Lymphatics in Tumor Progression and Immunomodulation
Abstract
:1. Introduction
2. Cellular Origins of Tumor-Associated Lymphatics
3. Lymphangiogenesis in the Primary Tumor
4. Lymphatics in Tumor Dissemination
5. Lymphatics and the Anti-Tumor Immune Response
6. Targeting Lymphatics for Cancer Therapeutics
7. Conclusions and Future Directions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Farnsworth, R.H.; Achen, M.G.; Stacker, S.A. The evolving role of lymphatics in cancer metastasis. Curr. Opin. Immunol. 2018, 53, 64–73. [Google Scholar] [CrossRef] [PubMed]
- Garnier, L.; Gkountidi, A.O.; Hugues, S. Tumor-Associated Lymphatic Vessel Features and Immunomodulatory Functions. Front. Immunol. 2019, 10, 720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baluk, P.; Fuxe, J.; Hashizume, H.; Romano, T.; Lashnits, E.; Butz, S.; Vestweber, D.; Corada, M.; Molendini, C.; Dejana, E.; et al. Functionally specialized junctions between endothelial cells of lymphatic vessels. J. Exp. Med. 2007, 204, 2349–2362. [Google Scholar] [CrossRef]
- Leak, L.V.; Burke, J.F. Fine structure of the lymphatic capillary and the adjoining connective tissue area. Am. J. Anat. 1966, 118, 785–809. [Google Scholar] [CrossRef] [PubMed]
- Muthuchamy, M.; Zawieja, D. Molecular regulation of lymphatic contractility. Ann. N. Y. Acad. Sci. 2008, 1131, 89–99. [Google Scholar] [CrossRef] [PubMed]
- Ma, Q.; Dieterich, L.C.; Detmar, M. Multiple roles of lymphatic vessels in tumor progression. Curr. Opin. Immunol. 2018, 53, 7–12. [Google Scholar] [CrossRef]
- Vaahtomeri, K.; Alitalo, K. Lymphatic vessels in tumor dissemination versus immunotherapy. Cancer Res. 2020, 80, 3463–3465. [Google Scholar] [CrossRef]
- Stachura, J.; Wachowska, M.; Kilarski, W.W.; Güç, E.; Golab, J.; Muchowicz, A. The dual role of tumor lymphatic vessels in dissemination of metastases and immune response development. Oncoimmunology 2016, 5, e1182278. [Google Scholar] [CrossRef] [Green Version]
- Alitalo, A.; Detmar, M. Interaction of tumor cells and lymphatic vessels in cancer progression. Oncogene 2012, 31, 4499–4508. [Google Scholar] [CrossRef] [Green Version]
- Harrell, M.I.; Iritani, B.M.; Ruddell, A. Tumor-Induced Sentinel Lymph Node Lymphangiogenesis and Increased Lymph Flow Precede Melanoma Metastasis. Am. J. Pathol. 2007, 170, 774–786. [Google Scholar] [CrossRef] [Green Version]
- Ma, Q.; Dieterich, L.C.; Ikenberg, K.; Bachmann, S.B.; Mangana, J.; Proulx, S.T.; Amann, V.C.; Levesque, M.P.; Dummer, R.; Baluk, P.; et al. Unexpected contribution of lymphatic vessels to promotion of distant metastatic tumor spread. Sci. Adv. 2018, 4, eaat4758. [Google Scholar] [CrossRef] [Green Version]
- Tewalt, E.F.; Cohen, J.N.; Rouhani, S.J.; Guidi, C.J.; Qiao, H.; Fahl, S.P.; Conaway, M.R.; Bender, T.P.; Tung, K.S.; Vella, A.T.; et al. Lymphatic endothelial cells induce tolerance via PD-L1 and lack of costimulation leading to high-level PD-1 expression on CD8 T cells. Blood 2012, 120, 4772–4782. [Google Scholar] [CrossRef]
- Rouhani, S.J.; Eccles, J.D.; Riccardi, P.; Peske, J.D.; Tewalt, E.F.; Cohen, J.N.; Liblau, R.; Makinen, T.; Engelhard, V.H. Roles of lymphatic endothelial cells expressing peripheral tissue antigens in CD4 T-cell tolerance induction. Nat. Commun. 2015, 6, 6771. [Google Scholar] [CrossRef] [Green Version]
- Lund, A.W.; Duraes, F.V.; Hirosue, S.; Raghavan, V.R.; Nembrini, C.; Thomas, S.N.; Issa, A.; Hugues, S.; Swartz, M.A. VEGF-C Promotes Immune Tolerance in B16 Melanomas and Cross-Presentation of Tumor Antigen by Lymph Node Lymphatics. Cell Rep. 2012, 1, 191–199. [Google Scholar] [CrossRef]
- Lane, R.S.; Femel, J.; Breazeale, A.P.; Loo, C.P.; Thibault, G.; Kaempf, A.; Mori, M.; Tsujikawa, T.; Chang, Y.H.; Lund, A.W. IFNγ-activated dermal lymphatic vessels inhibit cytotoxic T cells in melanoma and inflamed skin. J. Exp. Med. 2018, 215, 3057–3074. [Google Scholar] [CrossRef]
