Phytochemicals as Chemo-Preventive Agents and Signaling Molecule Modulators: Current Role in Cancer Therapeutics and Inflammation
Abstract
:1. Introduction
2. Mechanisms of Cancer Chemoprevention
2.1. Signal Transduction Mechanisms
2.2. Key Signaling Pathways
2.3. Angiogenesis Inhibition and Tumor Dissemination/Metastasis
2.4. Cell Death and Cell Cycle Halt
2.5. Current Cancer Therapy Involving Phytochemicals
2.6. Vinca Alkaloids
2.7. Taxanes (Paclitaxel)
2.8. Camptothecin (CPT)
2.9. PTOX
2.10. Additional Anticancer Drugs Derived from Plants
2.11. Inflammation vs. Cancer
2.12. Colorectal Cancer
2.13. Prostate Cancer
3. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
PTOX | Podophyllotoxin |
CPT | Camptothecin |
ROS | Reactive oxygen species |
IL-1 | Interleukin 1 |
TNF-α | Tumor necrosis factor |
NO | Nitric oxide |
EGCG | Epigallocatechin gallate |
NSCLC | Non-small-cell lung carcinoma |
PKC | Protein kinase C |
FDA | US Food and Drug Administration |
MAPs | Microtubule associated proteins |
MIPs | Microtubule interacting proteins |
PTMs | Post translational modifications |
STAT3 | Signal Transducers and Activators of Transcription-3 |
VEGF | Vascular endothelial growth factor |
UCP-2 | Uncoupling protein 2 |
TLR-4 | Toll like receptor 4 |
JNK/SAPK | c-Jun N-terminal kinase/stress-activated protein kinase |
EPC | Endothelial progenitor cells |
MAPKs | Mitogen-activated protein kinases |
TME | Tumor microenvironment |
ERK1/2 | Extracellular signal-regulated protein kinase |
NPs | Nanoparticles |
CPT-11,4 | Irinotecan |
CKD-602,5 | Belotecan |
HCPT | 10-hydroxycamptothecin |
DMEP | 4’-demethylepipodophyllotoxin |
PPP | Picropodophyllotoxin |
IGF-1R | Insulin-like growth factor-1 receptor |
IR | Insulin receptor |
pErk1/2 | Phosphorylated extracellular signal-regulated kinases 1 and 2 |
IM | Ingenol mebutate |
HHT | Homoharringtonine |
CMML | Myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia |
EGF | Epidermal growth factor |
FOXO3a | Fork head box O3a |
GSK3 | Glycogen synthase kinase 3 |
HUVECs | Human umbilical vein endothelial cells |
CDC | The Centers for Disease Control and Prevention |
CXCL-1,2 | C-X-C motif ligands 1 and 2 |
References
- Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, H.; Khor, T.O.; Shu, L.; Su, Z.Y.; Fuentes, F.; Lee, J.H.; Kong, A.N. Plants vs. cancer: A review on natural phytochemicals in preventing and treating cancers and their druggability. Anti-Cancer Agents Med. Chem. 2012, 12, 1281–1305. [Google Scholar] [CrossRef]
- De Flora, S.; Ferguson, L.R. Overview of mechanisms of cancer chemopreventive agents. Mutat. Res. 2005, 591, 8–15. [Google Scholar] [CrossRef]
- Nussbaumer, S.; Bonnabry, P.; Veuthey, J.L.; Fleury-Souverain, S. Analysis of anticancer drugs: A review. Talanta 2011, 85, 2265–2289. [Google Scholar] [CrossRef]
- Sharifi-Rad, J.; Dey, A.; Koirala, N.; Shaheen, S.; El Omari, N.; Salehi, B.; Goloshvili, T.; Cirone Silva, N.C.; Bouyahya, A.; Vitalini, S.; et al. Cinnamomum Species: Bridging Phytochemistry Knowledge, Pharmacological Properties and Toxicological Safety for Health Benefits. Front. Pharmacol. 2021, 12, 600139. [Google Scholar] [CrossRef]
- Bellik, Y.; Boukraâ, L.; Alzahrani, H.A.; Bakhotmah, B.A.; Abdellah, F.; Hammoudi, S.M.; Iguer-Ouada, M. Molecular mechanism underlying anti-inflammatory and anti-allergic activities of phytochemicals: An update. Molecules 2012, 18, 322–353. [Google Scholar] [CrossRef] [Green Version]
- Zhu, F.; Du, B.; Xu, B. Anti-inflammatory effects of phytochemicals from fruits, vegetables, and food legumes: A review. Crit. Rev. Food Sci. Nutr. 2018, 58, 1260–1270. [Google Scholar] [CrossRef]
- Zhao, R.; Liang, H.; Clarke, E.; Jackson, C.; Xue, M. Inflammation in Chronic Wounds. Int. J. Mol. Sci. 2016, 17, 2085. [Google Scholar] [CrossRef]
- Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef] [PubMed]
- Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018, 9, 7204–7218. [Google Scholar] [CrossRef]
- Furman, D.; Campisi, J.; Verdin, E.; Carrera-Bastos, P.; Targ, S.; Franceschi, C.; Ferrucci, L.; Gilroy, D.W.; Fasano, A.; Miller, G.W.; et al. Chronic inflammation in the etiology of disease across the life span. Nat. Med. 2019, 25, 1822–1832. [Google Scholar] [CrossRef]
- Cote, B.; Carlson, L.J.; Rao, D.A.; Alani, A.W.G. Combinatorial resveratrol and quercetin polymeric micelles mitigate doxorubicin induced cardiotoxicity in vitro and in vivo. J. Control. Release Off. J. Control Release Soc. 2015, 213, 128–133. [Google Scholar] [CrossRef] [PubMed]
- Kocaadam, B.; Şanlier, N. Curcumin, an active component of turmeric (Curcuma longa), and its effects on health. Crit. Rev. Food Sci. Nutr. 2017, 57, 2889–2895. [Google Scholar] [CrossRef] [PubMed]
- Meghwal, M.; Goswami, T.K. Piper nigrum and piperine: An update. Phytother. Res. PTR 2013, 27, 1121–1130. [Google Scholar] [CrossRef]
- Vázquez Cisneros, L.C.; López-Uriarte, P.; López-Espinoza, A.; Navarro Meza, M.; Espinoza-Gallardo, A.C.; Guzmán Aburto, M.B. Effects of green tea and its epigallocatechin (EGCG) content on body weight and fat mass in humans: A systematic review. Nutr. Hosp. 2017, 34, 731–737. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mohd Yusof, Y.A. Gingerol and Its Role in Chronic Diseases. Adv. Exp. Med. Biol. 2016, 929, 177–207. [Google Scholar]
- Pérez-Ruiz, E.; Melero, I.; Kopecka, J.; Sarmento-Ribeiro, A.B.; García-Aranda, M.; De Las Rivas, J. Cancer immunotherapy resistance based on immune checkpoints inhibitors: Targets, biomarkers, and remedies. Drug Resist. Updates Rev. Comment. Antimicrob. Anticancer Chemother. 2020, 53, 100718. [Google Scholar] [CrossRef]
- 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]
- Gasparri, M.L.; Bardhi, E.; Ruscito, I.; Papadia, A.; Farooqi, A.A.; Marchetti, C.; Bogani, G.; Ceccacci, I.; Mueller, M.D.; Benedetti Panici, P. PI3K/AKT/mTOR Pathway in Ovarian Cancer Treatment: Are We on the Right Track? Geburtshilfe Frauenheilkd. 2017, 77, 1095–1103. [Google Scholar] [CrossRef] [Green Version]
- Haque, A.; Brazeau, D.; Amin, A.R. Perspectives on natural compounds in chemoprevention and treatment of cancer: An update with new promising compounds. Eur. J. Cancer 2021, 149, 165–183. [Google Scholar] [CrossRef]
- Rizeq, B.; Gupta, I.; Ilesanmi, J.; AlSafran, M.; Rahman, M.M.; Ouhtit, A. The Power of Phytochemicals Combination in Cancer Chemoprevention. J. Cancer 2020, 11, 4521–4533. [Google Scholar] [CrossRef] [PubMed]
- Chen, C.; Kong, A.N. Dietary cancer-chemopreventive compounds: From signaling and gene expression to pharmacological effects. Trends Pharmacol. Sci. 2005, 26, 318–326. [Google Scholar] [CrossRef] [PubMed]
- Kotecha, R.; Takami, A.; Espinoza, J.L. Dietary phytochemicals and cancer chemoprevention: A review of the clinical evidence. Oncotarget 2016, 7, 52517–52529. [Google Scholar] [CrossRef] [Green Version]
- Michl, C.; Vivarelli, F.; Weigl, J.; De Nicola, G.R.; Canistro, D.; Paolini, M.; Iori, R.; Rascle, A. The Chemopreventive Phytochemical Moringin Isolated from Moringa oleifera Seeds Inhibits JAK/STAT Signaling. PLoS ONE 2016, 11, e0157430. [Google Scholar] [CrossRef] [Green Version]
- Muhammad, N.; Usmani, D.; Tarique, M.; Naz, H.; Ashraf, M.; Raliya, R.; Tabrez, S.; Zughaibi, T.A.; Alsaieedi, A.; Hakeem, I.J.; et al. The Role of Natural Products and Their Multitargeted Approach to Treat Solid Cancer. Cells 2022, 11, 2209. [Google Scholar] [CrossRef]
- Pralea, I.E.; Petrache, A.M.; Tigu, A.B.; Gulei, D.; Moldovan, R.C.; Ilieș, M.; Nicoară, R.; Hegheș, S.C.; Uifălean, A.; Iuga, C.A. Phytochemicals as Regulators of Tumor Glycolysis and Hypoxia Signaling Pathways: Evidence from In Vitro Studies. Pharmaceuticals 2022, 15, 808. [Google Scholar] [CrossRef]
- Rahman, M.A.; Hannan, M.A.; Dash, R.; Rahman, M.H.; Islam, R.; Uddin, M.J.; Sohag, A.A.M.; Rahman, M.H.; Rhim, H. Phytochemicals as a Complement to Cancer Chemotherapy: Pharmacological Modulation of the Autophagy-Apoptosis Pathway. Front. Pharmacol. 2021, 12, 639628. [Google Scholar] [CrossRef] [PubMed]
- Khan, A.; Siddiqui, S.; Husain, S.A.; Mazurek, S.; Iqbal, M.A. Phytocompounds Targeting Metabolic Reprogramming in Cancer: An Assessment of Role, Mechanisms, Pathways, and Therapeutic Relevance. J. Agric. Food Chem. 2021, 69, 6897–6928. [Google Scholar] [CrossRef]
- Wee, P.; Wang, Z. Epidermal Growth Factor Receptor Cell Proliferation Signaling Pathways. Cancers 2017, 9, 52. [Google Scholar] [CrossRef] [Green Version]
- Hu, X.; Li, J.; Fu, M.; Zhao, X.; Wang, W. The JAK/STAT signaling pathway: From bench to clinic. Signal Transduct. Target. Ther. 2021, 6, 402. [Google Scholar] [CrossRef]
- Chen, K.; Li, Y.; Zhang, X.; Ullah, R.; Tong, J.; Shen, Y. The role of the PI3K/AKT signalling pathway in the corneal epithelium: Recent updates. Cell Death Dis. 2022, 13, 513. [Google Scholar] [CrossRef] [PubMed]
- Shiau, J.P.; Chuang, Y.T.; Tang, J.Y.; Yang, K.H.; Chang, F.R.; Hou, M.F.; Yen, C.Y.; Chang, H.W. The Impact of Oxidative Stress and AKT Pathway on Cancer Cell Functions and Its Application to Natural Products. Antioxidants 2022, 11, 1845. [Google Scholar] [CrossRef] [PubMed]
