AccScience Publishing / TD / Volume 1 / Issue 1 / DOI: 10.36922/td.v1i1.46
REVIEW

Advances in the study of the pathogenesis of cancer-related cognitive impairment

Jiwei Jiang1,2† Zhongli Du3† Yanli Wang1,2 Hanping Shi4 Wenyi Li1,2 Yuan Zhang1,2 Mengfan Sun1,2 Zhimin Bian5* Jun Xu1,2*
Show Less
1 Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
2 China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
3 National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, China
4 Department of Gastrointestinal Surgery/Clinical Nutrition, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
5 Comprehensive Department, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
Tumor Discovery 2022, 1(1), 46 https://doi.org/10.36922/td.v1i1.46
Submitted: 2 December 2021 | Accepted: 7 March 2022 | Published: 22 March 2022
© 2022 by the Authors. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution 4.0 International License ( https://creativecommons.org/licenses/by/4.0/ )
Abstract

Advances in diagnostic and therapeutic strategies have significantly contributed to an increase in the survival rate of cancer patients. Recently, several studies suggested that cancer patients may exhibit symptoms of cognitive impairment before, during and even many years after the completion of therapies, negatively impacting the quality of life and functional independence of cancer survivors. Clinically, the coexistence of cancer and cognitive impairment reminds scientists of paraneoplastic syndrome, especially limbic encephalitis. However, some cancer patients show symptoms of cognition deterioration after treatment, without any typical psychiatric symptoms, epileptic seizures or positive antineuronal antibodies, suggesting that the relationship between cancer and cognitive deficits is more common than previously anticipated. Most importantly, many aspects of the association between cancer and cognitive impairment remain uncertain. The definitive connection between systemic cancer and central nervous system is yet to be established. Therefore, this review summarizes the current evidence on the potential pathophysiology in these patients with cancer-related cognitive impairment, and reviews the knowledge gaps and the potential counteracting strategies.

Keywords
Cancer
Cognitive impairment
Mechanism
Chemotherapy
Funding
National Natural Science Foundation of China
Beijing Youth Talent Team Support Program
References
[1]

Zhou M, Wang H, Zeng X, et al., 2019, Mortality, morbidity, and risk factors in China and its provinces, 1990-2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet, 394: 1145–1158. https://doi.org/10.1016/S0140-6736(19)30427-1

[2]

Jia L, Du Y, Chu L, Zhang Z, Li F, Lyu D, et al., 2020, Prevalence, risk factors, and management of dementia and mild cognitive impairment in adults aged 60 years or older in China: A cross-sectional study. Lancet Public Health, 5: e661–e671. https://doi.org/10.1016/S2468-2667(20)30185-7

[3]

Siegel RL, Miller KD, Jemal A, 2019, Cancer statistics, 2019. CA Cancer J Clin, 69: 7–34. https://doi.org/10.3322/caac.21551

[4]

Ren X, Boriero D, Chaiswing L, et al., 2019. 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, 1865(6): 1088–1097. https://doi.org/10.1016/j.bbadis.2019.02.007

[5]

Ahles TA, Root JC, 2018, Cognitive effects of cancer and cancer treatments. Annu Rev Clin Psychol, 14: 425–451. https://doi.org/10.1146/annurev-clinpsy-050817-084903

[6]

Wefel JS, Kesler SR, Noll KR, et al., 2015, Clinical characteristics, pathophysiology, and management of noncentral nervous system cancer-related cognitive impairment in adults. CA Cancer J Clin, 65: 123–138. https://doi.org/10.3322/caac.21258

[7]

Lange M, Joly F, Vardy J, et al., 2019, Cancer-related cognitive impairment: An update on state of the art, detection, and management strategies in cancer survivors. Ann Oncol, 30(12): 1925–1940. https://doi.org/10.1093/annonc/mdz410

[8]

Janelsins MC, Heckler CE, Peppone LJ, et al., 2017, Cognitive complaints in survivors of breast cancer after chemotherapy compared with age-matched controls: An analysis from a nationwide, multicenter, prospective longitudinal study. J Clin Oncol, 35(5): 506–514. https://doi.org/10.1200/jco.2016.68.5826

[9]