- Pullinger, B.D.; Florey, H.W. Proliferation of lymphatics in inflammation. J. Pathol. Bacteriol. 1937, 45, 157–170. [Google Scholar] [CrossRef]
- Gutierrez-Miranda, L.; Yaniv, K. Cellular Origins of the Lymphatic Endothelium: Implications for Cancer Lymphangiogenesis. Front. Physiol. 2020, 11, 577584. [Google Scholar] [CrossRef]
- He, Y.; Rajantie, I.; Ilmonen, M.; Makinen, T.; Karkkainen, M.J.; Haiko, P.; Salven, P.; Alitalo, K. Preexisting lymphatic endothelium but not endothelial progenitor cells are essential for tumor lymphangiogenesis and lymphatic metastasis. Cancer Res. 2004, 64, 3737–3740. [Google Scholar] [CrossRef] [Green Version]
- Religa, P.; Cao, R.; Bjorndahl, M.; Zhou, Z.; Zhu, Z.; Cao, Y. Presence of bone marrow–derived circulating progenitor endothelial cells in the newly formed lymphatic vessels. Blood 2005, 106, 4184–4190. [Google Scholar] [CrossRef] [Green Version]
- Jiang, S.; Bailey, A.S.; Goldman, D.C.; Swain, J.R.; Wong, M.H.; Streeter, P.R.; Fleming, W.H. Hematopoietic stem cells contribute to lymphatic endothelium. PLoS ONE 2008, 3, e3812. [Google Scholar] [CrossRef] [Green Version]
- Lee, J.Y.; Park, C.; Cho, Y.P.; Lee, E.; Kim, H.; Kim, P.; Yun, S.H.; Yoon, Y.S. Podoplanin-expressing cells derived from bone marrow play a crucial role in postnatal lymphatic neovascularization. Circulation 2010, 122, 1413–1425. [Google Scholar] [CrossRef]
- Zumsteg, A.; Baeriswyl, V.; Imaizumi, N.; Schwendener, R.; Rüegg, C.; Christofori, G. Myeloid cells contribute to tumor lymphangiogenesis. PLoS ONE 2009, 4, e7067. [Google Scholar] [CrossRef]
- Volk-Draper, L.; Patel, R.; Bhattarai, N.; Yang, J.; Wilber, A.; DeNardo, D.; Ran, S. Myeloid-Derived Lymphatic Endothelial Cell Progenitors Significantly Contribute to Lymphatic Metastasis in Clinical Breast Cancer. Am. J. Pathol. 2019, 189, 2269–2292. [Google Scholar] [CrossRef]
- Volk-Draper, L.D.; Hall, K.L.; Wilber, A.C.; Ran, S. Lymphatic endothelial progenitors originate from plastic myeloid cells activated by toll-like receptor-4. PLoS ONE 2017, 12, e0179257. [Google Scholar] [CrossRef]
- Gordon, E.J.; Rao, S.; Pollard, J.W.; Nutt, S.L.; Lang, R.A.; Harvey, N.L. Macrophages define dermal lymphatic vessel calibre during development by regulating lymphatic endothelial cell proliferation. Development 2010, 137, 3899–3910. [Google Scholar] [CrossRef] [Green Version]
- Beasley, N.J.P.; Prevo, R.; Banerji, S.; Leek, R.D.; Moore, J.; van Trappen, P.; Cox, G.; Harris, A.L.; Jackson, D.G. Intratumoral Lymphangiogenesis and Lymph Node Metastasis in Head and Neck Cancer. Cancer Res. 2002, 62, 1315–1320. [Google Scholar]
- Roma, A.A.; Magi-Galluzzi, C.; Kral, M.A.; Jin, T.T.; Klein, E.A.; Zhou, M. Peritumoral lymphatic invasion is associated with regional lymph node metastases in prostate adenocarcinoma. Mod. Pathol. 2006, 19, 392–398. [Google Scholar] [CrossRef] [Green Version]
- Thelen, A.; Scholz, A.; Benckert, C.; Weichert, W.; Dietz, E.; Wiedenmann, B.; Neuhaus, P.; Jonas, S. Tumor-Associated Lymphangiogenesis Correlates with Lymph Node Metastases and Prognosis in Hilar Cholangiocarcinoma. Ann. Surg. Oncol. 2008, 15, 791–799. [Google Scholar] [CrossRef]
- Gombos, Z.; Xu, X.; Chu, C.S.; Zhang, P.J.; Acs, G. Peritumoral Lymphatic Vessel Density and Vascular Endothelial Growth Factor C Expression in Early-Stage Squamous Cell Carcinoma of the Uterine Cervix. Clin. Cancer Res. 2005, 11, 8364–8371. [Google Scholar] [CrossRef] [Green Version]
- Dadras, S.S.; Lange-Asschenfeldt, B.; Velasco, P.; Nguyen, L.; Vora, A.; Muzikansky, A.; Jahnke, K.; Hauschild, A.; Hirakawa, S.; Mihm, M.C.; et al. Tumor lymphangiogenesis predicts melanoma metastasis to sentinel lymph nodes. Mod. Pathol. 2005, 18, 1232–1242. [Google Scholar] [CrossRef] [Green Version]
- Mlecnik, B.; Bindea, G.; Kirilovsky, A.; Angell, H.K.; Obenauf, A.C.; Tosolini, M.; Church, S.E.; Maby, P.; Vasaturo, A.; Angelova, M.; et al. The tumor microenvironment and Immunoscore are critical determinants of dissemination to distant metastasis. Sci. Transl. Med. 2016, 8, 327ra326. [Google Scholar] [CrossRef]
