Xanthomicrol: Effective therapy for cancer treatment

Nenhuma Miniatura disponível
Citações na Scopus
3
Tipo de produção
article
Data de publicação
2023
Título da Revista
ISSN da Revista
Título do Volume
Editora
ELSEVIER INC.
Autores
POORMOLAIE, N.
MOHAMMADI, M.
MIR, A.
ASADI, M.
KARAROUDI, A. N.
RASHIDI, M.
MAROUFI, N. F.
Citação
TOXICOLOGY REPORTS, v.10, p.436-440, 2023
Projetos de Pesquisa
Unidades Organizacionais
Fascículo
Resumo
Cancer treatment is one of the main challenges of global health. For decades, researchers have been trying to find anti-cancer compounds with minimal side effects. In recent years, flavonoids, as a group of polyphenolic compounds, have attracted the attention of researchers due to their beneficial effects on health. Xanthomicrol is one of the flavonoids that has the ability to inhibit growth, proliferation, survival and cell invasion and ultimately tumor progression. Xanthomicrol, as active anti-cancer compounds, can be effective in the prevention and treatment of cancer. Therefore, the use of flavonoids can be suggested as a treatment along with other medicinal agents. It is obvious that additional investigations in cellular levels and animal models are still needed. In this review article, the effects of xanthomicrol on various cancers have been reviewed.
Palavras-chave
Apoptosis, Cancer, Treatment, Xanthomicrol
Referências
  1. Pourmohammad P., Et al., Potential therapeutic effects of melatonin mediate via miRNAs in cancer, Biochem. Genet., 60, pp. 1-23, (2021)
  2. Hajipour H., Et al., Improved anticancer effects of epigallocatechin gallate using RGD-containing nanostructured lipid carriers, Artif. Cells Nanomed., Biotechnol., 46, sup1, pp. 283-292, (2018)
  3. Maroufi N.F., Et al., The apatinib inhibits breast cancer cell line MDA-MB-231 in vitro by inducing apoptosis, cell cycle arrest, and regulating nuclear factor-κB (NF-κB) and mitogen-activated protein kinase (MAPK) signaling pathways, Breast Cancer, 27, 4, pp. 613-620, (2020)
  4. Mohammadian J., Et al., Formulation of Stattic as STAT3 inhibitor in nanostructured lipid carriers (NLCs) enhances efficacy of doxorubicin in melanoma cancer cells, Naunyn-Schmiede 'S. Arch. Pharmacol., 393, 12, pp. 2315-2323, (2020)
  5. Talib W.H., Et al., Melatonin in cancer treatment: current knowledge and future opportunities, Molecules, 26, 9, (2021)
  6. van den Heerik A.S.V., Et al., Adjuvant therapy for endometrial cancer in the era of molecular classification: radiotherapy, chemoradiation and novel targets for therapy, Int. J. Gynecol. Cancer, 31, (2021)
  7. Haiaty S., Et al., Targeting vasculogenic mimicry by phytochemicals: a potential opportunity for cancer therapy, IUBMB life, 72, 5, pp. 825-841, (2020)
  8. Yuan M., Et al., The role of bioactive compounds in natural products extracted from plants in cancer treatment and their mechanisms related to anticancer effects, Oxid. Med. Cell. Longev., 2022, pp. 1-19, (2022)
  9. Amin A.R., Et al., Perspectives for cancer prevention with natural compounds, J. Clin. Oncol., 27, 16, (2009)
  10. Wen X., Walle T., Methylated flavonoids have greatly improved intestinal absorption and metabolic stability, Drug Metab. Dispos., 34, 10, pp. 1786-1792, (2006)
  11. Koirala N., Et al., Methylation of flavonoids: chemical structures, bioactivities, progress and perspectives for biotechnological production, Enzym. Microb. Technol., 86, pp. 103-116, (2016)
  12. Mottaghi S., Abbaszadeh H., The anticarcinogenic and anticancer effects of the dietary flavonoid, morin: Current status, challenges, and future perspectives, Phytother. Res., 35, 12, pp. 6843-6861, (2021)
  13. Attari F., Et al., Inhibitory effect of flavonoid xanthomicrol on triple-negative breast tumor via regulation of cancer-associated microRNAs, Phytother. Res., 35, 4, pp. 1967-1982, (2021)
