1
|
Li ZZ, Zhou K, Wu Q, Liu B, Bu LL. Lymph node metastasis in cancer: Clearing the clouds to see the dawn. Crit Rev Oncol Hematol 2024; 204:104536. [PMID: 39426554 DOI: 10.1016/j.critrevonc.2024.104536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 09/26/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024] Open
Abstract
Lymph node metastasis (LNM) is often regarded as an indicator of poor prognosis in various cancers. Despite over three centuries of exploration since its discovery, the molecular mechanisms underlying LNM remain inconclusive. This review summarizes the molecular mechanisms of LNM, using the "PUMP+" principle for clarification. Pathological examination remains the gold standard for LNM diagnosis, yet there is a need to explore early diagnostic strategies that can effectively improve patient outcomes. With the advent of immunotherapy, discussions on the fate of lymph nodes (LN) have emerged, emphasizing the importance of preserving LN integrity prior to immunotherapy. This, in turn, poses higher demands for diagnostic accuracy and precision treatment of LNM. This review comprehensively discusses the molecular mechanisms, diagnostic methods, and treatment strategies for cancer lymph node metastasis, along with current bottlenecks and future directions in this field.
Collapse
Affiliation(s)
- Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Kan Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China.
| |
Collapse
|
2
|
Zhu L, Bai Y, Li A, Wan J, Sun M, Lou X, Duan X, Sheng Y, Lei N, Qin Z. IFN-γ-responsiveness of lymphatic endothelial cells inhibits melanoma lymphatic dissemination via AMPK-mediated metabolic control. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167314. [PMID: 38936516 DOI: 10.1016/j.bbadis.2024.167314] [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: 12/06/2023] [Revised: 05/28/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024]
Abstract
The integrity of the lymphatic system is critical for preventing the dissemination of tumor cells, such as melanoma, to distant parts of the body. IFN-γ is well studied as a negative regulator for lymphangiogenesis, which is strongly associated with cancer metastasis. However, the exact mechanisms underlying this process remain unclear. In the present study, we investigated whether IFN-γ signaling in lymphatic endothelial cells (LECs) affects tumor cell dissemination by regulating the barrier function of tumor-associated lymphatic vessels. Using LEC-specific IFN-γ receptor (IFN-γR) knockout mice, we found that the loss of IFN-γR in LECs increased the dissemination of melanoma cells into the draining lymph nodes. Notably, IFN-γ signaling in LECs inhibited trans-lymphatic endothelial cell migration of melanoma cells, indicating its regulation of lymphatic barrier function. Further investigations revealed that IFN-γ upregulated the expression of the tight junction protein Claudin-3 in LECs, while knockdown of Claudin-3 in LECs abolished IFN-γ-induced inhibition of trans-lymphatic endothelial migration activity. Mechanistically, IFN-γ inhibits AMPK signaling activation, which is involved in the regulation of fatty acid metabolism. Modulating fatty acid metabolism and AMPK activation in LECs also affected the lymphatic dissemination of melanoma cells, further confirming that this process is involved in IFN-γ-induced regulation of lymphatic barrier function. These results provide novel insights into how IFN-γ modulates tight junctions in LECs, inhibiting the dissemination of melanoma cells via the lymphatic vessels.
Collapse
Affiliation(s)
- Linyu Zhu
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Yueyue Bai
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Anqi Li
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Jiajia Wan
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Mengyao Sun
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China; School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaohan Lou
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Xixi Duan
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqiao Sheng
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China
| | - Ningjing Lei
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, Henan, China.
| | - Zhihai Qin
- Medical Research Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan, China; Key Laboratory of Protein and Peptide Pharmaceuticals, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
3
|
Nakanishi M, Ibe A, Morishita K, Shinagawa K, Yamamoto Y, Takahashi H, Ikemori K, Muragaki Y, Ehata S. Acid-sensing receptor GPR4 plays a crucial role in lymphatic cancer metastasis. Cancer Sci 2024; 115:1551-1563. [PMID: 38410865 PMCID: PMC11093208 DOI: 10.1111/cas.16098] [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: 08/15/2023] [Revised: 01/08/2024] [Accepted: 01/17/2024] [Indexed: 02/28/2024] Open
Abstract
Cancer tissues exhibit an acidic microenvironment owing to the accumulation of protons and lactic acid produced by cancer and inflammatory cells. To examine the role of an acidic microenvironment in lymphatic cancer metastasis, gene expression profiling was conducted using human dermal lymphatic endothelial cells (HDLECs) treated with a low pH medium. Microarray and gene set enrichment analysis revealed that acid treatment induced the expression of inflammation-related genes in HDLECs, including genes encoding chemokines and adhesion molecules. Acid treatment-induced chemokines C-X3-C motif chemokine ligand 1 (CX3CL1) and C-X-C motif chemokine ligand 6 (CXCL6) autocrinally promoted the growth and tube formation of HDLECs. The expression of vascular cell adhesion molecule 1 (VCAM-1) increased in HDLECs after acid treatment in a time-dependent manner, which, in turn, enhanced their adhesion to melanoma cells. Among various acid-sensing receptors, HDLECs basally expressed G protein-coupled receptor 4 (GPR4), which was augmented under the acidic microenvironment. The induction of chemokines or VCAM-1 under acidic conditions was attenuated by GPR4 knockdown in HDLECs. In addition, lymph node metastases in a mouse melanoma model were suppressed by administering an anti-VCAM-1 antibody or a GPR4 antagonist. These results suggest that an acidic microenvironment modifies the function of lymphatic endothelial cells via GPR4, thereby promoting lymphatic cancer metastasis. Acid-sensing receptors and their downstream molecules might serve as preventive or therapeutic targets in cancer.
