Heterogeneity of Vascular Endothelial Cells, De Novo Arteriogenesis and Therapeutic Implications in Pancreatic Neuroendocrine Tumors
Abstract
:1. Introduction
2. Heterogeneity of Vascular Endothelial Cells
3. De Novo Arteriogenesis, an Emerging Concept of Formation of New Vascular Networks
4. Tumor Arteriogenesis: Potential Target in pNETs
5. Antiangiogenic Therapy in pNETs: Challenges and Prospective
5.1. Notch Pathway: The Potential Target in pNETs
5.2. Control of pNET Progression by Targeting Vascular Niche and CSC Plasticity
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Halfdanarson, T.R.; Rabe, K.G.; Rubin, J.; Petersen, G.M. Pancreatic neuroendocrine tumors (PNETs): Incidence, prognosis and recent trend toward improved survival. Ann. Oncol. 2008, 19, 1727–1733. [Google Scholar] [CrossRef] [PubMed]
- Terris, B.; Scoazec, J.Y.; Rubbia, L.; Bregeaud, L.; Pepper, M.S.; Ruszniewski, P.; Belghiti, J.; Flejou, J.; Degott, C. Expression of vascular endothelial growth factor in digestive neuroendocrine tumours. Histopathology 1998, 32, 133–138. [Google Scholar] [CrossRef] [PubMed]
- Casanovas, O.; Hicklin, D.J.; Bergers, G.; Hanahan, D. Drug resistance by evasion of antiangiogenic targeting of VEGF signaling in late-stage pancreatic islet tumors. Cancer Cell 2005, 8, 299–309. [Google Scholar] [CrossRef] [PubMed]
- Gomez, D.; Malik, H.Z.; Al-Mukthar, A.; Menon, K.V.; Toogood, G.J.; Lodge, J.P.; Prasad, K.R. Hepatic resection for metastatic gastrointestinal and pancreatic neuroendocrine tumours: Outcome and prognostic predictors. HPB (Oxford) 2007, 9, 345–351. [Google Scholar] [CrossRef]
- De Dosso, S.; Grande, E.; Barriuso, J.; Castellano, D.; Tabernero, J.; Capdevila, J. The targeted therapy revolution in neuroendocrine tumors: In search of biomarkers for patient selection and response evaluation. Cancer Metastasis Rev. 2013, 32, 465–477. [Google Scholar] [CrossRef]
- Sennino, B.; Ishiguro-Oonuma, T.; Wei, Y.; Naylor, R.M.; Williamson, C.W.; Bhagwandin, V.; Tabruyn, S.P.; You, W.K.; Chapman, H.A.; Christensen, J.G.; et al. Suppression of tumor invasion and metastasis by concurrent inhibition of c-Met and VEGF signaling in pancreatic neuroendocrine tumors. Cancer Discov. 2012, 2, 270–287. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef]
- Lenzi, P.; Bocci, G.; Natale, G. John Hunter and the origin of the term “angiogenesis”. Angiogenesis 2016, 19, 255–256. [Google Scholar] [CrossRef]
- Folkman, J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat. Med. 1995, 1, 27–31. [Google Scholar] [CrossRef]
- Carmeliet, P. Mechanisms of angiogenesis and arteriogenesis. Nat. Med. 2000, 6, 389–395. [Google Scholar] [CrossRef]
- Shalaby, F.; Rossant, J.; Yamaguchi, T.P.; Gertsenstein, M.; Wu, X.F.; Breitman, M.L.; Schuh, A.C. Failure of blood-island formation and vasculogenesis in Flk-1-deficient mice. Nature 1995, 376, 62–66. [Google Scholar] [CrossRef] [PubMed]
- Dvorak, H.F.; Brown, L.F.; Detmar, M.; Dvorak, A.M. Vascular permeability factor/vascular endothelial growth factor, microvascular hyperpermeability, and angiogenesis. Am. J. Pathol. 1995, 146, 1029–1039. [Google Scholar] [PubMed]
- Bernardi, R.; Guernah, I.; Jin, D.; Grisendi, S.; Alimonti, A.; Teruya-Feldstein, J.; Cordon-Cardo, C.; Simon, M.C.; Rafii, S.; Pandolfi, P.P. PML inhibits HIF-1alpha translation and neoangiogenesis through repression of mTOR. Nature 2006, 442, 779–785. [Google Scholar] [CrossRef] [PubMed]
- Potente, M.; Gerhardt, H.; Carmeliet, P. Basic and therapeutic aspects of angiogenesis. Cell 2011, 146, 873–887. [Google Scholar] [CrossRef]
- Carmeliet, P.; Jain, R.K. Molecular mechanisms and clinical applications of angiogenesis. Nature 2011, 473, 298–307. [Google Scholar] [CrossRef]
- Simons, M.; Ware, J.A. Therapeutic angiogenesis in cardiovascular disease. Nat. Rev. Drug Discov. 2003, 2, 863–871. [Google Scholar] [CrossRef]
- Mac Gabhann, F.; Peirce, S.M. Collateral capillary arterialization following arteriolar ligation in murine skeletal muscle. Microcirculation 2010, 17, 333–347. [Google Scholar] [CrossRef]
- Ren, B.; Best, B.; Weihrauch, D.; Jones, D.W.; Dong, L.; Opansky, C.; Yuan, R.; Pritchard, K.A.; Silverstein, R. Abstract 15673: LPA/PKD-1-FoxO1-CD36 Signaling Axis Regulates Capillary Arterialization in Ischemic Conditions. Circulation 2016, 134, A15673. [Google Scholar] [CrossRef]
- Ren, B.; Deng, Y.; Mukhopadhyay, A.; Lanahan, A.A.; Zhuang, Z.W.; Moodie, K.L.; Mulligan-Kehoe, M.J.; Byzova, T.V.; Peterson, R.T.; Simons, M. ERK1/2-Akt1 crosstalk regulates arteriogenesis in mice and zebrafish. J. Clin. Invest. 2010, 120, 1217–1228. [Google Scholar] [CrossRef]
- Dong, L.; Yuan, Y.; Opansky, C.; Chen, Y.; Aguilera-Barrantes, I.; Wu, S.; Yuan, R.; Cao, Q.; Cheng, Y.C.; Sahoo, D.; et al. Diet-induced obesity links to ER positive breast cancer progression via LPA/PKD-1-CD36 signaling-mediated microvascular remodeling. Oncotarget 2017, 8, 22550–22562. [Google Scholar] [CrossRef]
- Ren, B.; Best, B.; Ramakrishnan, D.P.; Walcott, B.P.; Storz, P.; Silverstein, R.L. LPA/PKD-1-FoxO1 Signaling Axis Mediates Endothelial Cell CD36 Transcriptional Repression and Proangiogenic and Proarteriogenic Reprogramming. Arter. Thromb. Vasc. Biol. 2016, 36, 1197–1208. [Google Scholar] [CrossRef] [PubMed]
- Ren, B.; Hale, J.; Srikanthan, S.; Silverstein, R.L. Lysophosphatidic acid suppresses endothelial cell CD36 expression and promotes angiogenesis via a PKD-1-dependent signaling pathway. Blood 2011, 117, 6036–6045. [Google Scholar] [CrossRef] [PubMed]
- Aitman, T.J.; Glazier, A.M.; Wallace, C.A.; Cooper, L.D.; Norsworthy, P.J.; Wahid, F.N.; Al-Majali, K.M.; Trembling, P.M.; Mann, C.J.; Shoulders, C.C.; et al. Identification of Cd36 (Fat) as an insulin-resistance gene causing defective fatty acid and glucose metabolism in hypertensive rats. Nat. Genet. 1999, 21, 76–83. [Google Scholar] [CrossRef] [PubMed]
- Yuan, Y.; Kohlenberg, J.D.; Chen, Y.; Komas, S.; Xin, G.; Yuan, G.; Cui, W.; Wu, S.; Ren, B. Abstract A09: Diet-induced obesity promotes breast cancer progression by LPA-signaling-mediated functional changes of mitochondria and angiogenesis. Cancer Res. 2015, 75, A09. [Google Scholar] [CrossRef]
- Pascual, G.; Avgustinova, A.; Mejetta, S.; Martin, M.; Castellanos, A.; Attolini, C.S.; Berenguer, A.; Prats, N.; Toll, A.; Hueto, J.A.; et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 2017, 541, 41–45. [Google Scholar] [CrossRef]
- Hale, J.S.; Otvos, B.; Sinyuk, M.; Alvarado, A.G.; Hitomi, M.; Stoltz, K.; Wu, Q.; Flavahan, W.; Levison, B.; Johansen, M.L.; et al. Cancer stem cell-specific scavenger receptor CD36 drives glioblastoma progression. Stem Cells 2014, 32, 1746–1758. [Google Scholar] [CrossRef]
- Nagy, J.A.; Dvorak, H.F. Heterogeneity of the tumor vasculature: The need for new tumor blood vessel type-specific targets. Clin. Exp. Metastasis 2012, 29, 657–662. [Google Scholar] [CrossRef]
- Sitohy, B.; Nagy, J.A.; Jaminet, S.C.; Dvorak, H.F. Tumor-surrogate blood vessel subtypes exhibit differential susceptibility to anti-VEGF therapy. Cancer Res. 2011, 71, 7021–7028. [Google Scholar] [CrossRef]
- Ribatti, D. The involvement of endothelial progenitor cells in tumor angiogenesis. J. Cell Mol. Med. 2004, 8, 294–300. [Google Scholar] [CrossRef]
- Wang, R.; Chadalavada, K.; Wilshire, J.; Kowalik, U.; Hovinga, K.E.; Geber, A.; Fligelman, B.; Leversha, M.; Brennan, C.; Tabar, V. Glioblastoma stem-like cells give rise to tumour endothelium. Nature 2010, 468, 829–833. [Google Scholar] [CrossRef]
- Yuan, L.; Chan, G.C.; Beeler, D.; Janes, L.; Spokes, K.C.; Dharaneeswaran, H.; Mojiri, A.; Adams, W.J.; Sciuto, T.; Garcia-Cardena, G.; et al. A role of stochastic phenotype switching in generating mosaic endothelial cell heterogeneity. Nat. Commun. 2016, 7, 10160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Regan, E.R.; Aird, W.C. Dynamical systems approach to endothelial heterogeneity. Circ. Res. 2012, 111, 110–130. [Google Scholar] [CrossRef] [PubMed]
