Brain Vascular Microenvironments in Cancer Metastasis
Abstract
:1. Introduction
2. Central Nervous System Vasculature and Anatomy
2.1. CNS Anatomy and Blood Supply
2.2. Arteriovenous Zonation
2.3. CNS Drainage
3. The Brain Metastatic Cascade and Tumour Microenvironment
3.1. Clinical Presentation
3.2. The Brain Pre-Metastatic Niche and Organotropism
3.3. Metastatic Colonisation of the Brain
3.4. Tumour Drainage
4. Future Research and Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Lambert, A.W.; Pattabiraman, D.R.; Weinberg, R.A. Emerging Biological Principles of Metastasis. Cell 2017, 168, 670–691. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lowery, F.J.; Yu, D. Brain metastasis: Unique challenges and open opportunities. BBA Rev. Cancer 2017, 1867, 49–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eichler, A.F.; Chung, E.; Kodack, D.P.; Loeffler, J.S.; Fukumura, D.; Jain, R.K. The biology of brain metastases-translation to new therapies. Nat. Rev. Clin. Oncol. 2011, 8, 344–356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ostrom, Q.T.; Wright, C.H.; Barnholtz-Sloan, J.S. Brain metastases: Epidemiology. Handb. Clin. Neurol. 2018, 149, 27–42. [Google Scholar] [CrossRef] [PubMed]
- Cagney, D.N.; Martin, A.M.; Catalano, P.J.; Redig, A.J.; Lin, N.U.; Lee, E.Q.; Wen, P.Y.; Dunn, I.F.; Bi, W.L.; Weiss, S.E.; et al. Incidence and prognosis of patients with brain metastases at diagnosis of systemic malignancy: A population-based study. Neuro-Oncol. 2017, 19, 1511–1521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef] [PubMed]
- Pardridge, W.M. The Blood-Brain Barrier: Bottleneck in Brain Drug Development. NeuroRX 2005, 2, 3–14. [Google Scholar] [CrossRef]
- Carvalho, R.; Paredes, J.; Ribeiro, A.S. Impact of breast cancer cells secretome on the brain metastatic niche remodeling. Semin. Cancer Biol. 2020, 60, 294–301. [Google Scholar] [CrossRef]
- Custodio-Santos, T.; Videira, M.; Brito, M.A. Brain metastasization of breast cancer. Biochim. Biophys. Acta Rev. Cancer 2017, 1868, 132–147. [Google Scholar] [CrossRef]
- Peinado, H.; Zhang, H.; Matei, I.R.; Costa-Silva, B.; Hoshino, A.; Rodrigues, G.; Psaila, B.; Kaplan, R.N.; Bromberg, J.F.; Kang, Y.; et al. Pre-metastatic niches: Organ-specific homes for metastases. Nat. Rev. Cancer 2017, 17, 302–317. [Google Scholar] [CrossRef]
- Rodrigues, G.; Hoshino, A.; Kenific, C.M.; Matei, I.R.; Steiner, L.; Freitas, D.; Kim, H.S.; Oxley, P.R.; Scandariato, I.; Casanova-Salas, I.; et al. Tumour exosomal CEMIP protein promotes cancer cell colonization in brain metastasis. Nat. Cell Biol. 2019, 21, 1403–1412. [Google Scholar] [CrossRef]
- Lorger, M. Tumor microenvironment in the brain. Cancers 2012, 4, 218–243. [Google Scholar] [CrossRef]
- Quail, D.F.; Joyce, J.A. The Microenvironmental Landscape of Brain Tumors. Cancer Cell 2017, 31, 326–341. [Google Scholar] [CrossRef] [Green Version]
- Ferrer, I.; Vidal, N. Neuropathology of cerebrovascular diseases. Handb. Clin. Neurol. 2017, 145, 79–114. [Google Scholar] [CrossRef]
- Hendrikse, J.; Hartkamp, M.J.; Hillen, B.; Mali, W.P.T.M.; Grond, J.V.D. Collateral Ability of the Circle of Willis in Patients with Unilateral Internal Carotid Artery Occlusion: Border Zone Infarcts and Clinical Symptoms. Stroke J. Am. Heart Assoc. 2001, 32, 2768–2773. [Google Scholar] [CrossRef]
- Furtado, D.; Bjornmalm, M.; Ayton, S.; Bush, A.I.; Kempe, K.; Caruso, F. Overcoming the Blood-Brain Barrier: The Role of Nanomaterials in Treating Neurological Diseases. Adv. Mater. 2018, 30, e1801362. [Google Scholar] [CrossRef] [Green Version]
- Mastorakos, P.; McGavern, D. The anatomy and immunology of vasculature in the central nervous system. Sci. Immunol. 2019, 4, eaav0492. [Google Scholar] [CrossRef]
