Honey and Cancer: Current Status and Future Directions
Abstract
:1. Introduction
2. Honey and Cancer
2.1. The Antiproliferative Properties of Honey
2.2. Modulation of Growth Factor Signalling by Honey
2.3. The Apoptotic Properties of Honey
2.4. The Anti-Inflammatory and Immunomodulatory Properties of Honey
2.5. The Anti-Angiogenic Potential of Honey
2.6. The Anti-Invasive Properties of Honey
3. Honey for Chemoprevention and as an Adjunct to Anticancer Drugs
4. Safety
5. Conclusions
Acknowledgments
Conflicts of Interest
References
- Siegel, R.L.; Miller, K.D.; Jemal, A. Cancer Statistics, 2016. CA Cancer J. Clin. 2016, 66, 7–30. [Google Scholar] [CrossRef] [PubMed]
- Anand, P.; Kunnumakkara, A.B.; Sundaram, C.; Harikumar, K.B.; Tharakan, S.T.; Lai, O.S.; Sung, B.; Aggarwal, B.B. Cancer is a preventable disease that requires major lifestyle changes. Pharm. Res. 2008, 25, 2097–2116. [Google Scholar] [CrossRef] [PubMed]
- Irigaray, P.; Newby, J.A.; Clapp, R.; Hardell, L.; Howard, V.; Montagnier, L.; Epstein, S.; Belpomme, D. Lifestyle-related factors and environmental agents causing cancer: An overview. Biomed. Pharmacother. 2007, 61, 640–658. [Google Scholar] [CrossRef] [PubMed]
- Alvarez-Suarez, J.M.; Tulipani, S.; Díaz, D.; Estevez, Y.; Romandini, S.; Giampieri, F.; Damiani, E.; Astolfi, P.; Bompadre, S.; Battino, M. Antioxidant and antimicrobial capacity of several monofloral Cuban honeys and their correlation with color, polyphenol content and other chemical compounds. Food Chem. Toxicol. 2010, 48, 2490–2499. [Google Scholar] [CrossRef] [PubMed]
- Alvarez-Suarez, J.M.; González-Paramás, A.M.; Santos-Buelga, C.; Battino, M. Antioxidant characterization of native monofloral Cuban honeys. J. Agric. Food Chem. 2010, 58, 9817–9824. [Google Scholar] [CrossRef] [PubMed]
- Da Silva, P.M.; Gauche, C.; Gonzaga, L.V.; Costa, A.C.O.; Fett, R. Honey: Chemical composition, stability and authenticity. Food Chem. 2016, 196, 309–323. [Google Scholar] [CrossRef] [PubMed]
- Pasini, F.; Gardini, S.; Marcazzan, G.L.; Caboni, M.F. Buckwheat honeys: Screening of composition and properties. Food Chem. 2013, 141, 2802–2811. [Google Scholar] [CrossRef] [PubMed]
- Liou, G.-Y.; Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 2010, 44, 479–496. [Google Scholar] [CrossRef] [PubMed]
- Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef] [PubMed]
- Gorjanović, S.Ž.; Alvarez-Suarez, J.M.; Novaković, M.M.; Pastor, F.T.; Pezo, L.; Battino, M.; Sužnjević, D.Ž. Comparative analysis of antioxidant activity of honey of different floral sources using recently developed polarographic and various spectrophotometric assays. J. Food Compos. Anal. 2013, 30, 13–18. [Google Scholar] [CrossRef]
- Alvarez-Suarez, J.M.; Giampieri, F.; González-Paramás, A.M.; Damiani, E.; Astolfi, P.; Martinez-Sanchez, G.; Bompadre, S.; Quiles, J.L.; Santos-Buelga, C.; Battino, M. Phenolics from monofloral honeys protect human erythrocyte membranes against oxidative damage. Food Chem. Toxicol. 2012, 50, 1508–1516. [Google Scholar] [CrossRef] [PubMed]
- Kassim, M.; Achoui, M.; Mustafa, M.R.; Mohd, M.A.; Yusoff, K.M. Ellagic acid, phenolic acids, and flavonoids in Malaysian honey extracts demonstrate in vitro anti-inflammatory activity. Nutr. Res. 2010, 30, 650–659. [Google Scholar] [CrossRef] [PubMed]
- Bertoncelj, J.; Dobersek, U.; Jamnik, M.; Golob, T. Evaluation of the phenolic content, antioxidant activity and colour of Slovenian honey. Food Chem. 2007, 105, 822–828. [Google Scholar] [CrossRef]
- Vallianou, N.G.; Gounari, P.; Skourtis, A.; Panagos, J.; Kazazis, C. Honey and its anti-inflammatory, anti-bacterial and anti-oxidant properties. Gen. Med. 2014, 2, 132. [Google Scholar] [CrossRef]
- Van den Berg, A.J.J.; van den Worm, E.; van Ufford, H.C.Q.; Halkes, S.B.A.; Hoekstra, M.J.; Beukelman, C.J. An in vitro examination of the antioxidant and anti-inflammatory properties of buckwheat honey. J. Wound Care 2008, 17, 172–179. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, S.; Othman, N. Review of the medicinal effects of tualang honey and a comparison with manuka honey. Malays. J. Med. Sci. 2013, 20, 6–13. [Google Scholar] [PubMed]
- Sergiel, I.; Pohl, P.; Biesaga, M. Characterisation of honeys according to their content of phenolic compounds using high performance liquid chromatography/tandem mass spectrometry. Food Chem. 2014, 145, 404–408. [Google Scholar] [CrossRef] [PubMed]
- Chan, C.W.; Deadman, B.J.; Manley-Harris, M.; Wilkins, A.L.; Alber, D.G.; Harry, E. Analysis of the flavonoid component of bioactive New Zealand mānuka (Leptospermum scoparium) honey and the isolation, characterisation and synthesis of an unusual pyrrole. Food Chem. 2013, 141, 1772–1781. [Google Scholar] [CrossRef] [PubMed]
- Moniruzzaman, M.; Sulaiman, S.A.; Gan, S.H. Phenolic acid and flavonoid composition of malaysian honeys. J. Food Biochem. 2016. [Google Scholar] [CrossRef]
- Khalil, M.I.; Alam, N.; Moniruzzaman, M.; Sulaiman, S.A.; Gan, S.H. Phenolic acid composition and antioxidant properties of Malaysian honeys. J. Food Sci. 2011, 76, C921–C928. [Google Scholar] [CrossRef] [PubMed]
- Wieczorek, J.; Pietrzak, M.; Pomianowski, J.; Wieczorec, Z. Honey as a source of bioactive compounds. Pol. J. Nat. Sci. 2014, 29, 275–285. [Google Scholar]
- Yao, L.; Datta, N.; Tomás-Barberán, F.A.; Ferreres, F.; Martos, I.; Singanusong, R. Flavonoids, phenolic acids and abscisic acid in Australian and New Zealand Leptospermum honeys. Food Chem. 2003, 81, 159–168. [Google Scholar] [CrossRef]
- Ismail, N.I.; Kadir, M.R.A.; Zulkifli, R.M. Isolation and identification of potential antineoplastic bioactive phenolic compounds in malaysian honeys. J. Appl. Pharm. Sci. 2015, 5, 59–66. [Google Scholar] [CrossRef]
- Moniruzzaman, M.; An, C.Y.; Rao, P.V.; Hawlader, M.N.I.; Azlan, S.A.B.M.; Sulaiman, S.A.; Gan, S.H. Identification of phenolic acids and flavonoids in monofloral honey from bangladesh by high performance liquid chromatography: Determination of antioxidant capacity. Biomed. Res. Int. 2014, 2014, 737490. [Google Scholar] [CrossRef] [PubMed]