- Wan Mohd Tajuddin, W.N.B.; Lajis, N.H.; Abas, F.; Othman, I.; Naidu, R. Mechanistic Understanding of Curcumin’s Therapeutic Effects in Lung Cancer. Nutrients 2019, 11, 2989. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Raina, R.; Pramodh, S.; Rais, N.; Haque, S.; Shafarin, J.; Bajbouj, K.; Hamad, M.; Hussain, A. Luteolin inhibits proliferation, triggers apoptosis and modulates Akt/mTOR and MAP kinase pathways in HeLa cells. Oncol. Lett. 2021, 21, 192. [Google Scholar] [CrossRef]
- Lin, C.H.; Chang, C.Y.; Lee, K.R.; Lin, H.J.; Chen, T.H.; Wan, L. Flavones inhibit breast cancer proliferation through the Akt/FOXO3a signaling pathway. BMC Cancer 2015, 15, 958. [Google Scholar] [CrossRef] [Green Version]
- Abid, R.; Ghazanfar, S.; Farid, A.; Sulaman, S.M.; Idrees, M.; Amen, R.A.; Muzammal, M.; Shahzad, M.K.; Mohamed, M.O.; Khaled, A.A.; et al. Pharmacological Properties of 4′,5,7-Trihydroxyflavone (Apigenin) and Its Impact on Cell Signaling Pathways. Molecules 2022, 27, 4304. [Google Scholar] [CrossRef]
- Wang, Y.; Wu, H.; Dong, N.; Su, X.; Duan, M.; Wei, Y.; Wei, J.; Liu, G.; Peng, Q.; Zhao, Y. Sulforaphane induces S-phase arrest and apoptosis via p53-dependent manner in gastric cancer cells. Sci. Rep. 2021, 11, 2504. [Google Scholar] [CrossRef]
- Bharadwaj, U.; Kasembeli, M.M.; Robinson, P.; Tweardy, D.J. Targeting Janus Kinases and Signal Transducer and Activator of Transcription 3 to Treat Inflammation, Fibrosis, and Cancer: Rationale, Progress, and Caution. Pharmacol. Rev. 2020, 72, 486–526. [Google Scholar] [CrossRef] [Green Version]
- Michalkova, R.; Mirossay, L.; Gazdova, M.; Kello, M.; Mojzis, J. Molecular Mechanisms of Antiproliferative Effects of Natural Chalcones. Cancers 2021, 13, 2730. [Google Scholar] [CrossRef]
- Tian, T.; Sun, J.; Wang, J.; Liu, Y.; Liu, H. Isoliquiritigenin inhibits cell proliferation and migration through the PI3K/AKT signaling pathway in A549 lung cancer cells. Oncol. Lett. 2018, 16, 6133–6139. [Google Scholar] [CrossRef] [Green Version]
- Li, B.; Xu, N.; Wan, Z.; Ma, L.; Li, H.; Cai, W.; Chen, X.; Huang, Z.; He, Z. Isobavachalcone exerts anti-proliferative and pro-apoptotic effects on human liver cancer cells by targeting the ERKs/RSK2 signaling pathway. Oncol. Rep. 2019, 41, 3355–3366. [Google Scholar] [CrossRef] [PubMed]
- Liu, J.; Xiao, Q.; Xiao, J.; Niu, C.; Li, Y.; Zhang, X.; Zhou, Z.; Shu, G.; Yin, G. Wnt/β-catenin signalling: Function, biological mechanisms, and therapeutic opportunities. Signal Transduct. Target. Ther. 2022, 7, 3. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Y.; Wang, X. Targeting the Wnt/β-catenin signaling pathway in cancer. J. Hematol. Oncol. 2020, 13, 165. [Google Scholar] [CrossRef]
- Driehuis, E.; Clevers, H. WNT signalling events near the cell membrane and their pharmacological targeting for the treatment of cancer. Br. J. Pharmacol. 2017, 174, 4547–4563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Oliveira, L.F.S.; Predes, D.; Borges, H.L.; Abreu, J.G. Therapeutic Potential of Naturally Occurring Small Molecules to Target the Wnt/β-Catenin Signaling Pathway in Colorectal Cancer. Cancers 2022, 14, 403. [Google Scholar] [CrossRef]
- Vallée, A. Curcumin and Wnt/β-catenin signaling in exudative age-related macular degeneration (Review). Int. J. Mol. Med. 2022, 49, 1–13. [Google Scholar] [CrossRef]
- Ge, H.; Xu, C.; Chen, H.; Liu, L.; Zhang, L.; Wu, C.; Lu, Y.; Yao, Q. Traditional Chinese Medicines as Effective Reversals of Epithelial-Mesenchymal Transition Induced-Metastasis of Colorectal Cancer: Molecular Targets and Mechanisms. Front. Pharmacol. 2022, 13, 842295. [Google Scholar] [CrossRef]
- Zhu, J.; Jiang, Y.; Yang, X.; Wang, S.; Xie, C.; Li, X.; Li, Y.; Chen, Y.; Wang, X.; Meng, Y.; et al. Wnt/β-catenin pathway mediates (-)-Epigallocatechin-3-gallate (EGCG) inhibition of lung cancer stem cells. Biochem. Biophys. Res. Commun. 2017, 482, 15–21. [Google Scholar] [CrossRef]
- Miyashita, T.; Reed, J.C. Tumor suppressor p53 is a direct transcriptional activator of the human bax gene. Cell 1995, 80, 293–299. [Google Scholar]
- Kumar, A.; Kaur, S.; Dhiman, S.; Singh, P.P.; Bhatia, G.; Thakur, S.; Tuli, H.S.; Sharma, U.; Kumar, S.; Almutary, A.G.; et al. Targeting Akt/NF-κB/p53 Pathway and Apoptosis Inducing Potential of 1,2-Benzenedicarboxylic Acid, Bis (2-Methyl Propyl) Ester Isolated from Onosma bracteata Wall. against Human Osteosarcoma (MG-63) Cells. Molecules 2022, 27, 3478. [Google Scholar] [CrossRef]
- Aubrey, B.J.; Kelly, G.L.; Janic, A.; Herold, M.J.; Strasser, A. How does p53 induce apoptosis and how does this relate to p53-mediated tumour suppression? Cell Death Differ. 2018, 25, 104–113. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, H.; Park, S.; Zhang, H.; Park, S.; Kwon, W.; Kim, E.; Zhang, X.; Jang, S.; Yoon, D.; Choi, S.K.; et al. Targeting AKT with costunolide suppresses the growth of colorectal cancer cells and induces apoptosis in vitro and in vivo. J. Exp. Clin. Cancer Res. CR 2021, 40, 114. [Google Scholar] [CrossRef]
- Cordero-Herrera, I.; Martín, M.A.; Bravo, L.; Goya, L.; Ramos, S. Epicatechin gallate induces cell death via p53 activation and stimulation of p38 and JNK in human colon cancer SW480 cells. Nutr. Cancer 2013, 65, 718–728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.; Chen, B.; Xu, D.; Li, Z.; Sui, Y.; Lin, X. Delicaflavone Reverses Cisplatin Resistance via Endoplasmic Reticulum Stress Signaling Pathway in Non-Small Cell Lung Cancer Cells. OncoTargets Ther. 2020, 13, 10315–10322. [Google Scholar] [CrossRef] [PubMed]
- Javaid, A.; Zahra, D.; Rashid, F.; Mashraqi, M.; Alzamami, A.; Khurshid, M.; Ali Ashfaq, U. Regulation of micro-RNA, epigenetic factor by natural products for the treatment of cancers: Mechanistic insight and translational association. Saudi J. Biol. Sci. 2022, 29, 103255. [Google Scholar] [CrossRef]
- Xie, Y.H.; Chen, Y.X.; Fang, J.Y. Comprehensive review of targeted therapy for colorectal cancer. Signal Transduct. Target. Ther. 2020, 5, 22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, Y.; Li, N.; Wang, T.M.; Di, L. Natural Products with Activity against Lung Cancer: A Review Focusing on the Tumor Microenvironment. Int. J. Mol. Sci. 2021, 22, 10827. [Google Scholar] [CrossRef]
- García-Caballero, M.; Torres-Vargas, J.A.; Marrero, A.D.; Martínez-Poveda, B.; Medina, M.; Quesada, A.R. Angioprevention of Urologic Cancers by Plant-Derived Foods. Pharmaceutics 2022, 14, 256. [Google Scholar] [CrossRef]
- Sharifi-Rad, M.; Pezzani, R.; Redaelli, M.; Zorzan, M.; Imran, M.; Ahmed Khalil, A.; Salehi, B.; Sharopov, F.; Cho, W.C.; Sharifi-Rad, J. Preclinical Pharmacological Activities of Epigallocatechin-3-gallate in Signaling Pathways: An Update on Cancer. Molecules 2020, 25, 467. [Google Scholar] [CrossRef] [Green Version]
- Kim, J.D.; Liu, L.; Guo, W.; Meydani, M. Chemical structure of flavonols in relation to modulation of angiogenesis and immune-endothelial cell adhesion. J. Nutr. Biochem. 2006, 17, 165–176. [Google Scholar] [CrossRef]
- Maurya, A.K.; Vinayak, M. Quercetin Attenuates Cell Survival, Inflammation, and Angiogenesis via Modulation of AKT Signaling in Murine T-Cell Lymphoma. Nutr. Cancer 2017, 69, 470–480. [Google Scholar] [CrossRef] [PubMed]
- Fares, J.; Fares, M.Y.; Khachfe, H.H.; Salhab, H.A.; Fares, Y. Molecular principles of metastasis: A hallmark of cancer revisited. Signal Transduct. Target. Ther. 2020, 5, 28. [Google Scholar] [CrossRef] [PubMed]
- Ci, Y.; Qiao, J.; Han, M. Molecular Mechanisms and Metabolomics of Natural Polyphenols Interfering with Breast Cancer Metastasis. Molecules 2016, 21, 1634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Amawi, H.; Ashby, C.R.; Samuel, T.; Peraman, R.; Tiwari, A.K. Polyphenolic Nutrients in Cancer Chemoprevention and Metastasis: Role of the Epithelial-to-Mesenchymal (EMT) Pathway. Nutrients 2017, 9, 911. [Google Scholar] [CrossRef] [Green Version]
- Costea, T.; Hudiță, A.; Ciolac, O.A.; Gălățeanu, B.; Ginghină, O.; Costache, M.; Ganea, C.; Mocanu, M.M. Chemoprevention of Colorectal Cancer by Dietary Compounds. Int. J. Mol. Sci. 2018, 19, 3787. [Google Scholar] [CrossRef] [Green Version]
- Gaikwad, S.; Srivastava, S.K. Role of Phytochemicals in Perturbation of Redox Homeostasis in Cancer. Antioxidants 2021, 10, 83. [Google Scholar] [CrossRef]
- Rajendran, P.; Ho, E.; Williams, D.E.; Dashwood, R.H. Dietary phytochemicals, HDAC inhibition, and DNA damage/repair defects in cancer cells. Clin. Epigenetics 2011, 3, 4. [Google Scholar] [CrossRef] [Green Version]
- Manna, S.; Banerjee, S.; Mukherjee, S.; Das, S.; Panda, C.K.J.A. Epigallocatechin gallate induced apoptosis in Sarcoma180 cells in vivo: Mediated by p53 pathway and inhibition in U1B, U4-U6 UsnRNAs expression. Apoptosis 2006, 11, 2267–2276. [Google Scholar] [CrossRef]
- Hastak, K.; Agarwal, M.K.; Mukhtar, H.; Agarwal, M.L. Ablation of either p21 or Bax prevents p53-dependent apoptosis induced by green tea polyphenol epigallocatechin-3-gallate. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2005, 19, 789–791. [Google Scholar] [CrossRef]
