You're using an outdated browser. Please upgrade to a modern browser for the best experience.
Submitted Successfully!
Thank you for your contribution! You can also upload a video entry or images related to this topic. For video creation, please contact our Academic Video Service.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2970 2023-04-24 05:39:31 |
2 format correct + 3 word(s) 2973 2023-04-24 05:45:21 |

Video Upload Options

We provide professional Academic Video Service to translate complex research into visually appealing presentations. Would you like to try it?

Confirm

Are you sure to Delete?
Yes No
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Fatma, H.; Jameel, M.; Siddique, H.R. Phytochemicals in Redox Homeostasis. Encyclopedia. Available online: https://encyclopedia.pub/entry/43356 (accessed on 10 July 2025).
Fatma H, Jameel M, Siddique HR. Phytochemicals in Redox Homeostasis. Encyclopedia. Available at: https://encyclopedia.pub/entry/43356. Accessed July 10, 2025.
Fatma, Homa, Mohd Jameel, Hifzur R. Siddique. "Phytochemicals in Redox Homeostasis" Encyclopedia, https://encyclopedia.pub/entry/43356 (accessed July 10, 2025).
Fatma, H., Jameel, M., & Siddique, H.R. (2023, April 24). Phytochemicals in Redox Homeostasis. In Encyclopedia. https://encyclopedia.pub/entry/43356
Fatma, Homa, et al. "Phytochemicals in Redox Homeostasis." Encyclopedia. Web. 24 April, 2023.
Phytochemicals in Redox Homeostasis
Edit

Redox homeostasis, a dynamic process ensuring a balance between cellular oxidizing and reducing reactions, is crucial for maintaining healthy cellular physiology and regulating many biological processes, requiring continuous monitoring and fine-tuning. Reactive species play a critical role in intra/intercellular signaling, and each cell has a specific system guarding cellular redox homeostasis. Reactive oxygen species (ROS) signaling and oxidative stress are involved in cancer initiation and progression.

redox homeostasis antioxidant reactive species phytochemicals

1. Phytochemicals as a Chemopreventive Agent

Reactive species are known to have a double edge role in carcinogenesis. The level of reactive species is said to play an important role in cancer progression or tumor suppression. It has been observed that at a certain level, reactive species facilitate cancer growth and proliferation, while above that level, reactive species are implicated in the suppression of tumor cells (Table 1). Several studies have pointed toward the cytotoxic role of reactive species in tumor cells. Dietary phytochemicals have drawn much attention because of their extensive therapeutic effects in preventing the onset and progression of a disease. Phytochemicals and their derivatives have been thought to be involved in chemoprevention and chemosensitization, and their therapeutic efficacy has been extensively studied [1]. Many plants have shown anticancer properties owing to the phytochemicals present in them. More than 15 Allium spp. have shown anticancer properties due to their secondary metabolites. Allium spp. contain S-allyl mercaptocysteine, Quercetin, Flavanoids, and Ajoene, which facilitate cytotoxicity, immunomodulation, anti-inflammation, and apoptosis in various cancers. Moreover, these compounds also balance cellular redox homeostasis by scavenging ROS, reducing macromolecular damage, and increasing GST activity [2]. Phytochemicals are found to exert an antioxidant effect in the normal cell, while in cancer cells, phytochemicals tend to increase reactive species beyond the threshold level at which the survival adaptation of the cell is rendered futile. Dietary phytochemicals exert anticancer, chemopreventive, and chemosensitizing properties by generating excessive reactive species levels at which the oxidative stress inside the cell is so high that the cell becomes destined to die (Figure 1).
Figure 1. Role of oxidant–antioxidant levels in cancer cells and cancer cell death. Phytochemicals exert their anticancer property by targeting the redox status of cancer cells. They exert cytotoxic action by scavenging reactive species or generating reactive species burst inside the cells. Caspase-med-Apoptosis: Caspase-mediate Apoptosis; COX: Cyclooxygenase; Cyt-C: Cytochrome C; GGT: γ-Glutamyl Transpeptidase; GST: Glutathione S-Transferase; HDAC2: Histone Deacetylase; HIF: Hypoxia-Induced Factor; HO: Hemeoxygenase-1; LPO: Lipid Peroxidation; MDA: Malondialdehyde; MMP: Mitochondrial Metalloproteinase; NAF: Nutrient-deprivation Autophagy Factor-1; NRF-2: Nuclear Factor Erythroid 2–Related Factor 2; NF-κB: Nuclear Factor kappa-B; NQO1: NADPH Quinone Oxidoreductase; PI3K: Phosphoinositide 3-Kinase; QR: Quinone Reductase; RNS: Reactive Nitrogen Species; ROS: Reactive Oxygen Species.
