploited to unveil the transcriptional profile of activated LECs. Our data demonstrate that both recombinant and tumor cell-mediated activation trigger significant molecular pathways associated with endothelial activation, morphogenesis, and cytokine-mediated signaling. In addition, this system provides information on new genes to be further investigated in the lymphangiogenesis process and open the possibility for further exploitation in other tumor contexts where lymphatic dissemination plays a relevant role.
Collapse
Affiliation(s)
- Marta Turati
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| | - Gianluca Mattei
- Department of Information Engineering, University of Florence, 50139 Florence, Italy; (G.M.); (A.M.)
| | - Alessia Boaretto
- Department of Pediatric Hematology-Oncology, A. Meyer Children’s Hospital, Scientific Institute for Research, Hospitalisation and Health Care (IRCCS), 50139 Florence, Italy; (A.B.); (M.C.)
| | - Alberto Magi
- Department of Information Engineering, University of Florence, 50139 Florence, Italy; (G.M.); (A.M.)
| | - Maura Calvani
- Department of Pediatric Hematology-Oncology, A. Meyer Children’s Hospital, Scientific Institute for Research, Hospitalisation and Health Care (IRCCS), 50139 Florence, Italy; (A.B.); (M.C.)
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, 25123 Brescia, Italy;
| |
Collapse
|
9
|
Wang J, Gao W, Lu M, Yao X, Yang D. Development of an interpretable machine learning model for Ki-67 prediction in breast cancer using intratumoral and peritumoral ultrasound radiomics features. Front Oncol 2023; 13:1290313. [PMID: 38044998 PMCID: PMC10691503 DOI: 10.3389/fonc.2023.1290313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
Background Traditional immunohistochemistry assessment of Ki-67 in breast cancer (BC) via core needle biopsy is invasive, inaccurate, and nonrepeatable. While machine learning (ML) provides a promising alternative, its effectiveness depends on extensive data. Although the current mainstream MRI-centered radiomics offers sufficient data, its unsuitability for repeated examinations, along with limited accessibility and an intratumoral focus, constrain the application of predictive models in evaluating Ki-67 levels. Objective This study aims to explore ultrasound (US) image-based radiomics, incorporating both intra- and peritumoral features, to develop an interpretable ML model for predicting Ki-67 expression in BC patients. Methods A retrospective analysis was conducted on 263 BC patients, divided into training and external validation cohorts. From intratumoral and peritumoral regions of interest (ROIs) in US images, 849 distinctive radiomics features per ROI were derived. These features underwent systematic selection to analyze Ki-67 expression relationships. Four ML models-logistic regression, random forests, support vector machine (SVM), and extreme gradient boosting-were formulated and internally validated to identify the optimal predictive model. External validation was executed to ascertain the robustness of the optimal model, followed by employing Shapley Additive Explanations (SHAP) to reveal the significant features of the model. Results Among 231 selected BC patients, 67.5% exhibited high Ki-67 expression, with consistency observed across both training and validation cohorts as well as other clinical characteristics. Of the 1698 radiomics features identified, 15 were significantly correlated with Ki-67 expression. The SVM model, utilizing combined ROI, demonstrated the highest accuracy [area under the receiver operating characteristic curve (AUROC): 0.88], making it the most suitable for predicting Ki-67 expression. External validation sustained an AUROC of 0.82, affirming the model's robustness above a 40% threshold. SHAP analysis identified five influential features from intra- and peritumoral ROIs, offering insight into individual prediction. Conclusion This study emphasized the potential of SVM model using radiomics features from both intra- and peritumoral US images, for predicting elevated Ki-67 levels in BC patients. The model exhibited strong performance in validations, indicating its promise as a noninvasive tool to enable personalized decision-making in BC care.
Collapse
|
10
|
Bian X, Du S, Yue Z, Gao S, Zhao R, Huang G, Guo L, Peng C, Zhang L. Potential Antihuman Epidermal Growth Factor Receptor 2 Target Therapy Beneficiaries: The Role of MRI-Based Radiomics in Distinguishing Human Epidermal Growth Factor Receptor 2-Low Status of Breast Cancer. J Magn Reson Imaging 2023; 58:1603-1614. [PMID: 36763035 DOI: 10.1002/jmri.28628] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/21/2023] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
BACKGROUND Multiparametric MRI radiomics could distinguish human epidermal growth factor receptor 2 (HER2)-positive from HER2-negative breast cancers. However, its value for further distinguishing HER2-low from HER2-negative breast cancers has not been investigated. PURPOSE To investigate whether multiparametric MRI-based radiomics can distinguish HER2-positive from HER2-negative breast cancers (task 1) and HER2-low from HER2-negative breast cancers (task 2). STUDY TYPE Retrospective. POPULATION Task 1: 310 operable breast cancer patients from center 1 (97 HER2-positive and 213 HER2-negative); task 2: 213 HER2-negative patients (108 HER2-low and 105 HER2-zero); 59 patients from center 2 (16 HER2-positive, 27 HER2-low and 16 HER2-zero) for external validation. FIELD STRENGTH/SEQUENCE A 3.0 T/T1-weighted contrast-enhanced imaging (T1CE), diffusion-weighted imaging (DWI)-derived apparent diffusion coefficient (ADC). ASSESSMENT Patients in center 1 were assigned to a training and internal validation cohort at a 2:1 ratio. Intratumoral and peritumoral features were extracted from T1CE and ADC. After dimensionality reduction, the radiomics signatures (RS) of two tasks were developed using features from T1CE (RS-T1CE), ADC (RS-ADC) alone and T1CE + ADC combination (RS-Com). STATISTICAL TESTS Mann-Whitney U tests, the least absolute shrinkage and selection operator, receiver operating characteristic (ROC) curve, calibration curve, and decision curve analysis (DCA). RESULTS For task 1, RS-ADC yielded higher area under the ROC curve (AUC) in the training, internal, and external validation of 0.767/0.725/0.746 than RS-T1CE (AUC = 0.733/0.674/0.641). For task 2, RS-T1CE yielded higher AUC of 0.765/0.755/0.678 than RS-ADC (AUC = 0.706/0.608/0.630). For both of task 1 and task 2, RS-Com achieved the best performance with AUC of 0.793/0.778/0.760 and 0.820/0.776/0.711, respectively, and obtained higher clinical benefit in DCA compared with RS-T1CE and RS-ADC. The calibration curves of all RS demonstrated a good fitness. DATA CONCLUSION Multiparametric MRI radiomics could noninvasively and robustly distinguish HER2-positive from HER2-negative breast cancers and further distinguish HER2-low from HER2-negative breast cancers. EVIDENCE LEVEL 3. TECHNICAL EFFICACY Stage 2.
Collapse
Affiliation(s)
- Xiaoqian Bian
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Siyao Du
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhibin Yue
- School of Intelligent Medicine, China Medical University, Shenyang, China
| | - Si Gao
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Ruimeng Zhao
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Guoliang Huang
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Liangcun Guo
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Can Peng
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Lina Zhang
- Department of Radiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
11
|
Solorzano E, Alejo AL, Ball HC, Robinson GT, Solorzano AL, Safadi R, Douglas J, Kelly M, Safadi FF. The Lymphatic Endothelial Cell Secretome Inhibits Osteoblast Differentiation and Bone Formation. Cells 2023; 12:2482. [PMID: 37887326 PMCID: PMC10605748 DOI: 10.3390/cells12202482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 10/28/2023] Open
Abstract
Complex lymphatic anomalies (CLAs) are a set of rare diseases with unique osteopathic profiles. Recent efforts have identified how lymphatic-specific somatic activating mutations can induce abnormal lymphatic formations that are capable of invading bone and inducing bone resorption. The abnormal bone resorption in CLA patients has been linked to overactive osteoclasts in areas with lymphatic invasions. Despite these findings, the mechanism associated with progressive bone loss in CLAs remains to be elucidated. In order to determine the role of osteoblasts in CLAs, we sought to assess osteoblast differentiation and bone formation when exposed to the lymphatic endothelial cell secretome. When treated with lymphatic endothelial cell conditioned medium (L-CM), osteoblasts exhibited a significant decrease in proliferation, differentiation, and function. Additionally, L-CM treatment also inhibited bone formation through a neonatal calvaria explant culture. These findings are the first to reveal how osteoblasts may be actively suppressed during bone lymphatic invasion in CLAs.
Collapse
Affiliation(s)
- Ernesto Solorzano
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, OH 44272, USA; (E.S.); (A.L.A.); (H.C.B.); (G.T.R.); (A.L.S.)
- Musculoskeletal Research Group, NEOMED, Rootstown, OH 44272, USA;
- Basic and Translational Biomedicine (BTB) Graduate Program, College of Graduate Studies, NEOMED, Rootstown, OH 44272, USA;
| | - Andrew L. Alejo
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, OH 44272, USA; (E.S.); (A.L.A.); (H.C.B.); (G.T.R.); (A.L.S.)
| | - Hope C. Ball
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, OH 44272, USA; (E.S.); (A.L.A.); (H.C.B.); (G.T.R.); (A.L.S.)
- Musculoskeletal Research Group, NEOMED, Rootstown, OH 44272, USA;
- Basic and Translational Biomedicine (BTB) Graduate Program, College of Graduate Studies, NEOMED, Rootstown, OH 44272, USA;
| | - Gabrielle T. Robinson
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, OH 44272, USA; (E.S.); (A.L.A.); (H.C.B.); (G.T.R.); (A.L.S.)
- Musculoskeletal Research Group, NEOMED, Rootstown, OH 44272, USA;
- Basic and Translational Biomedicine (BTB) Graduate Program, College of Graduate Studies, NEOMED, Rootstown, OH 44272, USA;
| | - Andrea L. Solorzano
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, OH 44272, USA; (E.S.); (A.L.A.); (H.C.B.); (G.T.R.); (A.L.S.)
| | - Rama Safadi
- College of Arts and Sciences, Kent State University, Kent, OH 44243, USA;
| | - Jacob Douglas
- Musculoskeletal Research Group, NEOMED, Rootstown, OH 44272, USA;
| | - Michael Kelly
- Basic and Translational Biomedicine (BTB) Graduate Program, College of Graduate Studies, NEOMED, Rootstown, OH 44272, USA;
- Department of Pediatric Hematology Oncology and Blood, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Fayez F. Safadi
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, OH 44272, USA; (E.S.); (A.L.A.); (H.C.B.); (G.T.R.); (A.L.S.)
- Musculoskeletal Research Group, NEOMED, Rootstown, OH 44272, USA;
- Basic and Translational Biomedicine (BTB) Graduate Program, College of Graduate Studies, NEOMED, Rootstown, OH 44272, USA;
- Rebecca D. Considine Research Institute, Akron Children’s Hospital, Akron, OH 44308, USA
| |
Collapse
|
12
|
Qiu Y, Liu YF, Shu X, Qiao XF, Ai GY, He XJ. Peritumoral Radiomics Strategy Based on Ensemble Learning for the Prediction of Gleason Grade Group of Prostate Cancer. Acad Radiol 2023; 30 Suppl 1:S1-S13. [PMID: 37393175 DOI: 10.1016/j.acra.2023.06.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 07/03/2023]
Abstract
RATIONALE AND OBJECTIVES To develop and evaluate a peritumoral radiomic-based machine learning model to differentiate low-Gleason grade group (L-GGG) and high-GGG (H-GGG) prostate lesions. MATERIALS AND METHODS In this retrospective study, a total of 175 patients with prostate cancer (PCa) confirmed by puncture biopsy were recruited and included 59 patients with L-GGG and 116 patients with H-GGG. The original PCa regions of interest (ROIs) were delineated on T2-weighted (T2WI), diffusion-weighted imaging (DWI), and apparent diffusion coefficient (ADC) maps, and then centra-tumoral and peritumoral ROIs were defined. Features were meticulously extracted from each ROI to establish radiomics models, employing distinct sequence datasets. Peritumoral radiomics models were specifically developed for both the peripheral zone (PZ) and transitional zone (TZ), utilizing dedicated PZ and TZ datasets, respectively. The performances of the models were evaluated by using the receiver operating characteristic (ROC) curve and precision-recall curve. RESULTS The classification model with combined peritumoral features based on T2 + DWI + ADC sequence dataset demonstrated superior performance compared to the original tumor and centra-tumoral classification models. It achieved an area under the ROC curve (AUC) of 0.850 [95% confidence interval, 0.849, 0.860] and an average accuracy of 0.950. The combined peritumoral model outperformed the regional peritumoral models with AUC of 0.85 versus 0.75 for PZ lesions and 0.88 versus 0.69 for TZ lesions, respectively. The peritumoral classification models exhibit greater efficacy in predicting PZ lesions as opposed to TZ lesions. CONCLUSION The peritumoral radiomics features showed excellent performance in predicting GGG in PCa patients and might be a valuable addition to the non-invasive assessment of PCa aggressiveness.