- Ren, B. Endothelial Cells: A Key Player in Angiogenesis and Lymphangiogenesis. MOJ Cell Sci. Rep. 2015, 2. [Google Scholar] [CrossRef] [Green Version]
- Chi, J.T.; Chang, H.Y.; Haraldsen, G.; Jahnsen, F.L.; Troyanskaya, O.G.; Chang, D.S.; Wang, Z.; Rockson, S.G.; van de Rijn, M.; Botstein, D.; et al. Endothelial cell diversity revealed by global expression profiling. Proc. Natl. Acad. Sci. USA 2003, 100, 10623–10628. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Best, B.; Moran, P.; Ren, B. VEGF/PKD-1 signaling mediates arteriogenic gene expression and angiogenic responses in reversible human microvascular endothelial cells with extended lifespan. Mol. Cell Biochem. 2018, 446, 199–207. [Google Scholar] [CrossRef] [PubMed]
- Nolan, D.J.; Ginsberg, M.; Israely, E.; Palikuqi, B.; Poulos, M.G.; James, D.; Ding, B.S.; Schachterle, W.; Liu, Y.; Rosenwaks, Z.; et al. Molecular signatures of tissue-specific microvascular endothelial cell heterogeneity in organ maintenance and regeneration. Dev. Cell 2013, 26, 204–219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marcu, R.; Choi, Y.J.; Xue, J.; Fortin, C.L.; Wang, Y.; Nagao, R.J.; Xu, J.; MacDonald, J.W.; Bammler, T.K.; Murry, C.E.; et al. Human Organ-Specific Endothelial Cell Heterogeneity. iScience 2018, 4, 20–35. [Google Scholar] [CrossRef] [Green Version]
- Bentley, K.; Franco, C.A.; Philippides, A.; Blanco, R.; Dierkes, M.; Gebala, V.; Stanchi, F.; Jones, M.; Aspalter, I.M.; Cagna, G.; et al. The role of differential VE-cadherin dynamics in cell rearrangement during angiogenesis. Nat. Cell Biol. 2014, 16, 309–321. [Google Scholar] [CrossRef]
- Ren, B. FoxO1 transcriptional activities in VEGF expression and beyond: A key regulator in functional angiogenesis? J. Pathol. 2018, 245, 255–257. [Google Scholar] [CrossRef] [Green Version]
- Ren, B. Protein Kinase D1 Signaling in Angiogenic Gene Expression and VEGF-Mediated Angiogenesis. Front. Cell Dev. Biol. 2016, 4, 37. [Google Scholar] [CrossRef] [Green Version]
- Baek, K.I.; Packard, R.R.S.; Hsu, J.J.; Saffari, A.; Ma, Z.; Luu, A.P.; Pietersen, A.; Yen, H.; Ren, B.; Ding, Y.; et al. Ultrafine Particle Exposure Reveals the Importance of FOXO1/Notch Activation Complex for Vascular Regeneration. Antioxid. Redox Signal. 2018, 28, 1209–1223. [Google Scholar] [CrossRef]
- Denekamp, J. Endothelial cell proliferation as a novel approach to targeting tumour therapy. Br. J. Cancer 1982, 45, 136–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Morikawa, S.; Baluk, P.; Kaidoh, T.; Haskell, A.; Jain, R.K.; McDonald, D.M. Abnormalities in pericytes on blood vessels and endothelial sprouts in tumors. Am. J. Pathol. 2002, 160, 985–1000. [Google Scholar] [CrossRef] [Green Version]
- Orimo, A.; Weinberg, R.A. Stromal fibroblasts in cancer: A novel tumor-promoting cell type. Cell Cycle 2006, 5, 1597–1601. [Google Scholar] [CrossRef] [PubMed]
- Franklin, R.A.; Liao, W.; Sarkar, A.; Kim, M.V.; Bivona, M.R.; Liu, K.; Pamer, E.G.; Li, M.O. The cellular and molecular origin of tumor-associated macrophages. Science 2014, 344, 921–925. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ronca, R.; Van Ginderachter, J.A.; Turtoi, A. Paracrine interactions of cancer-associated fibroblasts, macrophages and endothelial cells: Tumor allies and foes. Curr. Opin. Oncol. 2018, 30, 45–53. [Google Scholar] [CrossRef]
- Wilson, W.R.; Hay, M.P. Targeting hypoxia in cancer therapy. Nat. Rev. Cancer 2011, 11, 393–410. [Google Scholar] [CrossRef]
- Krock, B.L.; Skuli, N.; Simon, M.C. Hypoxia-induced angiogenesis: Good and evil. Genes Cancer 2011, 2, 1117–1133. [Google Scholar] [CrossRef] [Green Version]
- Senger, D.R.; Galli, S.J.; Dvorak, A.M.; Perruzzi, C.A.; Harvey, V.S.; Dvorak, H.F. Tumor cells secrete a vascular permeability factor that promotes accumulation of ascites fluid. Science 1983, 219, 983–985. [Google Scholar] [CrossRef]
- Ren, B.; Hoti, N.; Rabasseda, X.; Wang, Y.Z.; Wu, M. The antiangiogenic and therapeutic implications of endostatin. Methods Find. Exp. Clin. Pharm. 2003, 25, 215–224. [Google Scholar] [CrossRef]
- Ren, B.; Yee, K.O.; Lawler, J.; Khosravi-Far, R. Regulation of tumor angiogenesis by thrombospondin-1. Biochim. Biophys. Acta 2006, 1765, 178–188. [Google Scholar] [CrossRef] [PubMed]
- Ren, B.; Song, K.; Parangi, S.; Jin, T.; Ye, M.; Humphreys, R.; Duquette, M.; Zhang, X.; Benhaga, N.; Lawler, J.; et al. A double hit to kill tumor and endothelial cells by TRAIL and antiangiogenic 3TSR. Cancer Res. 2009, 69, 3856–3865. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Simons, M.; Gordon, E.; Claesson-Welsh, L. Mechanisms and regulation of endothelial VEGF receptor signalling. Nat. Rev. Mol. Cell Biol. 2016, 17, 611–625. [Google Scholar] [CrossRef] [PubMed]
- Nagy, J.A.; Chang, S.H.; Shih, S.C.; Dvorak, A.M.; Dvorak, H.F. Heterogeneity of the tumor vasculature. Semin. Thromb. Hemost. 2010, 36, 321–331. [Google Scholar] [CrossRef] [Green Version]
- Dvorak, H.F. Tumor Stroma, Tumor Blood Vessels, and Antiangiogenesis Therapy. Cancer J. 2015, 21, 237–243. [Google Scholar] [CrossRef]
- Hida, K.; Hida, Y.; Amin, D.N.; Flint, A.F.; Panigrahy, D.; Morton, C.C.; Klagsbrun, M. Tumor-associated endothelial cells with cytogenetic abnormalities. Cancer Res. 2004, 64, 8249–8255. [Google Scholar] [CrossRef] [Green Version]
- Ricci-Vitiani, L.; Pallini, R.; Biffoni, M.; Todaro, M.; Invernici, G.; Cenci, T.; Maira, G.; Parati, E.A.; Stassi, G.; Larocca, L.M.; et al. Tumour vascularization via endothelial differentiation of glioblastoma stem-like cells. Nature 2010, 468, 824–828. [Google Scholar] [CrossRef]
- Samson, T.; Welch, C.; Monaghan-Benson, E.; Hahn, K.M.; Burridge, K. Endogenous RhoG is rapidly activated after epidermal growth factor stimulation through multiple guanine-nucleotide exchange factors. Mol. Biol. Cell 2010, 21, 1629–1642. [Google Scholar] [CrossRef] [Green Version]
- Franses, J.W.; Edelman, E.R. The evolution of endothelial regulatory paradigms in cancer biology and vascular repair. Cancer Res. 2011, 71, 7339–7344. [Google Scholar] [CrossRef]
- Franses, J.W.; Baker, A.B.; Chitalia, V.C.; Edelman, E.R. Stromal endothelial cells directly influence cancer progression. Sci. Transl. Med. 2011, 3, 66ra65. [Google Scholar] [CrossRef] [Green Version]
- Jain, R.K.; Carmeliet, P. SnapShot: Tumor angiogenesis. Cell 2012, 149, 1408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jain, R.K. Antiangiogenesis strategies revisited: From starving tumors to alleviating hypoxia. Cancer Cell 2014, 26, 605–622. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Folkman, J. Tumor angiogenesis: Therapeutic implications. N. Engl. J. Med. 1971, 285, 1182–1186. [Google Scholar] [CrossRef] [PubMed]
- Pluda, J.M. Tumor-associated angiogenesis: Mechanisms, clinical implications, and therapeutic strategies. Semin. Oncol. 1997, 24, 203–218. [Google Scholar]
- Brem, H.; Folkman, J. Inhibition of tumor angiogenesis mediated by cartilage. J. Exp. Med. 1975, 141, 427–439. [Google Scholar] [CrossRef]
- Hainaud, P.; Contreres, J.O.; Villemain, A.; Liu, L.X.; Plouet, J.; Tobelem, G.; Dupuy, E. The role of the vascular endothelial growth factor-Delta-like 4 ligand/Notch4-ephrin B2 cascade in tumor vessel remodeling and endothelial cell functions. Cancer Res. 2006, 66, 8501–8510. [Google Scholar] [CrossRef]
- Heil, M.; Eitenmuller, I.; Schmitz-Rixen, T.; Schaper, W. Arteriogenesis versus angiogenesis: Similarities and differences. J. Cell Mol. Med. 2006, 10, 45–55. [Google Scholar] [CrossRef] [Green Version]