- Lachman, J.; Hejtmánková, A.; Sýkora, J.; Karban, J.; Orsák, M.; Rygerová, B. Contents of major phenolic and flavonoid antioxidants in selected Czech honey. Czech J. Food Sci. 2010, 28, 412–426. [Google Scholar]
- Schramm, D.; Karim, M.; Schrader, H.; Holt, R.; Cardetti, M.; Keen, C. Honey with high levels of antioxidants can provide protection in healthy human subjects. J. Agric. Food Chem. 2003, 51, 1732–1735. [Google Scholar] [CrossRef] [PubMed]
- Jaganathan, S.K.; Balaji, A.; Vellayappan, M.V.; Asokan, M.K.; Subramanian, A.P.; John, A.A.; Supriyanto, E.; Razak, S.I.; Marvibaigi, M. A review on antiproliferative and apoptotic activities of natural honey. Anticancer Agents Med. Chem. 2015, 15, 48–56. [Google Scholar] [CrossRef] [PubMed]
- Subramanian, A.P.; John, A.A.; Vellayappan, M.V.; Balaji, A.; Jaganathan, S.K.; Mandal, M.; Supriyanto, E. Honey and its phytochemicals: Plausible agents in combating colon cancer through its diversified actions. J. Food Biochem. 2016, 40, 613–629. [Google Scholar] [CrossRef]
- Erejuwa, O.; Sulaiman, S.; Wahab, M. Effects of honey and its mechanisms of action on the development and progression of cancer. Molecules 2014, 19, 2497–2522. [Google Scholar] [CrossRef] [PubMed]
- Allan, J.M.; Travis, L.B. Mechanisms of therapy-related carcinogenesis. Nat. Rev. Cancer 2005, 5, 943–955. [Google Scholar] [CrossRef] [PubMed]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [PubMed]
- Swellam, T.; Miyanaga, N.; Onozawa, M.; Hattori, K.; Kawai, K.; Shimazui, T.; Akaza, H. Antineoplastic activity of honey in an experimental bladder cancer implantation model: In vivo and in vitro studies. Int. J. Urol. 2003, 10, 213–219. [Google Scholar] [CrossRef] [PubMed]
- Jaganathan, S.K.; Mandal, M. Honey Constituents and their apoptotic effect in colon cancer cells. J. ApiProd. ApiMed. Sci. 2009, 1, 29–36. [Google Scholar] [CrossRef]
- Pichichero, E.; Cicconi, R.; Mattei, M.; Muzi, M.G.; Canini, A. Acacia honey and chrysin reduce proliferation of melanoma cells through alterations in cell cycle progression. Int. J. Oncol. 2010, 37, 973–981. [Google Scholar] [PubMed]
- Fernandez-Cabezudo, M.J.; El-Kharrag, R.; Torab, F.; Bashir, G.; George, J.A.; El-Taji, H.; Al-Ramadi, B.K. Intravenous administration of manuka honey inhibits tumor growth and improves host survival when used in combination with chemotherapy in a melanoma mouse model. PLoS ONE 2013, 8, e55993. [Google Scholar] [CrossRef] [PubMed]
- Ghashm, A.A.; Othman, N.H.; Khattak, M.N.; Ismail, N.M.; Saini, R. Antiproliferative effect of Tualang honey on oral squamous cell carcinoma and osteosarcoma cell lines. BMC Complement. Altern. Med. 2010, 10, 49. [Google Scholar] [CrossRef] [PubMed]
- Fauzi, A.N.; Norazmi, M.N.; Yaacob, N.S. Tualang honey induces apoptosis and disrupts the mitochondrial membrane potential of human breast and cervical cancer cell lines. Food Chem. Toxicol. 2011, 49, 871–878. [Google Scholar] [CrossRef] [PubMed]
- Nik Man, N.M.K.; Hassan, R.; Ang, C.Y.; Abdullah, A.D.; Mohd Radzi, M.A.R.; Sulaiman, S.A. Antileukemic effect of tualang honey on acute and chronic leukemia cell lines. Biomed. Res. Int. 2015, 2015, 307094. [Google Scholar] [CrossRef] [PubMed]
- Jubri, Z.; Narayanan, N.; Karim, N.; Ngah, W. Antiproliferative activity and apoptosis induction by gelam honey on liver cancer cell line. Int. J. Appl. Sci. Technol. 2012, 2, 135–141. [Google Scholar]
- Abu, N.M.; Salleh, M.A.M.; Radzman, N.H.M.; Ismail, W.I.W.; Yusof, R.M.; Hassan, H.F. Insulin Sensitivity Enhancement of the Mixture of Tinospora Crispa and Gelam (Melaleuca Cajuputi) Honey and Its Antiproliferative Activity on Hepatocellular Carcinoma, HepG2: A Preliminary Study. J. Med. Res. Dev. 2013, 2, 48–54. [Google Scholar]
- Tsiapara, A.V.; Jaakkola, M.; Chinou, I.; Graikou, K.; Tolonen, T.; Virtanen, V. Bioactivity of Greek honey extracts on breast cancer (MCF-7), prostate cancer (PC-3) and endometrial cancer (Ishikawa) cells: Profile analysis of extracts. Food Chem. 2009, 116, 702–708. [Google Scholar] [CrossRef]
- Van der Woude, H.; Gliszczyńska-Swigło, A.; Struijs, K.; Smeets, A.; Alink, G.M.; Rietjens, I.M.C.M. Biphasic modulation of cell proliferation by quercetin at concentrations physiologically relevant in humans. Cancer Lett. 2003, 200, 41–47. [Google Scholar] [CrossRef]
- Wen, C.T.P.; Hussein, S.Z.; Abdullah, S.; Karim, N.A.; Makpol, S.; Yusof, Y.A.M. Gelam and Nenas Honeys Inhibit Proliferation of HT 29 Colon Cancer Cells by Inducing DNA Damage and Apoptosis while Suppressing Inflammation. Asian Pac. J Cancer Prev. 2012, 13, 1605–1610. [Google Scholar] [CrossRef] [PubMed]
- Kishore, R.K.; Halim, A.S.; Syazana, M.S.N.; Sirajudeen, K.N.S. Tualang honey has higher phenolic content and greater radical scavenging activity compared with other honey sources. Nutr. Res. 2011, 31, 322–325. [Google Scholar] [CrossRef] [PubMed]
- Aliyou, M.; Odunola, O.; Farooq, A.; Mesaik, A.; Choudhary, M.; Fatima, B.; Qureshi, T.A.; Erukainure, O.L. Acacia honey modulates cell cycle progression, pro-inflammatory cytokines and calcium Ions secretion in PC-3 cell line. J. Cancer Sci. Ther. 2012, 4, 401–407. [Google Scholar] [CrossRef]
- Wang, X.-H.; Andrae, L.; Engeseth, N.J. Antimutagenic effect of various honeys and sugars against Trp-p-1. J. Agric. Food Chem. 2002, 50, 6923–6928. [Google Scholar] [CrossRef] [PubMed]
- Medina, R.A.; Owen, G.I. Glucose transporters: Expression, regulation and cancer. Biol. Res. 2002, 35, 9–26. [Google Scholar] [CrossRef] [PubMed]
- Khoo, B.Y.; Chua, S.L.; Balaram, P. Apoptotic effects of chrysin in human cancer cell lines. Int. J. Mol. Sci. 2010, 11, 2188–2199. [Google Scholar] [CrossRef] [PubMed]
- Xuan, H.; Zhang, J.; Wang, Y.; FU, C.; Zhang, W. Anti-tumor activity evaluation of novel chrysin-organotin compound in MCF-7 cells. Bioorg. Med. Chem. Lett. 2016, 26, 570–574. [Google Scholar] [CrossRef] [PubMed]
