Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-mwx4w Total loading time: 0 Render date: 2024-06-24T14:10:06.048Z Has data issue: false hasContentIssue false

7 - Cognitive impairment: basic science

from Section 2 - Cancer Symptom Mechanisms and Models: Clinical and Basic Science

Published online by Cambridge University Press:  05 August 2011

Perry N. Fuchs
Affiliation:
The University of Texas at Arlington
Jessica A. Boyette-Davis
Affiliation:
The University of Texas M. D. Anderson Cancer Center
Adrian J. Dunn
Affiliation:
University of Hawaii at Manoa
Charles S. Cleeland
Affiliation:
University of Texas, M. D. Anderson Cancer Center
Michael J. Fisch
Affiliation:
University of Texas, M. D. Anderson Cancer Center
Adrian J. Dunn
Affiliation:
University of Hawaii, Manoa
Get access

Summary

Cognitive dysfunction is increasingly recognized by practitioners and researchers as one of the symptoms that cause great distress for patients with cancer, their families, and their health care providers. The prevalence of cognitive impairment varies and is based on several factors, including the amount of time the patient has had cancer, the type of cancer, the length of treatment, and the treatment used, and is estimated to affect anywhere between 17% and 75% of patients with cancer. In a series of experiments, Wefel et al. noted signs of cognitive impairment in 33% to 35% of cancer patients prior to treatment with chemotherapy and in approximately 61% of patients after chemotherapeutic treatment.

Findings such as these, coupled with the fact that treatment for cancer begins soon after diagnosis, has led to the characterization of chemotherapy-related cognitive impairments as “chemofog” or “chemobrain.” Although the nature of the impairment seems to vary among patients, those who experience chemobrain generally report subtle changes in the ability to maintain focus and engage in routine daily activities. Some investigations performed before and after the initiation of chemotherapeutic treatment find that memory, motor dexterity, and executive function (frontal subcortical components) tend to be impaired, with attention and psychomotor speed remaining unimpaired. Other studies have shown that working memory, or the ability to process information and do multiple tasks, is often impaired, whereas hippocampal components of memory, such as retention and consolidation, frequently are not.

Type
Chapter
Information
Cancer Symptom Science
Measurement, Mechanisms, and Management
, pp. 60 - 69
Publisher: Cambridge University Press
Print publication year: 2010

