Thymoquinone: A Promising Therapeutic Agent for the Treatment of Colorectal Cancer
Abstract
:1. Introduction
2. Nigella sativa: The Main Source of Thymoquinone
3. Characterization, Pharmacognostic Isolation and Purification of Thymoquinone
4. Properties and Pharmacological Features of Thymoquinone
5. Preclinical and Clinical Studies on Thymoquinone and Its Main Source Nigella sativa
6. Brief on the Anticancer Properties of Thymoquinone
7. Thymoquinone a Promising Candidate for the Treatment of Colorectal Cancer
7.1. Cytotoxic Effect of Thymoquinone on Colorectal Cancer Cells
7.2. Influence of Thymoquinone on Apoptosis and Pathways Involved in Colorectal Cancer Cell Survival and Proliferation
7.3. Effect of Thymoquinone on Oxidative Stress-Related Damage in Colorectal Cancer
8. Simultaneous Stimulation with Thymoquinone and Known Chemotherapeutics: An Opportunity to Increase Treatment Efficacy
9. Future Directions
10. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Li, J.; Ma, X.; Chakravarti, D.; Shalapour, S.; DePinho, R.A. Genetic and biological hallmarks of colorectal cancer. Genes Dev. 2021, 35, 787–820. [Google Scholar] [CrossRef] [PubMed]
- Sninsky, J.A.; Shore, B.M.; Lupu, G.V.; Crockett, S.D. Risk factors for colorectal polyps and cancer. Gastrointest. Endosc. Clin. N. Am. 2022, 32, 195–213. [Google Scholar] [CrossRef] [PubMed]
- Klimeck, L.; Heisser, T.; Hoffmeister, M.; Brenner, H. Colorectal cancer. A health and economic problem. Best Pract. Res. Clin. Gastroenterol. 2023, 66, 195–213. [Google Scholar] [CrossRef] [PubMed]
- Thanikachalam, K.; Khan, G. Colorectal cancer and nutrition. Nutrients 2019, 11, 164. [Google Scholar] [CrossRef] [PubMed]
- Kanth, P.; Inadomi, J.M. Screening and prevention of colorectal cancer. BMJ 2021, 374, n1855. [Google Scholar] [CrossRef] [PubMed]
- Dariya, B.; Aliya, S.; Merchant, N.; Alam, A.; Nagaraju, G.P. Colorectal cancer biology, diagnosis, and therapeutic approaches. Crit. Rev. Oncog. 2020, 25, 71–94. [Google Scholar] [CrossRef] [PubMed]
- Hossain, M.S.; Karuniawati, H.; Jairoun, A.A.; Urbi, Z.; Ooi, J.; John, A.; Lim, Y.C.; Kibria, K.M.K.; Mohiuddin, A.K.M.; Ming, L.C.; et al. Colorectal cancer: A review of carcinogenesis, global epidemiology, current challenges, risk factors, preventive and treatment strategies. Cancers 2022, 14, 1732. [Google Scholar] [CrossRef]
- Bosman, F.; Yan, P. Molecular pathology of colorectal cancer. Pol. J. Pathol. 2014, 65, 257–266. [Google Scholar] [CrossRef]
- Disoma, C.; Zhou, Y.; Li, S.; Peng, J.; Xia, Z. Wnt/β-catenin signaling in colorectal cancer: Is therapeutic targeting even possible? Biochimie 2022, 195, 39–53. [Google Scholar] [CrossRef]
- Zhao, H.; Ming, T.; Tang, S.; Ren, S.; Yang, H.; Liu, M.; Tao, Q.; Xu, H. Wnt signaling in colorectal cancer: Pathogenic role and therapeutic target. Mol. Cancer 2022, 21, 144. [Google Scholar] [CrossRef]
- Kundu, S.; Ali, M.A.; Handin, N.; Conway, L.P.; Rendo, V.; Artursson, P.; He, L.; Globisch, D.; Sjöblom, T. Common and mutation specific phenotypes of KRAS and BRAF mutations in colorectal cancer cells revealed by integrative-omics analysis. J. Exp. Clin. Cancer Res. CR 2021, 40, 225. [Google Scholar] [CrossRef] [PubMed]
- Midthun, L.; Shaheen, S.; Deisch, J.; Senthil, M.; Tsai, J.; Hsueh, C.T. Concomitant KRAS and BRAF mutations in colorectal cancer. J. Gastrointest. Oncol. 2019, 10, 577–581. [Google Scholar] [CrossRef] [PubMed]
- Fang, J.Y.; Richardson, B.C. The MAPK signalling pathways and colorectal cancer. Lancet. Oncol. 2005, 6, 322–327. [Google Scholar] [CrossRef] [PubMed]
- Michel, M.; Kaps, L.; Maderer, A.; Galle, P.R.; Moehler, M. The role of p53 dysfunction in colorectal cancer and its implication for therapy. Cancers 2021, 13, 2296. [Google Scholar] [CrossRef] [PubMed]
- Papageorgis, P.; Cheng, K.; Ozturk, S.; Gong, Y.; Lambert, A.W.; Abdolmaleky, H.M.; Zhou, J.R.; Thiagalingam, S. Smad4 inactivation promotes malignancy and drug resistance of colon cancer. Cancer Res. 2011, 71, 998–1008. [Google Scholar] [CrossRef] [PubMed]
- Mármol, I.; Sánchez-de-Diego, C.; Pradilla Dieste, A.; Cerrada, E.; Rodriguez Yoldi, M. Colorectal carcinoma: A general overview and future perspectives in colorectal cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef] [PubMed]
- Vodenkova, S.; Buchler, T.; Cervena, K.; Veskrnova, V.; Vodicka, P.; Vymetalkova, V. 5-fluorouracil and other fluoropyrimidines in colorectal cancer: Past, present and future. Pharmacol. Ther. 2020, 206, 107447. [Google Scholar] [CrossRef]
- Ahmad, A.; Husain, A.; Mujeeb, M.; Khan, S.A.; Najmi, A.K.; Siddique, N.A.; Damanhouri, Z.A.; Anwar, F. A review on therapeutic potential of Nigella sativa: A miracle herb. Asian Pac. J. Trop. Biomed. 2013, 3, 337–352. [Google Scholar] [CrossRef]
- Rajsekhar, S.; Kuldeep, B. Pharmacognosy and pharmacology of Nigella sativa—A review. Int. Res. J. Pharm. 2011, 2, 36–39. [Google Scholar]
