Chemotherapy-Mediated Neuronal Aberration
Abstract
:1. Introduction
2. Mechanism of Chemotherapy-Induced Cancer Cell Death and Inflammation
3. Role of Commonly Used Chemotherapeutic Drugs in Neuronal Function
3.1. Doxorubicin
3.2. Effect of Doxorubicin-Mediated Oxidative Stress and Inflammation in Cognitive Impairment
3.3. Role of Cisplatin in Chemobrain
3.4. Molecular Mechanism of Cisplatin-Mediated Inflammation
3.5. Role of Paclitaxel in Chemobrain
3.6. Paclitaxel and Inflammation in Chemobrain
3.7. 5-Fluorouracil in Chemobrain
3.8. Inflammatory Role of 5-FU in Chemobrain
4. Combinatorial Chemotherapy in Chemobrain
5. Possible Therapeutic Intervention for Chemotherapy-Mediated Chemobrain
5.1. Phytochemical as Chemobrain Therapeutic Intervention
5.2. Anti-Inflammatory Drug in Chemobrain Therapy
6. Conclusions and Future Prospective
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Chiang, A.C.A.; Huo, X.; Kavelaars, A.; Heijnen, C.J. Chemotherapy accelerates age-related development of tauopathy and results in loss of synaptic integrity and cognitive impairment. Brain Behav. Immun. 2019, 79, 319–325. [Google Scholar] [CrossRef] [PubMed]
- Chiang, A.C.A.; Seua, A.V.; Singhmar, P.; Arroyo, L.D.; Mahalingam, R.; Hu, J.; Kavelaars, A.; Heijnen, C.J. Bexarotene normalizes chemotherapy-induced myelin decompaction and reverses cognitive and sensorimotor deficits in mice. Acta Neuropathol. Commun. 2020, 8, 193. [Google Scholar] [CrossRef]
- Ongnok, B.; Chattipakorn, N.; Chattipakorn, S.C. Doxorubicin and cisplatin induced cognitive impairment: The possible mechanisms and interventions. Exp. Neurol. 2020, 324, 113118. [Google Scholar] [CrossRef]
- Kovalchuk, A.; Kolb, B. Chemo brain: From discerning mechanisms to lifting the brain fog-An aging connection. Cell Cycle 2017, 16, 1345–1349. [Google Scholar] [CrossRef]
- John, J.; Kinra, M.; Mudgal, J.; Viswanatha, G.L.; Nandakumar, K. Animal models of chemotherapy-induced cognitive decline in preclinical drug development. Psychopharmacology 2021, 238, 3025–3053. [Google Scholar] [CrossRef]
- Was, H.; Borkowska, A.; Bagues, A.; Tu, L.; Liu, J.Y.H.; Lu, Z.; Rudd, J.A.; Nurgali, K.; Abalo, R. Mechanisms of Chemotherapy-Induced Neurotoxicity. Front. Pharmacol. 2022, 13, 750507. [Google Scholar] [CrossRef]
- Janelsins, M.C.; Kesler, S.R.; Ahles, T.A.; Morrow, G.R. Prevalence, mechanisms, and management of cancer-related cognitive impairment. Int. Rev. Psychiatry 2014, 26, 102–113. [Google Scholar] [CrossRef] [PubMed]
- Henderson, F.M.; Cross, A.J.; Baraniak, A.R. ‘A new normal with chemobrain’: Experiences of the impact of chemotherapy-related cognitive deficits in long-term breast cancer survivors. Health Psychol. Open 2019, 6, 2055102919832234. [Google Scholar] [CrossRef] [PubMed]
- Magge, R.S.; DeAngelis, L.M. The double-edged sword: Neurotoxicity of chemotherapy. Blood Rev. 2015, 29, 93–100. [Google Scholar] [CrossRef]
- Soffietti, R.; Trevisan, E.; Rudà, R. Chapter 80—Neurologic complications of chemotherapy and other newer and experimental approaches. In Handbook of Clinical Neurology; Biller, J., Ferro, J.M., Eds.; Elsevier: Amsterdam, The Netherlands, 2014; Volume 121, pp. 1199–1218. [Google Scholar]
- Demaria, M.; O’Leary, M.N.; Chang, J.; Shao, L.; Liu, S.; Alimirah, F.; Koenig, K.; Le, C.; Mitin, N.; Deal, A.M.; et al. Cellular Senescence Promotes Adverse Effects of Chemotherapy and Cancer Relapse. Cancer Discov. 2017, 7, 165–176. [Google Scholar] [CrossRef]
- Amidi, A.; Hosseini, S.M.H.; Leemans, A.; Kesler, S.R.; Agerbæk, M.; Wu, L.M.; Zachariae, R. Changes in Brain Structural Networks and Cognitive Functions in Testicular Cancer Patients Receiving Cisplatin-Based Chemotherapy. JNCI J. Natl. Cancer Inst. 2017, 109, djx085. [Google Scholar] [CrossRef] [PubMed]
- Bromis, K.; Gkiatis, K.; Karanasiou, I.; Matsopoulos, G.; Karavasilis, E.; Papathanasiou, M.; Efstathopoulos, E.; Kelekis, N.; Kouloulias, V. Altered Brain Functional Connectivity in Small-Cell Lung Cancer Patients after Chemotherapy Treatment: A Resting-State fMRI Study. Comput. Math. Methods Med. 2017, 2017, 1403940. [Google Scholar] [CrossRef] [PubMed]
- Simó, M.; Root, J.C.; Vaquero, L.; Ripollés, P.; Jové, J.; Ahles, T.; Navarro, A.; Cardenal, F.; Bruna, J.; Rodríguez-Fornells, A. Cognitive and Brain Structural Changes in a Lung Cancer Population. J. Thorac. Oncol. 2015, 10, 38–45. [Google Scholar] [CrossRef] [PubMed]