Collapse
Affiliation(s)
- Yang Qiu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Yun-Fan Liu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Xin Shu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Xiao-Feng Qiao
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Guang-Yong Ai
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | - Xiao-Jing He
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
13
|
Huang CL, Achudhan D, Liu PI, Lin YY, Liu SC, Guo JH, Liu CL, Wu CY, Wang SW, Tang CH. Visfatin upregulates VEGF-C expression and lymphangiogenesis in esophageal cancer by activating MEK1/2-ERK and NF-κB signaling. Aging (Albany NY) 2023; 15:204762. [PMID: 37286356 DOI: 10.18632/aging.204762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/17/2023] [Indexed: 06/09/2023]
Abstract
Lymph node metastasis is a recognized prognostic factor in esophageal cancer. Adipokines, including visfatin, and the molecule vascular endothelial growth factor (VEGF)-C, are implicated in lymphangiogenesis, but whether any association exists between esophageal cancer, adipokines and VEGF-C is unknown. We examined the relevance of adipokines and VEGF-C in esophageal squamous cell carcinoma (ESCC) in the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. We found significantly higher levels of visfatin and VEGF-C expression in esophageal cancer tissue than in normal tissue. Immunohistochemistry (IHC) staining identified that higher levels of visfatin and VEGF-C expression were correlated with advanced stage ESCC. Visfatin treatment of ESCC cell lines upregulated VEGF-C expression and VEGF-C-dependent lymphangiogenesis in lymphatic endothelial cells. Visfatin induced increases in VEGF-C expression by activating the mitogen-activated protein kinase kinases1/2-extracellular signal-regulated kinase (MEK1/2-ERK) and Nuclear Factor Kappa B (NF-κB) signaling cascades. Transfecting ESCC cells with MEK1/2-ERK and NF-κB inhibitors (PD98059, FR180204, PDTC, and TPCK) and siRNAs inhibited visfatin-induced increases in VEGF-C expression. It appears that visfatin and VEGF-C are promising therapeutic targets in the inhibition of lymphangiogenesis in esophageal cancer.
Collapse
Affiliation(s)
- Chang-Lun Huang
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Surgery, Division of Thoracic Surgery, Changhua Christian Hospital, Changhua 500, Taiwan
| | - David Achudhan
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Po-I Liu
- Department of General Thoracic Surgery, Asia University Hospital, Taichung 41354, Taiwan
- Department of Physical Therapy, Asia University, Taichung 41354, Taiwan
| | - Yen-You Lin
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan
| | - Shan-Chi Liu
- Department of Medical Education and Research, China Medical University Beigang Hospital, Yunlin 65152, Taiwan
| | - Jeng-Hung Guo
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Neurosurgery, China Medical University Hospital, Taichung 404327, Taiwan
| | - Chun-Lin Liu
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Neurosurgery, China Medical University Hospital, Taichung 404327, Taiwan
| | - Chih-Ying Wu
- Department of Neurosurgery, China Medical University Hospital, Taichung 404327, Taiwan
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 406040, Taiwan
- Department of Neurosurgery, China Medical University Hsinchu Hospital, Hsinchu 302, Taiwan
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei 252, Taiwan
- College of Pharmacy, Graduate Institute of Natural Products, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Biomedical Science, College of Medicine, China Medical University, Taichung 40402, Taiwan
- Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 406040, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Health Science, Asia University, Taichung 41354, Taiwan
- Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu 302, Taiwan
| |
Collapse
|
14
|
Sansone C, Pistelli L, Calabrone L, Del Mondo A, Fontana A, Festa M, Noonan DM, Albini A, Brunet C. The Carotenoid Diatoxanthin Modulates Inflammatory and Angiogenesis Pathways In Vitro in Prostate Cancer Cells. Antioxidants (Basel) 2023; 12:antiox12020359. [PMID: 36829917 PMCID: PMC9952135 DOI: 10.3390/antiox12020359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/23/2023] [Accepted: 01/27/2023] [Indexed: 02/05/2023] Open
Abstract
Xanthophylls, a group of carotenoids, have attracted attention as human health benefit compounds thanks to their functionality and bioavailability. The great antioxidant and anti-inflammatory abilities of diatoxanthin (Dt), a photoprotective xanthophyll synthetized by diatoms, were recently documented. This study investigates the capacity of Dt to intercept prostate cancer progression in vitro on different human cell lines, exploring its role against cancer proliferation and angiogenesis. Our results highlighted the chemopreventive role of Dt already at low concentration (44.1 pM) and suggest that the Dt-induced cancer cell death occurred through oxidative stress mechanisms. This hypothesis was supported by variations on the expression of key genes and proteins. Oxidative stress cell deaths (e.g., ferroptosis) are recently described types of cell death that are closely related to the pathophysiological processes of many diseases, such as tumors. Nonetheless, the interest of Dt was further strengthened by its ability to inhibit angiogenesis. The results are discussed considering the actual progress and requirements in cancer therapy, notably for prostate cancer.
Collapse
Affiliation(s)
- Clementina Sansone
- Stazione Zoologica Anton Dohrn, Sede Molosiglio Marina Acton, 80133 Napoli, Italy
- Laboratory of Vascular Biology and Angiogenesis, IRCCS MultiMedica, 20138 Milan, Italy
- Correspondence: (C.S.); (C.B.)
| | - Luigi Pistelli
- Stazione Zoologica Anton Dohrn, Sede Molosiglio Marina Acton, 80133 Napoli, Italy
| | - Luana Calabrone
- Laboratory of Vascular Biology and Angiogenesis, IRCCS MultiMedica, 20138 Milan, Italy
| | - Angelo Del Mondo
- Stazione Zoologica Anton Dohrn, Sede Molosiglio Marina Acton, 80133 Napoli, Italy
| | - Angelo Fontana
- Institute of Biomolecular Chemistry, Italian National Research Council (CNR), 80078 Pozzuoli, Italy
| | - Marco Festa
- Laboratory of Vascular Biology and Angiogenesis, IRCCS MultiMedica, 20138 Milan, Italy
| | - Douglas M. Noonan
- Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy
- Unit of Molecular Pathology, Biochemistry and Immunology, IRCCS MultiMedica, 20138 Milan, Italy
| | - Adriana Albini
- IRCSS European Institute of Oncology (IEO), 20141 Milan, Italy
| | - Christophe Brunet
- Stazione Zoologica Anton Dohrn, Sede Molosiglio Marina Acton, 80133 Napoli, Italy
- Correspondence: (C.S.); (C.B.)
| |
Collapse
|
15
|
Roy R, Singh SK, Misra S. Advancements in Cancer Immunotherapies. Vaccines (Basel) 2022; 11:vaccines11010059. [PMID: 36679904 PMCID: PMC9861770 DOI: 10.3390/vaccines11010059] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Recent work has suggested involvement of the immune system in biological therapies specifically targeting tumor microenvironment. Substantial advancement in the treatment of malignant tumors utilizing immune cells, most importantly T cells that play a key role in cell-mediated immunity, have led to success in clinical trials. Therefore, this article focuses on the therapeutic approaches and developmental strategies to treat cancer. This review emphasizes the immunomodulatory response, the involvement of key tumor-infiltrating cells, the mechanistic aspects, and prognostic biomarkers. We also cover recent advancements in therapeutic strategies.
Collapse
Affiliation(s)
- Ruchi Roy
- UICentre for Drug Discovery, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
- Correspondence:
| | - Sunil Kumar Singh
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Sweta Misra
- UICentre for Drug Discovery, College of Pharmacy, University of Illinois at Chicago, Chicago, IL 60607, USA
| |
Collapse
|
16
|
Nurlaila I, Roh K, Yeom CH, Kang H, Lee S. Acquired lymphedema: Molecular contributors and future directions for developing intervention strategies. Front Pharmacol 2022; 13:873650. [PMID: 36386144 PMCID: PMC9640931 DOI: 10.3389/fphar.2022.873650] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 10/13/2022] [Indexed: 08/05/2023] Open
Abstract
Lymphedema is a debilitating chronic disease that mostly develops as an adverse reaction to cancer treatment modalities such as chemotherapy, surgery, and radiotherapy. Lymphedema also appears to be a deteriorating consequence of roundworm infections, as best represented by filariasis. According to its origin, lymphedema is classified as primary lymphedema and acquired lymphedema. The latter is an acquired condition that, hitherto, received a considerably low attention owing to the less number of fatal cases been reported. Notably, despite the low mortality rate in lymphedema, it has been widely reported to reduce the disease-free survival and thus the quality of life of affected patients. Hence, in this review, we focused on acquired lymphedema and orchestration of molecular interplays associated with either stimulation or inhibition of lymphedema development that were, in vast majority, clearly depicted in animal models with their specific and distinct technical approaches. We also discussed some recent progress made in phytochemical-based anti-lymphedema intervention strategies and the specific mechanisms underlying their anti-lymphedema properties. This review is crucial to understand not only the comprehensive aspects of the disease but also the future directions of the intervention strategies that can address the quality of life of affected patients rather than alleviating apparent symptoms only.
Collapse
Affiliation(s)
- Ika Nurlaila
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
- Department of Vaccine and Drugs, The National Research and Innovation Agency, Jakarta, Indonesia
| | - Kangsan Roh
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Division of Cardiology and Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | | | - Hee Kang
- Humanitas College, Kyung Hee University, Yongin, South Korea
| | - Sukchan Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
17
|
Visible Lymph Affluents in the D3 Volume: An MDCTA Pictorial Essay. Diagnostics (Basel) 2022; 12:diagnostics12102441. [PMID: 36292133 PMCID: PMC9600001 DOI: 10.3390/diagnostics12102441] [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: 09/18/2022] [Revised: 10/05/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND There seems to be a gap in knowledge of the anatomy of mesenteric lymphatics between the superior mesenteric nodes and the intestinal trunk. To our knowledge, these central lymph vessels were not hitherto systematically searched for, described, or morphometrically analyzed. Our aim was to identify those vessels on the routine multidetector computerized tomography angiography (MDCTA), performed prior to right colectomy for cancer, with extended mesenterectomy, central vascular ligation, and D3 lymphadenectomy. METHODS A total of 420 MDCTA datasets were analyzed utilizing manual segmentation and 3D reconstruction, with the aid of image processing software Osirix, Mimics, and 3-matic. The 3D models and masks underwent a detailed topographic and morphometric analysis. RESULTS Significant vascular-like structures, having neither origin nor termination on the blood vessels, were noted in 18 cases (4.3%) in the D3 volume. The dimensions of visible lymph vessels varied, their mean diameter was 1.81 ± 0.61 mm, and the mean length was 38.07 ± 22.19 mm. In the vast majority of cases, the lymph vessels were situated in front of the superior mesenteric artery (SMA), coursing either longitudinally cranially (13 cases) or transversely/obliquely to the left (5 cases). In all cases but one, the lymph vessel passed at the left-hand side of the middle colic artery. As for the course shape, in seven cases, the lymph vessel appeared highly serpiginous. CONCLUSIONS The regular MDCTA can provide valuable information on mesenteric lymphatics and aid in surgical planning.