Curcumin is a naturally occurring polyphenol found in the rhizome of turmeric (Curcuma longa). Curcumin can target multiple signaling molecules to exert its biological efficacy, such as antioxidant, antimicrobial, anti-inflammatory, anti-mutagenic, antimicrobial, and anticancerous properties [3]. Curcumin (diferuloylmethane) neutralizes ROS generated from chemical carcinogens by inducing GST (a phase II metabolizing enzyme) and Quinine reductase. Not only does Curcumin scavenge ROS by inducing ROS-scavenging enzymes such as GST, Hemeoxygenase-1, and redox-sensitive inducible enzyme, but Curcumin can also exploit ROS generation to kill cancer cells [4]. Curcumin is found to generate ROS beyond the threshold level to induce apoptosis by downregulating anti-apoptotic protein, NF-κB, and COX-2 and upregulating the activity of tumor suppressor p53 [5]. In colon cancer, Curcumin induces apoptosis by increasing the production of ROS, Ca2+, upregulating the expression of proapoptotic proteins (BAX, Cytochrome C, p53, and p21), Caspase 3, and reducing the mitochondrial membrane potential [1][6]. Moreover, Curcumin exerts its anticancerous activity by inhibiting proinflammatory enzymes such as inducible NOS (iNOS) and tumor necrosis factor-α (TNF-α). In addition, Curcumin can inhibit hypoxia-induced ROS in hepatic carcinoma cells (HCC) by downregulating the expression of hypoxia-inducible factor-1 (HIF-1) [7].
Another polyphenol, Resveratrol (3,5,4′-trihydroxy-trans-stilbene), belongs to the stilbenoids groups and comprises two phenol rings linked by an ethylene bridge. Resveratrol is commonly found in the skin and seeds of grapes and many other plant species. Resveratrol has several biological efficacies, including antioxidant and anticancer properties. It has been found that Resveratrol induces its anticancer efficacy by numerous mechanisms, including but not limited to the overproduction of reactive species. It has been found that Resveratrol suppresses the expression of NAF-1 by facilitating NRF-2 signaling and overproducing ROS in pancreatic cancer cells [8]. Moreover, in colon cancer cells, Resveratrol inhibits iNOS expression along with post-translational modification and translocation of NF-κB, resulting in the inhibition of inflammation associated with cancer cells [8].
Apigenin (4′,5′,7-trihydroxyflavone) is a flavone extracted from plants that are abundantly found in vegetables, fruits, medicinal plants, etc., that has shown its biological efficacy as an antioxidant, organ protector, and anticancer agent [9]. Apigenin was found to exert its anticancer properties through apoptosis, cell cycle arrest, immune response, and ROS. Administration of Apigenin (12.5–50 μM) in the papillary thyroid carcinoma cells leads to cell cycle arrest via inhibiting the expression of CDC25c and overproduction of ROS, which ultimately causes DNA damage [10]. Apigenin is also used as a ROS amplifier to enhance the cytotoxic effect of Metformin in vitro and in vivo. The combination of Metformin (5 μM) and Apigenin (20 μM) is found to induce ROS-dependent severe DNA damage and apoptosis in human pancreatic cells. Moreover, in vivo combination of Metformin (75 mg/kg b.w.) and Apigenin (5 mg/kg b.w.) is found to have a profound effect on tumor weight [11]. Quercetin (3′,3′,4′,5′,7 pentahydroxyflavone) belongs to the flavonol group and is abundant in nature and common to the human diet. Quercetin exhibits chemopreventive properties owing to its antioxidant properties. It has been observed that the presence of an OH group and double bonds in Quercetin has provided antioxidant capability to the Quercetin molecules. Quercetin can scavenge both ROS and RNS [12]. Zhang et al. [13] observed that the antioxidant properties of Quercetin (15) μM helps in enhancing the therapeutic efficacy of Paclitaxel (12.5 μM) against CaP by inducing endoplasmic reticulum stress and intracellular ROS leading to CaP cell cycle arrest and death. Moreover, Quercetin can form Quercetin radicals to scavenge peroxyl radicals. The formation of Quercetin radicals can overall increase the intracellular ROS level. Quercetin is also observed to induce free radical-mediated apoptosis by p38/ASK1/AMPKα1/COX-2 [14].