- Nakada, T.; Kwee, I.L. Fluid Dynamics Inside the Brain Barrier: Current Concept of Interstitial Flow, Glymphatic Flow, and Cerebrospinal Fluid Circulation in the Brain. Neuroscientist 2019, 25, 155. [Google Scholar] [CrossRef] [Green Version]
- Bonasia, S.; Smajda, S.; Ciccio, G.; Robert, T. Middle Meningeal Artery: Anatomy and Variations. AJNR Am. J. Neuroradiol. 2020, 41, 1777–1785. [Google Scholar] [CrossRef]
- Castelli, W.A.; Huelke, D.F. The arterial supply of the dura mater of the Rhesus monkey. Anat. Rec. 1965, 152, 155–160. [Google Scholar] [CrossRef] [Green Version]
- Alcolado, R.; Weller, R.O.; Parrish, E.P.; Garrod, D. The cranial arachnoid and pia mater in man: Anatomical and ultrastructural observations. Neuropathol. Appl. Neurobiol. 1988, 14, 1–17. [Google Scholar] [CrossRef]
- Da Mesquita, S.; Fu, Z.; Kipnis, J. The Meningeal Lymphatic System: A New Player in Neurophysiology. Neuron 2018, 100, 375–388. [Google Scholar] [CrossRef] [Green Version]
- Engelhardt, B.; Carare, R.O.; Bechmann, I.; Flugel, A.; Laman, J.D.; Weller, R.O. Vascular, glial, and lymphatic immune gateways of the central nervous system. Acta Neuropathol. 2016, 132, 317–338. [Google Scholar] [CrossRef] [Green Version]
- Rasmussen, M.K.; Mestre, H.; Nedergaard, M. The glymphatic pathway in neurological disorders. Lancet Neurol. 2018, 17, 1016–1024. [Google Scholar] [CrossRef] [Green Version]
- Halpern, K.B.; Shenhav, R.; Matcovitch-Natan, O.; Toth, B.; Lemze, D.; Golan, M.; Massasa, E.E.; Baydatch, S.; Landen, S.; Moor, A.E.; et al. Single-cell spatial reconstruction reveals global division of labour in the mammalian liver. Nature 2017, 542, 352–356. [Google Scholar] [CrossRef]
- Corada, M.; Morini, M.F.; Dejana, E. Signaling pathways in the specification of arteries and veins. Arter. Thromb. Vasc. Biol. 2014, 34, 2372–2377. [Google Scholar] [CrossRef]
- Fang, J.; Hirschi, K. Molecular regulation of arteriovenous endothelial cell specification. F1000Research 2019, 8, 1208. [Google Scholar] [CrossRef] [Green Version]
- Vanlandewijck, M.; He, L.; Mae, M.A.; Andrae, J.; Ando, K.; del Gaudio, F.; Nahar, K.; Lebouvier, T.; Lavina, B.; Gouveia, L.; et al. A molecular atlas of cell types and zonation in the brain vasculature. Nature 2018, 554, 475–480. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Da Mesquita, S.; Papadopoulos, Z.; Dykstra, T.; Brase, L.; Farias, F.G.; Wall, M.; Jiang, H.; Kodira, C.D.; de Lima, K.A.; Herz, J.; et al. Meningeal lymphatics affect microglia responses and anti-Aβ immunotherapy. Nature 2021, 593, 255–260. [Google Scholar] [CrossRef] [PubMed]
- Rustenhoven, J.; Drieu, A.; Mamuladze, T.; de Lima, K.A.; Dykstra, T.; Wall, M.; Papadopoulos, Z.; Kanamori, M.; Salvador, A.F.; Baker, W.; et al. Functional characterization of the dural sinuses as a neuroimmune interface. Cell 2021, 184, 1000–1016.e27. [Google Scholar] [CrossRef] [PubMed]
- Kalucka, J.; de Rooij, L.; Goveia, J.; Rohlenova, K.; Dumas, S.J.; Meta, E.; Conchinha, N.V.; Taverna, F.; Teuwen, L.A.; Veys, K.; et al. Single-Cell Transcriptome Atlas of Murine Endothelial Cells. Cell 2020, 180, 764–779.e20. [Google Scholar] [CrossRef]
- Damkier, H.H.; Brown, P.D.; Praetorius, J. Cerebrospinal fluid secretion by the choroid plexus. Physiol. Rev. 2013, 93, 1847–1892. [Google Scholar] [CrossRef] [Green Version]
- Yu, J.; Xu, N.; Zhao, Y.; Yu, J. Clinical importance of the anterior choroidal artery: A review of the literature. Int. J. Med. Sci. 2018, 15, 368–375. [Google Scholar] [CrossRef] [Green Version]
- Brinker, T.; Stopa, E.; Morrison, J.; Klinge, P. A new look at cerebrospinal fluid circulation. Fluids Barriers CNS 2014, 11, 1–32. [Google Scholar] [CrossRef] [Green Version]
