Blood Vessels and Peripheral Nerves as Key Players in Cancer Progression and Therapy Resistance
Abstract
:Simple Summary
Abstract
1. Introduction: The Heterogeneous Microenvironment of Tumors
2. The Role of Blood Vessels and Peripheral Nerves in Tumor Initiation
3. Angiogenesis and Axonogenesis
4. The Role of Blood Vessels and Peripheral Nerves in Local Tumor Progression
5. Blood Vessels and Peripheral Nerves in Metastatic Progression
6. Blood Vessels and Peripheral Nerves in Cancer Treatment
6.1. Blood Vessels
6.2. Peripheral Nerves
7. Concluding Remarks
Author Contributions
Funding
Conflicts of Interest
References
- Cao, Y. Tumorigenesis as a process of gradual loss of original cell identity and gain of properties of neural precursor/progenitor cells. Cell Biosci. 2017, 7, 61. [Google Scholar] [CrossRef] [Green Version]
- Hanahan, D.; Weinberg, R.A. Hallmarks of Cancer: The Next Generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Roda, N.; Gambino, V.; Giorgio, M. Metabolic Constrains Rule Metastasis Progression. Cells 2020, 9, 2081. [Google Scholar] [CrossRef]
- Belli, C.; Trapani, D.; Viale, G.; D’Amico, P.; Duso, B.A.; Della Vigna, P.; Orsi, F.; Curigliano, G. Targeting the microenvironment in solid tumors. Cancer Treat. Rev. 2018, 65, 22–32. [Google Scholar] [CrossRef] [PubMed]
- Baghban, R.; Roshangar, L.; Jahanban-Esfahlan, R.; Seidi, K.; Ebrahimi-Kalan, A.; Jaymand, M.; Kolahian, S.; Javaheri, T.; Zare, P. Tumor microenvironment complexity and therapeutic implications at a glance. Cell Commun. Signal. 2020, 18, 59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Balkwill, F.R.; Capasso, M.; Hagemann, T. The tumor microenvironment at a glance. J. Cell Sci. 2012, 125, 5591–5596. [Google Scholar] [CrossRef] [Green Version]
- Jin, M.-Z.; Jin, W.-L. The updated landscape of tumor microenvironment and drug repurposing. Signal Transduct. Target. Ther. 2020, 5, 166. [Google Scholar] [CrossRef]
- Hanahan, D.; Coussens, L.M. Accessories to the Crime: Functions of Cells Recruited to the Tumor Microenvironment. Cancer Cell 2012, 21, 309–322. [Google Scholar] [CrossRef] [Green Version]
- Brassart-Pasco, S.; Brézillon, S.; Brassart, B.; Ramont, L.; Oudart, J.-B.; Monboisse, J.C. Tumor Microenvironment: Extracellular Matrix Alterations Influence Tumor Progression. Front. Oncol. 2020, 10, 397. [Google Scholar] [CrossRef] [Green Version]
- Henke, E.; Nandigama, R.; Ergün, S. Extracellular Matrix in the Tumor Microenvironment and Its Impact on Cancer Therapy. Front. Mol. Biosci. 2020, 6, 160. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deryugina, E.I.; Quigley, J.P. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 2006, 25, 9–34. [Google Scholar] [CrossRef] [PubMed]
- Jabłońska-Trypuć, A.; Matejczyk, M.; Rosochacki, S. Matrix metalloproteinases (MMPs), the main extracellular matrix (ECM) enzymes in collagen degradation, as a target for anticancer drugs. J. Enzym. Inhib. Med. Chem. 2016, 31, 177–183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yoon, S.-O.; Park, S.-J.; Yun, C.-H.; Chung, A.-S. Roles of Matrix Metalloproteinases in Tumor Metastasis and Angiogenesis. BMB Rep. 2003, 36, 128–137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Monteran, L.; Erez, N. The Dark Side of Fibroblasts: Cancer-Associated Fibroblasts as Mediators of Immunosuppression in the Tumor Microenvironment. Front. Immunol. 2019, 10, 1835. [Google Scholar] [CrossRef] [Green Version]
- Sahai, E.; Astsaturov, I.; Cukierman, E.; DeNardo, D.G.; Egeblad, M.; Evans, R.; Fearon, D.; Greten, F.R.; Hingorani, S.R.; Hunter, T.; et al. A framework for advancing our understanding of cancer-associated fibroblasts. Nat. Rev. Cancer 2020, 20, 174–186. [Google Scholar] [CrossRef] [Green Version]
- Xing, F. Cancer associated fibroblasts (CAFs) in tumor microenvironment. Front. Biosci. 2010, 15, 166–179. [Google Scholar] [CrossRef] [Green Version]
- Najafi, M.; Farhood, B.; Mortezaee, K. Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J. Cell. Biochem. 2019, 120, 2782–2790. [Google Scholar] [CrossRef]
- Bauer, M.; Su, G.; Casper, C.; He, R.; Rehrauer, W.; Friedl, A. Heterogeneity of gene expression in stromal fibroblasts of human breast carcinomas and normal breast. Oncogene 2010, 29, 1732–1740. [Google Scholar] [CrossRef] [Green Version]
- Yavuz, B.G.; Gunaydin, G.; Gedik, M.E.; Kosemehmetoglu, K.; Karakoc, D.; Ozgür, F.F.; Guc, D. Cancer associated fibroblasts sculpt tumour microenvironment by recruiting monocytes and inducing immunosuppressive PD-1+ TAMs. Sci. Rep. 2019, 9, 3172. [Google Scholar] [CrossRef]
- Cohen, N.; Shani, O.; Raz, Y.; Sharon, Y.; Hoffman, D.; Abramovitz, L.; Erez, N. Fibroblasts drive an immunosuppressive and growth-promoting microenvironment in breast cancer via secretion of Chitinase 3-like 1. Oncogene 2017, 36, 4457–4468. [Google Scholar] [CrossRef] [Green Version]
- Öhlund, D.; Handly-Santana, A.; Biffi, G.; Elyada, E.; Almeida, A.S.; Ponz-Sarvise, M.; Corbo, V.; Oni, T.E.; Hearn, S.A.; Lee, E.J.; et al. Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J. Exp. Med. 2017, 214, 579–596. [Google Scholar] [CrossRef]
- Biffi, G.; Oni, T.E.; Spielman, B.; Hao, Y.; Elyada, E.; Park, Y.; Preall, J.; Tuveson, D.A. IL1-Induced JAK/STAT Signaling Is Antagonized by TGFβ to Shape CAF Heterogeneity in Pancreatic Ductal Adenocarcinoma. Cancer Discov. 2019, 9, 282–301. [Google Scholar] [CrossRef] [Green Version]
- Hao, N.-B.; Lü, M.-H.; Fan, Y.-H.; Cao, Y.-L.; Zhang, Z.-R.; Yang, S.-M. Macrophages in Tumor Microenvironments and the Progression of Tumors. Clin. Dev. Immunol. 2012, 2012, 948098. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hendry, S.; Salgado, R.; Gevaert, T.; Russell, P.A.; John, T.; Thapa, B.; Christie, M.; van de Vijver, K.; Estrada, M.V.; Ericsson, P.G.; et al. Assessing Tumor-infiltrating Lymphocytes in Solid Tumors: A Practical Review for Pathologists and Proposal for a Standardized Method from the International Immunooncology Biomarkers Working Group: Part 1: Assessing the Host Immune Response, TILs in Invasive Breast Carcinoma and Ductal Carcinoma in Situ, Metastatic Tumor Deposits and Areas for Further Research. Adv. Anat. Pathol. 2017, 24, 235–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hendry, S.; Salgado, R.; Gevaert, T.; Russell, P.A.; John, T.; Thapa, B.; Christie, M.; van de Vijver, K.; Estrada, M.V.; Ericsson, P.G.; et al. Assessing Tumor-Infiltrating Lymphocytes in Solid Tumors: A Practical Review for Pathologists and Proposal for a Standardized Method from the International Immuno-Oncology Biomarkers Working Group: Part 2: TILs in Melanoma, Gastrointestinal Tract Carcinomas, Non–Small Cell Lung Carcinoma and Mesothelioma, Endometrial and Ovarian Carcinomas, Squamous Cell Carcinoma of the Head and Neck, Genitourinary Carcinomas, and Primary Brain Tumors. Adv. Anat. Pathol. 2017, 24, 311–335. [Google Scholar] [CrossRef]
- Lin, Y.; Xu, J.; Lan, H. Tumor-associated macrophages in tumor metastasis: Biological roles and clinical therapeutic applications. J. Hematol. Oncol. 2019, 12, 76. [Google Scholar] [CrossRef] [PubMed]
- Wang, F.; Zhang, S.; Jeon, R.; Vuckovic, I.; Jiang, X.; Lerman, A.; Folmes, C.; Dzeja, P.D.; Herrmann, J. Interferon Gamma Induces Reversible Metabolic Reprogramming of M1 Macrophages to Sustain Cell Viability and Pro-Inflammatory Activity. EBioMedicine 2018, 30, 303–316. [Google Scholar] [CrossRef] [Green Version]
- Mantovani, A.; Sozzani, S.; Locati, M.; Allavena, P.; Sica, A. Macrophage polarization: Tumor-associated macrophages as a paradigm for polarized M2 mononuclear phagocytes. Trends Immunol. 2002, 23, 549–555. [Google Scholar] [CrossRef]
- Martinez, V.M.G.; Rubio, C.; Martínez-Fernández, M.; Segovia, C.; López-Calderón, F.; Garin, M.I.; Teijeira, A.; Munera-Maravilla, E.; Varas, A.; Sacedón, R.; et al. BMP4 Induces M2 Macrophage Polarization and Favors Tumor Progression in Bladder Cancer. Clin. Cancer Res. 2017, 23, 7388–7399. [Google Scholar] [CrossRef] [Green Version]
- Sumitomo, R.; Hirai, T.; Fujita, M.; Murakami, H.; Otake, Y.; Huang, C. M2 tumor-associated macrophages promote tumor progression in non-small-cell lung cancer. Exp. Ther. Med. 2019, 18, 4490–4498. [Google Scholar] [CrossRef] [Green Version]
- Wagner, J.; Rapsomaniki, M.A.; Chevrier, S.; Anzeneder, T.; Langwieder, C.; Dykgers, A.; Rees, M.; Ramaswamy, A.; Muenst, S.; Soysal, S.D.; et al. A Single-Cell Atlas of the Tumor and Immune Ecosystem of Human Breast Cancer. Cell 2019, 177, 1330–1345.e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, P.; Fu, Y.-X. Tumor-infiltrating T lymphocytes: Friends or foes? Lab. Investig. 2006, 86, 231–245. [Google Scholar] [CrossRef] [Green Version]