Collapse
Affiliation(s)
- Masako Nakanishi
- Department of Pathology, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Akiya Ibe
- Department of Pathology, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Kiyoto Morishita
- Department of Pathology, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Kazutaka Shinagawa
- Department of Pathology, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Yushi Yamamoto
- Department of Pathology, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Hibiki Takahashi
- Department of Pathology, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Kyoka Ikemori
- Department of Pathology, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Yasuteru Muragaki
- Department of Pathology, School of MedicineWakayama Medical UniversityWakayamaJapan
| | - Shogo Ehata
- Department of Pathology, School of MedicineWakayama Medical UniversityWakayamaJapan
| |
Collapse
|
4
|
Zheng H, An M, Luo Y, Diao X, Zhong W, Pang M, Lin Y, Chen J, Li Y, Kong Y, Zhao Y, Yin Y, Ai L, Huang J, Chen C, Lin T. PDGFRα +ITGA11 + fibroblasts foster early-stage cancer lymphovascular invasion and lymphatic metastasis via ITGA11-SELE interplay. Cancer Cell 2024; 42:682-700.e12. [PMID: 38428409 DOI: 10.1016/j.ccell.2024.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 11/20/2023] [Accepted: 02/05/2024] [Indexed: 03/03/2024]
Abstract
Cancer-associated fibroblasts (CAFs) exhibit considerable heterogeneity in advanced cancers; however, the functional annotation and mechanism of CAFs in early-stage cancers remain elusive. Utilizing single-cell RNA sequencing and spatial transcriptomic, we identify a previously unknown PDGFRα+ITGA11+ CAF subset in early-stage bladder cancer (BCa). Multicenter clinical analysis of a 910-case cohort confirms that PDGFRα+ITGA11+ CAFs are associated with lymphovascular invasion (LVI) and poor prognosis in early-stage BCa. These CAFs facilitate LVI and lymph node (LN) metastasis in early-stage BCa, as evidenced in a PDGFRα+ITGA11+ CAFs-specific deficient mouse model. Mechanistically, PDGFRα+ITGA11+ CAFs promote lymphangiogenesis via recognizing ITGA11 surface receptor SELE on lymphatic endothelial cells to activate SRC-p-VEGFR3-MAPK pathway. Further, CHI3L1 from PDGFRα+ITGA11+ CAFs aligns the surrounding matrix to assist cancer cell intravasation, fostering early-stage BCa LVI and LN metastasis. Collectively, our study reveals the crucial role of PDGFRα+ITGA11+ CAFs in shaping metastatic landscape, informing the treatment of early-stage BCa LVI.
Collapse
Affiliation(s)
- Hanhao Zheng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China
| | - Mingjie An
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China
| | - Yuming Luo
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, P.R. China
| | - Xiayao Diao
- Department of Thoracic Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wenlong Zhong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China
| | - Mingrui Pang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China
| | - Yan Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China
| | - Jiancheng Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China
| | - Yuanlong Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China
| | - Yao Kong
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong, P.R. China
| | - Yue Zhao
- Department of Tumor Intervention, Sun Yat-sen University First Affiliated Hospital, Guangzhou, Guangdong, P.R. China
| | - Yina Yin
- Department of Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, P.R. China
| | - Le Ai
- Department of Oncology, Sun Yat-sen Memorial Hospital, Guangzhou, Guangdong, P.R. China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China
| | - Changhao Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China.
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, P.R. China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, P.R. China.
| |
Collapse
|
5
|
Zeng W, Wang Y, Zhang Q, Hu C, Li J, Feng J, Hu C, Su Y, Lou J, Long L, Zhou X. Neutrophil Nanodecoys Inhibit Tumor Metastasis by Blocking the Interaction between Tumor Cells and Neutrophils. ACS NANO 2024; 18:7363-7378. [PMID: 38422392 DOI: 10.1021/acsnano.3c08946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Cancer metastasis is the main cause of cancer-related deaths and involves the interaction between tumor cells and neutrophils. In this study, we developed activated neutrophil membrane-coated nanoparticles (aNEM NPs) as nanodecoys to block neutrophil-mediated cancer metastasis. The aNEM NPs were fabricated by cloaking poly(lactic acid) nanoparticles with membranes derived from activated neutrophils and inherited the functional proteins of activated neutrophils. We demonstrated that aNEM NPs could interfere with the recruitment of neutrophils to the primary tumor and premetastatic niches, inhibit the adhesion of neutrophils to tumor vascular endothelium and circulating tumor cells (CTCs), and disrupt the formation of CTC-neutrophil clusters in vitro and in vivo. In 4T1-bearing mice, aNEM NPs could effectively reduce breast cancer metastasis to various organs in mice. Our results suggest that aNEM NPs are a promising nanomedicine for preventing or treating cancer metastasis by acting as neutrophil nanodecoys.