- Hannan, M.A.; Rahman, M.A.; Sohag, A.A.M.; Uddin, M.J.; Dash, R.; Sikder, M.H.; Rahman, M.S.; Timalsina, B.; Munni, Y.A.; Sarker, P.P.; et al. Black cumin (Nigella sativa L.): A comprehensive review on phytochemistry, health benefits, molecular pharmacology, and safety. Nutrients 2021, 13, 1784. [Google Scholar] [CrossRef]
- Almajali, B.; Al-Jamal, H.A.N.; Taib, W.R.W.; Ismail, I.; Johan, M.F.; Doolaanea, A.A.; Ibrahim, W.N. Thymoquinone, as a novel therapeutic candidate of cancers. Pharmaceuticals 2021, 14, 369. [Google Scholar] [CrossRef] [PubMed]
- Butt, A.S.; Nisar, N.; Ghani, N.; Altaf, I.; Mughal, T.A. Isolation of thymoquinone from Nigella sativa L. and Thymus vulgaris L., and its anti-proliferative effect on HeLa cancer cell lines. Trop. J. Pharm. Res. 2019, 18, 37–42. [Google Scholar] [CrossRef]
- Butnariu, M.; Quispe, C.; Herrera-Bravo, J.; Helon, P.; Kukula-Koch, W.; López, V.; Les, F.; Vergara, C.V.; Alarcón-Zapata, P.; Alarcón-Zapata, B.; et al. The effects of thymoquinone on pancreatic cancer: Evidence from preclinical studies. Biomed. Pharmacother. 2022, 153, 113364. [Google Scholar] [CrossRef] [PubMed]
- Ahmad, A.; Mishra, R.K.; Vyawahare, A.; Kumar, A.; Rehman, M.U.; Qamar, W.; Khan, A.Q.; Khan, R. Thymoquinone (2-Isoprpyl-5-methyl-1, 4-benzoquinone) as a chemopreventive/anticancer agent: Chemistry and biological effects. Saudi Pharm. J. 2019, 27, 1113–1126. [Google Scholar] [CrossRef] [PubMed]
- Goyal, S.N.; Prajapati, C.P.; Gore, P.R.; Patil, C.R.; Mahajan, U.B.; Sharma, C.; Talla, S.P.; Ojha, S.K. Therapeutic potential and pharmaceutical development of thymoquinone: A multitargeted molecule of natural origin. Front. Pharmacol. 2017, 8, 656. [Google Scholar] [CrossRef] [PubMed]
- Burits, M.; Bucar, F. Antioxidant activity of Nigella sativa essential oil. Phytother. Res. 2000, 14, 323–328. [Google Scholar] [CrossRef] [PubMed]
- Solati, Z.; Baharin, B.S.; Bagheri, H. Supercritical carbon dioxide (SC-CO2) extraction of Nigella sativa L. oil using full factorial design. Ind. Crops. Prod. 2012, 36, 519–523. [Google Scholar] [CrossRef]
- Ghanavi, Z.; Velayati, A.A.; Frania, P.; Naji, A.M.; Kalatehjari, S. Extraction and purification of anticancer thymoquinone from seeds of Nigella sativa by preparative high-performance liquid chromatography. JMPB 2020, 9, 73–79. [Google Scholar]
- Sovova, H.; Sajfrtova, M.; Topiar, M. Supercritical CO2 extraction of volatile thymoquinone from Monarda didyma and M. fistulosa herbs. J. Supercrit. Fluids 2015, 105, 29–34. [Google Scholar] [CrossRef]
- Iqbal, M.S.; Ahmad, A.; Pandey, B. Solvent based optimization for extraction and stability of thymoquinone from Nigella sativa Linn. and its quantification using RP-HPLC. Physiol. Mol. Biol. Plants 2018, 24, 1209–1219. [Google Scholar] [CrossRef]
- Rasoul-Amini, S.; Fotooh-Abadi, E.; Ghasemi, Y. Biotransformation of monoterpenes by immobilized microalgae. J. Appl. Physiol. 2011, 23, 975–981. [Google Scholar] [CrossRef]
- Nejabati, F.; Ebrahimzadeh, H. Electrospun nanofibers for extraction of thymoquinone from Nigella-Stevia prior to detection using electrochemical biosensor based on GCE/rGO/CuO. Microchem. J. 2023, 189, 108545. [Google Scholar] [CrossRef]
- Salem, M.L. Immunomodulatory and therapeutic properties of the Nigella sativa L. seed. Int. Immunopharmacol. 2005, 5, 1749–1770. [Google Scholar] [CrossRef] [PubMed]
- Salmani, J.M.; Asghar, S.; Lv, H.; Zhou, J. Aqueous solubility and degradation kinetics of the phytochemical anticancer thy-moquinone; probing the effects of solvents, pH and light. Molecules 2014, 19, 5925–5939. [Google Scholar] [CrossRef] [PubMed]
- Darakhshan, S.; Pour, A.B.; Colagar, A.H.; Sisakhtnezhad, S. Thymoquinone and its therapeutic potentials. Pharmacol. Res. 2015, 95–96, 138–158. [Google Scholar] [CrossRef] [PubMed]
- Nagi, M.N.; Almakki, H.A. Thymoquinone supplementation induces quinone reductase and glutathione transferase in mice liver: Possible role in protection against chemical carcinogenesis and toxicity. Phytother. Res. 2009, 23, 1295–1298. [Google Scholar] [CrossRef] [PubMed]
- Abukhader, M.M. The effect of route of administration in thymoquinone toxicity in male and female rats. Indian J. Pharm. Sci. 2012, 74, 195–200. [Google Scholar] [CrossRef]
- Rahmani, A.H.; Alzohairy, M.A.; Khan, M.A.; Aly, S.M. Therapeutic implications of black seed and its constituent thymo-quinone in the prevention of cancer through inactivation and activation of molecular pathways. Evid. Based Complement Alternat. Med. 2014, 2014, 724658. [Google Scholar] [CrossRef]
- Ahmad, A.; Alqahtani, S.; Jan, B.L.; Raish, M.; Rabba, A.K.; Alkharfy, K.M. Gender effect on the pharmacokinetics of thymoquinone: Preclinical investigation and in silico modelling in male and female rats. Saudi Pharm. J. 2020, 28, 403–408. [Google Scholar] [CrossRef]