- Cai, W.J.; Koltai, S.; Kocsis, E.; Scholz, D.; Kostin, S.; Luo, X.; Schaper, W.; Schaper, J. Remodeling of the adventitia during coronary arteriogenesis. Am. J. Physiol.-Heart Circ. Physiol. 2003, 284, H31–H40. [Google Scholar] [CrossRef] [Green Version]
- Lanahan, A.; Zhang, X.; Fantin, A.; Zhuang, Z.; Rivera-Molina, F.; Speichinger, K.; Prahst, C.; Zhang, J.; Wang, Y.; Davis, G.; et al. The neuropilin 1 cytoplasmic domain is required for VEGF-A-dependent arteriogenesis. Dev. Cell 2013, 25, 156–168. [Google Scholar] [CrossRef] [Green Version]
- Moraes, F.; Paye, J.; Mac Gabhann, F.; Zhuang, Z.W.; Zhang, J.; Lanahan, A.A.; Simons, M. Endothelial cell-dependent regulation of arteriogenesis. Circ. Res. 2013, 113, 1076–1086. [Google Scholar] [CrossRef] [Green Version]
- Moran, P.; Guo, Y.; Yuan, R.; Barnekow, N.; Palmer, J.; Beck, A.; Ren, B. Translating Ribosome Affinity Purification (TRAP) for RNA Isolation from Endothelial Cells In vivo. J. Vis. Exp. 2019, e59624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moran, P.; Opansky, C.; Weihrauch, D.; Yuan, R.; Jones, D.W.; Ramchandran, R.; Ren, B. Abstract 14944: Transcriptional Reprogramming of Endothelial Cells for Arteriolar Differentiation by Small Chemical Molecule via Protein Kinase D1 Signaling Pathway. Circulation 2017, 136, A14944. [Google Scholar] [CrossRef]
- Wang, H.U.; Chen, Z.F.; Anderson, D.J. Molecular distinction and angiogenic interaction between embryonic arteries and veins revealed by ephrin-B2 and its receptor Eph-B4. Cell 1998, 93, 741–753. [Google Scholar] [CrossRef] [Green Version]
- Weinstein, B.M.; Stemple, D.L.; Driever, W.; Fishman, M.C. Gridlock, a localized heritable vascular patterning defect in the zebrafish. Nat. Med. 1995, 1, 1143–1147. [Google Scholar] [CrossRef]
- Zhong, T.P.; Rosenberg, M.; Mohideen, M.A.; Weinstein, B.; Fishman, M.C. gridlock, an HLH gene required for assembly of the aorta in zebrafish. Science 2000, 287, 1820–1824. [Google Scholar] [CrossRef]
- Zhong, T.P.; Childs, S.; Leu, J.P.; Fishman, M.C. Gridlock signalling pathway fashions the first embryonic artery. Nature 2001, 414, 216–220. [Google Scholar] [CrossRef]
- Lawson, N.D.; Scheer, N.; Pham, V.N.; Kim, C.H.; Chitnis, A.B.; Campos-Ortega, J.A.; Weinstein, B.M. Notch signaling is required for arterial-venous differentiation during embryonic vascular development. Development 2001, 128, 3675–3683. [Google Scholar]
- Weinstein, B.M.; Lawson, N.D. Arteries, veins, Notch, and VEGF. Cold Spring Harb. Symp. Quant. Biol. 2002, 67, 155–162. [Google Scholar] [CrossRef]
- Mukouyama, Y.S.; Shin, D.; Britsch, S.; Taniguchi, M.; Anderson, D.J. Sensory nerves determine the pattern of arterial differentiation and blood vessel branching in the skin. Cell 2002, 109, 693–705. [Google Scholar] [CrossRef] [Green Version]
- Visconti, R.P.; Richardson, C.D.; Sato, T.N. Orchestration of angiogenesis and arteriovenous contribution by angiopoietins and vascular endothelial growth factor (VEGF). Proc. Natl. Acad. Sci. USA 2002, 99, 8219–8224. [Google Scholar] [CrossRef] [Green Version]
- Hong, C.C.; Peterson, Q.P.; Hong, J.Y.; Peterson, R.T. Artery/vein specification is governed by opposing phosphatidylinositol-3 kinase and MAP kinase/ERK signaling. Curr. Biol. 2006, 16, 1366–1372. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peterson, R.T.; Shaw, S.Y.; Peterson, T.A.; Milan, D.J.; Zhong, T.P.; Schreiber, S.L.; MacRae, C.A.; Fishman, M.C. Chemical suppression of a genetic mutation in a zebrafish model of aortic coarctation. Nat. Biotechnol. 2004, 22, 595–599. [Google Scholar] [CrossRef] [PubMed]
- Kazerounian, S.; Duquette, M.; Reyes, M.A.; Lawler, J.T.; Song, K.; Perruzzi, C.; Primo, L.; Khosravi-Far, R.; Bussolino, F.; Rabinovitz, I.; et al. Priming of the vascular endothelial growth factor signaling pathway by thrombospondin-1, CD36, and spleen tyrosine kinase. Blood 2011, 117, 4658–4666. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dewey, J.F. Regional tectonics. Science 1981, 214, 550–551. [Google Scholar] [CrossRef] [PubMed]
- Deindl, E.; Hoefer, I.E.; Fernandez, B.; Barancik, M.; Heil, M.; Strniskova, M.; Schaper, W. Involvement of the fibroblast growth factor system in adaptive and chemokine-induced arteriogenesis. Circ. Res. 2003, 92, 561–568. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Aragones, J.; Fraisl, P.; Baes, M.; Carmeliet, P. Oxygen sensors at the crossroad of metabolism. Cell Metab. 2009, 9, 11–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Helisch, A.; Schaper, W. Arteriogenesis: The development and growth of collateral arteries. Microcirculation 2003, 10, 83–97. [Google Scholar] [CrossRef]
- Dong, L.; Yuan, Y.; Aguilera-Barrantes, I.; Chen, Y.; Sturich, A.; Yuan, R.; Wu, S.; Silverstein, R.; Ren, B. Abstract 482: Signaling Lipid Lysophosphatidic Acid Is a Critical Link to Diet-induced Obesity, Cellular Bioenergetics and Breast Cancer Angiogenesis. Arterioscler. Thromb. Vasc. Biol. 2015, 35, A482. [Google Scholar]
- Yu, J.L.; Rak, J.W. Host microenvironment in breast cancer development: Inflammatory and immune cells in tumour angiogenesis and arteriogenesis. Breast Cancer Res. 2003, 5, 83–88. [Google Scholar] [CrossRef] [Green Version]
- Skuli, N.; Majmundar, A.J.; Krock, B.L.; Mesquita, R.C.; Mathew, L.K.; Quinn, Z.L.; Runge, A.; Liu, L.; Kim, M.N.; Liang, J.; et al. Endothelial HIF-2alpha regulates murine pathological angiogenesis and revascularization processes. J. Clin. Invest. 2012, 122, 1427–1443. [Google Scholar] [CrossRef]
- Skuli, N.; Liu, L.; Runge, A.; Wang, T.; Yuan, L.; Patel, S.; Iruela-Arispe, L.; Simon, M.C.; Keith, B. Endothelial deletion of hypoxia-inducible factor-2alpha (HIF-2alpha) alters vascular function and tumor angiogenesis. Blood 2009, 114, 469–477. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Eberhard, A.; Kahlert, S.; Goede, V.; Hemmerlein, B.; Plate, K.H.; Augustin, H.G. Heterogeneity of angiogenesis and blood vessel maturation in human tumors: Implications for antiangiogenic tumor therapies. Cancer Res. 2000, 60, 1388–1393. [Google Scholar] [PubMed]
- Buschmann, I.; Heil, M.; Jost, M.; Schaper, W. Influence of inflammatory cytokines on arteriogenesis. Microcirculation 2003, 10, 371–379. [Google Scholar] [CrossRef] [PubMed]
- Rissanen, T.T.; Korpisalo, P.; Markkanen, J.E.; Liimatainen, T.; Orden, M.R.; Kholova, I.; de Goede, A.; Heikura, T.; Grohn, O.H.; Yla-Herttuala, S. Blood flow remodels growing vasculature during vascular endothelial growth factor gene therapy and determines between capillary arterialization and sprouting angiogenesis. Circulation 2005, 112, 3937–3946. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schechter, J.; Goldsmith, P.; Wilson, C.; Weiner, R. Morphological evidence for the presence of arteries in human prolactinomas. J. Clin. Endocrinol. Metab. 1988, 67, 713–719. [Google Scholar] [CrossRef] [PubMed]
- Tinsley, J.H.; Teasdale, N.R.; Yuan, S.Y. Involvement of PKCdelta and PKD in pulmonary microvascular endothelial cell hyperpermeability. Am. J. Physiol. Cell Physiol. 2004, 286, C105–C111. [Google Scholar] [CrossRef] [Green Version]
- Shin, D.; Garcia-Cardena, G.; Hayashi, S.; Gerety, S.; Asahara, T.; Stavrakis, G.; Isner, J.; Folkman, J.; Gimbrone, M.A., Jr.; Anderson, D.J. Expression of ephrinB2 identifies a stable genetic difference between arterial and venous vascular smooth muscle as well as endothelial cells, and marks subsets of microvessels at sites of adult neovascularization. Dev. Biol. 2001, 230, 139–150. [Google Scholar] [CrossRef] [Green Version]
- Elias, D.; Lasser, P.; Ducreux, M.; Duvillard, P.; Ouellet, J.F.; Dromain, C.; Schlumberger, M.; Pocard, M.; Boige, V.; Miquel, C.; et al. Liver resection (and associated extrahepatic resections) for metastatic well-differentiated endocrine tumors: A 15-year single center prospective study. Surgery 2003, 133, 375–382. [Google Scholar] [CrossRef]