- Rudie, J.D.; Rauschecker, A.M.; Nabavizadeh, S.A.; Mohan, S. Neuroimaging of Dilated Perivascular Spaces: From Benign and Pathologic Causes to Mimics. J. Neuroimaging 2018, 28, 139–149. [Google Scholar] [CrossRef]
- Jessen, N.A.; Munk, A.S.; Lundgaard, I.; Nedergaard, M. The Glymphatic System: A Beginner’s Guide. Neurochem. Res. 2015, 40, 2583–2599. [Google Scholar] [CrossRef] [Green Version]
- Guo, X.B.; Wei, S.; Guan, S. Intracranial Venous Pressures Manometry for Patients with Idiopathic Intracranial Hypertension: Under Awake Setting or General Anesthesia. Front. Neurol. 2019, 10, 751. [Google Scholar] [CrossRef] [Green Version]
- Shulman, K.; Yarnell, P.; Ransohoff, J. Dural Sinus Pressure: In Normal and Hydrocephalic Dogs. Arch. Neurol. 1964, 10, 575–580. [Google Scholar] [CrossRef]
- Tsutsumi, S.; Ono, H.; Ishii, H. Inferior sagittal sinus: Magnetic resonance imaging study. Surg. Radiol. Anat. 2021, 43, 1353–1357. [Google Scholar] [CrossRef]
- Tsutsumi, S.; Ono, H.; Ishii, H. Arachnoid granulations bulging into the transverse sinus, sigmoid sinus, straight sinus, and confluens sinuum: A magnetic resonance imaging study. Surg. Radiol. Anat. 2021, 43, 1311–1318. [Google Scholar] [CrossRef]
- Doepp, F.; Schreiber, S.J.; von Munster, T.; Rademacher, J.; Klingebiel, R.; Valdueza, J.M. How does the blood leave the brain? A systematic ultrasound analysis of cerebral venous drainage patterns. Neuroradiology 2004, 46, 565–570. [Google Scholar] [CrossRef] [PubMed]
- Louveau, A.; Smirnov, I.; Keyes, T.J.; Eccles, J.D.; Rouhani, S.J.; Peske, J.D.; Derecki, N.C.; Castle, D.; Mandell, J.W.; Lee, K.S.; et al. Structural and functional features of central nervous system lymphatic vessels. Nature 2015, 523, 337–341. [Google Scholar] [CrossRef] [PubMed]
- Aspelund, A.; Antila, S.; Proulx, S.T.; Karlsen, T.V.; Karaman, S.; Detmar, M.; Wiig, H.; Alitalo, K. A dural lymphatic vascular system that drains brain interstitial fluid and macromolecules. J. Exp. Med. 2015, 212, 991–999. [Google Scholar] [CrossRef] [PubMed]
- Louveau, A.; Herz, J.; Alme, M.N.; Salvador, A.F.; Dong, M.Q.; Viar, K.E.; Herod, S.G.; Knopp, J.; Setliff, J.C.; Lupi, A.L.; et al. CNS lymphatic drainage and neuroinflammation are regulated by meningeal lymphatic vasculature. Nat. Neurosci. 2018, 21, 1380–1391. [Google Scholar] [CrossRef]
- Ma, Q.; Ineichen, B.V.; Detmar, M.; Proulx, S.T. Outflow of cerebrospinal fluid is predominantly through lymphatic vessels and is reduced in aged mice. Nat. Commun. 2017, 8, 1434. [Google Scholar] [CrossRef] [Green Version]
- Paget, S. The distribution of secondary growths in cancer of the breast. 1889. Cancer Metastasis Rev. 1989, 8, 98–101. [Google Scholar]
- Filipa, M.; Katia, L.; Filipa, P.; Nadine, S.; Nuno, B.; Luísa, P.; Francisco, G. Bone metastases: An overview. Oncol. Rev. 2017, 11, 321. [Google Scholar] [CrossRef]
- Frinton, E.; Tong, D.; Tan, J.; Read, G.; Kumar, V.; Kennedy, S.; Lim, C.; Board, R.E. Metastatic melanoma: Prognostic factors and survival in patients with brain metastases. J. Neurooncol. 2017, 135, 507–512. [Google Scholar] [CrossRef] [Green Version]
- Villanueva-Meyer, J.E.; Mabray, M.C.; Cha, S. Current Clinical Brain Tumor Imaging. Neurosurgery 2017, 81, 397–415. [Google Scholar] [CrossRef] [Green Version]
- Patchell, R.A. The management of brain metastases. Cancer Treat. Rev. 2003, 29, 533–540. [Google Scholar] [CrossRef]
- Hwang, T.L.; Close, T.P.; Grego, J.M.; Brannon, W.L.; Gonzales, L. Predilection of Brain Metastasis in Gray and White Matter Junction and Vascular Border Zones. Interdiscip. Int. J. Am. Cancer Soc. 1996, 77, 1551–1555. [Google Scholar] [CrossRef]
- Suh, J.H.; Kotecha, R.; Chao, S.T.; Ahluwalia, M.S.; Sahgal, A.; Chang, E.L. Current approaches to the management of brain metastases. Nat. Rev. Clin. Oncol. 2020, 17, 279–299. [Google Scholar] [CrossRef]