Collapse
|
18
|
Yan L, Wen C, Lu Q, Jing L, Mao W, Shen X, Zheng F, Wang W, Ma Y, Huang B. Quantitative Indicators of Retraction Phenomenon on an Automated Breast Volume Scanner: Initial Study in the Diagnosis and Prognostic Prediction of Breast Tumors. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1496-1508. [PMID: 35618533 DOI: 10.1016/j.ultrasmedbio.2022.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/15/2022] [Accepted: 03/19/2022] [Indexed: 06/15/2023]
Abstract
Retraction phenomenon is a unique sign on an automated breast volume scanner coronal plane image and has high specificity in differentiating benign lesions from malignant breast cancer. The purpose of this study was to quantify the retraction phenomenon by setting different rules to describe connected regions from different dimensions. In total, six quantitative indicators (FΩ1,FΓ,FS,FΩ2,FΩ3and FL) were obtained. FΩ1, FΩ2 and FΩ3 represent the relative areas of the connected region under different rules. FΓandFS represent the number ratio and absolute area of the connected region, respectively. FL represents the ratio of edge numbers. Two hundred fourteen patients with 214 lesions (90 benign and 124 malignant) were enrolled in this study. All quantitative indicators in the malignant group were significantly higher than those in the benign group (all p values <0.001). The indicator FΓ achieved the highest area under the receiver operating characteristic curve (AUC) (0.701, 95% confidence interval: 0.631-0.771). Both FΓ and FS had significant associations with axillary lymph node metastasis (p = 0.023 and 0.049). Compared with the classic texture feature gray-level co-occurrence matrix, retraction phenomenon quantization improved the AUC by 8.3%. The results indicate that retraction phenomenon quantitative indicators have certain value in distinguishing benign and malignant breast lesions and seem to be associated with axillary lymph node metastasis.
Collapse
Affiliation(s)
- Lixia Yan
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China
| | - Chuan Wen
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Qing Lu
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China
| | - Luxia Jing
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China
| | - Wujian Mao
- Department of Nuclear Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xinmeng Shen
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Fengyang Zheng
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China
| | - Wenping Wang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China
| | - Yu Ma
- School of Information Science and Technology, Fudan University, Shanghai, China
| | - Beijian Huang
- Department of Ultrasound, Zhongshan Hospital, Fudan University, Shanghai, China; Shanghai Institute of Medical Imaging, Shanghai, China.
| |
Collapse
|
19
|
Singla B, Aithabathula RV, Kiran S, Kapil S, Kumar S, Singh UP. Reactive Oxygen Species in Regulating Lymphangiogenesis and Lymphatic Function. Cells 2022; 11:1750. [PMID: 35681445 PMCID: PMC9179518 DOI: 10.3390/cells11111750] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/18/2022] [Accepted: 05/24/2022] [Indexed: 11/17/2022] Open
Abstract
The lymphatic system is pivotal for immunosurveillance and the maintenance of tissue homeostasis. Lymphangiogenesis, the formation of new lymphatic vessels from pre-existing vessels, has both physiological and pathological roles. Recent advances in the molecular mechanisms regulating lymphangiogenesis have opened a new area of research on reparative lymphangiogenesis for the treatment of various pathological disorders comprising neurological disorders, cardiac repair, autoimmune disease, obesity, atherosclerosis, etc. Reactive oxygen species (ROS) produced by the various cell types serve as signaling molecules in several cellular mechanisms and regulate various aspects of growth-factor-mediated responses, including lymphangiogenesis. The ROS, including superoxide anion, hydrogen peroxide, and nitric oxide, play both beneficial and detrimental roles depending upon their levels and cellular microenvironment. Low ROS levels are essential for lymphangiogenesis. On the contrary, oxidative stress due to enhanced ROS generation and/or reduced levels of antioxidants suppresses lymphangiogenesis via promoting lymphatic endothelial cell apoptosis and death. In this review article, we provide an overview of types and sources of ROS, discuss the role of ROS in governing lymphangiogenesis and lymphatic function, and summarize the role of lymphatics in various diseases.
Collapse
Affiliation(s)
- Bhupesh Singla
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Ravi Varma Aithabathula
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Sonia Kiran
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Shweta Kapil
- Division of Gastroenterology, Hepatology and Nutrition, Cincinnati Children′s Hospital Medical Center, Cincinnati, OH 45229, USA;
| | - Santosh Kumar
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| | - Udai P. Singh
- Department of Pharmaceutical Sciences, The University of Tennessee Health Science Center, Memphis, TN 38017, USA; (R.V.A.); (S.K.); (S.K.); (U.P.S.)
| |
Collapse
|
20
|
Harris AR, Esparza S, Azimi MS, Cornelison R, Azar FN, Llaneza DC, Belanger M, Mathew A, Tkachenko S, Perez MJ, Rosean CB, Bostic RR, Cornelison RC, Tate KM, Peirce-Cottler SM, Paquette C, Mills A, Landen CN, Saucerman J, Dillon PM, Pompano RR, Rutkowski MA, Munson JM. Platinum Chemotherapy Induces Lymphangiogenesis in Cancerous and Healthy Tissues That Can be Prevented With Adjuvant Anti-VEGFR3 Therapy. Front Oncol 2022; 12:801764. [PMID: 35372032 PMCID: PMC8970967 DOI: 10.3389/fonc.2022.801764] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/31/2022] [Indexed: 11/16/2022] Open
Abstract
Chemotherapy has been used to inhibit cancer growth for decades, but emerging evidence shows it can affect the tumor stroma, unintentionally promoting cancer malignancy. After treatment of primary tumors, remaining drugs drain via lymphatics. Though all drugs interact with the lymphatics, we know little of their impact on them. Here, we show a previously unknown effect of platinums, a widely used class of chemotherapeutics, to directly induce systemic lymphangiogenesis and activation. These changes are dose-dependent, long-lasting, and occur in healthy and cancerous tissue in multiple mouse models of breast cancer. We found similar effects in human ovarian and breast cancer patients whose treatment regimens included platinums. Carboplatin treatment of healthy mice prior to mammary tumor inoculation increased cancer metastasis as compared to no pre-treatment. These platinum-induced phenomena could be blocked by VEGFR3 inhibition. These findings have implications for cancer patients receiving platinums and may support the inclusion of anti-VEGFR3 therapy into treatment regimens or differential design of treatment regimens to alter these potential effects.
Collapse
Affiliation(s)
- Alexandra R Harris
- Department of Obstetrics and Gynecology, Gynecologic Oncology Division, University of Virginia, Charlottesville, VA, United States.,Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Savieay Esparza
- Department of Biomedical Engineering & Mechanics, Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Mohammad S Azimi
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Robert Cornelison
- Department of Obstetrics and Gynecology, Gynecologic Oncology Division, University of Virginia, Charlottesville, VA, United States.,Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Francesca N Azar
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Danielle C Llaneza
- Department of Obstetrics and Gynecology, Gynecologic Oncology Division, University of Virginia, Charlottesville, VA, United States
| | - Maura Belanger
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Alexander Mathew
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Svyatoslav Tkachenko
- Department of Genetics & Genome Sciences, Lerner Research Institute, Cleveland, OH, United States
| | - Matthew J Perez
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Claire Buchta Rosean
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Raegan R Bostic
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - R Chase Cornelison
- Department of Biomedical Engineering & Mechanics, Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Kinsley M Tate
- Department of Biomedical Engineering & Mechanics, Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, VA, United States
| | - Shayn M Peirce-Cottler
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Cherie Paquette
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States.,Department of Pathology and Laboratory Medicine, Women & Infants Hospital of Rhode Island, Providence, RI, United States
| | - Anne Mills
- Department of Pathology, University of Virginia, Charlottesville, VA, United States
| | - Charles N Landen
- Department of Obstetrics and Gynecology, Gynecologic Oncology Division, University of Virginia, Charlottesville, VA, United States
| | - Jeff Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Patrick M Dillon
- Department of Hematology and Oncology, University of Virginia, Charlottesville, VA, United States
| | - Rebecca R Pompano
- Department of Chemistry, University of Virginia, Charlottesville, VA, United States
| | - Melanie A Rutkowski
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, United States
| | - Jennifer M Munson
- Department of Biomedical Engineering & Mechanics, Fralin Biomedical Research Institute, Virginia Polytechnic Institute and State University, Roanoke, VA, United States.,Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, United States
| |
Collapse
|
21
|
Energy Sources for Exosome Communication in a Cancer Microenvironment. Cancers (Basel) 2022; 14:cancers14071698. [PMID: 35406470 PMCID: PMC8996881 DOI: 10.3390/cancers14071698] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Exosomal communication in the tumor microenvironment plays a crucial role in cancer development, progression, and metastasis, and is achieved by either short-distance communication with neighboring cells or long-distance communication with distant organs. Nevertheless, how exosomes gain energy to establish such communication and the different sources of energy are unclear. Recently, a handful of studies have demonstrated the presence of mitochondria, adenosine triphosphate, and glycolytic enzymes, which may serve as potential energy sources for exosomes. This review clarifies how exosomes maintain their structural integrity and stability during their intracellular communication, and reviews evidence of their energy source. Abstract Exosomes are crucial extracellular vesicles (EVs) with a diameter of approximately 30–200 nm. They are released by most cell types in their extracellular milieu and carry various biomolecules, including proteins and nucleic acids. Exosomes are increasingly studied in various diseases, including cancer, due to their role in local and distant cell–cell communication in which they can promote tumor growth, cancer progression, and metastasis. Interestingly, a tremendous number of exosomes is released by malignant cancer cells, and these are then taken up by autologous and heterologous recipient stromal cells such as immune cells, cancer stem cells, and endothelial cells. All these events demand an enormous amount of energy and require that exosomes remain stable while having the capacity to reach distant sites and cross physical barriers. Nevertheless, there is a dearth of research pertaining to the energy sources of exosomes, and questions remain about how they maintain their motility in the tumor microenvironment (TME) and beyond. Moreover, exosomes can produce adenosine triphosphate (ATP), an important energy molecule required by all cells, and mitochondria have been identified as one of the exosomal cargoes. These findings strengthen the prospect of exosomal communication via transfer of mitochondria and the bioenergetics of target recipient cells. In the TME, the accumulation of ATP and lactate may facilitate the entry of exosomes into cancer cells to promote metastasis, as well as help to target cancer cells at the tumor site. This review highlights how exosomes obtain sufficient energy to thrive in the TME and communicate with distant physiological destinations.
Collapse
|
22
|
Bekisz S, Baudin L, Buntinx F, Noël A, Geris L. In Vitro, In Vivo, and In Silico Models of Lymphangiogenesis in Solid Malignancies. Cancers (Basel) 2022; 14:1525. [PMID: 35326676 PMCID: PMC8946816 DOI: 10.3390/cancers14061525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/24/2022] [Accepted: 03/08/2022] [Indexed: 12/04/2022] Open
Abstract
Lymphangiogenesis (LA) is the formation of new lymphatic vessels by lymphatic endothelial cells (LECs) sprouting from pre-existing lymphatic vessels. It is increasingly recognized as being involved in many diseases, such as in cancer and secondary lymphedema, which most often results from cancer treatments. For some cancers, excessive LA is associated with cancer progression and metastatic dissemination to the lymph nodes (LNs) through lymphatic vessels. The study of LA through in vitro, in vivo, and, more recently, in silico models is of paramount importance in providing novel insights and identifying the key molecular actors in the biological dysregulation of this process under pathological conditions. In this review, the different biological (in vitro and in vivo) models of LA, especially in a cancer context, are explained and discussed, highlighting their principal modeled features as well as their advantages and drawbacks. Imaging techniques of the lymphatics, complementary or even essential to in vivo models, are also clarified and allow the establishment of the link with computational approaches. In silico models are introduced, theoretically described, and illustrated with examples specific to the lymphatic system and the LA. Together, these models constitute a toolbox allowing the LA research to be brought to the next level.
Collapse
Affiliation(s)
- Sophie Bekisz
- Biomechanics Research Unit, GIGA In silico Medicine, ULiège, 4000 Liège, Belgium;
| | - Louis Baudin
- Laboratory of Biology of Tumor and Development, GIGA Cancer, ULiège, 4000 Liège, Belgium; (L.B.); (F.B.); (A.N.)
| | - Florence Buntinx
- Laboratory of Biology of Tumor and Development, GIGA Cancer, ULiège, 4000 Liège, Belgium; (L.B.); (F.B.); (A.N.)
| | - Agnès Noël
- Laboratory of Biology of Tumor and Development, GIGA Cancer, ULiège, 4000 Liège, Belgium; (L.B.); (F.B.); (A.N.)