- Kasala, E.R.; Bodduluru, L.N.; Madana, R.M.; Athira, K.V.; Gogoi, R.; Barua, C.C. Chemopreventive and therapeutic potential of chrysin in cancer: Mechanistic perspectives. Toxicol. Lett. 2015, 233, 214–225. [Google Scholar] [CrossRef] [PubMed]
- Ronnekleiv-Kelly, S.; Nukaya, M.; Diaz-Diaz, C.; Megna, B.; Carney, P.; Geiger, P.; Kennedy, G.D. Aryl hydrocarbon receptor-dependent apoptotic cell death induced by the flavonoid chrysin in human colorectal cancer cells. Cancer Lett. 2016, 370, 91–99. [Google Scholar] [CrossRef] [PubMed]
- Kang, T.B.; Liang, N.C. Studies on the inhibitory effects of quercetin on the growth of HL-60 leukemia cells. Biochem. Pharmacol. 1997, 54, 1013–1018. [Google Scholar] [CrossRef]
- Choi, J.A.; Kim, J.Y.; Lee, J.Y.; Kang, C.M.; Kwon, H.J.; Yoo, Y.D.; Kim, T.W.; Lee, Y.S.; Lee, S.J. Induction of cell cycle arrest and apoptosis in human breast cancer cells by quercetin. Int. J. Oncol. 2001, 19, 837–844. [Google Scholar] [CrossRef] [PubMed]
- Salucci, M.; Stivala, L.A.; Maiani, G.; Bugianesi, R.; Vannini, V. Flavonoids uptake and their effect on cell cycle of human colon adenocarcinoma cells (CaCO2). Br. J. Cancer 2002, 86, 1645–1651. [Google Scholar] [CrossRef] [PubMed]
- Nair, H.K.; Rao, K.V.K.; Aalinkeel, R.; Mahajan, S.; Chawda, R.; Schwartz, S.A. Inhibition of prostate cancer cell colony formation by the flavonoid quercetin correlates with modulation of specific regulatory genes. Clin. Diagn. Lab. Immunol. 2004, 11, 63–69. [Google Scholar] [CrossRef] [PubMed]
- Elattar, T.M.; Virji, A.S. The inhibitory effect of curcumin, genistein, quercetin and cisplatin on the growth of oral cancer cells in vitro. Anticancer Res. 2000, 20, 1733–1738. [Google Scholar] [PubMed]
- Kaneuchi, M.; Sasaki, M.; Tanaka, Y.; Sakuragi, N.; Fujimoto, S.; Dahiya, R. Quercetin regulates growth of Ishikawa cells through the suppression of EGF and cyclin D1. Int. J. Oncol. 2003, 22, 159–164. [Google Scholar] [CrossRef] [PubMed]
- Demiroglu-Zergeroglu, A.; Basara-Cigerim, B.; Kililc, E.; Yanikkaya, G. The investigation of effects of quercetin and Its combination with cisplatin on malignant mesothelioma cells In vitro. J. Biomed. Biotechnol. 2010, 2010, 851589. [Google Scholar] [CrossRef] [PubMed]
- Jaganathan, S.K.; Mandal, M. Antiproliferative effects of honey and of its polyphenols: A review. J. Biomed. Biotechnol. 2009, 2009, 830616. [Google Scholar] [CrossRef] [PubMed]
- Lin, Y.; Shi, R.; Wang, X.; Shen, H.-M. Luteolin, a flavonoid with potential for cancer prevention and therapy. Curr. Cancer Drug Targets. 2008, 8, 634–646. [Google Scholar] [CrossRef] [PubMed]
- Aaronson, S.A. Growth factors and cancer. Science 1991, 254, 1146–1153. [Google Scholar] [CrossRef] [PubMed]
- Witsch, E.; Sela, M.; Yarden, Y. Roles for growth factors in cancer progression. Physiology 2010, 25, 85–101. [Google Scholar] [CrossRef] [PubMed]
- Wagner, E.F.; Nebreda, Á.R. Signal integration by JNK and p38 MAPK pathways in cancer development. Nat. Rev. Cancer 2009, 9, 537–549. [Google Scholar] [CrossRef] [PubMed]
- Tahir, A.A.; Sani, N.F.A.; Murad, N.A.; Makpol, S.; Ngah, W.Z.W.; Yusof, Y.A.M. Combined ginger extract & Gelam honey modulate Ras/ERK and PI3K/AKT pathway genes in colon cancer HT29 cells. Nutr. J. 2015, 14, 1–10. [Google Scholar]
- Alvarez-Suarez, J.M.; Giampieri, F.; Cordero, M.; Gasparrini, M.; Forbes-Hernández, T.Y.; Mazzoni, L.; Afrin, S.; Beltrán-Ayala, P.; González-Paramás, A.M.; Santos-Buelga, C.; et al. Activation of AMPK/Nrf2 signalling by Manuka honey protects human dermal fibroblasts against oxidative damage by improving antioxidant response and mitochondrial function promoting wound healing. J. Funct. Foods 2016, 25, 38–49. [Google Scholar] [CrossRef]
- Wu, J.; Omene, C.; Karkoszka, J.; Bosland, M.; Eckard, J.; Klein, C.B.; Frenkel, K. Caffeic acid phenethyl ester (CAPE), derived from a honeybee product propolis, exhibits a diversity of anti-tumor effects in pre-clinical models of human breast cancer. Cancer Lett. 2011, 308, 43–53. [Google Scholar] [CrossRef] [PubMed]
- Narayanan, B.A.; Re, G.G. IGF-II down regulation associated cell cycle arrest in colon cancer cells exposed to phenolic antioxidant ellagic acid. Anticancer Res. 2001, 21, 359–364. [Google Scholar] [PubMed]
- Nalini, N.; Aranganathan, S.; Kabalimurthy, J. Chemopreventive efficacy of hesperetin (citrus flavonone) against 1,2-dimethylhydrazine-induced rat colon carcinogenesis. Toxicol. Mech. Methods 2012, 22, 397–408. [Google Scholar] [CrossRef] [PubMed]
- Elmore, S. Apoptosis: A review of programmed cell death. Toxicol. Pathol. 2007, 35, 495–516. [Google Scholar] [CrossRef] [PubMed]
- Forbes-Hernández, T.Y.; Giampieri, F.; Gasparrini, M.; Mazzoni, L.; Quiles, J.L.; Alvarez-Suarez, J.M.; Battino, M. The effects of bioactive compounds from plant foods on mitochondrial function: A focus on apoptotic mechanisms. Food Chem. Toxicol. 2014, 68, 154–182. [Google Scholar] [CrossRef] [PubMed]
- Morales, P.; Haza, A.I. Antiproliferative and apoptotic effects of spanish honeys. Pharmacogn. Mag. 2013, 9, 231–237. [Google Scholar] [PubMed]
- Samarghandian, S.; Afshari, J.T.; Davoodi, S. Honey induces apoptosis in renal cell carcinoma. Pharmacogn. Mag. 2011, 7, 46–52. [Google Scholar] [PubMed]
- Tomasin, R.; Gomes-Marcondes, M.C.C. Oral administration of Aloe vera and honey reduces Walker tumour growth by decreasing cell proliferation and increasing apoptosis in tumour tissue. Phyther Res. 2011, 25, 619–623. [Google Scholar] [CrossRef] [PubMed]
- Jaganathan, S.K.; Mandal, M. Involvement of non-protein thiols, mitochondrial dysfunction, reactive oxygen species and p53 in honey-induced apoptosis. Investig. New Drugs 2010, 28, 624–633. [Google Scholar] [CrossRef] [PubMed]
- Sadeghi-Aliabadi, H.; Hamzeh, J.; Mirian, M. Investigation of Astragalus honey and propolis extract’s cytotoxic effect on two human cancer cell lines and their oncogen and proapoptotic gene expression profiles. Adv. Biomed. Res. 2015, 4, 42. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Ma, S.; Liu, B.; Liu, J.; Zhu, R.; Li, M. Chrysin induces cell apoptosis via activation of the p53/Bcl-2/caspase-9 pathway in hepatocellular carcinoma cells. Exp. Ther. Med. 2016, 12, 469–474. [Google Scholar] [CrossRef] [PubMed]