Collapse
Affiliation(s)
- Weiya Zeng
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Ying Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
- Leibo County People's Hospital, Sichuan 616500, China
| | - Qing Zhang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Chengyi Hu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Jing Li
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Jinwei Feng
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Chenglu Hu
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Yong Su
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Jie Lou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
| | - Ling Long
- Department of Oncology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China
| | - Xing Zhou
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, China
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| |
Collapse
|
6
|
Ping J, Liu W, Chen Z, Li C. Lymph node metastases in breast cancer: Mechanisms and molecular imaging. Clin Imaging 2023; 103:109985. [PMID: 37757640 DOI: 10.1016/j.clinimag.2023.109985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/29/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023]
Abstract
Breast cancer is the most common malignant disease of women in the world. Breast cancer often metastasizes to axillary lymph nodes. Accurate assessment of the status of axillary lymph nodes is crucial to the staging and treatment of breast cancer. None of the methods used clinically for preoperative noninvasive examination of axillary lymph nodes can accurately identify cancer cells from a molecular level. In recent years, with the in-depth study of lymph node metastases, the mechanisms and molecular imaging of lymph node metastases in breast cancer have been reported. In this review, we highlight the new progress in the study of the main mechanisms of lymph node metastases in breast cancer. In addition, we analyze the advantages and disadvantages of traditional preoperative axillary lymph node imaging methods for breast cancer, and list molecular imaging methods that can accurately identify breast cancer cells in lymph nodes.
Collapse
Affiliation(s)
- Jieyi Ping
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China
| | - Wei Liu
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China
| | - Zhihui Chen
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China
| | - Cuiying Li
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, No. 300 Guangzhou Road, Nanjing 210029, China.
| |
Collapse
|
7
|
Chang A, Wang Y, Guo X, Sun Z, Ling J, Pan J, Zhuo X. Identification of immune-related genes in the prognosis of head and neck cancer using a novel prognostic signature model. Oral Surg Oral Med Oral Pathol Oral Radiol 2023; 136:478-489. [PMID: 37620228 DOI: 10.1016/j.oooo.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 06/11/2023] [Accepted: 07/02/2023] [Indexed: 08/26/2023]
Abstract
BACKGROUND Increasing evidence indicates that the immune response plays a critical role in the development of head and neck cancer (HNC). We aimed to develop an immune-related gene signature and evaluate its prognostic value in patients with HNC. METHODS We retrieved an HNC cohort from The Cancer Genome Atlas database and divided the samples into high-risk and low-risk groups based on the median of the immune and stromal scores. We performed Venn and Cox analyses to identify the immune-related DEGs to use in our prognostic model. We evaluated the correlation between the model and immune-cell infiltration and validated the prognostic value of the model by applying it to 2 external HNC cohorts. RESULTS We identified 7 DEGs-CCR4, WDFY4, VCAM1, LYZ, VSIG4, XIRP1, and CMKLR1-to use in our prognostic model and validated the model by applying it to 2 external HNC cohorts. We found that risk scores based on the model could reflect the status of the tumor microenvironment and that VSIG4 might be associated with lymph node metastasis in HNC. CONCLUSIONS We developed a highly accurate immune-related prognostic 7-gene model in HNC predication, indicating that these 7 genes play critical roles in the tumor microenvironment.
Collapse
Affiliation(s)
- Aoshuang Chang
- Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yan Wang
- Department of Respiratory Medicine, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Xiaopeng Guo
- Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhen Sun
- Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| | - Junjun Ling
- Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jigang Pan
- School of Basic Medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Xianlu Zhuo
- Affiliated Hospital, Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
8
|
Viúdez-Pareja C, Kreft E, García-Caballero M. Immunomodulatory properties of the lymphatic endothelium in the tumor microenvironment. Front Immunol 2023; 14:1235812. [PMID: 37744339 PMCID: PMC10512957 DOI: 10.3389/fimmu.2023.1235812] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/08/2023] [Indexed: 09/26/2023] Open
Abstract
The tumor microenvironment (TME) is an intricate complex and dynamic structure composed of various cell types, including tumor, stromal and immune cells. Within this complex network, lymphatic endothelial cells (LECs) play a crucial role in regulating immune responses and influencing tumor progression and metastatic dissemination to lymph node and distant organs. Interestingly, LECs possess unique immunomodulatory properties that can either promote or inhibit anti-tumor immune responses. In fact, tumor-associated lymphangiogenesis can facilitate tumor cell dissemination and metastasis supporting immunoevasion, but also, different molecular mechanisms involved in LEC-mediated anti-tumor immunity have been already described. In this context, the crosstalk between cancer cells, LECs and immune cells and how this communication can shape the immune landscape in the TME is gaining increased interest in recent years. In this review, we present a comprehensive and updated report about the immunomodulatory properties of the lymphatic endothelium within the TME, with special focus on primary tumors and tumor-draining lymph nodes. Furthermore, we outline emerging research investigating the potential therapeutic strategies targeting the lymphatic endothelium to enhance anti-tumor immune responses. Understanding the intricate mechanisms involved in LEC-mediated immune modulation in the TME opens up new possibilities for the development of innovative approaches to fight cancer.