Rutin (3,3′,4′,5,7-pentahydroxyflavone-3-rhamnoglucoside) is a flavonol commonly found in passionflower, buckwheat tea, apple, and many other plants. Rutin prohibits liver cancer cell proliferation at the IC50 value of 52.7 μM/L and enhances cell death. Moreover, Rutin treatment significantly alters the expression of drug-metabolizing CYP3A4 and CYP1A1 and phase II reaction catalyzing enzyme NADPH Quinone oxidoreductase I (NQO1) and GST variant P1 [15]. In another study, the administration of Rutin is followed by a reduction in cell viability due to enhanced ROS generation and dose-dependent nuclear condensation in cervical cancer cells [16].
Caffeic acid (3,4-Dihydroxycinnamic acid) belongs to a subgroup of hydroxycinnamic acids of polyphenol groups and is believed to possess antioxidant properties that help in many biological activities [17]. Caffeic acid (0–500 μM) induces cell death in the colon and cervical cancer by inhibiting Histone Deacetylases (HDAC) 2. Furthermore, inhibition of HDAC2 leads to the overproduction of ROS, cell cycle arrest, and caspase-3-mediated apoptosis in the cancer cells [18]. Caffeic and Ferulic acid are found to chelate RNS and form stable intermediates with these reactive species [19]. The scavenging capacity of Ferulic acid (0–100 μM) facilitates cytoprotection by inhibiting DNA damage, inflammation, lipid peroxidation, and stimulating apoptosis [20]. Sinapic acid (3-(4-hydroxy-3,5-dimethoxyphenyl) prop-2-enoic acid) exhibits a chemopreventive effect on colon carcinogenesis. The authors observed that Sinapic acid (40 mg/kg b.w.) could decrease tumor prevalence, modulate LPO markers, and increase antioxidant defense by regulating phase I and II detoxifying enzymes [21]. In another study, Sinapic acid showed anticancer properties both in free and nano-capsulated form. It was observed that Sinapic acid (125.23 µM) showed an apparent increase in the ROS level in HeP-2 cells, leading to oxidative stress and a mitochondria-dependent pathway of apoptosis [22].
Table 1. Occurrence and anticancer mechanism of the phytochemicals.
Gallic acid (3,4,5-trihydroxybenzoic acid) is grouped with phenolic acid and found in hazel, tea leaves, oak barks, etc. Gallic acid (0–50 μM & 100–200 μM) induces lung cancer apoptosis by increasing ROS levels and decreasing GSH levels, leading to the loss of mitochondrial membrane potential. Moreover, Gallic acid-induced ROS at 50 g/mL facilitates c-Jun-NH2 kinase (JNK) mediate apoptosis in lung fibroblast cells. Formation and accumulation of H2O2 lead to the activation of the p53 pathway and JNK pathway, culminating in apoptosis [23]. Furthermore, Gallic acid exerts anticancer properties against CaP, leukemia, esophageal cancer, and cervical cancer through the ROS burst and antioxidant defense system [23].