- Li, X.; Wang, J.-N.; Huang, J.-M.; Xiong, X.-K.; Chen, M.-F.; Ong, C.-N.; Shen, H.M.; Yang, X.F. Chrysin promotes tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) induced apoptosis in human cancer cell lines. Toxicol. Vitr. 2011, 25, 630–635. [Google Scholar] [CrossRef] [PubMed]
- Samarghandian, S.; Afshari, J.T.; Davoodi, S. Chrysin reduces proliferation and induces apoptosis in the human prostate cancer cell line pc-3. Clinics (São Paulo) 2011, 66, 1073–1079. [Google Scholar] [CrossRef] [PubMed]
- Shao, J.; Zhang, A.; Qin, W.; Zheng, L.; Zhu, Y.; Chen, X. AMP-activated protein kinase (AMPK) activation is involved in chrysin-induced growth inhibition and apoptosis in cultured A549 lung cancer cells. Biochem. Biophys. Res. Commun. 2012, 423, 448–453. [Google Scholar] [CrossRef] [PubMed]
- Huang, C.; Wei, Y.-X.; Shen, M.-C.; Tu, Y.-H.; Wang, C.-C.; Huang, H.-C. Chrysin, Abundant in Morinda citrifolia Fruit Water-EtOAc Extracts, Combined with Apigenin Synergistically Induced Apoptosis and Inhibited Migration in Human Breast and Liver Cancer Cells. J. Agric. Food Chem. 2016, 64, 4235–4245. [Google Scholar] [CrossRef] [PubMed]
- Woo, K.J.; Jeong, Y.-J.; Park, J.-W.; Kwon, T.K. Chrysin-induced apoptosis is mediated through caspase activation and Akt inactivation in U937 leukemia cells. Biochem. Biophys. Res. Commun. 2004, 325, 1215–1222. [Google Scholar] [CrossRef] [PubMed]
- Su, Q.; Peng, M.; Zhang, Y.; Xu, W.; Darko, K.O.; Tao, T.; Huang, Y.; Tao, X.; Yang, X. Quercetin induces bladder cancer cells apoptosis by activation of AMPK signaling pathway. Am. J. Cancer Res. 2016, 6, 498–508. [Google Scholar] [PubMed]
- Luo, C.; Liu, Y.; Wang, P.; Song, C.; Wang, K.; Dai, L.; Zhang, J.Y.; Ye, H. The effect of quercetin nanoparticle on cervical cancer progression by inducing apoptosis, autophagy and anti-proliferation via JAK2 suppression. Biomed. Pharmacother. 2016, 82, 595–605. [Google Scholar] [CrossRef] [PubMed]
- Yi, L.; Zongyuan, Y.; Cheng, G.; Lingyun, Z.; Guilian, Y.; Wei, G. Quercetin enhances apoptotic effect of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in ovarian cancer cells through reactive oxygen species (ROS) mediated CCAAT enhancer-binding protein homologous protein (CHOP)-death receptor 5 pathway. Cancer Sci. 2014, 105, 520–527. [Google Scholar] [CrossRef] [PubMed]
- Ranganathan, S.; Halagowder, D.; Sivasithambaram, N.D. Quercetin Suppresses Twist to Induce Apoptosis in MCF-7 Breast Cancer Cells. PLoS ONE 2015, 10, e0141370. [Google Scholar] [CrossRef] [PubMed]
- Mishra, S.; Vinayak, M. Ellagic acid induces novel and atypical PKC isoforms and promotes caspase-3 dependent apoptosis by blocking energy metabolism. Nutr. Cancer 2014, 66, 675–681. [Google Scholar] [CrossRef] [PubMed]
- Ho, C.-C.; Huang, A.-C.; Yu, C.-S.; Lien, J.-C.; Wu, S.-H.; Huang, Y.-P.; Huang, H.Y.; Kuo, J.H.; Liao, W.Y.; Yang, J.S. Ellagic acid induces apoptosis in TSGH8301 human bladder cancer cells through the endoplasmic reticulum stress- and mitochondria-dependent signaling pathways. Environ. Toxicol. 2014, 29, 1262–1274. [Google Scholar] [CrossRef] [PubMed]
- Umesalma, S.; Nagendraprabhu, P.; Sudhandiran, G. Ellagic acid inhibits proliferation and induced apoptosis via the Akt signaling pathway in HCT-15 colon adenocarcinoma cells. Mol. Cell. Biochem. 2015, 399, 303–313. [Google Scholar] [CrossRef] [PubMed]
- Dang, Q.; Song, W.; Xu, D.; Ma, Y.; Li, F.; Zeng, J.; Zhu, G.; Wang, X.; Chang, L.S.; He, D.; Li, L. Kaempferol suppresses bladder cancer tumor growth by inhibiting cell proliferation and inducing apoptosis. Mol. Carcinog. 2015, 54, 831–840. [Google Scholar] [CrossRef] [PubMed]
- Xie, F.; Su, M.; Qiu, W.; Zhang, M.; Guo, Z.; Su, B.; Liu, J.; Li, X.; Zhou, L. Kaempferol promotes apoptosis in human bladder cancer cells by inducing the tumor suppressor, PTEN. Int. J. Mol. Sci. 2013, 14, 21215–21226. [Google Scholar] [CrossRef] [PubMed]
- Lee, H.S.; Cho, H.J.; Yu, R.; Lee, K.W.; Chun, H.S.; Park, J.H.Y. Mechanisms underlying apoptosis-inducing effects of Kaempferol in HT-29 human colon cancer cells. Int. J. Mol. Sci. 2014, 15, 2722–2737. [Google Scholar] [CrossRef] [PubMed]
- Tu, L.-Y.; Bai, H.-H.; Cai, J.-Y.; Deng, S.-P. The mechanism of kaempferol induced apoptosis and inhibited proliferation in human cervical cancer SiHa cell: From macro to nano. Scanning 2016. [Google Scholar] [CrossRef] [PubMed]
- Luo, H.; Rankin, G.O.; Li, Z.; Depriest, L.; Chen, Y.C. Kaempferol induces apoptosis in ovarian cancer cells through activating p53 in the intrinsic pathway. Food Chem. 2011, 128, 513–519. [Google Scholar] [CrossRef] [PubMed]
- Kang, G.-Y.; Lee, E.-R.; Kim, J.-H.; Jung, J.W.; Lim, J.; Kim, S.K.; Cho, S.G.; Kim, K.P. Downregulation of PLK-1 expression in kaempferol-induced apoptosis of MCF-7 cells. Eur. J. Pharmacol. 2009, 611, 17–21. [Google Scholar] [CrossRef] [PubMed]
- Mantovani, A. Cancer: Inflaming metastasis. Nature 2009, 457, 36–37. [Google Scholar] [CrossRef] [PubMed]
- Friis, S.; Riis, A.H.; Erichsen, R.; Baron, J.A.; Sørensen, H.T. Low-Dose Aspirin or Nonsteroidal Anti-inflammatory Drug Use and Colorectal Cancer Risk: A Population-Based, Case-Control Study. Ann. Intern. Med. 2015, 163, 347–355. [Google Scholar] [CrossRef] [PubMed]
- Jacobs, E.J.; Rodriguez, C.; Mondul, A.M.; Connell, C.J.; Henley, S.J.; Calle, E.E.; Thun, M.J. A large cohort study of aspirin and other nonsteroidal anti-inflammatory drugs and prostate cancer incidence. J. Natl. Cancer Inst. 2005, 97, 975–980. [Google Scholar] [CrossRef] [PubMed]
- Baandrup, L.; Faber, M.T.; Christensen, J.; Jensen, A.; Andersen, K.K.; Friis, S.; Kjaer, S.K. Nonsteroidal anti-inflammatory drugs and risk of ovarian cancer: Systematic review and meta-analysis of observational studies. Acta Obstet. Gynecol. Scand. 2013, 92, 245–255. [Google Scholar] [CrossRef] [PubMed]