- Mansour, M.A.; Ginawi, O.T.; El-Hadiyah, T.; El-Khatib, A.S.; Al-Shabanah, O.A.; Al-Sawaf, H.A. Effects of volatile oil con-stituents of Nigella sativa on carbon tetrachloride-induced hepatotoxicity in mice: Evidence for antioxidant effects of thymo-quinone. Res. Commun. Mol. Pathol. Pharmacol. 2001, 110, 239–252. [Google Scholar]
- Kanter, M. Thymoquinone attenuates lung injury induced by chronic toluene exposure in rats. Toxicol. Ind. Heal. 2010, 27, 387–395. [Google Scholar] [CrossRef] [PubMed]
- Alkharfy, K.M.; Ahmad, A.; Khan, R.M.; Al-Shagha, W.M. Pharmacokinetic plasma behaviors of intravenous and oral bioavailability of thymoquinone in a rabbit model. Eur. J. Drug Metab. Pharmacokinet. 2015, 40, 319–323. [Google Scholar] [CrossRef] [PubMed]
- Mahmoud, Y.K.; Abdelrazek, H.M.A. Cancer: Thymoquinone antioxidant/pro-oxidant effect as potential anticancer remedy. Biomed. Pharmacother. 2019, 115, 108783. [Google Scholar] [CrossRef] [PubMed]
- Elmowafy, M.; Samy, A.; Raslan, M.A.; Salama, A.; Said, R.A.; Abdelaziz, A.E.; El-Eraky, W.; El Awdan, S.; Viitala, T. Enhancement of bioavailability and pharmacodynamic effects of thymoquinone via nanostructured lipid carrier (NLC) formulation. AAPS PharmSciTech 2015, 17, 663–672. [Google Scholar] [CrossRef] [PubMed]
- Khan, A.; Alsahli, M.A.; Aljasir, M.A.; Maswadeh, H.; Mobark, M.A.; Azam, F.; Allemailem, K.S.; Alrumaihi, F.; Alhumaydhi, F.A.; Alwashmi, A.S.S.; et al. Safety, stability, and therapeutic efficacy of long-circulating TQ-incorporated liposomes: Implication in the treatment of lung cancer. Pharmaceutics 2022, 14, 153. [Google Scholar] [CrossRef] [PubMed]
- Hatami Nemati, S.; Bigdeli, M.R.; Mortazavi Moghadam, F.; Sharifi, K. Neuroprotective effects of niosomes loaded with thymoquinone in the cerebral ischemia model of male Wistar rats. Nanomedicine 2023, 48, 102637. [Google Scholar] [CrossRef]
- Alam, M.; Zameer, S.; Najmi, A.K.; Ahmad, F.J.; Imam, S.S.; Akhtar, M. Thymoquinone loaded solid lipid nanoparticles demonstrated antidepressant-like activity in rats via indoleamine 2, 3-dioxygenase pathway. Drug Res. 2020, 70, 206–213. [Google Scholar] [CrossRef]
- Manjunatha, V.; Nixon, J.E.; Mathis, G.F.; Lumpkins, B.S.; Güzel-Seydim, Z.B.; Seydim, A.C.; Greene, A.K.; Jiang, X. Nigella sativa as an antibiotic alternative to promote growth and enhance health of broilers challenged with Eimeria maxima and Clostridium perfringens. Poult. Sci. 2023, 102, 102831. [Google Scholar] [CrossRef]
- Barashkova, A.S.; Smirnov, A.N.; Zorina, E.S.; Rogozhin, E.A. Diversity of cationic antimicrobial peptides in black cumin (Nigella sativa L.) seeds. Int. J. Mol. Sci. 2023, 24, 8066. [Google Scholar] [CrossRef]
- Alam, T.; Naseem, S.; Shahabuddin, F.; Abidi, S.; Parwez, I.; Khan, F. Oral administration of Nigella sativa oil attenuates arsenic-induced redox imbalance, DNA damage, metabolic distress, and histopathological alterations in rat intestine. J. Trace. Elem. Med. Biol. 2023, 79, 127238. [Google Scholar] [CrossRef]
- Ciesielska-Figlon, K.; Wojciechowicz, K.; Daca, A.; Kokotkiewicz, A.; Łuczkiewicz, M.; Witkowski, J.M.; Lisowska, K.A. The impact of Nigella sativa essential oil on T cells in women with Hashimoto’s thyroiditis. Antioxidants 2023, 12, 1246. [Google Scholar] [CrossRef] [PubMed]
- Ali Bakr, E.H.; Saad Alyamani, R.A. Immunomodulatory protective effects of Nigella sativa and Lactuca sativa oils on liver intoxication in experimental animals. Pak. J. Biol. Sci. 2023, 26, 434–441. [Google Scholar] [CrossRef] [PubMed]
- Younus, H.; Sawhney (Eds.) Molecular and Therapeutic: Actions of Thymoquinone; Springer: Berlin/Heidelberg, Germany, 2018. [Google Scholar]
- Gholamnezhad, Z.; Havakhah, S.; Boskabady, M.H. Preclinical and clinical effects of Nigella sativa and its constituent, thymoquinone: A review. J. Ethnopharmacol. 2016, 190, 372–386. [Google Scholar] [CrossRef] [PubMed]
- Dalli, M.; Bekkouch, O.; Azizi, S.E.; Azghar, A.; Gseyra, N.; Kim, B. Nigella sativa L. phytochemistry and pharmacological activities: A review (2019–2021). Biomolecules 2022, 12, 20. [Google Scholar] [CrossRef] [PubMed]
- Jarmakiewicz-Czaja, S.; Zielińska, M.; Helma, K.; Sokal, A.; Filip, R. Effect of Nigella sativa on selected gastrointestinal diseases. Curr. Issues Mol. Biol. 2023, 45, 3016–3034. [Google Scholar] [CrossRef] [PubMed]
- Azami, S.; Forouzanfar, F. Potential role of Nigella sativa and its constituent (thymoquinone) in ischemic stroke. Curr. Mol. Med. 2023. [CrossRef] [PubMed]
- Alabdullah, M.; Kara Beit, Z.Z.; Shehada, A. Comparative clinical study of the effect of Nigella sativa oil on soft tissue healing and inflammation reduction compared to Eugenol in the context of dry socket. Cureus 2023, 15, e35375. [Google Scholar] [CrossRef] [PubMed]
- Palanisamy, C.P.; Alugoju, P.; Jayaraman, S.; Poompradub, S. Nigella sativa L. seed extracts promote wound healing progress by activating VEGF and PDGF signaling pathways: An in vitro and in silico study. F1000Research 2023, 12, 436. [Google Scholar] [CrossRef]