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA In silico Medicine, ULiège, 4000 Liège, Belgium;
- Biomechanics Section, KU Leuven, 3000 Leuven, Belgium
- Skeletal Biology and Engineering Research Center, KU Leuven, 3000 Leuven, Belgium
| |
Collapse
|
23
|
Huang L, Lin W, Xie D, Yu Y, Cao H, Liao G, Wu S, Yao L, Wang Z, Wang M, Wang S, Wang G, Zhang D, Yao S, He Z, Cho WCS, Chen D, Zhang Z, Li W, Qiao G, Chan LWC, Zhou H. Development and validation of a preoperative CT-based radiomic nomogram to predict pathology invasiveness in patients with a solitary pulmonary nodule: a machine learning approach, multicenter, diagnostic study. Eur Radiol 2022; 32:1983-1996. [PMID: 34654966 PMCID: PMC8831242 DOI: 10.1007/s00330-021-08268-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 07/23/2021] [Accepted: 08/06/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVES To develop and validate a preoperative CT-based nomogram combined with radiomic and clinical-radiological signatures to distinguish preinvasive lesions from pulmonary invasive lesions. METHODS This was a retrospective, diagnostic study conducted from August 1, 2018, to May 1, 2020, at three centers. Patients with a solitary pulmonary nodule were enrolled in the GDPH center and were divided into two groups (7:3) randomly: development (n = 149) and internal validation (n = 54). The SYSMH center and the ZSLC Center formed an external validation cohort of 170 patients. The least absolute shrinkage and selection operator (LASSO) algorithm and logistic regression analysis were used to feature signatures and transform them into models. RESULTS The study comprised 373 individuals from three independent centers (female: 225/373, 60.3%; median [IQR] age, 57.0 [48.0-65.0] years). The AUCs for the combined radiomic signature selected from the nodular area and the perinodular area were 0.93, 0.91, and 0.90 in the three cohorts. The nomogram combining the clinical and combined radiomic signatures could accurately predict interstitial invasion in patients with a solitary pulmonary nodule (AUC, 0.94, 0.90, 0.92) in the three cohorts, respectively. The radiomic nomogram outperformed any clinical or radiomic signature in terms of clinical predictive abilities, according to a decision curve analysis and the Akaike information criteria. CONCLUSIONS This study demonstrated that a nomogram constructed by identified clinical-radiological signatures and combined radiomic signatures has the potential to precisely predict pathology invasiveness. KEY POINTS • The radiomic signature from the perinodular area has the potential to predict pathology invasiveness of the solitary pulmonary nodule. • The new radiomic nomogram was useful in clinical decision-making associated with personalized surgical intervention and therapeutic regimen selection in patients with early-stage non-small-cell lung cancer.
Collapse
Affiliation(s)
- Luyu Huang
- Division of Thoracic Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, The Second School of Clinical Medicine, Southern Medical University, Shantou University Medical College, Guangzhou, China
| | - Weihuan Lin
- Division of Thoracic Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, The Second School of Clinical Medicine, Southern Medical University, Shantou University Medical College, Guangzhou, China
| | - Daipeng Xie
- Division of Thoracic Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, The Second School of Clinical Medicine, Southern Medical University, Shantou University Medical College, Guangzhou, China
| | - Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
- AI & Digital Media Concentration Program, Division of Science and Technology, Beijing Normal University-Hong Kong Baptist University United International College, Zhuhai, China
| | - Hanbo Cao
- Department of Radiology, Zhoushan Hospital, Zhoushan City, Zhejiang Province, China
| | - Guoqing Liao
- Division of Thoracic Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, The Second School of Clinical Medicine, Southern Medical University, Shantou University Medical College, Guangzhou, China
| | - Shaowei Wu
- Division of Thoracic Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, The Second School of Clinical Medicine, Southern Medical University, Shantou University Medical College, Guangzhou, China
| | - Lintong Yao
- Division of Thoracic Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, The Second School of Clinical Medicine, Southern Medical University, Shantou University Medical College, Guangzhou, China
| | - Zhaoyu Wang
- Department of Pathology, Zhoushan Hospital, Zhoushan City, Zhejiang Province, China
| | - Mei Wang
- Department of Radiology, Department of PET Center, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Siyun Wang
- Department of Radiology, Department of PET Center, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guangyi Wang
- Department of Radiology, Department of PET Center, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Dongkun Zhang
- Division of Thoracic Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, The Second School of Clinical Medicine, Southern Medical University, Shantou University Medical College, Guangzhou, China
| | - Su Yao
- Department of Pathology, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zifan He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Phase I Clinical Trial Centre, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | | | - Duo Chen
- Department of Respiratory and Critical Care Medicine, Beijing Institute of Respiratory Medicine, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Zhengjie Zhang
- Division of Thoracic Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, The Second School of Clinical Medicine, Southern Medical University, Shantou University Medical College, Guangzhou, China
| | - Wanshan Li
- Clinical Medicine, Zhongshan School of Medicine, Yat-Sen University, Guangzhou, China
| | - Guibin Qiao
- Division of Thoracic Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, The Second School of Clinical Medicine, Southern Medical University, Shantou University Medical College, Guangzhou, China.
| | - Lawrence Wing-Chi Chan
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China.
| | - Haiyu Zhou
- Division of Thoracic Surgery, Guangdong Provincial People's Hospital & Guangdong Academy of Medical Sciences, The Second School of Clinical Medicine, Southern Medical University, Shantou University Medical College, Guangzhou, China.
| |
Collapse
|
24
|
Martin SG, Rakha E, Storr SJ. Quantifying Lymphatic Vessel Density in Human Tissue Samples. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2441:183-189. [PMID: 35099737 DOI: 10.1007/978-1-0716-2059-5_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The development of new blood and lymphatic vessels, through the process of angiogenesis and lymphangiogenesis, respectively, is critical to the development and growth of tumors, and integral to the process of metastasis. Lymphatic vessel density can be assessed as a surrogate measure of lymphangiogenesis in human tissue samples. Lymphatic vessel density has been shown to be associated with lymph node metastasis and patient survival in various solid tumor types. Here we describe a method for quantifying the number of lymphatic vessels within tumor tissue that can also be used to assess lymphatic vessel invasion, and compare with blood vessel density and invasion.
Collapse
Affiliation(s)
- Stewart G Martin
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Emad Rakha
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK
| | - Sarah J Storr
- Nottingham Breast Cancer Research Centre, Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham, UK.
| |
Collapse
|
25
|
Cho W, Mittal SK, Elbasiony E, Chauhan SK. Ocular surface mast cells promote inflammatory lymphangiogenesis. Microvasc Res 2022; 141:104320. [PMID: 35031275 PMCID: PMC8923954 DOI: 10.1016/j.mvr.2022.104320] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/16/2021] [Accepted: 01/06/2022] [Indexed: 12/01/2022]
Abstract
Mast cells, sentinel immune cells, are most abundantly expressed in vascularized tissues that interface the external environment, such as the skin and ocular surface. Our previous reports have shown mast cells reside closely with vascular endothelial cells and mediate the pathogenic angiogenic response. However, the contribution of mast cells and their underlying mechanisms on lymphangiogenesis have not been fully deciphered. Using a murine model of inflammatory corneal angiogenesis, we observed adjacent migration of activated mast cells with new lymph vessel growth. Our in vitro co-culture assays demonstrate that mast cells express high levels of of VEGF-D and directly promote lymphatic endothelial cell tube formation and proliferation. Moreover, our loss-of-function approaches, using mast cell knockout mice and cromolyn-mediated mast cell inhibition, showed mast cell deficiency suppresses the induction of inflammatory lymphangiogenesis and VEGF-D expression at the ocular surface following corneal tissue insult. Our findings suggest blockade of mast cells as a potential therapeutic strategy to inhibit pathological lymphangiogenesis.
Collapse
Affiliation(s)
- WonKyung Cho
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Sharad K Mittal
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Elsayed Elbasiony
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA
| | - Sunil K Chauhan
- Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Ding J, Chen S, Serrano Sosa M, Cattell R, Lei L, Sun J, Prasanna P, Liu C, Huang C. Optimizing the Peritumoral Region Size in Radiomics Analysis for Sentinel Lymph Node Status Prediction in Breast Cancer. Acad Radiol 2022; 29 Suppl 1:S223-S228. [PMID: 33160860 PMCID: PMC9583077 DOI: 10.1016/j.acra.2020.10.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/05/2020] [Accepted: 10/10/2020] [Indexed: 01/03/2023]
Abstract
RATIONALE AND OBJECTIVES Peritumoral features have been suggested to be useful in improving the prediction performance of radiomic models. The aim of this study is to systematically investigate the prediction performance improvement for sentinel lymph node (SLN) status in breast cancer from peritumoral features in radiomic analysis by exploring the effect of peritumoral region sizes. MATERIALS AND METHODS This retrospective study was performed using dynamic contrast-enhanced MRI scans of 162 breast cancer patients. The effect of peritumoral features was evaluated in a radiomics pipeline for predicting SLN metastasis in breast cancer. Peritumoral regions were generated by dilating the tumor regions-of-interest (ROIs) manually annotated by two expert radiologists, with thicknesses of 2 mm, 4 mm, 6 mm, and 8 mm. The prediction models were established in the training set (∼67% of cases) using the radiomics pipeline with and without peritumoral features derived from different peritumoral thicknesses. The prediction performance was tested in an independent validation set (the remaining ∼33%). RESULTS For this specific application, the accuracy in the validation set when using the two radiologists' ROIs could be both improved from 0.704 to 0.796 by incorporating peritumoral features. The choice of the peritumoral size could affect the level of improvement. CONCLUSION This study systematically investigates the effect of peritumoral region sizes in radiomic analysis for prediction performance improvement. The choice of the peritumoral size is dependent on the ROI drawing and would affect the final prediction performance of radiomic models, suggesting that peritumoral features should be optimized in future radiomics studies.
Collapse
Affiliation(s)
- Jie Ding
- Department of Biomedical Engineering, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA; Department of Radiation Oncology, Medical College of Wisconsin, 8701 W Watertown Plank Rd, Wauwatosa, WI 53226, USA
| | - Shenglan Chen
- Department of Biomedical Engineering, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA
| | - Mario Serrano Sosa
- Department of Biomedical Engineering, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA
| | - Renee Cattell
- Department of Biomedical Engineering, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA
| | - Lan Lei
- Pogram in Program in Public Health, Renaissance School of Medicine, Stony Brook University, 101 Nicolls Rd, Stony Brook, NY 11794, USA
| | - Junqi Sun
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, 101 Nicolls Rd, Stony Brook, NY 11794, USA; Department of Radiology, Yuebei People's Hospital, 133 Huimin S Rd, Shaoguan, Guangdong 512025, China
| | - Prateek Prasanna
- Department of Biomedical Informatics, Renaissance School of Medicine, Stony Brook University, 101 Nicolls Rd, Stony Brook, NY 11794, USA
| | - Chunling Liu
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, 101 Nicolls Rd, Stony Brook, NY 11794, USA; Department of Radiology, Guangdong General Hospital/Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Rd, Guangzhou, Guangdong 510080, China
| | - Chuan Huang
- Department of Biomedical Engineering, Stony Brook University, 100 Nicolls Rd, Stony Brook, NY 11794, USA; Department of Radiology, Renaissance School of Medicine, Stony Brook University, 101 Nicolls Rd, Stony Brook, NY 11794, USA; Department of Psychiatry, Renaissance School of Medicine, Stony Brook University, 101 Nicolls Rd, Stony Brook, NY 11794, USA.
| |
Collapse
|
27
|
The Role of Lymphatic Marker Prox-1 in Relation to Brain Tumours. FOLIA VETERINARIA 2021. [DOI: 10.2478/fv-2021-0040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
The homeobox gene, Prox-1 is a transcription factor essential for lymphatic development (lymphangiogenesis) during embryogenesis. It also performs different functions in various tissues such as: retina, lens, liver, pancreas and the central nervous system. Intense expression of Prox-1 has been demonstrated in the developing spinal cord and brain. In adulthood its expression continues in the hippocampus and cerebellum. In adult tissues the process of lymphatic vasculature formation is accompanied under certain pathological conditions such as inflammation, tissue repair and tumour growth. Prox-1 expression is typical for lymphatic vessels; thus it belongs to one of the most specific and widely used mammalian lymphatic endothelial marker in the detection of lymphangiogenesis and lymphatic vessel invasion in oncogenesis. It has been shown that Prox-1 is involved in cancer development and progression. It’s tumour suppressive and oncogenic properties are proven in several human cancers, including brain tumours. Among all body cancers the brain tumours represent the most feared tumours with very limited treatment options and a poor diagnosis. The aim of this paper was to show the current knowledge of the gene Prox-1 with an emphasis on brain tumours, especially in gliomas.
