Clearing the Air: A Review of Our Current Understanding of “Chemo Fog”

10
PALLIATIVE MEDICINE (A JATOI, SECTION EDITOR) Clearing the Air: A Review of Our Current Understanding of Chemo FogErin OFarrell & Joyce MacKenzie & Barbara Collins Published online: 13 March 2013 # Springer Science+Business Media New York 2013 Abstract An increasing number of cancer survivors has led to a greater interest in the long-term side effects of cancer treat- ments and their impact on quality of life. In particular, cognitive impairments have been frequently reported by cancer survivors as an adverse effect which they attribute to the neurotoxicity of chemotherapy and have dubbed chemobrainor chemo fog.Research within the past 1520 years has explored the many factors thought to contribute to cancer-related cognitive decline in an attempt to determine a potential cause. In spite of many confounding factors, there is growing evidence that the neuro- toxicity of chemotherapy does contribute to cognitive changes. This review examines the evolution of chemo fogresearch with a look at methodological issues, the status of our current understanding, and suggestions for future research. Keywords Chemo fog . Chemobrain . Chemotherapy-related cognitive impairment . Cognitive function . Adjuvant chemotherapy Introduction Advances in cancer treatments have led to increased survival rates and a growing number of individuals who are living with the long-term side effects of cancer therapies. Cognitive de- cline is one of the most frequently reported adverse effects among cancer survivors [1]. Patients generally attribute these cognitive disturbances to toxic effects of chemotherapy, as implied by their use of terms such as chemo fogand chemobrain.However, in the early years of the adjuvant chemotherapy era, such symptoms were generally dismissed by the medical community as psychological rather than neu- rological. Although it was recognized that many chemothera- peutic agents could be acutely neurotoxic when delivered directly to the central nervous system, it was believed that most of these agents could not penetrate the bloodbrain barrier and thus were unlikely to cause neurotoxicity when administered systemically in the adjuvant setting. Most studies of the cognitive effects of adjuvant systemic chemotherapy have been conducted primarily in breast cancer (BC) patients. These studies began to appear in the literature in the latter half of the 1990s [2, 3], shortly following the establishment of adjuvant chemotherapy as the new standard of care for treatment of most breast tumors. The study of chemotherapy-related cognitive impairment (CRCI) has large- ly focused on patients with BC for several reasons, primarily its high prevalence and generally good prognosis, such that many BC survivors expect to resume active, cognitively de- manding lives following treatment. These early efforts were generally small, local, retrospective, cross-sectional studies and, although they were important in validating the cognitive complaints of cancer patients and impelling the research en- deavor, uncontrolled confounding factors prevented isolation of the specific effects of chemotherapy on cognition. One of the most troublesome confounding factors was this issue of psychological distresscancer patients, particularly those patients requiring chemotherapy, are at elevated risk of anxiety and depression, both of which can undermine cognitive function. Other confounding factors include fatigue and effects of anesthetic and surgery. One particularly important confound- er is the effect of medications and treatments given in conjunc- tion with cytostatic drugs that may, in and of themselves, affect cognition. Among these are palliative medications designed to mitigate the side effects of chemotherapy, including anti- E. OFarrell : B. Collins School of Psychology, University of Ottawa, Ottawa, ON, Canada J. MacKenzie : B. Collins (*) The Ottawa Hospital Civic Campus, 1053 Carling Ave, Room A628, Ottawa, ON K1Y 4E9, Canada e-mail: [email protected] Curr Oncol Rep (2013) 15:260269 DOI 10.1007/s11912-013-0307-7

Transcript of Clearing the Air: A Review of Our Current Understanding of “Chemo Fog”

PALLIATIVE MEDICINE (A JATOI, SECTION EDITOR)

Clearing the Air: A Review of Our Current Understandingof “Chemo Fog”

Erin O’Farrell & Joyce MacKenzie & Barbara Collins

Published online: 13 March 2013# Springer Science+Business Media New York 2013

Abstract An increasing number of cancer survivors has led toa greater interest in the long-term side effects of cancer treat-ments and their impact on quality of life. In particular, cognitiveimpairments have been frequently reported by cancer survivorsas an adverse effect which they attribute to the neurotoxicity ofchemotherapy and have dubbed “chemobrain” or “chemo fog.”Research within the past 15–20 years has explored the manyfactors thought to contribute to cancer-related cognitive declinein an attempt to determine a potential cause. In spite of manyconfounding factors, there is growing evidence that the neuro-toxicity of chemotherapy does contribute to cognitive changes.This review examines the evolution of “chemo fog” researchwith a look at methodological issues, the status of our currentunderstanding, and suggestions for future research.