- Dromain, C.; de Baere, T.; Baudin, E.; Galline, J.; Ducreux, M.; Boige, V.; Duvillard, P.; Laplanche, A.; Caillet, H.; Lasser, P.; et al. MR imaging of hepatic metastases caused by neuroendocrine tumors: Comparing four techniques. Am. J. Roentgenol. 2003, 180, 121–128. [Google Scholar] [CrossRef]
- Calderone, A. The Biological Role of Nestin((+))-Cells in Physiological and Pathological Cardiovascular Remodeling. Front. Cell Dev. Biol. 2018, 6, 15. [Google Scholar] [CrossRef] [Green Version]
- Yazdani, S.; Kasajima, A.; Tamaki, K.; Nakamura, Y.; Fujishima, F.; Ohtsuka, H.; Motoi, F.; Unno, M.; Watanabe, M.; Sato, Y.; et al. Angiogenesis and vascular maturation in neuroendocrine tumors. Hum. Pathol. 2014, 45, 866–874. [Google Scholar] [CrossRef] [PubMed]
- Hall, S.M.; Hislop, A.A.; Pierce, C.M.; Haworth, S.G. Prenatal origins of human intrapulmonary arteries: Formation and smooth muscle maturation. Am. J. Respir. Cell Mol. Biol. 2000, 23, 194–203. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Helmlinger, G.; Netti, P.A.; Lichtenbeld, H.C.; Melder, R.J.; Jain, R.K. Solid stress inhibits the growth of multicellular tumor spheroids. Nat. Biotechnol. 1997, 15, 778–783. [Google Scholar] [CrossRef] [PubMed]
- Faivre, S.; Niccoli, P.; Castellano, D.; Valle, J.W.; Hammel, P.; Raoul, J.L.; Vinik, A.; Van Cutsem, E.; Bang, Y.J.; Lee, S.H.; et al. Sunitinib in pancreatic neuroendocrine tumors: Updated progression-free survival and final overall survival from a phase III randomized study. Ann. Oncol. 2017, 28, 339–343. [Google Scholar] [CrossRef] [PubMed]
- Gu, J.W.; Rizzo, P.; Pannuti, A.; Golde, T.; Osborne, B.; Miele, L. Notch signals in the endothelium and cancer “stem-like” cells: Opportunities for cancer therapy. Vasc. Cell 2012, 4, 7. [Google Scholar] [CrossRef] [Green Version]
- Rafii, S.; Butler, J.M.; Ding, B.S. Angiocrine functions of organ-specific endothelial cells. Nature 2016, 529, 316–325. [Google Scholar] [CrossRef] [Green Version]
- Awgulewitsch, C.P.; Trinh, L.T.; Hatzopoulos, A.K. The Vascular Wall: A Plastic Hub of Activity in Cardiovascular Homeostasis and Disease. Curr. Cardiol. Rep. 2017, 19, 51. [Google Scholar] [CrossRef]
- Kohlenberg, J.D.; Chen, Y.; Best, B.; Storz, P.; Peterson, R.T.; Silverstein, R.; Ren, B. Abstract LB-338: A novel LPA-PKD1-FoxO1 pathway in endothelial cells provides an angiogenic switch via down-regulation of CD36 transcription and induction of arteriogenic responses. Cancer Res. 2013, 73. [Google Scholar] [CrossRef]
- Opansky, C.; Best, B.; Yuan, R.; Cao, Q.; Ren, B. Protein Kinase D1 Signaling is the Key to Arterial Differentiation of Vascular Endothelial Cells. Circulation 2016, 134, A14437. [Google Scholar]
- Yao, J.C.; Eisner, M.P.; Leary, C.; Dagohoy, C.; Phan, A.; Rashid, A.; Hassan, M.; Evans, D.B. Population-based study of islet cell carcinoma. Ann. Surg. Oncol. 2007, 14, 3492–3500. [Google Scholar] [CrossRef] [Green Version]
- Metz, D.C.; Jensen, R.T. Gastrointestinal neuroendocrine tumors: Pancreatic endocrine tumors. Gastroenterology 2008, 135, 1469–1492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lawrence, B.; Gustafsson, B.I.; Chan, A.; Svejda, B.; Kidd, M.; Modlin, I.M. The epidemiology of gastroenteropancreatic neuroendocrine tumors. Endocrinol. Metab. Clin. N. Am. 2011, 40, 1–18. [Google Scholar] [CrossRef] [PubMed]
- Cueto, A.; Burigana, F.; Nicolini, A.; Lugnani, F. Neuroendocrine tumors of the lung: Hystological classification, diagnosis, traditional and new therapeutic approaches. Curr. Med. Chem. 2014, 21, 1107–1116. [Google Scholar] [CrossRef] [PubMed]
- Liakakos, T.; Roukos, D.H. Everolimus and sunitinib: From mouse models to treatment of pancreatic neuroendocrine tumors. Future Oncol. 2011, 7, 1025–1029. [Google Scholar] [CrossRef]
- Hori, T.; Takaori, K.; Uemoto, S. Pancreatic neuroendocrine tumor accompanied with multiple liver metastases. World J. Hepatol. 2014, 6, 596–600. [Google Scholar] [CrossRef]
- Rindi, G.; Klersy, C.; Albarello, L.; Baudin, E.; Bianchi, A.; Buchler, M.W.; Caplin, M.; Couvelard, A.; Cros, J.; de Herder, W.W.; et al. Competitive Testing of the WHO 2010 versus the WHO 2017 Grading of Pancreatic Neuroendocrine Neoplasms: Data from a Large International Cohort Study. Neuroendocrinology 2018, 107, 375–386. [Google Scholar] [CrossRef]
- Proye, C. Natural history of liver metastasis of gastroenteropancreatic neuroendocrine tumors: Place for chemoembolization. World J. Surg. 2001, 25, 685–688. [Google Scholar] [CrossRef]
- Fendrich, V.; Waldmann, J.; Bartsch, D.K.; Langer, P. Surgical management of pancreatic endocrine tumors. Nat. Rev. Clin. Oncol. 2009, 6, 419–428. [Google Scholar] [CrossRef]
- Nguyen, S.Q.; Angel, L.P.; Divino, C.M.; Schluender, S.; Warner, R.R. Surgery in malignant pancreatic neuroendocrine tumors. J. Surg. Oncol. 2007, 96, 397–403. [Google Scholar] [CrossRef]
- Villaume, K.; Blanc, M.; Gouysse, G.; Walter, T.; Couderc, C.; Nejjari, M.; Vercherat, C.; Cordier-Bussat, M.; Roche, C.; Scoazec, J.Y. VEGF secretion by neuroendocrine tumor cells is inhibited by octreotide and by inhibitors of the PI3K/AKT/mTOR pathway. Neuroendocrinology 2010, 91, 268–278. [Google Scholar] [CrossRef]
- Zhang, J.; Cao, R.; Zhang, Y.; Jia, T.; Cao, Y.; Wahlberg, E. Differential roles of PDGFR-alpha and PDGFR-beta in angiogenesis and vessel stability. FASEB J. 2009, 23, 153–163. [Google Scholar] [CrossRef] [PubMed]
- Raymond, E.; Dahan, L.; Raoul, J.L.; Bang, Y.J.; Borbath, I.; Lombard-Bohas, C.; Valle, J.; Metrakos, P.; Smith, D.; Vinik, A.; et al. Sunitinib malate for the treatment of pancreatic neuroendocrine tumors. N. Engl. J. Med. 2011, 364, 501–513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Valle, J.W.; Borbath, I.; Rosbrook, B.; Fernandez, K.; Raymond, E. Sunitinib in patients with pancreatic neuroendocrine tumors: Update of safety data. Future Oncol. 2019, 15. [Google Scholar] [CrossRef] [PubMed]
- Capozzi, M.; VON Arx, C.; DE Divitiis, C.; Ottaiano, A.; Tatangelo, F.; Romano, G.M.; Tafuto, S. Antiangiogenic Therapy in Pancreatic Neuroendocrine Tumors. Anticancer Res. 2016, 36, 5025–5030. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Berruti, A.; Fazio, N.; Ferrero, A.; Brizzi, M.P.; Volante, M.; Nobili, E.; Tozzi, L.; Bodei, L.; Torta, M.; D’Avolio, A.; et al. Bevacizumab plus octreotide and metronomic capecitabine in patients with metastatic well-to-moderately differentiated neuroendocrine tumors: The XELBEVOCT study. BMC Cancer 2014, 14, 184. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chan, J.A.; Stuart, K.; Earle, C.C.; Clark, J.W.; Bhargava, P.; Miksad, R.; Blaszkowsky, L.; Enzinger, P.C.; Meyerhardt, J.A.; Zheng, H.; et al. Prospective study of bevacizumab plus temozolomide in patients with advanced neuroendocrine tumors. J. Clin. Oncol. 2012, 30, 2963–2968. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Helfrich, I.; Scheffrahn, I.; Bartling, S.; Weis, J.; von Felbert, V.; Middleton, M.; Kato, M.; Ergun, S.; Augustin, H.G.; Schadendorf, D. Resistance to antiangiogenic therapy is directed by vascular phenotype, vessel stabilization, and maturation in malignant melanoma. J. Exp. Med. 2010, 207, 491–503. [Google Scholar] [CrossRef] [Green Version]
- Erber, R.; Thurnher, A.; Katsen, A.D.; Groth, G.; Kerger, H.; Hammes, H.P.; Menger, M.D.; Ullrich, A.; Vajkoczy, P. Combined inhibition of VEGF and PDGF signaling enforces tumor vessel regression by interfering with pericyte-mediated endothelial cell survival mechanisms. FASEB J. 2004, 18, 338–340. [Google Scholar] [CrossRef]
- Sitohy, B.; Nagy, J.A.; Dvorak, H.F. Anti-VEGF/VEGFR therapy for cancer: Reassessing the target. Cancer Res. 2012, 72, 1909–1914. [Google Scholar] [CrossRef] [Green Version]