Collapse
|
28
|
Czajka-Francuz P, Cisoń-Jurek S, Czajka A, Kozaczka M, Wojnar J, Chudek J, Francuz T. Systemic Interleukins' Profile in Early and Advanced Colorectal Cancer. Int J Mol Sci 2021; 23:124. [PMID: 35008550 PMCID: PMC8745135 DOI: 10.3390/ijms23010124] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/18/2021] [Accepted: 12/20/2021] [Indexed: 02/05/2023] Open
Abstract
Tumor microenvironment (TME) is characterized by mutual interactions of the tumor, stromal and immune cells. Early and advanced colorectal tumors differ in structure and present altered serum cytokine levels. Mutual crosstalk among TME infiltrating cells may shift the balance into immune suppressive or pro-inflammatory, antitumor response this way influencing patients' prognosis. Cancer-related inflammation affects all the body and this way, the systemic level of cytokines could reflect TME processes. Despite numerous studies, it is still not known how systemic cytokines levels change during colorectal cancer (CRC) tumor development. Better understanding tumor microenvironment processes could help in planning therapeutic interventions and more accurate patient prognosis. To contribute to the comprehension of these processes within TME, we reviewed cytokines levels from clinical trials in early and advanced colorectal cancer. Presented data were analyzed in the context of experimental studies and studies analyzing tumor infiltration with immune cells. The review summarizes clinical data of cytokines secreted by tumor microenvironment cells: lymphocytes T helper 1 (Th1), lymphocytes T helper 2 (Th2), lymphocytes T helper 17 (Th17), regulatory T cells (Treg cells), regulatory T cells (Breg cells), M1/M2 macrophages, N1/N2 neutrophils, myeloid-derived suppressor cells (MDSC), dendritic cells (DC), innate lymphoid cells (ILC) natural killer (NK) cells and tumor cells.
Collapse
Affiliation(s)
- Paulina Czajka-Francuz
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Sylwia Cisoń-Jurek
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Aleksander Czajka
- Department of General Surgery, Vascular Surgery, Angiology and Phlebology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-635 Katowice, Poland;
| | - Maciej Kozaczka
- Department of Radiotherapy and Chemotherapy, National Institute of Oncology, Public Research Institute in Gliwice, 44-101 Gliwice, Poland;
| | - Jerzy Wojnar
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Jerzy Chudek
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
| | - Tomasz Francuz
- Department of Internal Medicine and Oncological Chemotherapy, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-027 Katowice, Poland; (S.C.-J.); (J.W.); (J.C.); (T.F.)
- Department of Biochemistry, Faculty of Medical Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
| |
Collapse
|
29
|
Verteporfin-Loaded Mesoporous Silica Nanoparticles' Topical Applications Inhibit Mouse Melanoma Lymphangiogenesis and Micrometastasis In Vivo. Int J Mol Sci 2021; 22:ijms222413443. [PMID: 34948239 PMCID: PMC8705243 DOI: 10.3390/ijms222413443] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 01/04/2023] Open
Abstract
Photodynamic therapy (PDT) has been pointed out as a candidate for improving melanoma treatment. Nanotechnology application in PDT has increased its efficacy by reducing side effects. Herein, mesoporous silica nanoparticles (MSNs) conjugated with verteporfin (Ver-MSNs), in use with PDT, were administered in mice to evaluate their efficacy on lymphoangiogenesis and micrometastasis in melanoma. Melanoma was induced in mice by the subcutaneous injection of B16-F10 cells. The mice were transcutaneously treated with MSNs, Ver-MSNs, or glycerol and exposed to red light. The treatment was carried out four times until day 20. Lymphangiogenesis and micrometastasis were identified by the immunohistochemical method. Lymphoangiogenesis was halved by MSN treatment compared with the control animals, whereas the Ver-MSN treatment almost abolished it. A similar reduction was also observed in lung micrometastasis. PDT with topically administrated Ver-MSNs reduced melanoma lymphoangiogenesis and lung micrometastasis, as well as tumor mass and angiogenesis, and therefore their use could be an innovative and useful tool in melanoma clinical therapy.
Collapse
|
30
|
Cho Y, Na K, Jun Y, Won J, Yang JH, Chung S. Three-Dimensional In Vitro Lymphangiogenesis Model in Tumor Microenvironment. Front Bioeng Biotechnol 2021; 9:697657. [PMID: 34671596 PMCID: PMC8520924 DOI: 10.3389/fbioe.2021.697657] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/30/2021] [Indexed: 11/25/2022] Open
Abstract
Lymphangiogenesis is a stage of new lymphatic vessel formation in development and pathology, such as inflammation and tumor metastasis. Physiologically relevant models of lymphatic vessels have been in demand because studies on lymphatic vessels are required for understanding the mechanism of tumor metastasis. In this study, a new three-dimensional lymphangiogenesis model in a tumor microenvironment is proposed, using a newly designed macrofluidic platform. It is verified that controllable biochemical and biomechanical cues, which contribute to lymphangiogenesis, can be applied in this platform. In particular, this model demonstrates that a reconstituted lymphatic vessel has an in vivo–like lymphatic vessel in both physical and biochemical aspects. Since biomechanical stress with a biochemical factor influences robust directional lymphatic sprouting, whether our model closely approximates in vivo, the initial lymphatics in terms of the morphological and genetic signatures is investigated. Furthermore, attempting an incorporation with a tumor spheroid, this study successfully develops a complex tumor microenvironment model for use in lymphangiogenesis and reveals the microenvironment factors that contribute to tumor metastasis. As a first attempt at a coculture model, this reconstituted model is a novel system with a fully three-dimensional structure and can be a powerful tool for pathological drug screening or disease model.
Collapse
Affiliation(s)
- Youngkyu Cho
- Department of IT Convergence, Korea University, Seoul, South Korea.,Samsung Research, Samsung Electronics Co. Ltd., Seoul, South Korea
| | - Kyuhwan Na
- School of Mechanical Engineering, Korea University, Seoul, South Korea
| | - Yesl Jun
- Departments of Pediatrics and Cellular & Molecular Medicine, Pediatric Diabetes Research Center, University of California, La Jolla, CA, United States.,Drug Discovery Platform Research Center, Therapeutics and Biotechnology Division, Korea Research Institute of Chemical Technology, Daejeon, South Korea
| | - Jihee Won
- School of Mechanical Engineering, Korea University, Seoul, South Korea
| | - Ji Hun Yang
- School of Mechanical Engineering, Korea University, Seoul, South Korea.,Next&Bio Inc., Seoul, South Korea
| | - Seok Chung
- Department of IT Convergence, Korea University, Seoul, South Korea.,School of Mechanical Engineering, Korea University, Seoul, South Korea
| |
Collapse
|
31
|
Yusof KM, Groen K, Rosli R, Avery-Kiejda KA. Crosstalk Between microRNAs and the Pathological Features of Secondary Lymphedema. Front Cell Dev Biol 2021; 9:732415. [PMID: 34733847 PMCID: PMC8558478 DOI: 10.3389/fcell.2021.732415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/01/2021] [Indexed: 01/07/2023] Open
Abstract
Secondary lymphedema is characterized by lymphatic fluid retention and subsequent tissue swelling in one or both limbs that can lead to decreased quality of life. It often arises after loss, obstruction, or blockage of lymphatic vessels due to multifactorial modalities, such as lymphatic insults after surgery, immune system dysfunction, deposition of fat that compresses the lymphatic capillaries, fibrosis, and inflammation. Although secondary lymphedema is often associated with breast cancer, the condition can occur in patients with any type of cancer that requires lymphadenectomy such as gynecological, genitourinary, or head and neck cancers. MicroRNAs demonstrate pivotal roles in regulating gene expression in biological processes such as lymphangiogenesis, angiogenesis, modulation of the immune system, and oxidative stress. MicroRNA profiling has led to the discovery of the molecular mechanisms involved in the pathophysiology of auto-immune, inflammation-related, and metabolic diseases. Although the role of microRNAs in regulating secondary lymphedema is yet to be elucidated, the crosstalk between microRNAs and molecular factors involved in the pathological features of lymphedema, such as skin fibrosis, inflammation, immune dysregulation, and aberrant lipid metabolism have been demonstrated in several studies. MicroRNAs have the potential to serve as biomarkers for diseases and elucidation of their roles in lymphedema can provide a better understanding or new insights of the mechanisms underlying this debilitating condition.
Collapse
Affiliation(s)
- Khairunnisa’ Md Yusof
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Kira Groen
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
| | - Rozita Rosli
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
- UPM-MAKNA Cancer Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Malaysia
| | - Kelly A. Avery-Kiejda
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
32
|
CD147 Promotes Tumor Lymphangiogenesis in Melanoma via PROX-1. Cancers (Basel) 2021; 13:cancers13194859. [PMID: 34638342 PMCID: PMC8508014 DOI: 10.3390/cancers13194859] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 01/19/2023] Open
Abstract
Simple Summary Melanoma is one of the most aggressive skin cancers, characterized by metastasis to the lymph nodes and a high capacity to develop drug resistance. There is a lack of knowledge on the mechanisms contributing to lymphatic vessel formation and metastasis regulation in malignant melanoma. We previously reported the involvement of CD147, a transmembrane glycoprotein overexpressed in melanoma, in the regulation of the tumor microenvironment and angiogenesis. The aim of our study was to further determine how CD147 is involved in lymphangiogenesis regulation. Our results revealed that high CD147 expression is correlated with the number of lymphatic vessels in the human melanoma lymph nodes and that paracrine CD147 upregulates lymphangiogenesis through lymphangiogenic mediators in vitro and in vivo, suggesting that CD147 could be a promising target for melanoma-associated lymphangiogenesis inhibition. Abstract Malignant melanoma is one of the most aggressive skin cancers and is characterized by early lymph node metastasis and the capacity to develop resistance to therapies. Hence, understanding the regulation of lymphangiogenesis through mechanisms contributing to lymphatic vessel formation represents a treatment strategy for metastatic cancer. We have previously shown that CD147, a transmembrane glycoprotein overexpressed in melanoma, regulates the angiogenic process in endothelial cells. In this study, we show a correlation between high CD147 expression levels and the number of lymphatic vessels expressing LYVE-1, Podoplanin, and VEGFR-3 in human melanoma lymph nodes. CD147 upregulates in vitro lymphangiogenesis and its related mediators through the PROX-1 transcription factor. In vivo studies in a melanoma model confirmed that CD147 is involved in metastasis through a similar mechanism as in vitro. This study, demonstrating the paracrine role of CD147 in the lymphangiogenesis process, suggests that CD147 could be a promising target for the inhibition of melanoma-associated lymphangiogenesis.
Collapse
|
33
|
Kang MJ, Lee S, Jung U, Mandal C, Park H, Stetler-Stevenson WG, Kim YS, Moon JW, Park SH, Oh J. Inhibition of Hepatic Stellate Cell Activation Suppresses Tumorigenicity of Hepatocellular Carcinoma in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:2219-2230. [PMID: 34428424 PMCID: PMC8747013 DOI: 10.1016/j.ajpath.2021.08.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/27/2021] [Accepted: 08/06/2021] [Indexed: 12/18/2022]
Abstract
Transdifferentiation (or activation) of hepatic stellate cells (HSCs) to myofibroblasts is a key event in liver fibrosis. Activated HSCs in the tumor microenvironment reportedly promote tumor progression. This study analyzed the effect of an inhibitor of HSC activation, retinol-binding protein–albumin domain III fusion protein (R-III), on protumorigenic functions of HSCs. Although conditioned medium collected from activated HSCs enhanced the migration, invasion, and proliferation of the hepatocellular carcinoma cell line Hepa-1c1c7, this effect was not observed in Hepa-1c1c7 cells treated with conditioned medium from R-III–exposed HSCs. In a subcutaneous tumor model, larger tumors with increased vascular density were formed in mice transplanted with Hepa-1c1c7+HSC than in mice transplanted with Hepa-1c1c7 cells alone. Intriguingly, when Hepa-1c1c7+HSC–transplanted mice were injected intravenously with R-III, a reduction in vascular density and extended tumor necrosis were observed. In an orthotopic tumor model, co-transplantation of HSCs enhanced tumor growth, angiogenesis, and regional metastasis accompanied by increased peritumoral lymphatic vessel density, which was abolished by R-III. In vitro study showed that R-III treatment affected the synthesis of pro-angiogenic and anti-angiogenic factors in activated HSCs, which might be the potential mechanism underlying the R-III effect. These findings suggest that the inhibition of HSC activation abrogates HSC-induced tumor angiogenesis and growth, which represents an attractive therapeutic strategy.