- Fan, Y.; Mao, R.; Yang, J. NF-κB and STAT3 signaling pathways collaboratively link inflammation to cancer. Protein Cell 2013, 4, 176–185. [Google Scholar] [CrossRef] [PubMed]
- Saitoh, Y.; Martínez Bruyn, V.J.; Uota, S.; Hasegawa, A.; Yamamoto, N.; Imoto, I.; Inazawa, J.; Yamaoka, S. Overexpression of NF-κB inducing kinase underlies constitutive NF-κB activation in lung cancer cells. Lung Cancer 2010, 70, 263–270. [Google Scholar] [CrossRef] [PubMed]
- Compagno, M.; Lim, W.K.; Grunn, A.; Nandula, S.V.; Brahmachary, M.; Shen, Q.; Bertoni, F.; Ponzoni, M.; Scandurra, M.; Califano, A.; et al. Mutations of multiple genes cause deregulation of NF-kappaB in diffuse large B-cell lymphoma. Nature 2009, 459, 717–721. [Google Scholar] [CrossRef] [PubMed]
- Kendellen, M.F.; Bradford, J.W.; Lawrence, C.L.; Clark, K.S.; Baldwin, A.S. Canonical and non-canonical NF-κB signaling promotes breast cancer tumor-initiating cells. Oncogene 2014, 33, 1297–1305. [Google Scholar] [CrossRef] [PubMed]
- Zha, W.J.; Qian, Y.; Shen, Y.; Du, Q.; Chen, F.F.; Wu, Z.Z.; Li, X.; Huang, M. Galangin Abrogates Ovalbumin-Induced Airway Inflammation via Negative Regulation of NF-kappa, B. Evid.-Based Complement. Altern. Med. 2013, 2013, 767689. [Google Scholar] [CrossRef] [PubMed]
- Khan, M.S.; Devaraj, H.; Devaraj, N. Chrysin abrogates early hepatocarcinogenesis and induces apoptosis in N-nitrosodiethylamine-induced preneoplastic nodules in rats. Toxicol. Appl. Pharmacol. 2011, 251, 85–94. [Google Scholar] [CrossRef] [PubMed]
- Natarajan, K.; Singh, S.; Burke, T.R.; Grunberger, D.; Aggarwal, B.B. Caffeic acid phenethyl ester is a potent and specific inhibitor of activation of nuclear transcription factor NF-kappa, B. Proc. Natl. Acad. Sci. USA 1996, 93, 9090–9095. [Google Scholar] [CrossRef] [PubMed]
- Batumalaie, K.; Zaman Safi, S.; Mohd Yusof, K.; Shah Ismail, I.; Devi Sekaran, S.; Qvist, R. Effect of gelam honey on the oxidative stress-induced signaling pathways in pancreatic hamster cells. Int. J. Endocrinol. 2013, 2013, 367312. [Google Scholar] [CrossRef] [PubMed]
- Safi, S.Z.; Batumalaie, K.; Qvist, R.; Mohd Yusof, K.; Ismail, I.S. Gelam honey attenuates the oxidative stress-induced inflammatory pathways in pancreatic hamster cells. Evid.-Based Complement. Altern. Med. 2016, 2016, 5843615. [Google Scholar] [CrossRef] [PubMed]
- Candiracci, M.; Piatti, E.; Dominguez-Barragán, M.; García-Antrás, D.; Morgado, B.; Ruano, D.; Gutiérrez, J.F.; Parrado, J.; Castaño, A. Anti-inflammatory activity of a honey flavonoid extract on lipopolysaccharide-activated N13 microglial cells. J. Agric. Food Chem. 2012, 60, 12304–12311. [Google Scholar] [CrossRef] [PubMed]
- Al-Waili, N.S. Natural honey lowers plasma glucose, C-reactive protein, homocysteine, and blood lipids in healthy, diabetic, and hyperlipidemic subjects: Comparison with dextrose and sucrose. J. Med. Food 2004, 7, 100–107. [Google Scholar] [CrossRef] [PubMed]
- Al-Waili, N.S.; Boni, N.S. Natural honey lowers plasma prostaglandin concentrations in normal individuals. J. Med. Food. 2003, 6, 129–133. [Google Scholar] [CrossRef] [PubMed]
- Morteau, O. Prostaglandins and inflammation: The cyclooxygenase controversy. Arch. Immunol. Ther. Exp. (Warsz) 2000, 48, 473–480. [Google Scholar] [PubMed]
- Kassim, M.; Achoui, M.; Mansor, M.; Yusoff, K.M. The inhibitory effects of Gelam honey and its extracts on nitric oxide and prostaglandin E(2) in inflammatory tissues. Fitoterapia 2010, 81, 1196–1201. [Google Scholar] [CrossRef] [PubMed]
- Michaluart, P.; Masferrer, J.L.; Carothers, A.M.; Subbaramaiah, K.; Zweifel, B.S.; Koboldt, C.; Mestre, J.R.; Grunberger, D.; Sacks, P.G.; Tanabe, T.; et al. Inhibitory effects of caffeic acid phenethyl ester on the activity and expression of cyclooxygenase-2 in human oral epithelial cells and in a rat model of inflammation. Cancer Res. 1999, 59, 2347–2352. [Google Scholar] [PubMed]
- García-Mediavilla, V.; Crespo, I.; Collado, P.S.; Esteller, A.; Sánchez-Campos, S.; Tuñón, M.J.; González-Gallego, J. The anti-inflammatory flavones quercetin and kaempferol cause inhibition of inducible nitric oxide synthase, cyclooxygenase-2 and reactive C-protein, and down-regulation of the nuclear factor kappaB pathway in Chang Liver cells. Eur. J. Pharmacol. 2007, 557, 221–229. [Google Scholar] [CrossRef] [PubMed]
- Xie, K. Interleukin-8 and human cancer biology. Cytokine Growth Factor Rev. 2001, 12, 375–391. [Google Scholar] [CrossRef]
- Liu, J.-R.; Ye, Y.-L.; Lin, T.-Y.; Wang, Y.-W.; Peng, C.-C. Effect of floral sources on the antioxidant, antimicrobial, and anti-inflammatory activities of honeys in Taiwan. Food Chem. 2013, 139, 938–943. [Google Scholar] [CrossRef] [PubMed]
- Attia, W.Y.; Gabry, M.S.; El-Shaikh, K.A.; Othman, G.A. The anti-tumor effect of bee honey in Ehrlich ascite tumor model of mice is coincided with stimulation of the immune cells. Egypt. J. Immunol. 2008, 15, 169–183. [Google Scholar] [PubMed]
- Al-Waili, N.S. Effects of daily consumption of honey solution on hematological indices and blood levels of minerals and enzymes in normal individuals. J. Med. Food 2003, 6, 135–140. [Google Scholar] [CrossRef] [PubMed]
- Fukuda, M.; Kobayashi, K.; Hirono, Y.; Miyagawa, M.; Ishida, T.; Ejiogu, E.C.; Sawai, M.; Pinkerton, K.E.; Takeuchi, M. Jungle honey enhances immune function and antitumor activity. Evid.-Based Complement. Altern. Med. 2011, 2011, 908743. [Google Scholar] [CrossRef] [PubMed]
- Tonks, A.; Cooper, R.; Jones, K.; Parton, J.; Tonks, A. Honey stimulates inflammatory cytokine production from monocytes. Cytokine 2003, 21, 242–247. [Google Scholar] [CrossRef]
- Gannabathula, S.; Skinner, M.A.; Rosendale, D.; Greenwood, J.M.; Mutukumira, A.N.; Steinhorn, G.; Stephens, J.; Krissansen, G.W.; Schlothauer, R.C. Arabinogalactan proteins contribute to the immunostimulatory properties of New Zealand honeys. Immunopharmacol. Immunotoxicol. 2012, 34, 598–607. [Google Scholar] [CrossRef] [PubMed]