- Thomas, D.A.; Massagué, J. TGF-β directly targets cytotoxic T cell functions during tumor evasion of immune surveillance. Cancer Cell 2005, 8, 369–380. [Google Scholar] [CrossRef] [Green Version]
- Farhood, B.; Najafi, M.; Mortezaee, K. CD8 + cytotoxic T lymphocytes in cancer immunotherapy: A review. J. Cell. Physiol. 2018, 234, 8509–8521. [Google Scholar] [CrossRef] [PubMed]
- Beatty, G.L.; Paterson, Y. IFN-γ-Dependent Inhibition of Tumor Angiogenesis by Tumor-Infiltrating CD4+T Cells Requires Tumor Responsiveness to IFN-γ. J. Immunol. 2001, 166, 2276–2282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yu, P.; Spiotto, M.T.; Lee, Y.; Schreiber, H.; Fu, Y.-X. Complementary Role of CD4+ T Cells and Secondary Lymphoid Tissues for Cross-presentation of Tumor Antigen to CD8+ T Cells. J. Exp. Med. 2003, 197, 985–995. [Google Scholar] [CrossRef] [Green Version]
- Han, S.; Zhang, C.; Li, Q.; Dong, J.; Liu, Y.; Huang, Y.; Jiang, T.; Wu, A. Tumour-infiltrating CD4+ and CD8+ lymphocytes as predictors of clinical outcome in glioma. Br. J. Cancer 2014, 110, 2560–2568. [Google Scholar] [CrossRef]
- Kroemer, M.; Turco, C.; Spehner, L.; Viot, J.; Idirène, I.; Bouard, A.; Renaude, E.; Deschamps, M.; Godet, Y.; Adotévi, O.; et al. Investigation of the prognostic value of CD4 T cell subsets expanded from tumor-infiltrating lymphocytes of colorectal cancer liver metastases. J. Immunother. Cancer 2020, 8, e001478. [Google Scholar] [CrossRef]
- Lugano, R.; Ramachandran, M.; Dimberg, A. Tumor angiogenesis: Causes, consequences, challenges and opportunities. Cell. Mol. Life Sci. 2020, 77, 1745–1770. [Google Scholar] [CrossRef] [Green Version]
- Martinet, L.; Garrido, I.; Filleron, T.; Le Guellec, S.; Bellard, E.; Fournie, J.-J.; Rochaix, P.; Girard, J.-P. Human Solid Tumors Contain High Endothelial Venules: Association with T- and B-Lymphocyte Infiltration and Favorable Prognosis in Breast Cancer. Cancer Res. 2011, 71, 5678–5687. [Google Scholar] [CrossRef] [Green Version]
- Schepers, K.; Campbell, T.B.; Passegué, E. Normal and Leukemic Stem Cell Niches: Insights and Therapeutic Opportunities. Cell Stem Cell 2015, 16, 254–267. [Google Scholar] [CrossRef] [Green Version]
- Boilly, B.; Faulkner, S.; Jobling, P.; Hondermarck, H. Nerve Dependence: From Regeneration to Cancer. Cancer Cell 2017, 31, 342–354. [Google Scholar] [CrossRef] [Green Version]
- Wang, W.; Li, L.; Chen, N.; Niu, C.; Li, Z.; Hu, J.; Cui, J. Nerves in the Tumor Microenvironment: Origin and Effects. Front. Cell Dev. Biol. 2020, 8, 1630. [Google Scholar] [CrossRef] [PubMed]
- Tan, X.; Sivakumar, S.; Bednarsch, J.; Wiltberger, G.; Kather, J.N.; Niehues, J.; de Vos-Geelen, J.; Iersel, L.V.-V.; Kintsler, S.; Roeth, A.; et al. Nerve fibers in the tumor microenvironment in neurotropic cancer—Pancreatic cancer and cholangiocarcinoma. Oncogene 2021, 40, 899–908. [Google Scholar] [CrossRef] [PubMed]
- Nishida, N.; Yano, H.; Nishida, T.; Kamura, T.; Kojiro, M. Angiogenesis in cancer. Vasc. Health Risk Manag. 2006, 2, 213–219. [Google Scholar] [CrossRef]
- Faulkner, S.; Jobling, P.; March, B.; Jiang, C.C.; Hondermarck, H. Tumor Neurobiology and the War of Nerves in Cancer. Cancer Discov. 2019, 9, 702–710. [Google Scholar] [CrossRef] [Green Version]
- Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
- Freire, M.; Van Dyke, T. Natural resolution of inflammation. Periodontology 2000 2013, 63, 149–164. [Google Scholar] [CrossRef] [Green Version]
- Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, Inflammation, and Cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef] [Green Version]
- Liou, G.-Y.; Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 2010, 44, 479–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giorgio, M.; Dellino, G.I.; Gambino, V.; Roda, N.; Pelicci, P.G. On the epigenetic role of guanosine oxidation. Redox Biol. 2020, 29, 101398. [Google Scholar] [CrossRef]
- Ernst, P.B.; Gold, B.D. The Disease Spectrum of Helicobacter Pylori: The Immunopathogenesis of Gastroduodenal Ulcer and Gastric Cancer. Annu. Rev. Microbiol. 2000, 54, 615–640. [Google Scholar] [CrossRef] [PubMed]
- Correa, P.; Houghton, J. Carcinogenesis of Helicobacter pylori. Gastroenterology 2007, 133, 659–672. [Google Scholar] [CrossRef]
- Mannick, E.E.; Bravo, L.E.; Zarama, G.; Realpe, J.L.; Zhang, X.J.; Ruiz, B.; Fontham, E.T.; Mera, R.; Miller, M.J.; Correa, P. Inducible nitric oxide synthase, nitrotyrosine, and apoptosis in Helicobacter pylori gastritis: Effect of antibiotics and antioxidants. Cancer Res. 1996, 56, 3238–3243. [Google Scholar]
- Baik, S.C.; Youn, H.S.; Chung, M.H.; Lee, W.K.; Cho, M.J.; Ko, G.H.; Park, C.K.; Kasai, H.; Rhee, K.H. Increased Oxidative DNA Damage in Helicobacter /Ty/on-Infected Human Gastric Mucosa. Cancer Res 1996, 56, 1279–1282. [Google Scholar] [PubMed]
- Mancino, M.; Ametller, E.; Gascón, P.; Almendro, V. The neuronal influence on tumor progression. Biochim. Biophys. Acta (BBA) Bioenerg. 2011, 1816, 105–118. [Google Scholar] [CrossRef] [Green Version]
- Magnon, C.; Hall, S.J.; Lin, J.; Xue, X.; Gerber, L.; Freedland, S.J.; Frenette, P.S. Autonomic Nerve Development Contributes to Prostate Cancer Progression. Science 2013, 341, 1236361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mauffrey, P.; Tchitchek, N.; Barroca, V.; Bemelmans, A.-P.; Firlej, V.; Allory, Y.; Roméo, P.-H.; Magnon, C. Progenitors from the central nervous system drive neurogenesis in cancer. Nature 2019, 569, 672–678. [Google Scholar] [CrossRef]
- Renz, B.W.; Takahashi, R.; Tanaka, T.; Macchini, M.; Hayakawa, Y.; Dantes, Z.; Maurer, H.C.; Chen, X.; Jiang, Z.; Westphalen, C.B.; et al. β2 Adrenergic-Neurotrophin Feedforward Loop Promotes Pancreatic Cancer. Cancer Cell 2018, 33, 75–90.e7. [Google Scholar] [CrossRef] [Green Version]
- Saloman, J.; Albers, K.M.; Li, D.; Hartman, D.J.; Crawford, H.C.; Muha, E.A.; Rhim, A.D.; Davis, B. Ablation of sensory neurons in a genetic model of pancreatic ductal adenocarcinoma slows initiation and progression of cancer. Proc. Natl. Acad. Sci. USA 2016, 113, 3078–3083. [Google Scholar] [CrossRef] [Green Version]
- Stopczynski, R.E.; Normolle, D.P.; Hartman, D.J.; Ying, H.; DeBerry, J.; Bielefeldt, K.; Rhim, A.D.; DePinho, R.; Albers, K.; Davis, B. Neuroplastic changes occur early in the development of pancreatic ductal adenocarcinoma. Cancer Res. 2014, 74, 1718–1727. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhao, C.-M.; Hayakawa, Y.; Kodama, Y.; Muthupalani, S.; Westphalen, C.B.; Andersen, G.T.; Flatberg, A.; Johannessen, H.; Friedman, R.A.; Renz, B.W.; et al. Denervation suppresses gastric tumorigenesis. Sci. Transl. Med. 2014, 6, 250ra115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Peterson, S.C.; Eberl, M.; Vagnozzi, A.N.; Belkadi, A.; Veniaminova, N.A.; Verhaegen, M.E.; Bichakjian, C.K.; Ward, N.L.; Dlugosz, A.A.; Wong, S.Y. Basal Cell Carcinoma Preferentially Arises from Stem Cells within Hair Follicle and Mechanosensory Niches. Cell Stem Cell 2015, 16, 400–412. [Google Scholar] [CrossRef] [Green Version]
- Sherwood, L.M.; Parris, E.E.; Folkman, J. Tumor Angiogenesis: Therapeutic Implications. N. Engl. J. Med. 1971, 285, 1182–1186. [Google Scholar] [CrossRef] [PubMed]
- Folkman, J. Role of angiogenesis in tumor growth and metastasis. Semin. Oncol. 2002, 29, 15–18. [Google Scholar] [CrossRef]
- Folkman, J.; Watson, K.; Ingber, D.; Hanahan, D. Induction of angiogenesis during the transition from hyperplasia to neoplasia. Nature 1989, 339, 58–61. [Google Scholar] [CrossRef] [PubMed]
- Schmidt, T.; Carmeliet, P. Angiogenesis: A Target in Solid Tumors, Also in Leukemia? Hematology 2011, 2011, 1–8. [Google Scholar] [CrossRef] [Green Version]
- Liao, D.; Johnson, R.S. Hypoxia: A key regulator of angiogenesis in cancer. Cancer Metastasis Rev. 2007, 26, 281–290. [Google Scholar] [CrossRef]
- Semenza, G.L. Hydroxylation of HIF-1: Oxygen Sensing at the Molecular Level. Physiology 2004, 19, 176–182. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Holash, J.; Maisonpierre, P.C.; Compton, D.; Boland, P.; Alexander, C.R.; Zagzag, D.; Yancopoulos, G.D.; Wiegand, S.J. Vessel Cooption, Regression, and Growth in Tumors Mediated by Angiopoietins and VEGF. Science 1999, 284, 1994–1998. [Google Scholar] [CrossRef] [Green Version]