- Ren, X.; Clair, D.K.S.; Butterfield, D.A. Dysregulation of cytokine mediated chemotherapy induced cognitive impairment. Pharmacol. Res. 2017, 117, 267–273. [Google Scholar] [CrossRef]
- Lomeli, N.; Lepe, J.; Gupta, K.; Bota, D.A. Cognitive complications of cancer and cancer-related treatments—Novel paradigms. Neurosci. Lett. 2021, 749, 135720. [Google Scholar] [CrossRef]
- Koppelmans, V.; Breteler, M.M.; Boogerd, W.; Seynaeve, C.; Gundy, C.; Schagen, S.B. Neuropsychological performance in survivors of breast cancer more than 20 years after adjuvant chemotherapy. J. Clin. Oncol. 2012, 30, 1080–1086. [Google Scholar] [CrossRef]
- van Schaik, T.A.; Chen, K.S.; Shah, K. Therapy-Induced Tumor Cell Death: Friend or Foe of Immunotherapy? Front. Oncol. 2021, 11, 678562. [Google Scholar] [CrossRef]
- Pan, S.T.; Li, Z.L.; He, Z.X.; Qiu, J.X.; Zhou, S.F. Molecular mechanisms for tumour resistance to chemotherapy. Clin. Exp. Pharmacol. Physiol. 2016, 43, 723–737. [Google Scholar] [CrossRef]
- Seitz, S.J.; Schleithoff, E.S.; Koch, A.; Schuster, A.; Teufel, A.; Staib, F.; Stremmel, W.; Melino, G.; Krammer, P.H.; Schilling, T.; et al. Chemotherapy-induced apoptosis in hepatocellular carcinoma involves the p53 family and is mediated via the extrinsic and the intrinsic pathway. Int. J. Cancer 2010, 126, 2049–2066. [Google Scholar] [CrossRef]
- Stevenson, L.; Allen, W.L.; Proutski, I.; Stewart, G.; Johnston, L.; McCloskey, K.; Wilson, P.M.; Longley, D.B.; Johnston, P.G. Calbindin 2 (CALB2) regulates 5-fluorouracil sensitivity in colorectal cancer by modulating the intrinsic apoptotic pathway. PLoS ONE 2011, 6, e20276. [Google Scholar] [CrossRef]
- Kim, J.S.; Lee, J.H.; Jeong, W.W.; Choi, D.H.; Cha, H.J.; Kim, D.H.; Kwon, J.K.; Park, S.E.; Park, J.H.; Cho, H.R.; et al. Reactive oxygen species-dependent EndoG release mediates cisplatin-induced caspase-independent apoptosis in human head and neck squamous carcinoma cells. Int. J. Cancer 2008, 122, 672–680. [Google Scholar] [CrossRef] [PubMed]
- Onoda, T.; Ono, T.; Dhar, D.K.; Yamanoi, A.; Nagasue, N. Tetracycline analogues (doxycycline and COL-3) induce caspase-dependent and -independent apoptosis in human colon cancer cells. Int. J. Cancer 2006, 118, 1309–1315. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.F.; Chen, C.Y.; Chung, S.F.; Chiou, Y.H.; Lo, H.R. Involvement of oxidative stress and caspase activation in paclitaxel-induced apoptosis of primary effusion lymphoma cells. Cancer Chemother. Pharmacol. 2004, 54, 322–330. [Google Scholar] [CrossRef] [PubMed]
- Bertheloot, D.; Latz, E.; Franklin, B.S. Necroptosis, pyroptosis and apoptosis: An intricate game of cell death. Cell. Mol. Immunol. 2021, 18, 1106–1121. [Google Scholar] [CrossRef] [PubMed]
- Brown, M.F.; Leibowitz, B.J.; Chen, D.; He, K.; Zou, F.; Sobol, R.W.; Beer-Stolz, D.; Zhang, L.; Yu, J. Loss of caspase-3 sensitizes colon cancer cells to genotoxic stress via RIP1-dependent necrosis. Cell Death Dis. 2015, 6, e1729. [Google Scholar] [CrossRef]
- Xu, Y.; Lin, Z.; Zhao, N.; Zhou, L.; Liu, F.; Cichacz, Z.; Zhang, L.; Zhan, Q.; Zhao, X. Receptor interactive protein kinase 3 promotes Cisplatin-triggered necrosis in apoptosis-resistant esophageal squamous cell carcinoma cells. PLoS ONE 2014, 9, e100127. [Google Scholar] [CrossRef]
- Perillo, B.; Di Donato, M.; Pezone, A.; Di Zazzo, E.; Giovannelli, P.; Galasso, G.; Castoria, G.; Migliaccio, A. ROS in cancer therapy: The bright side of the moon. Exp. Mol. Med. 2020, 52, 192–203. [Google Scholar] [CrossRef]
- Ren, X.; Boriero, D.; Chaiswing, L.; Bondada, S.; St Clair, D.K.; Butterfield, D.A. Plausible biochemical mechanisms of chemotherapy-induced cognitive impairment (“chemobrain”), a condition that significantly impairs the quality of life of many cancer survivors. Biochim. Biophys. Acta Mol. Basis Dis. 2019, 1865, 1088–1097. [Google Scholar] [CrossRef]
- Pun, P.B.; Lu, J.; Moochhala, S. Involvement of ROS in BBB dysfunction. Free Radic. Res. 2009, 43, 348–364. [Google Scholar] [CrossRef]
- Wardill, H.R.; Mander, K.A.; Van Sebille, Y.Z.; Gibson, R.J.; Logan, R.M.; Bowen, J.M.; Sonis, S.T. Cytokine-mediated blood brain barrier disruption as a conduit for cancer/chemotherapy-associated neurotoxicity and cognitive dysfunction. Int. J. Cancer 2016, 139, 2635–2645. [Google Scholar] [CrossRef]
- Vaure, C.; Liu, Y. A comparative review of toll-like receptor 4 expression and functionality in different animal species. Front. Immunol. 2014, 5, 316. [Google Scholar] [CrossRef] [PubMed]