- Tavakoli-Rouzbehani, O.M.; Abbasnezhad, M.; Kheirouri, S.; Alizadeh, M. Efficacy of Nigella sativa oil on endothelial function and atherogenic indices in patients with coronary artery diseases: A randomized, double-blind, placebo-control clinical trial. Phytother. Res. 2022, 36, 4516–4526. [Google Scholar] [CrossRef]
- Khodaie, S.A.; Nikkhah, H.; Namiranian, N.; Abotorabi, M.; Askari, M.; Khalilzadeh, S.H.; Khatibi Aghda, A.; Kamalinejad, M. Topical Nigella sativa L. product: A new candidate for the management of diabetic peripheral neuropathy. Inflammopharmacology 2023. [Google Scholar] [CrossRef]
- Yousefnejad, H.; Mohammadi, F.; Alizadeh-Naini, M.; Hejazi, N. Nigella sativa powder for helicobacter pylori infected patients: A randomized, double-blinded, placebo-controlled clinical trial. BMC Complement. Med. Ther. 2023, 23, 123. [Google Scholar] [CrossRef] [PubMed]
- Balasubramanian, R.; Maideen, N.M.P.; Muthusamy, S.; Gobinath, M. A review of clinical and preclinical studies on the therapeutic potential of black seeds (Nigella sativa) in the management of polycystic ovarian syndrome (PCOS). J. Pharmacopunct. 2023, 26, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Thomas, J.V.; Mohan, M.E.; Prabhakaran, P.; Das, S.S.; Maliakel, B.; Krishnakumar, I.M. A phase I clinical trial to evaluate the safety of thymoquinone-rich black cumin oil (BlaQmax®) on healthy subjects: Randomized, double-blinded, placebo-controlled prospective study. Toxicol. Rep. 2022, 9, 999–1007. [Google Scholar] [CrossRef] [PubMed]
- Ali, S.M.; Chen, P.; Sheikh, S.; Ahmad, A.; Ahmad, M.; Paithankar, M.; Desai, B.; Patel, P.; Khan, M.; Chaturvedi, A.; et al. Thymoquinone with metformin decreases fasting, post prandial glucose, and HbA1c in type 2 diabetic patients. Drug Res. 2021, 71, 302–306. [Google Scholar] [CrossRef]
- Akhondian, J.; Kianifar, H.; Raoofziaee, M.; Moayedpour, A.; Toosi, M.B.; Khajedaluee, M. The effect of thymoquinone on intractable pediatric seizures (pilot study). Epilepsy Res. 2011, 93, 39–43. [Google Scholar] [CrossRef]
- Rajput, S.; Kumar, B.P.; Dey, K.K.; Pal, I.; Parekh, A.; Mandal, M. Molecular targeting of Akt by thymoquinone promotes G1 arrest through translation inhibition of cyclin D1 and induces apoptosis in breast cancer cells. Life Sci. 2013, 93, 783–790. [Google Scholar] [CrossRef]
- Şahin, C.; Maytalman, E.; Nemutlu Samur, D.; Doğan, B. The effect of thymoquinone and propranolol combination on epidermoid laryngeal carcinoma cell. Eur. Arch. Otorhinolaryngol. 2023, 280, 2849–2858. [Google Scholar] [CrossRef]
- Almajali, B.; Al-Jamal, H.A.N.; Wan Taib, W.R.; Ismail, I.; Johan, M.F.; Doolaanea, A.A.; Ibrahim, W.N.; Tajudin, S.A. Thymoquinone suppresses cell proliferation and enhances apoptosis of HL60 leukemia cells through re-expression of JAK/STAT negative regulators. Asian Pac. J. Cancer Prev. 2021, 22, 879–885. [Google Scholar] [CrossRef]
- Al-Rawashde, F.A.; Al-Sanabra, O.M.; Alqaraleh, M.; Jaradat, A.Q.; Al-Wajeeh, A.S.; Johan, M.F.; Wan Taib, W.R.; Ismail, I.; Al-Jamal, H.A.N. Thymoquinone enhances apoptosis of K562 chronic myeloid leukemia cells through hypomethylation of SHP-1 and inhibition of JAK/STAT signaling pathway. Pharmaceuticals 2023, 16, 884. [Google Scholar] [CrossRef]
- Zhang, Y.; Liu, X.; Dang, W.; Liu, L. Thymoquinone inhibits lung cancer stem cell properties via triggering YAP degradation. Carcinogenesis 2023, 44, 426–435. [Google Scholar] [CrossRef]
- Nithya, G.; Santhanasabapathy, R.; Vanitha, M.K.; Anandakumar, P.; Sakthisekaran, D. Antioxidant, antiproliferative, and apoptotic activity of thymoquinone against benzo(a)pyrene-induced experimental lung cancer. J. Biochem. Mol. Toxicol. 2023, 37, e23230. [Google Scholar] [CrossRef] [PubMed]
- Sanapour, N.; Malakoti, F.; Shanebandi, D.; Targhazeh, N.; Yousefi, B.; Soleimanpour, J.; Majidinia, M. Thymoquinone augments methotrexate-induced apoptosis on osteosarcoma cells. Drug Res. 2022, 72, 220–225. [Google Scholar] [CrossRef] [PubMed]
- Khyavi, P.A.; Valizadeh, A.; Shanehbandi, D.; Yousefi, B.; Soleimanpour, J. Thymoquinone potentiates methotrexate mediated-apoptosis in Saos-2 osteosarcoma cell line. Drug Res. 2022, 72, 390–395. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Dong, J.; Cai, W.; Pan, Y.; Li, R.; Li, B. The effect of thymoquinone on apoptosis of SK-OV-3 ovarian cancer cell by regulation of Bcl-2 and Bax. Int. J. Gynecol. Cancer 2017, 27, 1596–1601. [Google Scholar] [CrossRef] [PubMed]
- Khurshid, Y.; Syed, B.; Simjee, S.U.; Beg, O.; Ahmed, A. Antiproliferative and apoptotic effects of proteins from black seeds (Nigella sativa) on human breast MCF-7 cancer cell line. BMC Complement. Med. Ther. 2020, 20, 5. [Google Scholar] [CrossRef] [PubMed]