Janelsins MC, Kesler SR, Ahles TA, et al., 2014, Prevalence, mechanisms, and management of cancer-related cognitive impairment. Int Rev Psychiatry, 26(1): 102–113. https://doi.org/10.3109/09540261.2013.864260

[10]

Rummel NG, Chaiswing L, Bondada S, et al., 2021, Chemotherapy-induced cognitive impairment: focus on the intersection of oxidative stress and TNFα. Cell Mol Life Sci, 78(19–20): 6533–6540. https://doi.org/10.1007/s00018-021-03925-4

[11]

Ren X, Keeney JT, Miriyala S, et al., 2019, The triangle of death of neurons: oxidative damage, mitochondrial dysfunction, and loss of choline-containing biomolecules in brains of mice treated with doxorubicin: Advanced insights into mechanisms of chemotherapy induced cognitive impairment (“chemobrain”) involving TNF-α. Free Radic Biol Med, 134: 1–8. https://doi.org/10.1016/j.freeradbiomed.2018.12.029

[12]

Tönnies E, Trushina E, 2017, Oxidative stress, synaptic dysfunction, and Alzheimer’s disease. J Alzheimers Dis, 57: 1105–1121. https://doi.org/10.3233/jad-161088

[13]

Williams AM, van Wijngaarden E, Seplaki CL, et al., 2020, Cognitive function in patients with chronic lymphocytic leukemia: A cross-sectional study examining effects of disease and treatment. Leuk Lymphoma, 61(7): 1627–1635. https://doi.org/10.1080/10428194.2020.1728748

[14]

Kaiser J, Dietrich J, Amiri M, et al., 2020, Cognitive performance and psychological distress in breast cancer patients at disease onset. Front Psychol, 10: 2584. https://doi.org/10.3389/fpsyg.2019.02584

[15]

Shiroishi MS, Gupta V, Bigjahan B, et al., 2017, Brain cortical structural differences between non-central nervous system cancer patients treated with and without chemotherapy compared to non-cancer controls: A cross-sectional pilot MRI study using clinically-indicated scans. Proc SPIE Int Soc Opt Eng, 10572: 105720G. https://doi.org/10.1117/12.2285971

[16]

Olson B, Marks DL, 2019, Pretreatment cancer-related cognitive impairment-mechanisms and outlook. Cancers (Basel), 11(5): 687. https://doi.org/10.3390/cancers11050687

[17]

Patel SK, Wong AL, Wong FL, et al., 2015, Inflammatory biomarkers, comorbidity, and neurocognition in women with newly diagnosed breast cancer. J Natl Cancer Inst, 107(8): djv131. https://doi.org/10.1093/jnci/djv131

[18]

Lyon DE, Cohen R, Chen H, et al., 2016, Relationship of systemic cytokine concentrations to cognitive function over two years in women with early stage breast cancer. J Neuroimmunol, 301: 74–82. https://doi.org/10.1016/j.jneuroim.2016.11.002

[19]

Wardill HR, Mander KA, Van Sebille YZ, et al., 2016, Cytokine-mediated blood brain barrier disruption as a conduit for cancer/chemotherapy-associated neurotoxicity and cognitive dysfunction. Int J Cancer, 139(12): 2635–2645. https://doi.org/10.1002/ijc.30252 

[20]

Santos JC, Pyter LM, 2018, Neuroimmunology of behavioral comorbidities associated with cancer and cancer treatments. Front Immunol, 9: 1195. https://doi.org/10.3389/fimmu.2018.01195 

[21]

Kaur D, Sharma V, Deshmukh R, 2019, Activation of microglia and astrocytes: a roadway to neuroinflammation and Alzheimer’s disease. Inflammopharmacology, 27(4): 663–677. https://doi.org/10.1007/s10787-019-00580-x

[22]

Capuron L, Miller AH, 2011, Immune system to brain signaling: neuropsychopharmacological implications. Pharmacol Ther, 130(2): 226–238. https://doi.org/10.1016/j.pharmthera.2011.01.014

[23]

Fischer HG, Reichmann G, 2001, Brain dendritic cells and macrophages/microglia in central nervous system inflammation. J Immunol, 166(4): 2717–2726. https://doi.org/10.4049/jimmunol.166.4.2717

[24]

Clancy J, D’Souza-Schorey C, 2018, Extracellular vesicles in cancer: purpose and promise. Cancer J, 24(2): 65–69. https://doi.org/10.1097/ppo.0000000000000306