- Leung, D.; Cachianes, G.; Kuang, W.; Goeddel, D.; Ferrara, N. Vascular endothelial growth factor is a secreted angiogenic mitogen. Science 1989, 246, 1306–1309. [Google Scholar] [CrossRef] [PubMed]
- Bernatchez, P.N.; Soker, S.; Sirois, M.G. Vascular Endothelial Growth Factor Effect on Endothelial Cell Proliferation, Migration, and Platelet-activating Factor Synthesis Is Flk-1-dependent. J. Biol. Chem. 1999, 274, 31047–31054. [Google Scholar] [CrossRef] [Green Version]
- Weidenaar, A.C.; Ter Elst, A.; Koopmans-Klein, G.; Rosati, S.; Dunnen, W.F.A.D.; Boer, T.M.-D.; Kamps, W.A.; Vellenga, E.; De Bont, E.S.J.M. High Acute Myeloid Leukemia derived VEGFA levels are associated with a specific vascular morphology in the leukemic bone marrow. Cell. Oncol. 2011, 34, 289–296. [Google Scholar] [CrossRef] [Green Version]
- Padró, T.; Ruiz, S.; Bieker, R.; Bürger, H.; Steins, M.; Kienast, J.; Büchner, T.; Berdel, W.E.; Mesters, R.M. Increased angiogenesis in the bone marrow of patients with acute myeloid leukemia. Blood 2000, 95, 2637–2644. [Google Scholar] [CrossRef] [PubMed]
- Aguayo, A.; Kantarjian, H.; Manshouri, T.; Gidel, C.; Estey, E.; Thomas, D.; Koller, C.; Estrov, Z.; O’Brien, S.; Keating, M. Angiogenesis in Acute and Chronic Leukemias and Myelodysplastic Syndromes. Blood 2000, 6, 2240–2245. [Google Scholar] [CrossRef]
- Hiramatsu, A.; Miwa, H.; Shikami, M.; Ikai, T.; Tajima, E.; Yamamoto, H.; Imai, N.; Hattori, A.; Kyo, T.; Watarai, M.; et al. Disease-specific expression of VEGF and its receptors in AML cells: Possible autocrine pathway of VEGF/type1 receptor of VEGF in t(15;17) AML and VEGF/type2 receptor of VEGF in t(8;21) AML. Leuk. Lymphoma 2006, 47, 89–95. [Google Scholar] [CrossRef] [PubMed]
- Silverman, D.A.; Martinez, V.K.; Dougherty, P.M.; Myers, J.N.; Calin, G.A.; Amit, M. Cancer-Associated Neurogenesis and Nerve-Cancer Cross-talk. Cancer Res. 2021, 81, 1431–1440. [Google Scholar] [CrossRef] [PubMed]
- Zahalka, A.H.; Frenette, P.S. Nerves in cancer. Nat. Rev. Cancer 2020, 20, 143–157. [Google Scholar] [CrossRef]
- Ayala, G.E.; Dai, H.; Powell, M.; Li, R.; Ding, Y.; Wheeler, T.M.; Shine, D.; Kadmon, D.; Thompson, T.; Miles, B.J.; et al. Cancer-Related Axonogenesis and Neurogenesis in Prostate Cancer. Clin. Cancer Res. 2008, 14, 7593–7603. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Biankin, A.V.; Initiative, A.P.C.G.; Waddell, N.; Kassahn, K.S.; Gingras, M.-C.; Muthuswamy, L.B.; Johns, A.L.; Miller, D.K.; Wilson, P.J.; Patch, A.-M.; et al. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 2012, 491, 399–405. [Google Scholar] [CrossRef] [PubMed]
- Dollé, L.; EL Yazidi-Belkoura, I.; Adriaenssens, E.; Nurcombe, V.; Hondermarck, H. Nerve growth factor overexpression and autocrine loop in breast cancer cells. Oncogene 2003, 22, 5592–5601. [Google Scholar] [CrossRef] [Green Version]
- Bloom, A.P.; Jimenez-Andrade, J.M.; Taylor, R.N.; Castañeda-Corral, G.; Kaczmarska, M.J.; Freeman, K.T.; Coughlin, K.A.; Ghilardi, J.R.; Kuskowski, M.A.; Mantyh, P.W. Breast Cancer-Induced Bone Remodeling, Skeletal Pain, and Sprouting of Sensory Nerve Fibers. J. Pain 2011, 12, 698–711. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Han, H.; Yang, C.; Zhang, Y.; Han, C.; Zhang, G. Vascular Endothelial Growth Factor Mediates the Sprouted Axonogenesis of Breast Cancer in Rat. Am. J. Pathol. 2021, 191, 515–526. [Google Scholar] [CrossRef]
- Pundavela, J.; Demont, Y.; Jobling, P.; Lincz, L.F.; Roselli, S.; Thorne, R.F.; Bond, D.; Bradshaw, R.A.; Walker, M.M.; Hondermarck, H. ProNGF Correlates with Gleason Score and Is a Potential Driver of Nerve Infiltration in Prostate Cancer. Am. J. Pathol. 2014, 184, 3156–3162. [Google Scholar] [CrossRef] [PubMed]
- Jimenez-Andrade, J.M.; Bloom, A.P.; Stake, J.I.; Mantyh, W.; Taylor, R.N.; Freeman, K.T.; Ghilardi, J.R.; Kuskowski, M.A.; Mantyh, P.W. Pathological Sprouting of Adult Nociceptors in Chronic Prostate Cancer-Induced Bone Pain. J. Neurosci. 2010, 30, 14649–14656. [Google Scholar] [CrossRef]
- Allen, J.K.; Armaiz-Pena, G.N.; Nagaraja, A.S.; Sadaoui, N.C.; Ortiz, T.; Dood, R.; Ozcan, M.; Herder, D.M.; Haemmerle, M.; Gharpure, K.M.; et al. Sustained Adrenergic Signaling Promotes Intratumoral Innervation through BDNF Induction. Cancer Res. 2018, 78, 3233–3242. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Y.; Liu, Y.; Liu, H.; Tang, W.H. Exosomes: Biogenesis, biologic function and clinical potential. Cell Biosci. 2019, 9, 19. [Google Scholar] [CrossRef] [PubMed]
- Madeo, M.; Colbert, P.L.; Vermeer, D.W.; Lucido, C.T.; Cain, J.T.; Vichaya, E.G.; Grossberg, A.J.; Muirhead, D.; Rickel, A.P.; Hong, Z.; et al. Cancer exosomes induce tumor innervation. Nat. Commun. 2018, 9, 4284. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lucido, C.; Wynja, E.; Madeo, M.; Williamson, C.S.; Schwartz, L.E.; Imblum, B.A.; Drapkin, R.; Vermeer, P.D. Innervation of cervical carcinoma is mediated by cancer-derived exosomes. Gynecol. Oncol. 2019, 154, 228–235. [Google Scholar] [CrossRef] [Green Version]
- Amit, M.; Takahashi, H.; Dragomir, M.P.; Lindemann, A.; Netto, F.O.G.; Pickering, C.R.; Anfossi, S.; Osman, A.A.; Cai, Y.; Wang, R.; et al. Loss of p53 drives neuron reprogramming in head and neck cancer. Nature 2020, 578, 449–454. [Google Scholar] [CrossRef]
- Méndez-Ferrer, S.; Michurina, T.V.; Ferraro, F.; Mazloom, A.R.; MacArthur, B.; Lira, S.A.; Scadden, D.T.; Ma’Ayan, A.; Enikolopov, G.; Frenette, P.S. Mesenchymal and haematopoietic stem cells form a unique bone marrow niche. Nature 2010, 466, 829–834. [Google Scholar] [CrossRef]
- Arranz, L.; Sánchez-Aguilera, A.; Pérez, D.M.; Isern, J.; Langa, X.; Tzankov, A.; Lundberg, P.; Muntión, S.; Tzeng, Y.-S.; Lai, D.-M.; et al. Neuropathy of haematopoietic stem cell niche is essential for myeloproliferative neoplasms. Nature 2014, 512, 78–81. [Google Scholar] [CrossRef]
- Hanoun, M.; Zhang, D.; Mizoguchi, T.; Pinho, S.; Pierce, H.; Kunisaki, Y.; Lacombe, J.; Armstrong, S.A.; Dührsen, U.; Frenette, P.S. Acute Myelogenous Leukemia-Induced Sympathetic Neuropathy Promotes Malignancy in an Altered Hematopoietic Stem Cell Niche. Cell Stem Cell 2014, 15, 365–375. [Google Scholar] [CrossRef] [Green Version]
- Salavati, H.; Soltani, M.; Amanpour, S. The pivotal role of angiogenesis in a multi-scale modeling of tumor growth exhibiting the avascular and vascular phases. Microvasc. Res. 2018, 119, 105–116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grimes, D.R.; Kannan, P.; McIntyre, A.; Kavanagh, A.; Siddiky, A.; Wigfield, S.; Harris, A.; Partridge, M. The Role of Oxygen in Avascular Tumor Growth. PLoS ONE 2016, 11, e0153692. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sefidgar, M.; Soltani, M.; Raahemifar, K.; Sadeghi, M.; Bazmara, H.; Bazargan, M.; Naeenian, M.M. Numerical modeling of drug delivery in a dynamic solid tumor microvasculature. Microvasc. Res. 2015, 99, 43–56. [Google Scholar] [CrossRef] [PubMed]
- Phillips, C.M.; Lima, E.A.B.F.; Woodall, R.T.; Brock, A.; Yankeelov, T.E. A hybrid model of tumor growth and angiogenesis: In silico experiments. PLoS ONE 2020, 15, e0231137. [Google Scholar] [CrossRef]
- Soltani, M.; Chen, P. Numerical Modeling of Interstitial Fluid Flow Coupled with Blood Flow through a Remodeled Solid Tumor Microvascular Network. PLoS ONE 2013, 8, e67025. [Google Scholar] [CrossRef] [PubMed]
- Sullivan, M.R.; Heiden, M.G.V. Determinants of nutrient limitation in cancer. Crit. Rev. Biochem. Mol. Biol. 2019, 54, 193–207. [Google Scholar] [CrossRef]
- Finicle, B.T.; Jayashankar, V.; Edinger, A.L. Nutrient scavenging in cancer. Nat. Rev. Cancer 2018, 18, 619–633. [Google Scholar] [CrossRef] [PubMed]
- Lyssiotis, C.A.; Kimmelman, A.C. Metabolic Interactions in the Tumor Microenvironment. Trends Cell Biol. 2017, 27, 863–875. [Google Scholar] [CrossRef] [Green Version]
- Zhou, C.; Qian, W.; Li, J.; Ma, J.; Chen, X.; Jiang, Z.; Cheng, L.; Duan, W.; Wang, Z.; Wu, Z.; et al. High glucose microenvironment accelerates tumor growth via SREBP1-autophagy axis in pancreatic cancer. J. Exp. Clin. Cancer Res. 2019, 38, 302. [Google Scholar] [CrossRef] [PubMed]
- Bertero, T.; Oldham, W.M.; Grasset, E.M.; Bourget, I.; Boulter, E.; Pisano, S.; Hofman, P.; Bellvert, F.; Meneguzzi, G.; Bulavin, D.V.; et al. Tumor-Stroma Mechanics Coordinate Amino Acid Availability to Sustain Tumor Growth and Malignancy. Cell Metab. 2019, 29, 124–140.e10. [Google Scholar] [CrossRef] [Green Version]