Collapse
Affiliation(s)
- Cristina Viúdez-Pareja
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| | - Ewa Kreft
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| | - Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| |
Collapse
|
9
|
Cazzola A, Calzón Lozano D, Menne DH, Dávila Pedrera R, Liu J, Peña-Jiménez D, Fontenete S, Halin C, Perez-Moreno M. Lymph Vessels Associate with Cancer Stem Cells from Initiation to Malignant Stages of Squamous Cell Carcinoma. Int J Mol Sci 2023; 24:13615. [PMID: 37686421 PMCID: PMC10488284 DOI: 10.3390/ijms241713615] [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: 08/08/2023] [Revised: 08/28/2023] [Accepted: 09/01/2023] [Indexed: 09/10/2023] Open
Abstract
Tumor-associated lymph vessels and lymph node involvement are critical staging criteria in several cancers. In skin squamous cell carcinoma, lymph vessels play a role in cancer development and metastatic spread. However, their relationship with the cancer stem cell niche at early tumor stages remains unclear. To address this gap, we studied the lymph vessel localization at the cancer stem cell niche and observed an association from benign skin lesions to malignant stages of skin squamous cell carcinoma. By co-culturing lymphatic endothelial cells with cancer cell lines representing the initiation and promotion stages, and conducting RNA profiling, we observed a reciprocal induction of cell adhesion, immunity regulation, and vessel remodeling genes, suggesting dynamic interactions between lymphatic and cancer cells. Additionally, imaging analyses of the cultured cells revealed the establishment of heterotypic contacts between cancer cells and lymph endothelial cells, potentially contributing to the observed distribution and maintenance at the cancer stem cell niche, inducing downstream cellular responses. Our data provide evidence for an association of lymph vessels from the early stages of skin squamous cell carcinoma development, opening new avenues for better comprehending their involvement in cancer progression.
Collapse
Affiliation(s)
- Anna Cazzola
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - David Calzón Lozano
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Dennis Hirsch Menne
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Raquel Dávila Pedrera
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Jingcheng Liu
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Daniel Peña-Jiménez
- Unidad de Investigación Biomédica, Universidad Alfonso X el Sabio (UAX), Avenida de la Universidad 1, Villanueva de la Cañada, 28691 Madrid, Spain
| | - Silvia Fontenete
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, 8093 Zurich, Switzerland;
| | - Mirna Perez-Moreno
- Section for Cell Biology and Physiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
10
|
Kuhn E, Gambini D, Despini L, Asnaghi D, Runza L, Ferrero S. Updates on Lymphovascular Invasion in Breast Cancer. Biomedicines 2023; 11:biomedicines11030968. [PMID: 36979946 PMCID: PMC10046167 DOI: 10.3390/biomedicines11030968] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Traditionally, lymphovascular invasion (LVI) has represented one of the foremost pathological features of malignancy and has been associated with a worse prognosis in different cancers, including breast carcinoma. According to the most updated reporting protocols, the assessment of LVI is required in the pathology report of breast cancer surgical specimens. Importantly, strict histological criteria should be followed for LVI assessment, which nevertheless is encumbered by inconsistency in interpretation among pathologists, leading to significant interobserver variability and scarce reproducibility. Current guidelines for breast cancer indicate biological factors as the main determinants of oncological and radiation therapy, together with TNM staging and age. In clinical practice, the widespread use of genomic assays as a decision-making tool for hormone receptor-positive, HER2-negative breast cancer and the subsequent availability of a reliable prognostic predictor have likely scaled back interest in LVI's predictive value. However, in selected cases, the presence of LVI impacts adjuvant therapy. This review summarizes current knowledge on LVI in breast cancer with regard to definition, histopathological assessment, its biological understanding, clinicopathological association, and therapeutic implications.
Collapse
Affiliation(s)
- Elisabetta Kuhn
- Department of Biomedical Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
- Pathology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Donatella Gambini
- Department of Neurorehabilitation Sciences, Casa di Cura Igea, 20129 Milan, Italy
| | - Luca Despini
- Breast Surgery Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Dario Asnaghi
- Radiotherapy Unit, ASST Grande Ospedale Metropolitano Niguarda, 20162 Milan, Italy
| | - Letterio Runza
- Pathology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Stefano Ferrero
- Department of Biomedical Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy
- Pathology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
11
|
Abstract
The lymphatic system, composed of initial and collecting lymphatic vessels as well as lymph nodes that are present in almost every tissue of the human body, acts as an essential transport system for fluids, biomolecules and cells between peripheral tissues and the central circulation. Consequently, it is required for normal body physiology but is also involved in the pathogenesis of various diseases, most notably cancer. The important role of tumor-associated lymphatic vessels and lymphangiogenesis in the formation of lymph node metastasis has been elucidated during the last two decades, whereas the underlying mechanisms and the relation between lymphatic and peripheral organ dissemination of cancer cells are incompletely understood. Lymphatic vessels are also important for tumor-host communication, relaying molecular information from a primary or metastatic tumor to regional lymph nodes and the circulatory system. Beyond antigen transport, lymphatic endothelial cells, particularly those residing in lymph node sinuses, have recently been recognized as direct regulators of tumor immunity and immunotherapy responsiveness, presenting tumor antigens and expressing several immune-modulatory signals including PD-L1. In this review, we summarize recent discoveries in this rapidly evolving field and highlight strategies and challenges of therapeutic targeting of lymphatic vessels or specific lymphatic functions in cancer patients.
Collapse
Affiliation(s)
- Lothar C Dieterich
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Carlotta Tacconi
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,Department of Biosciences, University of Milan, Milan, Italy
| | - Luca Ducoli
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Michael Detmar
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Hypoxia orchestrates the lymphovascular–immune ensemble in cancer. Trends Cancer 2022; 8:771-784. [DOI: 10.1016/j.trecan.2022.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/31/2022] [Accepted: 04/25/2022] [Indexed: 11/18/2022]
|
13
|
Angiotensin II Induces Cardiac Edema and Hypertrophic Remodeling through Lymphatic-Dependent Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:5044046. [PMID: 35222798 PMCID: PMC8881141 DOI: 10.1155/2022/5044046] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/17/2021] [Accepted: 01/25/2022] [Indexed: 12/14/2022]
Abstract
Cardiac lymphatic vessel growth (lymphangiogenesis) and integrity play an essential role in maintaining tissue fluid balance. Inhibition of lymphatic lymphangiogenesis is involved in cardiac edema and cardiac remodeling after ischemic injury or pressure overload. However, whether lymphatic vessel integrity is disrupted during angiotensin II- (Ang II-) induced cardiac remodeling remains to be investigated. In this study, cardiac remodeling models were established by Ang II (1000 ng/kg/min) in VEGFR-3 knockdown (Lyve-1Cre VEGFR-3f/−) and wild-type (VEGFR-3f/f) littermates. Our results indicated that Ang II infusion not only induced cardiac lymphangiogenesis and upregulation of VEGF-C and VEGFR-3 expression in the time-dependent manner but also enhanced proteasome activity, MKP5 and VE-cadherin degradation, p38 MAPK activation, and lymphatic vessel hyperpermeability. Moreover, VEGFR-3 knockdown significantly inhibited cardiac lymphangiogenesis in mice, resulting in exacerbation of tissue edema, hypertrophy, fibrosis superoxide production, inflammation, and heart failure (HF). Conversely, administration of epoxomicin (a selective proteasome inhibitor) markedly mitigated Ang II-induced cardiac edema, remodeling, and dysfunction; upregulated MKP5 and VE-cadherin expression; inactivated p38 MAPK; and reduced lymphatic vessel hyperpermeability in WT mice, indicating that inhibition of proteasome activity is required to maintain lymphatic endothelial cell (LEC) integrity. Our results show that both cardiac lymphangiogenesis and lymphatic barrier hyperpermeability are implicated in Ang II-induced adaptive hypertrophic remodeling and dysfunction. Proteasome-mediated hyperpermeability of LEC junctions plays a predominant role in the development of cardiac remodeling. Selective stimulation of lymphangiogenesis or inhibition of proteasome activity may be a potential therapeutic option for treating hypertension-induced cardiac remodeling.
Collapse
|
14
|
Leary N, Walser S, He Y, Cousin N, Pereira P, Gallo A, Collado‐Diaz V, Halin C, Garcia‐Silva S, Peinado H, Dieterich LC. Melanoma-derived extracellular vesicles mediate lymphatic remodelling and impair tumour immunity in draining lymph nodes. J Extracell Vesicles 2022; 11:e12197. [PMID: 35188342 PMCID: PMC8859913 DOI: 10.1002/jev2.12197] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/14/2022] [Accepted: 01/20/2022] [Indexed: 12/12/2022] Open
Abstract
Tumour-draining lymph nodes (LNs) undergo massive remodelling including expansion of the lymphatic sinuses, a process that has been linked to lymphatic metastasis by creation of a pre-metastatic niche. However, the signals leading to these changes have not been completely understood. Here, we found that extracellular vesicles (EVs) derived from melanoma cells are rapidly transported by lymphatic vessels to draining LNs, where they selectively interact with lymphatic endothelial cells (LECs) as well as medullary sinus macrophages. Interestingly, uptake of melanoma EVs by LN-resident LECs was partly dependent on lymphatic VCAM-1 expression, and induced transcriptional changes as well as proliferation of those cells. Furthermore, melanoma EVs shuttled tumour antigens to LN LECs for cross-presentation on MHC-I, resulting in apoptosis induction in antigen-specific CD8+ T cells. In conclusion, our data identify EV-mediated melanoma-LN LEC communication as a new pathway involved in tumour progression and tumour immune inhibition, suggesting that EV uptake or effector mechanisms in LECs might represent a new target for melanoma therapy.
Collapse
Affiliation(s)
- Noelle Leary
- Institute of Pharmaceutical SciencesSwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Sarina Walser
- Institute of Pharmaceutical SciencesSwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Yuliang He
- Institute of Pharmaceutical SciencesSwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Nikola Cousin
- Institute of Pharmaceutical SciencesSwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Paulo Pereira
- Institute of Pharmaceutical SciencesSwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Alessandro Gallo
- Institute of Pharmaceutical SciencesSwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Victor Collado‐Diaz
- Institute of Pharmaceutical SciencesSwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Cornelia Halin
- Institute of Pharmaceutical SciencesSwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| | - Susana Garcia‐Silva
- Microenvironment and Metastasis LaboratorySpanish National Cancer Research CentreMadridSpain
| | - Hector Peinado
- Microenvironment and Metastasis LaboratorySpanish National Cancer Research CentreMadridSpain
| | - Lothar C. Dieterich
- Institute of Pharmaceutical SciencesSwiss Federal Institute of Technology (ETH) ZurichZurichSwitzerland
| |
Collapse
|
15
|
Fujimoto N, Dieterich LC. Mechanisms and Clinical Significance of Tumor Lymphatic Invasion. Cells 2021; 10:cells10102585. [PMID: 34685565 PMCID: PMC8533989 DOI: 10.3390/cells10102585] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/20/2021] [Accepted: 09/25/2021] [Indexed: 12/17/2022] Open
Abstract
Tumor-associated lymphatic vessels play an important role in tumor progression, mediating lymphatic dissemination of malignant cells to tumor-draining lymph nodes and regulating tumor immunity. An early, necessary step in the lymphatic metastasis cascade is the invasion of lymphatic vessels by tumor cell clusters or single tumor cells. In this review, we discuss our current understanding of the underlying cellular and molecular mechanisms, which include tumor-specific as well as normal, developmental and immunological processes “hijacked” by tumor cells to gain access to the lymphatic system. Furthermore, we summarize the prognostic value of lymphatic invasion, discuss its relationship with local recurrence, lymph node and distant metastasis, and highlight potential therapeutic options and challenges.
Collapse
Affiliation(s)
- Noriki Fujimoto
- Department of Dermatology, Shiga University of Medical Science, Otsu 520-2192, Japan;
| | - Lothar C. Dieterich
- Institute of Pharmaceutical Sciences, Swiss Federal Institute of Technology (ETH) Zurich, Vladimir-Prelog-Weg 1-5/10, 8093 Zurich, Switzerland
- Correspondence:
| |
Collapse
|
16
|
Rezzola S, Sigmund EC, Halin C, Ronca R. The lymphatic vasculature: An active and dynamic player in cancer progression. Med Res Rev 2021; 42:576-614. [PMID: 34486138 PMCID: PMC9291933 DOI: 10.1002/med.21855] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 04/29/2021] [Accepted: 08/26/2021] [Indexed: 12/16/2022]
Abstract
The lymphatic vasculature has been widely described and explored for its key functions in fluid homeostasis and in the organization and modulation of the immune response. Besides transporting immune cells, lymphatic vessels play relevant roles in tumor growth and tumor cell dissemination. Cancer cells that have invaded into afferent lymphatics are propagated to tumor‐draining lymph nodes (LNs), which represent an important hub for metastatic cell arrest and growth, immune modulation, and secondary dissemination to distant sites. In recent years many studies have reported new mechanisms by which the lymphatic vasculature affects cancer progression, ranging from induction of lymphangiogenesis to metastatic niche preconditioning or immune modulation. In this review, we provide an up‐to‐date description of lymphatic organization and function in peripheral tissues and in LNs and the changes induced to this system by tumor growth and progression. We will specifically focus on the reported interactions that occur between tumor cells and lymphatic endothelial cells (LECs), as well as on interactions between immune cells and LECs, both in the tumor microenvironment and in tumor‐draining LNs. Moreover, the most recent prognostic and therapeutic implications of lymphatics in cancer will be reported and discussed in light of the new immune‐modulatory roles that have been ascribed to LECs.
Collapse
Affiliation(s)
- Sara Rezzola
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Elena C Sigmund
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Cornelia Halin
- Institute of Pharmaceutical Sciences, ETH Zurich, Zurich, Switzerland
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
17
|
Gillot L, Baudin L, Rouaud L, Kridelka F, Noël A. The pre-metastatic niche in lymph nodes: formation and characteristics. Cell Mol Life Sci 2021; 78:5987-6002. [PMID: 34241649 PMCID: PMC8316194 DOI: 10.1007/s00018-021-03873-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 05/10/2021] [Accepted: 06/05/2021] [Indexed: 02/06/2023]
Abstract
Lymph node metastasis is a crucial prognostic parameter in many different types of cancers and a gateway for further dissemination to distant organs. Prior to metastatic dissemination, the primary tumor prepares for the remodeling of the draining (sentinel) lymph node by secreting soluble factors or releasing extracellular vesicles that are transported by lymphatic vessels. These important changes occur before the appearance of the first metastatic cell and create what is known as a pre-metastatic niche giving rise to the subsequent survival and growth of metastatic cells. In this review, the lymph node structure, matrix composition and the emerging heterogeneity of cells forming it are described. Current knowledge of the major cellular and molecular processes associated with nodal pre-metastatic niche formation, including lymphangiogenesis, extracellular matrix remodeling, and immunosuppressive cell enlisting in lymph nodes are additionally summarized. Finally, future directions that research could possibly take and the clinical impact are discussed.
Collapse
Affiliation(s)
- Lionel Gillot
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Avenue Hippocrate 13, 4000 Liege, Belgium
| | - Louis Baudin
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Avenue Hippocrate 13, 4000 Liege, Belgium
| | - Loïc Rouaud
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Avenue Hippocrate 13, 4000 Liege, Belgium
| | - Frédéric Kridelka
- Department of Obstetrics and Gynecology, CHU of Liege, 4000 Liege, Belgium
| | - Agnès Noël
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Avenue Hippocrate 13, 4000 Liege, Belgium
| |
Collapse
|
18
|
Kumaravel S, Abbey CA, Bayless KJ, Chakraborty S. The β 1-integrin plays a key role in LEC invasion in an optimized 3-D collagen matrix model. Am J Physiol Cell Physiol 2020; 319:C1045-C1058. [PMID: 33052069 DOI: 10.1152/ajpcell.00299.2020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Lymphangiogenesis, or formation of new lymphatic vessels, is a tightly regulated process that is controlled by growth factor signaling and biomechanical cues. Lymphatic endothelial cells (LECs) undergo remodeling, migration, and proliferation to invade the surrounding extracellular matrix (ECM) during both physiological and pathological lymphangiogenesis. This study optimized conditions for an in vitro three-dimensional (3-D) collagen-based model that induced LEC invasion and recapitulated physiological formation of lymphatic capillaries with lumens. Invasion of LECs was enhanced in the presence of sphingosine 1-phosphate (S1P). Effects of various known lymphangiogenic factors, vascular endothelial growth factor (VEGF)-A, basic fibroblast growth factor (bFGF), interleukin (IL)-8, and hepatocyte growth factor (HGF), were tested on LEC sprout formation synergistically with VEGF-C. Several of these growth factors significantly enhanced LEC invasion, and synergistic effects of some of these further enhanced the sprouting density and lumen volume. To determine the contribution of specific ECM components, we analyzed the expression of different integrin subunits. Basal expressions of the integrin α5- and integrin β1-subunits were high in LECs. The addition of fibronectin, which mediates cellular responses through these integrins, enhanced LEC sprouting density and sprout length dose-dependently. siRNA-mediated knockdown of the integrin β1-subunit suppressed LEC invasion and also inhibited VEGF receptor (VEGFR)3 and ERK activation. Furthermore, exposing LECs to the inflammatory mediator lipopolysaccharide (LPS) inhibited sprouting. This optimized model for LEC invasion includes S1P, VEGF-C, and fibronectin within a 3-D collagen matrix, along with VEGF-C, VEGF-A, bFGF, and HGF in the culture medium, and provides a useful tool to investigate the functional effect of various lymphangiogenic factors and inhibitors.
Collapse
Affiliation(s)
- Subhashree Kumaravel
- Department of Medical Physiology, Texas A&M Health Science Center, College of Medicine, Bryan, Texas
| | - Colette A Abbey
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - Kayla J Bayless
- Department of Molecular and Cellular Medicine, Texas A&M Health Science Center, Bryan, Texas
| | - Sanjukta Chakraborty
- Department of Medical Physiology, Texas A&M Health Science Center, College of Medicine, Bryan, Texas
| |
Collapse
|
19
|
Gengenbacher N, Singhal M, Mogler C, Hai L, Milde L, Pari AAA, Besemfelder E, Fricke C, Baumann D, Gehrs S, Utikal J, Felcht M, Hu J, Schlesner M, Offringa R, Chintharlapalli SR, Augustin HG. Timed Ang2-Targeted Therapy Identifies the Angiopoietin-Tie Pathway as Key Regulator of Fatal Lymphogenous Metastasis. Cancer Discov 2020; 11:424-445. [PMID: 33106316 DOI: 10.1158/2159-8290.cd-20-0122] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 08/13/2020] [Accepted: 10/09/2020] [Indexed: 11/16/2022]
Abstract
Recent clinical and preclinical advances have highlighted the existence of a previously hypothesized lymphogenous route of metastasis. However, due to a lack of suitable preclinical modeling tools, its contribution to long-term disease outcome and relevance for therapy remain controversial. Here, we established a genetically engineered mouse model (GEMM) fragment-based tumor model uniquely sustaining a functional network of intratumoral lymphatics that facilitates seeding of fatal peripheral metastases. Multiregimen survival studies and correlative patient data identified primary tumor-derived Angiopoietin-2 (Ang2) as a potent therapeutic target to restrict lymphogenous tumor cell dissemination. Mechanistically, tumor-associated lymphatic endothelial cells (EC), in contrast to blood vascular EC, were found to be critically addicted to the Angiopoietin-Tie pathway. Genetic manipulation experiments in combination with single-cell mapping revealed agonistically acting Ang2-Tie2 signaling as key regulator of lymphatic maintenance. Correspondingly, acute presurgical Ang2 neutralization was sufficient to prolong survival by regressing established intratumoral lymphatics, hence identifying a therapeutic regimen that warrants further clinical evaluation. SIGNIFICANCE: Exploiting multiple mouse tumor models including a unique GEMM-derived allograft system in combination with preclinical therapy designs closely matching the human situation, this study provides fundamental insight into the biology of tumor-associated lymphatic EC and defines an innovative presurgical therapeutic window of migrastatic Ang2 neutralization to restrict lymphogenous metastasis.This article is highlighted in the In This Issue feature, p. 211.
Collapse
Affiliation(s)
- Nicolas Gengenbacher
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Faculty of Biosciences, Heidelberg University, Mannheim, Germany
| | - Mahak Singhal
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Faculty of Biosciences, Heidelberg University, Mannheim, Germany
| | - Carolin Mogler
- Institute of Pathology, TUM School of Medicine, Munich, Germany
| | - Ling Hai
- Junior Group Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Laura Milde
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Faculty of Biosciences, Heidelberg University, Mannheim, Germany
| | - Ashik Ahmed Abdul Pari
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Faculty of Biosciences, Heidelberg University, Mannheim, Germany
| | - Eva Besemfelder
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Claudine Fricke
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany
| | - Daniel Baumann
- Faculty of Biosciences, Heidelberg University, Mannheim, Germany.,Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephanie Gehrs
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany.,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Faculty of Biosciences, Heidelberg University, Mannheim, Germany
| | - Jochen Utikal
- Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Moritz Felcht
- Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,Department of Dermatology, Venereology and Allergology, University Medical Center Mannheim, Heidelberg University, Mannheim, Germany
| | - Junhao Hu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, China
| | - Matthias Schlesner
- Junior Group Bioinformatics and Omics Data Analytics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Rienk Offringa
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | | | - Hellmut G Augustin
- Division of Vascular Oncology and Metastasis, German Cancer Research Center (DKFZ-ZMBH Alliance), Heidelberg, Germany. .,Department of Vascular Biology and Tumor Angiogenesis, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.,German Cancer Consortium, Heidelberg, Germany
| |
Collapse
|
20
|
Van de Velde M, Ebroin M, Durré T, Joiret M, Gillot L, Blacher S, Geris L, Kridelka F, Noel A. Tumor exposed-lymphatic endothelial cells promote primary tumor growth via IL6. Cancer Lett 2020; 497:154-164. [PMID: 33080310 PMCID: PMC7723984 DOI: 10.1016/j.canlet.2020.10.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/14/2020] [Accepted: 10/14/2020] [Indexed: 12/22/2022]
Abstract
Solid tumors are composed of tumor cells and stromal cells including lymphatic endothelial cells (LEC), which are mainly viewed as cells forming lymphatic vessels involved in the transport of metastatic and immune cells. We here reveal a new mechanism by which tumor exposed-LEC (teLEC) exert mitogenic effects on tumor cells. Our conclusions are supported by morphological and molecular changes induced in teLEC that in turn enhance cancer cell invasion in 3D cultures and tumor cell proliferation in vivo. The characterization of teLEC secretome by RNA-Sequencing and cytokine array revealed that interleukine-6 (IL6) is one of the most modulated molecules in teLEC, whose production was negligible in unexposed LEC. Notably, neutralizing anti-human IL6 antibody abrogated teLEC-mediated mitogenic effects in vivo, when LEC were mixed with tumor cells in the ear sponge assay. We here assign a novel function to teLEC that is beyond their role of lymphatic vessel formation. This work highlights a new paradigm, in which teLEC exert “fibroblast-like properties”, contribute in a paracrine manner to the control of tumor cell properties and are worth considering as key stromal determinant in future studies. teLEC, but not normal LEC, produce huge amount of IL6. IL6-derived teLEC exert mitogenic effect on tumor cells, in the primary tumor. teLEC act as fibroblast-like cells in the tumor microenvironment. It warrants to revisit the “vascular-centric view” of LECs.
Collapse
Affiliation(s)
- Maureen Van de Velde
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, B23, Avenue Hippocrate 13, Sart-Tilman, B-4000, Liege, Belgium
| | - Marie Ebroin
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, B23, Avenue Hippocrate 13, Sart-Tilman, B-4000, Liege, Belgium
| | - Tania Durré
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, B23, Avenue Hippocrate 13, Sart-Tilman, B-4000, Liege, Belgium
| | - Marc Joiret
- Biomechanics Research Unit, GIGA-In Silico Medicine, Liege University, B34, Sart-Tilman, 4000, Liège, Belgium
| | - Lionel Gillot
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, B23, Avenue Hippocrate 13, Sart-Tilman, B-4000, Liege, Belgium
| | - Silvia Blacher
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, B23, Avenue Hippocrate 13, Sart-Tilman, B-4000, Liege, Belgium
| | - Liesbet Geris
- Biomechanics Research Unit, GIGA-In Silico Medicine, Liege University, B34, Sart-Tilman, 4000, Liège, Belgium
| | - Frédéric Kridelka
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, B23, Avenue Hippocrate 13, Sart-Tilman, B-4000, Liege, Belgium; Department of Obstetrics and Gynecology, CHU Liege, Sart-Tilman, 4000, Liege, Belgium
| | - Agnès Noel
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, B23, Avenue Hippocrate 13, Sart-Tilman, B-4000, Liege, Belgium.
| |
Collapse
|
21
|
Dieterich LC, Bikfalvi A. The tumor organismal environment: Role in tumor development and cancer immunotherapy. Semin Cancer Biol 2020; 65:197-206. [DOI: 10.1016/j.semcancer.2019.12.021] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/03/2019] [Accepted: 12/22/2019] [Indexed: 02/07/2023]
|
22
|
Blei F. Update April 2020. Lymphat Res Biol 2020. [DOI: 10.1089/lrb.2020.29083.fb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
23
|
Parallels of Resistance between Angiogenesis and Lymphangiogenesis Inhibition in Cancer Therapy. Cells 2020; 9:cells9030762. [PMID: 32244922 PMCID: PMC7140636 DOI: 10.3390/cells9030762] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/13/2020] [Accepted: 03/18/2020] [Indexed: 12/24/2022] Open
Abstract
Metastasis is the primary cause of cancer-related mortality. Cancer cells primarily metastasize via blood and lymphatic vessels to colonize lymph nodes and distant organs, leading to worse prognosis. Thus, strategies to limit blood and lymphatic spread of cancer have been a focal point of cancer research for several decades. Resistance to FDA-approved anti-angiogenic therapies designed to limit blood vessel growth has emerged as a significant clinical challenge. However, there are no FDA-approved drugs that target tumor lymphangiogenesis, despite the consequences of metastasis through the lymphatic system. This review highlights several of the key resistance mechanisms to anti-angiogenic therapy and potential challenges facing anti-lymphangiogenic therapy. Blood and lymphatic vessels are more than just conduits for nutrient, fluid, and cancer cell transport. Recent studies have elucidated how these vasculatures often regulate immune responses. Vessels that are abnormal or compromised by tumor cells can lead to immunosuppression. Therapies designed to improve lymphatic vessel function while limiting metastasis may represent a viable approach to enhance immunotherapy and limit cancer progression.
Collapse
|