[25]

Xu R, Rai A, Chen M, et al., 2018, Extracellular vesicles in cancer: Implications for future improvements in cancer care. Nat Rev Clin Oncol, 15(10): 617–638. https://doi.org/10.1038/s41571-018-0036-9

[26]

Kok VC, Yu CC, 202, Cancer-derived exosomes: Their role in cancer biology and biomarker development. Int J Nanomed, 15: 8019–8036. https://doi.org/10.2147/IJN.S272378

[27]

Delpech JC, Herron S, Botros MB, et al., 2019, Neuroimmune crosstalk through extracellular vesicles in health and disease. Trends Neurosci, 42(5): 361–372. https://doi.org/10.1016/j.tins.2019.02.007

[28]

Li JJ, Wang B, Kodali MC, et al., 2018, In vivo evidence for the contribution of peripheral circulating inflammatory exosomes to neuroinflammation. J Neuroinflammation, 15(1): 8. https://doi.org/10.1186/s12974-017-1038-8

[29]

Pascual M, Ibáñez F, Guerri C, 2020, Exosomes as mediators of neuron-glia communication in neuroinflammation. Neural Regen Res, 15(5): 796–801. https://doi.org/10.4103/1673-5374.268893

[30]

Frühbeis C, Kuo-Elsner WP, Müller C, et al., 2002. Oligodendrocytes support axonal transport and maintenance via exosome secretion. PLoS Biol, 18(12): e3000621. https://doi.org/10.1371/journal.pbio.3000621

[31]

Gharbi T, Zhang Z, Yang GY, 2020, The function of astrocyte mediated extracellular vesicles in central nervous system diseases. Front Cell Dev Biol, 8: 568889. https://doi.org/10.3389/fcell.2020.568889

[32]

Antonucci F, Turola E, Riganti L, et al., 2012, Microvesicles released from microglia stimulate synaptic activity via enhanced sphingolipid metabolism. EMBO J, 31(5): 1231–1240. https://doi.org/10.1038/emboj.2011.489

[33]

Zhang G, Yang P, 2018, A novel cell-cell communication mechanism in the nervous system: exosomes. J Neurosci Res, 96(1): 45–52. https://doi.org/10.1002/jnr.24113

[34]

Treps L, Edmond S, Harford-Wright E, et al., 2016, Extracellular vesicle-transported Semaphorin3A promotes vascular permeability in glioblastoma. Oncogene, 35(20): 2615–2623. https://doi.org/10.1038/onc.2015.317

[35]

Zhang Z, Yin J, Lu C, et al., 2019, Exosomal transfer of long non-coding RNA SBF2-AS1 enhances chemoresistance to temozolomide in glioblastoma. J Exp Clin Cancer Res, 38(1): 166. https://doi.org/10.1186/s13046-019-1139-6 

[36]

Koh YQ, Tan CJ, Toh YL, et al., 2020, Role of exosomes in cancer-related cognitive impairment. Int J Mol Sci, 21(8):2755. https://doi.org/10.3390/ijms21082755

[37]

Sweeney MD, Zhao Z, Montagne A, et al., 2019, Blood-brain barrier: from physiology to disease and back. Physiol Rev, 99(1): 21–78. https://doi.org/10.1152/physrev.00050.2017

[38]

Liebner S, Dijkhuizen RM, Reiss Y, et al., 2018, Functional morphology of the blood-brain barrier in health and disease. Acta Neuropathol, 135(3): 311–336. https://doi.org/10.1007/s00401-018-1815-1

[39]

Morad G, Carman CV, Hagedorn EJ, et al., 2019, Tumor-derived extracellular vesicles breach the intact blood-brain barrier via transcytosis. ACS Nano, 13(12): 13853–13865. https://doi.org/10.1021/acsnano.9b04397

[40]

Sweeney MD, Sagare AP, Zlokovic BV, 2018, Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorder. Nat Rev Neurol, 14(3): 133–150. https://doi.org/10.1038/nrneurol.2017.188

[41]

Hanahan D, Weinberg RA, 2011, Hallmarks of cancer: The next generation. Cell, 144(5): 646–674. https://doi.org/10.1016/j.cell.2011.02.013

[42]

Saharinen P, Eklund L, Alitalo K, 2017, Therapeutic targeting of the angiopoietin-TIE pathway. Nat Rev Drug Discov, 16(9): 635–661.