- Kundu, T.; Dey, S.; Roy, M.; Siddiqi, M.; Bhattacharya, R.K. Induction of apoptosis in human leukemia cells by black tea and its polyphenol theaflavin. Cancer Lett. 2005, 230, 111–121. [Google Scholar] [CrossRef]
- Kalra, N.; Seth, K.; Prasad, S.; Singh, M.; Pant, A.B.; Shukla, Y. Retraction notice to “Theaflavins induced apoptosis of LNCaP cells is mediated through induction of p53, down-regulation of NF-kappa B and mitogen-activated protein kinases pathways” [Life Sci. 18/23 (2007) 2137-2146]. Life Sci. 2019, 219, 364. [Google Scholar] [CrossRef] [PubMed]
- Wu, Q.K.; Koponen, J.M.; Mykkänen, H.M.; Törrönen, A.R. Berry phenolic extracts modulate the expression of p21(WAF1) and Bax but not Bcl-2 in HT-29 colon cancer cells. J. Agric. Food Chem. 2007, 55, 1156–1163. [Google Scholar] [CrossRef] [PubMed]
- Favot, L.; Martin, S.; Keravis, T.; Andriantsitohaina, R.; Lugnier, C. Involvement of cyclin-dependent pathway in the inhibitory effect of delphinidin on angiogenesis. Cardiovasc. Res. 2003, 59, 479–487. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Martin, S.; Favot, L.; Matz, R.; Lugnier, C.; Andriantsitohaina, R. Delphinidin inhibits endothelial cell proliferation and cell cycle progression through a transient activation of ERK-1/-2. Biochem. Pharmacol. 2003, 65, 669–675. [Google Scholar] [CrossRef]
- Juric, V.; Murphy, B.J.C.D.R. Cyclin-dependent kinase inhibitors in brain cancer: Current state and future directions. Cancer Drug Resist. 2020, 3, 48. [Google Scholar] [CrossRef] [Green Version]
- Liu, E.; Wu, J.; Cao, W.; Zhang, J.; Liu, W.; Jiang, X.; Zhang, X. Curcumin induces G2/M cell cycle arrest in a p53-dependent manner and upregulates ING4 expression in human glioma. J. Neuro-Oncol. 2007, 85, 263–270. [Google Scholar] [CrossRef]
- Su, C.C.; Wang, M.J.; Chiu, T.L. The anti-cancer efficacy of curcumin scrutinized through core signaling pathways in glioblastoma. Int. J. Mol. Med. 2010, 26, 217–224. [Google Scholar]
- Nocito, M.C.; De Luca, A.; Prestia, F.; Avena, P.; La Padula, D.; Zavaglia, L.; Sirianni, R.; Casaburi, I.; Puoci, F.; Chimento, A.J.B. Antitumoral Activities of Curcumin and Recent Advances to ImProve Its Oral Bioavailability. Biomedicines 2021, 9, 1476. [Google Scholar] [CrossRef]
- Shan, J.; Dudenhausen, E.; Kilberg, M.S. Induction of early growth response gene 1 (EGR1) by endoplasmic reticulum stress is mediated by the extracellular regulated kinase (ERK) arm of the MAPK pathways. Biochim. Et Biophys. Acta (BBA)-Mol. Cell Res. 2019, 1866, 371–381. [Google Scholar] [CrossRef]
- Choi, B.H.; Kim, C.G.; Bae, Y.-S.; Lim, Y.; Lee, Y.H.; Shin, S.Y. p21Waf1/Cip1 expression by curcumin in U-87MG human glioma cells: Role of early growth response-1 expression. Cancer Res. 2008, 68, 1369–1377. [Google Scholar] [CrossRef] [Green Version]
- Wu, B.; Yao, H.; Wang, S.; Xu, R. DAPK1 modulates a curcumin-induced G2/M arrest and apoptosis by regulating STAT3, NF-κB, and caspase-3 activation. Biochem. Biophys. Res. Commun. 2013, 434, 75–80. [Google Scholar] [CrossRef] [PubMed]
- Jiang, S.; Fu, Y.; Zhang, X.; Yu, T.; Lu, B.; Du, J. Research Progress of Carrier-Free Antitumor Nanoparticles Based on Phytochemicals. Front. Bioeng. Biotechnol. 2021, 9, 799806. [Google Scholar] [CrossRef]
- Karatoprak, G.; Küpeli Akkol, E.; Genç, Y.; Bardakci, H.; Yücel, Ç.; Sobarzo-Sánchez, E. Combretastatins: An Overview of Structure, Probable Mechanisms of Action and Potential Applications. Molecules 2020, 25, 2560. [Google Scholar] [CrossRef] [PubMed]
- Lee, K.S. Happy Accidents: Serendipity in Modern Medical Breakthroughs. Korean J. Radiol. 2007, 8, 263. [Google Scholar] [CrossRef]
- Martino, E.; Casamassima, G.; Castiglione, S.; Cellupica, E.; Pantalone, S.; Papagni, F.; Rui, M.; Siciliano, A.M.; Collina, S. Vinca alkaloids and analogues as anti-cancer agents: Looking back, peering ahead. Bioorganic Med. Chem. Lett. 2018, 28, 2816–2826. [Google Scholar] [CrossRef]
- Farnsworth, N. Plants and Modern Medicine: Where Science and Folklore Meets; World Health Forum: Geneva, Switzerland, 1985; pp. 76–80. [Google Scholar]
- Arora, R.; Malhotra, P.; Mathur, A.K.; Mathur, A.; Govil, C.; Ahuja, P.S. Therapeutic Perspective. In Anticancer Alkaloids of Catharanthus Roseus: Transition from Traditional to Modern Medicine; Jaypee Brothers Medical Publishers Pvt. Ltd., N.D.: New Delhi, India, 2010; pp. 292–310. [Google Scholar]
- Foye, W.O. Cancer Chemotherapeutic Agents; Amer Chemical Society: New York, NY, USA, 1995. [Google Scholar]
- Goa, K.L.; Faulds, D.J.D. Vinorelbine. Drugs Aging 1994, 5, 200–234. [Google Scholar] [CrossRef]
- Bennouna, J.; Delord, J.P.; Campone, M.; Nguyen, L. Vinflunine: A new microtubule inhibitor agent. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2008, 14, 1625–1632. [Google Scholar] [CrossRef] [Green Version]
- Kotsakis, A.; Matikas, A.; Koinis, F.; Kentepozidis, N.; Varthalitis, II; Karavassilis, V.; Samantas, E.; Katsaounis, P.; Dermitzaki, E.K.; Hatzidaki, D.; et al. A multicentre phase II trial of cabazitaxel in patients with advanced non-small-cell lung cancer progressing after docetaxel-based chemotherapy. Br. J. Cancer 2016, 115, 784–788. [Google Scholar] [CrossRef] [Green Version]
- Oudard, S.; Fizazi, K.; Sengeløv, L.; Daugaard, G.; Saad, F.; Hansen, S.; Hjälm-Eriksson, M.; Jassem, J.; Thiery-Vuillemin, A.; Caffo, O.; et al. Cabazitaxel Versus Docetaxel As First-Line Therapy for Patients with Metastatic Castration-Resistant Prostate Cancer: A Randomized Phase III Trial-FIRSTANA. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2017, 35, 3189–3197. [Google Scholar] [CrossRef]
- Hertzberg, R.P.; Caranfa, M.J.; Hecht, S.M. On the mechanism of topoisomerase I inhibition by camptothecin: Evidence for binding to an enzyme-DNA complex. Biochemistry 1989, 28, 4629–4638. [Google Scholar] [CrossRef]
- Cao, B.; Chen, H.; Gao, Y.; Niu, C.; Zhang, Y.; Li, L. CIP-36, a novel topoisomerase II-targeting agent, induces the apoptosis of multidrug-resistant cancer cells in vitro. Int. J. Mol. Med. 2015, 35, 771–776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Itokawa, H.; Takeya, K.; Lee, K.-H. Anticancer compounds from higher plants. In Biomaterials from Aquatic and Terrestrial Organisms; CRC Press: Boca Raton, FL, USA, 2006; pp. 267–296. [Google Scholar]
- Skroza, N.; Bernardini, N.; Proietti, I.; Potenza, C. Clinical utility of ingenol mebutate in the management of actinic keratosis: Perspectives from clinical practice. Ther. Clin. Risk Manag. 2018, 14, 1879–1885. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tozer, G.M.; Kanthou, C.; Parkins, C.S.; Hill, S.A. The biology of the combretastatins as tumour vascular targeting agents. Int. J. Exp. Pathol. 2002, 83, 21–38. [Google Scholar] [CrossRef]
- Kumar, S.; Singh, B.; Singh, R. Catharanthus roseus (L.) G. Don: A review of its ethnobotany, phytochemistry, ethnopharmacology and toxicities. J. Ethnopharmacol. 2022, 284, 114647. [Google Scholar] [CrossRef] [PubMed]
- Pham, H.N.T.; Vuong, Q.V.; Bowyer, M.C.; Scarlett, C.J.J.T. Phytochemicals derived from Catharanthus roseus and their health benefits. Technologies 2020, 8, 80. [Google Scholar] [CrossRef]
- Taher, Z.M.; Agouillal, F.; Marof, A.Q.; Dailin, D.J.; Nurjayadi, M.; Razif, E.N.; Gomaa, S.E.; El Enshasy, H.A. Anticancer molecules from Catharanthus roseus. Indones. J. Pharm. 2019, 30, 147. [Google Scholar] [CrossRef]
- Allamsetty, J.; Pedada, S.P.; Pedada, N.; Kolli, D. A basic review on Vinca rosea. Int. J. Pharmacogn. Chem. 2020, I, 31–36. [Google Scholar]
- Almagro, L.; Fernández-Pérez, F.; Pedreño, M.A. Indole alkaloids from Catharanthus roseus: Bioproduction and their effect on human health. Molecules 2015, 20, 2973–3000. [Google Scholar] [CrossRef] [Green Version]
- Mishra, J.N.; Verma, N.K. A brief study on Catharanthus roseus: A review. Intern. J. Res. Pharm. Pharm. Sci. 2017, 2, 20–23. [Google Scholar]
- Bailon-Moscoso, N.; Cevallos-Solorzano, G.; Romero-Benavides, J.C.; Orellana, M.I. Natural Compounds as Modulators of Cell Cycle Arrest: Application for Anticancer Chemotherapies. Curr. Genom. 2017, 18, 106–131. [Google Scholar] [CrossRef] [Green Version]
- Tiong, S.H.; Looi, C.Y.; Arya, A.; Wong, W.F.; Hazni, H.; Mustafa, M.R.; Awang, K. Vindogentianine, a hypoglycemic alkaloid from Catharanthus roseus (L.) G. Don (Apocynaceae). Fitoterapia 2015, 102, 182–188. [Google Scholar] [CrossRef]
- Zhang, T.; Chen, W.; Jiang, X.; Liu, L.; Wei, K.; Du, H.; Wang, H.; Li, J. Anticancer effects and underlying mechanism of Colchicine on human gastric cancer cell lines in vitro and in vivo. Biosci. Rep. 2019, 39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pernot, B.; Gyan, E.; Maillot, F.; Hodges, P.; Ertault, M.; Ferreira-Maldent, N. Lymphomas diagnosed in an internal medicine department compared to lymphomas diagnosed in other departments: Clinical and outcome differences. Medicine 2018, 97, e13228. [Google Scholar] [CrossRef] [PubMed]
- Schiller, G.J.; Damon, L.E.; Coutre, S.E.; Hsu, P.; Bhat, G.; Douer, D. High-Dose Vincristine Sulfate Liposome Injection, for Advanced, Relapsed, or Refractory Philadelphia Chromosome-Negative Acute Lymphoblastic Leukemia in an Adolescent and Young Adult Subgroup of a Phase 2 Clinical Trial. J. Adolesc. Young Adult Oncol. 2018, 7, 546–552. [Google Scholar] [CrossRef] [PubMed]
- Maloney, S.M.; Hoover, C.A.; Morejon-Lasso, L.V.; Prosperi, J.R. Mechanisms of Taxane Resistance. Cancers 2020, 12, 3323. [Google Scholar] [CrossRef] [PubMed]
- Akhmanova, A.; Steinmetz, M.O. Tracking the ends: A dynamic protein network controls the fate of microtubule tips. Nat. Reviews. Mol. Cell Biol. 2008, 9, 309–322. [Google Scholar] [CrossRef]
- Maiato, H.; Sampaio, P.; Sunkel, C.E. Microtubule-associated proteins and their essential roles during mitosis. Int. Rev. Cytol. 2004, 241, 53–153. [Google Scholar]
- Vemu, A.; Atherton, J.; Spector, J.O.; Moores, C.A.; Roll-Mecak, A. Tubulin isoform composition tunes microtubule dynamics. Mol. Biol. Cell 2017, 28, 3564–3572. [Google Scholar] [CrossRef]
- Portran, D.; Schaedel, L.; Xu, Z.; Théry, M.; Nachury, M.V. Tubulin acetylation protects long-lived microtubules against mechanical ageing. Nat. Cell Biol. 2017, 19, 391–398. [Google Scholar] [CrossRef] [Green Version]
- Prassanawar, S.S.; Panda, D. Tubulin heterogeneity regulates functions and dynamics of microtubules and plays a role in the development of drug resistance in cancer. Biochem. J. 2019, 476, 1359–1376. [Google Scholar] [CrossRef]
- Wloga, D.; Joachimiak, E.; Fabczak, H. Tubulin Post-Translational Modifications and Microtubule Dynamics. Int. J. Mol. Sci. 2017, 18, 2207. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xu, Z.; Schaedel, L.; Portran, D.; Aguilar, A.; Gaillard, J.; Marinkovich, M.P.; Théry, M.; Nachury, M.V. Microtubules acquire resistance from mechanical breakage through intralumenal acetylation. Science 2017, 356, 328–332. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mialhe, A.; Lafanechère, L.; Treilleux, I.; Peloux, N.; Dumontet, C.; Brémond, A.; Panh, M.H.; Payan, R.; Wehland, J.; Margolis, R.L.; et al. Tubulin detyrosination is a frequent occurrence in breast cancers of poor prognosis. Cancer Res. 2001, 61, 5024–5027. [Google Scholar] [PubMed]
- Lu, C.; Zhang, J.; He, S.; Wan, C.; Shan, A.; Wang, Y.; Yu, L.; Liu, G.; Chen, K.; Shi, J.; et al. Increased α-tubulin1b expression indicates poor prognosis and resistance to chemotherapy in hepatocellular carcinoma. Dig. Dis. Sci. 2013, 58, 2713–2720. [Google Scholar] [CrossRef]
- Blenk, S.; Engelmann, J.C.; Pinkert, S.; Weniger, M.; Schultz, J.; Rosenwald, A.; Müller-Hermelink, H.K.; Müller, T.; Dandekar, T. Explorative data analysis of MCL reveals gene expression networks implicated in survival and prognosis supported by explorative CGH analysis. BMC Cancer 2008, 8, 106. [Google Scholar] [CrossRef] [Green Version]
- Bai, R.L.; Lin, C.M.; Nguyen, N.Y.; Liu, T.Y.; Hamel, E. Identification of the cysteine residue of beta-tubulin alkylated by the antimitotic agent 2,4-dichlorobenzyl thiocyanate, facilitated by separation of the protein subunits of tubulin by hydrophobic column chromatography. Biochemistry 1989, 28, 5606–5612. [Google Scholar] [CrossRef]
- Guo, J.; Walss-Bass, C.; Ludueña, R.F. The beta isotypes of tubulin in neuronal differentiation. Cytoskeleton 2010, 67, 431–441. [Google Scholar] [CrossRef] [Green Version]
- Joe, P.A.; Banerjee, A.; Ludueña, R.F. The roles of cys124 and ser239 in the functional properties of human betaIII tubulin. Cell Motil. Cytoskelet. 2008, 65, 476–486. [Google Scholar] [CrossRef]
- Lebok, P.; Öztürk, M.; Heilenkötter, U.; Jaenicke, F.; Müller, V.; Paluchowski, P.; Geist, S.; Wilke, C.; Burandt, E.; Lebeau, A.; et al. High levels of class III β-tubulin expression are associated with aggressive tumor features in breast cancer. Oncol. Lett. 2016, 11, 1987–1994. [Google Scholar] [CrossRef] [Green Version]
- Liou, G.Y.; Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 2010, 44, 479–496. [Google Scholar] [CrossRef] [Green Version]
- Parness, J.; Horwitz, S.B. Taxol binds to polymerized tubulin in vitro. J. Cell Biol. 1981, 91 (2 Pt 1), 479–487. [Google Scholar] [CrossRef] [PubMed]
- Gjyrezi, A.; Xie, F.; Voznesensky, O.; Khanna, P.; Calagua, C.; Bai, Y.; Kung, J.; Wu, J.; Corey, E.; Montgomery, B.; et al. Taxane resistance in prostate cancer is mediated by decreased drug-target engagement. J. Clin. Investig. 2020, 130, 3287–3298. [Google Scholar] [CrossRef]
- Brito, D.A.; Rieder, C.L. The ability to survive mitosis in the presence of microtubule poisons differs significantly between human nontransformed (RPE-1) and cancer (U2OS, HeLa) cells. Cell Motil. Cytoskelet. 2009, 66, 437–447. [Google Scholar] [CrossRef] [Green Version]
- Musacchio, A.; Salmon, E.D. The spindle-assembly checkpoint in space and time. Nat. Reviews. Mol. Cell Biol. 2007, 8, 379–393. [Google Scholar] [CrossRef]
- Gascoigne, K.E.; Taylor, S.S. Cancer cells display profound intra- and interline variation following prolonged exposure to antimitotic drugs. Cancer Cell 2008, 14, 111–122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Visconti, R.; Della Monica, R.; Grieco, D. Cell cycle checkpoint in cancer: A therapeutically targetable double-edged sword. J. Exp. Clin. Cancer Res. CR 2016, 35, 153. [Google Scholar] [CrossRef]
- Serpico, A.F.; Grieco, D. Recent advances in understanding the role of Cdk1 in the Spindle Assembly Checkpoint. F1000Research 2020, 9. [Google Scholar] [CrossRef] [Green Version]
- Kuh, H.J.; Jang, S.H.; Wientjes, M.G.; Au, J.L. Computational model of intracellular pharmacokinetics of paclitaxel. J. Pharmacol. Exp. Ther. 2000, 293, 761–770. [Google Scholar]
- Rowinsky, E.K.; Donehower, R.C. The clinical pharmacology of paclitaxel (Taxol). Semin. Oncol. 1993, 20 (Suppl. 3), 16–25. [Google Scholar]
- Thrower, D.; Jordan, M.A.; Wilson, L. Quantitation of cellular tubulin in microtubules and tubulin pools by a competitive ELISA. J. Immunol. Methods 1991, 136, 45–51. [Google Scholar] [CrossRef] [PubMed]
- Derry, W.B.; Wilson, L.; Jordan, M.A. Substoichiometric binding of taxol suppresses microtubule dynamics. Biochemistry 1995, 34, 2203–2211. [Google Scholar] [CrossRef]
- Jordan, M.A.; Wendell, K.; Gardiner, S.; Derry, W.B.; Copp, H.; Wilson, L. Mitotic block induced in HeLa cells by low concentrations of paclitaxel (Taxol) results in abnormal mitotic exit and apoptotic cell death. Cancer Res. 1996, 56, 816–825. [Google Scholar] [PubMed]
- Zasadil, L.M.; Andersen, K.A.; Yeum, D.; Rocque, G.B.; Wilke, L.G.; Tevaarwerk, A.J.; Raines, R.T.; Burkard, M.E.; Weaver, B.A. Cytotoxicity of paclitaxel in breast cancer is due to chromosome missegregation on multipolar spindles. Sci. Transl. Med. 2014, 6, 229ra43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shannon, K.B.; Canman, J.C.; Ben Moree, C.; Tirnauer, J.S.; Salmon, E.D. Taxol-stabilized microtubules can position the cytokinetic furrow in mammalian cells. Mol. Biol. Cell 2005, 16, 4423–4436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, Z.; Kenny, A.E.; Brito, D.A.; Rieder, C.L. Cells satisfy the mitotic checkpoint in Taxol, and do so faster in concentrations that stabilize syntelic attachments. J. Cell Biol. 2009, 186, 675–684. [Google Scholar] [CrossRef] [Green Version]
- Rose, W.C. Taxol-based combination chemotherapy and other in vivo preclinical antitumor studies. J. Natl. Cancer Inst. Monogr. 1993, 47–53. [Google Scholar]
- Milross, C.G.; Mason, K.A.; Hunter, N.R.; Chung, W.K.; Peters, L.J.; Milas, L. Relationship of mitotic arrest and apoptosis to antitumor effect of paclitaxel. J. Natl. Cancer Inst. 1996, 88, 1308–1314. [Google Scholar] [CrossRef]
- Desai, N.; Trieu, V.; Yao, Z.; Louie, L.; Ci, S.; Yang, A.; Tao, C.; De, T.; Beals, B.; Dykes, D.; et al. Increased antitumor activity, intratumor paclitaxel concentrations, and endothelial cell transport of cremophor-free, albumin-bound paclitaxel, ABI-007, compared with cremophor-based paclitaxel. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2006, 12, 1317–1324. [Google Scholar] [CrossRef] [Green Version]
- Pineda, J.J.; Miller, M.A.; Song, Y.; Kuhn, H.; Mikula, H.; Tallapragada, N.; Weissleder, R.; Mitchison, T.J. Site occupancy calibration of taxane pharmacology in live cells and tissues. Proc. Natl. Acad. Sci. USA 2018, 115, e11406–e11414. [Google Scholar] [CrossRef]
- Symmans, W.F.; Volm, M.D.; Shapiro, R.L.; Perkins, A.B.; Kim, A.Y.; Demaria, S.; Yee, H.T.; McMullen, H.; Oratz, R.; Klein, P.; et al. Paclitaxel-induced apoptosis and mitotic arrest assessed by serial fine-needle aspiration: Implications for early prediction of breast cancer response to neoadjuvant treatment. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2000, 6, 4610–4617. [Google Scholar]
- Hu, Y.; Manasrah, B.K.; McGregor, S.M.; Lera, R.F.; Norman, R.X.; Tucker, J.B.; Scribano, C.M.; Yan, R.E.; Humayun, M.; Wisinski, K.B.; et al. Paclitaxel Induces Micronucleation and Activates Pro-Inflammatory cGAS-STING Signaling in Triple-Negative Breast Cancer. Mol. Cancer Ther. 2021, 20, 2553–2567. [Google Scholar] [CrossRef] [PubMed]
