Next Article in Journal
Bilateral Shifts in Deuterium Supply Similarly Change Physiology of the Pituitary–Thyroid Axis, but Differentially Influence Na+/I Symporter Production
Previous Article in Journal
Artificial Intelligence That Predicts Sensitizing Potential of Cosmetic Ingredients with Accuracy Comparable to Animal and In Vitro Tests—How Does the Infotechnomics Compare to Other “Omics” in the Cosmetics Safety Assessment?
Previous Article in Special Issue
Dietary Salt Administration Decreases Enterotoxigenic Bacteroides fragilis (ETBF)-Promoted Tumorigenesis via Inhibition of Colonic Inflammation
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Editorial

The Effect of Dietary Factors on Cancer

1
Department of Clinical Sciences and Translational Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy
2
Departmental Faculty of Medicine and Surgery, Saint Camillus International University of Health and Medical Sciences, 00131 Rome, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(7), 6802; https://doi.org/10.3390/ijms24076802
Submission received: 28 March 2023 / Accepted: 3 April 2023 / Published: 6 April 2023
(This article belongs to the Special Issue The Effect of Dietary Factors on Cancer 2.0)
The effects of dietary factors on cancer have been widely studied for several decades. The American Cancer Society estimates that in 2023, approximately 1,958,310 new cases of cancer will be diagnosed, and 609,820 cancer deaths will occur in the United States [1]. Diet has been identified as a key modifiable risk factor for several types of cancer, including colorectal, breast, prostate, and lung cancer. Indeed, the composition of an individual’s diet can affect nutritional biomarkers (biomarkers of exposure, effect/function, and health/disease and physiological status), which can, in turn, affect cancer risk [2,3]. This Special Issue, “The Effect of Dietary Factors on Cancer 2.0”, in the International Journal of Molecular Sciences, comprises eight original research papers and nine review articles. These papers highlight the current research on the anti-tumoral effects of several dietary factors, including their in vitro and in vivo mechanisms of action, in different types of cancers. The nutraceuticals, which are natural components of the diet, including purified substances from foods, plant extracts, dietary supplements, vitamins, and phytonutrients, possess many biological activities (anti-microbial, antioxidant, anti-inflammatory, anti-viral, anti-cancer, and immunomodulatory) that can influence the prevention and/or treatment of diseases, such as cancer [4,5,6,7,8].
The review by Matsushita et al. suggested that, although the relationship between diet, nutrition, and prostate cancer is not well-understood, an intervention targeting dietary patterns may be a potential avenue for prostate cancer prevention. Indeed, gut microbiota and nutrients, such as fat, protein, carbohydrates, vitamins, polyphenols, and also zinc, may affect the development and progression of prostate cancer [9,10]. Similarly, the review by Hayashi et al. pointed out how lifestyle and a pro-inflammatory diet could be risk factors for bladder cancer because of the induction of chronic systemic inflammation. On the other hand, fruit and vegetable intake play a protective role. Thus, the diet has the potential to modulate the carcinogenesis process [11]. It has been also reported that polyphenols present in foods and beverages of plant origin can efficiently modulate autophagy in several types of cancer. The role of autophagy in cancer is debated, as autophagy can both induce and suppress tumour growth. Accordingly, the modulation of autophagy by polyphenols could represent a suitable tool for developing novel therapies to fight cancer [12].