- Uri, I.; Grozinsky-Glasberg, S. Current treatment strategies for patients with advanced gastroenteropancreatic neuroendocrine tumors (GEP-NETs). Clin. Diabetes Endocrinol. 2018, 4, 16. [Google Scholar] [CrossRef] [Green Version]
- Raymond, E.; Kulke, M.H.; Qin, S.; Yu, X.; Schenker, M.; Cubillo, A.; Lou, W.; Tomasek, J.; Thiis-Evensen, E.; Xu, J.M.; et al. Efficacy and Safety of Sunitinib in Patients with Well-Differentiated Pancreatic Neuroendocrine Tumours. Neuroendocrinology 2018, 107, 237–245. [Google Scholar] [CrossRef] [PubMed]
- Rinzivillo, M.; Fazio, N.; Pusceddu, S.; Spallanzani, A.; Ibrahim, T.; Campana, D.; Marconcini, R.; Partelli, S.; Badalamenti, G.; Brizzi, M.P.; et al. Sunitinib in patients with pre-treated pancreatic neuroendocrine tumors: A real-world study. Pancreatology 2018, 18, 198–203. [Google Scholar] [CrossRef] [PubMed]
- Yao, J.C.; Shah, M.H.; Ito, T.; Bohas, C.L.; Wolin, E.M.; Van Cutsem, E.; Hobday, T.J.; Okusaka, T.; Capdevila, J.; de Vries, E.G.; et al. Everolimus for advanced pancreatic neuroendocrine tumors. N. Engl. J. Med. 2011, 364, 514–523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baldelli, R.; Barnabei, A.; Rizza, L.; Isidori, A.M.; Rota, F.; Di Giacinto, P.; Paoloni, A.; Torino, F.; Corsello, S.M.; Lenzi, A.; et al. Somatostatin analogs therapy in gastroenteropancreatic neuroendocrine tumors: Current aspects and new perspectives. Front. Endocrinol. 2014, 5, 7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Caplin, M.E.; Pavel, M.; Cwikla, J.B.; Phan, A.T.; Raderer, M.; Sedlackova, E.; Cadiot, G.; Wolin, E.M.; Capdevila, J.; Wall, L.; et al. Lanreotide in metastatic enteropancreatic neuroendocrine tumors. N. Engl. J. Med. 2014, 371, 224–233. [Google Scholar] [CrossRef]
- Woltering, E.A.; Barrie, R.; O’Dorisio, T.M.; Arce, D.; Ure, T.; Cramer, A.; Holmes, D.; Robertson, J.; Fassler, J. Somatostatin analogues inhibit angiogenesis in the chick chorioallantoic membrane. J. Surg. Res. 1991, 50, 245–251. [Google Scholar] [CrossRef]
- Grozinsky-Glasberg, S.; Shimon, I.; Korbonits, M.; Grossman, A.B. Somatostatin analogues in the control of neuroendocrine tumours: Efficacy and mechanisms. Endocr. Relat. Cancer 2008, 15, 701–720. [Google Scholar] [CrossRef] [Green Version]
- Dasgupta, P. Somatostatin analogues: Multiple roles in cellular proliferation, neoplasia, and angiogenesis. Pharmacol. Ther. 2004, 102, 61–85. [Google Scholar] [CrossRef]
- Albini, A.; Florio, T.; Giunciuglio, D.; Masiello, L.; Carlone, S.; Corsaro, A.; Thellung, S.; Cai, T.; Noonan, D.M.; Schettini, G. Somatostatin controls Kaposi’s sarcoma tumor growth through inhibition of angiogenesis. FASEB J. 1999, 13, 647–655. [Google Scholar] [CrossRef] [Green Version]
- Mentlein, R.; Eichler, O.; Forstreuter, F.; Held-Feindt, J. Somatostatin inhibits the production of vascular endothelial growth factor in human glioma cells. Int. J. Cancer 2001, 92, 545–550. [Google Scholar] [CrossRef]
- Boehm, T.; Folkman, J.; Browder, T.; O’Reilly, M.S. Antiangiogenic therapy of experimental cancer does not induce acquired drug resistance. Nature 1997, 390, 404–407. [Google Scholar] [CrossRef] [PubMed]
- Mauceri, H.J.; Hanna, N.N.; Beckett, M.A.; Gorski, D.H.; Staba, M.J.; Stellato, K.A.; Bigelow, K.; Heimann, R.; Gately, S.; Dhanabal, M.; et al. Combined effects of angiostatin and ionizing radiation in antitumour therapy. Nature 1998, 394, 287–291. [Google Scholar] [CrossRef] [PubMed]
- Friedlander, M.; Brooks, P.C.; Shaffer, R.W.; Kincaid, C.M.; Varner, J.A.; Cheresh, D.A. Definition of two angiogenic pathways by distinct alpha v integrins. Science 1995, 270, 1500–1502. [Google Scholar] [CrossRef] [PubMed]
- Ferrara, N.; Chen, H.; Davis-Smyth, T.; Gerber, H.P.; Nguyen, T.N.; Peers, D.; Chisholm, V.; Hillan, K.J.; Schwall, R.H. Vascular endothelial growth factor is essential for corpus luteum angiogenesis. Nat. Med. 1998, 4, 336–340. [Google Scholar] [CrossRef] [PubMed]
- Han, Z.B.; Ren, H.; Zhao, H.; Chi, Y.; Chen, K.; Zhou, B.; Liu, Y.J.; Zhang, L.; Xu, B.; Liu, B.; et al. Hypoxia-inducible factor (HIF)-1 alpha directly enhances the transcriptional activity of stem cell factor (SCF) in response to hypoxia and epidermal growth factor (EGF). Carcinogenesis 2008, 29, 1853–1861. [Google Scholar] [CrossRef] [PubMed]
- Cooke, V.G.; LeBleu, V.S.; Keskin, D.; Khan, Z.; O’Connell, J.T.; Teng, Y.; Duncan, M.B.; Xie, L.; Maeda, G.; Vong, S.; et al. Pericyte depletion results in hypoxia-associated epithelial-to-mesenchymal transition and metastasis mediated by met signaling pathway. Cancer Cell 2012, 21, 66–81. [Google Scholar] [CrossRef] [Green Version]
- Paez-Ribes, M.; Allen, E.; Hudock, J.; Takeda, T.; Okuyama, H.; Vinals, F.; Inoue, M.; Bergers, G.; Hanahan, D.; Casanovas, O. Antiangiogenic therapy elicits malignant progression of tumors to increased local invasion and distant metastasis. Cancer Cell 2009, 15, 220–231. [Google Scholar] [CrossRef] [Green Version]
- Manegold, C.; Dingemans, A.C.; Gray, J.E.; Nakagawa, K.; Nicolson, M.; Peters, S.; Reck, M.; Wu, Y.L.; Brustugun, O.T.; Crino, L.; et al. The Potential of Combined Immunotherapy and Antiangiogenesis for the Synergistic Treatment of Advanced NSCLC. J. Thorac. Oncol. 2017, 12, 194–207. [Google Scholar] [CrossRef] [Green Version]
- Mack, J.J.; Iruela-Arispe, M.L. NOTCH regulation of the endothelial cell phenotype. Curr. Opin. Hematol. 2018, 25, 212–218. [Google Scholar] [CrossRef]
- Noguera-Troise, I.; Daly, C.; Papadopoulos, N.J.; Coetzee, S.; Boland, P.; Gale, N.W.; Lin, H.C.; Yancopoulos, G.D.; Thurston, G. Blockade of Dll4 inhibits tumour growth by promoting non-productive angiogenesis. Nature 2006, 444, 1032–1037. [Google Scholar] [CrossRef]
- Ridgway, J.; Zhang, G.; Wu, Y.; Stawicki, S.; Liang, W.C.; Chanthery, Y.; Kowalski, J.; Watts, R.J.; Callahan, C.; Kasman, I.; et al. Inhibition of Dll4 signalling inhibits tumour growth by deregulating angiogenesis. Nature 2006, 444, 1083–1087. [Google Scholar] [CrossRef] [PubMed]
- Scehnet, J.S.; Jiang, W.; Kumar, S.R.; Krasnoperov, V.; Trindade, A.; Benedito, R.; Djokovic, D.; Borges, C.; Ley, E.J.; Duarte, A.; et al. Inhibition of Dll4-mediated signaling induces proliferation of immature vessels and results in poor tissue perfusion. Blood 2007, 109, 4753–4760. [Google Scholar] [CrossRef] [PubMed]
- Low, S.; Barnes, J.L.; Zammit, P.S.; Beauchamp, J.R. Delta-Like 4 Activates Notch 3 to Regulate Self-Renewal in Skeletal Muscle Stem Cells. Stem Cells 2018, 36, 458–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takahashi, Y.; Akishima-Fukasawa, Y.; Kobayashi, N.; Sano, T.; Kosuge, T.; Nimura, Y.; Kanai, Y.; Hiraoka, N. Prognostic value of tumor architecture, tumor-associated vascular characteristics, and expression of angiogenic molecules in pancreatic endocrine tumors. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2007, 13, 187–196. [Google Scholar] [CrossRef]
- Tan, G.; Cioc, A.M.; Perez-Montiel, D.; Ellison, E.C.; Frankel, W.L. Microvascular density does not correlate with histopathology and outcome in neuroendocrine tumors of the pancreas. Appl. Immunohistochem. Mol. Morphol. 2004, 12, 31–35. [Google Scholar] [CrossRef]
- Laitakari, J.; Nayha, V.; Stenback, F. Size, shape, structure, and direction of angiogenesis in laryngeal tumour development. J. Clin. Pathol. 2004, 57, 394–401. [Google Scholar] [CrossRef] [Green Version]
- Nayha, V.V.; Stenback, F.G. Increased angiogenesis is associated with poor prognosis of squamous cell carcinoma of the vulva. Acta Obstet. Gynecol. Scand. 2007, 86, 1392–1397. [Google Scholar] [CrossRef]
- Crabtree, J.S.; Singleton, C.S.; Miele, L. Notch Signaling in Neuroendocrine Tumors. Front. Oncol. 2016, 6, 94. [Google Scholar] [CrossRef] [Green Version]
- Radtke, F.; Raj, K. The role of Notch in tumorigenesis: Oncogene or tumour suppressor? Nat. Rev. Cancer 2003, 3, 756–767. [Google Scholar] [CrossRef]