- Skobe, M.; Hawighorst, T.; Jackson, D.G.; Prevo, R.; Janes, L.; Velasco, P.; Riccardi, L.; Alitalo, K.; Claffey, K.; Detmar, M. Induction of tumor lymphangiogenesis by VEGF-C promotes breast cancer metastasis. Nat. Med. 2001, 7, 192–198. [Google Scholar] [CrossRef]
- Mandriota, S.J.; Jussila, L.; Jeltsch, M.; Compagni, A.; Baetens, D.; Prevo, R.; Banerji, S.; Huarte, J.; Montesano, R.; Jackson, D.G.; et al. Vascular endothelial growth factor-C-mediated lymphangiogenesis promotes tumour metastasis. EMBO J. 2001, 20, 672–682. [Google Scholar] [CrossRef] [Green Version]
- Karpanen, T.; Egeblad, M.; Karkkainen, M.J.; Kubo, H.; Ylä-Herttuala, S.; Jäättelä, M.; Alitalo, K. Vascular endothelial growth factor C promotes tumor lymphangiogenesis and intralymphatic tumor growth. Cancer Res. 2001, 61, 1786–1790. [Google Scholar]
- Lin, J.; Lalani, A.S.; Harding, T.C.; Gonzalez, M.; Wu, W.W.; Luan, B.; Tu, G.H.; Koprivnikar, K.; VanRoey, M.J.; He, Y.; et al. Inhibition of lymphogenous metastasis using adeno-associated virus-mediated gene transfer of a soluble VEGFR-3 decoy receptor. Cancer Res. 2005, 65, 6901–6909. [Google Scholar] [CrossRef] [Green Version]
- He, Y.; Rajantie, I.; Pajusola, K.; Jeltsch, M.; Holopainen, T.; Yla-Herttuala, S.; Harding, T.; Jooss, K.; Takahashi, T.; Alitalo, K. Vascular Endothelial Cell Growth Factor Receptor 3–Mediated Activation of Lymphatic Endothelium Is Crucial for Tumor Cell Entry and Spread via Lymphatic Vessels. Cancer Res. 2005, 65, 4739–4746. [Google Scholar] [CrossRef] [Green Version]
- Burton, J.B.; Priceman, S.J.; Sung, J.L.; Brakenhielm, E.; An, D.S.; Pytowski, B.; Alitalo, K.; Wu, L. Suppression of prostate cancer nodal and systemic metastasis by blockade of the lymphangiogenic axis. Cancer Res. 2008, 68, 7828–7837. [Google Scholar] [CrossRef] [Green Version]
- Lahdenranta, J.; Hagendoorn, J.; Padera, T.P.; Hoshida, T.; Nelson, G.; Kashiwagi, S.; Jain, R.K.; Fukumura, D. Endothelial nitric oxide synthase mediates lymphangiogenesis and lymphatic metastasis. Cancer Res. 2009, 69, 2801–2808. [Google Scholar] [CrossRef] [Green Version]
- Hoshida, T.; Isaka, N.; Hagendoorn, J.; di Tomaso, E.; Chen, Y.L.; Pytowski, B.; Fukumura, D.; Padera, T.P.; Jain, R.K. Imaging steps of lymphatic metastasis reveals that vascular endothelial growth factor-C increases metastasis by increasing delivery of cancer cells to lymph nodes: Therapeutic implications. Cancer Res. 2006, 66, 8065–8075. [Google Scholar] [CrossRef] [Green Version]
- Stacker, S.A.; Caesar, C.; Baldwin, M.E.; Thornton, G.E.; Williams, R.A.; Prevo, R.; Jackson, D.G.; Nishikawa, S.-I.; Kubo, H.; Achen, M.G. VEGF-D promotes the metastatic spread of tumor cells via the lymphatics. Nat. Med. 2001, 7, 186–191. [Google Scholar] [CrossRef]
- Hirakawa, S.; Kodama, S.; Kunstfeld, R.; Kajiya, K.; Brown, L.F.; Detmar, M. VEGF-A induces tumor and sentinel lymph node lymphangiogenesis and promotes lymphatic metastasis. J. Exp. Med. 2005, 201, 1089–1099. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cao, R.; Björndahl, M.A.; Religa, P.; Clasper, S.; Garvin, S.; Galter, D.; Meister, B.; Ikomi, F.; Tritsaris, K.; Dissing, S.; et al. PDGF-BB induces intratumoral lymphangiogenesis and promotes lymphatic metastasis. Cancer Cell 2004, 6, 333–345. [Google Scholar] [CrossRef] [Green Version]
- Holopainen, T.; Saharinen, P.; D’Amico, G.; Lampinen, A.; Eklund, L.; Sormunen, R.; Anisimov, A.; Zarkada, G.; Lohela, M.; Heloterä, H.; et al. Effects of Angiopoietin-2-Blocking Antibody on Endothelial Cell–Cell Junctions and Lung Metastasis. JNCI J. Natl. Cancer Inst. 2012, 104, 461–475. [Google Scholar] [CrossRef] [PubMed]