- Delattre, J.Y.; Krol, G.; Thaler, H.T.; Posner, J.B. Distribution of Brain Metastases. Arch. Neurol. 1988, 45, 741–744. [Google Scholar] [CrossRef]
- Lasocki, A.; Khoo, C.; Lau, P.K.H.; Kok, D.L.; McArthur, G.A. High-resolution MRI demonstrates that more than 90% of small intracranial melanoma metastases develop in close relationship to the leptomeninges. Neuro-Oncol. 2020, 22, 423–432. [Google Scholar] [CrossRef]
- Bachmann, C.; Schmidt, S.; Staebler, A.; Fehm, T.; Fend, F.; Schittenhelm, J.; Wallwiener, D.; Grischke, E. CNS metastases in breast cancer patients: Prognostic implications of tumor subtype. Med. Oncol. 2015, 32, 400. [Google Scholar] [CrossRef]
- Berghoff, A.S.; Preusser, M. Anti-angiogenic therapies in brain metastases. memo Mag. Eur. Med. Oncol. 2018, 11, 14–17. [Google Scholar] [CrossRef] [Green Version]
- Glitza Oliva, I.; Tawbi, H.; Davies, M.A. Melanoma Brain Metastases: Current Areas of Investigation and Future Directions. Cancer J. 2017, 23, 68–74. [Google Scholar] [CrossRef] [Green Version]
- Ramanujam, S.; Schadendorf, D.; Long, G.V. Systemic therapies for melanoma brain metastases: Which drug for whom and when? Chin. Clin. Oncol. 2015, 4, 25. [Google Scholar] [CrossRef]
- Robey, R.W.; Pluchino, K.M.; Hall, M.D.; Fojo, A.T.; Bates, S.E.; Gottesman, M.M. Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat. Rev. Cancer 2018, 18, 452–464. [Google Scholar] [CrossRef]
- Schinkel, A.H.; Smit, J.J.M.; van Tellingen, O.; Beijnen, J.H.; Wagenaar, E.; van Deemter, L.; Mol, C.A.A.M.; van der Valk, T.A.; Robanus-Maandag, E.C.; te Riele, H.P.J.; et al. Disruption of the mouse mdr1a P-glycoprotein gene leads to a deficiency in the blood-brain barrier and to increased sensitivity to drugs. Cell 1994, 77, 491. [Google Scholar] [CrossRef]
- Löscher, W.; Potschka, H. Blood-Brain Barrier Active Efflux Transporters: ATP-Binding Cassette Gene Family. NeuroRX 2005, 2, 86–98. [Google Scholar] [CrossRef]
- Amin, S.; Baine, M.J.; Meza, J.L.; Lin, C. Association of Immunotherapy with Survival Among Patients with Brain Metastases Whose Cancer Was Managed with Definitive Surgery of the Primary Tumor. JAMA Netw. Open 2020, 3, e2015444. [Google Scholar] [CrossRef]
- Liu, Q.; Tong, X.; Wang, J. Management of brain metastases: History and the present. Chin. Neurosurg. J. 2019, 5, 1. [Google Scholar] [CrossRef]
- De Gooijer, M.C.; de Vries, N.A.; Buckle, T.; Buil, L.C.M.; Beijnen, J.H.; Boogerd, W.; van Tellingen, O. Improved Brain Penetration and Antitumor Efficacy of Temozolomide by Inhibition of ABCB1 and ABCG2. Neoplasia 2018, 20, 710–720. [Google Scholar] [CrossRef]
- Ferraris, C.; Cavalli, R.; Panciani, P.P.; Battaglia, L. Overcoming the Blood-Brain Barrier: Successes and Challenges in Developing Nanoparticle-Mediated Drug Delivery Systems for the Treatment of Brain Tumours. Int. J. Nanomed. 2020, 15, 2999–3022. [Google Scholar] [CrossRef]
- Long, G.V.; Atkinson, V.; Lo, S.; Sandhu, S.; Guminski, A.D.; Brown, M.P.; Wilmott, J.S.; Edwards, J.; Gonzalez, M.; Scolyer, R.A.; et al. Combination nivolumab and ipilimumab or nivolumab alone in melanoma brain metastases: A multicentre randomised phase 2 study. Lancet Oncol. 2018, 19, 672–681. [Google Scholar] [CrossRef]
- Tawbi, H.A.; Forsyth, P.A.; Algazi, A.; Hamid, O.; Hodi, F.S.; Moschos, S.J.; Khushalani, N.I.; Lewis, K.; Lao, C.D.; Postow, M.A.; et al. Combined Nivolumab and Ipilimumab in Melanoma Metastatic to the Brain. N. Engl. J. Med. 2018, 379, 722–730. [Google Scholar] [CrossRef]
- Pardoll, D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat. Rev. Cancer 2012, 12, 252–264. [Google Scholar] [CrossRef] [Green Version]