- Al-Amri, A.M.; Bamosa, A.O. Phase I safety and clinical activity of thymoquinone in patients with advanced refractory malignant disease. Shiraz. E-Med. J. 2009, 10, 107–111. [Google Scholar]
- Khan, M.A.; Tania, M.; Fu, J. Epigenetic role of thymoquinone: Impact on cellular mechanism and cancer therapeutics. Drug Discov. Today 2019, 24, 2315–2322. [Google Scholar] [CrossRef]
- Ansary, J.; Giampieri, F.; Forbes-Hernandez, T.Y.; Regolo, L.; Quinzi, D.; Gracia Villar, S.; Garcia Villena, E.; Tutusaus Pifarre, K.; Alvarez-Suarez, J.M.; Battino, M.; et al. Nutritional value and preventive role of Nigella sativa L. and its main component thymoquinone in cancer: An evidenced-based review of preclinical and clinical studies. Molecules 2021, 26, 2108. [Google Scholar] [CrossRef]
- Alhmied, F.; Alammar, A.; Alsultan, B.; Alshehri, M.; Pottoo, F.H. Molecular mechanisms of thymoquinone as anticancer agent. Comb. Chem. High Throughput Screen. 2021, 24, 1644–1653. [Google Scholar] [CrossRef]
- Khan, M.A.; Younus, H. Thymoquinone shows the diverse therapeutic actions by modulating multiple cell signaling pathways: Single drug for multiple targets. Curr. Pharm. Biotechnol. 2018, 19, 934–945. [Google Scholar] [CrossRef]
- Badary, O.; Al-Shabanah, O.A.; Nagi, M.N.; Al-Bekairi, A.M.; Elmazar, M.M. Acute and subchronic toxicity of thymoquinone in mice. Drug Dev. Res. 1998, 44, 56–61. [Google Scholar] [CrossRef]
- Abukhader, M.M. Thymoquinone in the clinical treatment of cancer: Fact or fiction? Pharmacogn. Rev. 2013, 7, 117–120. [Google Scholar] [CrossRef] [PubMed]
- Park, E.J.; Chauhan, A.K.; Min, K.J.; Park, D.C.; Kwon, T.K. Thymoquinone induces apoptosis through downregulation of c-FLIP and Bcl-2 in renal carcinoma Caki cells. Oncol. Rep. 2016, 36, 2261–2267. [Google Scholar] [CrossRef] [PubMed]
- Gurung, R.L.; Lim, S.N.; Khaw, A.K.; Soon, J.F.; Shenoy, K.; Mohamed Ali, S.; Jayapal, M.; Sethu, S.; Baskar, R.; Hande, M.P. Thymoquinone induces telomere shortening, DNA damage and apoptosis in human glioblastoma cells. PLoS ONE 2010, 5, e12124. [Google Scholar] [CrossRef] [PubMed]
- El-Najjar, N.; Chatila, M.; Moukadem, H.; Vuorela, H.; Ocker, M.; Gandesiri, M.; Schneider-Stock, R.; Gali-Muhtasib, H. Reactive oxygen species mediate thymoquinone-induced apoptosis and activate ERK and JNK signaling. Apoptosis 2010, 15, 183–195. [Google Scholar] [CrossRef]
- Kou, B.; Liu, W.; Zhao, W.; Duan, P.; Yang, Y.; Yi, Q.; Guo, F.; Li, J.; Zhou, J.; Kou, Q. Thymoquinone inhibits epithelial-mesenchymal transition in prostate cancer cells by negatively regulating the TGF-β/Smad2/3 signaling pathway. Oncol. Rep. 2017, 38, 3592–3598. [Google Scholar] [CrossRef] [PubMed]
- Karimian, A.; Majidinia, M.; Moliani, A.; Alemi, F.; Asemi, Z.; Yousefi, B.; Fazlollahpour Naghibi, A. The modulatory effects of two bioflavonoids, quercetin and thymoquinone on the expression levels of DNA damage and repair genes in human breast, lung and prostate cancer cell lines. Pathol. Res. Pract. 2022, 240, 154143. [Google Scholar] [CrossRef] [PubMed]
- Chu, S.C.; Hsieh, Y.S.; Yu, C.C.; Lai, Y.Y.; Chen, P.N. Thymoquinone induces cell death in human squamous carcinoma cells via caspase activation-dependent apoptosis and LC3-II activation-dependent autophagy. PLoS ONE 2014, 9, e101579. [Google Scholar] [CrossRef]
- Imran, M.; Rauf, A.; Khan, I.A.; Shahbaz, M.; Qaisrani, T.B.; Fatmawati, S.; Abu-Izneid, T.; Imran, A.; Rahman, K.U.; Gondal, T.A. Thymoquinone: A novel strategy to combat cancer: A review. Biomed. Pharmacother. 2018, 106, 390–402. [Google Scholar] [CrossRef]
- El-Far, A.H.; Tantawy, M.A.; Al Jaouni, S.K.; Mousa, S.A. Thymoquinone-chemotherapeutic combinations: New regimen to combat cancer and cancer stem cells. Naunyn Schmiedebergs Arch. Pharmacol. 2020, 393, 1581–1598. [Google Scholar] [CrossRef]
- Idris, S.; Refaat, B.; Almaimani, R.A.; Ahmed, H.G.; Ahmad, J.; Alhadrami, M.; El-Readi, M.Z.; Elzubier, M.E.; Alaufi, H.A.A.; Al-Amin, B.; et al. Enhanced in vitro tumoricidal effects of 5-Fluorouracil, thymoquinone, and active vitamin D3 triple therapy against colon cancer cells by attenuating the PI3K/AKT/mTOR pathway. Life Sci. 2022, 296, 120442. [Google Scholar] [CrossRef] [PubMed]
- Farrash, W.F.; Aslam, A.; Almaimani, R.; Minshawi, F.; Almasmoum, H.; Alsaegh, A.; Iqbal, M.S.; Tabassum, A.; Elzubier, M.E.; El-Readi, M.Z.; et al. Metformin and thymoquinone co-treatment enhance 5-fluorouracil cytotoxicity by suppressing the PI3K/mTOR/HIF1α pathway and increasing oxidative stress in colon cancer cells. Biofactors 2023, 49, 831–848. [Google Scholar] [CrossRef] [PubMed]
- Fatfat, Z.; Fatfat, M.; Gali-Muhtasib, H. Therapeutic potential of thymoquinone in combination therapy against cancer and cancer stem cells. World J. Clin. Oncol. 2021, 12, 522–543. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; Du, H.; Wang, L.; Yue, Y.; Zhang, P.; Huang, Z.; Lv, W.; Ma, J.; Shao, Q.; Ma, M.; et al. Thymoquinone suppresses invasion and metastasis in bladder cancer cells by reversing EMT through the Wnt/β-catenin signaling pathway. Chem. Biol. Interact. 2020, 320, 109022. [Google Scholar] [CrossRef] [PubMed]
- Koveitypour, Z.; Panahi, F.; Vakilian, M.; Peymani, M.; Forootan, F.S.; Esfahani, M.H.N.; Ghaedi, K. Signaling pathways involved in colorectal cancer progression. Cell Biosci. 2019, 9, 97. [Google Scholar] [CrossRef] [PubMed]
- Chen, M.C.; Lee, N.H.; Hsu, H.H.; Ho, T.J.; Tu, C.C.; Chen, R.J.; Lin, Y.M.; Viswanadha, V.P.; Kuo, W.W.; Huang, C.Y. Inhibition of NF-κB and metastasis in irinotecan (CPT-11)-resistant LoVo colon cancer cells by thymoquinone via JNK and p38. Environ. Toxicol. 2017, 32, 669–678. [Google Scholar] [CrossRef] [PubMed]
- Gali-Muhtasib, H.; Ocker, M.; Kuester, D.; Krueger, S.; El-Hajj, Z.; Diestel, A.; Evert, M.; El-Najjar, N.; Peters, B.; Jurjus, A.; et al. Thymoquinone reduces mouse colon tumor cell invasion and inhibits tumor growth in murine colon cancer models. J. Cell Mol. Med. 2008, 12, 330–342. [Google Scholar] [CrossRef]
- Al Bitar, S.; Ballout, F.; Monzer, A.; Kanso, M.; Saheb, N.; Mukherji, D.; Faraj, W.; Tawil, A.; Doughan, S.; Hussein, M.; et al. Thymoquinone radiosensitizes human colorectal cancer cells in 2D and 3D culture models. Cancers 2022, 14, 1363. [Google Scholar] [CrossRef]
- Osorio-Pérez, S.M.; Estrada-Meza, C.; Ruiz-Manriquez, L.M.; Arvizu-Espinosa, M.G.; Srivastava, A.; Sharma, A.; Paul, S. Thymoquinone potentially modulates the expression of key onco- and tumor suppressor miRNAs in prostate and colon cancer cell lines: Insights from PC3 and HCT-15 cells. Genes 2023, 14, 1730. [Google Scholar] [CrossRef]
- Özkoç, M.; Mutlu Altundag, E. Antiproliferative effect of thymoquinone on human colon cancer cells: Is it dependent on glycolytic pathway? Acibadem Univ. Saglik. Bilim. Derg. 2023, 14, 103–107. [Google Scholar] [CrossRef]
- Thabet, N.A.; El-Khouly, D.; Sayed-Ahmed, M.M.; Omran, M.M. Thymoquinone chemosensitizes human colorectal cancer cells to imatinib via uptake/efflux genes modulation. Clin. Exp. Pharmacol. Physiol. 2021, 48, 911–920. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, D.; Eide, P.W.; Eilertsen, I.A.; Danielsen, S.A.; Eknæs, M.; Hektoen, M.; Lind, G.E.; Lothe, R.A. Epigenetic and genetic features of 24 colon cancer cell lines. Oncogenesis 2013, 2, e71. [Google Scholar] [CrossRef] [PubMed]
- Bertrand, F.E.; Angus, C.W.; Partis, W.J.; Sigounas, G. Developmental pathways in colon cancer: Crosstalk between WNT, BMP, Hedgehog and Notch. Cell Cycle 2012, 11, 4344–4351. [Google Scholar] [CrossRef] [PubMed]
- Hon, K.W.; Zainal Abidin, S.A.; Othman, I.; Naidu, R. The crosstalk between signaling pathways and cancer metabolism in colorectal cancer. Front. Pharmacol. 2021, 12, 768861. [Google Scholar] [CrossRef] [PubMed]
- Farooqi, A.A.; de la Roche, M.; Djamgoz, M.B.A.; Siddik, Z.H. Overview of the oncogenic signaling pathways in colorectal cancer: Mechanistic insights. Semin. Cancer Biol. 2019, 58, 65–79. [Google Scholar] [CrossRef] [PubMed]
- Farooqi, A.A.; Attar, R.; Xu, B. Anticancer and anti-metastatic role of thymoquinone: Regulation of oncogenic signaling cascades by thymoquinone. Int. J. Mol. Sci. 2022, 23, 6311. [Google Scholar] [CrossRef] [PubMed]
- Zhang, L.; Bai, Y.; Yang, Y. Thymoquinone chemosensitizes colon cancer cells through inhibition of NF-κB. Oncol. Lett. 2016, 12, 2840–2845. [Google Scholar] [CrossRef] [PubMed]
- Kundu, J.; Choi, B.Y.; Jeong, C.H.; Kundu, J.K.; Chun, K.S. Thymoquinone induces apoptosis in human colon cancer HCT116 cells through inactivation of STAT3 by blocking JAK2- and Src-mediated phosphorylation of EGF receptor tyrosine kinase. Oncol. Rep. 2014, 32, 821–828. [Google Scholar] [CrossRef]
- Wirries, A.; Breyer, S.; Quint, K.; Schobert, R.; Ocker, M. Thymoquinone hydrazone derivatives cause cell cycle arrest in p53-competent colorectal cancer cells. Exp. Ther. Med. 2010, 1, 369–375. [Google Scholar] [CrossRef]
- Hsu, H.H.; Chen, M.C.; Day, C.H.; Lin, Y.M.; Li, S.Y.; Tu, C.C.; Padma, V.V.; Shih, H.N.; Kuo, W.W.; Huang, C.Y. Thymoquinone suppresses migration of LoVo human colon cancer cells by reducing prostaglandin E2 induced COX-2 activation. World J. Gastroenterol. 2017, 23, 1171–1179. [Google Scholar] [CrossRef]
- Rooney, S.; Ryan, M.F. Effects of alpha-hederin and thymoquinone, constituents of Nigella sativa, on human cancer cell lines. Anticancer Res. 2005, 25, 2199–2204. [Google Scholar] [PubMed]
- Gali-Muhtasib, H.; Diab-Assaf, M.; Boltze, C.; Al-Hmaira, J.; Hartig, R.; Roessner, A.; Schneider-Stock, R. Thymoquinone extracted from black seed triggers apoptotic cell death in human colorectal cancer cells via a p53-dependent mechanism. Int. J. Oncol. 2004, 25, 857–866. [Google Scholar] [PubMed]