- Nagahashi, M.; Ramachandran, S.; Kim, E.Y.; Allegood, J.C.; Rashid, O.M.; Yamada, A.; Zhao, R.; Milstien, S.; Zhou, H.; Spiegel, S.; et al. Sphingosine-1-Phosphate Produced by Sphingosine Kinase 1 Promotes Breast Cancer Progression by Stimulating Angiogenesis and Lymphangiogenesis. Cancer Res. 2012, 72, 726–735. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Karpinich, N.O.; Kechele, D.O.; Espenschied, S.T.; Willcockson, H.H.; Fedoriw, Y.; Caron, K.M. Adrenomedullin gene dosage correlates with tumor and lymph node lymphangiogenesis. FASEB J. 2013, 27, 590–600. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jian, M.; Qingfu, Z.; Yanduo, J.; Guocheng, J.; Xueshan, Q. Anti-lymphangiogenesis effects of a specific anti-interleukin 7 receptor antibody in lung cancer model in vivo. Mol. Carcinog. 2015, 54, 148–155. [Google Scholar] [CrossRef]
- Chen, C.; Luo, Y.; He, W.; Zhao, Y.; Kong, Y.; Liu, H.; Zhong, G.; Li, Y.; Li, J.; Huang, J.; et al. Exosomal long noncoding RNA LNMAT2 promotes lymphatic metastasis in bladder cancer. J. Clin. Investig. 2020, 130, 404–421. [Google Scholar] [CrossRef]
- Chen, C.; Zheng, H.; Luo, Y.; Kong, Y.; An, M.; Li, Y.; He, W.; Gao, B.; Zhao, Y.; Huang, H.; et al. SUMOylation promotes extracellular vesicle–mediated transmission of lncRNA ELNAT1 and lymph node metastasis in bladder cancer. J. Clin. Investig. 2021, 131, e146431. [Google Scholar] [CrossRef]
- Zhou, C.-F.; Ma, J.; Huang, L.; Yi, H.-Y.; Zhang, Y.-M.; Wu, X.-G.; Yan, R.-M.; Liang, L.; Zhong, M.; Yu, Y.-H.; et al. Cervical squamous cell carcinoma-secreted exosomal miR-221-3p promotes lymphangiogenesis and lymphatic metastasis by targeting VASH1. Oncogene 2019, 38, 1256–1268. [Google Scholar] [CrossRef] [Green Version]
- Watari, K.; Shibata, T.; Kawahara, A.; Sata, K.; Nabeshima, H.; Shinoda, A.; Abe, H.; Azuma, K.; Murakami, Y.; Izumi, H.; et al. Tumor-derived interleukin-1 promotes lymphangiogenesis and lymph node metastasis through M2-type macrophages. PLoS ONE 2014, 9, e99568. [Google Scholar] [CrossRef]
- Karnezis, T.; Shayan, R.; Caesar, C.; Roufail, S.; Harris, N.C.; Ardipradja, K.; Zhang, Y.F.; Williams, S.P.; Farnsworth, R.H.; Chai, M.G.; et al. VEGF-D promotes tumor metastasis by regulating prostaglandins produced by the collecting lymphatic endothelium. Cancer Cell 2012, 21, 181–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, C.K.; Jeong, S.H.; Jang, C.; Bae, H.; Kim, Y.H.; Park, I.; Kim, S.K.; Koh, G.Y. Tumor metastasis to lymph nodes requires YAP-dependent metabolic adaptation. Science 2019, 363, 644–649. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cho, H.J.; Jung, J.I.; Lim, D.Y.; Kwon, G.T.; Her, S.; Park, J.H.; Park, J.H.Y. Bone marrow-derived, alternatively activated macrophages enhance solid tumor growth and lung metastasis of mammary carcinoma cells in a Balb/C mouse orthotopic model. Breast Cancer Res. 2012, 14, R81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.; Keskin, D.; Sugimoto, H.; Kanasaki, K.; Phillips, P.E.; Bizarro, L.; Sharpe, A.; LeBleu, V.S.; Kalluri, R. Podoplanin+ tumor lymphatics are rate limiting for breast cancer metastasis. PLoS Biol. 2018, 16, e2005907. [Google Scholar] [CrossRef]
- Kimura, T.; Sugaya, M.; Oka, T.; Blauvelt, A.; Okochi, H.; Sato, S. Lymphatic dysfunction attenuates tumor immunity through impaired antigen presentation. Oncotarget 2015, 6, 18081–18093. [Google Scholar] [CrossRef] [Green Version]
- Naxerova, K.; Reiter, J.G.; Brachtel, E.; Lennerz, J.K.; van de Wetering, M.; Rowan, A.; Cai, T.; Clevers, H.; Swanton, C.; Nowak, M.A.; et al. Origins of lymphatic and distant metastases in human colorectal cancer. Science 2017, 357, 55–60. [Google Scholar] [CrossRef] [Green Version]
- Hong, M.K.H.; Macintyre, G.; Wedge, D.C.; Van Loo, P.; Patel, K.; Lunke, S.; Alexandrov, L.B.; Sloggett, C.; Cmero, M.; Marass, F.; et al. Tracking the origins and drivers of subclonal metastatic expansion in prostate cancer. Nat. Commun. 2015, 6, 6605. [Google Scholar] [CrossRef]
- Deutsch, A.; Lubach, D.; Nissen, S.; Neukam, D. Ultrastructural Studies on the Invasion of Melanomas in Initial Lymphatics of Human Skin. J. Investig. Dermatol. 1992, 98, 64–67. [Google Scholar] [CrossRef] [Green Version]