- Aquilanti, E.; Brastianos, P.K. Immune Checkpoint Inhibitors for Brain Metastases: A Primer for Neurosurgeons. Neurosurgery 2020, 87, E281–E288. [Google Scholar] [CrossRef]
- Buchbinder, E.I.; Desai, A. CTLA-4 and PD-1 Pathways: Similarities, Differences, and Implications of Their Inhibition. Am. J. Clin. Oncol. 2016, 39, 98–106. [Google Scholar] [CrossRef] [Green Version]
- Yshii, L.M.; Hohlfeld, R.; Liblau, R.S. Inflammatory CNS disease caused by immune checkpoint inhibitors: Status and perspectives. Nat. Rev. Neurol. 2017, 13, 755–763. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Achrol, A.S.; Rennert, R.C.; Anders, C.; Soffietti, R.; Ahluwalia, M.S.; Nayak, L.; Peters, S.; Arvold, N.D.; Harsh, G.R.; Steeg, P.S.; et al. Brain metastases. Nat. Rev. Dis. Primers 2019, 5, 5. [Google Scholar] [CrossRef] [PubMed]
- Thakkar, J.P.; Kumthekar, P.; Dixit, K.S.; Stupp, R.; Lukas, R.V. Leptomeningeal metastasis from solid tumors. J. Neurol. Sci. 2020, 411, 116706. [Google Scholar] [CrossRef]
- Le Rhun, E.; Taillibert, S.; Chamberlain, M.C. Carcinomatous meningitis: Leptomeningeal metastases in solid tumors. Surg. Neurol. Int. 2013, 4, S265–S288. [Google Scholar] [CrossRef]
- Ellison, D.W.; Aldape, K.D.; Capper, D.; Fouladi, M.; Gilbert, M.R.; Gilbertson, R.J.; Hawkins, C.; Merchant, T.E.; Pajtler, K.; Venneti, S.; et al. cIMPACT-NOW update 7: Advancing the molecular classification of ependymal tumors. Brain Pathol. 2020, 30, 863–866. [Google Scholar] [CrossRef]
- Doglioni, G.; Parik, S.; Fendt, S.M. Interactions in the (Pre)metastatic Niche Support Metastasis Formation. Front. Oncol. 2019, 9, 219. [Google Scholar] [CrossRef]
- Costa-Silva, B.; Aiello, N.M.; Ocean, A.J.; Singh, S.; Zhang, H.; Thakur, B.K.; Becker, A.; Hoshino, A.; Mark, M.T.; Molina, H.; et al. Pancreatic cancer exosomes initiate pre-metastatic niche formation in the liver. Nat. Cell Biol. 2015, 17, 816–826. [Google Scholar] [CrossRef]
- Kong, J.; Tian, H.; Zhang, F.; Zhang, Z.; Li, J.; Liu, X.; Li, X.; Liu, J.; Li, X.; Jin, D.; et al. Extracellular vesicles of carcinoma-associated fibroblasts creates a pre-metastatic niche in the lung through activating fibroblasts. Mol. Cancer 2019, 18, 175. [Google Scholar] [CrossRef]
- Kaplan, R.N.; Riba, R.D.; Zacharoulis, S.; Bramley, A.H.; Vincent, L.; Costa, C.; MacDonald, D.D.; Jin, D.K.; Shido, K.; Kerns, S.A.; et al. VEGFR1-positive haematopoietic bone marrow progenitors initiate the pre-metastatic niche. Nature 2005, 438, 820–827. [Google Scholar] [CrossRef]
- Carpenter, R.A.; Kwak, J.G.; Peyton, S.R.; Lee, J. Implantable pre-metastatic niches for the study of the microenvironmental regulation of disseminated human tumour cells. Nat. Biomed. Eng. 2018, 2, 915–929. [Google Scholar] [CrossRef]
- Huang, Y.; Song, N.; Ding, Y.; Yuan, S.; Li, X.; Cai, H.; Shi, H.; Luo, Y. Pulmonary vascular destabilization in the premetastatic phase facilitates lung metastasis. Cancer Res. 2009, 69, 7529–7537. [Google Scholar] [CrossRef] [Green Version]
- Hiratsuka, S.; Ishibashi, S.; Tomita, T.; Watanabe, A.; Akashi-Takamura, S.; Murakami, M.; Kijima, H.; Miyake, K.; Aburatani, H.; Maru, Y. Primary tumours modulate innate immune signalling to create pre-metastatic vascular hyperpermeability foci. Nat. Commun. 2013, 4, 1853. [Google Scholar] [CrossRef] [Green Version]
- Izraely, S.; Witz, I.P. Site-specific metastasis: A cooperation between cancer cells and the metastatic microenvironment. Int. J. Cancer 2021, 148, 1308. [Google Scholar] [CrossRef]
- Kim, S.H.; Redvers, R.P.; Chi, L.H.; Ling, X.; Lucke, A.J.; Reid, R.C.; Fairlie, D.P.; Martin, A.; Anderson, R.L.; Denoyer, D.; et al. Identification of brain metastasis genes and therapeutic evaluation of histone deacetylase inhibitors in a clinically relevant model of breast cancer brain metastasis. Dis. Model Mech. 2018, 11, DMM034850. [Google Scholar] [CrossRef] [Green Version]