- Aluise, C.D.; Miriyala, S.; Noel, T.; Sultana, R.; Jungsuwadee, P.; Taylor, T.J.; Cai, J.; Pierce, W.M.; Vore, M.; Moscow, J.A.; et al. 2-Mercaptoethane sulfonate prevents doxorubicin-induced plasma protein oxidation and TNF-α release: Implications for the reactive oxygen species-mediated mechanisms of chemobrain. Free Radic. Biol. Med. 2011, 50, 1630–1638. [Google Scholar] [CrossRef] [PubMed]
- Du, J.; Zhang, A.; Li, J.; Liu, X.; Wu, S.; Wang, B.; Wang, Y.; Jia, H. Doxorubicin-Induced Cognitive Impairment: The Mechanistic Insights. Front. Oncol. 2021, 11, 673340. [Google Scholar] [CrossRef] [PubMed]
- Blaser, H.; Dostert, C.; Mak, T.W.; Brenner, D. TNF and ROS Crosstalk in Inflammation. Trends Cell Biol. 2016, 26, 249–261. [Google Scholar] [CrossRef]
- Zhao, N.; Woodle, M.C.; Mixson, A.J. Advances in delivery systems for doxorubicin. J. Nanomed. Nanotechnol. 2018, 9, 519. [Google Scholar] [CrossRef]
- Dempke, W.C.M.; Zielinski, R.; Winkler, C.; Silberman, S.; Reuther, S.; Priebe, W. Anthracycline-induced cardiotoxicity—Are we about to clear this hurdle? Eur. J. Cancer 2023, 185, 94–104. [Google Scholar] [CrossRef]
- Andryszak, P.; Wiłkość, M.; Żurawski, B.; Izdebski, P. Verbal memory in breast cancer patients treated with chemotherapy with doxorubicin and cyclophosphamide. Eur. J. Cancer Care 2018, 27, e12749. [Google Scholar] [CrossRef]
- Crowgey, T.; Peters, K.B.; Hornsby, W.E.; Lane, A.; McSherry, F.; Herndon, J.E., 2nd; West, M.J.; Williams, C.L.; Jones, L.W. Relationship between exercise behavior, cardiorespiratory fitness, and cognitive function in early breast cancer patients treated with doxorubicin-containing chemotherapy: A pilot study. Appl. Physiol. Nutr. Metab. 2014, 39, 724–729. [Google Scholar] [CrossRef]
- Ramalingayya, G.V.; Cheruku, S.P.; Nayak, P.G.; Kishore, A.; Shenoy, R.; Rao, C.M.; Krishnadas, N. Rutin protects against neuronal damage in vitro and ameliorates doxorubicin-induced memory deficits in vivo in Wistar rats. Drug Des. Devel. Ther. 2017, 11, 1011–1026. [Google Scholar] [CrossRef]
- Manchon, J.F.; Dabaghian, Y.; Uzor, N.E.; Kesler, S.R.; Wefel, J.S.; Tsvetkov, A.S. Levetiracetam mitigates doxorubicin-induced DNA and synaptic damage in neurons. Sci. Rep. 2016, 6, 25705. [Google Scholar] [CrossRef]
- Wetzel, M.; Rosenberg, G.A.; Cunningham, L.A. Tissue inhibitor of metalloproteinases-3 and matrix metalloproteinase-3 regulate neuronal sensitivity to doxorubicin-induced apoptosis. Eur. J. Neurosci. 2003, 18, 1050–1060. [Google Scholar] [CrossRef] [PubMed]
- Shokoohinia, Y.; Hosseinzadeh, L.; Moieni-Arya, M.; Mostafaie, A.; Mohammadi-Motlagh, H.R. Osthole attenuates doxorubicin-induced apoptosis in PC12 cells through inhibition of mitochondrial dysfunction and ROS production. Biomed. Res. Int. 2014, 2014, 156848. [Google Scholar] [CrossRef] [PubMed]
- Keeney, J.T.R.; Ren, X.; Warrier, G.; Noel, T.; Powell, D.K.; Brelsfoard, J.M.; Sultana, R.; Saatman, K.E.; Clair, D.K.S.; Butterfield, D.A. Doxorubicin-induced elevated oxidative stress and neurochemical alterations in brain and cognitive decline: Protection by MESNA and insights into mechanisms of chemotherapy-induced cognitive impairment (“chemobrain”). Oncotarget 2018, 9, 30324–30339. [Google Scholar] [CrossRef]
- Finn, N.A.; Findley, H.W.; Kemp, M.L. A switching mechanism in doxorubicin bioactivation can be exploited to control doxorubicin toxicity. PLoS Comput. Biol. 2011, 7, e1002151. [Google Scholar] [CrossRef] [PubMed]
- Malik, A.; Sultana, M.; Qazi, A.; Qazi, M.H.; Parveen, G.; Waquar, S.; Ashraf, A.B.; Rasool, M. Role of Natural Radiosensitizers and Cancer Cell Radioresistance: An Update. Anal. Cell. Pathol. 2016, 2016, 6146595. [Google Scholar] [CrossRef]
- Zhu, H.; Sarkar, S.; Scott, L.; Danelisen, I.; Trush, M.A.; Jia, Z.; Li, Y.R. Doxorubicin Redox Biology: Redox Cycling, Topoisomerase Inhibition, and Oxidative Stress. React. Oxyg. Species 2016, 1, 189–198. [Google Scholar] [CrossRef]
- Renu, K.; Valsala Gopalakrishnan, A. Deciphering the molecular mechanism during doxorubicin-mediated oxidative stress, apoptosis through Nrf2 and PGC-1α in a rat testicular milieu. Reprod. Biol. 2019, 19, 22–37. [Google Scholar] [CrossRef]
- Lingappan, K. NF-κB in Oxidative Stress. Curr. Opin. Toxicol. 2018, 7, 81–86. [Google Scholar] [CrossRef]
- Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef]
- Morgan, M.J.; Liu, Z.-g. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef]