- Padera, T.P.; Kadambi, A.; di Tomaso, E.; Carreira, C.M.; Brown, E.B.; Boucher, Y.; Choi, N.C.; Mathisen, D.; Wain, J.; Mark, E.J.; et al. Lymphatic Metastasis in the Absence of Functional Intratumor Lymphatics. Science 2002, 296, 1883–1886. [Google Scholar] [CrossRef]
- Wong, S.Y.; Haack, H.; Crowley, D.; Barry, M.; Bronson, R.T.; Hynes, R.O. Tumor-secreted vascular endothelial growth factor-C is necessary for prostate cancer lymphangiogenesis, but lymphangiogenesis is unnecessary for lymph node metastasis. Cancer Res. 2005, 65, 9789–9798. [Google Scholar] [CrossRef] [Green Version]
- Ubellacker, J.M.; Tasdogan, A.; Ramesh, V.; Shen, B.; Mitchell, E.C.; Martin-Sandoval, M.S.; Gu, Z.; McCormick, M.L.; Durham, A.B.; Spitz, D.R.; et al. Lymph protects metastasizing melanoma cells from ferroptosis. Nature 2020, 585, 113–118. [Google Scholar] [CrossRef] [PubMed]
- Proulx, S.T.; Luciani, P.; Derzsi, S.; Rinderknecht, M.; Mumprecht, V.; Leroux, J.-C.; Detmar, M. Quantitative Imaging of Lymphatic Function with Liposomal Indocyanine Green. Cancer Res. 2010, 70, 7053–7062. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Proulx, S.T.; Luciani, P.; Christiansen, A.; Karaman, S.; Blum, K.S.; Rinderknecht, M.; Leroux, J.-C.; Detmar, M. Use of a PEG-conjugated bright near-infrared dye for functional imaging of rerouting of tumor lymphatic drainage after sentinel lymph node metastasis. Biomaterials 2013, 34, 5128–5137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kawada, K.; Sonoshita, M.; Sakashita, H.; Takabayashi, A.; Yamaoka, Y.; Manabe, T.; Inaba, K.; Minato, N.; Oshima, M.; Taketo, M.M. Pivotal role of CXCR3 in melanoma cell metastasis to lymph nodes. Cancer Res. 2004, 64, 4010–4017. [Google Scholar] [CrossRef] [Green Version]
- Lee, E.; Fertig, E.J.; Jin, K.; Sukumar, S.; Pandey, N.B.; Popel, A.S. Breast cancer cells condition lymphatic endothelial cells within pre-metastatic niches to promote metastasis. Nat. Commun. 2014, 5, 4715. [Google Scholar] [CrossRef] [Green Version]
- Wiley, H.E.; Gonzalez, E.B.; Maki, W.; Wu, M.T.; Hwang, S.T. Expression of CC chemokine receptor-7 and regional lymph node metastasis of B16 murine melanoma. J. Natl. Cancer Inst. 2001, 93, 1638–1643. [Google Scholar] [CrossRef] [Green Version]
- Shields, J.D.; Emmett, M.S.; Dunn, D.B.; Joory, K.D.; Sage, L.M.; Rigby, H.; Mortimer, P.S.; Orlando, A.; Levick, J.R.; Bates, D.O. Chemokine-mediated migration of melanoma cells towards lymphatics-a mechanism contributing to metastasis. Oncogene 2007, 26, 2997–3005. [Google Scholar] [CrossRef] [Green Version]
- Das, S.; Sarrou, E.; Podgrabinska, S.; Cassella, M.; Mungamuri, S.K.; Feirt, N.; Gordon, R.; Nagi, C.S.; Wang, Y.; Entenberg, D.; et al. Tumor cell entry into the lymph node is controlled by CCL1 chemokine expressed by lymph node lymphatic sinuses. J. Exp. Med. 2013, 210, 1509–1528. [Google Scholar] [CrossRef] [Green Version]
- Hirakawa, S.; Detmar, M.; Kerjaschki, D.; Nagamatsu, S.; Matsuo, K.; Tanemura, A.; Kamata, N.; Higashikawa, K.; Okazaki, H.; Kameda, K.; et al. Nodal lymphangiogenesis and metastasis: Role of tumor-induced lymphatic vessel activation in extramammary Paget’s disease. Am. J. Pathol. 2009, 175, 2235–2248. [Google Scholar] [CrossRef] [Green Version]
- Hirakawa, S.; Brown, L.F.; Kodama, S.; Paavonen, K.; Alitalo, K.; Detmar, M. VEGF-C–induced lymphangiogenesis in sentinel lymph nodes promotes tumor metastasis to distant sites. Blood 2006, 109, 1010–1017. [Google Scholar] [CrossRef] [Green Version]
- Quagliata, L.; Klusmeier, S.; Cremers, N.; Pytowski, B.; Harvey, A.; Pettis, R.J.; Thiele, W.; Sleeman, J.P. Inhibition of VEGFR-3 activation in tumor-draining lymph nodes suppresses the outgrowth of lymph node metastases in the MT-450 syngeneic rat breast cancer model. Clin. Exp. Metastasis 2014, 31, 351–365. [Google Scholar] [CrossRef] [PubMed]