Terpenoids or isoprenoids are diverse phytochemicals showing various biological efficacy, including chemopreventive and chemosensitizing effects in in vitro, preclinical, and clinical settings. Betulinic acid (3β-hydroxy-lup-20 (29)-en-28-oic acid) shows a chemopreventive effect by modulating xenobiotic and antioxidative enzyme activities. Betulinic acid (10 mg/kg b.w.) has been observed to easily neutralize the reactive species by upregulating the activity of phase II enzymes such as GST, γ-Glutamyl transpeptidase, and DT-diaphorase and reducing MDA levels [24]. Moreover, Betulinic acid interacts with xenobiotic metabolizing enzymes to prevent the development of skin papilloma and carcinomas in DMBA (10 mg/kg b.w.)-treated groups [24]. Another triterpene, Lupeol, exhibits anticancer potential against human lung carcinoma cells by excessive ROS generation, apoptosis, and downregulation of the mTOR/PI3K/AKT axis. Moreover, it has been observed that the cytotoxic potential of Lupeol was governed by the loss of MMP, which further leads to higher ROS levels and apoptosis [25]. The phytochemicals and their role in anticancer therapy by perturbing redox homeostasis of the cancer cells can be exploited extensively as one of the important benefits of using natural compounds is few to no side effects in normal cells.

2. Toxic Effect of Phytochemicals

More than 10,000 plant metabolites have been identified to date; however, the toxicological characterization of many of these secondary metabolites is not yet preclinically and clinically defined. Another challenge in phytochemical research is the contradictory effects reported in different setups. The contradictory effects of phytochemicals might be dose/concentration related. The different doses might elicit different effects. Few phytochemicals have anticancer properties and show chemopreventive activity when applied in different setups. For instance, Capsaicin acts as a co-carcinogen with DMBA/TPA to induce skin cancer. Applying phytochemicals such as Capsaicin on the dorsal skin of a DMBA-initiated TPA-promoted skin cancer model increases the number, size, and amount of cancer [26]. A recent study showed that continuous exposure of low dose Capsaicin facilitates colorectal cancer progression by further promoting abnormal expression of HDAC and histone modification leading to dysregulation of Toll-like receptor 4 (TLR4) [27].
Another phytochemical, Cycasin (methylazoxymethanol-D-glucoside), commonly found in cycad nuts, was reported to have carcinogenic properties. It was observed that Cycasin and its metabolite, Methylazoxymethanol, promotes neoplasia in the liver, kidneys, and intestines, which prompted the international agency of research on cancer to identify Cycasin and its metabolite as a carcinogen to humans [28]. Furthermore, oral administration of Acyclic monoterpene, β-myrcene (1000 mg/kg b.w.), which is commonly found in verbena, lemongrass, bay, rosemary, basil, cardamom, etc., proliferates liver and kidney (adenomas and carcinomas) cancer [28].
Alkylbenzenes such as Asarones, Elmicin, Estragole, and Safrole, found in essential oils or parts of the Aristolochiaceae plants, Artemisia dracunculus, nutmegs, are reported to have carcinogenic properties by forming DNA adducts, micronuclei, and malignant tumors. Moreover, benzopyrene, such as Coumarin, increases the incidence of renal tubule adenomas, alveolar/bronchiolar adenomas, alveolar/bronchiolar carcinoma, HCC, etc. [29]. Moreover, rodent studies have demonstrated that Safrole and methyleugenol act as hepatocarcinogens. In silico data also report that Myristicin might play a role in carcinogenesis, but studies specify that 2 mM/kg/day of Myristicin for 2 years would lead to a significant but weak increase in hepatic tumor burden. However, conclusive evaluation or data regarding the carcinogenic properties of Myristicin does not exist [30]. Safrole, methyleugenol, and betel quid are further implicated in increased HCC risk, genotoxicity, mutagenicity, and chromosomal aberrations [31].
Aristocholic acid or herbs containing Aristocholic acid are reported to promote HCC both in vitro and in vivo. Aristocholic acid increases HCC incidence of DNA damage, DNA adduct, and premalignant alterations in mice and canines. Moreover, Gingko biloba extracts increase the incidence of HCC, hepatocellular necrosis, and hepatoblastoma [31]. Hirose et al. [32] reported the cancer-promoting properties of isothiocyanates. Their study observed that Benzyl isothiocyanates and Phenylethyl isothiocyanates (0.1% of diet) could increase the incidences of papillary nodular hyperplasia and carcinoma in the urinary bladder of the DEN and N-butyl-N-(4hydroxybutyl) nitrosamine-treated rat model. However, several reports have also shown the anticancer activity of the isothiocyanates, suggesting that the dose [33], duration of treatment, or any other circumstance might play the defining role of a phytochemical as a carcinogen, co-carcinogen, or anti-carcinogen (Table 2).