In their original research paper, Colapietro et al. elucidated, for the first time, the in vitro and in vivo anti-cancer properties of crocetin on glioblastoma. Crocetin, a saffron extract, was demonstrated to possess anti-proliferative and pro-differentiative effects that are able to activate apoptosis and reduce cell migration and wound repair. Moreover, crocetin was able to counteract the growth of glioma cells in vivo in subcutaneous xenograft murine and orthotopic intra-brain tumour models [13]. Khan et al. demonstrated the role of the well-known polyphenol curcumin in papillary thyroid cancer (PTC). Curcumin had cytotoxic effects in PTC cell lines by stimulating apoptosis, inhibiting the constitutively active Janus Kinase/Signal Transducer and the Activator of the Transcription 3 (JAK/STAT3) signalling pathway and enhancing the anti-tumoral activity of cisplatin. These findings suggest that the use of curcumin in combination with chemotherapeutic agents could improve the clinical outcomes of PTC patients [14]. Indeed, preclinical and clinical studies have shown how dietary and medicinal plant polyphenols can overcome multidrug resistance to chemotherapeutic agents in breast, lung, colorectal, and prostate cancer by modulating different signalling pathways [15].
Two research articles demonstrate the inhibition of migratory properties of prostate cancer cells and triple-negative breast cancer cells by the grape-derived phytochemical resveratrol and by soybean phytoalexins glyceollins [16,17]. The first study shows how resveratrol in vitro inhibits the stroma–epithelium interaction mediated by the hepatocyte growth factor (HGF) and is thus able to control the epithelial-to-mesenchymal transition (EMT) and inhibit prostate cancer cells migration [16]. The second research paper demonstrates that glyceollins are novel aryl hydrocarbon receptor (AhR) ligands and show anti-migration activity in the MDA-MB-231 mammary cancer cells by decreasing N-cadherin gene expression and modulating the expression of Chemokine (C-C motif) ligand 2 (CCL2) and PDZ-LIM domain protein 4 (PDLIM4) genes [17]. In addition, Itkin et al. explore the role of flavonoids as natural AhR ligands in renal cancer, suggesting that aminoflavone (AFP 464) and benzothiazole (5F 203) have potential therapeutic effects in this type of cancer [18].
Different studies have reported the anti-cancer effects of dietary factors in colorectal cancer (CRC). The chemo-preventive potential of a combined treatment of microencapsulated probiotics (Bifidobacterium longum BAA-999, BF) and lycopene (LYC) in an azoxymethane (AOM)/dextran sulphate sodium (DSS)-induced CRC model has been evaluated. The combined treatment modulated the major determinant of CRC pathogenesis, the insulin-like growth factor-I/Insulin-like growth factor-I receptor (IGF-1/IGF-1R) system [19]. The study by Gao et al. suggests a novel mechanism of action in tumour suppression by 3,3′-Diindolylmethane (DIM), a naturally derived chemo-preventive compound formed in the acidic environment of the stomach after the ingestion of glucobrassicin: an indole glucosinolate enriched in cruciferous vegetables. DIM directly inhibits an oncogenic mouse double minute 2 homologue (MDM2) at mRNA and protein levels, inhibits cancer cell proliferation, and induces cell cycle arrest and apoptosis in CRC cells. Moreover, DIM showed synergistic anti-cancer effects when applied in combination with cis-imidazoline MDM2 inhibitors, thus providing a novel potential therapeutic strategy for CRC [20]. Hwang et al. demonstrated the anti-cancer potential of the oral administration of zerumbone, the main component of the subtropical ginger plant Zingiber zerumbet, which was able to reduce colonic inflammation, hyperplasia, colonic polyp number, and to prevent macroadenoma progression [21]. In the same in vivo model, high salt diet (HSD) consumption was also demonstrated to decrease enterotoxigenic Bacteroides fragilis (ETBF)-colitis and carcinogenesis in ETBF-colonized mice with AOM/DSS-induced tumorigenesis by affecting the gut microbiome and immune system. Thus, they suggested how an HSD, although not recommended, could be beneficial under certain conditions [22]. Indeed, as pointed out in the review by Bojková et al., a high fat diet (HFD) traditionally has been considered unhealthy. However, the harmful impacts of HFD on human health mainly stem from the excessive accumulation of adipose tissue, particularly visceral adipose tissue, and the resulting induction of inflammation. Human studies have indicated that a reduction in total fat intake does not significantly impact the risk of developing cancer. Rather, the type of fatty acids in the diet plays a crucial role. Thus, additional studies are needed to fully establish the role of fatty acids in regulating cell proliferation, thus leading to the development of an effective approach for cancer prevention and treatment [23].