- Schackert, G.; Fidler, I.J. Site-specific metastasis of mouse melanomas and a fibrosarcoma in the brain or meninges of syngeneic animals. Cancer Res. 1988, 48, 3478–3484. [Google Scholar]
- Gal, A.; Sjoblom, T.; Fedorova, L.; Imreh, S.; Beug, H.; Moustakas, A. Sustained TGF beta exposure suppresses Smad and non-Smad signalling in mammary epithelial cells, leading to EMT and inhibition of growth arrest and apoptosis. Oncogene 2008, 27, 1218–1230. [Google Scholar] [CrossRef] [Green Version]
- Voloshenyuk, T.G.; Landesman, E.S.; Khoutorova, E.; Hart, A.D.; Gardner, J.D. Induction of cardiac fibroblast lysyl oxidase by TGF-beta1 requires PI3K/Akt, Smad3, and MAPK signaling. Cytokine 2011, 55, 90–97. [Google Scholar] [CrossRef]
- Elmansuri, A.Z.; Tanino, M.A.; Mahabir, R.; Wang, L.; Kimura, T.; Nishihara, H.; Kinoshita, I.; Dosaka-Akita, H.; Tsuda, M.; Tanaka, S. Novel signaling collaboration between TGF-β and adaptor protein Crk facilitates EMT in human lung cancer. Oncotarget 2016, 7, 27094–27107. [Google Scholar] [CrossRef] [Green Version]
- Wu, D.M.; Deng, S.H.; Liu, T.; Han, R.; Zhang, T.; Xu, Y. TGF-beta-mediated exosomal lnc-MMP2-2 regulates migration and invasion of lung cancer cells to the vasculature by promoting MMP2 expression. Cancer Med. 2018, 7, 5118–5129. [Google Scholar] [CrossRef]
- Wu, D.; Deng, S.; Li, L.; Liu, T.; Zhang, T.; Li, J.; Yu, Y.; Xu, Y. TGF-beta1-mediated exosomal lnc-MMP2-2 increases blood-brain barrier permeability via the miRNA-1207-5p/EPB41L5 axis to promote non-small cell lung cancer brain metastasis. Cell Death Dis. 2021, 12, 721. [Google Scholar] [CrossRef]
- Hashimoto, A.; Hashimoto, S.; Sugino, H.; Yoshikawa, A.; Onodera, Y.; Handa, H.; Oikawa, T.; Sabe, H. ZEB1 induces EPB41L5 in the cancer mesenchymal program that drives ARF6-based invasion, metastasis and drug resistance. Oncogenesis 2016, 5, e259. [Google Scholar] [CrossRef] [Green Version]
- Jeong, M.H.; Park, S.Y.; Lee, S.H.; Seo, J.; Yoo, J.Y.; Park, S.H.; Kim, M.J.; Lee, S.; Jang, S.; Choi, H.K.; et al. EPB41L5 Mediates TGFbeta-Induced Metastasis of Gastric Cancer. Clin. Cancer Res. 2019, 25, 3617–3629. [Google Scholar] [CrossRef] [Green Version]
- Lv, T.; Miao, Y.; Xu, T.; Sun, W.; Sang, Y.; Jia, F.; Zhang, X. Circ-EPB41L5 regulates the host gene EPB41L5 via sponging miR-19a to repress glioblastoma tumorigenesis. Aging 2020, 12, 318–339. [Google Scholar] [CrossRef]
- Michael, I.P.; Saghafinia, S.; Tichet, M.; Zangger, N.; Marinoni, I.; Perren, A.; Hanahan, D. ALK7 Signaling Manifests a Homeostatic Tissue Barrier That Is Abrogated during Tumorigenesis and Metastasis. Dev. Cell 2019, 49, 409–424.e6. [Google Scholar] [CrossRef]
- Gan, D.X.; Wang, Y.B.; He, M.Y.; Chen, Z.Y.; Qin, X.X.; Miao, Z.W.; Chen, Y.H.; Li, B. Lung Cancer Cells-Controlled Dkk-1 Production in Brain Metastatic Cascade Drive Microglia to Acquire a Pro-tumorigenic Phenotype. Front. Cell Dev. Biol. 2020, 8, 591405. [Google Scholar] [CrossRef]
- Fong, M.Y.; Zhou, W.; Liu, L.; Alontaga, A.Y.; Chandra, M.; Ashby, J.; Chow, A.; O’Connor, S.T.; Li, S.; Chin, A.R.; et al. Breast-cancer-secreted miR-122 reprograms glucose metabolism in premetastatic niche to promote metastasis. Nat. Cell Biol. 2015, 17, 183–194. [Google Scholar] [CrossRef] [Green Version]
- Kienast, Y.; von Baumgarten, L.; Fuhrmann, M.; Klinkert, W.E.; Goldbrunner, R.; Herms, J.; Winkler, F. Real-time imaging reveals the single steps of brain metastasis formation. Nat. Med. 2010, 16, 116–122. [Google Scholar] [CrossRef]
- Winkler, F. Pathogenesis and biology. Handb. Clin. Neurol. 2018, 149, 43–56. [Google Scholar] [CrossRef]