- Kunnimalaiyaan, M.; Chen, H. Tumor suppressor role of Notch-1 signaling in neuroendocrine tumors. Oncologist 2007, 12, 535–542. [Google Scholar] [CrossRef] [Green Version]
- Kunnimalaiyaan, M.; Yan, S.; Wong, F.; Zhang, Y.W.; Chen, H. Hairy Enhancer of Split-1 (HES-1), a Notch1 effector, inhibits the growth of carcinoid tumor cells. Surgery 2005, 138, 1137–1142. [Google Scholar] [CrossRef] [PubMed]
- Kunnimalaiyaan, M.; Traeger, K.; Chen, H. Conservation of the Notch1 signaling pathway in gastrointestinal carcinoid cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2005, 289, G636–G642. [Google Scholar] [CrossRef] [PubMed]
- Kunnimalaiyaan, M.; Vaccaro, A.M.; Ndiaye, M.A.; Chen, H. Overexpression of the NOTCH1 intracellular domain inhibits cell proliferation and alters the neuroendocrine phenotype of medullary thyroid cancer cells. J. Biol. Chem. 2006, 281, 39819–39830. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakakura, E.K.; Sriuranpong, V.R.; Kunnimalaiyaan, M.; Hsiao, E.C.; Schuebel, K.E.; Borges, M.W.; Jin, N.; Collins, B.J.; Nelkin, B.D.; Chen, H.; et al. Regulation of neuroendocrine differentiation in gastrointestinal carcinoid tumor cells by notch signaling. J. Clin. Endocrinol. Metab. 2005, 90, 4350–4356. [Google Scholar] [CrossRef] [Green Version]
- Krampitz, G.W.; George, B.M.; Willingham, S.B.; Volkmer, J.P.; Weiskopf, K.; Jahchan, N.; Newman, A.M.; Sahoo, D.; Zemek, A.J.; Yanovsky, R.L.; et al. Identification of tumorigenic cells and therapeutic targets in pancreatic neuroendocrine tumors. Proc. Natl. Acad. Sci. USA 2016, 113, 4464–4469. [Google Scholar] [CrossRef] [Green Version]
- Wang, H.; Chen, Y.; Fernandez-Del Castillo, C.; Yilmaz, O.; Deshpande, V. Heterogeneity in signaling pathways of gastroenteropancreatic neuroendocrine tumors: A critical look at notch signaling pathway. Mod. Pathol. 2013, 26, 139–147. [Google Scholar] [CrossRef] [Green Version]
- Krausch, M.; Kroepil, F.; Lehwald, N.; Lachenmayer, A.; Schott, M.; Anlauf, M.; Cupisti, K.; Knoefel, W.T.; Raffel, A. Notch 1 tumor expression is lacking in highly proliferative pancreatic neuroendocrine tumors. Endocrine 2013, 44, 182–186. [Google Scholar] [CrossRef]
- Eliasz, S.; Liang, S.; Chen, Y.; De Marco, M.A.; Machek, O.; Skucha, S.; Miele, L.; Bocchetta, M. Notch-1 stimulates survival of lung adenocarcinoma cells during hypoxia by activating the IGF-1R pathway. Oncogene 2010, 29, 2488–2498. [Google Scholar] [CrossRef] [Green Version]
- Sriuranpong, V.; Borges, M.W.; Strock, C.L.; Nakakura, E.K.; Watkins, D.N.; Blaumueller, C.M.; Nelkin, B.D.; Ball, D.W. Notch signaling induces rapid degradation of achaete-scute homolog 1. Mol. Cell Biol. 2002, 22, 3129–3139. [Google Scholar] [CrossRef] [Green Version]
- Jaskula-Sztul, R.; Eide, J.; Tesfazghi, S.; Dammalapati, A.; Harrison, A.D.; Yu, X.M.; Scheinebeck, C.; Winston-McPherson, G.; Kupcho, K.R.; Robers, M.B.; et al. Tumor-suppressor role of Notch3 in medullary thyroid carcinoma revealed by genetic and pharmacological induction. Mol. Cancer Ther. 2015, 14, 499–512. [Google Scholar] [CrossRef] [Green Version]
- Somnay, Y.R.; Yu, X.M.; Lloyd, R.V.; Leverson, G.; Aburjania, Z.; Jang, S.; Jaskula-Sztul, R.; Chen, H. Notch3 expression correlates with thyroid cancer differentiation, induces apoptosis, and predicts disease prognosis. Cancer 2017, 123, 769–782. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lou, I.; Odorico, S.; Yu, X.M.; Harrison, A.; Jaskula-Sztul, R.; Chen, H. Notch3 as a novel therapeutic target in metastatic medullary thyroid cancer. Surgery 2018, 163, 104–111. [Google Scholar] [CrossRef] [PubMed]
- Zhou, M.; Jin, W.Y.; Fan, Z.W.; Han, R.C. Analysis of the expression of the Notch3 receptor protein in adult lung cancer. Oncol. Lett. 2013, 5, 499–504. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ito, T.; Udaka, N.; Yazawa, T.; Okudela, K.; Hayashi, H.; Sudo, T.; Guillemot, F.; Kageyama, R.; Kitamura, H. Basic helix-loop-helix transcription factors regulate the neuroendocrine differentiation of fetal mouse pulmonary epithelium. Development 2000, 127, 3913–3921. [Google Scholar] [PubMed]
- Marcucci, F.; Caserta, C.A.; Romeo, E.; Rumio, C. Antibody-Drug Conjugates (ADC) Against Cancer Stem-Like Cells (CSC)-Is There Still Room for Optimism? Front. Oncol. 2019, 9, 167. [Google Scholar] [CrossRef] [Green Version]
- Lashari, B.H.; Vallatharasu, Y.; Kolandra, L.; Hamid, M.; Uprety, D. Rovalpituzumab Tesirine: A Novel DLL3-Targeting Antibody-Drug Conjugate. Drugs R D 2018, 18, 255–258. [Google Scholar] [CrossRef] [Green Version]
- Saunders, L.R.; Bankovich, A.J.; Anderson, W.C.; Aujay, M.A.; Bheddah, S.; Black, K.; Desai, R.; Escarpe, P.A.; Hampl, J.; Laysang, A.; et al. A DLL3-targeted antibody-drug conjugate eradicates high-grade pulmonary neuroendocrine tumor-initiating cells in vivo. Sci. Transl. Med. 2015, 7, 302ra136. [Google Scholar] [CrossRef] [Green Version]
- Kuhnert, F.; Kirshner, J.R.; Thurston, G. Dll4-Notch signaling as a therapeutic target in tumor angiogenesis. Vasc. Cell 2011, 3, 20. [Google Scholar] [CrossRef] [Green Version]
- Driessens, G.; Beck, B.; Caauwe, A.; Simons, B.D.; Blanpain, C. Defining the mode of tumour growth by clonal analysis. Nature 2012, 488, 527–530. [Google Scholar] [CrossRef] [Green Version]
- Schepers, A.G.; Snippert, H.J.; Stange, D.E.; van den Born, M.; van Es, J.H.; van de Wetering, M.; Clevers, H. Lineage tracing reveals Lgr5+ stem cell activity in mouse intestinal adenomas. Science 2012, 337, 730–735. [Google Scholar] [CrossRef] [Green Version]
- Kozar, S.; Morrissey, E.; Nicholson, A.M.; van der Heijden, M.; Zecchini, H.I.; Kemp, R.; Tavare, S.; Vermeulen, L.; Winton, D.J. Continuous clonal labeling reveals small numbers of functional stem cells in intestinal crypts and adenomas. Cell Stem Cell 2013, 13, 626–633. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zomer, A.; Ellenbroek, S.I.; Ritsma, L.; Beerling, E.; Vrisekoop, N.; Van Rheenen, J. Intravital imaging of cancer stem cell plasticity in mammary tumors. Stem Cells 2013, 31, 602–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, J.; Li, Y.; Yu, T.S.; McKay, R.M.; Burns, D.K.; Kernie, S.G.; Parada, L.F. A restricted cell population propagates glioblastoma growth after chemotherapy. Nature 2012, 488, 522–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oshimori, N.; Oristian, D.; Fuchs, E. TGF-beta promotes heterogeneity and drug resistance in squamous cell carcinoma. Cell 2015, 160, 963–976. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Batlle, E.; Clevers, H. Cancer stem cells revisited. Nat. Med. 2017, 23, 1124–1134. [Google Scholar] [CrossRef] [PubMed]
- Mathis, R.A.; Sokol, E.S.; Gupta, P.B. Cancer cells exhibit clonal diversity in phenotypic plasticity. Open Biol. 2017, 7. [Google Scholar] [CrossRef]
- Gupta, P.B.; Fillmore, C.M.; Jiang, G.; Shapira, S.D.; Tao, K.; Kuperwasser, C.; Lander, E.S. Stochastic state transitions give rise to phenotypic equilibrium in populations of cancer cells. Cell 2011, 146, 633–644. [Google Scholar] [CrossRef] [Green Version]
- Hoeck, J.D.; Biehs, B.; Kurtova, A.V.; Kljavin, N.M.; de Sousa, E.M.F.; Alicke, B.; Koeppen, H.; Modrusan, Z.; Piskol, R.; de Sauvage, F.J. Stem cell plasticity enables hair regeneration following Lgr5(+) cell loss. Nat. Cell Biol. 2017, 19, 666–676. [Google Scholar] [CrossRef]
- Lenos, K.J.; Miedema, D.M.; Lodestijn, S.C.; Nijman, L.E.; van den Bosch, T.; Romero Ros, X.; Lourenco, F.C.; Lecca, M.C.; van der Heijden, M.; van Neerven, S.M.; et al. Stem cell functionality is microenvironmentally defined during tumour expansion and therapy response in colon cancer. Nat. Cell Biol. 2018, 20, 1193–1202. [Google Scholar] [CrossRef]
- de Sousa e Melo, F.; Kurtova, A.V.; Harnoss, J.M.; Kljavin, N.; Hoeck, J.D.; Hung, J.; Anderson, J.E.; Storm, E.E.; Modrusan, Z.; Koeppen, H.; et al. A distinct role for Lgr5(+) stem cells in primary and metastatic colon cancer. Nature 2017, 543, 676–680. [Google Scholar] [CrossRef]
- Drost, J.; van Boxtel, R.; Blokzijl, F.; Mizutani, T.; Sasaki, N.; Sasselli, V.; de Ligt, J.; Behjati, S.; Grolleman, J.E.; van Wezel, T.; et al. Use of CRISPR-modified human stem cell organoids to study the origin of mutational signatures in cancer. Science 2017, 358, 234–238. [Google Scholar] [CrossRef] [Green Version]
- Clevers, H. STEM CELLS. What is an adult stem cell? Science 2015, 350, 1319–1320. [Google Scholar] [CrossRef]
- Ball, C.R.; Oppel, F.; Ehrenberg, K.R.; Dubash, T.D.; Dieter, S.M.; Hoffmann, C.M.; Abel, U.; Herbst, F.; Koch, M.; Werner, J.; et al. Succession of transiently active tumor-initiating cell clones in human pancreatic cancer xenografts. EMBO Mol. Med. 2017, 9, 918–932. [Google Scholar] [CrossRef] [Green Version]
- McGranahan, N.; Swanton, C. Clonal Heterogeneity and Tumor Evolution: Past, Present, and the Future. Cell 2017, 168, 613–628. [Google Scholar] [CrossRef]
- Jagust, P.; de Luxan-Delgado, B.; Parejo-Alonso, B.; Sancho, P. Metabolism-Based Therapeutic Strategies Targeting Cancer Stem Cells. Front. Pharmacol. 2019, 10, 203. [Google Scholar] [CrossRef] [Green Version]
- Ryall, J.G.; Dell’Orso, S.; Derfoul, A.; Juan, A.; Zare, H.; Feng, X.; Clermont, D.; Koulnis, M.; Gutierrez-Cruz, G.; Fulco, M.; et al. The NAD(+)-dependent SIRT1 deacetylase translates a metabolic switch into regulatory epigenetics in skeletal muscle stem cells. Cell Stem Cell 2015, 16, 171–183. [Google Scholar] [CrossRef] [Green Version]
- Buffie, C.G.; Bucci, V.; Stein, R.R.; McKenney, P.T.; Ling, L.; Gobourne, A.; No, D.; Liu, H.; Kinnebrew, M.; Viale, A.; et al. Precision microbiome reconstitution restores bile acid mediated resistance to Clostridium difficile. Nature 2015, 517, 205–208. [Google Scholar] [CrossRef] [Green Version]
- Viale, A.; Pettazzoni, P.; Lyssiotis, C.A.; Ying, H.; Sanchez, N.; Marchesini, M.; Carugo, A.; Green, T.; Seth, S.; Giuliani, V.; et al. Oncogene ablation-resistant pancreatic cancer cells depend on mitochondrial function. Nature 2014, 514, 628–632. [Google Scholar] [CrossRef] [Green Version]
- Sancho, P.; Burgos-Ramos, E.; Tavera, A.; Bou Kheir, T.; Jagust, P.; Schoenhals, M.; Barneda, D.; Sellers, K.; Campos-Olivas, R.; Grana, O.; et al. MYC/PGC-1alpha Balance Determines the Metabolic Phenotype and Plasticity of Pancreatic Cancer Stem Cells. Cell Metab. 2015, 22, 590–605. [Google Scholar] [CrossRef] [Green Version]
- Barnekow, N.; Yuan, R.; Moran, P.; Ren, B. Abstract 16740: Foxo1-Activated Cd36 Transcription Switches Arteriolar Differentiation of Endothelial Cells. Circulation 2018, 138, A16740. [Google Scholar] [CrossRef]
- Ladanyi, A.; Mukherjee, A.; Kenny, H.A.; Johnson, A.; Mitra, A.K.; Sundaresan, S.; Nieman, K.M.; Pascual, G.; Benitah, S.A.; Montag, A.; et al. Adipocyte-induced CD36 expression drives ovarian cancer progression and metastasis. Oncogene 2018, 37, 2285–2301. [Google Scholar] [CrossRef]
- Ye, H.; Adane, B.; Khan, N.; Sullivan, T.; Minhajuddin, M.; Gasparetto, M.; Stevens, B.; Pei, S.; Balys, M.; Ashton, J.M.; et al. Leukemic Stem Cells Evade Chemotherapy by Metabolic Adaptation to an Adipose Tissue Niche. Cell Stem Cell 2016, 19, 23–37. [Google Scholar] [CrossRef] [Green Version]
- Fumagalli, A.; Drost, J.; Suijkerbuijk, S.J.; van Boxtel, R.; de Ligt, J.; Offerhaus, G.J.; Begthel, H.; Beerling, E.; Tan, E.H.; Sansom, O.J.; et al. Genetic dissection of colorectal cancer progression by orthotopic transplantation of engineered cancer organoids. Proc. Natl. Acad. Sci. USA 2017, 114, E2357–E2364. [Google Scholar] [CrossRef] [Green Version]
- Fujii, M.; Shimokawa, M.; Date, S.; Takano, A.; Matano, M.; Nanki, K.; Ohta, Y.; Toshimitsu, K.; Nakazato, Y.; Kawasaki, K.; et al. A Colorectal Tumor Organoid Library Demonstrates Progressive Loss of Niche Factor Requirements during Tumorigenesis. Cell Stem Cell 2016, 18, 827–838. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.; Yalcin, S.; Lee, D.F.; Yeh, T.Y.; Lee, S.M.; Su, J.; Mungamuri, S.K.; Rimmele, P.; Kennedy, M.; Sellers, R.; et al. FOXO1 is an essential regulator of pluripotency in human embryonic stem cells. Nat. Cell Biol. 2011, 13, 1092–1099. [Google Scholar] [CrossRef]
- Tothova, Z.; Gilliland, D.G. FoxO transcription factors and stem cell homeostasis: Insights from the hematopoietic system. Cell Stem Cell 2007, 1, 140–152. [Google Scholar] [CrossRef] [Green Version]
- Sykes, S.M.; Lane, S.W.; Bullinger, L.; Kalaitzidis, D.; Yusuf, R.; Saez, B.; Ferraro, F.; Mercier, F.; Singh, H.; Brumme, K.M.; et al. AKT/FOXO signaling enforces reversible differentiation blockade in myeloid leukemias. Cell 2011, 146, 697–708. [Google Scholar] [CrossRef] [Green Version]
- Paik, J.H.; Kollipara, R.; Chu, G.; Ji, H.; Xiao, Y.; Ding, Z.; Miao, L.; Tothova, Z.; Horner, J.W.; Carrasco, D.R.; et al. FoxOs are lineage-restricted redundant tumor suppressors and regulate endothelial cell homeostasis. Cell 2007, 128, 309–323. [Google Scholar] [CrossRef] [Green Version]
- Wilhelm, K.; Happel, K.; Eelen, G.; Schoors, S.; Oellerich, M.F.; Lim, R.; Zimmermann, B.; Aspalter, I.M.; Franco, C.A.; Boettger, T.; et al. FOXO1 couples metabolic activity and growth state in the vascular endothelium. Nature 2016, 529, 216–220. [Google Scholar] [CrossRef] [Green Version]
- Carrasco, P.; Zuazo-Gaztelu, I.; Casanovas, O. Sprouting strategies and dead ends in anti-angiogenic targeting of NETs. J. Mol. Endocrinol. 2017, 59, R77–R91. [Google Scholar] [CrossRef] [Green Version]
- Chu, X.; Gao, X.; Jansson, L.; Quach, M.; Skogseid, B.; Barbu, A. Multiple microvascular alterations in pancreatic islets and neuroendocrine tumors of a Men1 mouse model. Am. J. Pathol. 2013, 182, 2355–2367. [Google Scholar] [CrossRef]
- Wagenblast, E.; Soto, M.; Gutierrez-Angel, S.; Hartl, C.A.; Gable, A.L.; Maceli, A.R.; Erard, N.; Williams, A.M.; Kim, S.Y.; Dickopf, S.; et al. A model of breast cancer heterogeneity reveals vascular mimicry as a driver of metastasis. Nature 2015, 520, 358–362. [Google Scholar] [CrossRef] [Green Version]
- Hendrix, M.J.; Seftor, E.A.; Seftor, R.E.; Chao, J.T.; Chien, D.S.; Chu, Y.W. Tumor cell vascular mimicry: Novel targeting opportunity in melanoma. Pharmacol. Ther. 2016, 159, 83–92. [Google Scholar] [CrossRef] [Green Version]
- Zhou, L.; Liu, X.D.; Sun, M.; Zhang, X.; German, P.; Bai, S.; Ding, Z.; Tannir, N.; Wood, C.G.; Matin, S.F.; et al. Targeting MET and AXL overcomes resistance to sunitinib therapy in renal cell carcinoma. Oncogene 2016, 35, 2687–2697. [Google Scholar] [CrossRef]
- Capdevila, J.; Fazio, N.; López-López, C.; Teule, A.; Valle, J.W.; Tafuto, S.; Custodio, A.B.; Reed, N.; Raderer, M.; Grande, E.; et al. Progression-free survival (PFS) and subgroups analyses of lenvatinib in patients (pts) with G1/G2 advanced pancreatic (panNETs) and gastrointestinal (giNETs) neuroendocrine tumors (NETs): Updated results from the phase II TALENT trial (GETNE 1509). J. Clin. Oncol. 2019, 37, 332. [Google Scholar] [CrossRef]
- Capozzi, M.; De Divitiis, C.; Ottaiano, A.; von Arx, C.; Scala, S.; Tatangelo, F.; Delrio, P.; Tafuto, S. Lenvatinib, a molecule with versatile application: From preclinical evidence to future development in anti-cancer treatment. Cancer Manag. Res. 2019, 11, 3847–3860. [Google Scholar] [CrossRef] [Green Version]
- Grande, E.; Lopez, C.; Alonso-Gordoa, T.; Benavent, M.; Capdevila, J.; Teule, A.; Custodio, A.; Sevilla, I.; Gajate, P.; Molina-Cerrillo, J.; et al. The SUNEVO (GETNE-1408) trial to evaluate the activity and safety of thecombination of sunitinib with evofosfamide (TH-302) in patients with G1/G2 metastatic pancreatic neuroendocrine tumours (pNETs) naïve forsystemic treatment: A phase II study of the Spanish Task Force Group for Neuroendocrine and Endocrine Tumors (GETNE). J. Clin. Oncol. 2019, 37, 4105. [Google Scholar] [CrossRef]