- Paget, S. The distribution of secondary growths in cancer of the breast. Lancet 1889, 133, 571–573. [Google Scholar] [CrossRef] [Green Version]
- Hood, J.L.; San, R.S.; Wickline, S.A. Exosomes released by melanoma cells prepare sentinel lymph nodes for tumor metastasis. Cancer Res. 2011, 71, 3792–3801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, B.; Zhou, Y.; Fang, Y.; Li, Z.; Gu, X.; Xiang, J. Colorectal cancer exosomes induce lymphatic network remodeling in lymph nodes. Int. J. Cancer 2019, 145, 1648–1659. [Google Scholar] [CrossRef] [PubMed]
- Olmeda, D.; Cerezo-Wallis, D.; Riveiro-Falkenbach, E.; Pennacchi, P.C.; Contreras-Alcalde, M.; Ibarz, N.; Cifdaloz, M.; Catena, X.; Calvo, T.G.; Cañón, E.; et al. Whole-body imaging of lymphovascular niches identifies pre-metastatic roles of midkine. Nature 2017, 546, 676–680. [Google Scholar] [CrossRef] [PubMed]
- Van den Eynden, G.G.; Vandenberghe, M.K.; van Dam, P.-J.H.; Colpaert, C.G.; van Dam, P.; Dirix, L.Y.; Vermeulen, P.B.; Van Marck, E.A. Increased Sentinel Lymph Node Lymphangiogenesis is Associated with Nonsentinel Axillary Lymph Node Involvement in Breast Cancer Patients with a Positive Sentinel Node. Clin. Cancer Res. 2007, 13, 5391–5397. [Google Scholar] [CrossRef] [Green Version]
- Pastushenko, I.; Van den Eynden, G.G.; Vicente-Arregui, S.; Prieto-Torres, L.; Alvarez-Alegret, R.; Querol, I.; Dirix, L.Y.; Carapeto, F.J.; Vermeulen, P.B.; Van Laere, S.J. Increased Angiogenesis and Lymphangiogenesis in Metastatic Sentinel Lymph Nodes Is Associated With Nonsentinel Lymph Node Involvement and Distant Metastasis in Patients With Melanoma. Am. J. Dermatopathol. 2016, 38, 338–346. [Google Scholar] [CrossRef]
- Brown, M.; Assen, F.P.; Leithner, A.; Abe, J.; Schachner, H.; Asfour, G.; Bago-Horvath, Z.; Stein, J.V.; Uhrin, P.; Sixt, M.; et al. Lymph node blood vessels provide exit routes for metastatic tumor cell dissemination in mice. Science 2018, 359, 1408–1411. [Google Scholar] [CrossRef] [Green Version]
- Pereira, E.R.; Kedrin, D.; Seano, G.; Gautier, O.; Meijer, E.F.J.; Jones, D.; Chin, S.-M.; Kitahara, S.; Bouta, E.M.; Chang, J.; et al. Lymph node metastases can invade local blood vessels, exit the node, and colonize distant organs in mice. Science 2018, 359, 1403–1407. [Google Scholar] [CrossRef] [Green Version]
- Roberts, E.W.; Broz, M.L.; Binnewies, M.; Headley, M.B.; Nelson, A.E.; Wolf, D.M.; Kaisho, T.; Bogunovic, D.; Bhardwaj, N.; Krummel, M.F. Critical role for CD103+/CD141+ dendritic cells bearing CCR7 for tumor antigen trafficking and priming of T cell immunity in melanoma. Cancer Cell 2016, 30, 324–336. [Google Scholar] [CrossRef] [Green Version]
- Lund, A.W.; Wagner, M.; Fankhauser, M.; Steinskog, E.S.; Broggi, M.A.; Spranger, S.; Gajewski, T.F.; Alitalo, K.; Eikesdal, H.P.; Wiig, H.; et al. Lymphatic vessels regulate immune microenvironments in human and murine melanoma. J. Clin. Investig. 2016, 126, 3389–3402. [Google Scholar] [CrossRef] [PubMed]
- Kataru, R.P.; Ly, C.L.; Shin, J.; Park, H.J.; Baik, J.E.; Rehal, S.; Ortega, S.; Lyden, D.; Mehrara, B.J. Tumor lymphatic function regulates tumor inflammatory and immunosuppressive microenvironments. Cancer Immunol. Res. 2019, 7, 1345–1358. [Google Scholar] [CrossRef] [PubMed]
- Hatzioannou, A.; Nayar, S.; Gaitanis, A.; Barone, F.; Anagnostopoulos, C.; Verginis, P. Intratumoral accumulation of podoplanin-expressing lymph node stromal cells promote tumor growth through elimination of CD4+ tumor-infiltrating lymphocytes. OncoImmunology 2016, 5, e1216289. [Google Scholar] [CrossRef] [Green Version]
- Cousin, N.; Cap, S.; Dihr, M.; Tacconi, C.; Detmar, M.; Dieterich, L.C. Lymphatic PD-L1 Expression Restricts Tumor-Specific CD8+ T-cell Responses. Cancer Res. 2021, 81, 4133. [Google Scholar] [CrossRef]