- Gali-Muhtasib, H.; Kuester, D.; Mawrin, C.; Bajbouj, K.; Diestel, A.; Ocker, M.; Habold, C.; Foltzer-Jourdainne, C.; Schoenfeld, P.; Peters, B.; et al. Thymoquinone triggers inactivation of the stress response pathway sensor CHEK1 and contributes to apoptosis in colorectal cancer cells. Cancer Res. 2008, 68, 5609–5618. [Google Scholar] [CrossRef] [PubMed]
- Chen, M.C.; Lee, N.H.; Hsu, H.H.; Ho, T.J.; Tu, C.C.; Hsieh, D.J.; Lin, Y.M.; Chen, L.M.; Kuo, W.W.; Huang, C.Y. Thymoquinone induces caspase-independent, autophagic cell death in CPT-11-resistant lovo colon cancer via mitochondrial dysfunction and activation of JNK and p38. J. Agric. Food Chem. 2015, 63, 1540–1546. [Google Scholar] [CrossRef] [PubMed]
- Asfour, W.; Almadi, S.; Haffar, L. Thymoquinone suppresses cellular proliferation, inhibits VEGF production and obstructs tumor progression and invasion in the rat model of DMH-induced colon carcinogenesis. Pharm. Pharmacol. 2013, 4, 7–17. [Google Scholar] [CrossRef]
- Bardelčíková, A.; Šoltys, J.; Mojžiš, J. Oxidative stress, inflammation and colorectal cancer: An overview. Antioxidants 2023, 12, 901. [Google Scholar] [CrossRef] [PubMed]
- Boakye, D.; Jansen, L.; Schöttker, B.; Jansen, E.H.J.M.; Schneider, M.; Halama, N.; Gào, X.; Chang-Claude, J.; Hoffmeister, M.; Brenner, H. Blood markers of oxidative stress are strongly associated with poorer prognosis in colorectal cancer patients. Int. J. Cancer 2020, 147, 2373–2386. [Google Scholar] [CrossRef]
- Gackowski, D.; Banaszkiewicz, Z.; Rozalski, R.; Jawien, A.; Olinski, R. Persistent oxidative stress in colorectal carcinoma patients. Int. J. Cancer 2002, 101, 395–397. [Google Scholar] [CrossRef]
- Vodicka, P.; Urbanova, M.; Makovicky, P.; Tomasova, K.; Kroupa, M.; Stetina, R.; Opattova, A.; Kostovcikova, K.; Siskova, A.; Schneiderova, M.; et al. Oxidative damage in sporadic colorectal cancer: Molecular mapping of base excision repair glycosylases in colorectal cancer patients. Int. J. Mol. Sci. 2020, 21, 2473. [Google Scholar] [CrossRef]
- Al-Johar, D.; Shinwari, N.; Arif, J.; Al-Sanea, N.; Jabbar, A.A.; El-Sayed, R.; Mashhour, A.; Billedo, G.; El-Doush, I.; Al-Saleh, I. Role of Nigella sativa and a number of its antioxidant constituents towards azoxymethane-induced genotoxic effects and colon cancer in rats. Phytother. Res. 2008, 22, 1311–1323. [Google Scholar] [CrossRef]
- Jrah-Harzallah, H.; Ben-Hadj-Khalifa, S.; Almawi, W.Y.; Maaloul, A.; Houas, Z.; Mahjoub, T. Effect of thymoquinone on 1,2-dimethyl-hydrazine-induced oxidative stress during initiation and promotion of colon carcinogenesis. Eur. J. Cancer 2013, 49, 1127–1135. [Google Scholar] [CrossRef]
- Harzallah, H.J.; Grayaa, R.; Kharoubi, W.; Maaloul, A.; Hammami, M.; Mahjoub, T. Thymoquinone, the Nigella sativa bioactive compound, prevents circulatory oxidative stress caused by 1,2-dimethylhydrazine in erythrocyte during colon postinitiation carcinogenesis. Oxid. Med. Cell. Longev. 2012, 2012, 854065. [Google Scholar]
- Blondy, S.; David, V.; Verdier, M.; Mathonnet, M.; Perraud, A.; Christou, N. 5-Fluorouracil resistance mechanisms in colorectal cancer: From classical pathways to promising processes. Cancer Sci. 2020, 111, 3142–3154. [Google Scholar] [CrossRef]
- Pouya, F.D.; Gazouli, M.; Rasmi, Y.; Lampropoulou, D.I.; Nemati, M. MicroRNAs and drug resistance in colorectal cancer with special focus on 5-fluorouracil. Mol. Biol. Rep. 2022, 49, 5165–5178. [Google Scholar] [CrossRef]
- Gmeiner, W.H.; Okechukwu, C.C. Review of 5-FU resistance mechanisms in colorectal cancer: Clinical significance of attenuated on-target effects. Cancer Drug Resist. 2023, 6, 257–272. [Google Scholar] [CrossRef]
- Shibata, J.; Aiba, K.; Shibata, H.; Minowa, S.; Horikoshi, N. Detection and quantitation of thymidylate synthase mRNA in human colon adenocarcinoma cell line resistant to 5-fluorouracil by competitive PCR. Anticancer Res. 1998, 18, 1457–1463. [Google Scholar]
- Lv, L.; Liu, H.G.; Dong, S.Y.; Yang, F.; Wang, Q.X.; Guo, G.L.; Pan, Y.F.; Zhang, X.H. Upregulation of CD44v6 contributes to acquired chemoresistance via the modulation of autophagy in colon cancer SW480 cells. Tumour Biol. 2016, 37, 8811–8824. [Google Scholar] [CrossRef]
- Chen, J.; Na, R.; Xiao, C.; Wang, X.; Wang, Y.; Yan, D.; Song, G.; Liu, X.; Chen, J.; Lu, H.; et al. The loss of SHMT2 mediates 5-fluorouracil chemoresistance in colorectal cancer by upregulating autophagy. Oncogene 2021, 40, 3974–3988. [Google Scholar] [CrossRef]
- Romano, G.; Santi, L.; Bianco, M.R.; Giuffrè, M.R.; Pettinato, M.; Bugarin, C.; Garanzini, C.; Savarese, L.; Leoni, S.; Cerrito, M.G.; et al. The TGF-β pathway is activated by 5-fluorouracil treatment in drug resistant colorectal carcinoma cells. Oncotarget 2016, 7, 22077–22091. [Google Scholar] [CrossRef]
- Longley, D.B.; Johnston, P.G. Molecular mechanisms of drug resistance. J. Pathol. 2005, 205, 275–292. [Google Scholar] [CrossRef]
- Schiavoni, G.; Gabriele, L.; Mattei, F. The tumor microenvironment: A pitch for multiple players. Front. Oncol. 2013, 3, 90. [Google Scholar] [CrossRef]