[43]

Zajączkowska R, Kocot-Kępska M, Leppert W, et al., 2019, Mechanisms of chemotherapy-induced peripheral neuropathy. Int J Mol Sci, 20(6): 1451. https://doi.org/10.3390/ijms20061451

[44]

Vitali M, Ripamonti CI, Roila F, et al., 2017. Cognitive impairment and chemotherapy: A brief overview. Crit Rev Oncol Hematol, 118: 7–14. https://doi.org/10.1016/j.critrevonc.2017.08.001 

[45]

de Ruiter MB, Reneman L, Boogerd W, et al., 2011, Cerebral hyporesponsiveness and cognitive impairment 10 years after chemotherapy for breast cancer. Hum Brain Mapp, 32(8): 1206–1219. https://doi.org/10.1002/hbm.21102

[46]

Janelsins MC, Heckler CE, Peppone LJ, et al., 2018, Longitudinal trajectory and characterization of cancer-related cognitive impairment in a nationwide cohort study. J Clin Oncol, 36(32): JCO2018786624. https://doi.org/10.1200/jco.2018.78.6624

[47]

Collins B, MacKenzie J, Tasca GA, et al., 2013, Cognitive effects of chemotherapy in breast cancer patients: A dose-response study. Psychooncology, 22(7): 1517–1527. https://doi.org/10.1002/pon.3163

[48]

Mounier NM, Abdel-Maged AE, Wahdan SA, et al., 2020, Chemotherapy-induced cognitive impairment (CICI): An overview of etiology and pathogenesis. Life Sci, 258: 118071. https://doi.org/10.1016/j.lfs.2020.118071

[49]

Bagnall-Moreau C, Chaudhry S, Salas-Ramirez K, et al., 2019, Chemotherapy-induced cognitive impairment is associated with increased inflammation and oxidative damage in the hippocampus. Mol Neurobiol, 56(10): 7159–7172. https://doi.org/10.1007/s12035-019-1589-z

[50]

Michalak S, Rybacka-Mossakowska J, Ambrosius W, et al., 2016, The markers of glutamate metabolism in peripheral blood mononuclear cells and neurological complications in lung cancer patients. Dis Markers, 2016: 2895972. https://doi.org/10.1155/2016/2895972

[51]

Gibson EM, Purger D, Mount CW, et al., 2014, Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain. Science, 344(6183): 1252304. https://doi.org/10.1126/science.1252304

[52]

Briones TL, Woods J, 2014, Dysregulation in myelination mediated by persistent neuroinflammation: Possible mechanisms in chemotherapy-related cognitive impairment. Brain Behav Immun, 35: 23–32. https://doi.org/10.1016/j.bbi.2013.07.175

[53]

Berlin C, Lange K, Lekaye HC, et al., 2020, Long-term clinically relevant rodent model of methotrexate-induced cognitive impairment. Neuro Oncol, 22(8): 1126-1137. https://doi.org/10.1093/neuonc/noaa086

[54]

Sirichoat A, Krutsri S, Suwannakot K, et al., 2019, Melatonin protects against methotrexate-induced memory deficit and hippocampal neurogenesis impairment in a rat model. Biochem Pharmacol, 163: 225–233.

[55]

Sirichoat A, Suwannakot K, Chaisawang P, et al., 2020, Melatonin attenuates 5-fluorouracil-induced spatial memory and hippocampal neurogenesis impairment in adult rats. Life Sci, 248: 117468. https://doi.org/10.1016/j.lfs.2020.117468

[56]

El-Agamy SE, Abdel-Aziz AK, Wahdan S, et al., 2018, Astaxanthin ameliorates doxorubicin-induced cognitive impairment (chemobrain) in experimental rat model: Impact on oxidative, inflammatory, and apoptotic machineries. Mol Neurobiol, 55(7): 5727–5740. https://doi.org/10.1007/s12035-017-0797-7

[57]

Keeney JT, Miriyala S, Noel T, et al., 2015, Superoxide induces protein oxidation in plasma and TNF-α elevation in macrophage culture: Insights into mechanisms of neurotoxicity following doxorubicin chemotherapy. Cancer Lett, 367(2): 157–161. https://doi.org/10.1016/j.canlet.2015.07.023