- Dieterich, L.C.; Ikenberg, K.; Cetintas, T.; Kapaklikaya, K.; Hutmacher, C.; Detmar, M. Tumor-Associated Lymphatic Vessels Upregulate PDL1 to Inhibit T-Cell Activation. Front. Immunol. 2017, 8, 66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gkountidi, A.O.; Garnier, L.; Dubrot, J.; Angelillo, J.; Harlé, G.; Brighouse, D.; Wrobel, L.J.; Pick, R.; Scheiermann, C.; Swartz, M.A.; et al. MHC Class II Antigen Presentation by Lymphatic Endothelial Cells in Tumors Promotes Intratumoral Regulatory T cell-Suppressive Functions. Cancer Immunol. Res. 2021, 9, 748–764. [Google Scholar] [CrossRef] [PubMed]
- Podgrabinska, S.; Kamalu, O.; Mayer, L.; Shimaoka, M.; Snoeck, H.; Randolph, G.J.; Skobe, M. Inflamed lymphatic endothelium suppresses dendritic cell maturation and function via Mac-1/ICAM-1-dependent mechanism. J. Immunol. 2009, 183, 1767–1779. [Google Scholar] [CrossRef]
- Nörder, M.; Gutierrez, M.G.; Zicari, S.; Cervi, E.; Caruso, A.; Guzmán, C.A. Lymph node-derived lymphatic endothelial cells express functional costimulatory molecules and impair dendritic cell-induced allogenic T-cell proliferation. FASEB J. 2012, 26, 2835–2846. [Google Scholar] [CrossRef]
- He, Y.; Kozaki, K.; Karpanen, T.; Koshikawa, K.; Yla-Herttuala, S.; Takahashi, T.; Alitalo, K. Suppression of tumor lymphangiogenesis and lymph node metastasis by blocking vascular endothelial growth factor receptor 3 signaling. J. Natl. Cancer Inst. 2002, 94, 819–825. [Google Scholar] [CrossRef] [Green Version]
- Krishnan, J.; Kirkin, V.; Steffen, A.; Hegen, M.; Weih, D.; Tomarev, S.; Wilting, J.; Sleeman, J.P. Differential Expression of Vascular Endothelial Growth Factor (VEGF)-C and VEGF-D in Tumors and Its Relationship to Lymphatic Metastasis in Immunocompetent Rats. Cancer Res. 2003, 63, 713–722. [Google Scholar]
- Laakkonen, P.; Waltari, M.; Holopainen, T.; Takahashi, T.; Pytowski, B.; Steiner, P.; Hicklin, D.; Persaud, K.; Tonra, J.R.; Witte, L.; et al. Vascular Endothelial Growth Factor Receptor 3 Is Involved in Tumor Angiogenesis and Growth. Cancer Res. 2007, 67, 593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Roberts, N.; Kloos, B.; Cassella, M.; Podgrabinska, S.; Persaud, K.; Wu, Y.; Pytowski, B.; Skobe, M. Inhibition of VEGFR-3 Activation with the Antagonistic Antibody More Potently Suppresses Lymph Node and Distant Metastases than Inactivation of VEGFR-2. Cancer Res. 2006, 66, 2650. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shimizu, K.; Kubo, H.; Yamaguchi, K.; Kawashima, K.; Ueda, Y.; Matsuo, K.; Awane, M.; Shimahara, Y.; Takabayashi, A.; Yamaoka, Y.; et al. Suppression of VEGFR-3 signaling inhibits lymph node metastasis in gastric cancer. Cancer Sci. 2004, 95, 328–333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Saif, M.W.; Knost, J.A.; Chiorean, E.G.; Kambhampati, S.R.; Yu, D.; Pytowski, B.; Qin, A.; Kauh, J.S.; O’Neil, B.H. Phase 1 study of the anti-vascular endothelial growth factor receptor 3 monoclonal antibody LY3022856/IMC-3C5 in patients with advanced and refractory solid tumors and advanced colorectal cancer. Cancer Chemother. Pharm. 2016, 78, 815–824. [Google Scholar] [CrossRef] [PubMed]
- Qin, S.; Li, A.; Yi, M.; Yu, S.; Zhang, M.; Wu, K. Recent advances on anti-angiogenesis receptor tyrosine kinase inhibitors in cancer therapy. J. Hematol. Oncol. 2019, 12, 27. [Google Scholar] [CrossRef] [Green Version]
- Alam, A.; Blanc, I.; Gueguen-Dorbes, G.; Duclos, O.; Bonnin, J.; Barron, P.; Laplace, M.-C.; Morin, G.; Gaujarengues, F.; Dol, F.; et al. SAR131675, a Potent and Selective VEGFR-3–TK Inhibitor with Antilymphangiogenic, Antitumoral, and Antimetastatic Activities. Mol. Cancer Ther. 2012, 11, 1637. [Google Scholar] [CrossRef] [Green Version]
- Fankhauser, M.; Broggi, M.A.S.; Potin, L.; Bordry, N.; Jeanbart, L.; Lund, A.W.; Da Costa, E.; Hauert, S.; Rincon-Restrepo, M.; Tremblay, C.; et al. Tumor lymphangiogenesis promotes T cell infiltration and potentiates immunotherapy in melanoma. Sci. Transl. Med. 2017, 9, eaal4712. [Google Scholar] [CrossRef] [Green Version]