- Bos, P.D.; Zhang, X.H.; Nadal, C.; Shu, W.; Gomis, R.R.; Nguyen, D.X.; Minn, A.J.; van de Vijver, M.J.; Gerald, W.L.; Foekens, J.A.; et al. Genes that mediate breast cancer metastasis to the brain. Nature 2009, 459, 1005–1009. [Google Scholar] [CrossRef] [PubMed]
- Miyamoto, S.; Yagi, H.; Yotsumoto, F.; Kawarabayashi, T.; Mekada, E. Heparin-binding epidermal growth factor-like growth factor as a novel targeting molecule for cancer therapy. Cancer Sci. 2006, 97, 341–347. [Google Scholar] [CrossRef] [PubMed]
- De Vries, H.E.; Blom-Roosemalen, M.C.; van Oosten, M.; de Boer, A.G.; van Berkel, T.J.; Breimer, D.D.; Kuiper, J. The influence of cytokines on the integrity of the blood-brain barrier in vitro. J. Neuroimmunol. 1996, 64, 37–43. [Google Scholar] [CrossRef]
- Rodewald, A.K.; Rushing, E.J.; Kirschenbaum, D.; Mangana, J.; Mittmann, C.; Moch, H.; Lugassy, C.; Barnhill, R.L.; Mihic-Probst, D. Eight autopsy cases of melanoma brain metastases showing angiotropism and pericytic mimicry. Implications for extravascular migratory metastasis. J. Cutan. Pathol. 2019, 46, 570–578. [Google Scholar] [CrossRef] [PubMed]
- Holash, J.; Maisonpierre, P.C.; Compton, D.; Boland, P.; Alexander, C.R.; Zagzag, D.; Yancopoulos, G.D.; Wiegand, S.J.; Holash, J.; Maisonpierre, P.C.; et al. Vessel Cooption, Regression, and Growth in Tumors Mediated by Angiopoietins and VEGF. Science 1999, 284, 1994–1998. [Google Scholar] [CrossRef] [Green Version]
- Küsters, B.; Westphal, J.R.; Smits, D.; Ruiter, D.J.; Wesseling, P.; Keilholz, U.; de Waal, R.M. The pattern of metastasis of human melanoma to the central nervous system is not influenced by integrin alpha(v) beta(3) expression. Int. J. Cancer 2001, 92, 176–180. [Google Scholar] [CrossRef]
- Zhang, C.; Zhang, F.; Tsan, R.; Fidler, I.J. Transforming growth factor-beta2 is a molecular determinant for site-specific melanoma metastasis in the brain. Cancer Res. 2009, 69, 828–835. [Google Scholar] [CrossRef] [Green Version]
- Schackert, G.; Price, J.E.; Zhang, R.D.; Bucana, C.D.; Itoh, K.; Fidler, I.J. Regional growth of different human melanomas as metastases in the brain of nude mice. Am. J. Pathol. 1990, 136, 95–102. [Google Scholar]
- Cruz-Munoz, W.; Kerbel, R.S. Preclinical approaches to study the biology and treatment of brain metastases. Semin. Cancer Biol. 2011, 21, 123–130. [Google Scholar] [CrossRef] [Green Version]
- Remsik, J.; Chi, Y.; Tong, X.; Sener, U.; Derderian, C.; Park, A.; Saadeh, F.; Bale, T.; Boire, A. Leptomeningeal metastatic cells adopt two phenotypic states. Cancer Rep. 2020, 369, e1236. [Google Scholar] [CrossRef]
- Chi, Y.; Remsik, J.; Kiseliovas, V.; Derderian, C.; Sener, U.; Alghader, M.; Saadeh, F.; Nikishina, K.; Bale, T.; Iacobuzio-Donahue, C.; et al. Cancer cells deploy lipocalin-2 to collect limiting iron in leptomeningeal metastasis. Science 2020, 369, 276–282. [Google Scholar] [CrossRef]
- Boire, A.; Zou, Y.; Shieh, J.; Macalinao, D.G.; Pentsova, E.; Massague, J. Complement Component 3 Adapts the Cerebrospinal Fluid for Leptomeningeal Metastasis. Cell 2017, 168, 1101–1113.e13. [Google Scholar] [CrossRef] [Green Version]
- Da Mesquita, S.; Louveau, A.; Vaccari, A.; Smirnov, I.; Cornelison, R.C.; Kingsmore, K.M.; Contarino, C.; Onengut-Gumuscu, S.; Farber, E.; Raper, D.; et al. Functional aspects of meningeal lymphatics in ageing and Alzheimer’s disease. Nature 2018, 560, 185–191. [Google Scholar] [CrossRef]
- Louveau, A.; Harris, T.H.; Kipnis, J. Revisiting the Mechanisms of CNS Immune Privilege. Trends Immunol. 2015, 36, 569–577. [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]
- Baslan, T.; Hicks, J. Unravelling biology and shifting paradigms in cancer with single-cell sequencing. Nat. Rev. Cancer 2017, 17, 557–569. [Google Scholar] [CrossRef]