- Hiramatsu, K.; Serada, S.; Enomoto, T.; Takahashi, Y.; Nakagawa, S.; Nojima, S.; Morimoto, A.; Matsuzaki, S.; Yokoyama, T.; Takahashi, T.; et al. LSR Antibody Therapy Inhibits Ovarian Epithelial Tumor Growth by Inhibiting Lipid Uptake. Cancer Res. 2018, 78, 516–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Witsch, E.; Sela, M.; Yarden, Y. Roles for Growth Factors in Cancer Progression. Physiology 2010, 25, 85–101. [Google Scholar] [CrossRef] [Green Version]
- Aaronson, S. Growth factors and cancer. Science 1991, 254, 1146–1153. [Google Scholar] [CrossRef] [PubMed]
- Reznikov, A. Hormonal impact on tumor growth and progression. Exp. Oncol. 2015, 37, 162–172. [Google Scholar] [CrossRef]
- Jiang, S.-H.; Hu, L.; Wang, X.; Li, J.; Zhang, Z.-G. Neurotransmitters: Emerging targets in cancer. Oncogene 2020, 39, 503–515. [Google Scholar] [CrossRef]
- Kappos, E.A.; Engels, P.E.; Tremp, M.; Sieber, P.K.; von Felten, S.; Madduri, S.; zu Schwabedissen, M.M.; Fischmann, A.; Schaefer, D.J.; Kalbermatten, D.F. Denervation leads to volume regression in breast cancer. J. Plast. Reconstr. Aesthetic Surg. 2018, 71, 833–839. [Google Scholar] [CrossRef] [Green Version]
- Coelho, M.; Soares-Silva, C.; Brandão, D.; Marino, F.; Cosentino, M.; Ribeiro, L. β-Adrenergic modulation of cancer cell proliferation: Available evidence and clinical perspectives. J. Cancer Res. Clin. Oncol. 2017, 143, 275–291. [Google Scholar] [CrossRef]
- Thaker, P.H.; Han, L.Y.; Kamat, A.A.; Arevalo, J.M.; Takahashi, R.; Lu, C.; Jennings, N.B.; Armaiz-Pena, G.N.; Bankson, J.; Ravoori, M.; et al. Chronic stress promotes tumor growth and angiogenesis in a mouse model of ovarian carcinoma. Nat. Med. 2006, 12, 939–944. [Google Scholar] [CrossRef]
- Zahalka, A.H.; Arnal-Estapé, A.; Maryanovich, M.; Nakahara, F.; Cruz, C.D.; Finley, L.W.S.; Frenette, P.S. Adrenergic nerves activate an angio-metabolic switch in prostate cancer. Science 2017, 358, 321–326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kamiya, A.; Hayama, Y.; Kato, S.; Shimomura, A.; Shimomura, T.; Irie, K.; Kaneko, R.; Yanagawa, Y.; Kobayashi, K.; Ochiya, T. Genetic manipulation of autonomic nerve fiber innervation and activity and its effect on breast cancer progression. Nat. Neurosci. 2019, 22, 1289–1305. [Google Scholar] [CrossRef]
- Basu, S.; Sarkar, C.; Chakroborty, D.; Nagy, J.; Mitra, R.B.; Dasgupta, P.S.; Mukhopadhyay, D. Ablation of Peripheral Dopaminergic Nerves Stimulates Malignant Tumor Growth by Inducing Vascular Permeability Factor/Vascular Endothelial Growth Factor-Mediated Angiogenesis. Cancer Res. 2004, 64, 5551–5555. [Google Scholar] [CrossRef] [Green Version]
- Basu, S.; Nagy, J.A.; Pal, S.; Vasile, E.; Eckelhoefer, I.A.; Bliss, V.S.; Manseau, E.J.; Dasgupta, P.S.; Dvorak, H.F.; Mukhopadhyay, D. The neurotransmitter dopamine inhibits angiogenesis induced by vascular permeability factor/vascular endothelial growth factor. Nat. Med. 2001, 7, 569–574. [Google Scholar] [CrossRef] [PubMed]
- Chakroborty, D.; Sarkar, C.; Mitra, R.B.; Banerjee, S.; Dasgupta, P.S.; Basu, S. Depleted Dopamine in Gastric Cancer Tissues. Clin. Cancer Res. 2004, 10, 4349–4356. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chakroborty, D.; Chowdhury, U.R.; Sarkar, C.; Baral, R.; Dasgupta, P.S.; Basu, S. Dopamine regulates endothelial progenitor cell mobilization from mouse bone marrow in tumor vascularization. J. Clin. Investig. 2008, 118, 1380–1389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moreno-Smith, M.; Lu, C.; Shahzad, M.M.; Armaiz-Pena, G.N.; Allen, J.K.; Stone, R.L.; Mangala, L.S.; Han, H.D.; Kim, H.S.; Farley, D.; et al. Dopamine Blocks Stress-Mediated Ovarian Carcinoma Growth. Clin. Cancer Res. 2011, 17, 3649–3659. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dolma, S.; Selvadurai, H.J.; Lan, X.; Lee, L.; Kushida, M.; Voisin, V.; Whetstone, H.; So, M.; Aviv, T.; Park, N.; et al. Inhibition of Dopamine Receptor D4 Impedes Autophagic Flux, Proliferation, and Survival of Glioblastoma Stem Cells. Cancer Cell 2016, 29, 859–873. [Google Scholar] [CrossRef] [Green Version]
- Soll, C.; Jang, J.H.; Riener, M.-O.; Moritz, W.; Wild, P.J.; Graf, R.; Clavien, P.-A. Serotonin promotes tumor growth in human hepatocellular cancer. Hepatology 2009, 51, 1244–1254. [Google Scholar] [CrossRef]
- Nocito, A.; Dahm, F.; Jochum, W.; Jang, J.H.; Georgiev, P.; Bader, M.; Graf, R.; Clavien, P.-A. Serotonin Regulates Macrophage-Mediated Angiogenesis in a Mouse Model of Colon Cancer Allografts. Cancer Res. 2008, 68, 5152–5158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pfitzinger, P.L.; Fangmann, L.; Wang, K.; Demir, E.; Gürlevik, E.; Fleischmann-Mundt, B.; Brooks, J.; D’Haese, J.G.; Teller, S.; Hecker, A.; et al. Indirect cholinergic activation slows down pancreatic cancer growth and tumor-associated inflammation. J. Exp. Clin. Cancer Res. 2020, 39, 289. [Google Scholar] [CrossRef]
- Yu, H.; Xia, H.; Tang, Q.; Xu, H.; Wei, G.; Chen, Y.; Dai, X.; Gong, Q.; Bi, F. Acetylcholine acts through M3 muscarinic receptor to activate the EGFR signaling and promotes gastric cancer cell proliferation. Sci. Rep. 2017, 7, 40802. [Google Scholar] [CrossRef]
- Amit, M.; Na’Ara, S.; Gil, Z. Mechanisms of cancer dissemination along nerves. Nat. Rev. Cancer 2016, 16, 399–408. [Google Scholar] [CrossRef] [PubMed]
- Bielenberg, D.R.; Zetter, B.R. The Contribution of Angiogenesis to the Process of Metastasis. Cancer J. 2015, 21, 267–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Butler, P.; Gullino, M. Quantitation of Cell Shedding into Efferent Blood of Mammary. Cancer Res. 1975, 35, 512–516. [Google Scholar]
- Zetter, B.R. Angiogenesis and tumor metastasis. Annu. Rev. Med. 1998, 49, 407–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tonini, T.; Rossi, F.; Claudio, P.P. Molecular basis of angiogenesis and cancer. Oncogene 2003, 22, 6549–6556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Farnsworth, R.; Lackmann, M.; Achen, M.; Stacker, S. Vascular remodeling in cancer. Oncogene 2014, 33, 3496–3505. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ellis, L.; Fidler, I. Angiogenesis and metastasis. Eur. J. Cancer 1996, 32, 2451–2460. [Google Scholar] [CrossRef]
- Hasan, J.; Byers, R.; Jayson, G.C. Intra-tumoural microvessel density in human solid tumours. Br. J. Cancer 2002, 86, 1566–1577. [Google Scholar] [CrossRef]
- Liotta, L.A.; Kleinerman, J.; Saidel, G.M. Quantitative Relationships of Intravascular Tumor Cells, Tumor Vessels, and Pulmonary Metastases following Tumor Implantation. Cancer Res. 1974, 34, 997–1004. [Google Scholar] [PubMed]
- Srivastava, A.; Laidler, P.; Davies, R.P.; Horgan, K.; Hughes, L.E. The Prognostic Significance of Tumor Vascularity in Intermediate-Thickness (0.76–4.0 Mm Thick) Skin Melanoma. Am. J. Pathol. 1988, 133, 419–423. [Google Scholar]
- Demirkesen, C.; Buyukpinarbasili, N.; Ramazanoglu, R.; Oguz, O.; Mandel, N.M.; Kaner, G. The correlation of angiogenesis with metastasis in primary cutaneous melanoma: A comparative analysis of microvessel density, expression of vascular endothelial growth factor and basic fibroblastic growth factor. Pathology 2006, 38, 132–137. [Google Scholar] [CrossRef] [PubMed]
- Horak, E.; Klenk, N.; Leek, R.; LeJeune, S.; Smith, K.; Stuart, N.; Harris, A.; Greenall, M.; Stepniewska, K. Angiogenesis, assessed by platelet/endothelial cell adhesion molecule antibodies, as indicator of node metastases and survival in breast cancer. Lancet 1992, 340, 1120–1124. [Google Scholar] [CrossRef]
- Gasparini, G.; Weidner, N.; Bevilacqua, P.; Maluta, S.; Palma, P.D.; Caffo, O.; Barbareschi, M.; Boracchi, P.; Marubini, E.; Pozza, F. Tumor microvessel density, p53 expression, tumor size, and peritumoral lymphatic vessel invasion are relevant prognostic markers in node-negative breast carcinoma. J. Clin. Oncol. 1994, 12, 454–466. [Google Scholar] [CrossRef]
- Weidner, N.; Semple, J.P.; Welch, W.R.; Folkman, J. Tumor Angiogenesis and Metastasis—Correlation in Invasive Breast Carcinoma. N. Engl. J. Med. 1991, 324, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Weidner, N.; Carroll, P.R.; Flax, J.; Blumenfeld, W.; Folkman, J. Tumor angiogenesis correlates with metastasis in invasive prostate carcinoma. Am. J. Pathol. 1993, 143, 401–409. [Google Scholar]
- Macchiarini, P.; Fontanini, G.; Squartini, F.; Angeletti, C.; Hardin, M. Relation of neovascularisation to metastasis of non-small-cell lung cancer. Lancet 1992, 340, 145–146. [Google Scholar] [CrossRef]
- Xiangming, C.; Hokita, S.; Natsugoe, S.; Tanabe, G.; Baba, M.; Takao, S.; Kuroshima, K.; Aikou, T. Angiogenesis as an unfavorable factor related to lymph node metastasis in early gastric cancer. Ann. Surg. Oncol. 1998, 5, 585–589. [Google Scholar] [CrossRef]
- Entschladen, F.; Drell, T.L.; Lang, K.; Joseph, J.; Zaenker, K.S. Tumour-cell migration, invasion, and metastasis: Navigation by neurotransmitters. Lancet Oncol. 2004, 5, 254–258. [Google Scholar] [CrossRef]
- Kuol, N.; Stojanovska, L.; Apostolopoulos, V.; Nurgali, K. Role of the nervous system in cancer metastasis. J. Exp. Clin. Cancer Res. 2018, 37, 5. [Google Scholar] [CrossRef] [Green Version]
- Albo, D.; Akay, C.L.; Marshall, C.L.; Wilks, J.; Verstovsek, G.; Liu, H.; Agarwal, N.; Berger, D.H.; Ayala, G.E. Neurogenesis in colorectal cancer is a marker of aggressive tumor behavior and poor outcomes. Cancer 2011, 117, 4834–4845. [Google Scholar] [CrossRef]
- Zhao, Q.; Yang, Y.; Liang, X.; Du, G.; Liu, L.; Lu, L.; Dong, J.; Han, H.; Zhang, G. The clinicopathological significance of neurogenesis in breast cancer. BMC Cancer 2014, 14, 484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pundavela, J.; Roselli, S.; Faulkner, S.; Attia, J.; Scott, R.J.; Thorne, R.F.; Forbes, J.F.; Bradshaw, R.A.; Walker, M.M.; Jobling, P.; et al. Nerve fibers infiltrate the tumor microenvironment and are associated with nerve growth factor production and lymph node invasion in breast cancer. Mol. Oncol. 2015, 9, 1626–1635. [Google Scholar] [CrossRef] [Green Version]
- Zhang, L.; Department of General Surgery; Guo, L.; Tao, M.; Fu, W.; Xiu, D. Parasympathetic neurogenesis is strongly associated with tumor budding and correlates with an adverse prognosis in pancreatic ductal adenocarcinoma. Chin. J. Cancer Res. 2016, 28, 180–186. [Google Scholar] [CrossRef] [PubMed]
- Rowe, C.W.; Dill, T.; Griffin, N.; Jobling, P.; Faulkner, S.; Paul, J.W.; King, S.; Smith, R.; Hondermarck, H. Innervation of papillary thyroid cancer and its association with extra-thyroidal invasion. Sci. Rep. 2020, 10, 1539. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim-Fuchs, C.; Le, C.P.; Pimentel, M.A.; Shackleford, D.; Ferrari, D.; Angst, E.; Hollande, F.; Sloan, E.K. Chronic stress accelerates pancreatic cancer growth and invasion: A critical role for beta-adrenergic signaling in the pancreatic microenvironment. Brain Behav. Immun. 2014, 40, 40–47. [Google Scholar] [CrossRef] [Green Version]
- Masur, K.; Niggemann, B.; Zanker, K.S.; Entschladen, F. Norepinephrine-induced migration of SW 480 colon carcinoma cells is inhibited by beta-blockers. Cancer Res. 2001, 61, 2866–2869. [Google Scholar]
- Drell, T.; Joseph, J.; Lang, K.; Niggemann, B.; Zaenker, K.; Entschladen, F. Effects of Neurotransmitters on the Chemokinesis and Chemotaxis of MDA-MB-468 Human Breast Carcinoma Cells. Breast Cancer Res. Treat. 2003, 80, 63–70. [Google Scholar] [CrossRef]
- Sloan, E.K.; Priceman, S.J.; Cox, B.F.; Yu, S.; Pimentel, M.A.; Tangkanangnukul, V.; Arevalo, J.M.; Morizono, K.; Karanikolas, B.D.; Wu, L.; et al. The Sympathetic Nervous System Induces a Metastatic Switch in Primary Breast Cancer. Cancer Res. 2010, 70, 7042–7052. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Barbieri, A.; Bimonte, S.; Palma, G.; Luciano, A.; Rea, D.; Giudice, A.; Scognamiglio, G.; La Mantia, E.; Franco, R.; Perdonà, S.; et al. The stress hormone norepinephrine increases migration of prostate cancer cells in vitro and in vivo. Int. J. Oncol. 2015, 47, 527–534. [Google Scholar] [CrossRef] [Green Version]
- Sood, A.K.; Armaiz-Pena, G.N.; Halder, J.; Nick, A.M.; Stone, R.L.; Hu, W.; Carroll, A.R.; Spannuth, W.A.; Deavers, M.T.; Allen, J.K.; et al. Adrenergic modulation of focal adhesion kinase protects human ovarian cancer cells from anoikis. J. Clin. Investig. 2010, 120, 1515–1523. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Madden, K.S.; Szpunar, M.J.; Brown, E.B. β-Adrenergic receptors (β-AR) regulate VEGF and IL-6 production by divergent pathways in high β-AR-expressing breast cancer cell lines. Breast Cancer Res. Treat. 2011, 130, 747–758. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, H.; Liu, D.; Yang, Z.; Sun, L.; Deng, Q.; Yang, S.; Qian, L.; Guo, L.; Yu, M.; Hu, M.; et al. Adrenergic signaling promotes angiogenesis through endothelial cell-tumor cell crosstalk. Endocr. Relat. Cancer 2014, 21, 783–795. [Google Scholar] [CrossRef] [Green Version]
- Lutgendorf, S.K.; Cole, S.; Costanzo, E.; Bradley, S.; Coffin, J.; Jabbari, S.; Rainwater, K.; Ritchie, J.M.; Yang, M.; Sood, A.K. Stress-related mediators stimulate vascular endothelial growth factor secretion by two ovarian cancer cell lines. Clin. Cancer Res. 2003, 9, 281. [Google Scholar]
- Zhang, X.; Zhang, Y.; He, Z.; Yin, K.; Li, B.; Zhang, L.; Xu, Z. Chronic stress promotes gastric cancer progression and metastasis: An essential role for ADRB2. Cell Death Dis. 2019, 10, 788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liebig, C.; Ayala, G.; Wilks, J.; Berger, D.H.; Albo, D. Perineural invasion in cancer. Cancer 2009, 115, 3379–3391. [Google Scholar] [CrossRef]
- Li, S.; Sun, Y.; Gao, D. Role of the nervous system in cancer metastasis. Oncol. Lett. 2013, 5, 1101–1111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Duchalais, E.; Guilluy, C.; Nedellec, S.; Touvron, M.; Bessard, A.; Touchefeu, Y.; Bossard, C.; Boudin, H.; Louarn, G.; Neunlist, M.; et al. Colorectal Cancer Cells Adhere to and Migrate along the Neurons of the Enteric Nervous System. Cell. Mol. Gastroenterol. Hepatol. 2017, 5, 31–49. [Google Scholar] [CrossRef] [Green Version]
- Zhou, Y.; Wang, H.; Gong, H.; Cao, M.; Zhang, G.; Wang, Y. Clinical significance of perineural invasion in stages II and III colorectal cancer. Pathol. Res. Pract. 2015, 211, 839–844. [Google Scholar] [CrossRef]
- Poeschl, E.M.; Pollheimer, M.J.; Kornprat, P.; Lindtner, R.A.; Schlemmer, A.; Rehak, P.; Vieth, M.; Langner, C. Perineural Invasion: Correlation with Aggressive Phenotype and Independent Prognostic Variable in Both Colon and Rectum Cancer. J. Clin. Oncol. 2010, 28, e358–e360. [Google Scholar] [CrossRef]
- Peng, J.; Sheng, W.; Huang, D.; Venook, A.P.; Xu, Y.; Guan, Z.; Cai, S. Perineural invasion in pT3N0 rectal cancer. Cancer 2011, 117, 1415–1421. [Google Scholar] [CrossRef] [PubMed]
- Ceyhan, G.O.; Liebl, F.; Maak, M.; Schuster, T.; Becker, K.; Langer, R.; Demir, I.E.; Hartel, M.; Friess, H.; Rosenberg, R. The Severity of Neural Invasion Is a Crucial Prognostic Factor in Rectal Cancer Independent of Neoadjuvant Radiochemotherapy. Ann. Surg. 2010, 252, 797–804. [Google Scholar] [CrossRef] [PubMed]
- Okazaki, I.; Kotani, A.; Honjo, T. Role of AID in Tumorigenesis. Adv. immunol. 2007, 94, 245–273. [Google Scholar] [CrossRef] [PubMed]
- Ayala, G.E.; Dai, H.; Ittmann, M.; Li, R.; Powell, M.; Frolov, A.; Wheeler, T.M.; Thompson, T.C.; Rowley, D. Growth and Survival Mechanisms Associated with Perineural Invasion in Prostate Cancer. Cancer Res. 2004, 64, 6082–6090. [Google Scholar] [CrossRef] [Green Version]
- Yu, H.-H.; Song, D.Y.; Tsai, Y.-Y.; Thompson, T.; Frassica, D.A.; DeWeese, T.L. Perineural Invasion Affects Biochemical Recurrence-Free Survival in Patients with Prostate Cancer Treated with Definitive External Beam Radiotherapy. Urology 2007, 70, 111–116. [Google Scholar] [CrossRef] [PubMed]
- Tai, S.-K.; Li, W.-Y.; Chu, P.-Y.; Chang, S.-Y.; Tsai, T.-L.; Wang, Y.-F.; Huang, J.-L. Risks and clinical implications of perineural invasion in T1-2 oral tongue squamous cell carcinoma. Head Neck 2012, 34, 994–1001. [Google Scholar] [CrossRef]
- Bilici, A.; Seker, M.; Ustaalioglu, B.B.O.; Kefeli, U.; Yildirim, E.; Yavuzer, D.; Aydin, F.M.; Salepci, T.; Oncel, M.; Gumus, M. Prognostic Significance of Perineural Invasion in Patients with Gastric Cancer Who Underwent Curative Resection. Ann. Surg. Oncol. 2010, 17, 2037–2044. [Google Scholar] [CrossRef]
- Jhawer, M.; Coit, D.; Brennan, M.; Qin, L.-X.; Gonen, M.; Klimstra, D.; Tang, L.; Kelsen, D.P.; Shah, M.A. Perineural Invasion After Preoperative Chemotherapy Predicts Poor Survival in Patients with Locally Advanced Gastric Cancer. Am. J. Clin. Oncol. 2009, 32, 356–362. [Google Scholar] [CrossRef] [PubMed]
- Duraker, N.; Çaynak, Z.; Türköz, K. Perineural invasion has no prognostic value in patients with invasive breast carcinoma. Breast 2006, 15, 629–634. [Google Scholar] [CrossRef]