Table 2. Anticancer and carcinogenic effect of phytochemicals doses and toxicity models used.

References

  1. Fatma, H.; Siddique, H.R. Research and Patents Status of Selected Phytochemicals Against Cancer: How Close and How Far? Recent Pat. Anticancer Drug Discov. 2022, 18.
  2. Asemani, Y.; Zamani, N.; Bayat, M.; Amirghofran, Z. Allium vegetables for possible future of cancer treatment. Phytother. Res. 2019, 33, 3019–3039.
  3. Hewlings, S.J.; Kalman, D.S. Curcumin: A Review of Its’ Effects on Human Health. Foods 2017, 6, 92.
  4. Park, W.; Amin, A.R.; Chen, Z.G.; Shin, D.M. New Perspectives of Curcumin in Cancer Prevention. Cancer Prev. Res. 2013, 6, 387–400.
  5. Mortezaee, K.; Salehi, E.; Mirtavoos-Mahyari, H.; Motevaseli, E.; Najafi, M.; Farhood, B.; Rosengren, R.J.; Sahebkar, A. Mechanisms of apoptosis modulation by Curcumin: Implications for cancer therapy. J. Cell. Physiol. 2019, 234, 12537–12550.
  6. Su, C.C.; Lin, J.G.; Li, T.M.; Chung, J.G.; Yang, J.S.; Ip, S.W.; Lin, W.C.; Chen, G.W. Curcumin-induced apoptosis of human colon cancer colo 205 cells through the production of R.O.S., Ca2+ and the activation of caspase-3. Anticancer Res. 2006, 26, 4379–4389.
  7. Gupta, N.; Verma, K.; Nalla, S.; Kulshreshtha, A.; Lall, R.; Prasad, S. Free Radicals as a Double-Edged Sword: The Cancer Preventive and Therapeutic Roles of Curcumin. Molecules 2020, 25, 5390.
  8. Salehi, B.; Mishra, A.P.; Nigam, M.; Sener, B.; Kilic, M.; Sharifi-Rad, M.; Fokou, P.V.T.; Martins, N.; Sharifi-Rad, J. Resveratrol: A Double-Edged Sword in Health Benefits. Biomedicines 2018, 6, 91.
  9. Singh, D.; Khan, M.A.; Siddique, H.R. Apigenin, A Plant Flavone Playing Noble Roles in Cancer Prevention Via Modulation of Key Cell Signaling Networks. Recent Pat. Anticancer Drug Discov. 2019, 14, 298–311.
  10. Yan, X.; Qi, M.; Li, P.; Zhan, Y.; Shao, H. Apigenin in cancer therapy: Anticancer effects and mechanisms of action. Cell Biosci. 2017, 7, 50.
  11. Warkad, M.S.; Kim, C.H.; Kang, B.G.; Park, S.H.; Jung, J.S.; Feng, J.H.; Inci, G.; Kim, S.C.; Suh, H.W.; Lim, S.S.; et al. Metformin-induced R.O.S. upregulation as amplified by Apigenin causes profound anticancer activity while sparing normal cells. Sci. Rep. 2021, 11, 14002.
  12. Deepika; Maurya, P.K. Health Benefits of Quercetin in Age-Related Diseases. Molecules 2022, 27, 2498.
  13. Zhang, X.; Huang, J.; Yu, C.; Xiang, L.; Li, L.; Shi, D.; Lin, F. Quercetin Enhanced Paclitaxel Therapeutic Effects Towards PC-3 Prostate Cancer Through ER Stress Induction and ROS Production. OncoTargets Ther. 2020, 13, 513–523.
  14. Biswas, P.; Dey, D.; Biswas, P.K.; Rahaman, T.I.; Saha, S.; Parvez, A.; Khan, D.A.; Lily, N.J.; Saha, K.; Sohel, M.; et al. A Comprehensive Analysis and Anti-Cancer Activities of Quercetin in ROS-Mediated Cancer and Cancer Stem Cells. Int. J. Mol. Sci. 2022, 23, 11746.
  15. Karakurt, S. Modulatory effects of Rutin on the expression of cytochrome P450s and antioxidant enzymes in human hepatoma cells. Acta Pharm. 2016, 66, 491–502.