- Tangpong, J.; Cole, M.P.; Sultana, R.; Joshi, G.; Estus, S.; Vore, M.; St Clair, W.; Ratanachaiyavong, S.; St Clair, D.K.; Butterfield, D.A. Adriamycin-induced, TNF-alpha-mediated central nervous system toxicity. Neurobiol. Dis. 2006, 23, 127–139. [Google Scholar] [CrossRef] [PubMed]
- Tangpong, J.; Miriyala, S.; Noel, T.; Sinthupibulyakit, C.; Jungsuwadee, P.; St Clair, D.K. Doxorubicin-induced central nervous system toxicity and protection by xanthone derivative of Garcinia mangostana. Neuroscience 2011, 175, 292–299. [Google Scholar] [CrossRef] [PubMed]
- Hyka, N.; Dayer, J.M.; Modoux, C.; Kohno, T.; Edwards, C.K., 3rd; Roux-Lombard, P.; Burger, D. Apolipoprotein A-I inhibits the production of interleukin-1beta and tumor necrosis factor-alpha by blocking contact-mediated activation of monocytes by T lymphocytes. Blood 2001, 97, 2381–2389. [Google Scholar] [CrossRef] [PubMed]
- Osburg, B.; Peiser, C.; Dömling, D.; Schomburg, L.; Ko, Y.T.; Voigt, K.; Bickel, U. Effect of endotoxin on expression of TNF receptors and transport of TNF-alpha at the blood-brain barrier of the rat. Am. J. Physiol. Endocrinol. Metab. 2002, 283, E899–E908. [Google Scholar] [CrossRef] [PubMed]
- Pan, W.; Kastin, A.J. TNFalpha transport across the blood-brain barrier is abolished in receptor knockout mice. Exp. Neurol. 2002, 174, 193–200. [Google Scholar] [CrossRef]
- Wu, Y.Q.; Dang, R.L.; Tang, M.M.; Cai, H.L.; Li, H.D.; Liao, D.H.; He, X.; Cao, L.J.; Xue, Y.; Jiang, P. Long Chain Omega-3 Polyunsaturated Fatty Acid Supplementation Alleviates Doxorubicin-Induced Depressive-Like Behaviors and Neurotoxicity in Rats: Involvement of Oxidative Stress and Neuroinflammation. Nutrients 2016, 8, 243. [Google Scholar] [CrossRef]
- Mander, P.K.; Jekabsone, A.; Brown, G.C. Microglia proliferation is regulated by hydrogen peroxide from NADPH oxidase. J. Immunol. 2006, 176, 1046–1052. [Google Scholar] [CrossRef]
- Butterfield, D.A. The 2013 SFRBM discovery award: Selected discoveries from the butterfield laboratory of oxidative stress and its sequela in brain in cognitive disorders exemplified by Alzheimer disease and chemotherapy induced cognitive impairment. Free Radic. Biol. Med. 2014, 74, 157–174. [Google Scholar] [CrossRef]
- Sheng, W.S.; Hu, S.; Feng, A.; Rock, R.B. Reactive oxygen species from human astrocytes induced functional impairment and oxidative damage. Neurochem. Res. 2013, 38, 2148–2159. [Google Scholar] [CrossRef]
- Mohamed, R.H.; Karam, R.A.; Amer, M.G. Epicatechin attenuates doxorubicin-induced brain toxicity: Critical role of TNF-α, iNOS and NF-κB. Brain Res. Bull. 2011, 86, 22–28. [Google Scholar] [CrossRef]
- Holley, A.K.; Bakthavatchalu, V.; Velez-Roman, J.M.; St Clair, D.K. Manganese superoxide dismutase: Guardian of the powerhouse. Int. J. Mol. Sci. 2011, 12, 7114–7162. [Google Scholar] [CrossRef] [PubMed]
- Tangpong, J.; Cole, M.P.; Sultana, R.; Estus, S.; Vore, M.; St Clair, W.; Ratanachaiyavong, S.; St Clair, D.K.; Butterfield, D.A. Adriamycin-mediated nitration of manganese superoxide dismutase in the central nervous system: Insight into the mechanism of chemobrain. J. Neurochem. 2007, 100, 191–201. [Google Scholar] [CrossRef] [PubMed]
- Andres, A.L.; Gong, X.; Di, K.; Bota, D.A. Low-doses of cisplatin injure hippocampal synapses: A mechanism for ‘chemo’ brain? Exp. Neurol. 2014, 255, 137–144. [Google Scholar] [CrossRef] [PubMed]
- Horky, L.L.; Gerbaudo, V.H.; Zaitsev, A.; Plesniak, W.; Hainer, J.; Govindarajulu, U.; Kikinis, R.; Dietrich, J. Systemic chemotherapy decreases brain glucose metabolism. Ann. Clin. Transl. Neurol. 2014, 1, 788–798. [Google Scholar] [CrossRef]
- Michalak, S.; Rybacka-Mossakowska, J.; Ambrosius, W.; Gazdulska, J.; Gołda-Gocka, I.; Kozubski, W.; Ramlau, R. The Markers of Glutamate Metabolism in Peripheral Blood Mononuclear Cells and Neurological Complications in Lung Cancer Patients. Dis. Markers 2016, 2016, 2895972. [Google Scholar] [CrossRef]
- Chiu, G.S.; Boukelmoune, N.; Chiang, A.C.A.; Peng, B.; Rao, V.; Kingsley, C.; Liu, H.L.; Kavelaars, A.; Kesler, S.R.; Heijnen, C.J. Nasal administration of mesenchymal stem cells restores cisplatin-induced cognitive impairment and brain damage in mice. Oncotarget 2018, 9, 35581–35597. [Google Scholar] [CrossRef]
- Zhou, W.; Kavelaars, A.; Heijnen, C.J. Metformin Prevents Cisplatin-Induced Cognitive Impairment and Brain Damage in Mice. PLoS ONE 2016, 11, e0151890. [Google Scholar] [CrossRef]
- Manohar, S.; Jamesdaniel, S.; Salvi, R. Cisplatin inhibits hippocampal cell proliferation and alters the expression of apoptotic genes. Neurotox. Res. 2014, 25, 369–380. [Google Scholar] [CrossRef]