- Narayan, P.; Flynn, J.; Zhang, Z.; Gillespie, E.F.; Mueller, B.; Xu, A.J.; Cuaron, J.; McCormick, B.; Khan, A.J.; Cahlon, O.; et al. Perineural invasion as a risk factor for locoregional recurrence of invasive breast cancer. Sci. Rep. 2021, 11, 12781. [Google Scholar] [CrossRef] [PubMed]
- He, S.; Chen, C.-H.; Chernichenko, N.; Bakst, R.L.; Barajas, F.; Deborde, S.; Allen, P.J.; Vakiani, E.; Yu, Z.; Wong, R.J. GFR 1 released by nerves enhances cancer cell perineural invasion through GDNF-RET signaling. Proc. Natl. Acad. Sci. USA 2014, 111, E2008–E2017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Esseghir, S.; Todd, S.K.; Hunt, T.; Poulsom, R.; Plaza-Menacho, I.; Reis-Filho, J.S.; Isacke, C. A Role for Glial Cell Derived Neurotrophic Factor Induced Expression by Inflammatory Cytokines and RET/GFR 1 Receptor Up-regulation in Breast Cancer. Cancer Res. 2007, 67, 11732–11741. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Iwahashi, N.; Nagasaka, T.; Tezel, G.; Iwashita, T.; Asai, N.; Murakumo, Y.; Kiuchi, K.; Sakata, K.; Nimura, Y.; Takahashi, M. Expression of glial cell line-derived neurotrophic factor correlates with perineural invasion of bile duct carcinoma. Cancer 2002, 94, 167–174. [Google Scholar] [CrossRef]
- Scanlon, C.S.; Banerjee, R.; Inglehart, R.C.; Liu, M.; Russo, N.; Hariharan, A.; Van Tubergen, E.A.; Corson, S.L.; Asangani, I.; Mistretta, C.M.; et al. Galanin modulates the neural niche to favour perineural invasion in head and neck cancer. Nat. Commun. 2015, 6, 6885. [Google Scholar] [CrossRef] [Green Version]
- Dankbaar, J.W.; Pameijer, F.A.; Hendrikse, J.; Schmalfuss, I.M. Easily detected signs of perineural tumour spread in head and neck cancer. Insights Into Imaging 2018, 9, 1089–1095. [Google Scholar] [CrossRef] [Green Version]
- Bakst, R.L.; Glastonbury, C.M.; Parvathaneni, U.; Katabi, N.; Hu, K.S.; Yom, S. Perineural Invasion and Perineural Tumor Spread in Head and Neck Cancer. Int. J. Radiat. Oncol. 2019, 103, 1109–1124. [Google Scholar] [CrossRef] [Green Version]
- Amit, M.; Eran, A.; Billan, S.; Fridman, E.; Na’Ara, S.; Charas, T.; Gil, Z. Perineural Spread in Noncutaneous Head and Neck Cancer: New Insights into an Old Problem. J. Neurol. Surg. Part B Skull Base 2016, 77, 086–095. [Google Scholar] [CrossRef] [Green Version]
- Paes, F.M.; Singer, A.D.; Checkver, A.N.; Palmquist, R.A.; De La Vega, G.; Sidani, C. Perineural Spread in Head and Neck Malignancies: Clinical Significance and Evaluation with18F-FDG PET/CT. RadioGraphics 2013, 33, 1717–1736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parangi, S.; O’Reilly, M.; Christofori, G.; Holmgren, L.; Grosfeld, J.; Folkman, J.; Hanahan, D. Antiangiogenic therapy of transgenic mice impairs de novo tumor growth. Proc. Natl. Acad. Sci. USA 1996, 93, 2002–2007. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Atzori, M.G.; Tentori, L.; Ruffini, F.; Ceci, C.; Bonanno, E.; Scimeca, M.; Lacal, P.M.; Graziani, G. The Anti–Vascular Endothelial Growth Factor Receptor-1 Monoclonal Antibody D16F7 Inhibits Glioma Growth and Angiogenesis In Vivo. J. Pharmacol. Exp. Ther. 2018, 364, 77–86. [Google Scholar] [CrossRef]
- Prewett, M.; Huber, J.; Li, Y.; Santiago, A.; O’Connor, W.; King, K.; Overholser, J.; Hooper, A.; Pytowski, B.; Witte, L.; et al. Antivascular endothelial growth factor receptor (fetal liver kinase 1) monoclonal antibody inhibits tumor angiogenesis and growth of several mouse and human tumors. Cancer Res. 1999, 59, 5209–5218. [Google Scholar]
- Hashizume, H.; Falcón, B.L.; Kuroda, T.; Baluk, P.; Coxon, A.; Yu, D.; Bready, J.V.; Oliner, J.D.; McDonald, D.M. Complementary Actions of Inhibitors of Angiopoietin-2 and VEGF on Tumor Angiogenesis and Growth. Cancer Res. 2010, 70, 2213–2223. [Google Scholar] [CrossRef] [Green Version]
- Abou-Elkacem, L.; Arns, S.; Brix, G.; Gremse, F.; Zopf, D.; Kiessling, F.; Lederle, W. Regorafenib Inhibits Growth, Angiogenesis, and Metastasis in a Highly Aggressive, Orthotopic Colon Cancer Model. Mol. Cancer Ther. 2013, 12, 1322–1331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mazzieri, R.; Pucci, F.; Moi, D.; Zonari, E.; Ranghetti, A.; Berti, A.; Politi, L.S.; Gentner, B.; Brown, J.L.; Naldini, L.; et al. Targeting the ANG2/TIE2 Axis Inhibits Tumor Growth and Metastasis by Impairing Angiogenesis and Disabling Rebounds of Proangiogenic Myeloid Cells. Cancer Cell 2011, 19, 512–526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jayson, G.C.; Kerbel, R.; Ellis, L.M.; Harris, A. Antiangiogenic therapy in oncology: Current status and future directions. Lancet 2016, 388, 518–529. [Google Scholar] [CrossRef]
- Mancuso, M.R.; Davis, R.; Norberg, S.M.; O’Brien, S.; Sennino, B.; Nakahara, T.; Yao, V.J.; Inai, T.; Brooks, P.; Freimark, B.; et al. Rapid vascular regrowth in tumors after reversal of VEGF inhibition. J. Clin. Investig. 2006, 116, 2610–2621. [Google Scholar] [CrossRef] [Green Version]
- Scappaticci, F.A.; Skillings, J.R.; Holden, S.N.; Gerber, H.-P.; Miller, K.; Kabbinavar, F.; Bergsland, E.; Ngai, J.; Holmgren, E.; Wang, J.; et al. Arterial Thromboembolic Events in Patients with Metastatic Carcinoma Treated with Chemotherapy and Bevacizumab. J. Natl. Cancer Inst. 2007, 99, 1232–1239. [Google Scholar] [CrossRef] [Green Version]
- Elice, F.; Rodeghiero, F. Side effects of anti-angiogenic drugs. Thromb. Res. 2012, 129, S50–S53. [Google Scholar] [CrossRef]
- Allen, J.A.; Adlakha, A.; Bergethon, P. Reversible Posterior Leukoencephalopathy Syndrome after Bevacizumab/FOLFIRI Regimen for Metastatic Colon Cancer. Arch. Neurol. 2006, 63, 1475–1478. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tlemsani, C.; Mir, O.; Boudou-Rouquette, P.; Huillard, O.; Maley, K.; Ropert, S.; Coriat, R.; Goldwasser, F. Posterior reversible encephalopathy syndrome induced by anti-VEGF agents. Target. Oncol. 2011, 6, 253–258. [Google Scholar] [CrossRef]
- Vasudev, N.S.; Reynolds, A.R. Anti-angiogenic therapy for cancer: Current progress, unresolved questions and future directions. Angiogenesis 2014, 17, 471–494. [Google Scholar] [CrossRef] [Green Version]
- Jain, R.K. Normalization of Tumor Vasculature: An Emerging Concept in Antiangiogenic Therapy. Science 2005, 307, 58–62. [Google Scholar] [CrossRef] [PubMed]
- Goel, S.; Duda, D.G.; Xu, L.; Munn, L.L.; Boucher, Y.; Fukumura, D.; Jain, R.K. Normalization of the Vasculature for Treatment of Cancer and Other Diseases. Physiol. Rev. 2011, 91, 1071–1121. [Google Scholar] [CrossRef]
- Vasir, J.K.; Labhasetwar, V. Targeted Drug Delivery in Cancer Therapy. Technol. Cancer Res. Treat. 2005, 4, 363–374. [Google Scholar] [CrossRef] [PubMed]
- Sriraman, S.K.; Aryasomayajula, B.; Torchilin, V.P. Barriers to drug delivery in solid tumors. Tissue Barriers 2014, 2, e29528. [Google Scholar] [CrossRef] [Green Version]
- Di Paolo, A.; Bocci, G. Drug distribution in tumors: Mechanisms, role in drug resistance, and methods for modification. Curr. Oncol. Rep. 2007, 9, 109–114. [Google Scholar] [CrossRef] [PubMed]
- Zhang, B.; Hu, Y.; Pang, Z. Modulating the Tumor Microenvironment to Enhance Tumor Nanomedicine Delivery. Front. Pharmacol. 2017, 8, 952. [Google Scholar] [CrossRef]
- Hobbs, S.K.; Monsky, W.L.; Yuan, F.; Roberts, W.G.; Griffith, L.; Torchilin, V.P.; Jain, R.K. Regulation of transport pathways in tumor vessels: Role of tumor type and microenvironment. Proc. Natl. Acad. Sci. USA 1998, 95, 4607–4612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bottaro, D.; Liotta, L.A. Out of air is not out of action. Nature 2003, 423, 593–595. [Google Scholar] [CrossRef] [PubMed]
- Mpekris, F.; Voutouri, C.; Baish, J.W.; Duda, D.G.; Munn, L.L.; Stylianopoulos, T.; Jain, R.K. Combining microenvironment normalization strategies to improve cancer immunotherapy. Proc. Natl. Acad. Sci. USA 2020, 117, 3728–3737. [Google Scholar] [CrossRef] [Green Version]
- Kamoun, W.S.; Ley, C.D.; Farrar, C.; Duyverman, A.M.; Lahdenranta, J.; Lacorre, D.A.; Batchelor, T.T.; Di Tomaso, E.; Duda, D.G.; Munn, L.L.; et al. Edema Control by Cediranib, a Vascular Endothelial Growth Factor Receptor–Targeted Kinase Inhibitor, Prolongs Survival Despite Persistent Brain Tumor Growth in Mice. J. Clin. Oncol. 2009, 27, 2542–2552. [Google Scholar] [CrossRef]
- Batchelor, T.T.; Gerstner, E.R.; Emblem, K.; Duda, D.G.; Kalpathy-Cramer, J.; Snuderl, M.; Ancukiewicz, M.; Polaskova, P.; Pinho, M.; Jennings, D.; et al. Improved tumor oxygenation and survival in glioblastoma patients who show increased blood perfusion after cediranib and chemoradiation. Proc. Natl. Acad. Sci. USA 2013, 110, 19059–19064. [Google Scholar] [CrossRef] [Green Version]
- Batchelor, T.T.; Sorensen, A.G.; di Tomaso, E.; Zhang, W.-T.; Duda, D.G.; Cohen, K.S.; Kozak, K.R.; Cahill, D.; Chen, P.-J.; Zhu, M.; et al. AZD2171, a Pan-VEGF Receptor Tyrosine Kinase Inhibitor, Normalizes Tumor Vasculature and Alleviates Edema in Glioblastoma Patients. Cancer Cell 2007, 11, 83–95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tong, R.T.; Boucher, Y.; Kozin, S.V.; Winkler, F.; Hicklin, D.J.; Jain, R.K. Vascular Normalization by Vascular Endothelial Growth Factor Receptor 2 Blockade Induces a Pressure Gradient Across the Vasculature and Improves Drug Penetration in Tumors. Cancer Res. 2004, 64, 3731–3736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, Q.; Guo, P.; Gallo, J.M. Impact of Angiogenesis Inhibition by Sunitinib on Tumor Distribution of Temozolomide. Clin. Cancer Res. 2008, 14, 1540–1549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wildiers, H.; Guetens, G.; De Boeck, G.; Verbeken, E.K.; Landuyt, B.; Landuyt, W.; De Bruijn, E.A.; Van Oosterom, A.T. Effect of antivascular endothelial growth factor treatment on the intratumoral uptake of CPT-11. Br. J. Cancer 2003, 88, 1979–1986. [Google Scholar] [CrossRef] [Green Version]
- Winkler, F.; Kozin, S.; Tong, R.; Chae, S.; Booth, M.; Garkavtsev, I.; Xu, L.; Hicklin, D.; Fukumura, D.; DiTOMASO, E. Kinetics of vascular normalization by VEGFR2 blockade governs brain tumor response to radiationRole of oxygenation, angiopoietin-1, and matrix metalloproteinases. Cancer Cell 2004, 6, 553–563. [Google Scholar] [CrossRef] [Green Version]
- Fenton, B.M.; Paoni, S.F. The Addition of AG-013736 to Fractionated Radiation Improves Tumor Response without Functionally Normalizing the Tumor Vasculature. Cancer Res. 2007, 67, 9921–9928. [Google Scholar] [CrossRef] [Green Version]
- Escudier, B.; Eisen, T.; Stadler, W.M.; Szczylik, C.; Oudard, S.; Siebels, M.; Negrier, S.; Chevreau, C.; Solska, E.; Desai, A.A.; et al. Sorafenib in Advanced Clear-Cell Renal-Cell Carcinoma. N. Engl. J. Med. 2007, 356, 125–134. [Google Scholar] [CrossRef] [PubMed]
- Llovet, J.M.; Ricci, S.; Mazzaferro, V.M.; Hilgard, P.; Gane, E.; Blanc, J.-F.; De Oliveira, A.C.; Santoro, A.; Raoul, J.-L.; Forner, A.; et al. Sorafenib in Advanced Hepatocellular Carcinoma. N. Engl. J. Med. 2008, 359, 378–390. [Google Scholar] [CrossRef] [PubMed]
- Motzer, R.J.; Hutson, T.; Tomczak, P.; Michaelson, D.; Bukowski, R.M.; Rixe, O.; Oudard, S.; Negrier, S.; Szczylik, C.; Kim, S.T.; et al. Sunitinib versus Interferon Alfa in Metastatic Renal-Cell Carcinoma. N. Engl. J. Med. 2007, 356, 115–124. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Motzer, R.J.; Hutson, T.; Cella, D.; Reeves, J.; Hawkins, R.; Guo, J.; Nathan, P.; Staehler, M.; De Souza, P.; Merchan, J.R.; et al. Pazopanib versus Sunitinib in Metastatic Renal-Cell Carcinoma. N. Engl. J. Med. 2013, 369, 722–731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rini, B.I.; Escudier, B.; Tomczak, P.; Kaprin, A.; Szczylik, C.; Hutson, T.; Michaelson, D.; Gorbunova, V.A.; Gore, M.E.; Rusakov, I.G.; et al. Comparative effectiveness of axitinib versus sorafenib in advanced renal cell carcinoma (AXIS): A randomised phase 3 trial. Lancet 2011, 378, 1931–1939. [Google Scholar] [CrossRef]
- Perren, T.; Swart, A.M.; Pfisterer, J.; Ledermann, J.A.; Pujade-Lauraine, E.; Kristensen, G.; Carey, M.S.; Beale, P.; Cervantes, A.; Kurzeder, C.; et al. A Phase 3 Trial of Bevacizumab in Ovarian Cancer. N. Engl. J. Med. 2011, 365, 2484–2496. [Google Scholar] [CrossRef] [Green Version]
- Sandler, A.; Gray, R.; Perry, M.C.; Brahmer, J.; Schiller, J.H.; Dowlati, A.; Lilenbaum, R.; Johnson, D.H. Paclitaxel–Carboplatin Alone or with Bevacizumab for Non–Small-Cell Lung Cancer. N. Engl. J. Med. 2006, 355, 2542–2550. [Google Scholar] [CrossRef] [Green Version]
- Willett, C.G.; Boucher, Y.; Di Tomaso, E.; Duda, D.G.; Munn, L.L.; Tong, R.T.; Chung, D.C.; Sahani, D.V.; Kalva, S.P.; Kozin, S.V.; et al. Direct evidence that the VEGF-specific antibody bevacizumab has antivascular effects in human rectal cancer. Nat. Med. 2004, 10, 145–147. [Google Scholar] [CrossRef]
- Hurwitz, H.; Fehrenbacher, L.; Novotny, W.; Cartwright, T.; Hainsworth, J.; Heim, W.; Berlin, J.; Baron, A.; Griffing, S.; Holmgren, E.; et al. Bevacizumab plus Irinotecan, Fluorouracil, and Leucovorin for Metastatic Colorectal Cancer. N. Engl. J. Med. 2004, 350, 2335–2342. [Google Scholar] [CrossRef] [Green Version]
- Bear, H.D.; Tang, G.; Rastogi, P.; Geyer, C.E.; Robidoux, A.; Atkins, J.N.; Baez-Diaz, L.; Brufsky, A.M.; Mehta, R.S.; Fehrenbacher, L.; et al. Bevacizumab Added to Neoadjuvant Chemotherapy for Breast Cancer. N. Engl. J. Med. 2012, 366, 310–320. [Google Scholar] [CrossRef] [Green Version]
- Von Minckwitz, G.; Eidtmann, H.; Rezai, M.; Fasching, P.A.; Tesch, H.; Eggemann, H.; Schrader, I.; Kittel, K.; Hanusch, C.; Kreienberg, R.; et al. Neoadjuvant Chemotherapy and Bevacizumab for HER2-Negative Breast Cancer. N. Engl. J. Med. 2012, 366, 299–309. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Miller, K.; Wang, M.; Gralow, J.; Dickler, M.; Cobleigh, M.; Perez, E.A.; Shenkier, T.; Cella, D.; Davidson, N.E. Paclitaxel plus Bevacizumab versus Paclitaxel Alone for Metastatic Breast Cancer. N. Engl. J. Med. 2007, 357, 2666–2676. [Google Scholar] [CrossRef] [Green Version]
- Demir, I.E.; Tieftrunk, E.; Schorn, S.; Friess, H.; Ceyhan, G.O. Nerve growth factor & TrkA as novel therapeutic targets in cancer. Biochim. Biophys. Acta (BBA) Bioenerg. 2016, 1866, 37–50. [Google Scholar] [CrossRef]
- Brodeur, G.M. Neuroblastoma: Biological insights into a clinical enigma. Nat. Rev. Cancer 2003, 3, 203–216. [Google Scholar] [CrossRef] [PubMed]
- Adriaenssens, E.; Vanhecke, E.; Saule, P.; Mougel, A.; Page, A.; Romon, R.; Nurcombe, V.; Le Bourhis, X.; Hondermarck, H. Nerve Growth Factor Is a Potential Therapeutic Target in Breast Cancer. Cancer Res. 2008, 68, 346–351. [Google Scholar] [CrossRef] [Green Version]
- Watanabe, T.; Katayama, Y.; Kimura, S.; Yoshino, A. Control of proliferation and survival of C6 glioma cells with modification of the nerve growth factor autocrine system. J. Neuro-Oncol. 1999, 41, 121–128. [Google Scholar] [CrossRef]
- Dionne, C.A.; Camoratto, A.M.; Jani, J.P.; Emerson, E.; Neff, N.; Vaught, J.L.; Murakata, C.; Djakiew, D.; Lamb, J.; Bova, S.; et al. Cell cycle-independent death of prostate adenocarcinoma is induced by the trk tyrosine kinase inhibitor CEP-751 (KT6587). Clin. Cancer Res. 1998, 4, 1887–1898. [Google Scholar] [PubMed]
- Evans, A.E.; Kisselbach, K.D.; Yamashiro, D.J.; Ikegaki, N.; Camoratto, A.M.; Dionne, C.A.; Brodeur, G.M. Antitumor activity of CEP-751 (KT-6587) on human neuroblastoma and medulloblastoma xenografts. Clin. Cancer Res. 1999, 5, 3594–3602. [Google Scholar] [PubMed]
- Miknyoczki, S.J.; Dionne, C.A.; Ruggeri, B.A.; Klein-Szanto, A.J. The Novel Trk Receptor Tyrosine Kinase Inhibitor CEP-701 (KT-5555) Exhibits Antitumor Efficacy against Human Pancreatic Carcinoma (Panc1) Xenograft Growth and In Vivo Invasiveness. Ann. N. Y. Acad. Sci. 1999, 880, 252–262. [Google Scholar] [CrossRef] [PubMed]
- Romon, R.; Adriaenssens, E.; Lagadec, C.; Germain, E.; Hondermarck, H.; Le Bourhis, X. Nerve growth factor promotes breast cancer angiogenesis by activating multiple pathways. Mol. Cancer 2010, 9, 157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bapat, A.A.; Munoz, R.M.; Von Hoff, D.D.; Han, H. Blocking Nerve Growth Factor Signaling Reduces the Neural Invasion Potential of Pancreatic Cancer Cells. PLoS ONE 2016, 11, e0165586. [Google Scholar] [CrossRef] [Green Version]
- Lei, Y.; Tang, L.; Xie, Y.; Xianyu, Y.; Zhang, L.; Wang, P.; Hamada, Y.; Jiang, K.; Zheng, W.; Jiang, X. Gold nanoclusters-assisted delivery of NGF siRNA for effective treatment of pancreatic cancer. Nat. Commun. 2017, 8, 15130. [Google Scholar] [CrossRef]
- Hayakawa, Y.; Sakitani, K.; Konishi, M.; Asfaha, S.; Niikura, R.; Tomita, H.; Renz, B.W.; Tailor, Y.; Macchini, M.; Middelhoff, M.; et al. Nerve Growth Factor Promotes Gastric Tumorigenesis through Aberrant Cholinergic Signaling. Cancer Cell 2017, 31, 21–34. [Google Scholar] [CrossRef] [Green Version]
- Mantyh, W.; Jimenez-Andrade, J.M.; Stake, J.; Bloom, A.; Kaczmarska, M.; Taylor, R.; Freeman, K.; Ghilardi, J.; Kuskowski, M.; Mantyh, P. Blockade of nerve sprouting and neuroma formation markedly attenuates the development of late stage cancer pain. Neuroscience 2010, 171, 588–598. [Google Scholar] [CrossRef] [Green Version]