  16. Imani, A.; Maleki, N.; Bohlouli, S.; Kouhsoltani, M.; Sharifi, S.; Maleki Dizaj, S. Molecular mechanisms of anticancer effect of Rutin. Phytother. Res. 2021, 35, 2500–2513.
  17. Birková, A.; Hubková, B.; Bolerázska, B.; Mareková, M.; Čižmárová, B. Caffeic acid: A brief overview of its presence, metabolism, and bioactivity. Bioact. Compd. Health Dis. 2020, 3, 74–81.
  18. Anantharaju, P.G.; Reddy, D.B.; Padukudru, M.A.; Chitturi, C.M.K.; Vimalambike, M.G.; Madhunapantula, S.V. Induction of colon and cervical cancer cell death by cinnamic acid derivatives is mediated through the inhibition of Histone Deacetylases (HDAC). PLoS ONE 2017, 12, e0186208.
  19. Damasceno, S.S.; Dantas, B.B.; Ribeiro-Filho, J.; Antônio, M.; Araújo, D.; da Costa, J.G.M. Chemical Properties of Caffeic and Ferulic Acids in Biological System: Implications in Cancer Therapy: A Review. Curr. Pharm. Des. 2017, 23, 3015–3023.
  20. Zhang, X.; Lin, D.; Jiang, R.; Li, H.; Wan, J.; Li, H. Ferulic acid exerts antitumor activity and inhibits metastasis in breast cancer cells by regulating epithelial to mesenchymal transition. Oncol. Rep. 2016, 36, 271–278.
  21. Balaji, C.; Muthukumaran, J.; Nalini, N. Chemopreventive effect of sinapic acid on 1,2-dimethylhydrazine-induced experimental rat colon carcinogenesis. Hum. Exp. Toxicol. 2014, 33, 1253–1268.
  22. Badr, D.A.; Amer, M.E.; Abd-Elhay, W.M.; Nasr, M.S.; Abuamara, T.M.; Ali, H.; Mohamed, A.F.; Youssef, M.A.; Awwad, N.S.; Ju, Y.H.; et al. Histopathological and genetic changes proved the anticancer potential of free and nano-capsulated sinapic acid. Appl. Biol. Chem. 2019, 62, 59.
  23. Subramanian, A.P.; John, A.A.; Vellayappan, M.V.; Balaji, A.; Jaganathan, S.K.; Supriyanto, E.; Yusof, M. Gallic acid: Prospects and molecular mechanisms of its anticancer activity. RSC Adv. 2015, 5, 35608–35621.
  24. Kaur, R.; Arora, S. Interactions of betulinic acid with xenobiotic metabolizing and antioxidative enzymes in DMBA-treated Sprague Dawley female rats. Free Radic. Biol. Med. 2013, 65, 131–142.
  25. He, W.; Li, X.; Xia, S. Lupeol triterpene exhibits potent antitumor effects in A427 human lung carcinoma cells via mitochondrial mediated apoptosis, ROS generation, loss of mitochondrial membrane potential and downregulation of m-TOR/PI3Ksol; AKT signalling pathway. J. BUON 2018, 23, 635–640.
  26. Liu, Z.; Zhu, P.; Tao, Y.; Shen, C.; Wang, S.; Zhao, L.; Wu, H.; Fan, F.; Lin, C.; Chen, C.; et al. Cancer-promoting effect of Capsaicin on DMBA/TPA-induced skin tumorigenesis by modulating inflammation, Erk and p38 in mice. Food Chem. Toxicol. 2015, 81, 1–8.
  27. Qin, J.; Li, H.; Yu, W.; Wei, L.; Wen, B. Effect of cold exposure and Capsaicin on the expression of histone acetylation and Toll-like receptors in 1,2-dimethylhydrazine-induced colon carcinogenesis. Environ. Sci. Pollut. Res. 2021, 28, 60981–60992.
  28. Kobets, T.; Smith, B.P.C.; Williams, G.M. Food-Borne Chemical Carcinogens and the Evidence for Human Cancer Risk. Foods 2022, 11, 2828.