- AlJanahi, A.A.; Danielsen, M.; Dunbar, C.E. An Introduction to the Analysis of Single-Cell RNA-Sequencing Data. Mol. Ther. Methods Clin. Dev. 2018, 10, 189–196. [Google Scholar] [CrossRef] [Green Version]
- Xie, Y.; He, L.; Lugano, R.; Zhang, Y.; Cao, H.; He, Q.; Chao, M.; Liu, B.; Cao, Q.; Wang, J.; et al. Key molecular alterations in endothelial cells in human glioblastoma uncovered through single-cell RNA sequencing. JCI Insight 2021, 6, e150861. [Google Scholar] [CrossRef]
- Carlson, J.C.; Cantu Gutierrez, M.; Lozzi, B.; Huang-Hobbs, E.; Turner, W.D.; Tepe, B.; Zhang, Y.; Herman, A.M.; Rao, G.; Creighton, C.J.; et al. Identification of diverse tumor endothelial cell populations in malignant glioma. Neuro-Oncol. 2021, 23, 932–944. [Google Scholar] [CrossRef]
- Schulz, M.; Michels, B.; Niesel, K.; Stein, S.; Farin, H.; Rodel, F.; Sevenich, L. Cellular and Molecular Changes of Brain Metastases-Associated Myeloid Cells during Disease Progression and Therapeutic Response. iScience 2020, 23, 101178. [Google Scholar] [CrossRef]
- Hunter, M.V.; Moncada, R.; Weiss, J.M.; Yanai, I.; White, R.M. Spatially resolved transcriptomics reveals the architecture of the tumor-microenvironment interface. Nat. Commun. 2021, 12, 6278. [Google Scholar] [CrossRef]
- 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 2007, 109, 1010–1017. [Google Scholar] [CrossRef] [Green Version]
- Farnsworth, R.H.; Karnezis, T.; Shayan, R.; Matsumoto, M.; Nowell, C.J.; Achen, M.G.; Stacker, S.A. A role for bone morphogenetic protein-4 in lymph node vascular remodeling and primary tumor growth. Cancer Res. 2011, 71, 6547–6557. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Stacker, S.A.; Williams, S.P.; Karnezis, T.; Shayan, R.; Fox, S.B.; Achen, M.G. Lymphangiogenesis and lymphatic vessel remodelling in cancer. Nat. Rev. Cancer 2014, 14, 159–172. [Google Scholar] [CrossRef]
- Williams, S.P.; Odell, A.F.; Karnezis, T.; Farnsworth, R.H.; Gould, C.M.; Li, J.; Paquet-Fifield, S.; Harris, N.C.; Walter, A.; Gregory, J.L.; et al. Genome-wide functional analysis reveals central signaling regulators of lymphatic endothelial cell migration and remodeling. Sci. Signal. 2017, 10, eaal2987. [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]
- He, M.Y.; Halford, M.M.; Liu, R.; Roy, J.P.; Grant, Z.L.; Coultas, L.; Thio, N.; Gilan, O.; Chan, Y.C.; Dawson, M.A.; et al. Three-dimensional CRISPR screening reveals epigenetic interaction with anti-angiogenic therapy. Commun. Biol. 2021, 4, 878. [Google Scholar] [CrossRef]
Endothelial Cell Specialization | Arterial | Capillary | Venous | Choroid Plexus | Meningeal | Meningeal Lymphatic |
---|---|---|---|---|---|---|
BBB characteristics | Yes | Yes | Yes | No | No | No |
Molecular/transcriptomic markers | Pecam1 Cldn5 Cd34 Bmx Vcam1 Vegfc Efnb2 Sema3g | Pecam1 Cldn5 Cd34 Tfrc Mfsd2a Slc16a1 | Pecam1 Cldn5 Cd34 Vwf Vcam1 Slc38a5 Nr2f2 | Pecam1 Cd34 Plvap Plpp3 Esm1 Cd24a | Pecam1 Cd34 Plvap | Pecam1 Lyve1 Flt4 Ccl21b Prox1 |
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
Tobar, L.E.; Farnsworth, R.H.; Stacker, S.A. Brain Vascular Microenvironments in Cancer Metastasis. Biomolecules 2022, 12, 401. https://doi.org/10.3390/biom12030401
Tobar LE, Farnsworth RH, Stacker SA. Brain Vascular Microenvironments in Cancer Metastasis. Biomolecules. 2022; 12(3):401. https://doi.org/10.3390/biom12030401
Chicago/Turabian StyleTobar, Lucas E., Rae H. Farnsworth, and Steven A. Stacker. 2022. "Brain Vascular Microenvironments in Cancer Metastasis" Biomolecules 12, no. 3: 401. https://doi.org/10.3390/biom12030401
APA StyleTobar, L. E., Farnsworth, R. H., & Stacker, S. A. (2022). Brain Vascular Microenvironments in Cancer Metastasis. Biomolecules, 12(3), 401. https://doi.org/10.3390/biom12030401