- Ikezono, Y.; Koga, H.; Akiba, J.; Abe, M.; Yoshida, T.; Wada, F.; Nakamura, T.; Iwamoto, H.; Masuda, A.; Sakaue, T.; et al. Pancreatic Neuroendocrine Tumors and EMT Behavior Are Driven by the CSC Marker DCLK1. Mol. Cancer Res. 2017, 15, 744–752. [Google Scholar] [CrossRef] [Green Version]
- Rigamonti, N.; Kadioglu, E.; Keklikoglou, I.; Wyser Rmili, C.; Leow, C.C.; De Palma, M. Role of angiopoietin-2 in adaptive tumor resistance to VEGF signaling blockade. Cell Rep. 2014, 8, 696–706. [Google Scholar] [CrossRef] [Green Version]
- Biel, N.M.; Siemann, D.W. Targeting the Angiopoietin-2/Tie-2 axis in conjunction with VEGF signal interference. Cancer Lett. 2016, 380, 525–533. [Google Scholar] [CrossRef] [Green Version]
- Maione, F.; Capano, S.; Regano, D.; Zentilin, L.; Giacca, M.; Casanovas, O.; Bussolino, F.; Serini, G.; Giraudo, E. Semaphorin 3A overcomes cancer hypoxia and metastatic dissemination induced by antiangiogenic treatment in mice. J. Clin. Invest. 2012, 122, 1832–1848. [Google Scholar] [CrossRef] [Green Version]
- Allen, E.; Walters, I.B.; Hanahan, D. Brivanib, a dual FGF/VEGF inhibitor, is active both first and second line against mouse pancreatic neuroendocrine tumors developing adaptive/evasive resistance to VEGF inhibition. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2011, 17, 5299–5310. [Google Scholar] [CrossRef]
- Hilfenhaus, G.; Gohrig, A.; Pape, U.F.; Neumann, T.; Jann, H.; Zdunek, D.; Hess, G.; Stassen, J.M.; Wiedenmann, B.; Detjen, K.; et al. Placental growth factor supports neuroendocrine tumor growth and predicts disease prognosis in patients. Endocr. Relat. Cancer 2013, 20, 305–319. [Google Scholar] [CrossRef] [Green Version]
- Fischer, C.; Jonckx, B.; Mazzone, M.; Zacchigna, S.; Loges, S.; Pattarini, L.; Chorianopoulos, E.; Liesenborghs, L.; Koch, M.; De Mol, M.; et al. Anti-PlGF inhibits growth of VEGF(R)-inhibitor-resistant tumors without affecting healthy vessels. Cell 2007, 131, 463–475. [Google Scholar] [CrossRef] [Green Version]
- Marconcini, R.; Faviana, P.; Campani, D.; Galli, L.; Antonuzzo, A.; Orlandini, C.; Falcone, A.; Ricci, S. Enhancer of zest homolog 2 (EZH2) expression in well and moderately differentiated pancreatic neuroendocrine tumor (pNET). Ann. Oncol. 2016, 27. [Google Scholar] [CrossRef]
- Yang, L.; Yu, X.; Yang, Y. Autotaxin upregulated by STAT3 activation contributes to invasion in pancreatic neuroendocrine neoplasms. Endocr. Connect. 2018, 7, 1299–1307. [Google Scholar] [CrossRef] [Green Version]
- Chen, Y.; Ramakrishnan, D.P.; Ren, B. Regulation of angiogenesis by phospholipid lysophosphatidic acid. Front. Biosci. (Landmark Ed) 2013, 18, 852–861. [Google Scholar]
- Liu, S.; Umezu-Goto, M.; Murph, M.; Lu, Y.; Liu, W.; Zhang, F.; Yu, S.; Stephens, L.C.; Cui, X.; Murrow, G.; et al. Expression of autotaxin and lysophosphatidic acid receptors increases mammary tumorigenesis, invasion, and metastases. Cancer Cell 2009, 15, 539–550. [Google Scholar] [CrossRef] [Green Version]
- Liu, Y.; An, S.; Ward, R.; Yang, Y.; Guo, X.X.; Li, W.; Xu, T.R. G protein-coupled receptors as promising cancer targets. Cancer Lett. 2016, 376, 226–239. [Google Scholar] [CrossRef]
- Zecchin, A.; Kalucka, J.; Dubois, C.; Carmeliet, P. How Endothelial Cells Adapt Their Metabolism to Form Vessels in Tumors. Front. Immunol. 2017, 8, 1750. [Google Scholar] [CrossRef] [Green Version]
Name | Mechanisms & Functions | Reference(s) |
---|---|---|
CVM-1118 | Phase II clinical trial for patients with advanced NETs, including pNETs; de novo development of vascular networks via vascular mimicry, which is associated with a malignant phenotype and a poor clinical outcome. Vascular mimicry was presented in pNET animal models. | https://clinicaltrials.gov/ct2/show/NCT03600233; Chu et al., Am J Pathol 2013; Wagenblast, et al., Nature 2015; Hendrix, et al., Pharmacol Ther 2016 [211,212,213] |
Cabozantinib | Phase III clinical trial of in patients with advanced neuroendocrine tumors after progression on everolimus (CABINET) A VEGFR, c-MET and AXL inhibitor; attenuate sunitinib therapy-mediated pro-metastasis in xenograft mouse tumor models of renal cell carcinoma. | https://clinicaltrials.gov/ct2/show/NCT03375320; Zhou et al., Oncogene, 2015 [214] |
Lenvatinib | A multi-kinase inhibitor with a preferential antiangiogenic activity. | Capdevila, et al. J Clin Oncol, 2019; Capozzi, et al., Cancer Manag Res 2019 [215,216] |
TH-302 (evofosfamide) | A hypoxia-activated prodrug, which is metabolized to its active form, bromo-isophosphoramide mustard (Br-IPM), under hypoxic conditions. Used in combination with sunitinib. | Grande, et al., abstract. J Clin Oncol, 2019 [217] |
Doublecortin-like kinase 1 (DCLK1) | A potential marker for pNET CSCs; induce epithelial-mesenchymal transition (EMT). | Ikezono, et al., Mol Cancer Res, 2017 [218] |
MEDI3617 | A monoclonal antibody targeting angiopoietin-2; in combination with VEGF-targeted therapies. | Rigamonti, et al., Cell Rep 2014; Biel, et al., Cancer Letters, 2016 [219,220] |
Sema3A (Semaphorin 3A) | Overcome cancer hypoxia and metastatic dissemination induced by sunitinib treatment in mice. In combination with sunitinib, Sema3A synergistically enhanced RIP-Tag2 mouse survival. | Maione, F, et al., J Clin Invest, 2012 [221] |
Brivanib | A dual FGF/VEGF inhibitor; vascular inhibition and tumor stability. | Allen, et al., Clin Cancer Res, 2011 [222] |
PlGF signaling inhibitor? | High PIGF expression in pNET patients with poor outcome; overcome resistance to antiangiogenic factors. | Fischer et al., Cell, 2007; Hilfenhaus, et al., Endocr Relat Cancer, 2013 [223,224] |
EZH2 inhibitor? | Sensitize tumor to sunitinib in cell lines and PDX models. | Marconcini, et al., 2016 [225] |
Autotaxin (ATX) inhibitors? LPA receptor-specific antagonists? | High ATX expression in pNET tissues, which was associated with higher tumor grade, TNM staging and lymph node metastasis; ATX drives LPA expression, which is also linked to tumor angiogenesis and arteriolar differentiation and malignancy of tumor cells. | Liu et al., Cancer Cell, 2009; Chen, et al., Front Biosci (Landmark Ed) 2013; Ren, et al., Arterioscler Thromb Vasc Biol, 2016; Liu et al., Cancer Lett 2016; Dong et al., Oncotarget, 2017 [20]; Yang et al., Endocr. Connect, 2018 [21,226,227,228,229] |
© 2019 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 (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Ren, B.; Rose, J.B.; Liu, Y.; Jaskular-Sztul, R.; Contreras, C.; Beck, A.; Chen, H. Heterogeneity of Vascular Endothelial Cells, De Novo Arteriogenesis and Therapeutic Implications in Pancreatic Neuroendocrine Tumors. J. Clin. Med. 2019, 8, 1980. https://doi.org/10.3390/jcm8111980
Ren B, Rose JB, Liu Y, Jaskular-Sztul R, Contreras C, Beck A, Chen H. Heterogeneity of Vascular Endothelial Cells, De Novo Arteriogenesis and Therapeutic Implications in Pancreatic Neuroendocrine Tumors. Journal of Clinical Medicine. 2019; 8(11):1980. https://doi.org/10.3390/jcm8111980
Chicago/Turabian StyleRen, Bin, J. Bart Rose, Yehe Liu, Renata Jaskular-Sztul, Carlo Contreras, Adam Beck, and Herbert Chen. 2019. "Heterogeneity of Vascular Endothelial Cells, De Novo Arteriogenesis and Therapeutic Implications in Pancreatic Neuroendocrine Tumors" Journal of Clinical Medicine 8, no. 11: 1980. https://doi.org/10.3390/jcm8111980
APA StyleRen, B., Rose, J. B., Liu, Y., Jaskular-Sztul, R., Contreras, C., Beck, A., & Chen, H. (2019). Heterogeneity of Vascular Endothelial Cells, De Novo Arteriogenesis and Therapeutic Implications in Pancreatic Neuroendocrine Tumors. Journal of Clinical Medicine, 8(11), 1980. https://doi.org/10.3390/jcm8111980