[58]

Lv L, Mao S, Dong H, et al., 2020, Pathogenesis, assessments, and management of chemotherapy-related cognitive impairment (CRCI): An updated literature review. J Oncol, 2020: 3942439. https://doi.org/10.1155/2020/3942439

[59]

Ren X, St Clair DK, Butterfield DA, 2017, Dysregulation of cytokine mediated chemotherapy induced cognitive impairment. Pharmacol Res, 117: 267–273. https://doi.org/10.1016/j.phrs.2017.01.001

[60]

Gibson EM, Monje M, 2019, Emerging mechanistic underpinnings and therapeutic targets for chemotherapy-related cognitive impairment. Curr Opin Oncol, 31(6): 531–539. https://doi.org/10.1097/cco.0000000000000578

[61]

Lyon D, Elmore L, Aboalela N, et al., 2014, Potential epigenetic mechanism(s) associated with the persistence of psychoneurological symptoms in women receiving chemotherapy for breast cancer: A hypothesis. Biol Res Nurs, 16(2): 160-174.

[62]

Cardoso S, Santos RX, Carvalho C, et al., 2008, Doxorubicin increases the susceptibility of brain mitochondria to Ca(2+)- induced permeability transition and oxidative damage. Free Radic Biol Med, 45(10): 1395–1402.

[63]

Uzar E, Koyuncuoglu HR, Uz E, et al., 2006, The activities of antioxidant enzymes and the level of malondialdehyde in cerebellum of rats subjected to methotrexate: Protective effect of caffeic acid phenethyl ester. Mol Cell Biochem, 291(1-2): 63–68. https://doi.org/10.1007/s11010-006-9196-5

[64]

Cauli O, 2021, Oxidative stress and cognitive alterations induced by cancer chemotherapy drugs: A scoping review. Antioxidants (Basel), 10(7): 1116. https://doi.org/10.3390/antiox10071116

[65]

Nyunt T, Britton M, Wanichthanarak K, et al., 2019, Mitochondrial oxidative stress-induced transcript variants of ATF3 mediate lipotoxic brain microvascular injury. Free Radic Biol Med, 143: 25–46. https://doi.org/10.1016/j.freeradbiomed.2019.07.024

[66]

Jebahi F, Sharma S, Bloss JE, et al., 2021, Effects of tamoxifen on cognition and language in women with breast cancer: A systematic search and a scoping review. Psychooncology, 30(8): 1262–1277. https://doi.org/10.1002/pon.5696 

[67]

Taleat Z, Larsson A, Ewing AG, 2019, Anticancer drug tamoxifen affects catecholamine transmitter release and storage from single cells. ACS Chem Neurosci, 10(4): 2060–2069. https://doi.org/10.1021/acschemneuro.8b00714

[68]

Gervais NJ, Remage-Healey L, Starrett JR, et al., 2019, Adverse effects of aromatase inhibition on the brain and behavior in a nonhuman primate. J Neurosci, 39(5): 918–928. https://doi.org/10.1523/jneurosci.0353-18.2018

[69]

Joly F, Heutte N, Duclos B, et al., 2016, Prospective evaluation of the impact of antiangiogenic treatment on cognitive functions in metastatic renal cancer. Eur Urol Focus, 2(6): 642–649. https://doi.org/10.1016/j.euf.2016.04.009

[70]

Mulder SF, Bertens D, Desar IM, et al., 2014, Impairment of cognitive functioning during sunitinib or sorafenib treatment in cancer patients: A cross sectional study. BMC Cancer, 14: 219. https://doi.org/10.1186/1471-2407-14-219

[71]

Abdel-Aziz AK, Mantawy EM, Said RS, et al., 2016, The tyrosine kinase inhibitor, sunitinib malate, induces cognitive impairment in vivo via dysregulating VEGFR signaling, apoptotic and autophagic machineries. Exp Neurol, 283(Pt A): 129–141.