Keywords Chemo fog . Chemobrain .

Chemotherapy-related cognitive impairment . Cognitivefunction . Adjuvant chemotherapy

Introduction

Advances in cancer treatments have led to increased survivalrates and a growing number of individuals who are living withthe long-term side effects of cancer therapies. Cognitive de-cline is one of the most frequently reported adverse effectsamong cancer survivors [1]. Patients generally attribute thesecognitive disturbances to toxic effects of chemotherapy, as

implied by their use of terms such as “chemo fog” and“chemobrain.” However, in the early years of the adjuvantchemotherapy era, such symptoms were generally dismissedby the medical community as psychological rather than neu-rological. Although it was recognized that many chemothera-peutic agents could be acutely neurotoxic when delivereddirectly to the central nervous system, it was believed thatmost of these agents could not penetrate the blood–brainbarrier and thus were unlikely to cause neurotoxicity whenadministered systemically in the adjuvant setting.

Most studies of the cognitive effects of adjuvant systemicchemotherapy have been conducted primarily in breast cancer(BC) patients. These studies began to appear in the literature inthe latter half of the 1990s [2, 3], shortly following theestablishment of adjuvant chemotherapy as the new standardof care for treatment of most breast tumors. The study ofchemotherapy-related cognitive impairment (CRCI) has large-ly focused on patients with BC for several reasons, primarilyits high prevalence and generally good prognosis, such thatmany BC survivors expect to resume active, cognitively de-manding lives following treatment. These early efforts weregenerally small, local, retrospective, cross-sectional studiesand, although they were important in validating the cognitivecomplaints of cancer patients and impelling the research en-deavor, uncontrolled confounding factors prevented isolation ofthe specific effects of chemotherapy on cognition.

One of the most troublesome confounding factors was thisissue of psychological distress—cancer patients, particularlythose patients requiring chemotherapy, are at elevated risk ofanxiety and depression, both of which can undermine cognitivefunction. Other confounding factors include fatigue and effectsof anesthetic and surgery. One particularly important confound-er is the effect of medications and treatments given in conjunc-tion with cytostatic drugs that may, in and of themselves, affectcognition. Among these are palliative medications designed tomitigate the side effects of chemotherapy, including anti-

E. O’Farrell :B. CollinsSchool of Psychology, University of Ottawa, Ottawa, ON, Canada

J. MacKenzie : B. Collins (*)The Ottawa Hospital – Civic Campus, 1053 Carling Ave,Room A628,Ottawa, ON K1Y 4E9, Canadae-mail: [email protected]

Curr Oncol Rep (2013) 15:260–269DOI 10.1007/s11912-013-0307-7

nauseants, anti-emetics, and anti-inflammatories, as well asother adjuvant treatments. In some two thirds of BC patients,their tumors express estrogen and/or progesterone receptorsand grow in the presence of these hormones [4]. An importantadjunctive treatment for these tumors is long-term (5 years ormore) anti-estrogen therapy (with or without cytostatic drugs).It is well established that estrogen acts in the brain to influencecognition [5, 6], and there is growing evidence to suggest thatthe hormonal therapies, which work either by blocking estro-gen receptors or by preventing estrogen synthesis, have adetrimental effect on cognitive functioning [7–17].

Careful attention to study design and methodology is re-quired to address these many confounding factors. Therefore,key researchers in this field formed the International Cognitionand Cancer Task Force (ICCTF) [18] in 2006 to establishguidelines for harmonizing study methodologies, thereby pro-moting more comparable findings among studies [19•]. As aresult, we are now able to provide at least partial answers tosome of the most frequently asked questions concerning CRCI.This article will review current methodological issues andrecommendations and the status of our knowledge with regardto these frequently asked questions.

Methodological Considerations

Cross-Sectional Versus Longitudinal Designs

One of the major limitations to the early studies on CRCI wasthat they were retrospective and cross-sectional in design andthus could not distinguish chemotherapy-induced cognitivechanges from pretreatment cognitive disturbances related tohost or disease factors. These studies were prey to bothoverestimation and underestimation of CRCI. Overestimationwas a particular risk in studies that used a healthy controlgroup because, as later prospective studies demonstrated, upto 35 % of BC patients showed pretreatment cognitive impair-ment which would have been misattributed to chemotherapyin these cross-sectional designs [20–22]. On the other hand,cross-sectional designs failed to detect pretreatment-to-posttreatment cognitive decline in up to 46 % of individualswho, because of high premorbid function, still scored withinthe normal range on posttreatment cognitive testing [22]. Inmost cases, the ICCTF recommends prospective longitudinalstudies [19•].