- Crea, F.; Nobili, S.; Paolicchi, E.; Perrone, G.; Napoli, C.; Landini, I.; Danesi, R.; Mini, E. Epigenetics and chemoresistance in colorectal cancer: An opportunity for treatment tailoring and novel therapeutic strategies. Drug Resist. Updat. 2011, 14, 280–296. [Google Scholar] [CrossRef]
- Fazzone, W.; Wilson, P.M.; LaBonte, M.J.; Lenz, H.-J.; Ladner, R.D. Histone deacetylase inhibitors suppress thymidylate synthase gene expression and synergize with the fluoropyrimidines in colon cancer cells. Int. J. Cancer 2009, 125, 463–473. [Google Scholar] [CrossRef]
- Kensara, O.A.; El-Shemi, A.G.; Mohamed, A.M.; Refaat, B.; Idris, S.; Ahmad, J. Thymoquinone subdues tumor growth and potentiates the chemopreventive effect of 5-fluorouracil on the early stages of colorectal carcinogenesis in rats. Drug Des. Dev. Ther. 2016, 10, 2239–2253. [Google Scholar]
- Eftekhar, S.P.; Kazemi, S.; Moghadamnia, A.A. Effect of thymoquinone on pharmacokinetics of 5-fluorouracil in rats and its effect on human cell line in vitro. Hum. Exp. Toxicol. 2022, 41, 9603271221145422. [Google Scholar] [CrossRef]
- Ballout, F.; Monzer, A.; Fatfat, M.; Ouweini, H.E.; Jaffa, M.A.; Abdel-Samad, R.; Darwiche, N.; Abou-Kheir, W.; Gali-Muhtasib, H. Thymoquinone induces apoptosis and DNA damage in 5-Fluorouracil-resistant colorectal cancer stem/progenitor cells. Oncotarget 2020, 11, 2959–2972. [Google Scholar] [CrossRef]
- Ndreshkjana, B.; Çapci, A.; Klein, V.; Chanvorachote, P.; Muenzner, J.K.; Huebner, K.; Steinmann, S.; Erlenbach-Wuensch, K.; Geppert, C.I.; Agaimy, A.; et al. Combination of 5-fluorouracil and thymoquinone targets stem cell gene signature in colorectal cancer cells. Cell Death Dis. 2019, 10, 379. [Google Scholar] [CrossRef]
- Norwood, A.A.; Tucci, M.; Benghuzzi, H. A comparison of 5-fluorouracil and natural chemotherapeutic agents, EGCG and thymoquinone, delivered by sustained drug delivery on colon cancer cells. Biomed. Sci. Instrum. 2007, 43, 272–277. [Google Scholar]
- Zaki, S.M.; Waggas, D.S. Protective effect of Nigella sativa and onion extract against 5-fluorouracil-induced hepatic toxicity. Nutr. Cancer 2022, 74, 2657–2670. [Google Scholar] [CrossRef]
- Madani, F.; Kazemi, S.; Shirafkan, F.; Lotfi, M.; Hosseini, S.M.; Moghadamnia, A.A. Thymoquinone protects against 5-fluorouracil-induced mucositis by NF-κβ and HIF-1 mechanisms in mice. J. Biochem. Mol. Toxicol. 2023, 37, e23405. [Google Scholar] [CrossRef]
- Effenberger-Neidnicht, K.; Schobert, R. Combinatorial effects of thymoquinone on the anti-cancer activity of doxorubicin. Cancer Chemother. Pharmacol. 2011, 67, 867–874. [Google Scholar] [CrossRef] [PubMed]
- Khalife, R.; Hodroj, M.H.; Fakhoury, R.; Rizk, S. Thymoquinone from Nigella sativa seeds promotes the antitumor activity of noncytotoxic doses of topotecan in human colorectal cancer cells in vitro. Planta Med. 2016, 82, 312–321. [Google Scholar] [CrossRef] [PubMed]
CRC Line | IC50 (µM) | Exposure Time (h) | Ref. |
---|---|---|---|
LoVo | 6–8 | 24 | [97] |
C26 | 40 | 24 | [98] |
HT-29 | 112 | 24 | [99] |
DLD-1 | 61 | ||
HCT-116 | 51.73 | 48 | |
HT-29 | 99.46 | ||
HCT-15 | 82.59 | 24 | [100] |
HCT-116 | 68 | 24 | [101] |
HCT-116 | 20 | 24 | [102] |
10.3 | 48 | ||
6.5 | 72 |
Chemotherapeutic Agent | Mechanism of Action | CRC Line | Effect of Combination Treatment | Ref. |
---|---|---|---|---|
IM | Protein tyrosine kinase inhibitor | HCT116 | TQ-augmented cytotoxic activity of IM and synergized IM effect; TQ + IM decreases the expression of ABCB1, ABCG2 and hOCT1; TQ + IM increases the uptake/efflux ratio of imatinib | [102] |
DOX | DNA intercalation; Inhibitor of topoisomerase II; Generation of free radicals | HT-29 | DOX + TQ was more effective than DOX alone against HT-29 cells desensitized by repeated DOX exposure | [142] |
TP | Inhibitor of topoisomerase I | HT-29 | Increased production of fragmented DNA; Inhibiting proliferation and lowering toxicity through p53- and Bax/Bcl2-independent mechanisms | [143] |
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. |
© 2023 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 (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kurowska, N.; Madej, M.; Strzalka-Mrozik, B. Thymoquinone: A Promising Therapeutic Agent for the Treatment of Colorectal Cancer. Curr. Issues Mol. Biol. 2024, 46, 121-139. https://doi.org/10.3390/cimb46010010
Kurowska N, Madej M, Strzalka-Mrozik B. Thymoquinone: A Promising Therapeutic Agent for the Treatment of Colorectal Cancer. Current Issues in Molecular Biology. 2024; 46(1):121-139. https://doi.org/10.3390/cimb46010010
Chicago/Turabian StyleKurowska, Natalia, Marcel Madej, and Barbara Strzalka-Mrozik. 2024. "Thymoquinone: A Promising Therapeutic Agent for the Treatment of Colorectal Cancer" Current Issues in Molecular Biology 46, no. 1: 121-139. https://doi.org/10.3390/cimb46010010