  14. Fattahi M., Et al., Xanthomicrol: a comprehensive review of its chemistry, distribution, biosynthesis and pharmacological activity, Mini Rev. Med. Chem., 14, 9, pp. 725-733, (2014)
  15. Jalezadeh A., Et al., Investigation of structural, electronic, and antioxidant properties of calycopetrin and xanthomicrol as two polymethoxylated flavones using DFT calculations, Struct. Chem., pp. 1-10, (2022)
  16. Soureshjani E.H., Babaheydari A.K., Saberi E., DNA Methyltransferases directed anti-cancerous plant medicine (Xanthomicrol and Galloyl) based molecular docking and dynamics simulation, Comput. Mol. Biosci., 5, 2, (2015)
  17. Varisli B., Et al., Hesperidin attenuates oxidative stress, inflammation, apoptosis, and cardiac dysfunction in sodium fluoride-Induced cardiotoxicity in rats, Cardiovasc. Toxicol., 22, 8, pp. 727-735, (2022)
  18. Varisli B., Et al., Chrysin mitigates diclofenac-induced hepatotoxicity by modulating oxidative stress, apoptosis, autophagy and endoplasmic reticulum stress in rats, Mol. Biol. Rep., pp. 1-10, (2022)
  19. Caglayan C., Et al., Neuroprotective effects of 18β-glycyrrhetinic acid against bisphenol A-induced neurotoxicity in rats: involvement of neuronal apoptosis, endoplasmic reticulum stress and JAK1/STAT1 signaling pathway, Metab. brain Dis., 37, 6, pp. 1931-1940, (2022)
  20. Zamani S.-S., Et al., Pharmacokinetics of calycopterin and xanthmicrol, two polymethoxylated hydroxyflavones with anti-angiogenic activities from Dracocephalum kotschyi Bioss. DARU, J. Pharm. Sci., 24, 1, pp. 1-10, (2016)
  21. Sotillo W.S., Et al., Breast cancer cell line toxicity of a flavonoid isolated from Baccharis densiflora, BMC Complement. Med. Ther., 21, 1, pp. 1-11, (2021)
  22. Jodynis-Liebert J., Kujawska M., Biphasic dose-response induced by phytochemicals: experimental evidence, J. Clin. Med., 9, 3, (2020)
  23. Croom E., Metabolism of xenobiotics of human environments, Prog. Mol. Biol. Transl. Sci., 112, pp. 31-88, (2012)
  24. Nair M.P., Et al., The flavonoid quercetin inhibits proinflammatory cytokine (tumor necrosis factor alpha) gene expression in normal peripheral blood mononuclear cells via modulation of the NF-κβ system, Clin. Vaccin. Immunol., 13, 3, pp. 319-328, (2006)
  25. Yu M., Et al., Targeting transmembrane TNF-α suppresses breast cancer growthanti-tmTNF-α antibody with antitumor activities, Cancer Res., 73, 13, pp. 4061-4074, (2013)
  26. Soheilyfar S., Et al., In vivo and in vitro impact of miR-31 and miR-143 on the suppression of metastasis and invasion in breast cancer, J. BUON, 23, 5, pp. 1290-1296, (2018)
  27. Ma Y., Et al., miR-27a regulates the growth, colony formation and migration of pancreatic cancer cells by targeting Sprouty2, Cancer Lett., 298, 2, pp. 150-158, (2010)
  28. Yin H., Et al., Progress on the relationship between miR-125 family and tumorigenesis, Exp. Cell Res., 339, 2, pp. 252-260, (2015)
  29. Zhang Z., Et al., Upregulation of p72 enhances malignant migration and invasion of glioma cells by repressing Beclin1 expression, Biochem. (Mosc.), 81, 6, pp. 574-582, (2016)
  30. Wang R., Et al., Functional role of miR-34 family in human cancer, Curr. Drug Targets, 14, 10, pp. 1185-1191, (2013)
  31. Nieddu M., Et al., Xanthomicrol activity in cancer HeLa cells: comparison with other natural methoxylated flavones, Molecules, 28, 2, (2023)
  32. Bergers G., Hanahan D., Modes of resistance to anti-angiogenic therapy, Nat. Rev. Cancer, 8, 8, pp. 592-603, (2008)
  33. Maroufi N.F., Et al., Therapeutic potentials of Apatinib in cancer treatment: Possible mechanisms and clinical relevance, Life Sci., 241, (2020)
  34. Sagar S., Yance D., Wong R., Natural health products that inhibit angiogenesis: a potential source for investigational new agents to treat cancer—Part 1, Curr. Oncol., 13, 1, pp. 14-26, (2006)