- Mohiuddin, M.; Kasahara, K. The Mechanisms of the Growth Inhibitory Effects of Paclitaxel on Gefitinib-resistant Non-small Cell Lung Cancer Cells. Cancer Genom. Proteom. 2021, 18, 661–673. [Google Scholar] [CrossRef] [PubMed]
- Chan, J.; Adderley, H.; Alameddine, M.; Armstrong, A.; Arundell, D.; Fox, R.; Harries, M.; Lim, J.; Salih, Z.; Tetlow, C.; et al. Permanent hair loss associated with taxane chemotherapy use in breast cancer: A retrospective survey at two tertiary UK cancer centres. Eur. J. Cancer Care 2021, 30, e13395. [Google Scholar] [CrossRef] [PubMed]
- Liu, Y.; Hiramoto, B.; Kwok, J.; Ibrahim, A.; Tatishchev, S.; Kang, I.; Modi, R. Taxane-Induced Upper Gastrointestinal Bleeding. Case Rep. Oncol. 2021, 14, 1373–1379. [Google Scholar] [CrossRef] [PubMed]
- Thomas, J.; Warrier, A.; Kachare, N. Docetaxel-Induced Myositis. J. Clin. Rheumatol. Pract. Rep. Rheum. Musculoskelet. Dis. 2020, 26, e62–e63. [Google Scholar] [CrossRef]
- Rochigneux, P.; Schleinitz, N.; Ebbo, M.; Aymonier, M.; Pourroy, B.; Boissier, R.; Salas, S.; Deville, J.L. Acute myositis: An unusual and severe side effect of docetaxel: A case report and literature review. Anti-Cancer Drugs 2018, 29, 477–481. [Google Scholar] [CrossRef]
- Pobel, C.; Auclin, E.; Teyssonneau, D.; Laguerre, B.; Cancel, M.; Boughalem, E.; Noel, J.; Brachet, P.E.; Maillet, D.; Barthelemy, P.; et al. Cabazitaxel multiple rechallenges in metastatic castration-resistant prostate cancer. Cancer Med. 2021, 10, 6304–6309. [Google Scholar] [CrossRef]
- Liu, S.; Kwon, M.; Mannino, M.; Yang, N.; Renda, F.; Khodjakov, A.; Pellman, D. Nuclear envelope assembly defects link mitotic errors to chromothripsis. Nature 2018, 561, 551–555. [Google Scholar] [CrossRef]
- Mitchison, T.J.; Pineda, J.; Shi, J.; Florian, S. Is inflammatory micronucleation the key to a successful anti-mitotic cancer drug? Open Biol. 2017, 7, 170182. [Google Scholar] [CrossRef] [Green Version]
- Serpico, A.F.; Visconti, R.; Grieco, D. Exploiting immune-dependent effects of microtubule-targeting agents to improve efficacy and tolerability of cancer treatment. Cell Death Dis. 2020, 11, 361. [Google Scholar] [CrossRef]
- Bates, D.; Eastman, A. Microtubule destabilising agents: Far more than just antimitotic anticancer drugs. Br. J. Clin. Pharmacol. 2017, 83, 255–268. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fong, A.; Durkin, A.; Lee, H. The Potential of Combining Tubulin-Targeting Anticancer Therapeutics and Immune Therapy. Int. J. Mol. Sci. 2019, 20, 586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kampan, N.C.; Madondo, M.T.; McNally, O.M.; Quinn, M.; Plebanski, M. Paclitaxel and Its Evolving Role in the Management of Ovarian Cancer. BioMed Res. Int. 2015, 2015, 413076. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, T.H.; Wang, H.S.; Soong, Y.K. Paclitaxel-induced cell death: Where the cell cycle and apoptosis come together. Cancer 2000, 88, 2619–2628. [Google Scholar] [CrossRef]
- Huisman, C.; Ferreira, C.G.; Bröker, L.E.; Rodriguez, J.A.; Smit, E.F.; Postmus, P.E.; Kruyt, F.A.; Giaccone, G. Paclitaxel triggers cell death primarily via caspase-independent routes in the non-small cell lung cancer cell line NCI-H460. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2002, 8, 596–606. [Google Scholar]
- Hüsemann, L.C.; Reese, A.; Radine, C.; Piekorz, R.P.; Budach, W.; Sohn, D.; Jänicke, R.U. The microtubule targeting agents eribulin and paclitaxel activate similar signaling pathways and induce cell death predominantly in a caspase-independent manner. Cell Cycle (Georget. Tex.) 2020, 19, 464–478. [Google Scholar] [CrossRef]
- Huang, S.; Zhang, Y.; Wang, L.; Liu, W.; Xiao, L.; Lin, Q.; Gong, T.; Sun, X.; He, Q.; Zhang, Z.; et al. Improved melanoma suppression with target-delivered TRAIL and Paclitaxel by a multifunctional nanocarrier. J. Control. Release Off. J. Control. Release Soc. 2020, 325, 10–24. [Google Scholar] [CrossRef]
- Bocci, G.; Di Paolo, A.; Danesi, R. The pharmacological bases of the antiangiogenic activity of paclitaxel. Angiogenesis 2013, 16, 481–492. [Google Scholar] [CrossRef] [Green Version]
- Muta, M.; Yanagawa, T.; Sai, Y.; Saji, S.; Suzuki, E.; Aruga, T.; Kuroi, K.; Matsumoto, G.; Toi, M.; Nakashima, E. Effect of low-dose Paclitaxel and docetaxel on endothelial progenitor cells. Oncology 2009, 77, 182–191. [Google Scholar] [CrossRef]
- Cui, Q.; Wang, J.Q.; Assaraf, Y.G.; Ren, L.; Gupta, P.; Wei, L.; Ashby, C.R., Jr.; Yang, D.H.; Chen, Z.S. Modulating ROS to overcome multidrug resistance in cancer. Drug Resist. Updates Rev. Comment. Antimicrob. Anticancer Chemother. 2018, 41, 1–25. [Google Scholar] [CrossRef]
- Nita, M.; Grzybowski, A. The Role of the Reactive Oxygen Species and Oxidative Stress in the Pathomechanism of the Age-Related Ocular Diseases and Other Pathologies of the Anterior and Posterior Eye Segments in Adults. Oxidative Med. Cell. Longev. 2016, 2016, 3164734. [Google Scholar] [CrossRef] [PubMed]
- Su, W.P.; Lo, Y.C.; Yan, J.J.; Liao, I.C.; Tsai, P.J.; Wang, H.C.; Yeh, H.H.; Lin, C.C.; Chen, H.H.; Lai, W.W.; et al. Mitochondrial uncoupling protein 2 regulates the effects of paclitaxel on Stat3 activation and cellular survival in lung cancer cells. Carcinogenesis 2012, 33, 2065–2075. [Google Scholar] [CrossRef] [PubMed]
- Huang, J.; Peng, W.; Zheng, Y.; Hao, H.; Li, S.; Yao, Y.; Ding, Y.; Zhang, J.; Lyu, J.; Zeng, Q. Upregulation of UCP2 Expression Protects against LPS-Induced Oxidative Stress and Apoptosis in Cardiomyocytes. Oxidative Med. Cell. Longev. 2019, 2019, 2758262. [Google Scholar] [CrossRef] [PubMed]
- Pandya, A.D.; Jäger, E.; Bagheri Fam, S.; Höcherl, A.; Jäger, A.; Sincari, V.; Nyström, B.; Štěpánek, P.; Skotland, T.; Sandvig, K.; et al. Paclitaxel-loaded biodegradable ROS-sensitive nanoparticles for cancer therapy. Int. J. Nanomed. 2019, 14, 6269–6285. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chiu, Y.H.; Hsu, S.H.; Hsu, H.W.; Huang, K.C.; Liu, W.; Wu, C.Y.; Huang, W.P.; Chen, J.Y.; Chen, B.H.; Chiu, C.C. Human non-small cell lung cancer cells can be sensitized to camptothecin by modulating autophagy. Int. J. Oncol. 2018, 53, 1967–1979. [Google Scholar] [CrossRef] [Green Version]
- Lichota, A.; Gwozdzinski, K. Anticancer Activity of Natural Compounds from Plant and Marine Environment. Int. J. Mol. Sci. 2018, 19, 3533. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chou, H.L.; Fong, Y.; Wei, C.K.; Tsai, E.M.; Chen, J.Y.; Chang, W.T.; Wu, C.Y.; Huang, H.W.; Chiu, C.C. A Quinone-Containing Compound Enhances Camptothecin-Induced Apoptosis of Lung Cancer through Modulating Endogenous ROS and ERK Signaling. Arch. Immunol. Et Ther. Exp. 2017, 65, 241–252. [Google Scholar] [CrossRef]
- Liu, Y.Q.; Li, W.Q.; Morris-Natschke, S.L.; Qian, K.; Yang, L.; Zhu, G.X.; Wu, X.B.; Chen, A.L.; Zhang, S.Y.; Nan, X.; et al. Perspectives on biologically active camptothecin derivatives. Med. Res. Rev. 2015, 35, 753–789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hevener, K.; Verstak, T.A.; Lutat, K.E.; Riggsbee, D.L.; Mooney, J.W. Recent developments in topoisomerase-targeted cancer chemotherapy. Acta Pharm. Sinica. B 2018, 8, 844–861. [Google Scholar] [CrossRef]
- Wen, T.; Song, L.; Hua, S. Perspectives and controversies regarding the use of natural products for the treatment of lung cancer. Cancer Med. 2021, 10, 2396–2422. [Google Scholar] [CrossRef]
- Wilkes, G.M. Targeted Therapy: Attacking Cancer with Molecular and Immunological Targeted Agents. Asia-Pac. J. Oncol. Nurs. 2018, 5, 137–155. [Google Scholar] [CrossRef]
- Hsiang, Y.H.; Liu, L.F. Identification of mammalian DNA topoisomerase I as an intracellular target of the anticancer drug camptothecin. Cancer Res. 1988, 48, 1722–1726. [Google Scholar] [PubMed]
- Li, Q.Y.; Zu, Y.G.; Shi, R.Z.; Yao, L.P. Review camptothecin: Current perspectives. Curr. Med. Chem. 2006, 13, 2021–2039. [Google Scholar] [CrossRef] [PubMed]
- Soepenberg, O.; Sparreboom, A.; Verweij, J. Clinical studies of camptothecin and derivatives. Alkaloids. Chem. Biol. 2003, 60, 1–50. [Google Scholar] [PubMed]
- Ghanbari-Movahed, M.; Kaceli, T.; Mondal, A.; Farzaei, M.H.; Bishayee, A. Recent Advances in Improved Anticancer Efficacies of Camptothecin Nano-Formulations: A Systematic Review. Biomedicines 2021, 9, 480. [Google Scholar] [CrossRef]
- Beretta, G.L.; Gatti, L.; Perego, P.; Zaffaroni, N. Camptothecin resistance in cancer: Insights into the molecular mechanisms of a DNA-damaging drug. Curr. Med. Chem. 2013, 20, 1541–1565. [Google Scholar] [CrossRef]
- Pommier, Y. Topoisomerase I inhibitors: Camptothecins and beyond. Nat. Reviews. Cancer 2006, 6, 789–802. [Google Scholar] [CrossRef] [Green Version]
- Lorusso, D.; Pietragalla, A.; Mainenti, S.; Masciullo, V.; Di Vagno, G.; Scambia, G. Review role of topotecan in gynaecological cancers: Current indications and perspectives. Crit. Rev. Oncol./Hematol. 2010, 74, 163–174. [Google Scholar] [CrossRef]
- Venditto, V.J.; Simanek, E.E. Cancer therapies utilizing the camptothecins: A review of the in vivo literature. Mol. Pharm. 2010, 7, 307–349. [Google Scholar] [CrossRef] [Green Version]