Two review papers reporting preclinical and clinical studies demonstrated that the primary compounds found in the Cannabis sativa L. plant, known as cannabinoids, reduce tumour growth and trigger apoptosis in melanoma [24] and prostate cancer [25]. Even though the use of cannabis is prohibited in many countries, there has been a renewed interest in exploring the potential health benefits of these plant-derived compounds in recent years.
Collectively, these studies support the role of dietary factors in the prevention and treatment of cancer. However, the effects of dietary factors on cancer are complex and multifaceted. While many dietary components have been shown to possess anti-cancer properties, their effectiveness in vivo can be limited by their poor bioavailability. The effectiveness of these components could be increased through the application of novel techniques, such as nanotechnology, to develop novel formulations with increased bioavailability, solubility, and stability in the human body, such as nanosuspensions, solid lipid nanoparticles, liposomes, gold nanoparticles, polymeric nanoparticles. Epidemiological studies have also identified the role of nutrition in cancer risk and survival, and the development of functional foods enriched with anti-cancer compounds can provide a convenient and effective modality for individuals to improve their diet and reduce their risk of cancer. Further studies are needed to completely understand the mechanisms of action of dietary factors on cancer cells and to develop effective strategies for cancer prevention and treatment.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Siegel, R.L.; Miller, K.D.; Wagle, N.S.; Jemal, A. Cancer statistics, 2023. CA Cancer J. Clin. 2023, 73, 17–48. [Google Scholar] [CrossRef] [PubMed]
  2. Picó, C.; Serra, F.; Rodríguez, A.M.; Keijer, J.; Palou, A. Biomarkers of Nutrition and Health: New Tools for New Approaches. Nutrients 2019, 11, 1092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Zheng, J.; Wu, F.; Wang, F.; Cheng, J.; Zou, H.; Li, Y.; Du, J.; Kan, J. Biomarkers of Micronutrients and Phytonutrients and Their Application in Epidemiological Studies. Nutrients 2023, 15, 970. [Google Scholar] [CrossRef]
  4. Fantini, M.; Benvenuto, M.; Masuelli, L.; Frajese, G.V.; Tresoldi, I.; Modesti, A.; Bei, R. In vitro and in vivo antitumoral effects of combinations of polyphenols, or polyphenols and anticancer drugs: Perspectives on cancer treatment. Int. J. Mol. Sci. 2015, 16, 9236–9282. [Google Scholar] [CrossRef] [Green Version]
  5. Focaccetti, C.; Izzi, V.; Benvenuto, M.; Fazi, S.; Ciuffa, S.; Giganti, M.G.; Potenza, V.; Manzari, V.; Modesti, A.; Bei, R. Polyphenols as Immunomodulatory Compounds in the Tumor Microenvironment: Friends or Foes? Int. J. Mol. Sci. 2019, 20, 1714. [Google Scholar] [CrossRef] [Green Version]
  6. Masuelli, L.; Marzocchella, L.; Focaccetti, C.; Tresoldi, I.; Palumbo, C.; Izzi, V.; Benvenuto, M.; Fantini, M.; Lista, F.; Tarantino, U.; et al. Resveratrol and diallyl disulfide enhance curcumin-induced sarcoma cell apoptosis. Front. Biosci. (Landmark Ed.) 2012, 17, 498–508. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Masuelli, L.; Benvenuto, M.; Focaccetti, C.; Ciuffa, S.; Fazi, S.; Bei, A.; Miele, M.T.; Piredda, L.; Manzari, V.; Modesti, A.; et al. Targeting the tumor immune microenvironment with “nutraceuticals”: From bench to clinical trials. Pharmacol. Ther. 2021, 219, 107700. [Google Scholar] [CrossRef]
  8. Benvenuto, M.; Focaccetti, C.; Ciuffa, S.; Fazi, S.; Bei, A.; Miele, M.T.; Albonici, L.; Cifaldi, L.; Masuelli, L.; Bei, R. Polyphenols affect the humoral response in cancer, infectious and allergic diseases and autoimmunity by modulating the activity of TH1 and TH2 cells. Curr. Opin. Pharmacol. 2021, 60, 315–330. [Google Scholar] [CrossRef]
  9. Matsushita, M.; Fujita, K.; Nonomura, N. Influence of Diet and Nutrition on Prostate Cancer. Int. J. Mol. Sci. 2020, 21, 1447. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. To, P.K.; Do, M.H.; Cho, J.H.; Jung, C. Growth Modulatory Role of Zinc in Prostate Cancer and Application to Cancer Therapeutics. Int. J. Mol. Sci. 2020, 21, 2991. [Google Scholar] [CrossRef] [Green Version]
  11. Hayashi, T.; Fujita, K.; Hayashi, Y.; Hatano, K.; Kawashima, A.; McConkey, D.J.; Nonomura, N. Mutational Landscape and Environmental Effects in Bladder Cancer. Int. J. Mol. Sci. 2020, 21, 6072. [Google Scholar] [CrossRef]