  35. Kikuchi S., Et al., Involvement of extracellular vesicles in vascular-related functions in cancer progression and metastasis, Int. J. Mol. Sci., 20, 10, (2019)
  36. Ahmadi M., Rezaie J., Tumor cells derived-exosomes as angiogenenic agents: possible therapeutic implications, J. Transl. Med., 18, 1, pp. 1-17, (2020)
  37. Melincovici C.S., Et al., Vascular endothelial growth factor (VEGF)-key factor in normal and pathological angiogenesis, Rom. J. Morphol. Embryol., 59, 2, pp. 455-467, (2018)
  38. Quintero-Fabian S., Et al., Role of matrix metalloproteinases in angiogenesis and cancer, Front. Oncol., 9, (2019)
  39. Ghazizadeh F., Et al., Xanthomicrol exerts antiangiogenic and antitumor effects in a mouse melanoma (B16F10) allograft model, Evid. -Based Complement. Altern. Med., (2020)
  40. Lin Z.-Z., Et al., Xanthomicrol suppresses human hepatocellular carcinoma cells migration and invasion ability via Μu-opioid receptor, J. Pharm. Pharmacol., 74, 1, pp. 139-146, (2022)
  41. Patel S., Emerging adjuvant therapy for cancer: propolis and its constituents, J. Diet. Suppl., 13, 3, pp. 245-268, (2016)
  42. Agha A., Tarhini A.A., Adjuvant therapy for melanoma, Curr. Oncol. Rep., 19, 5, pp. 1-9, (2017)
  43. Bagri A., Et al., Use of anti-VEGF adjuvant therapy in cancer: challenges and rationale, Trends Mol. Med., 16, 3, pp. 122-132, (2010)
  44. Plate K.H., Scholz A., Dumont D.J., Tumor angiogenesis and anti-angiogenic therapy in malignant gliomas revisited, Acta Neuropathol., 124, 6, pp. 763-775, (2012)
  45. Hendrix M.J., Et al., Tumor cell vascular mimicry: novel targeting opportunity in melanoma, Pharmacol. Ther., 159, pp. 83-92, (2016)
  46. Maniotis A.J., Et al., Vascular channel formation by human melanoma cells in vivo and in vitro: vasculogenic mimicry, Am. J. Pathol., 155, 3, pp. 739-752, (1999)
  47. Xu Y., Et al., Short-term anti-vascular endothelial growth factor treatment elicits vasculogenic mimicry formation of tumors to accelerate metastasis, J. Exp. Clin. Cancer Res., 31, 1, pp. 1-7, (2012)
  48. Fathi Maroufi N., Et al., Vascular mimicry: changing the therapeutic paradigms in cancer, Mol. Biol. Rep., 47, 6, pp. 4749-4765, (2020)
  49. Hayes D.F., Bevacizumab treatment for solid tumors: boon or bust?, Jama, 305, 5, pp. 506-508, (2011)
  50. Wang M., Et al., Myricetin reverses epithelial–endothelial transition and inhibits vasculogenic mimicry and angiogenesis of hepatocellular carcinoma by directly targeting PAR1, Phytother. Res., 36, 4, pp. 1807-1821, (2022)
  51. Zang M., Et al., Luteolin suppresses angiogenesis and vasculogenic mimicry formation through inhibiting Notch1-VEGF signaling in gastric cancer, Biochem. Biophys. Res. Commun., 490, 3, pp. 913-919, (2017)
  52. Chiablaem K., Et al., Curcumin suppresses vasculogenic mimicry capacity of hepatocellular carcinoma cells through STAT3 and PI3K/AKT inhibition, Anticancer Res., 34, 4, pp. 1857-1864, (2014)
  53. Wu Q., Et al., Multi-drug resistance in cancer chemotherapeutics: mechanisms and lab approaches, Cancer Lett., 347, 2, pp. 159-166, (2014)
  54. Avner B.S., Fialho A.M., Chakrabarty A.M., Overcoming drug resistance in multi-drug resistant cancers and microorganisms: a conceptual framework, Bioengineered, 3, 5, pp. 262-270, (2012)
  55. Bugde P., Et al., The therapeutic potential of targeting ABC transporters to combat multi-drug resistance, Expert Opin. Ther. Targets, 21, 5, pp. 511-530, (2017)
  56. Niu S., Et al., A novel chitosan-based nanomedicine for multi-drug resistant breast cancer therapy, Chem. Eng. J., 369, pp. 134-149, (2019)
  57. Liskova A., Et al., Flavonoids as an effective sensitizer for anti-cancer therapy: insights into multi-faceted mechanisms and applicability towards individualized patient profiles, EPMA J., 12, 2, pp. 155-176, (2021)