- Liu, L.F.; Desai, S.D.; LI, T.K.; Mao, Y.; Sun, M.E.I.; SIM, S.P. Mechanism of action of camptothecin. Ann. N. Y. Acad. Sci. 2000, 922, 1–10. [Google Scholar] [CrossRef]
- Martino, E.; Della Volpe, S.; Terribile, E.; Benetti, E.; Sakaj, M.; Centamore, A.; Sala, A.; Collina, S. The long story of camptothecin: From traditional medicine to drugs. Bioorganic Med. Chem. Lett. 2017, 27, 701–707. [Google Scholar] [CrossRef] [PubMed]
- Zou, J.; Li, S.; Chen, Z.; Lu, Z.; Gao, J.; Zou, J.; Lin, X.; Li, Y.; Zhang, C.; Shen, L. A novel oral camptothecin analog, gimatecan, exhibits superior antitumor efficacy than irinotecan toward esophageal squamous cell carcinoma in vitro and in vivo. Cell Death Dis. 2018, 9, 1–10. [Google Scholar] [CrossRef] [PubMed]
- Ferguson, L.R.; Baguley, B.C. Mutagenicity of anticancer drugs that inhibit topoisomerase enzymes. Mutat. Res./Fundam. Mol. Mech. Mutagenesis 1996, 355, 91–101. [Google Scholar] [CrossRef] [PubMed]
- Nitiss, J.L.; Soans, E.; Rogojina, A.; Seth, A.; Mishina, M. Topoisomerase assays. Curr. Protoc. Pharmacol. 2012, 57, 3.3.1–3.3.27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stahl, M.; Kasimir-Bauer, S.; Harstrick, A. Down-regulation of topoisomerase II by camptothecin does not prevent additive activity of the topoisomerase II inhibitor etoposide in vitro. Anti-Cancer Drugs 1997, 8, 671–676. [Google Scholar] [CrossRef]
- Li, F.; Jiang, T.; Li, Q.; Ling, X. Camptothecin (CPT) and its derivatives are known to target topoisomerase I (Top1) as their mechanism of action: Did we miss something in CPT analogue molecular targets for treating human disease such as cancer? Am. J. Cancer Res. 2017, 7, 2350–2394. [Google Scholar]
- Paz-Ares, L.; Dvorkin, M.; Chen, Y.; Reinmuth, N.; Hotta, K.; Trukhin, D.; Statsenko, G.; Hochmair, M.J.; Özgüroğlu, M.; Ji, J.H.; et al. Durvalumab plus platinum-etoposide versus platinum-etoposide in first-line treatment of extensive-stage small-cell lung cancer (CASPIAN): A randomised, controlled, open-label, phase 3 trial. Lancet 2019, 394, 1929–1939. [Google Scholar] [CrossRef]
- Bugg, B.Y.; Danks, M.K.; Beck, W.T.; Suttle, D.P. Expression of a mutant DNA topoisomerase II in CCRF-CEM human leukemic cells selected for resistance to teniposide. Proc. Natl. Acad. Sci. USA 1991, 88, 7654–7658. [Google Scholar] [CrossRef] [Green Version]
- Vassetzky, Y.S.; Alghisi, G.C.; Roberts, E.; Gasser, S.M. Ectopic expression of inactive forms of yeast DNA topoisomerase II confers resistance to the anti-tumour drug, etoposide. Br. J. Cancer 1996, 73, 1201–1209. [Google Scholar] [CrossRef]
- Podpeskar, A.; Crazzolara, R.; Kropshofer, G.; Hetzer, B.; Meister, B.; Müller, T.; Salvador, C. Omega-3 Fatty Acids and Their Role in Pediatric Cancer. Nutrients 2021, 13, 1800. [Google Scholar] [CrossRef]
- Zi, C.T.; Yang, L.; Gao, W.; Li, Y.; Zhou, J.; Ding, Z.T.; Hu, J.M.; Jiang, Z.H. Click Glycosylation for the Synthesis of 1,2,3-Triazole-Linked Picropodophyllotoxin Glycoconjugates and Their Anticancer Activity. ChemistrySelect 2017, 2, 5038–5044. [Google Scholar] [CrossRef]
- Linder, S.; Shoshan, M.C.; Gupta, R.S. Picropodophyllotoxin or podophyllotoxin does not induce cell death via insulin-like growth factor-I receptor. Cancer Res. 2007, 67, 2899–2900. [Google Scholar] [CrossRef] [PubMed]
- Rigiracciolo, D.C.; Nohata, N.; Lappano, R.; Cirillo, F.; Talia, M.; Scordamaglia, D.; Gutkind, J.S.; Maggiolini, M. Correction: Rigiracciolo, D.C.; et al. IGF-1/IGF-1R/FAK/YAP Transduction Signaling Prompts Growth Effects in Triple-Negative Breast Cancer (TNBC) Cells. Cells 2020, 9, 1010. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Huang, F.; Shi, Q.; Li, Y.; Xu, L.; Xu, C.; Chen, F.; Wang, H.; Liao, H.; Chang, Z.; Liu, F.; et al. HER2/EGFR-AKT Signaling Switches TGFβ from Inhibiting Cell Proliferation to Promoting Cell Migration in Breast Cancer. Cancer Res. 2018, 78, 6073–6085. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yin, S.; Girnita, A.; Strömberg, T.; Khan, Z.; Andersson, S.; Zheng, H.; Ericsson, C.; Axelson, M.; Nistér, M.; Larsson, O.; et al. Targeting the insulin-like growth factor-1 receptor by picropodophyllin as a treatment option for glioblastoma. Neuro-Oncol. 2010, 12, 19–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carrasco-Garcia, E.; Martinez-Lacaci, I.; Mayor-López, L.; Tristante, E.; Carballo-Santana, M.; García-Morales, P.; Ventero Martin, M.P.; Fuentes-Baile, M.; Rodriguez-Lescure, Á.; Saceda, M. PDGFR and IGF-1R Inhibitors Induce a G2/M Arrest and Subsequent Cell Death in Human Glioblastoma Cell Lines. Cells 2018, 7, 131. [Google Scholar] [CrossRef] [Green Version]
- Shah, Z.; Gohar, U.F.; Jamshed, I.; Mushtaq, A.; Mukhtar, H.; Zia-Ui-Haq, M.; Toma, S.I.; Manea, R.; Moga, M.; Popovici, B. Podophyllotoxin: History, Recent Advances and Future Prospects. Biomolecules 2021, 11, 603. [Google Scholar] [CrossRef]
- Mazumder, K.; Aktar, A.; Roy, P.; Biswas, B.; Hossain, M.E.; Sarkar, K.K.; Bachar, S.C.; Ahmed, F.; Monjur-Al-Hossain, A.S.M.; Fukase, K. A Review on Mechanistic Insight of Plant Derived Anticancer Bioactive Phytocompounds and Their Structure Activity Relationship. Molecules 2022, 27, 3036. [Google Scholar] [CrossRef]
- Yang, Z.; Zhou, Z.; Luo, X.; Luo, X.; Luo, H.; Luo, L.; Yang, W. Design and Synthesis of Novel Podophyllotoxins Hybrids and the Effects of Different Functional Groups on Cytotoxicity. Molecules 2021, 27, 220. [Google Scholar] [CrossRef]
- Menu, E.; Jernberg-Wiklund, H.; Stromberg, T.; De Raeve, H.; Girnita, L.; Larsson, O.; Axelson, M.; Asosingh, K.; Nilsson, K.; Van Camp, B.; et al. Inhibiting the IGF-1 receptor tyrosine kinase with the cyclolignan PPP: An in vitro and in vivo study in the 5T33MM mouse model. Blood 2006, 107, 655–660. [Google Scholar] [CrossRef] [Green Version]
- Uzma, F.; Mohan, C.D.; Hashem, A.; Konappa, N.M.; Rangappa, S.; Kamath, P.V.; Singh, B.P.; Mudili, V.; Gupta, V.K.; Siddaiah, C.N.; et al. Endophytic Fungi-Alternative Sources of Cytotoxic Compounds: A Review. Front. Pharmacol. 2018, 9, 309. [Google Scholar] [CrossRef] [PubMed]
- Kumar, S.; Aharwal, R.P.; Jain, R.; Sandhu, S.S. Bioactive molecules of endophytic fungi and their potential in anticancer drug development. Curr. Pharmacol. Rep. 2021, 7, 27–41. [Google Scholar] [CrossRef]
- Pérard-Viret, J.; Quteishat, L.; Alsalim, R.; Royer, J.; Dumas, F. Cephalotaxus alkaloids. Alkaloids Chem. Biol. 2017, 78, 205–352. [Google Scholar] [PubMed]
- Gilles, A.; Frechin, L.; Natchiar, K.; Biondani, G.; Loeffelholz, O.V.; Holvec, S.; Malaval, J.L.; Winum, J.Y.; Klaholz, B.P.; Peyron, J.F. Targeting the Human 80S Ribosome in Cancer: From Structure to Function and Drug Design for Innovative Adjuvant Therapeutic Strategies. Cells 2020, 9, 629. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Short, N.J.; Jabbour, E.; Naqvi, K.; Patel, A.; Ning, J.; Sasaki, K.; Nogueras-Gonzalez, G.M.; Bose, P.; Kornblau, S.M.; Takahashi, K.; et al. A phase II study of omacetaxine mepesuccinate for patients with higher-risk myelodysplastic syndrome and chronic myelomonocytic leukemia after failure of hypomethylating agents. Am. J. Hematol. 2019, 94, 74–79. [Google Scholar] [CrossRef] [Green Version]
- McLoughlin, E.C.; O’Boyle, N.M. Colchicine-Binding Site Inhibitors from Chemistry to Clinic: A Review. Pharmaceuticals 2020, 13, 8. [Google Scholar] [CrossRef] [Green Version]
- Bhurta, D.; Bharate, S.B. Styryl Group, a Friend or Foe in Medicinal Chemistry. ChemMedChem 2022, 17, e202100706. [Google Scholar] [CrossRef] [PubMed]
- Zweifel, M.; Jayson, G.C.; Reed, N.S.; Osborne, R.; Hassan, B.; Ledermann, J.; Shreeves, G.; Poupard, L.; Lu, S.P.; Balkissoon, J.; et al. Phase II trial of combretastatin A4 phosphate, carboplatin, and paclitaxel in patients with platinum-resistant ovarian cancer. Ann. Oncol. Off. J. Eur. Soc. Med. Oncol. 2011, 22, 2036–2041. [Google Scholar] [CrossRef]
- Okada, F. Inflammation-related carcinogenesis: Current findings in epidemiological trends, causes and mechanisms. Yonago Acta Med. 2014, 57, 65. [Google Scholar]
- Balamurugan, K. HIF-1 at the crossroads of hypoxia, inflammation, and cancer. Int. J. Cancer 2016, 138, 1058–1066. [Google Scholar] [CrossRef] [Green Version]
- Pan, M.-G.; Xiong, Y.; Chen, F. NFAT gene family in inflammation and cancer. Curr. Mol. Med. 2013, 13, 543–554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, P.; Montes de Oca, M.K.; Alkeswani, A.R.; McClees, S.F.; Das, T.; Elmets, C.A.; Afaq, F. Tea polyphenols for the prevention of UVB-induced skin cancer. Photodermatol. Photoimmunol. Photomed. 2018, 34, 50–59. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maru, G.B.; Gandhi, K.; Ramchandani, A.; Kumar, G. The role of inflammation in skin cancer. Adv. Exp. Med. Biol. 2014, 816, 437–469. [Google Scholar] [PubMed]
- Park, K.K.; Chun, K.S.; Lee, J.M.; Lee, S.S.; Surh, Y.J. Inhibitory effects of [6]-gingerol, a major pungent principle of ginger, on phorbol ester-induced inflammation, epidermal ornithine decarboxylase activity and skin tumor promotion in ICR mice. Cancer Lett. 1998, 129, 139–144. [Google Scholar] [CrossRef]