  29. Pauzi, N.A.M.; Cheema, M.S.; Ismail, A.; Ghazali, A.R.; Abdullah, R. Are Genotoxic and Carcinogenic Compounds Present in Malaysian Traditional Medicines and Herbal Supplements? A Review Based on the Malaysian Herbal Monograph. Malays. J. Med. Health Sci. 2021, 17, 207–222.
  30. Götz, M.E.; Sachse, B.; Schäfer, B.; Eisenreich, A. Myristicin and Elemicin: Potentially Toxic Alkenylbenzenes in Food. Foods 2022, 11, 1988.
  31. Khalil, M.; Calasso, M.; Bonfrate, L.; Di Ciaula, A.; De Angelis, M.; Wang, D.Q.H.; Portincasa, P. The pros and cons of biological effects of herbs and herb-derived compounds on liver tumorigenesis. Hepatoma Res. 2022, 8, 23.
  32. Hirose, M.; Yamaguchi, T.; Kimoto, N.; Ogawa, K.; Futakuchi, M.; Sano, M.; Shirai, T. Strong promoting activity of phenylethyl isothiocyanate and benzyl isothiocyanate on urinary bladder carcinogenesis in F344 male rats. Int. J. Cancer. 1998, 77, 773–777.
  33. Ranjan, A.; Ramachandran, S.; Gupta, N.; Kaushik, I.; Wright, S.; Srivastava, S.; Das, H.; Srivastava, S.; Prasad, S.; Srivastava, S.K. Role of Phytochemicals in Cancer Prevention. Int. J. Mol. Sci. 2019, 20, 4981.
  34. Mukerjee, A.; Ranjan, A.P.; Vishwanatha, J.K. Targeted Nanocurcumin Therapy Using Annexin A2 Antibody Improves Tumor Accumulation and Therapeutic Efficacy Against Highly Metastatic Breast Cancer. J. Biomed. Nanotechnol. 2016, 12, 1374–1392.
  35. Annaji, M.; Poudel, I.; Boddu, S.H.S.; Arnold, R.D.; Tiwari, A.K.; Babu, R.J. Resveratrol-loaded nanomedicines for cancer applications. Cancer Rep. 2021, 4, e1353.
  36. Sen, R.; Ganguly, S.; Ganguly, S.; Debnath, M.C.; Chakraborty, S.; Mukherjee, B.; Chattopadhyay, D. Apigenin-Loaded PLGA-DMSA Nanoparticles: A Novel Strategy to Treat Melanoma Lung Metastasis. Mol. Pharm. 2021, 18, 1920–1938.
  37. Baksi, R.; Singh, D.P.; Borse, S.P.; Rana, R.; Sharma, V.; Nivsarkar, M. In vitro and in vivo anticancer efficacy potential of Quercetin loaded polymeric nanoparticles. Biomed. Pharmacother. 2018, 106, 1513–1526.
  38. Hoai, T.T.; Yen, P.T.; Dao, T.T.B.; Long, L.H.; Anh, D.X.; Minh, L.H.; Anh, B.Q.; Thuong, N.T. Evaluation of the Cytotoxic Effect of Rutin Prenanoemulsion in Lung and Colon Cancer Cell Lines. J. Nanomater. 2020, 2020, 8867669.
  39. Pandey, P.; Rahman, M.; Bhatt, P.C.; Beg, S.; Paul, B.; Hafeez, A.; Al-Abbasi, F.A.; Nadeem, M.S.; Baothman, O.; Anwar, F.; et al. Implication of nano-antioxidant therapy for treatment of hepatocellular carcinoma using PLGA nanoparticles of Rutin. Nanomedicine 2018, 13, 849–870.
  40. Firouzai-Amandi, A.; Tarahomi, M.; Rahmani Youshanlouie, H.; Mosaddeghi Heris, R.; Jafari-Gharabaghlou, D.; Zarghami, N.; Dadashpour, M. Development, Characterization, and In Vitro Evaluation of Cytotoxic Activity of Rutin Loaded PCL-PEG Nanoparticles Against Skov3 Ovarian Cancer Cell. Asian Pac. J. Cancer Prev. 2022, 23, 1951–1957.
More
Upload a video for this entry
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , ,
View Times: 947
Revisions: 2 times (View History)
Update Date: 24 Apr 2023
1000/1000
Hot Most Recent
Academic Video Service