- Hu, X.; Deng, Q.; Ma, L.; Li, Q.; Chen, Y.; Liao, Y.; Zhou, F.; Zhang, C.; Shao, L.; Feng, J.; et al. Meningeal lymphatic vessels regulate brain tumor drainage and immunity. Cell Res. 2020, 30, 229–243. [Google Scholar] [CrossRef] [Green Version]
- Song, E.; Mao, T.; Dong, H.; Boisserand, L.S.B.; Antila, S.; Bosenberg, M.; Alitalo, K.; Thomas, J.-L.; Iwasaki, A. VEGF-C-driven lymphatic drainage enables immunosurveillance of brain tumours. Nature 2020, 577, 689–694. [Google Scholar] [CrossRef]
- Sasso, M.S.; Mitrousis, N.; Wang, Y.; Briquez, P.S.; Hauert, S.; Ishihara, J.; Hubbell, J.A.; Swartz, M.A. Lymphangiogenesis-inducing vaccines elicit potent and long-lasting T cell immunity against melanomas. Sci. Adv. 2021, 7, eabe4362. [Google Scholar] [CrossRef]
- Commerford, C.D.; Dieterich, L.C.; He, Y.; Hell, T.; Montoya-Zegarra, J.A.; Noerrelykke, S.F.; Russo, E.; Röcken, M.; Detmar, M. Mechanisms of Tumor-Induced Lymphovascular Niche Formation in Draining Lymph Nodes. Cell Rep. 2018, 25, 3554–3563.e3554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Melia, M.J.; Manspeaker, M.P.; Thomas, S.N. Tumor-draining lymph nodes are survival niches that support T cell priming against lymphatic transported tumor antigen and effects of immune checkpoint blockade in TNBC. Cancer Immunol. Immunother. 2021, 70, 2179–2195. [Google Scholar] [CrossRef] [PubMed]
Molecule | Target cell | Target Pathway | Function Tumor | Origin | Refs |
---|---|---|---|---|---|
Cytokines | |||||
IL-1 | MФ | IKKβ/NF-кB | Recuitment and activation of lymphangiogenic M2 MФ | Lung | [36] |
IL-6 | LECs | pStat3-pcJun-pATF-2 ternary complex | Increased lymphatic chemctactic cues, increased lung vascular permeability, increased LN angiogenesis | Breast | [50] |
IL-7 | Tumor ; | cFos and c-Jun heterodimer | Increased tumor lymphangiogenic | Lung | [32] |
Grouth factors | |||||
VEGF-A | LECs | - | Increased tumor lymphangiogenic, increased LN angiogenesis | Cutaneous SCC | [27] |
VEGF-C | LECs | eNOS | Increased tumor lymphangiogenic, increased LN angiogenesis | Breast, pancreas, prostate, lung, melanoma, fibrosarcoma | [18,19,20,21,22,23,24] |
VEGF-D | LECs | Prostaglandin production | Increased tumor lymphangiogenesis, dilation of collecting efferent lymphatic vessel | Breast | [26,46] |
PDGF-BB | LECs | MAP Kinases Erk1/2 and Akt | Increased tumor lymphangiogenic | fibrosarcoma | [28] |
Exosome-derived | |||||
Long noncoding RNALINC00858 | LECs | Prax-1 transcription | Increased tumor lymphangiogenic | Bladder | [33] |
Long noncoding RNASNHG16 | LECs | SOX18 transcription | Increased tumor lymphangiogenic | Bladder | [34] |
miRNA-221-3p | LECs | VASHI/ERK/AKT | Increased tumor lymphangiogenic | Cervical SCC | [35] |
IRF-2 M | MФ | - | Increased LN angiogenesis through MФ secreted VEGF-C | Colorectal carcinoma | [51] |
Other | |||||
Adrenomedullin | LECs | - | Increased tumor lymphangiogenic | Lung | [31] |
LECs | mTOR | Increased tumor lymphangiogenic, increased LN angiogenesis, increased distant organ lymphangiogenesis | Melanoma | [52] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Li, C.Y.; Brown, S.; Mehrara, B.J.; Kataru, R.P. Lymphatics in Tumor Progression and Immunomodulation. Int. J. Mol. Sci. 2022, 23, 2127. https://doi.org/10.3390/ijms23042127
Li CY, Brown S, Mehrara BJ, Kataru RP. Lymphatics in Tumor Progression and Immunomodulation. International Journal of Molecular Sciences. 2022; 23(4):2127. https://doi.org/10.3390/ijms23042127
Chicago/Turabian StyleLi, Claire Y., Stav Brown, Babak J. Mehrara, and Raghu P. Kataru. 2022. "Lymphatics in Tumor Progression and Immunomodulation" International Journal of Molecular Sciences 23, no. 4: 2127. https://doi.org/10.3390/ijms23042127
APA StyleLi, C. Y., Brown, S., Mehrara, B. J., & Kataru, R. P. (2022). Lymphatics in Tumor Progression and Immunomodulation. International Journal of Molecular Sciences, 23(4), 2127. https://doi.org/10.3390/ijms23042127