- Chtourou, Y.; Gargouri, B.; Kebieche, M.; Fetoui, H. Naringin Abrogates Cisplatin-Induced Cognitive Deficits and Cholinergic Dysfunction Through the Down-Regulation of AChE Expression and iNOS Signaling Pathways in Hippocampus of Aged Rats. J. Mol. Neurosci. 2015, 56, 349–362. [Google Scholar] [CrossRef]
- Jangra, A.; Kwatra, M.; Singh, T.; Pant, R.; Kushwah, P.; Ahmed, S.; Dwivedi, D.; Saroha, B.; Lahkar, M. Edaravone alleviates cisplatin-induced neurobehavioral deficits via modulation of oxidative stress and inflammatory mediators in the rat hippocampus. Eur. J. Pharmacol. 2016, 791, 51–61. [Google Scholar] [CrossRef]
- Chiu, G.S.; Maj, M.A.; Rizvi, S.; Dantzer, R.; Vichaya, E.G.; Laumet, G.; Kavelaars, A.; Heijnen, C.J. Pifithrin-μ Prevents Cisplatin-Induced Chemobrain by Preserving Neuronal Mitochondrial Function. Cancer Res. 2017, 77, 742–752. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Wei, Y.; Pu, X.; Zhao, L. Preclinical studies for the combination of paclitaxel and curcumin in cancer therapy (Review). Oncol. Rep. 2017, 37, 3159–3166. [Google Scholar] [CrossRef]
- Klein, I.; Lehmann, H.C. Pathomechanisms of Paclitaxel-Induced Peripheral Neuropathy. Toxics 2021, 9, 229. [Google Scholar] [CrossRef] [PubMed]
- Li, Z.; Zhao, S.; Zhang, H.L.; Liu, P.; Liu, F.F.; Guo, Y.X.; Wang, X.L. Proinflammatory Factors Mediate Paclitaxel-Induced Impairment of Learning and Memory. Mediat. Inflamm. 2018, 2018, 3941840. [Google Scholar] [CrossRef]
- Tanimukai, H.; Kanayama, D.; Omi, T.; Takeda, M.; Kudo, T. Paclitaxel induces neurotoxicity through endoplasmic reticulum stress. Biochem. Biophys. Res. Commun. 2013, 437, 151–155. [Google Scholar] [CrossRef]
- Tang, M.; Zhao, S.; Liu, J.X.; Liu, X.; Guo, Y.X.; Wang, G.Y.; Wang, X.L. Paclitaxel induces cognitive impairment via necroptosis, decreased synaptic plasticity and M1 polarisation of microglia. Pharm. Biol. 2022, 60, 1556–1565. [Google Scholar] [CrossRef]
- Sekiguchi, F.; Domoto, R.; Nakashima, K.; Yamasoba, D.; Yamanishi, H.; Tsubota, M.; Wake, H.; Nishibori, M.; Kawabata, A. Paclitaxel-induced HMGB1 release from macrophages and its implication for peripheral neuropathy in mice: Evidence for a neuroimmune crosstalk. Neuropharmacology 2018, 141, 201–213. [Google Scholar] [CrossRef]
- Li, Y.; Zhang, H.; Kosturakis, A.K.; Cassidy, R.M.; Zhang, H.; Kennamer-Chapman, R.M.; Jawad, A.B.; Colomand, C.M.; Harrison, D.S.; Dougherty, P.M. MAPK signaling downstream to TLR4 contributes to paclitaxel-induced peripheral neuropathy. Brain Behav. Immun. 2015, 49, 255–266. [Google Scholar] [CrossRef]
- D’Souza, S.; Alinauskas, K.; McCrea, E.; Goodyer, C.; Antel, J.P. Differential susceptibility of human CNS-derived cell populations to TNF-dependent and independent immune-mediated injury. J. Neurosci. 1995, 15, 7293–7300. [Google Scholar] [CrossRef]
- Maqbool, A.; Lattke, M.; Wirth, T.; Baumann, B. Sustained, neuron-specific IKK/NF-κB activation generates a selective neuroinflammatory response promoting local neurodegeneration with aging. Mol. Neurodegener. 2013, 8, 40. [Google Scholar] [CrossRef]
- Miura, K.; Kinouchi, M.; Ishida, K.; Fujibuchi, W.; Naitoh, T.; Ogawa, H.; Ando, T.; Yazaki, N.; Watanabe, K.; Haneda, S.; et al. 5-fu metabolism in cancer and orally-administrable 5-fu drugs. Cancers 2010, 2, 1717–1730. [Google Scholar] [CrossRef] [PubMed]
- Groves, T.R.; Farris, R.; Anderson, J.E.; Alexander, T.C.; Kiffer, F.; Carter, G.; Wang, J.; Boerma, M.; Allen, A.R. 5-Fluorouracil chemotherapy upregulates cytokines and alters hippocampal dendritic complexity in aged mice. Behav. Brain Res. 2017, 316, 215–224. [Google Scholar] [CrossRef] [PubMed]
- Elbeltagy, M.; Mustafa, S.; Umka, J.; Lyons, L.; Salman, A.; Dormon, K.; Allcock, C.; Bennett, G.; Wigmore, P. The effect of 5-fluorouracil on the long term survival and proliferation of cells in the rat hippocampus. Brain Res. Bull. 2012, 88, 514–518. [Google Scholar] [CrossRef]
- Mustafa, S.; Walker, A.; Bennett, G.; Wigmore, P.M. 5-Fluorouracil chemotherapy affects spatial working memory and newborn neurons in the adult rat hippocampus. Eur. J. Neurosci. 2008, 28, 323–330. [Google Scholar] [CrossRef]
- Briones, T.L.; Woods, J. Dysregulation in myelination mediated by persistent neuroinflammation: Possible mechanisms in chemotherapy-related cognitive impairment. Brain Behav. Immun. 2014, 35, 23–32. [Google Scholar] [CrossRef] [PubMed]