  12. Benvenuto, M.; Albonici, L.; Focaccetti, C.; Ciuffa, S.; Fazi, S.; Cifaldi, L.; Miele, M.T.; De Maio, F.; Tresoldi, I.; Manzari, V.; et al. Polyphenol-Mediated Autophagy in Cancer: Evidence of In Vitro and In Vivo Studies. Int. J. Mol. Sci. 2020, 21, 6635. [Google Scholar] [CrossRef]
  13. Colapietro, A.; Mancini, A.; Vitale, F.; Martellucci, S.; Angelucci, A.; Llorens, S.; Mattei, V.; Gravina, G.L.; Alonso, G.L.; Festuccia, C. Crocetin Extracted from Saffron Shows Antitumor Effects in Models of Human Glioblastoma. Int. J. Mol. Sci. 2020, 21, 423. [Google Scholar] [CrossRef] [Green Version]
  14. Khan, A.Q.; Ahmed, E.I.; Elareer, N.; Fathima, H.; Prabhu, K.S.; Siveen, K.S.; Kulinski, M.; Azizi, F.; Dermime, S.; Ahmad, A.; et al. Curcumin-Mediated Apoptotic Cell Death in Papillary Thyroid Cancer and Cancer Stem-Like Cells through Targeting of the JAK/STAT3 Signaling Pathway. Int. J. Mol. Sci. 2020, 21, 438. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Costea, T.; Vlad, O.C.; Miclea, L.C.; Ganea, C.; Szöllősi, J.; Mocanu, M.M. Alleviation of Multidrug Resistance by Flavonoid and Non-Flavonoid Compounds in Breast, Lung, Colorectal and Prostate Cancer. Int. J. Mol. Sci. 2020, 21, 401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Hsieh, T.C.; Wu, J.M. Resveratrol Suppresses Prostate Cancer Epithelial Cell Scatter/Invasion by Targeting Inhibition of Hepatocyte Growth Factor (HGF) Secretion by Prostate Stromal Cells and Upregulation of E-cadherin by Prostate Cancer Epithelial Cells. Int. J. Mol. Sci. 2020, 21, 1760. [Google Scholar] [CrossRef] [Green Version]
  17. Pham, T.H.; Lecomte, S.; Le Guevel, R.; Lardenois, A.; Evrard, B.; Chalmel, F.; Ferriere, F.; Balaguer, P.; Efstathiou, T.; Pakdel, F. Characterization of Glyceollins as Novel Aryl Hydrocarbon Receptor Ligands and Their Role in Cell Migration. Int. J. Mol. Sci. 2020, 21, 1368. [Google Scholar] [CrossRef] [Green Version]
  18. Itkin, B.; Breen, A.; Turyanska, L.; Sandes, E.O.; Bradshaw, T.D.; Loaiza-Perez, A.I. New Treatments in Renal Cancer: The AhR Ligands. Int. J. Mol. Sci. 2020, 21, 3551. [Google Scholar] [CrossRef] [PubMed]
  19. Valadez-Bustos, N.; Escamilla-Silva, E.M.; García-Vázquez, F.J.; Gallegos-Corona, M.A.; Amaya-Llano, S.L.; Ramos-Gómez, M. Oral Administration of Microencapsulated B. Longum BAA-999 and Lycopene Modulates IGF-1/IGF-1R/IGFBP3 Protein Expressions in a Colorectal Murine Model. Int. J. Mol. Sci. 2019, 20, 4275. [Google Scholar] [CrossRef] [Green Version]
  20. Gao, X.; Liu, J.; Cho, K.B.; Kedika, S.; Guo, B. Chemopreventive Agent 3,3’-Diindolylmethane Inhibits MDM2 in Colorectal Cancer Cells. Int. J. Mol. Sci. 2020, 21, 4642. [Google Scholar] [CrossRef]
  21. Hwang, S.; Jo, M.; Hong, J.E.; Park, C.O.; Lee, C.G.; Rhee, K.J. Protective Effects of Zerumbone on Colonic Tumorigenesis in Enterotoxigenic Bacteroides fragilis (ETBF)-Colonized AOM/DSS BALB/c Mice. Int. J. Mol. Sci. 2020, 21, 857. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Hwang, S.; Yi, H.C.; Hwang, S.; Jo, M.; Rhee, K.J. Dietary Salt Administration Decreases Enterotoxigenic Bacteroides fragilis (ETBF)-Promoted Tumorigenesis via Inhibition of Colonic Inflammation. Int. J. Mol. Sci. 2020, 21, 8034. [Google Scholar] [CrossRef]
  23. Bojková, B.; Winklewski, P.J.; Wszedybyl-Winklewska, M. Dietary Fat and Cancer-Which Is Good, Which Is Bad, and the Body of Evidence. Int. J. Mol. Sci. 2020, 21, 4114. [Google Scholar] [CrossRef] [PubMed]
  24. Bachari, A.; Piva, T.J.; Salami, S.A.; Jamshidi, N.; Mantri, N. Roles of Cannabinoids in Melanoma: Evidence from In Vivo Studies. Int. J. Mol. Sci. 2020, 21, 6040. [Google Scholar] [CrossRef] [PubMed]
  25. Singh, K.; Jamshidi, N.; Zomer, R.; Piva, T.J.; Mantri, N. Cannabinoids and Prostate Cancer: A Systematic Review of Animal Studies. Int. J. Mol. Sci. 2020, 21, 6265. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Benvenuto, M.; Bei, R. The Effect of Dietary Factors on Cancer. Int. J. Mol. Sci. 2023, 24, 6802. https://doi.org/10.3390/ijms24076802

AMA Style

Benvenuto M, Bei R. The Effect of Dietary Factors on Cancer. International Journal of Molecular Sciences. 2023; 24(7):6802. https://doi.org/10.3390/ijms24076802

Chicago/Turabian Style

Benvenuto, Monica, and Roberto Bei. 2023. "The Effect of Dietary Factors on Cancer" International Journal of Molecular Sciences 24, no. 7: 6802. https://doi.org/10.3390/ijms24076802

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop