96
Views
0
CrossRef citations to date
0
Altmetric
Research Articles

Thebaine Derivatives as a New Regulator of Tumor Angiogenesis

, ORCID Icon, ORCID Icon, ORCID Icon, & ORCID Icon
Pages 4501-4519 | Received 12 Jul 2020, Accepted 17 Feb 2021, Published online: 17 May 2021

References

  • Ingo Flamme, Thomas Frölich, and Werner Risau, “Molecular Mechanisms of Vasculogenesis and Embryonic Angiogenesis,” Journal of Cellular Physiology 173, no. 2 (1997): 206–10.
  • Judah Folkman, “Role of Angiogenesis in Tumor Growth and Metastasis,” Seminars in Oncology 29, no. 6Q (2002): 15–8.
  • Rakesh K. Jain, “Normalizing Tumor Microenvironment to Treat Cancer: Bench to Bedside to Biomarkers,” Journal of Clinical Oncology 31, no. 17 (2013): 2205–18.
  • Rakesh K. Jain, “Antiangiogenesis Strategies Revisited: From Starving Tumors to Alleviating Hypoxia,” Cancer Cell 26, no. 5 (2014): 605–22.
  • Kyrre E. Emblem, Kim Mouridsen, Atle Bjornerud, Christian T. Farrar, Dominique Jennings, Ronald J. H. Borra, Patrick Y. Wen, Percy Ivy, Tracy T. Batchelor, Bruce R. Rosen, et al. “Vessel Architectural Imaging Identifies Cancer Patient Responders to anti-Angiogenic Therapy,” Nature Medicine 19, no. 9 (2013): 1178–83.
  • Melissa M. Keenan, and Jen-Tsan Chi, “Alternative Fuels for Cancer Cells,” Cancer Journal (Sudbury, Mass.) 21, no. 2 (2015): 49–55.
  • Elizabeth C. Finger, and Amato J. Giaccia, “Hypoxia, Inflammation, and the Tumor Microenvironment in Metastatic Disease,” Cancer Metastasis Reviews 29, no. 2 (2010): 285–93.
  • Gregg L. Semenza, “Hypoxia-Inducible Factors: Mediators of Cancer Progression and Targets for Cancer Therapy,” Trends in Pharmacological Sciences 33, no. 4 (2012): 207–14.
  • Astrid A. M. Van der Veldt, Mark Lubberink, Idris Bahce, Maudy Walraven, Michiel P. de Boer, Henri N. J. M. Greuter, N. Harry Hendrikse, Jonas Eriksson, Albert D. Windhorst, Pieter E. Postmus, et al. “Rapid Decrease in Delivery of Chemotherapy to Tumors after anti-VEGF Therapy: Implications for Scheduling of Anti-Angiogenic Drugs,” Cancer Cell 21, no. 1 (2012): 82–91.
  • Vikash P. Chauhan, John D. Martin, Hao Liu, Delphine A. Lacorre, Saloni R. Jain, Sergey V. Kozin, Triantafyllos Stylianopoulos, Ahmed S. Mousa, Xiaoxing Han, Pichet Adstamongkonkul, et al. “Angiotensin Inhibition Enhances Drug Delivery and Potentiates Chemotherapy by Decompressing Tumour Blood Vessels,” Nature Communications 4, no. 4 (2013): 2516.,
  • Vasiliki Gkretsi, Andreas Stylianou, Panagiotis Papageorgis, Christiana Polydorou, and Triantafyllos Stylianopoulos, “Remodeling Components of the Tumor Microenvironment to Enhance Cancer Therapy,” Frontiers in Oncology 5, no. 5 (2015): 214.
  • Tracy T. Batchelor, Elizabeth R. Gerstner, Kyrre E. Emblem, Dan G. Duda, Jayashree Kalpathy-Cramer, Matija Snuderl, Marek Ancukiewicz, Pavlina Polaskova, Marco C. Pinho, Dominique Jennings, et al. “Improved Tumor Oxygenation and Survival in Glioblastoma Patients Who Show Increased Blood Perfusion after Cediranib and Chemoradiation,” Proceedings of the National Academy of Sciences of the United States of America 110, no. 47 (2013): 19059–64.
  • Argelia Lara-Solares, Marisol Ahumada Olea, Amparito de Los Ángeles Basantes Pinos, Sara Bistre Cohén, Patricia Bonilla Sierra, Eva Rossina Duarte Juárez, Omar A. Símon Escudero, Juan Guillermo Santacruz Escudero, and José Alberto Flores Cantisani, “Latin-American Guidelines for Cancer Pain Management,” Pain Management 7, no. 4 (2017): 287–98.
  • P. A. Singleton, M. W. Lingen, M. J. Fekete, J. G. N. Garcia, and J. Moss, “Methylnaltrexone Inhibits Opiate and VEGF-Induced Angiogenesis: Role of Receptor Transactivation,” Microvascular Research 72, no. 1–2 (2006): 3–11.
  • J. Nguyen, K. Luk, D. Vang, W. Soto, L. Vincent, S. Robiner, R. Saavedra, Y. Li, P. Gupta, and K. Gupta, “Morphine Stimulates Cancer Progression and Mast Cell Activation and Impairs Survival in Transgenic Mice with Breast Cancer,” British Journal of Anaesthesia 113, no. 113 (2014): i4–13.
  • M. Farooqui, Y. Li, T. Rogers, T. Poonawala, R. J. Griffin, C. W. Song, and K. Gupta, “COX-2 Inhibitor Celecoxib Prevents Chronic Morphine-Induced Promotion of Angiogenesis, Tumour Growth, Metastasis and Mortality, without Compromising Analgesia,” British Journal of Cancer 97, no. 11 (2007): 1523–31.
  • Raja M. Abdel-Majid, and Jean S. Marshall, “Prostaglandin E2 Induces Degranulation-Independent Production of Vascular Endothelial Growth Factor by Human Mast Cells,” Journal of Immunology (Baltimore, Md.: 1950) 172, no. 2 (2004): 1227–36.
  • Lisa Koodie, Hongyan Yuan, Jeffery A. Pumper, Haidong Yu, Richard Charboneau, Sundaram Ramkrishnan, and Sabita Roy, “Morphine Inhibits Migration of Tumor-Infiltrating Leukocytes and Suppresses Angiogenesis Associated with Tumor Growth in Mice,” The American Journal of Pathology 184, no. 4 (2014): 1073–84.
  • Samira Khabbazi, Zeyad D. Nassar, Yannick Goumon, and Marie-Odile Parat, “Morphine Decreases the Pro-Angiogenic Interaction between Breast Cancer Cells and Macrophages in Vitro,” Scientific Reports 6, no. 6 (2016): 31572.
  • Lisa Koodie, Sundaram Ramakrishnan, and Sabita Roy, “Morphine Suppresses Tumor Angiogenesis through a HIF-1alpha/p38MAPK Pathway,” The American Journal of Pathology 177, no. 2 (2010): 984–97.
  • Sudha Balasubramanian, Sundaram Ramakrishnan, Richard Charboneau, Jinghua Wang, Roderick A. Barke, and Sabita Roy, “Morphine Sulfate Inhibits Hypoxia-Induced Vascular Endothelial Growth Factor Expression in Endothelial Cells and Cardiac Myocytes,” Journal of Molecular and Cellular Cardiology 33, no. 12 (2001): 2179–87.
  • Abbas Shafiee, Massoud Amanlou, Hassan Farsam, Ahmad Reza Dehpour, F. Mir-Ershadi, and Alireza Mani, “Synthesis and Pharmacological Activity of Thebaine-Derived μ-Opioid Receptor Agonists,” Pharmaceutica Acta Helvetiae 73, no. 5 (1999): 251–4.
  • Niusha Sharifi, Effat Souri, Seyed Ali Ziai, Gholamreza Amin, Mohsen Amini, and Massoud Amanlou, “Isolation, Identification and Molecular Docking Studies of a New Isolated Compound, from Onopordon Acanthium: A Novel Angiotensin Converting Enzyme (ACE) Inhibitor,” Journal of Ethnopharmacology 148, no. 3 (2013): 934–9.
  • Garrett M. Morris, Ruth Huey, William Lindstrom, Michel F. Sanner, Richard K. Belew, David S. Goodsell, and Arthur J. Olson, “Autodock4 and AutoDockTools4: Automated Docking with Selective Receptor Flexiblity,” Journal of Computational Chemistry 30, no. 16 (2009): 2785–91.
  • Xu Dai, Hong-jin Song, Shi-gang Cui, Ting Wang, Qian Liu, and Rui Wang, “The Stimulative Effects of Endogenous Opioids on Endothelial Cell Proliferation, Migration and Angiogenesis in Vitro,” European Journal of Pharmacology 628, no. 1–3 (2010): 42–50.
  • Michel Barrot, “Tests and Models of Nociception and Pain in Rodents,” Neuroscience 211, no. 211 (2012): 39–50.
  • Tsutomu Suzuki, Takuko Sawada, Koji Kawai, and Yasunobu Ishihara, “Pharmacological Profile of TAN-452, a Novel Peripherally Acting Opioid Receptor Antagonist for the Treatment of Opioid-Induced Bowel Syndromes,” Life Sciences 215, no. 215 (2018): 246–52.
  • E. E. Shults, M. M. Shakirov, G. A. Tolstikov, V. N. Kalinin, and G. Schmidhammer, “Thebaine Adducts with Maleimides. Synthesis and Transformations,” Russian Journal of Organic Chemistry 41, no. 8 (2005): 1132–44.
  • Teresa Kaserer, Aquilino Lantero, Helmut Schmidhammer, Mariana Spetea, and Daniela Schuster, “μ Opioid Receptor: Novel Antagonists and Structural Modeling,” Scientific Reports 6, no. 6 (2016): 21548.
  • Parker W. De Waal, Jingjing Shi, Erli You, Xiaoxi Wang, Karsten Melcher, Yi Jiang, H. Eric Xu, and Bradley M. Dickson, “Molecular Mechanisms of Fentanyl Mediated β-Arrestin Biased Signaling,” PLoS Computational Biology 16, no. 4 (2020): e1007394. ‏
  • John Blebea, James E. Mazo, Todd K. Kihara, Jonathan-Hien Vu, Patricia J. McLaughlin, Robert G. Atnip, and Ian S. Zagon, “Opioid Growth Factor Modulates Angiogenesis,” Journal of Vascular Surgery 32, no. 2 (2000): 364–73.
  • Andrew P. Feinberg, Ian Creese, and Solomon H. Snyder, “The Opiate Receptor: A Model Explaining Structure-Activity Relationships of Opiate Agonists and Antagonists,” Proceedings of the National Academy of Sciences of the United States of America 73, no. 11 (1976): 4215–9.
  • Yan Wu, Patricia J. Mclaughlin, and Ian S. Zagon, “Ontogeny of the Opioid Growth Factor,[Met5]‐Enkephalin, Preproenkephalin Gene Expression, and the ζ Opioid Receptor in the Developing and Adult Aorta of Rat,” Developmental Dynamics 211, no. 4 (1998): 327–37.
  • Ian S. Zagon, Yan Wu, and Patricia J. McLaughlin, “Opioid Growth Factor-Dependent DNA Synthesis in the Neonatal Rat Aorta,” The American Journal of Physiology 270, no. 1 Pt 2 (1996): R22–32.
  • Xu Dai, Shi-gang Cui, Ting Wang, Qian Liu, Hong-jin Song, and Rui Wang, “Endogenous Opioid Peptides, Endomorphin-1 and -2 and Deltorphin I, Stimulate Angiogenesis in the CAM Assay,” European Journal of Pharmacology 579, no. 1–3 (2008): 269–75.
  • Banafsheh Afsharimani, Peter Cabot, and Marie-Odile Parat, “Morphine and Tumor Growth and Metastasis,” Cancer Metastasis Reviews 30, no. 2 (2011): 225–38.
  • Martina Ondrovics, Andrea Hoelbl-Kovacic, and Daniela Alexandra Fux, “Opioids: Modulators of Angiogenesis in Wound Healing and Cancer,” Oncotarget 8, no. 15 (2017): 25783–96.
  • George B. Stefano, Alan Hartman, Thomas V. Bilfinger, Harold I. Magazine, Yu Liu, Federico Casares, and Michael S. Goligorsky, “Presence of the mu3 Opiate Receptor in Endothelial Cells Coupling to Nitric Oxide Production and Vasodilation,” The Journal of Biological Chemistry 270, no. 51 (1995): 30290–3.
  • Caterina Fimiani, Dwight Mattocks, Francesco Cavani, Michel Salzet, Dale G. Deutsch, Stephen Pryor, Thomas V. Bilfinger, and George B. Stefano, “Morphine and Anandamide Stimulate Intracellular Calcium Transients in Human Arterial Endothelial Cells: Coupling to Nitric Oxide Release,” Cellular Signalling 11, no. 3 (1999): 189–93.
  • Kalpna Gupta, Smita Kshirsagar, Liming Chang, Robert Schwartz, Ping-Y. Law, Doug Yee, and Robert P. Hebbel, “Morphine Stimulates Angiogenesis by Activating Proangiogenic and Survival-Promoting Signaling and Promotes Breast Tumor Growth,” Cancer Research no. 62 (2002): 4491–8.
  • Chen-Fuh Lam, Pei-Jung Chang, Yu-Sheng Huang, Yen-Hui Sung, Chien-Chi Huang, Ming-Wei Lin, Yen-Chin Liu, and Yu-Chuan Tsai, “Prolonged Use of High-Dose Morphine Impairs Angiogenesis and Mobilization of Endothelial Progenitor Cells in Mice,” Anesthesia and Analgesia 107, no. 2 (2008): 686–92.
  • Aurelio Pasi, Baoxi Qu, Rudolf Steiner, Hans-Jörg Senn, Walter Bär, and Fathy S. Messiha, “Angiogenesis: Modulation with Opioids,” General Pharmacology 22, no. 6 (1991): 1077–9.
  • Po-Ni Hsiao, Ming-Cheng Chang, Wen-Fang Cheng, Chi-An Chen, Han-Wei Lin, Chang-Yao Hsieh, and Wei-Zen Sun, “Morphine Induces Apoptosis of Human Endothelial Cells through Nitric Oxide and Reactive Oxygen Species Pathways,” Toxicology 256, no. 1–2 (2009): 83–91.
  • Negar Faramarzi, Ata Abbasi, Seyed M. Tavangar, Marjan Mazouchi, and Ahmad R. Dehpour, “Opioid Receptor Antagonist Promotes Angiogenesis in Bile Duct Ligated Rats,” Journal of Gastroenterology and Hepatology 24, no. 7 (2009): 1226–9.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.