- Kreukels, B.P.; van Dam, F.S.; Ridderinkhof, K.R.; Boogerd, W.; Schagen, S.B. Persistent neurocognitive problems after adjuvant chemotherapy for breast cancer. Clin. Breast Cancer 2008, 8, 80–87. [Google Scholar] [CrossRef]
- Mendiola, A.S.; Cardona, A.E. The IL-1β phenomena in neuroinflammatory diseases. J. Neural Transm. 2018, 125, 781–795. [Google Scholar] [CrossRef]
- Cheung, Y.T.; Lim, S.R.; Ho, H.K.; Chan, A. Cytokines as mediators of chemotherapy-associated cognitive changes: Current evidence, limitations and directions for future research. PLoS ONE 2013, 8, e81234. [Google Scholar] [CrossRef]
- Cheung, Y.T.; Ng, T.; Shwe, M.; Ho, H.K.; Foo, K.M.; Cham, M.T.; Lee, J.A.; Fan, G.; Tan, Y.P.; Yong, W.S.; et al. Association of proinflammatory cytokines and chemotherapy-associated cognitive impairment in breast cancer patients: A multi-centered, prospective, cohort study. Ann. Oncol. 2015, 26, 1446–1451. [Google Scholar] [CrossRef]
- Lee, K.S.; Chung, J.H.; Choi, T.K.; Suh, S.Y.; Oh, B.H.; Hong, C.H. Peripheral cytokines and chemokines in Alzheimer’s disease. Dement. Geriatr. Cogn. Disord. 2009, 28, 281–287. [Google Scholar] [CrossRef]
- Appay, V.; Rowland-Jones, S.L. RANTES: A versatile and controversial chemokine. Trends Immunol. 2001, 22, 83–87. [Google Scholar] [CrossRef]
- Valerio, A.; Ferrario, M.; Martinez, F.O.; Locati, M.; Ghisi, V.; Bresciani, L.G.; Mantovani, A.; Spano, P. Gene expression profile activated by the chemokine CCL5/RANTES in human neuronal cells. J. Neurosci. Res. 2004, 78, 371–382. [Google Scholar] [CrossRef] [PubMed]
- Plana, D.; Palmer, A.C.; Sorger, P.K. Independent Drug Action in Combination Therapy: Implications for Precision Oncology. Cancer Discov. 2022, 12, 606–624. [Google Scholar] [CrossRef] [PubMed]
- Anderson, J.E.; Trujillo, M.; McElroy, T.; Groves, T.; Alexander, T.; Kiffer, F.; Allen, A.R. Early Effects of Cyclophosphamide, Methotrexate, and 5-Fluorouracil on Neuronal Morphology and Hippocampal-Dependent Behavior in a Murine Model. Toxicol. Sci. 2020, 173, 156–170. [Google Scholar] [CrossRef]
- Jiang, Z.G.; Winocur, G.; Wojtowicz, J.M.; Shevtsova, O.; Fuller, S.; Ghanbari, H.A. PAN-811 prevents chemotherapy-induced cognitive impairment and preserves neurogenesis in the hippocampus of adult rats. PLoS ONE 2018, 13, e0191866. [Google Scholar] [CrossRef] [PubMed]
- Shi, D.-D.; Huang, Y.-H.; Lai, C.S.W.; Dong, C.M.; Ho, L.C.; Wu, E.X.; Li, Q.; Wang, X.-M.; Chung, S.K.; Sham, P.C.; et al. Chemotherapy-Induced Cognitive Impairment Is Associated with Cytokine Dysregulation and Disruptions in Neuroplasticity. Mol. Neurobiol. 2019, 56, 2234–2243. [Google Scholar] [CrossRef]
- Kang, S.; Lee, S.; Kim, J.; Kim, J.C.; Kim, S.H.; Son, Y.; Shin, T.; Youn, B.; Kim, J.S.; Wang, H.; et al. Chronic Treatment with Combined Chemotherapeutic Agents Affects Hippocampal Micromorphometry and Function in Mice, Independently of Neuroinflammation. Exp. Neurobiol. 2018, 27, 419–436. [Google Scholar] [CrossRef]
- Gahtori, R.; Tripathi, A.H.; Kumari, A.; Negi, N.; Paliwal, A.; Tripathi, P.; Joshi, P.; Rai, R.C.; Upadhyay, S.K. Anticancer plant-derivatives: Deciphering their oncopreventive and therapeutic potential in molecular terms. Future J. Pharm. Sci. 2023, 9, 14. [Google Scholar] [CrossRef]
- Jaiswara, P.K.; Gupta, V.K.; Sonker, P.; Rawat, S.G.; Tiwari, R.K.; Pathak, C.; Kumar, S.; Kumar, A. Nimbolide induces cell death in T lymphoma cells: Implication of altered apoptosis and glucose metabolism. Environ. Toxicol. 2021, 36, 628–641. [Google Scholar] [CrossRef]
- Choudhari, A.S.; Mandave, P.C.; Deshpande, M.; Ranjekar, P.; Prakash, O. Phytochemicals in Cancer Treatment: From Preclinical Studies to Clinical Practice. Front. Pharmacol. 2019, 10, 1614. [Google Scholar] [CrossRef] [PubMed]
- Xu, X.; Jia, L.; Ma, X.; Li, H.; Sun, C. Application Potential of Plant-Derived Medicines in Prevention and Treatment of Platinum-Induced Peripheral Neurotoxicity. Front. Pharmacol. 2021, 12, 792331. [Google Scholar] [CrossRef] [PubMed]
- Benavente-García, O.; Castillo, J. Update on uses and properties of citrus flavonoids: New findings in anticancer, cardiovascular, and anti-inflammatory activity. J. Agric. Food Chem. 2008, 56, 6185–6205. [Google Scholar] [CrossRef]
- Choe, S.C.; Kim, H.S.; Jeong, T.S.; Bok, S.H.; Park, Y.B. Naringin has an antiatherogenic effect with the inhibition of intercellular adhesion molecule-1 in hypercholesterolemic rabbits. J. Cardiovasc. Pharmacol. 2001, 38, 947–955. [Google Scholar] [CrossRef]