- Surh, Y.J. Anti-tumor promoting potential of selected spice ingredients with antioxidative and anti-inflammatory activities: A short review. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2002, 40, 1091–1097. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Hsieh, M.C.; Lo, C.Y.; Liu, C.B.; Sang, S.; Ho, C.T.; Pan, M.H. 6-Shogaol is more effective than 6-gingerol and curcumin in inhibiting 12-O-tetradecanoylphorbol 13-acetate-induced tumor promotion in mice. Mol. Nutr. Food Res. 2010, 54, 1296–1306. [Google Scholar] [CrossRef] [PubMed]
- Lu, Y.P.; Lou, Y.R.; Xie, J.G.; Peng, Q.Y.; Liao, J.; Yang, C.S.; Huang, M.T.; Conney, A.H. Topical applications of caffeine or (-)-epigallocatechin gallate (EGCG) inhibit carcinogenesis and selectively increase apoptosis in UVB-induced skin tumors in mice. Proc. Natl. Acad. Sci. USA 2002, 99, 12455–12460. [Google Scholar] [CrossRef] [Green Version]
- Nomura, M.; Ma, W.Y.; Huang, C.; Yang, C.S.; Bowden, G.T.; Miyamoto, K.; Dong, Z. Inhibition of ultraviolet B-induced AP-1 activation by theaflavins from black tea. Mol. Carcinog. 2000, 28, 148–155. [Google Scholar] [CrossRef]
- Katiyar, S.K.; Perez, A.; Mukhtar, H. Green tea polyphenol treatment to human skin prevents formation of ultraviolet light B-induced pyrimidine dimers in DNA. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2000, 6, 3864–3869. [Google Scholar]
- Gajewski, T.F.; Schreiber, H.; Fu, Y.X. Innate and adaptive immune cells in the tumor microenvironment. Nat. Immunol. 2013, 14, 1014–1022. [Google Scholar] [CrossRef] [Green Version]
- Corrales, L.; Matson, V.; Flood, B.; Spranger, S.; Gajewski, T.F. Innate immune signaling and regulation in cancer immunotherapy. Cell Res. 2017, 27, 96–108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
- Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef] [PubMed]
- Taniguchi, K.; Karin, M. NF-κB, inflammation, immunity and cancer: Coming of age. Nat. Reviews. Immunol. 2018, 18, 309–324. [Google Scholar] [CrossRef]
- Dawson, M.A.; Kouzarides, T. Cancer epigenetics: From mechanism to therapy. Cell 2012, 150, 12–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gupta, S.C.; Kunnumakkara, A.B.; Aggarwal, S.; Aggarwal, B.B. Inflammation, a Double-Edge Sword for Cancer and Other Age-Related Diseases. Front. Immunol. 2018, 9, 2160. [Google Scholar] [CrossRef] [Green Version]
- Murata, M. Inflammation and cancer. Environ. Health Prev. Med. 2018, 23, 50. [Google Scholar] [CrossRef] [Green Version]
- Zhang, D.; Tang, Z.; Huang, H.; Zhou, G.; Cui, C.; Weng, Y.; Liu, W.; Kim, S.; Lee, S.; Perez-Neut, M.; et al. Metabolic regulation of gene expression by histone lactylation. Nature 2019, 574, 575–580. [Google Scholar] [CrossRef]
- Ell, B.; Kang, Y. Transcriptional control of cancer metastasis. Trends Cell Biol. 2013, 23, 603–611. [Google Scholar] [CrossRef] [Green Version]
- Easwaran, H.; Tsai, H.C.; Baylin, S.B. Cancer epigenetics: Tumor heterogeneity, plasticity of stem-like states, and drug resistance. Mol. Cell 2014, 54, 716–727. [Google Scholar] [CrossRef] [Green Version]
- Iliopoulos, D.; Hirsch, H.A.; Struhl, K. An epigenetic switch involving NF-kappaB, Lin28, Let-7 MicroRNA, and IL6 links inflammation to cell transformation. Cell 2009, 139, 693–706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chiarugi, P.; Cirri, P. Metabolic exchanges within tumor microenvironment. Cancer Lett. 2016, 380, 272–280. [Google Scholar] [CrossRef] [PubMed]
- Fischer, K.; Hoffmann, P.; Voelkl, S.; Meidenbauer, N.; Ammer, J.; Edinger, M.; Gottfried, E.; Schwarz, S.; Rothe, G.; Hoves, S.; et al. Inhibitory effect of tumor cell-derived lactic acid on human T cells. Blood 2007, 109, 3812–3819. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gottfried, E.; Kunz-Schughart, L.A.; Ebner, S.; Mueller-Klieser, W.; Hoves, S.; Andreesen, R.; Mackensen, A.; Kreutz, M. Tumor-derived lactic acid modulates dendritic cell activation and antigen expression. Blood 2006, 107, 2013–2021. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bader, J.E.; Voss, K.; Rathmell, J.C. Targeting Metabolism to Improve the Tumor Microenvironment for Cancer Immunotherapy. Mol. Cell 2020, 78, 1019–1033. [Google Scholar] [CrossRef]
- Mu, X.; Shi, W.; Xu, Y.; Xu, C.; Zhao, T.; Geng, B.; Yang, J.; Pan, J.; Hu, S.; Zhang, C.; et al. Tumor-derived lactate induces M2 macrophage polarization via the activation of the ERK/STAT3 signaling pathway in breast cancer. Cell Cycle (Georget. Tex.) 2018, 17, 428–438. [Google Scholar] [CrossRef] [Green Version]
- Colegio, O.R.; Chu, N.Q.; Szabo, A.L.; Chu, T.; Rhebergen, A.M.; Jairam, V.; Cyrus, N.; Brokowski, C.E.; Eisenbarth, S.C.; Phillips, G.M.; et al. Functional polarization of tumour-associated macrophages by tumour-derived lactic acid. Nature 2014, 513, 559–563. [Google Scholar] [CrossRef] [Green Version]
- Hao, H.; Liu, M.; Wu, P.; Cai, L.; Tang, K.; Yi, P.; Li, Y.; Chen, Y.; Ye, D. Lipoxin A4 and its analog suppress hepatocellular carcinoma via remodeling tumor microenvironment. Cancer Lett. 2011, 309, 85–94. [Google Scholar] [CrossRef]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Jess, T.; Simonsen, J.; Jørgensen, K.T.; Pedersen, B.V.; Nielsen, N.M.; Frisch, M. Decreasing risk of colorectal cancer in patients with inflammatory bowel disease over 30 years. Gastroenterology 2012, 143, 375–381.e1; quiz e13–e14. [Google Scholar] [CrossRef]
- Keller, D.S.; Windsor, A.; Cohen, R.; Chand, M. Colorectal cancer in inflammatory bowel disease: Review of the evidence. Tech. Coloproctology 2019, 23, 3–13. [Google Scholar] [CrossRef]
- Pekow, J.; Hernandez, K.; Meckel, K.; Deng, Z.; Haider, H.I.; Khalil, A.; Zhang, C.; Talisila, N.; Siva, S.; Jasmine, F.; et al. IBD-associated Colon Cancers Differ in DNA Methylation and Gene Expression Profiles Compared with Sporadic Colon Cancers. J. Crohn’s Colitis 2019, 13, 884–893. [Google Scholar] [CrossRef]
- Bian, S.; Hou, Y.; Zhou, X.; Li, X.; Yong, J.; Wang, Y.; Wang, W.; Yan, J.; Hu, B.; Guo, H.; et al. Single-cell multiomics sequencing and analyses of human colorectal cancer. Science 2018, 362, 1060–1063. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gottlieb, M.; Nakitende, D. Comparison of Tamsulosin, Nifedipine, and Placebo for Ureteric Colic. Cjem 2017, 19, 156–158. [Google Scholar] [CrossRef]
- Loor, A.; Dumitraşcu, D.L. Helicobacter pylori Infection, Gastric Cancer and Gastropanel. Rom. J. Intern. Med. Rev. Roum. Med. Interne 2016, 54, 151–156. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bain, C.C.; Mowat, A.M. Macrophages in intestinal homeostasis and inflammation. Immunol. Rev. 2014, 260, 102–117. [Google Scholar] [CrossRef] [Green Version]
- Yang, H.; Wang, W.; Romano, K.A.; Gu, M.; Sanidad, K.Z.; Kim, D.; Yang, J.; Schmidt, B.; Panigrahy, D.; Pei, R.; et al. A common antimicrobial additive increases colonic inflammation and colitis-associated colon tumorigenesis in mice. Sci. Transl. Med. 2018, 10, eaan4116. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, Y.; Tian, Z. HBV-Induced Immune Imbalance in the Development of HCC. Front. Immunol. 2019, 10, 2048. [Google Scholar] [CrossRef] [Green Version]
- Grivennikov, S.I.; Greten, F.R.; Karin, M. Immunity, inflammation, and cancer. Cell 2010, 140, 883–899. [Google Scholar] [CrossRef] [Green Version]
- Schiffman, M.; Castle, P.E.; Jeronimo, J.; Rodriguez, A.C.; Wacholder, S. Human papillomavirus and cervical cancer. Lancet 2007, 370, 890–907. [Google Scholar] [CrossRef]
- Li, R.; Zhou, R.; Wang, H.; Li, W.; Pan, M.; Yao, X.; Zhan, W.; Yang, S.; Xu, L.; Ding, Y.; et al. Gut microbiota-stimulated cathepsin K secretion mediates TLR4-dependent M2 macrophage polarization and promotes tumor metastasis in colorectal cancer. Cell Death Differ. 2019, 26, 2447–2463. [Google Scholar] [CrossRef] [PubMed]
- Kalafati, L.; Kourtzelis, I.; Schulte-Schrepping, J.; Li, X.; Hatzioannou, A.; Grinenko, T.; Hagag, E.; Sinha, A.; Has, C.; Dietz, S.; et al. Innate Immune Training of Granulopoiesis Promotes Anti-tumor Activity. Cell 2020, 183, 771–785.e12. [Google Scholar] [CrossRef] [PubMed]
- Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly, Y.M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573. [Google Scholar] [CrossRef] [Green Version]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
- Arpaia, N.; Campbell, C.; Fan, X.; Dikiy, S.; van der Veeken, J.; deRoos, P.; Liu, H.; Cross, J.R.; Pfeffer, K.; Coffer, P.J.; et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature 2013, 504, 451–455. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [Green Version]
- Ma, C.; Han, M.; Heinrich, B.; Fu, Q.; Zhang, Q.; Sandhu, M.; Agdashian, D.; Terabe, M.; Berzofsky, J.A.; Fako, V.; et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 2018, 360, eaan5931. [Google Scholar] [CrossRef] [Green Version]
- Elkrief, A.; Derosa, L.; Zitvogel, L.; Kroemer, G.; Routy, B. The intimate relationship between gut microbiota and cancer immunotherapy. Gut Microbes 2019, 10, 424–428. [Google Scholar] [CrossRef] [Green Version]