Collapse
Affiliation(s)
- Min-Jung Kang
- Department of Biomedical Science, Korea University Graduate School, Seoul, Korea
| | - Soovin Lee
- Laboratory Animal Research Center, College of Medicine, Korea University, Seoul, Korea
| | - Usuk Jung
- Department of Biomedical Science, Korea University Graduate School, Seoul, Korea
| | - Chanchal Mandal
- Department of Biomedical Science, Korea University Graduate School, Seoul, Korea
| | - Heekyung Park
- Department of Biomedical Science, Korea University Graduate School, Seoul, Korea
| | - William G Stetler-Stevenson
- Extracellular Matrix Pathology Section, Laboratory of Pathology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Young-Sik Kim
- Department of Pathology, College of Medicine, Korea University, Seoul, Korea
| | - Ji Wook Moon
- Department of Anatomy, College of Medicine, Korea University, Seoul, Korea
| | - Sun-Hwa Park
- Department of Anatomy, College of Medicine, Korea University, Seoul, Korea
| | - Junseo Oh
- Department of Biomedical Science, Korea University Graduate School, Seoul, Korea; Department of Anatomy, College of Medicine, Korea University, Seoul, Korea.
| |
Collapse
|
34
|
Jakovija A, Chtanova T. Neutrophil Interactions with the Lymphatic System. Cells 2021; 10:cells10082106. [PMID: 34440875 PMCID: PMC8393351 DOI: 10.3390/cells10082106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 01/02/2023] Open
Abstract
The lymphatic system is a complex network of lymphatic vessels and lymph nodes designed to balance fluid homeostasis and facilitate host immune defence. Neutrophils are rapidly recruited to sites of inflammation to provide the first line of protection against microbial infections. The traditional view of neutrophils as short-lived cells, whose role is restricted to providing sterilizing immunity at sites of infection, is rapidly evolving to include additional functions at the interface between the innate and adaptive immune systems. Neutrophils travel via the lymphatics from the site of inflammation to transport antigens to lymph nodes. They can also enter lymph nodes from the blood by crossing high endothelial venules. Neutrophil functions in draining lymph nodes include pathogen control and modulation of adaptive immunity. Another facet of neutrophil interactions with the lymphatic system is their ability to promote lymphangiogenesis in draining lymph nodes and inflamed tissues. In this review, we discuss the significance of neutrophil migration to secondary lymphoid organs and within the lymphatic vasculature and highlight emerging evidence of the neutrophils’ role in lymphangiogenesis.
Collapse
Affiliation(s)
- Arnolda Jakovija
- Innate and Tumor Immunology Laboratory, Immunity Theme, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia;
- St Vincent’s School of Medicine, Faculty of Medicine, UNSW Sydney, Sydney, NSW 2052, Australia
| | - Tatyana Chtanova
- Innate and Tumor Immunology Laboratory, Immunity Theme, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia;
- School of Biotechnology and Biomolecular Sciences, Faculty of Science, UNSW Sydney, Sydney, NSW 2052, Australia
- Correspondence:
| |
Collapse
|
35
|
Palmini G, Brandi ML. microRNAs and bone tumours: Role of tiny molecules in the development and progression of chondrosarcoma, of giant cell tumour of bone and of Ewing's sarcoma. Bone 2021; 149:115968. [PMID: 33892177 DOI: 10.1016/j.bone.2021.115968] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 03/26/2021] [Accepted: 04/15/2021] [Indexed: 12/15/2022]
Abstract
The increasing interest on microRNAs (miRNAs), small non-coding RNA molecules containing about 22 nucleotides, about their biological functions led researchers to discover that they are actively involved in several biological processes. In the last decades, miRNAs become one of the most topic of cancer research. miRNAs, thanks to their function, are the perfect molecules to modulate multiple signaling pathways and gene expression in cancer, with the consequent capacity to modulate cancerous processes, such as cellular proliferation, invasion, metastasis and chemoresistance in various tumours. In the last years, several studies have demonstrated the role of miRNAs in their pathophysiology, but little we know about the underlying mechanism that lead to bone tumours like chondrosarcoma (COS), giant cell tumour of bone (GCTB) and Ewing sarcoma (EWS) to still be highly aggressive and resistant tumours. An exploration of the role of miRNAs in the biology of them will permit to researchers to find new molecular mechanisms that can be used to develop new and more effective therapies against these bone tumours. Here we present a comprehensive study of the latest discoveries which have been performed in relation to the role of miRNAs in the neoplastic processes which characterize COS, EWS and GCTB, demonstrating how these tiny molecules can act as tumour promoters or as tumour suppressors and how they can be used for improving therapeutic approaches.
Collapse
Affiliation(s)
- Gaia Palmini
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy.
| | - Maria Luisa Brandi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy; Fondazione Italiana Ricerca sulle Malattie dell'Osso, F.I.R.M.O Onlus, Florence, Italy.
| |
Collapse
|
36
|
McShane R, Arya S, Stewart AJ, Caie P, Bates M. Prognostic features of the tumour microenvironment in oesophageal adenocarcinoma. Biochim Biophys Acta Rev Cancer 2021; 1876:188598. [PMID: 34332022 DOI: 10.1016/j.bbcan.2021.188598] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 12/12/2022]
Abstract
Oesophageal adenocarcinoma (OAC) is a disease with an incredibly poor survival rate and a complex makeup. The growth and spread of OAC tumours are profoundly influenced by their surrounding microenvironment and the properties of the tumour itself. Constant crosstalk between the tumour and its microenvironment is key to the survival of the tumour and ultimately the death of the patient. The tumour microenvironment (TME) is composed of a complex milieu of cell types including cancer associated fibroblasts (CAFs) which make up the tumour stroma, endothelial cells which line blood and lymphatic vessels and infiltrating immune cell populations. These various cell types and the tumour constantly communicate through environmental cues including fluctuations in pH, hypoxia and the release of mitogens such as cytokines, chemokines and growth factors, many of which help promote malignant progression. Eventually clusters of tumour cells such as tumour buds break away and spread through the lymphatic system to nearby lymph nodes or enter the circulation forming secondary metastasis. Collectively, these factors need to be considered when assessing and treating patients clinically. This review aims to summarise the ways in which these various factors are currently assessed and how they relate to patient treatment and outcome at an individual level.
Collapse
Affiliation(s)
| | - Swati Arya
- School of Medicine, University of St Andrews, Fife, UK
| | | | - Peter Caie
- School of Medicine, University of St Andrews, Fife, UK
| | - Mark Bates
- Department of Surgery, Trinity Translational Medicine Institute, St. James's Hospital, Dublin 8, Ireland; Trinity St James's Cancer Institute, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
37
|
Naruse T, Goi T, Yamaguchi A. Prokineticin-1 induces normal lymphangiogenic activity and is involved in lymphangiogenesis and lymph node metastasis in colorectal cancer. Oncotarget 2021; 12:1388-1397. [PMID: 34262649 PMCID: PMC8274725 DOI: 10.18632/oncotarget.28016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
Background: Prokineticin family correlates with important roles in several biological processes, including homeostasis. We discovered novel functions of prokineticin1 (PROK1) in lymphangiogenesis and lymphnode metastasis in colorectal cancer. Materials and Methods: We examined changes in the number of lymphatic endothelial cells after PROK1 stimulation. PROK1 protein was stimulated with subcutaneously implanted in mice. Also a high-PROK1-expressing colorectal cancer cell line and anti-PROK1 antibody(Ab) were subcutaneously implanted in mice, and then examine lymphangiogenesis. PROK1 expression and the number of lymph vessels were examined in the primary lesion of 391 patients whose colorectal tumors had been resected. Results: When PROK1 was used as a stimulus, the number of lymphatic cells increased compared to unstimulated cells. And the number of lymph vessels in the skin of mice increased compared to mice implanted without PROK1. The number of lymph vessels in the primary tumor tissue increased when PROK1 was highly expressed compared to cases with non-detectable PROK1 expression. When PROK1 was expressed in human colorectal tumors, the rate of lymphnode metastasis was significantly higher than that in cases with non-detectable PROK1 expression. Conclusions: PROK1 is a lymphangiogenic factor involved in the formation of new lymph vessels and lymphnode metastasis in human colorectal cancer.
Collapse
Affiliation(s)
- Takayuki Naruse
- First Department of Surgery, University of Fukui, Fukui 9101193, Japan
| | - Takanori Goi
- First Department of Surgery, University of Fukui, Fukui 9101193, Japan
| | | |
Collapse
|
38
|
Li M, Xian HC, Tang YJ, Liang XH, Tang YL. Fatty acid oxidation: driver of lymph node metastasis. Cancer Cell Int 2021; 21:339. [PMID: 34217300 PMCID: PMC8254237 DOI: 10.1186/s12935-021-02057-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/27/2021] [Indexed: 02/08/2023] Open
Abstract
Fatty acid oxidation (FAO) is the emerging hallmark of cancer metabolism because certain tumor cells preferentially utilize fatty acids for energy. Lymph node metastasis, the most common way of tumor metastasis, is much indispensable for grasping tumor progression, formulating therapy measure and evaluating tumor prognosis. There is a plethora of studies showing different ways how tumor cells metastasize to the lymph nodes, but the role of FAO in lymph node metastasis remains largely unknown. Here, we summarize recent findings and update the current understanding that FAO may enable lymph node metastasis formation. Afterward, it will open innovative possibilities to present a distinct therapy of targeting FAO, the metabolic rewiring of cancer to terminal cancer patients.
Collapse
Affiliation(s)
- Mao Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hong-Chun Xian
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, 266237, China.
| | - Xin-Hua Liang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Ya-Ling Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, Department of Oral Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
39
|
Wang S, Sun Y, Li R, Mao N, Li Q, Jiang T, Chen Q, Duan S, Xie H, Gu Y. Diagnostic performance of perilesional radiomics analysis of contrast-enhanced mammography for the differentiation of benign and malignant breast lesions. Eur Radiol 2021; 32:639-649. [PMID: 34189600 DOI: 10.1007/s00330-021-08134-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 05/16/2021] [Accepted: 06/01/2021] [Indexed: 12/15/2022]
Abstract
OBJECTIVE To conduct perilesional region radiomics analysis of contrast-enhanced mammography (CEM) images to differentiate benign and malignant breast lesions. METHODS AND MATERIALS This retrospective study included patients who underwent CEM from November 2017 to February 2020. Lesion contours were manually delineated. Perilesional regions were automatically obtained. Seven regions of interest (ROIs) were obtained for each lesion, including the lesion ROI, annular perilesional ROIs (1 mm, 3 mm, 5 mm), and lesion + perilesional ROIs (1 mm, 3 mm, 5 mm). Overall, 4,098 radiomics features were extracted from each ROI. Datasets were divided into training and testing sets (1:1). Seven classification models using features from the seven ROIs were constructed using LASSO regression. Model performance was assessed by the AUC with 95% CI. RESULTS Overall, 190 women with 223 breast lesions (101 benign; 122 malignant) were enrolled. In the testing set, the annular perilesional ROI of 3-mm model showed the highest AUC of 0.930 (95% CI: 0.882-0.977), followed by the annular perilesional ROI of 1 mm model (AUC = 0.929; 95% CI: 0.881-0.978) and the lesion ROI model (AUC = 0.909; 95% CI: 0.857-0.961). A new model was generated by combining the predicted probabilities of the lesion ROI and annular perilesional ROI of 3-mm models, which achieved a higher AUC in the testing set (AUC = 0.940). CONCLUSIONS Annular perilesional radiomics analysis of CEM images is useful for diagnosing breast cancers. Adding annular perilesional information to the radiomics model built on the lesion information may improve the diagnostic performance. KEY POINTS • Radiomics analysis of the annular perilesional region of 3 mm in CEM images may provide valuable information for the differential diagnosis of benign and malignant breast lesions. • The radiomics information from the lesion region and the annular perilesional region may be complementary. Combining the predicted probabilities of the models constructed by the features from the two regions may improve the diagnostic performance of radiomics models.