- Wali, A.F.; Rashid, S.; Rashid, S.M.; Ansari, M.A.; Khan, M.R.; Haq, N.; Alhareth, D.Y.; Ahmad, A.; Rehman, M.U. Naringenin Regulates Doxorubicin-Induced Liver Dysfunction: Impact on Oxidative Stress and Inflammation. Plants 2020, 9, 550. [Google Scholar] [CrossRef] [PubMed]
- John, J.; Kinra, M.; Ranadive, N.; Keni, R.; Nayak, P.G.; Jagdale, R.N.; Ahmed, S.M.; Raghavendra, K.V.; Mudgal, J.; Nandakumar, K. Neuroprotective effect of Mulmina Mango against chemotherapy-induced cognitive decline in mouse model of mammary carcinoma. Sci. Rep. 2022, 12, 3072. [Google Scholar] [CrossRef]
- Feng, S.-T.; Wang, Z.-Z.; Yuan, Y.-H.; Sun, H.-M.; Chen, N.-H.; Zhang, Y. Mangiferin: A multipotent natural product preventing neurodegeneration in Alzheimer’s and Parkinson’s disease models. Pharmacol. Res. 2019, 146, 104336. [Google Scholar] [CrossRef]
- Imran, M.; Arshad, M.S.; Butt, M.S.; Kwon, J.H.; Arshad, M.U.; Sultan, M.T. Mangiferin: A natural miracle bioactive compound against lifestyle related disorders. Lipids Health Dis. 2017, 16, 84. [Google Scholar] [CrossRef]
- Howes, M.J.; Perry, E. The role of phytochemicals in the treatment and prevention of dementia. Drugs Aging 2011, 28, 439–468. [Google Scholar] [CrossRef]
- Lawrence, J.A.; Griffin, L.; Balcueva, E.P.; Groteluschen, D.L.; Samuel, T.A.; Lesser, G.J.; Naughton, M.J.; Case, L.D.; Shaw, E.G.; Rapp, S.R. A study of donepezil in female breast cancer survivors with self-reported cognitive dysfunction 1 to 5 years following adjuvant chemotherapy. J. Cancer Surviv. 2016, 10, 176–184. [Google Scholar] [CrossRef]
- Shaw, E.G.; Rosdhal, R.; D’Agostino, R.B., Jr.; Lovato, J.; Naughton, M.J.; Robbins, M.E.; Rapp, S.R. Phase II study of donepezil in irradiated brain tumor patients: Effect on cognitive function, mood, and quality of life. J. Clin. Oncol. 2006, 24, 1415–1420. [Google Scholar] [CrossRef]
- Winocur, G.; Binns, M.A.; Tannock, I. Donepezil reduces cognitive impairment associated with anti-cancer drugs in a mouse model. Neuropharmacology 2011, 61, 1222–1228. [Google Scholar] [CrossRef] [PubMed]
- El-Saadony, M.T.; Yang, T.; Korma, S.A.; Sitohy, M.; Abd El-Mageed, T.A.; Selim, S.; Al Jaouni, S.K.; Salem, H.M.; Mahmmod, Y.; Soliman, S.M.; et al. Impacts of turmeric and its principal bioactive curcumin on human health: Pharmaceutical, medicinal, and food applications: A comprehensive review. Front. Nutr. 2022, 9, 1040259. [Google Scholar] [CrossRef] [PubMed]
- Zhang, D.-w.; Fu, M.; Gao, S.-H.; Liu, J.-L. Curcumin and Diabetes: A Systematic Review. Evid.-Based Complement. Altern. Med. 2013, 2013, 636053. [Google Scholar] [CrossRef] [PubMed]
- Cox, K.H.; Pipingas, A.; Scholey, A.B. Investigation of the effects of solid lipid curcumin on cognition and mood in a healthy older population. J. Psychopharmacol. 2015, 29, 642–651. [Google Scholar] [CrossRef]
- Mythri, R.B.; Bharath, M.M. Curcumin: A potential neuroprotective agent in Parkinson’s disease. Curr. Pharm. Des. 2012, 18, 91–99. [Google Scholar] [CrossRef]
- Waseem, M.; Parvez, S. Mitochondrial dysfunction mediated cisplatin induced toxicity: Modulatory role of curcumin. Food Chem. Toxicol. 2013, 53, 334–342. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.; Huang, P.; Law, S.; Tian, H.; Leung, W.; Xu, C. Preventive Effect of Curcumin Against Chemotherapy-Induced Side-Effects. Front. Pharmacol. 2018, 9, 1374. [Google Scholar] [CrossRef]
- Hewlings, S.J.; Kalman, D.S. Curcumin: A Review of Its Effects on Human Health. Foods 2017, 6, 92. [Google Scholar] [CrossRef]
- Lao, C.D.; Ruffin, M.T.t.; Normolle, D.; Heath, D.D.; Murray, S.I.; Bailey, J.M.; Boggs, M.E.; Crowell, J.; Rock, C.L.; Brenner, D.E. Dose escalation of a curcuminoid formulation. BMC Complement. Altern. Med. 2006, 6, 10. [Google Scholar] [CrossRef]
- Alavi, M.; Farkhondeh, T.; Aschner, M.; Samarghandian, S. Resveratrol mediates its anti-cancer effects by Nrf2 signaling pathway activation. Cancer Cell Int. 2021, 21, 579. [Google Scholar] [CrossRef] [PubMed]
- Ko, J.H.; Sethi, G.; Um, J.Y.; Shanmugam, M.K.; Arfuso, F.; Kumar, A.P.; Bishayee, A.; Ahn, K.S. The Role of Resveratrol in Cancer Therapy. Int. J. Mol. Sci. 2017, 18, 2589. [Google Scholar] [CrossRef] [PubMed]
- Xiao, Q.; Zhu, W.; Feng, W.; Lee, S.S.; Leung, A.W.; Shen, J.; Gao, L.; Xu, C. A Review of Resveratrol as a Potent Chemoprotective and Synergistic Agent in Cancer Chemotherapy. Front. Pharmacol. 2018, 9, 1534. [Google Scholar] [CrossRef]
- Annaji, M.; Poudel, I.; Boddu, S.H.S.; Arnold, R.D.; Tiwari, A.K.; Babu, R.J. Resveratrol-loaded nanomedicines for cancer applications. Cancer Rep. 2021, 4, e1353. [Google Scholar] [CrossRef]