- Sopata, M.; Katz, N.; Carey, W.; Smith, M.D.; Keller, D.; Verburg, K.M.; West, C.R.; Wolfram, G.; Brown, M.T. Efficacy and safety of tanezumab in the treatment of pain from bone metastases. Pain 2015, 156, 1703–1713. [Google Scholar] [CrossRef] [PubMed]
- Qiao, G.; Chen, M.; Bucsek, M.J.; Repasky, E.A.; Hylander, B.L. Adrenergic Signaling: A Targetable Checkpoint Limiting Development of the Antitumor Immune Response. Front. Immunol. 2018, 9, 164. [Google Scholar] [CrossRef] [Green Version]
- McDonald, R.B.; Horwitz, B.A.; Hamilton, J.S.; Stern, J.S. Cold- and norepinephrine-induced thermogenesis in younger and older Fischer 344 rats. Am. J. Physiol. Integr. Comp. Physiol. 1988, 254, R457–R462. [Google Scholar] [CrossRef]
- Hsieh, A.C.L.; Carlson, L.D. Role of Adrenaline and Noradrenaline in Chemical Regulation of Heat Production. Am. J. Physiol. Content 1957, 190, 243–246. [Google Scholar] [CrossRef]
- Macdonald, C.R.; Bucsek, M.J.; Qiao, G.; Chen, M.; Evans, L.; Greenberg, D.J.; Uccello, T.; Battaglia, N.G.; Hylander, B.L.; Singh, A.K.; et al. Adrenergic Receptor Signaling Regulates the Response of Tumors to Ionizing Radiation. Radiat. Res. 2019, 191, 585–589. [Google Scholar] [CrossRef] [PubMed]
- Eng, J.; Reed, C.B.; Kokolus, K.; Pitoniak, R.; Utley, A.; Bucsek, M.J.; Ma, W.W.; Repasky, E.A.; Hylander, B.L. Housing temperature-induced stress drives therapeutic resistance in murine tumour models through β2-adrenergic receptor activation. Nat. Commun. 2015, 6, 6426. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shan, T.; Ma, Q.; Zhang, N.; Guo, K.; Liu, H.; Wang, F.; Wu, E. β2-adrenoceptor blocker synergizes with gemcitabine to inhibit the proliferation of pancreatic cancer cells via apoptosis induction. Eur. J. Pharmacol. 2011, 665, 1–7. [Google Scholar] [CrossRef]
- Chaudhary, K.R.; Yan, S.X.; Heilbroner, S.P.; Sonett, J.R.; Stoopler, M.B.; Shu, C.; Halmos, B.; Wang, T.J.; Hei, T.K.; Cheng, S.K. Effects of β-Adrenergic Antagonists on Chemoradiation Therapy for Locally Advanced Non-Small Cell Lung Cancer. J. Clin. Med. 2019, 8, 575. [Google Scholar] [CrossRef] [Green Version]
- Wolter, N.E.; Wolter, J.K.; Enepekides, D.J.; Irwin, M.S. Propranolol as a novel adjunctive treatment for head and neck squamous cell carcinoma. J. Otolaryngol. Head Neck Surg. 2012, 41, 3594–3602. [Google Scholar]
- Pasquier, E.; Ciccolini, J.; Carre, M.; Giacometti, S.; Fanciullino, R.; Pouchy, C.; Montero, M.-P.; Serdjebi, C.; Kavallaris, M.; André, N. Propranolol potentiates the anti-angiogenic effects and anti-tumor efficacy of chemotherapy agents: Implication in breast cancer treatment. Oncotarget 2011, 2, 797–809. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pasquier, E.; Street, J.; Pouchy, C.; Carre, M.; Gifford, A.; Murray, J.; Norris, M.D.; Trahair, T.; Andre, N.; Kavallaris, M. β-blockers increase response to chemotherapy via direct antitumour and anti-angiogenic mechanisms in neuroblastoma. Br. J. Cancer 2013, 108, 2485–2494. [Google Scholar] [CrossRef] [Green Version]
- Liao, X.; Che, X.; Zhao, W.; Zhang, D.; Long, H.; Chaudhary, P.; Li, H. Effects of propranolol in combination with radiation on apoptosis and survival of gastric cancer cells in vitro. Radiat. Oncol. 2010, 5, 98. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, F.; Liu, H.; Wang, F.; Xu, R.; Wang, P.; Tang, F.; Zhang, X.; Zhu, Z.; Lv, H.; Han, T. Propranolol suppresses the proliferation and induces the apoptosis of liver cancer cells. Mol. Med. Rep. 2018, 17, 5213–5221. [Google Scholar] [CrossRef] [Green Version]
- Kozanoglu, I.; Yandim, M.K.; Cincin, Z.B.; Ozdogu, H.; Cakmakoglu, B.; Baran, Y. New indication for therapeutic potential of an old well-known drug (propranolol) for multiple myeloma. J. Cancer Res. Clin. Oncol. 2013, 139, 327–335. [Google Scholar] [CrossRef] [Green Version]
- Léauté-Labrèze, C.; De La Roque, E.D.; Hubiche, T.; Boralevi, F.; Thambo, J.-B.; Taïeb, A. Propranolol for Severe Hemangiomas of Infancy. N. Engl. J. Med. 2008, 358, 2649–2651. [Google Scholar] [CrossRef]
- Spera, G.; Fresco, R.; Fung, H.; Dyck, J.R.B.; Pituskin, E.; Paterson, D.I.; Mackey, J.R. Beta blockers and improved progression-free survival in patients with advanced HER2 negative breast cancer: A retrospective analysis of the ROSE/TRIO-012 study. Ann. Oncol. 2017, 28, 1836–1841. [Google Scholar] [CrossRef]
- Watkins, J.L.; Thaker, P.H.; Nick, A.M.; Ramondetta, L.M.; Kumar, S.; Ms, D.L.U.; Matsuo, K.; Squires, K.C.; Coleman, R.L.; Lutgendorf, S.K.; et al. Clinical impact of selective and nonselective beta-blockers on survival in patients with ovarian cancer. Cancer 2015, 121, 3444–3451. [Google Scholar] [CrossRef]
- De Giorgi, V.; Gandini, S.; Grazzini, M.; Benemei, S.; Marchionni, N.; Geppetti, P. Effect of β-Blockers and Other Antihypertensive Drugs On the Risk of Melanoma Recurrence and Death. Mayo Clin. Proc. 2013, 88, 1196–1203. [Google Scholar] [CrossRef] [PubMed]
- Kim, S.-A.; Moon, H.; Roh, J.-L.; Kim, S.-B.; Choi, S.-H.; Nam, S.Y.; Kim, S.Y. Postdiagnostic use of β-blockers and other antihypertensive drugs and the risk of recurrence and mortality in head and neck cancer patients: An observational study of 10,414 person–years of follow-up. Clin. Transl. Oncol. 2017, 19, 826–833. [Google Scholar] [CrossRef]
- Holmes, S.; Griffith, E.J.; Musto, G.; Minuk, G.Y. Antihypertensive medications and survival in patients with cancer: A population-based retrospective cohort study. Cancer Epidemiol. 2013, 37, 881–885. [Google Scholar] [CrossRef] [PubMed]
- Yap, A.; Lopez-Olivo, M.; Dubowitz, J.; Pratt, G.; Hiller, J.; Gottumukkala, V.; Sloan, E.; Riedel, B.; Schier, R. Effect of beta-blockers on cancer recurrence and survival: A meta-analysis of epidemiological and perioperative studies. Br. J. Anaesth. 2018, 121, 45–57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kyprianou, N.; Benning, C.M. Suppression of human prostate cancer cell growth by alpha1-adrenoceptor antagonists doxazosin and terazosin via induction of apoptosis. Cancer Res. 2000, 60, 4550–4555. [Google Scholar]
- Park, M.S.; Kim, B.-R.; Kang, S.; Kim, D.-Y.; Rho, S.B. The antihypertension drug doxazosin suppresses JAK/STATs phosphorylation and enhances the effects of IFN-α/γ-induced apoptosis. Genes Cancer 2014, 5, 470–479. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hui, H.; Fernando, M.A.; Heaney, A.P. The α1-adrenergic receptor antagonist doxazosin inhibits EGFR and NF-κB signalling to induce breast cancer cell apoptosis. Eur. J. Cancer 2008, 44, 160–166. [Google Scholar] [CrossRef] [PubMed]
- Suzuki, S.; Yamamoto, M.; Sanomachi, T.; Togashi, K.; Sugai, A.; Seino, S.; Okada, M.; Yoshioka, T.; Kitanaka, C. Doxazosin, a Classic Alpha 1-Adrenoceptor Antagonist, Overcomes Osimertinib Resistance in Cancer Cells via the Upregulation of Autophagy as Drug Repurposing. Biomedicines 2020, 8, 273. [Google Scholar] [CrossRef]
- McConnell, J.D.; Roehrborn, C.G.; Bautista, O.M.; Andriole, G.L.; Dixon, C.M.; Kusek, J.W.; Lepor, H.; McVary, K.T.; Nyberg, L.M.; Clarke, H.S.; et al. The Long-Term Effect of Doxazosin, Finasteride, and Combination Therapy on the Clinical Progression of Benign Prostatic Hyperplasia. N. Engl. J. Med. 2003, 349, 2387–2398. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tahmatzopoulos, A.; Lagrange, C.A.; Zeng, L.; Mitchell, B.L.; Conner, W.T.; Kyprianou, N. Effect of terazosin on tissue vascularity and apoptosis in transitional cell carcinoma of bladder. Urology 2005, 65, 1019–1023. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Roda, N.; Blandano, G.; Pelicci, P.G. Blood Vessels and Peripheral Nerves as Key Players in Cancer Progression and Therapy Resistance. Cancers 2021, 13, 4471. https://doi.org/10.3390/cancers13174471
Roda N, Blandano G, Pelicci PG. Blood Vessels and Peripheral Nerves as Key Players in Cancer Progression and Therapy Resistance. Cancers. 2021; 13(17):4471. https://doi.org/10.3390/cancers13174471
Chicago/Turabian StyleRoda, Niccolò, Giada Blandano, and Pier Giuseppe Pelicci. 2021. "Blood Vessels and Peripheral Nerves as Key Players in Cancer Progression and Therapy Resistance" Cancers 13, no. 17: 4471. https://doi.org/10.3390/cancers13174471
APA StyleRoda, N., Blandano, G., & Pelicci, P. G. (2021). Blood Vessels and Peripheral Nerves as Key Players in Cancer Progression and Therapy Resistance. Cancers, 13(17), 4471. https://doi.org/10.3390/cancers13174471