- Dvorak, H.F. Tumors: Wounds that do not heal. Similarities between tumor stroma generation and wound healing. N. Engl. J. Med. 1986, 315, 1650–1659. [Google Scholar] [PubMed]
- Gupta, M.K.; Qin, R.-Y. Mechanism and its regulation of tumor-induced angiogenesis. World J. Gastroenterol. 2003, 9, 1144–1155. [Google Scholar] [CrossRef] [PubMed]
- Munshi, R.; Bhalerao, S.; Kalekar, S.; Patil, T. Exploration of the angiogenic potential of honey. Br. J. Pharmacol. Res. 2014, 4, 477–489. [Google Scholar] [CrossRef]
- Seno, H.; Oshima, M.; Ishikawa, T.; Oshima, H.; Takaku, K.; Chiba, T.; Narumiya, S.; Taketo, M.M. Cyclooxygenase 2- and Prostaglandin E2 Receptor EP2-dependent Angiogenesis in Apc{Delta}716 Mouse Intestinal Polyps. Cancer Res. 2002, 62, 506–511. [Google Scholar] [PubMed]
- Eteraf-Oskouei, T.; Najafi, M.; Gharehbagheri, A. Natural honey: A new and potent anti-angiogenic agent in the air-pouch model of inflammation. Drug Res. (Stuttg) 2014, 64, 530–536. [Google Scholar] [CrossRef] [PubMed]
- Kadir, E.A.; Sulaiman, S.A.; Yahya, N.K.; Othman, N.H. Inhibitory effects of Tualang Honey on experimental breast cancer in rats: A preliminary study. Asian Pac. J. Cancer Prev. 2013, 14, 2249–2254. [Google Scholar] [CrossRef] [PubMed]
- Lu, P.; Weaver, V.M.; Werb, Z. The extracellular matrix: A dynamic niche in cancer progression. J. Cell Biol. 2012, 196, 395–406. [Google Scholar] [CrossRef] [PubMed]
- Aziz, A.; Rady, H.; Amer, M.; Kiwan, H. Effect of Some Honey Bee Extracts on the Proliferation, Proteolytic and Gelatinolytic Activities of the Hepatocellular Carcinoma HepG2 Cell Line. Aust. J. Basic Appl. Sci. 2009, 3, 2754–2769. [Google Scholar]
- Dornelas, C.A.; Fechine-Jamacaru, F.V.; Albuquerque, I.L.; Magalhães, H.I.F.; Dias, T.A.; Faria, M.H.G.; Alves, M.K.; Rabenhorst, S.H.; de Almeida, P.R.; Lemos, T.L.; et al. Angiogenesis inhibition by green propolis and the angiogenic effect of L-lysine on bladder cancer in rats. Acta Cir. Bras. 2012, 27, 529–536. [Google Scholar] [CrossRef] [PubMed]
- Pedersen, T.X.; Leethanakul, C.; Patel, V.; Mitola, D.; Lund, L.R.; Danø, K.; Johnsen, M.; Gutkind, J.S.; Bugge, T.H. Laser capture microdissection-based in vivo genomic profiling of wound keratinocytes identifies similarities and differences to squamous cell carcinoma. Oncogene 2003, 22, 3964–3976. [Google Scholar] [CrossRef] [PubMed]
- Martin, T.A.; Ye, L.; Sanders, A.J.; Lane, J.; Jiang, W.G. Cancer Invasion and Metastasis: Molecular and Cellular Perspective. In Madam Curie Biosciences Database; Landes Bioscience: Austin, TX, USA, 2000. [Google Scholar]
- Price, J.T.; Thompson, E.W. Mechanisms of tumour invasion and metastasis: Emerging targets for therapy. Expert Opin. Ther. Targets 2002, 6, 217–233. [Google Scholar] [PubMed]
- Oršolić, N.; Bašić, I. Antimetastatic effect of honey. Mellifera 2004, 4, 38–43. [Google Scholar]
- Stetler-Stevenson, W.G. The role of matrix metalloproteinases in tumor invasion, metastasis, and angiogenesis. Surg. Oncol. Clin. N. Am. 2001, 10, 383–392. [Google Scholar] [PubMed]
- Deryugina, E.I.; Quigley, J.P. Matrix metalloproteinases and tumor metastasis. Cancer Metastasis Rev. 2006, 25, 9–34. [Google Scholar] [CrossRef] [PubMed]
- Halbersztadt, A.; Haloń, A.; Pajak, J.; Robaczyński, J.; Rabczynski, J.; St Gabryś, M. The role of matrix metalloproteinases in tumor invasion and metastasis. Ginekol. Polska 2006, 77, 63–71. [Google Scholar]
- Moskwa, J.; Borawska, M.H.; Markiewicz-Zukowska, R.; Puscion-Jakubik, A.; Naliwajko, S.K.; Socha, K.; Soroczynska, J. Polish natural bee honeys are anti-proliferative and anti-metastatic agents in human glioblastoma multiforme U87MG cell line. PLoS ONE 2014, 9, e90533. [Google Scholar] [CrossRef] [PubMed]
- Santos, B.L.; Oliveira, M.N.; Coelho, P.L.C.; Pitanga, B.P.S.; da Silva, A.B.; Adelita, T.; Silva, V.D.; Costa Mde, F.; El-Bachá, R.S.; Tardy, M. Flavonoids suppress human glioblastoma cell growth by inhibiting cell metabolism, migration, and by regulating extracellular matrix proteins and metalloproteinases expression. Chem. Biol. Interact. 2015, 242, 123–138. [Google Scholar] [CrossRef] [PubMed]
- Xia, Y.; Lian, S.; Khoi, P.N.; Yoon, H.J.; Joo, Y.E.; Chay, K.O.; Kim, K.K.; Do Jung, Y. Chrysin inhibits tumor promoter-induced MMP-9 expression by blocking AP-1 via suppression of ERK and JNK pathways in gastric cancer cells. PLoS ONE 2015, 10, e0124007. [Google Scholar] [CrossRef] [PubMed]
- Yang, B.; Huang, J.; Xiang, T.; Yin, X.; Luo, X.; Huang, J.; Luo, F.; Li, H.; Li, H.; Ren, G. Chrysin inhibits metastatic potential of human triple-negative breast cancer cells by modulating matrix metalloproteinase-10, epithelial to mesenchymal transition, and PI3K/Akt signaling pathway. J. Appl. Toxicol. 2014, 34, 105–112. [Google Scholar] [CrossRef] [PubMed]
- Spoerlein, C.; Mahal, K.; Schmidt, H.; Schobert, R. Effects of chrysin, apigenin, genistein and their homoleptic copper(II) complexes on the growth and metastatic potential of cancer cells. J. Inorg. Biochem. 2013, 127, 107–115. [Google Scholar] [CrossRef] [PubMed]
- Peng, C.-Y.; Yang, H.-W.; Chu, Y.-H.; Chang, Y.-C.; Hsieh, M.-J.; Chou, M.-Y.; Yeh, K.T.; Lin, Y.M.; Yang, S.F.; Lin, C.W. Caffeic Acid phenethyl ester inhibits oral cancer cell metastasis by regulating matrix metalloproteinase-2 and the mitogen-activated protein kinase pathway. Evid.-Based Complement. Altern. Med. 2012, 2012, 732578. [Google Scholar] [CrossRef] [PubMed]
- Dziedzic, A.; Kubina, R.; Kabała-Dzik, A.; Wojtyczka, R.D.; Morawiec, T.; Bułdak, R.J. Caffeic acid reduces the viability and migration rate of oral carcinoma cells (SCC-25) exposed to low concentrations of ethanol. Int. J. Mol. Sci. 2014, 15, 18725–18741. [Google Scholar] [CrossRef] [PubMed]
- Bouzaiene, N.N.; Jaziri, S.K.; Kovacic, H.; Chekir-Ghedira, L.; Ghedira, K.; Luis, J. The effects of caffeic, coumaric and ferulic acids on proliferation, superoxide production, adhesion and migration of human tumor cells in vitro. Eur. J. Pharmacol. 2015, 766, 99–105. [Google Scholar] [CrossRef] [PubMed]