- Shi, D.-D.; Dong, C.M.; Ho, L.C.; Lam, C.T.W.; Zhou, X.-D.; Wu, E.X.; Zhou, Z.-J.; Wang, X.-M.; Zhang, Z.-J. Resveratrol, a natural polyphenol, prevents chemotherapy-induced cognitive impairment: Involvement of cytokine modulation and neuroprotection. Neurobiol. Dis. 2018, 114, 164–173. [Google Scholar] [CrossRef] [PubMed]
- Liu, X.; Tang, M.; He, T.Y.; Zhao, S.; Li, H.Z.; Li, Z.; Guo, Y.X.; Wang, X.L. Resveratrol Improves Paclitaxel-Induced Cognitive Impairment in Mice by Activating SIRT1/PGC-1α Pathway to Regulate Neuronal State and Microglia Cell Polarization. Drug Des. Devel. Ther. 2023, 17, 1125–1138. [Google Scholar] [CrossRef]
- Shaito, A.; Posadino, A.M.; Younes, N.; Hasan, H.; Halabi, S.; Alhababi, D.; Al-Mohannadi, A.; Abdel-Rahman, W.M.; Eid, A.H.; Nasrallah, G.K.; et al. Potential Adverse Effects of Resveratrol: A Literature Review. Int. J. Mol. Sci. 2020, 21, 2084. [Google Scholar] [CrossRef]
- Brown, V.A.; Patel, K.R.; Viskaduraki, M.; Crowell, J.A.; Perloff, M.; Booth, T.D.; Vasilinin, G.; Sen, A.; Schinas, A.M.; Piccirilli, G.; et al. Repeat dose study of the cancer chemopreventive agent resveratrol in healthy volunteers: Safety, pharmacokinetics, and effect on the insulin-like growth factor axis. Cancer Res. 2010, 70, 9003–9011. [Google Scholar] [CrossRef]
- Salehi, B.; Mishra, A.P.; Nigam, M.; Sener, B.; Kilic, M.; Sharifi-Rad, M.; Fokou, P.V.T.; Martins, N.; Sharifi-Rad, J. Resveratrol: A Double-Edged Sword in Health Benefits. Biomedicines 2018, 6, 91. [Google Scholar] [CrossRef]
- Patel, K.R.; Scott, E.; Brown, V.A.; Gescher, A.J.; Steward, W.P.; Brown, K. Clinical trials of resveratrol. Ann. N. Y. Acad. Sci. 2011, 1215, 161–169. [Google Scholar] [CrossRef]
- Cheruku, S.P.; Ramalingayya, G.V.; Chamallamudi, M.R.; Biswas, S.; Nandakumar, K.; Nampoothiri, M.; Gourishetti, K.; Kumar, N. Catechin ameliorates doxorubicin-induced neuronal cytotoxicity in in vitro and episodic memory deficit in in vivo in Wistar rats. Cytotechnology 2018, 70, 245–259. [Google Scholar] [CrossRef] [PubMed]
- Nguyen, L.D.; Ehrlich, B.E. Cellular mechanisms and treatments for chemobrain: Insight from aging and neurodegenerative diseases. EMBO Mol. Med. 2020, 12, e12075. [Google Scholar] [CrossRef] [PubMed]
- Gibson, E.M.; Nagaraja, S.; Ocampo, A.; Tam, L.T.; Wood, L.S.; Pallegar, P.N.; Greene, J.J.; Geraghty, A.C.; Goldstein, A.K.; Ni, L.; et al. Methotrexate Chemotherapy Induces Persistent Tri-glial Dysregulation that Underlies Chemotherapy-Related Cognitive Impairment. Cell 2019, 176, 43–55.e13. [Google Scholar] [CrossRef] [PubMed]
- Jiang, Z.G.; Lebowitz, M.S.; Ghanbari, H.A. Neuroprotective activity of 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (PAN-811), a cancer therapeutic agent. CNS Drug Rev. 2006, 12, 77–90. [Google Scholar] [CrossRef] [PubMed]
- Chao, J.; Synold, T.W.; Morgan, R.J., Jr.; Kunos, C.; Longmate, J.; Lenz, H.J.; Lim, D.; Shibata, S.; Chung, V.; Stoller, R.G.; et al. A phase I and pharmacokinetic study of oral 3-aminopyridine-2-carboxaldehyde thiosemicarbazone (3-AP, NSC #663249) in the treatment of advanced-stage solid cancers: A California Cancer Consortium Study. Cancer Chemother. Pharmacol. 2012, 69, 835–843. [Google Scholar] [CrossRef]
- Nutting, C.M.; van Herpen, C.M.; Miah, A.B.; Bhide, S.A.; Machiels, J.P.; Buter, J.; Kelly, C.; de Raucourt, D.; Harrington, K.J. Phase II study of 3-AP Triapine in patients with recurrent or metastatic head and neck squamous cell carcinoma. Ann. Oncol. 2009, 20, 1275–1279. [Google Scholar] [CrossRef]
Drug for Chemobrain Intervention | Role in Chemobrain | References |
---|---|---|
Epicatechin |
| [61] |
Xanthone |
| [53] |
Naringin |
| [70] |
Mangiferin |
| [108] |
Curcumin |
| [119] |
Resveratrol |
| [126] |
Catechin |
| [132] |
Donepezil |
| [107] |
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
Jaiswara, P.K.; Shukla, S.K. Chemotherapy-Mediated Neuronal Aberration. Pharmaceuticals 2023, 16, 1165. https://doi.org/10.3390/ph16081165
Jaiswara PK, Shukla SK. Chemotherapy-Mediated Neuronal Aberration. Pharmaceuticals. 2023; 16(8):1165. https://doi.org/10.3390/ph16081165
Chicago/Turabian StyleJaiswara, Pradip Kumar, and Surendra Kumar Shukla. 2023. "Chemotherapy-Mediated Neuronal Aberration" Pharmaceuticals 16, no. 8: 1165. https://doi.org/10.3390/ph16081165
APA StyleJaiswara, P. K., & Shukla, S. K. (2023). Chemotherapy-Mediated Neuronal Aberration. Pharmaceuticals, 16(8), 1165. https://doi.org/10.3390/ph16081165