- Jin, S.; Chen, H.; Li, Y.; Zhong, H.; Sun, W.; Wang, J.; Zhang, T.; Ma, J.; Yan, S.; Zhang, J.; et al. Maresin 1 improves the Treg/Th17 imbalance in rheumatoid arthritis through miR-21. Ann. Rheum. Dis. 2018, 77, 1644–1652. [Google Scholar] [CrossRef]
- Tuomisto, A.E.; Mäkinen, M.J.; Väyrynen, J.P. Systemic inflammation in colorectal cancer: Underlying factors, effects, and prognostic significance. World J. Gastroenterol. 2019, 25, 4383–4404. [Google Scholar] [CrossRef]
- Han, M.; Song, Y.; Zhang, X. Quercetin Suppresses the Migration and Invasion in Human Colon Cancer Caco-2 Cells Through Regulating Toll-like Receptor 4/Nuclear Factor-kappa B Pathway. Pharmacogn. Mag. 2016, 12 (Suppl. 2), S237–S244. [Google Scholar]
- Zhao, H.; Wu, L.; Yan, G.; Chen, Y.; Zhou, M.; Wu, Y.; Li, Y. Inflammation and tumor progression: Signaling pathways and targeted intervention. Signal Transduct. Target. Ther. 2021, 6, 263. [Google Scholar] [CrossRef] [PubMed]
- Marjaneh, R.M.; Rahmani, F.; Hassanian, S.M.; Rezaei, N.; Hashemzehi, M.; Bahrami, A.; Ariakia, F.; Fiuji, H.; Sahebkar, A.; Avan, A.; et al. Phytosomal curcumin inhibits tumor growth in colitis-associated colorectal cancer. J. Cell. Physiol. 2018, 233, 6785–6798. [Google Scholar] [CrossRef] [PubMed]
- Kaur, H.; Kaur, G. A Critical Appraisal of Solubility Enhancement Techniques of Polyphenols. J. Pharm. 2014, 2014, 180845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Carroll, R.E.; Benya, R.V.; Turgeon, D.K.; Vareed, S.; Neuman, M.; Rodriguez, L.; Kakarala, M.; Carpenter, P.M.; McLaren, C.; Meyskens, F.L., Jr.; et al. Phase IIa clinical trial of curcumin for the prevention of colorectal neoplasia. Cancer Prev. Res. (Phila. Pa.) 2011, 4, 354–364. [Google Scholar] [CrossRef]
- Citronberg, J.; Bostick, R.; Ahearn, T.; Turgeon, D.K.; Ruffin, M.T.; Djuric, Z.; Sen, A.; Brenner, D.E.; Zick, S.M. Effects of ginger supplementation on cell-cycle biomarkers in the normal-appearing colonic mucosa of patients at increased risk for colorectal cancer: Results from a pilot, randomized, and controlled trial. Cancer Prev. Res. (Phila. Pa.) 2013, 6, 271–281. [Google Scholar] [CrossRef] [Green Version]
- Bhowmik, A.C.; Oksala, N.K.; Auriko, J.P.; Paavonen, T.; Mustonen, H.; Paimela, H. Structural signaling regulates inflammation-induced enhanced restitution and increased Mib-1 and Bax-indexes after superficial injury in isolated guinea pig gastric mucosa. Dig. Dis. Sci. 2004, 49, 1526–1530. [Google Scholar] [CrossRef]
- Warren, C.A.; Paulhill, K.J.; Davidson, L.A.; Lupton, J.R.; Taddeo, S.S.; Hong, M.Y.; Carroll, R.J.; Chapkin, R.S.; Turner, N.D. Quercetin may suppress rat aberrant crypt foci formation by suppressing inflammatory mediators that influence proliferation and apoptosis. J. Nutr. 2009, 139, 101–105. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.A.; Zhang, S.; Yin, Q.; Zhang, J. Quercetin induces human colon cancer cells apoptosis by inhibiting the nuclear factor-kappa B Pathway. Pharmacogn. Mag. 2015, 11, 404–409. [Google Scholar] [CrossRef] [Green Version]
- Jain, M.A.; Sapra, A. Prostate cancer screening. In StatPearls [Internet]; StatPearls Publishing: Tampa, FL, USA, 2021. [Google Scholar]
- Ide, H.; Tokiwa, S.; Sakamaki, K.; Nishio, K.; Isotani, S.; Muto, S.; Hama, T.; Masuda, H.; Horie, S. Combined inhibitory effects of soy isoflavones and curcumin on the production of prostate-specific antigen. Prostate 2010, 70, 1127–1133. [Google Scholar] [CrossRef]
- Ayob, A.Z.; Ramasamy, T.S. Cancer stem cells as key drivers of tumour progression. J. Biomed. Sci. 2018, 25, 20. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deng, Y.I.; Verron, E.; Rohanizadeh, R. Molecular Mechanisms of Anti-metastatic Activity of Curcumin. Anticancer Res. 2016, 36, 5639–5647. [Google Scholar] [CrossRef] [Green Version]
- Thomas, R.; Williams, M.; Sharma, H.; Chaudry, A.; Bellamy, P. A double-blind, placebo-controlled randomised trial evaluating the effect of a polyphenol-rich whole food supplement on PSA progression in men with prostate cancer—The U.K. NCRN Pomi-T study. Prostate Cancer Prostatic Dis. 2014, 17, 180–186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hejazi, J.; Rastmanesh, R.; Taleban, F.A.; Molana, S.H.; Hejazi, E.; Ehtejab, G.; Hara, N. Effect of Curcumin Supplementation During Radiotherapy on Oxidative Status of Patients with Prostate Cancer: A Double Blinded, Randomized, Placebo-Controlled Study. Nutr. Cancer 2016, 68, 77–85. [Google Scholar] [CrossRef] [PubMed]
Classes and Subclasses of Phytochemicals | Therapeutic Pharmacology | Distinct Cancers | Molecular Objectives | References |
---|---|---|---|---|
Vinca alkaloids | ||||
Vinblastine | Preventing microtubules from turning anti-mitotic by adhering to tubulin | Non-small-cell lung carcinoma (NSCLC), breast and lung cancers, leukemia, Hodgkin’s and non-Hodgkin’s lymphomas, testicular carcinoma, melanoma, head and neck cancer, uterine melanomas, Kaposi’s sarcoma, and transitional cell carcinoma | Microtubules and tubulin | [84,85,86,87,88,89,90] |
Vincristine | Mitosis inhibition through binding to tubulin dimer | |||
Vindesine | Anti-mitotic | |||
Vinflunine | Anti-mitotic | |||
Vinorelbine | Prevents cancer cells from beginning mitosis by blocking the transition from metaphase to anaphase; increases the rate of apoptosis | |||
Taxanes | ||||
Docetaxel | Microtubule inhibition causing cell cycle arrest and abnormal mitosis | Prostate, head and neck, breast cancers, gastric adenocarcinoma, and NSCLC | Microtubules and tubulin | [91,92] |
Cabazitaxel | ||||
Paclitaxel | ||||
Camptothecin | ||||
Irinotecan | Protects against deadly double-stranded DNA breaks by stabilizing topoisomerase I-DNA complex and halting religation of single-strand breaks | Small cell lung cancer (SCLC), ovarian, cervical, and colorectal cancers | Topoisomerase-I | [93] |
Topotecan | ||||
Podophyllotoxin | ||||
Etoposide | Forms a complex with topoisomerase II and DNA blocking the enzyme’s ability to synthesize new DNA | NSCLC, cervical, nasopharyngeal, colon, breast, testicular, osteosarcoma, and prostate cancer | Topoisomerase-II | [94] |
Teniposide | ||||
Other phytochemicals | ||||
Homoharringtonine | Interferes with chain elongation and inhibits protein synthesis by binding to the large ribosomal subunit | Chronic myeloid leukemia | Ribosomal protein | [95] |
Ingenol mebutate | Immediate cell death and inflammatory response activation | Actinic keratosis | Protein kinase C (PKC) | [96] |
Combretastatin A4 | Tumor endothelial cells are disrupted when tubulin polymerization is inhibited, resulting in loose tumor blood vessels | Anaplastic thyroid cancers | Tubulin | [97] |
Taxanes | Cancer Type | Days/Time | Concentration | Effects | References |
---|---|---|---|---|---|
Paclitaxel | triple-negative breast cancer (TNBC) | weekly | 80 (mg/m2) | ↑ stromal tumor-infiltrating lymphocytes and micronucleation | [145] |
gefitinib-resistant non-small-cell lung cancer (NSCLC) cells (PC9-MET) | 72 h | 50–100 nM | ↓ Sustainability and ↑ morphological signs of apoptosis. | [146] | |
Docetaxel and Paclitaxel | breast Cancer | 9 weeks and 12 weeks | 75–100 mg/m2 and 80 mg/m2 | Hair loss (alopecia) | [147] |
Docetaxel | breast invasive ductal carcinoma | 3 weeks for 6 cycles | 75–100 mg/m2 | Upper Gastrointestinal Bleeding | [148] |
Docetaxel | prostate adenocarcinoma | every 3 weeks | 75–100 mg/m2 | bilateral pain of the two inferior limbs and bilateral motor deficit | [149,150] |
Cabazitaxel | metastatic castration-resistant prostate cancer (mCRPC) | q3week and q2week | 25 mg/m2 and 16 mg/m2 | No cumulative grade ≥3 neuropathy or nail disorder and one case of febrile neutropenia. | [151] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Ahmed, M.B.; Islam, S.U.; Alghamdi, A.A.A.; Kamran, M.; Ahsan, H.; Lee, Y.S. Phytochemicals as Chemo-Preventive Agents and Signaling Molecule Modulators: Current Role in Cancer Therapeutics and Inflammation. Int. J. Mol. Sci. 2022, 23, 15765. https://doi.org/10.3390/ijms232415765
Ahmed MB, Islam SU, Alghamdi AAA, Kamran M, Ahsan H, Lee YS. Phytochemicals as Chemo-Preventive Agents and Signaling Molecule Modulators: Current Role in Cancer Therapeutics and Inflammation. International Journal of Molecular Sciences. 2022; 23(24):15765. https://doi.org/10.3390/ijms232415765
Chicago/Turabian StyleAhmed, Muhammad Bilal, Salman Ul Islam, Abdullah A. A. Alghamdi, Muhammad Kamran, Haseeb Ahsan, and Young Sup Lee. 2022. "Phytochemicals as Chemo-Preventive Agents and Signaling Molecule Modulators: Current Role in Cancer Therapeutics and Inflammation" International Journal of Molecular Sciences 23, no. 24: 15765. https://doi.org/10.3390/ijms232415765
APA StyleAhmed, M. B., Islam, S. U., Alghamdi, A. A. A., Kamran, M., Ahsan, H., & Lee, Y. S. (2022). Phytochemicals as Chemo-Preventive Agents and Signaling Molecule Modulators: Current Role in Cancer Therapeutics and Inflammation. International Journal of Molecular Sciences, 23(24), 15765. https://doi.org/10.3390/ijms232415765