- Fraser, S.P.; Hemsley, F.; Djamgoz, M.B.A. Caffeic acid phenethyl ester: Inhibition of metastatic cell behaviours via voltage-gated sodium channel in human breast cancer in vitro. Int. J. Biochem. Cell Biol. 2016, 71, 111–118. [Google Scholar] [CrossRef] [PubMed]
- Kuo, Y.-Y.; Jim, W.-T.; Su, L.-C.; Chung, C.-J.; Lin, C.-Y.; Huo, C.; Tseng, J.C.; Huang, S.H.; Lai, C.J.; Chen, B.C. Caffeic Acid phenethyl ester is a potential therapeutic agent for oral cancer. Int. J. Mol. Sci. 2015, 16, 10748–10766. [Google Scholar] [CrossRef] [PubMed]
- Lin, H.-P.; Lin, C.-Y.; Liu, C.-C.; Su, L.-C.; Huo, C.; Kuo, Y.-Y.; Tseng, J.C.; Hsu, J.M.; Chen, C.K.; Chuu, C.P. Caffeic Acid phenethyl ester as a potential treatment for advanced prostate cancer targeting akt signaling. Int. J. Mol. Sci. 2013, 14, 5264–5283. [Google Scholar] [CrossRef] [PubMed]
- Ho, H.-H.; Chang, C.-S.; Ho, W.-C.; Liao, S.-Y.; Wu, C.-H.; Wang, C.-J. Anti-metastasis effects of gallic acid on gastric cancer cells involves inhibition of NF-κB activity and downregulation of PI3K/AKT/small GTPase signals. Food Chem. Toxicol. 2010, 48, 2508–2516. [Google Scholar] [CrossRef] [PubMed]
- Ho, H.-H.; Chang, C.-S.; Ho, W.-C.; Liao, S.-Y.; Lin, W.-L.; Wang, C.-J. Gallic acid inhibits gastric cancer cells metastasis and invasive growth via increased expression of RhoB, downregulation of AKT/small GTPase signals and inhibition of NF-κB activity. Toxicol. Appl. Pharmacol. 2013, 266, 76–85. [Google Scholar] [CrossRef] [PubMed]
- Liao, C.-L.; Lai, K.-C.; Huang, A.-C.; Yang, J.-S.; Lin, J.-J.; Wu, S.-H.; Gibson Wood, W.; Lin, J.G.; Chung, J.G. Gallic acid inhibits migration and invasion in human osteosarcoma U-2 OS cells through suppressing the matrix metalloproteinase-2/-9, protein kinase B (PKB) and PKC signaling pathways. Food Chem. Toxicol. 2012, 50, 1734–1740. [Google Scholar] [CrossRef] [PubMed]
- Lu, Y.; Jiang, F.; Jiang, H.; Wu, K.; Zheng, X.; Cai, Y.; Katakowski, M.; Chopp, M.; To, S.S. Gallic acid suppresses cell viability, proliferation, invasion and angiogenesis in human glioma cells. Eur. J. Pharmacol. 2010, 641, 102–107. [Google Scholar] [CrossRef] [PubMed]
- Zhao, B.; Hu, M. Gallic acid reduces cell viability, proliferation, invasion and angiogenesis in human cervical cancer cells. Oncol. Lett. 2013, 6, 1749–1755. [Google Scholar] [PubMed]
- Chen, Y.-J.; Lin, K.-N.; Jhang, L.-M.; Huang, C.-H.; Lee, Y.-C.; Chang, L.-S. Gallic acid abolishes the EGFR/Src/Akt/Erk-mediated expression of matrix metalloproteinase-9 in MCF-7 breast cancer cells. Chem. Biol. Interact. 2016, 252, 131–140. [Google Scholar] [CrossRef] [PubMed]
- Jin, U.-H.; Lee, J.-Y.; Kang, S.-K.; Kim, J.-K.; Park, W.-H.; Kim, J.-G.; Moon, S.K.; Kim, C.H. A phenolic compound, 5-caffeoylquinic acid (chlorogenic acid), is a new type and strong matrix metalloproteinase-9 inhibitor: Isolation and identification from methanol extract of Euonymus alatus. Life Sci. 2005, 77, 2760–2769. [Google Scholar] [CrossRef] [PubMed]
- Narayanaswamy, R.; Kok Wai, L.; Ismail, I.S. In Silico Analysis of Selected Honey Constituents as Human Neutrophil Elastase (HNE) and Matrix Metalloproteinases (MMP 2 and 9) Inhibitors. Int. J. Food Prop. 2015, 18, 2155–2164. [Google Scholar] [CrossRef]
- Jeong, Y.-J.; Choi, Y.; Shin, J.-M.; Cho, H.-J.; Kang, J.-H.; Park, K.-K.; Choe, J.Y.; Bae, Y.S.; Han, S.M.; Kim, C.H.; et al. Melittin suppresses EGF-induced cell motility and invasion by inhibiting PI3K/Akt/mTOR signaling pathway in breast cancer cells. Food Chem. Toxicol. 2014, 68, 218–225. [Google Scholar] [CrossRef] [PubMed]
- Othman, N.H.; Ahmed, S.; Sulaiman, S.A. Inhibitory effects of Malaysian tualang honey and Australian/New Zealand Manuka honey in modulating experimental breast cancers induced by N-methyl-N-nitrosourea (mnu): A comparative study. Pathology 2016, 48 (Suppl. S1), S148. [Google Scholar] [CrossRef]
- Orsolic, N.; Terzic, S.; Sver, L.; Basic, I. Honey-bee products in prevention and/or therapy of murine transplantable tumours. J. Sci. Food Agric. 2005, 85, 363–370. [Google Scholar] [CrossRef]
- Yaacob, N.S.; Ismail, N.F. Comparison of cytotoxicity and genotoxicity of 4-hydroxytamoxifen in combination with Tualang honey in MCF-7 and MCF-10A cells. BMC Complement. Altern. Med. 2014, 14, 106. [Google Scholar] [CrossRef] [PubMed]
- Yaacob, S.N.; Nengsih, A.; Norazmi, M.N. Tualang honey promotes apoptotic cell death induced by tamoxifen in breast cancer cell lines. Evid.-Based Complement. Altern. Med. 2013, 2013, 989841. [Google Scholar] [CrossRef] [PubMed]
- Fahmy, M.A.; Hassan, N.H.A.; El-Fiky, S.A.; Elalfy, H.G. A mixture of honey bee products ameliorates the genotoxic side effects of cyclophosphamide. Asian Pac. J. Trop. Dis. 2015, 5, 638–644. [Google Scholar] [CrossRef]
- Münstedt, K.; Voss, B.; Kullmer, U.; Schneider, U.; Hübner, J. Bee pollen and honey for the alleviation of hot flushes and other menopausal symptoms in breast cancer patients. Mol. Clin. Oncol. 2015, 3, 869–874. [Google Scholar] [PubMed]
- Hamad, R.; Jayakumar, C.; Ranganathan, P.; Mohamed, R.; El-Hamamy, M.M.I.; Dessouki, A.A.; Ibrahim, A.; Ramesh, G. Honey feeding protects kidney against cisplatin nephrotoxicity through suppression of inflammation. Clin. Exp. Pharmacol. Physiol. 2015, 42, 843–848. [Google Scholar] [CrossRef] [PubMed]
- Bardy, J.; Slevin, N.J.; Mais, K.L.; Molassiotis, A. A systematic review of honey uses and its potential value within oncology care. J. Clin. Nurs. 2008, 17, 2604–2623. [Google Scholar] [CrossRef] [PubMed]
- Zidan, J.; Shetver, L.; Gershuny, A.; Abzah, A.; Tamam, S.; Stein, M.; Friedman, E. Prevention of chemotherapy-induced neutropenia by special honey intake. Med. Oncol. 2006, 23, 549–552. [Google Scholar] [CrossRef]
- Abdulrhman, M.A.; Hamed, A.A.; Mohamed, S.A.; Hassanen, N.A.A. Effect of honey on febrile neutropenia in children with acute lymphoblastic leukemia: A randomized crossover open-labeled study. Complement. Ther. Med. 2016, 25, 98–103. [Google Scholar] [CrossRef] [PubMed]