Collapse
Affiliation(s)
- Simin Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yuqi Sun
- Department of Biostatistics, School of Public Health, Fudan University, Shanghai, 200032, China
| | - Ruimin Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ning Mao
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Shandong, 264000, China
| | - Qin Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Tingting Jiang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Qianqian Chen
- GE Healthcare China, No. 1 Huatuo Road, Shanghai, 210000, China
| | - Shaofeng Duan
- GE Healthcare China, No. 1 Huatuo Road, Shanghai, 210000, China
| | - Haizhu Xie
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Shandong, 264000, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
40
|
Hsu M, Laaker C, Sandor M, Fabry Z. Neuroinflammation-Driven Lymphangiogenesis in CNS Diseases. Front Cell Neurosci 2021; 15:683676. [PMID: 34248503 PMCID: PMC8261156 DOI: 10.3389/fncel.2021.683676] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
The central nervous system (CNS) undergoes immunosurveillance despite the lack of conventional antigen presenting cells and lymphatic vessels in the CNS parenchyma. Additionally, the CNS is bathed in a cerebrospinal fluid (CSF). CSF is continuously produced, and consequently must continuously clear to maintain fluid homeostasis despite the lack of conventional lymphatics. During neuroinflammation, there is often an accumulation of fluid, antigens, and immune cells to affected areas of the brain parenchyma. Failure to effectively drain these factors may result in edema, prolonged immune response, and adverse clinical outcome as observed in conditions including traumatic brain injury, ischemic and hypoxic brain injury, CNS infection, multiple sclerosis (MS), and brain cancer. Consequently, there has been renewed interest surrounding the expansion of lymphatic vessels adjacent to the CNS which are now thought to be central in regulating the drainage of fluid, cells, and waste out of the CNS. These lymphatic vessels, found at the cribriform plate, dorsal dural meninges, base of the brain, and around the spinal cord have each been implicated to have important roles in various CNS diseases. In this review, we discuss the contribution of meningeal lymphatics to these processes during both steady-state conditions and neuroinflammation, as well as discuss some of the many still unknown aspects regarding the role of meningeal lymphatics in neuroinflammation. Specifically, we focus on the observed phenomenon of lymphangiogenesis by a subset of meningeal lymphatics near the cribriform plate during neuroinflammation, and discuss their potential roles in immunosurveillance, fluid clearance, and access to the CSF and CNS compartments. We propose that manipulating CNS lymphatics may be a new therapeutic way to treat CNS infections, stroke, and autoimmunity.
Collapse
Affiliation(s)
- Martin Hsu
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI, United States
| | - Collin Laaker
- Neuroscience Training Program, University of Wisconsin Madison, Madison, WI, United States
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, University of Wisconsin Madison, Madison, WI, United States
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, University of Wisconsin Madison, Madison, WI, United States
| |
Collapse
|
41
|
ECM Remodeling in Squamous Cell Carcinoma of the Aerodigestive Tract: Pathways for Cancer Dissemination and Emerging Biomarkers. Cancers (Basel) 2021. [DOI: 10.3390/cancers13112759
expr 955442319 + 839973387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
Squamous cell carcinomas (SCC) include a number of different types of tumors developing in the skin, in hollow organs, as well as the upper aerodigestive tract (UADT) including the head and neck region and the esophagus which will be dealt with in this review. These tumors are often refractory to current therapeutic approaches with poor patient outcome. The most important prognostic determinant of SCC tumors is the presence of distant metastasis, significantly correlating with low patient survival rates. Rapidly emerging evidence indicate that the extracellular matrix (ECM) composition and remodeling profoundly affect SSC metastatic dissemination. In this review, we will summarize the current knowledge on the role of ECM and its remodeling enzymes in affecting the growth and dissemination of UADT SCC. Taken together, these published evidence suggest that a thorough analysis of the ECM composition in the UADT SCC microenvironment may help disclosing the mechanism of resistance to the treatments and help defining possible targets for clinical intervention.
Collapse
|
42
|
ECM Remodeling in Squamous Cell Carcinoma of the Aerodigestive Tract: Pathways for Cancer Dissemination and Emerging Biomarkers. Cancers (Basel) 2021; 13:cancers13112759. [PMID: 34199373 PMCID: PMC8199582 DOI: 10.3390/cancers13112759] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/27/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Local and distant metastasis of patients affected by squamous cell carcinoma of the upper aerodigestive tract predicts poor prognosis. In the latest years, the introduction of new therapeutic approaches, including targeted and immune therapies, has improved the overall survival. However, a large number of these patients do not benefit from these treatments. Thus, the identification of suitable prognostic and predictive biomarkers, as well as the discovery of new therapeutic targets have emerged as a crucial clinical need. In this context, the extracellular matrix represents a suitable target for the development of such therapeutic tools. In fact, the extracellular matrix is composed by complex molecules able to interact with a plethora of receptors and growth factors, thus modulating the dynamic crosstalk between cancer cells and the tumor microenvironment. In this review, we summarize the current knowledge of the role of the extracellular matrix in affecting squamous cell carcinoma growth and dissemination. Despite extracellular matrix is known to affect the development of many cancer types, only a restricted number of these molecules have been recognized to impact on squamous cell carcinoma progression. Thus, we consider that a thorough analysis of these molecules may be key to develop new potential therapeutic targets/biomarkers. Abstract Squamous cell carcinomas (SCC) include a number of different types of tumors developing in the skin, in hollow organs, as well as the upper aerodigestive tract (UADT) including the head and neck region and the esophagus which will be dealt with in this review. These tumors are often refractory to current therapeutic approaches with poor patient outcome. The most important prognostic determinant of SCC tumors is the presence of distant metastasis, significantly correlating with low patient survival rates. Rapidly emerging evidence indicate that the extracellular matrix (ECM) composition and remodeling profoundly affect SSC metastatic dissemination. In this review, we will summarize the current knowledge on the role of ECM and its remodeling enzymes in affecting the growth and dissemination of UADT SCC. Taken together, these published evidence suggest that a thorough analysis of the ECM composition in the UADT SCC microenvironment may help disclosing the mechanism of resistance to the treatments and help defining possible targets for clinical intervention.
Collapse
|
43
|
Cheung SM, Husain E, Mallikourti V, Masannat Y, Heys S, He J. Intra-tumoural lipid composition and lymphovascular invasion in breast cancer via non-invasive magnetic resonance spectroscopy. Eur Radiol 2021; 31:3703-3711. [PMID: 33270144 PMCID: PMC8128855 DOI: 10.1007/s00330-020-07502-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 10/07/2020] [Accepted: 11/11/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Despite improved survival due to new treatments, the 10-year survival rate in patients with breast cancer is approximately 75%. Lymphovascular invasion (LVI), a prognostic marker independent from histological grade and stage, can only be fully determined at final histological examination. Lipid composition is deregulated in tumour via de novo lipogenesis, with alteration in lipogenic genes in LVI. We hypothesise alteration in lipid composition derived from novel non-invasive spectroscopy method is associated with LVI positivity. METHODS Thirty female patients (age 39-78) with invasive ductal carcinoma were enrolled, with 13 LVI negative and 17 LVI positive. Saturated, monounsaturated, polyunsaturated fatty acids and triglycerides (SFA, MUFA, PUFA and TRG) were quantified from ex vivo breast tumours freshly excised from patients on a 3 T clinical MRI scanner, and proliferative activity marker Ki-67 and serotonin derived histologically. RESULTS There were significantly lower MUFA (p = 0.0189) in LVI positive (median: 0.37, interquartile range (IQR): 0.25-0.64) than negative (0.63, 0.49-0.96). There were significantly lower TRG (p = 0.0226) in LVI positive (1.32, 0.95-2.43) than negative (2.5, 1.92-4.15). There was no significant difference in SFA (p = 0.6009) or PUFA (p = 0.1641). There was no significant correlation between lipid composition against Ki-67 or serotonin, apart from a borderline negative correlation between PUFA and serotonin (r = - 0.3616, p = 0.0496). CONCLUSION Lipid composition might provide a biomarker to study lymphovascular invasion in breast cancer. KEY POINTS • Monounsaturated fatty acids in lymphovascular invasion (LVI) positive invasive breast carcinoma were significantly lower than that in LVI negative. • Triglycerides in LVI positive invasive breast carcinoma were significantly lower than that in LVI negative. • Lipid composition from MR spectroscopy reflects the rate of de novo lipogenesis and provides a potential biomarker independent from histological grade and stage.
Collapse
Affiliation(s)
- Sai Man Cheung
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK.
| | - Ehab Husain
- Pathology Department, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Vasiliki Mallikourti
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| | | | - Steven Heys
- Breast Unit, Aberdeen Royal Infirmary, Aberdeen, UK
| | - Jiabao He
- Institute of Medical Sciences, School of Medicine, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
44
|
Li C, Song L, Yin J. Intratumoral and Peritumoral Radiomics Based on Functional Parametric Maps from Breast DCE-MRI for Prediction of HER-2 and Ki-67 Status. J Magn Reson Imaging 2021; 54:703-714. [PMID: 33955619 DOI: 10.1002/jmri.27651] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/04/2021] [Accepted: 04/05/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Radiomics has been applied to breast magnetic resonance imaging (MRI) for gene status prediction. However, the features of peritumoral regions were not thoroughly investigated. PURPOSE To evaluate the use of intratumoral and peritumoral regions from functional parametric maps based on breast dynamic contrast-enhanced MRI (DCE-MRI) for prediction of HER-2 and Ki-67 status. STUDY TYPE Retrospective. POPULATION A total of 351 female patients (average age, 51 years) with pathologically confirmed breast cancer were assigned to the training (n = 243) and validation (n = 108) cohorts. FIELD STRENGTH/SEQUENCE 3.0T, T1 gradient echo. ASSESSMENT Radiomic features were extracted from intratumoral and peritumoral regions on six functional parametric maps calculated using time-intensity curves of DCE-MRI. The intraclass correlation coefficients (ICCs) were used to determine the reproducibility of feature extraction. Based on the intratumoral, peritumoral, and combined intra- and peritumoral regions, three radiomics signatures (RSs) were built using the least absolute shrinkage and selection operator (LASSO) logistic regression model, respectively. STATISTICAL TESTS Wilcoxon rank-sum test, minimum redundancy maximum relevance, LASSO, receiver operating characteristic curve (ROC) analysis, and DeLong test. RESULTS The intratumoral and peritumoral RSs for prediction of HER-2 and Ki-67 status achieved areas under the ROC (AUCs) of 0.683 (95% confidence interval [CI], 0.574-0.793) and 0.690 (95% CI, 0.577-0.804), and 0.714 (95% CI, 0.616-0.812) and 0.692 (95% CI, 0.590-0.794) in the validation cohort, respectively. The combined RSs yielded AUCs of 0.713 (95% CI, 0.604-0.823) and 0.749 (95% CI, 0.656-0.841), respectively. There were no significant differences in prediction performance among intratumoral, peritumoral, and combined RSs. Most (69.7%) of the features had good agreement (ICCs >0.8). DATA CONCLUSION Radiomic features of intratumoral and peritumoral regions on functional parametric maps based on breast DCE-MRI had the potential to identify HER-2 and Ki-67 status. LEVEL OF EVIDENCE 3 Technical Efficacy Stage: 2.