[72]

Joly F, Castel H, Tron L, et al., 2020, Potential effect of immunotherapy agents on cognitive function in cancer patients. J Natl Cancer Inst, 112(2): 123–127. https://doi.org/10.1093/jnci/djz168

[73]

Greenbaum U, Kebriaei P, Srour SA, et al., 2021, Chimeric antigen receptor T-cell therapy toxicities. Br J Clin Pharmacol, 87(6): 2414–2424. https://doi.org/10.1111/bcp.14403

[74]

Cuzzubbo S, Belin C, Chouahnia K, et al., 2018, Assessing cognitive function in patients treated with immune checkpoint inhibitors: A feasibility study. Psychooncology, 27(7): 1861–1864. https://doi.org/10.1002/pon.4725

[75]

Neelapu SS, Locke FL, Bartlett NL, et al., 2017, Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl J Med, 377(26): 2531–2544. https://doi.org/10.1056/nejmoa1707447

[76]

Chen H, Wang F, Zhang P, et al., 2019, Management of cytokine release syndrome related to CAR-T cell therapy. Front Med, 13(5): 610–617. https://doi.org/10.1007/s11684-019-0714-8

[77]

Santomasso BD, Park JH, Salloum D, et al., 2018, Clinical and biologic correlates of neurotoxicity associated with CAR T cell therapy in patients with B-cell acute lymphoblastic leukemia. Cancer Discov, 8(8): 958–971. https://doi.org/10.1158/2159-8290.cd-17-1319 

[78]

Pazzaglia S, Briganti G, Mancuso M, et al., 2020, Neurocognitive decline following radiotherapy: Mechanisms and therapeutic implications. Cancers (Basel), 12: 146. https://doi.org/10.3390/cancers12010146

[79]

McDowell LJ, Ringash J, Xu W, et al., 2019, A cross sectional study in cognitive and neurobehavioral impairment in long-term nasopharyngeal cancer survivors treated with intensity-modulated radiotherapy. Radiother Oncol, 131: 179–185. https://doi.org/10.1016/j.radonc.2018.09.012

[80]

Haldbo-Classen L, Amidi A, Lukacova S, et al., 2020, Cognitive impairment following radiation to hippocampus and other brain structures in adults with primary brain tumours. Radiother Oncol, 148: 1–7. https://doi.org/10.1016/j.radonc.2020.03.023

[81]

Makale MT, McDonald CR, Hattangadi-Gluth JA, et al., 2017, Mechanisms of radiotherapy-associated cognitive disability in patients with brain tumours. Nat Rev Neurol, 13(1): 52–64. https://doi.org/10.1038/nrneurol.2016.185

[82]

McHugh D, Gil J, 2018, Senescence and aging: causes, consequences, and therapeutic avenues. J Cell Biol, 217(1): 65–77. https://doi.org/10.1083/jcb.201708092

[83]

Seo J, Park M, 2020, Molecular crosstalk between cancer and neurodegenerative diseases. Cell Mol Life Sci, 77(14): 2659–2680. https://doi.org/10.1007/s00018-019-03428-3

[84]

Lanni C, Masi M, Racchi M, et al., 2021, Cancer and Alzheimer’s disease inverse relationship: an age-associated diverging derailment of shared pathways. Mol Psychiatry, 26(1): 280–295. https://doi.org/10.1038/s41380-020-0760-2

[85]

Nebbioso A, Tambaro FP, Dell’Aversana C, et al., 2018, Cancer epigenetics: moving forward. PLoS Genet, 14(6): e1007362. https://doi.org/10.1371/journal.pgen.1007362

[86]

Rebeck GW, 2017, The role of APOE on lipid homeostasis and inflammation in normal brains. J Lipid Res, 58(8): 1493–1499. https://doi.org/10.1194/jlr.R075408

[87]

Ahles TA, Saykin AJ, Noll WW, et al., 2003, The relationship of APOE genotype to neuropsychological performance in long-term cancer survivors treated with standard dose chemotherapy. Psychooncology, 12(6): 612–619. https://doi.org/10.1002/pon.742

[88]

Fernandez HR, Varma A, Flowers SA, et al., 2020, Cancer chemotherapy related cognitive impairment and the impact of the Alzheimer’s disease risk factor APOE. Cancers (Basel), 12: 3842. https://doi.org/10.3390/cancers12123842

Conflict of interest
No conflict of interest was reported by all authors.
Share
Back to top
Tumor Discovery, Electronic ISSN: 2810-9775 Published by AccScience Publishing