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Wieneke, MH, Dienst, ER. Neuropsychological assessment of cognitive functioning following chemotherapy for breast cancer. Psychooncology 4(1):61–66, 1995.CrossRefGoogle Scholar
Wefel, JS, Lenzi, R, Theriault, R, Buzdar, AU, Cruickshank, S, Meyers, CA. ‘Chemobrain’ in breast carcinoma? A prologue. Cancer 101(3):466–475, 2004.CrossRefGoogle ScholarPubMed
Wefel, JS, Lenzi, R, Theriault, RL, Davis, RN, Meyers, CA. The cognitive sequelae of standard-dose adjuvant chemotherapy in women with breast carcinoma: results of a prospective, randomized, longitudinal trial. Cancer 100(11):2292–2299, 2004.CrossRefGoogle ScholarPubMed
Cleeland, CS. Cancer-related symptoms. Semin Radiat Oncol 10(3):175–190, 2000.CrossRefGoogle ScholarPubMed
Meyers, CA, Albitar, M, Estey, E. Cognitive impairment, fatigue, and cytokine levels in patients with acute myelogenous leukemia or myelodysplastic syndrome. Cancer 104(4):788–793, 2005.CrossRefGoogle ScholarPubMed
Wilson, CJ, Finch, CE, Cohen, HJ. Cytokines and cognition: the case for a head-to-toe inflammatory paradigm. J Am Geriatr Soc 50(12):2041–2056, 2002.CrossRefGoogle ScholarPubMed
Miller, GE, Cohen, S, Ritchey, AK. Chronic psychological stress and the regulation of pro-inflammatory cytokines: a glucocorticoid-resistance model. Health Psychol 21(6):531–541, 2002.CrossRefGoogle ScholarPubMed
Aubert, A, Goodall, G, Dantzer, R, Gheusi, G. Differential effects of lipopolysaccharide on pup retrieving and nest building in lactating mice. Brain Behav Immun 11(2):107–118, 1997.CrossRefGoogle ScholarPubMed
Larson, SJ, Dunn, AJ. Behavioral effects of cytokines. Brain Behav Immun 15(4):371–387, 2001.CrossRefGoogle ScholarPubMed
Gibertini, M, Newton, C, Friedman, H, Klein, TW. Spatial learning impairment in mice infected with Legionella pneumophila or administered exogenous interleukin-1-beta. Brain Behav Immun 9(2):113–128, 1995.CrossRefGoogle ScholarPubMed
Heyser, CJ, Masliah, E, Samimi, A, Campbell, IL, Gold, LH. Progressive decline in avoidance learning paralleled by inflammatory neurodegeneration in transgenic mice expressing interleukin 6 in the brain. Proc Natl Acad Sci U S A 94(4):1500–1505, 1997.CrossRefGoogle Scholar
Oitzl, MS, Oers, H, Schöbitz, B, Kloet, ER. Interleukin-1 beta, but not interleukin-6, impairs spatial navigation learning. Brain Res 613(1):160–163, 1993.CrossRefGoogle Scholar
Otterness, IG, Seymour, PA, Golden, HW, Reynolds, JA, Daumy, GO. The effects of continuous administration of murine interleukin-1 alpha in the rat. Physiol Behav 43(6):797–804, 1988.CrossRefGoogle ScholarPubMed
Dunn, AJ, Chapman, Y, Antoon, M. Endotoxin-induced behavioral changes of mice in the multicompartment chamber are distinct from those of interleukin-1. Neurosci Res Commun 10:63–69, 1992.Google Scholar
Dantzer, R. Cytokine-induced sickness behavior: where do we stand?Brain Behav Immun 15(1):7–24, 2001.CrossRefGoogle ScholarPubMed
Avitsur, R, Donchin, O, Barak, O, Cohen, E, Yirmiya, R. Behavioral effects of interleukin-1 beta: modulation by gender, estrus cycle, and progesterone. Brain Behav Immun 9(3):234–241, 1995.CrossRefGoogle ScholarPubMed
Krueger, JM, Majde, JA. Microbial products and cytokines in sleep and fever regulation. Crit Rev Immunol 14(3–4):355–379, 1994.CrossRefGoogle ScholarPubMed
Armario, A, Hernández, J, Bluethmann, H, Hidalgo, J. IL-6 deficiency leads to increased emotionality in mice: evidence in transgenic mice carrying a null mutation for IL-6. J Neuroimmunol 92(1–2):160–169, 1998.CrossRefGoogle ScholarPubMed
Alleva, E, Cirulli, F, Bianchi, M, et al. Behavioural characterization of interleukin-6 overexpressing or deficient mice during agonistic encounters. Eur J Neurosci 10(12):3664–3672, 1998.CrossRefGoogle ScholarPubMed
Swiergiel, AH, Dunn, AJ. Feeding, exploratory, anxiety- and depression-related behaviors are not altered in interleukin-6-deficient male mice. Behav Brain Res 171(1):94–108, 2006.CrossRefGoogle Scholar
Valentine, AD, Meyers, CA, Kling, MA, Richelson, E, Hauser, P. Mood and cognitive side effects of interferon-alpha therapy. Semin Oncol 25(1 Suppl 1):39–47, 1998.Google ScholarPubMed
Licinio, J, Kling, MA, Hauser, P. Cytokines and brain function: relevance to interferon-alpha-induced mood and cognitive changes. Semin Oncol 25(1 Suppl 1):30–38, 1998.Google ScholarPubMed
Glass, JD, Wesselingh, SL, Selnes, OA, McArthur, JC. Clinical-neuropathologic correlation in HIV-associated dementia. Neurology 43(11):2230–2237, 1993.CrossRefGoogle ScholarPubMed
Knoblach, SM, Fan, L, Faden, AI. Early neuronal expression of tumor necrosis factor-alpha after experimental brain injury contributes to neurological impairment. J Neuroimmunol 95(1–2):115–125, 1999.CrossRefGoogle ScholarPubMed
Suzuki, M, Umegaki, H, Ieda, S, Mogi, N, Iguchi, A. Factors associated with cognitive impairment in elderly patients with diabetes mellitus. J Am Geriatr Soc 54(3):558–559, 2006.CrossRefGoogle ScholarPubMed