Collapse
Affiliation(s)
- Chunli Li
- Department of Biomedical Engineering, School of Fundamental Sciences, China Medical University, Shenyang, China.,Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lirong Song
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jiandong Yin
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
45
|
Trac N, Chung EJ. Overcoming physiological barriers by nanoparticles for intravenous drug delivery to the lymph nodes. Exp Biol Med (Maywood) 2021; 246:2358-2371. [PMID: 33957802 DOI: 10.1177/15353702211010762] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The lymph nodes are major sites of cancer metastasis and immune activity, and thus represent important clinical targets. Although not as well-studied compared to subcutaneous administration, intravenous drug delivery is advantageous for lymph node delivery as it is commonly practiced in the clinic and has the potential to deliver therapeutics systemically to all lymph nodes. However, rapid clearance by the mononuclear phagocyte system, tight junctions of the blood vascular endothelium, and the collagenous matrix of the interstitium can limit the efficiency of lymph node drug delivery, which has prompted research into the design of nanoparticle-based drug delivery systems. In this mini review, we describe the physiological and biological barriers to lymph node targeting, how they inform nanoparticle design, and discuss the future outlook of lymph node targeting.
Collapse
Affiliation(s)
- Noah Trac
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| | - Eun Ji Chung
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA.,Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Division of Vascular Surgery and Endovascular Therapy, Department of Surgery, Keck School of Medicine, Los Angeles, CA 90033, USA.,Division of Nephrology and Hypertension, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.,Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA.,Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
46
|
Mutually exclusive lymphangiogenesis or perineural infiltration in human skin squamous-cell carcinoma. Oncotarget 2021; 12:638-648. [PMID: 33868585 PMCID: PMC8021034 DOI: 10.18632/oncotarget.27915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 02/26/2021] [Indexed: 11/25/2022] Open
Abstract
Although tumor-associated lymphangiogenesis correlates with metastasis and poor prognosis in several cancers, it also supports T cell infiltration into the tumor and predicts favorable outcome to immunotherapy. The role of lymphatic vessels in skin squamous-cell carcinoma (sSCC), the second most common form of skin cancer, remains mostly unknown. Although anti-PD-1 therapy is beneficial for some patients with advanced sSCC, a greater understanding of disease mechanisms is still needed to develop better therapies. Using quantitative multiplex immunohistochemistry, we analyzed sSCC sections from 36 patients. CD8+ T cell infiltration showed great differences between patients, whereby these cells were mainly excluded from the tumor mass. Similar to our data in melanoma, sSCC with high density of lymphatic endothelial cells showed increased CD8+ T cell density in tumor areas. An entirely new observation is that sSCC with perineural infiltration but without metastasis was characterized by low lymphatic endothelial cell density. Since both, metastasis and perineural infiltration are known to affect tumor progression and patients’ prognosis, it is important to identify the molecular drivers, opening future options for therapeutic targeting. Our data suggest that the mechanisms underlying perineural infiltration may be linked with the biology of lymphatic vessels and thus stroma.
Collapse
|
47
|
Decoding the Roles of Long Noncoding RNAs in Hepatocellular Carcinoma. Int J Mol Sci 2021; 22:ijms22063137. [PMID: 33808647 PMCID: PMC8003515 DOI: 10.3390/ijms22063137] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/15/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most prevalent malignancies worldwide. HCC is associated with several etiological factors, including HBV/HCV infections, cirrhosis, and fatty liver diseases. However, the molecular mechanism underlying HCC development remains largely elusive. The advent of high-throughput sequencing has unveiled an unprecedented discovery of a plethora of long noncoding RNAs (lncRNAs). Despite the lack of coding capacity, lncRNAs have key roles in gene regulation through interacting with various biomolecules. It is increasingly evident that the dysregulation of lncRNAs is inextricably linked to HCC cancer phenotypes, suggesting that lncRNAs are potential prognostic markers and therapeutic targets. In light of the emerging research in the study of the regulatory roles of lncRNAs in HCC, we discuss the association of lncRNAs with HCC. We link the biological processes influenced by lncRNAs to cancer hallmarks in HCC and describe the associated functional mechanisms. This review sheds light on future research directions, including the potential therapeutic applications of lncRNAs.
Collapse
|
48
|
Bae MG, Hwang-Bo J, Lee DY, Lee YH, Chung IS. Effects of 6,8-Diprenylgenistein on VEGF-A-Induced Lymphangiogenesis and Lymph Node Metastasis in an Oral Cancer Sentinel Lymph Node Animal Model. Int J Mol Sci 2021; 22:ijms22020770. [PMID: 33466636 PMCID: PMC7828717 DOI: 10.3390/ijms22020770] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The major determining factor of prognosis of oral squamous cell carcinoma is cervical lymph node metastasis. 6,8-Diprenylgenistein (6,8-DG), an isoflavonoid isolated from Cudrania tricuspidata has been reported to have anti-microbial and anti-obesity activities. However, its effects on lymphangiogenesis and lymph node metastasis in oral cancer have not yet been reported. METHODS To investigate the in vitro inhibitory effects of 6,8-DG on VEGF-A-induced lymphangiogenesis, we performed the proliferation, tube formation, and migration assay using human lymphatic microvascular endothelial cells (HLMECs). RT-PCR, Western blot, immunoprecipitation, ELISA and co-immunoprecipitation assays were used to investigate the expression levels of proteins, and mechanism of 6,8-DG. The in vivo inhibitory effects of 6,8-DG were investigated using an oral cancer sentinel lymph node (OCSLN) animal model. RESULTS 6,8-DG inhibited the proliferation, migration and tube formation of rhVEGF-A treated HLMECs. In addition, the in vivo lymphatic vessel formation stimulated by rhVEGF-A was significantly reduced by 6,8-DG. 6,8-DG inhibited the expression of VEGF-A rather than other lymphangiogenic factors in CoCl2-treated SCCVII cells. 6,8-DG inhibited the expression and activation of VEGFR-2 stimulated by rhVEGF-A in HLMECs. Also, 6,8-DG inhibited the activation of the lymphangiogenesis-related downstream signaling factors such as FAK, PI3K, AKT, p38, and ERK in rhVEGF-A-treated HLMECs. Additionally, 6,8-DG inhibited the expression of the hypoxia-inducible factor (HIF-1α), which is involved in the expression of VEGF-A in CoCl2-treated SCCVII cells, and 6,8-DG inhibited VEGF-A signaling via interruption of the binding of VEGF-A and VEGFR-2 in HLMECs. In the VEGF-A-induced OCSLN animal model, we confirmed that 6,8-DG suppressed tumor-induced lymphangiogenesis and SLN metastasis. CONCLUSION These data suggest that 6,8-DG inhibits VEGF-A-induced lymphangiogenesis and lymph node metastasis in vitro and in vivo. Furthermore, the inhibitory effects of 6,8-DG are probably mediated by inhibition of VEGF-A expression in cancer cells and suppression of the VEGF-A/VEGFR-2 signaling pathway in HLMEC. Thus, 6,8-DG could be novel and valuable therapeutic agents for metastasis prevention and treatment of oral cancer.
Collapse
Affiliation(s)
- Mun Gyeong Bae
- Department of Genetic Engineering and Graduate School of Biotechnology, Kyung Hee University, Yongin 446-701, Korea; (M.G.B.); (J.H.-B.)
| | - Jeon Hwang-Bo
- Department of Genetic Engineering and Graduate School of Biotechnology, Kyung Hee University, Yongin 446-701, Korea; (M.G.B.); (J.H.-B.)
| | - Dae Young Lee
- Department of Herbal Crop Research, National Institute of Horticulture and Herbal Science, RDA, Eumseong 27709, Korea;
| | - Youn-Hyung Lee
- Department of Horticultural Biotechnology, Kyung Hee University, Yongin 17104, Korea;
| | - In Sik Chung
- Department of Genetic Engineering and Graduate School of Biotechnology, Kyung Hee University, Yongin 446-701, Korea; (M.G.B.); (J.H.-B.)
- Correspondence:
| |
Collapse
|
49
|
Permana AD, Nainu F, Moffatt K, Larrañeta E, Donnelly RF. Recent advances in combination of microneedles and nanomedicines for lymphatic targeted drug delivery. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2021; 13:e1690. [PMID: 33401339 DOI: 10.1002/wnan.1690] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022]
Abstract
Numerous diseases have been reported to affect the lymphatic system. As such, several strategies have been developed to deliver chemotherapeutics to this specific network of tissues and associated organs. Nanotechnology has been exploited as one of the main approaches to improve the lymphatic uptake of drugs. Different nanoparticle approaches utilized for both active and passive targeting of the lymphatic system are discussed here. Specifically, due to the rich abundance of lymphatic capillaries in the dermis, particular attention is given to this route of administration, as intradermal administration could potentially result in higher lymphatic uptake compared to other routes of administration. Recently, progress in microneedle research has attracted particular attention as an alternative for the use of conventional hypodermic injections. The benefits of microneedles, when compared to intradermal injection, are subsequently highlighted. Importantly, microneedles exhibit particular benefit in relation to therapeutic targeting of the lymphatic system, especially when combined with nanoparticles, which are further discussed. However, despite the apparent benefits provided by this combination approach, further comprehensive preclinical and clinical studies are now necessary to realize the potential extent of this dual-delivery platform, further taking into consideration eventual usability and acceptability in the intended patient end-users. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies Nanotechnology Approaches to Biology > Nanoscale Systems in Biology.
Collapse
Affiliation(s)
| | - Firzan Nainu
- Faculty of Pharmacy, Hasanuddin University, Makassar, Indonesia
| | - Kurtis Moffatt
- School of Pharmacy, Queen's University Belfast, Belfast, UK
| | | | | |
Collapse
|
50
|
Md Yusof K, Rosli R, Abdullah M, Avery-Kiejda KA. The Roles of Non-Coding RNAs in Tumor-Associated Lymphangiogenesis. Cancers (Basel) 2020; 12:cancers12113290. [PMID: 33172072 PMCID: PMC7694641 DOI: 10.3390/cancers12113290] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/26/2020] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
Simple Summary The lymphatic system plays key roles in the bodies’ defence against disease, including cancer. The expansion of this system is termed lymphangiogenesis and it is orchestrated by factors and conditions within the microenvironment. One approach to prevent cancer progression is by interfering with these microenvironment factors that promote this process and that facilitate the spread of cancer cells to distant organs. One of these factors are non-coding RNAs. This review will summarize recent findings of the distinct roles played by non-coding RNAs in the lymphatic system within normal tissues and tumours. Understanding the mechanisms involved in this process can provide new avenues for therapeutic intervention for inhibiting the spread of cancer. Abstract Lymphatic vessels are regarded as the ”forgotten” circulation. Despite this, growing evidence has shown significant roles for the lymphatic circulation in normal and pathological conditions in humans, including cancers. The dissemination of tumor cells to other organs is often mediated by lymphatic vessels that serve as a conduit and is often referred to as tumor-associated lymphangiogenesis. Some of the most well-studied lymphangiogenic factors that govern tumor lymphangiogenesis are the vascular endothelial growth factor (VEGF-C/D and VEGFR-2/3), neuroplilin-2 (NRP2), fibroblast growth factor (FGF), and hepatocyte growth factor (HGF), to name a few. However, recent findings have illustrated that non-coding RNAs are significantly involved in regulating gene expression in most biological processes, including lymphangiogenesis. In this review, we focus on the regulation of growth factors and non-coding RNAs (ncRNAs) in the lymphatic development in normal and cancer physiology. Then, we discuss the lymphangiogenic factors that necessitate tumor-associated lymphangiogenesis, with regards to ncRNAs in various types of cancer. Understanding the different roles of ncRNAs in regulating lymphatic vasculature in normal and cancer conditions may pave the way towards the development of ncRNA-based anti-lymphangiogenic therapy.
Collapse
Affiliation(s)
- Khairunnisa’ Md Yusof
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia; (K.M.Y.); (R.R.)
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, NSW 2308, Australia
- Medical Genetics, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
| | - Rozita Rosli
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia; (K.M.Y.); (R.R.)
| | - Maha Abdullah
- Department of Pathology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Selangor 43400, Malaysia;
| | - Kelly A. Avery-Kiejda
- Priority Research Centre for Cancer Research, Innovation and Translation, School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Newcastle, NSW 2308, Australia
- Medical Genetics, Hunter Medical Research Institute, New Lambton Heights, NSW 2305, Australia
- Correspondence:
| |
Collapse
|