- Song, J.J.; Twumasi-Ankrah, P.; Salcido, R. Systematic review and meta-analysis on the use of honey to protect from the effects of radiation-induced oral mucositis. Adv. Skin Wound Care 2012, 25, 23–28. [Google Scholar] [CrossRef] [PubMed]
- Van den Wyngaert, T. Topical honey application to reduce radiation-induced oral mucositis: A therapy too sweet to ignore? J. Evid. Based Dent. Pract. 2012, 12, 203–205. [Google Scholar] [CrossRef] [PubMed]
- Sari Dogan, F.; Ozaydin, V.; Incealtin, O.; Guneysel, O.; Demireller, M. A case of acute hepatitis following mad honey ingestion. Turk. J. Emerg. Med. 2015, 15, 185–186. [Google Scholar] [CrossRef] [PubMed]
- Makpol, S.; Tengku Ahmad, T.A.F.; Jubri, Z.; Rejab, N.; Yusof, N.; Yusof, Y.A.M. Gelam honey acting as a radioprotectant agent in gamma-irradiated human diploid fibroblasts. J. Med. Plants Res. 2012, 6, 129–138. [Google Scholar]
In Vitro Effects of Honey | Cell Line | Honey Type | Reference |
---|---|---|---|
Antiproliferative effects of honey | |||
Decreases cell viability | T24, 253 J, RT4, MBT-2 | Pure unfractionated (Tokyo) | [32] |
MCF-7, PC3, ishikawa | Thyme (Greece) | [41] | |
MDA-MB-231, MCF-7, HeLa | Tualang (Malaysia) | [37] | |
Inhibits cell proliferation | MCF-7, B16.F1, CT26 | Manuka UMF 10+ (New Zealand) | [35] |
HOS (CRL-1543) | Tualang (Malaysia) | [36] | |
OSCC (CRL-1623) | Tualang (Malaysia) | [36] | |
K562, MV4-11 | Tualang (Malaysia) | [38] | |
HepG2 | Gelam (Malaysia) | [40] | |
HT29 | Gelam and Nenas (Malaysia) | [43] | |
B16-F1, A375 | Acacia (Unspecified) | [34] | |
PC3 | Acacia (Pakistan) | [45] | |
Apoptotic properties of honey | |||
Induces apoptosis | ACHN | Multifloral (Iran) | [72] |
T24 | Pure unfractionated (Tokyo) | [32] | |
HCT-15, HT-29 | Pure unfractionated (India) | [74] | |
Induces apoptosis via ROS-independent pathway | HL-60 | Heather, rosemary and polyfloral (Spain) | [71] |
Induces apoptosis via mitochondrial membrane depolarisation | MDA-MB-231, MCF-7, HeLa | Tualang (Malaysia) | [37] |
Increases caspase expression or activation | B16.F1, MCF-7, CT26 | Manuka UMF 10+ (New Zealand) | [35] |
MDA-MB-231, MCF-7, HeLa | Tualang (Malaysia) | [37] | |
HCT-15, HT-29 | Pure unfractionated (India) | [74] | |
HT29 | Gelam (Malaysia) | [64] | |
Increases expression of pro apoptotic proteins | HCT-15, HT-29 | Pure unfractionated (India) | [74] |
Decreases expression of anti-apoptotic proteins | B16.F1, MCF-7, CT26 | Manuka UMF 10+ (New Zealand) | [35] |
HCT-15, HT-29 | Pure unfractionated (India) | [74] | |
HepG2, 5637 | Astragalus (Iran) | [75] | |
Growth factor modulation by honey | |||
Downregulation of RAS/ERK and PI3K/Akt signalling | HT29 | Gelam (Malaysia) | [64] |
Anti-inflammatory and immune-modulatory effects of honey | |||
Reduce activation of NF-κB and MAPK | HIT-T15 | Gelam (Malaysia) | [106] |
Reduces expression of pro-inflammatory cytokines | HIT-T15 | Gelam (Malaysia) | [107] |
Inhibits expression of IL-8 | WiDr | Monofloral (D. longan, L. chinensis, C. maxima and A. formosana) and one multifloral honey (Taiwan) | [116] |
Increases expression of proinflamatory cytokines IL-1β, Il-6, TNF-α | MM6 | Manuka, Pasture (New Zealand) and Jelly bush (Australia) | [120] |
Anti-angiogenic effects of honey | |||
Inhibits extracellular protease and gelatinase activity | HepG2 | Unspecified (Saudi Arabia and Egypt) | [129] |
Anti-invasive effects of honey | |||
Decreases MMP-2 and MMP-9 activity | U87MG | Buckwheat, Multifloral light, Willow and Multifloral dark (Poland) | [138] |
HT29 | Gelam (Malaysia) | [64] |
In Vivo Effects of Honey | Animal Model | Honey Type | Reference |
---|---|---|---|
Apoptotic effects of honey | |||
Tumour growth inhibition | MBT-2 Mouse bladder tumour (C3H/He mice) | Pure unfractionated (Tokyo) (IL, Oral) | [32] |
Syngeneic mouse melanoma model (C57BL/6 mice) | Manuka UMF 10+ (IV) | [35] | |
Lewis Lung Carcinoma/2 (C57BL/6 mice) | Jungle (Nigeria) (IP) | [119] | |
Increases expression of Bax, inhibits expression of Bcl-2, Increases Bax/Bcl-2 ratio | Walker 256 carcinoma (Wistar rats) | Unspecified (Oral) | [73] |
Anti-inflammatory effects of honey | |||
Reduces swelling and oedema with decreased PGE2 levels | Rat paw oedema model (Sprague Dawley rats) | Gelam (Malaysia) (IP) | [112] |
Increases macrophage phagocytic activity, activates T-cells | Ehrlich ascites tumour (Swiss albino mince) | Bee honey (Oral) | [117] |
IL-1β induced neutrophil activation. Increased ROS | Lewis Lung Carcinoma/2 (C57BL/6 mice) | Jungle (Nigeria) (IP) | [119] |
Anti-angiogenic effects of Honey | |||
Reduces tumour growth, increases the number of apoptotic cells. Reduces VEGF, decreases vasculature around the tumour | DMBA-induced breast cancer (Sprague-Dawley rats) | Tualang (Malaysis) | [127] |
Anti-invasive effects of Honey | |||
Antimetastatic effects when used preventatively | Mammary carcinoma (CBA mice) Methylcholanthrene-induced fibrosarcoma (CBA mice) Anaplastic colon adenocarcinoma (Y59 rats) | Wildflower (Croatia) | [134] |
Human Studies | |||
Inflammation | |||
Reduces CRP | Eight subjects | Natural honey | [109] |
Reduces PGE2 | Twelve subjects | Natural unprocessed honey | [110] |
Increases peripheral blood monocyte, lymphocyte and eosinophil count | Ten subjects | Natural honey | [118] |
© 2016 by the authors; licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC-BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Porcza, L.M.; Simms, C.; Chopra, M. Honey and Cancer: Current Status and Future Directions. Diseases 2016, 4, 30. https://doi.org/10.3390/diseases4040030
Porcza LM, Simms C, Chopra M. Honey and Cancer: Current Status and Future Directions. Diseases. 2016; 4(4):30. https://doi.org/10.3390/diseases4040030
Chicago/Turabian StylePorcza, Laura M., Claire Simms, and Mridula Chopra. 2016. "Honey and Cancer: Current Status and Future Directions" Diseases 4, no. 4: 30. https://doi.org/10.3390/diseases4040030