Yamada, K, Iida, R, Miyamoto, Y, et al. Neurobehavioral alterations in mice with a targeted deletion of the tumor necrosis factor-alpha gene: implications for emotional behavior. J Neuroimmunol 111(1–2):131–138, 2000.CrossRefGoogle ScholarPubMed
Magnano, MD, Robinson, WH, Genovese, MC. Demyelination and inhibition of tumor necrosis factor (TNF). Clin Exp Rheumatol 22(5 Suppl 35):S134–S140, 2004.Google Scholar
O'Brien, JM, Wewers, MD, Moore, SA, Allen, JN. Taxol and colchicine increase LPS-induced pro-IL-1 beta production, but do not increase IL-1 beta secretion: a role for microtubules in the regulation of IL-1 beta production. J Immunol 154(8):4113–4122, 1995.Google Scholar
Gan, XH, Jewett, A, Bonavida, B. Activation of human peripheral-blood-derived monocytes by cis-diamminedichloroplatinum: enhanced tumoricidal activity and secretion of tumor necrosis factor-alpha. Nat Immun 11(3):144–155, 1992.Google ScholarPubMed
Zaks-Zilberman, M, Zaks, TZ, Vogel, SN. Induction of proinflammatory and chemokine genes by lipopolysaccharide and paclitaxel (Taxol) in murine and human breast cancer cell lines. Cytokine 15(3):156–165, 2001.CrossRefGoogle ScholarPubMed
Dina, OA, Chen, X, Reichling, D, Levine, JD. Role of protein kinase Cepsilon and protein kinase A in a model of paclitaxel-induced painful peripheral neuropathy in the rat. Neuroscience 108(3):507–515, 2001.CrossRefGoogle Scholar
Dudchenko, PA. An overview of the tasks used to test working memory in rodents. Neurosci Biobehav Rev 28(7):699–709, 2004.CrossRefGoogle ScholarPubMed
Morris, R. Developments of a water-maze procedure for studying spatial learning in the rat. J Neurosci Methods 11(1):47–60, 1984.CrossRefGoogle ScholarPubMed
Baldi, E, Efoudebe, M, Lorenzini, CA, Bucherelli, C. Spatial navigation in the Morris water maze: working and long lasting reference memories. Neurosci Lett 378(3):176–180, 2005.CrossRefGoogle ScholarPubMed
D'Hooge, R, Deyn, PP. Applications of the Morris water maze in the study of learning and memory. Brain Res Brain Res Rev 36(1):60–90, 2001.CrossRefGoogle Scholar
Gouirand, AM, Matuszewich, L. The effects of chronic unpredictable stress on male rats in the water maze. Physiol Behav 86(1–2):21–31, 2005.CrossRefGoogle ScholarPubMed
Sparkman, NL, Kohman, RA, Scott, VJ, Boehm, GW. Bacterial endotoxin-induced behavioral alterations in two variations of the Morris water maze. Physiol Behav 86(1–2):244–251, 2005.CrossRefGoogle ScholarPubMed
Martin, RS, Secchi, RL, Sung, E, et al. Effects of cannabinoid receptor ligands on psychosis-relevant behavior models in the rat. Psychopharmacology (Berl) 165(2):128–135, 2003.CrossRefGoogle ScholarPubMed
Borzan, J, LaGraize, SC, Fuchs, PN. Effect of chronic vincristine treatment on mechanical withdrawal response and pre-pulse inhibition in the rat. Neurosci Lett 364(2):110–113, 2004.CrossRefGoogle ScholarPubMed
Yanovski, JA, Packer, RJ, Levine, JD, Davidson, TL, Micalizzi, M, D'Angio, G. An animal model to detect the neuropsychological toxicity of anticancer agents. Med Pediatr Oncol 17(3):216–221, 1989.CrossRefGoogle ScholarPubMed
Stock, HS, Rosellini, RA, Abrahamsen, GC, McCaffrey, RJ, Ruckdeschel, JC. Methotrexate does not interfere with an appetitive Pavlovian conditioning task in Sprague-Dawley rats. Physiol Behav 58(5):969–973, 1995.CrossRefGoogle Scholar
Pickens, CL, Holland, PC. Conditioning and cognition. Neurosci Biobehav Rev 28(7):651–661, 2004.CrossRefGoogle ScholarPubMed
Lejeune, H, Hermans, I, Mocaër, E, Rettori, MC, Poignant, JC, Richelle, M. Amineptine, response timing, and time discrimination in the albino rat. Pharmacol Biochem Behav 51(2–3):165–173, 1995.CrossRefGoogle ScholarPubMed
Robbins, TW. The 5-choice serial reaction time task: behavioural pharmacology and functional neurochemistry. Psychopharmacology (Berl) 163(3–4):362–380, 2002.CrossRefGoogle ScholarPubMed
Inglis, WL, Olmstead, MC, Robbins, TW. Selective deficits in attentional performance on the 5-choice serial reaction time task following pedunculopontine tegmental nucleus lesions. Behav Brain Res 123(2):117–131, 2001.CrossRefGoogle ScholarPubMed
Boyette-Davis, JA, Thompson, CD, Fuchs, PN. Alterations in attentional mechanisms in response to acute inflammatory pain and morphine administration. Neuroscience 151(2):558–563, 2008.CrossRefGoogle ScholarPubMed
Carli, M, Robbins, TW, Evenden, JL, Everitt, BJ. Effects of lesions to ascending noradrenergic neurones on performance of a 5-choice serial reaction task in rats; implications for theories of dorsal noradrenergic bundle function based on selective attention and arousal. Behav Brain Res 9(3):361–380, 1983.CrossRefGoogle ScholarPubMed
Boyette-Davis, JA, Fuchs, PN. Differential effects of paclitaxel treatment on cognitive functioning and mechanical sensitivity. Neurosci Lett 453(3):170–174, 2009.CrossRefGoogle ScholarPubMed

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×