Control Groups

An important methodological consideration is the nature of thecontrol group used. In retrospective, cross-sectional studies inwhich patients are assessed at a single point in time aftercompleting chemotherapy, a disease control group is essentialto account for a number of factors besides treatment that may

influence cognition, such as constitutional and genetic riskfactors for cancer, psychological distress associated with acancer diagnosis, and biological changes associated with thedisease itself (as noted already, cancer patients are at increasedrisk of cognitive disturbance before any exposure to adjuvanttherapy). A disadvantage to the disease control group is the factthat those patients who are not receiving chemotherapy may bereceiving alternative treatments, such as anti-estrogen therapy,that may, in themselves, affect cognition [7–9, 11–17].

In prospective within-subject designs, repeated measure-ment of the same subject before and after treatment serves tocontrol for stable host and disease factors. One importantdisadvantage of this approach is that practice effects associatedwith repeated testing of an individual can mask subtle adverseeffects of treatment. Investigators attempt to mitigate this byusing parallel alternative forms of tests but practice effectsencompass far more than just content-based savings (e.g.,reduced anxiety, an established strategy) and, thus, it is stillimportant to include a control group to measure and accountfor these effects. In fact, without a control group, it may evenappear that cancer patients’ cognition improves after chemo-therapy [22]. The ICCTF recommends inclusion of both adisease control group and a healthy control group even inprospective longitudinal studies (in the event that the practiceeffect may be different in cancer patients from that in healthyindividuals) [19•]. However, we have empirically addressedthe issue of control groups. We obtained the same results withour longitudinal data whether we used a disease control group,a healthy control group, or published norms (unpublishedobservations).

Approach to Data Analysis

The effect sizes in this field are typically quite modest and,thus, the data analytic techniques used can significantly influ-ence the conclusions drawn from a particular study. One keydifference among studies is whether data are analyzed at theaggregate level (e.g., comparing group means) or at the levelof individual impairment or decline (comparing the frequencyof impairment or decline across groups). In that CRCI affectsonly a subgroup of patients, these subtle effects can be easilyobscured in comparing group means. Several studies havefailed to find group differences in mean cognitive functionbut have found significant differences in the frequency ofcognitive impairment or decline between the chemotherapyand control groups [22, 23]. However, as elegantly demon-strated by Shilling et al. [24] and Schilder et al. [21], the use ofdifferent definitions of impairment (e.g., the extent of negativechange in a given score as well as how many scores showingdecline would constitute impairment) greatly influences esti-mates of risk. The ICCTF generally recommends that, in orderto be considered impaired, the standardized scores on a givenmeasure should be −1.5 or lower and that the number of such

Curr Oncol Rep (2013) 15:260–269 261

scores required to consider a given participant cognitivelyimpaired should be determined in accordance with the prob-ability of obtaining such a result by chance given the size ofthe test battery [19•].

When data are analyzed at the aggregate level, longitudinalmodeling techniques are recommended over analysis of vari-ance or t tests, in that they allow for differing numbers andspacing of assessments across respondents and are more ro-bust to violation of assumptions [19•]. Different statisticalapproaches have been taken to evaluate decline at the individ-ual level. The ICCTF recommends approaches that accountfor practice effects, such as the reliable change index withpractice effects model or regression-based change models[19•]. We have argued in favor of the latter because it allowsthe inclusion of important covariates [25]. Differences amongstudies in the choice of covariates used in the analysis mayalso contribute to inconsistency in results. Most studies haveincluded, as a minimum, measures of mood and fatigue, andthis would seem advisable.

Cognitive Measures Used

Another critical issue concerns whether cognition is assessedsubjectively, by means of a self-report questionnaire, or ob-jectively, by means of performance-based neuropsychologicaltesting. In most cases, subjective measures of cognitive im-pairment correlate poorly, if at all, with objective measures,indicating a need to include both [26–30]. Among neuropsy-chological studies, the size and composition of the test batterydiffers widely. The selection of tests can contribute tooverestimation or underestimation of true cognitive impair-ment [24]. Many neuropsychological tests may not be sensi-tive enough to detect the subtle changes associated withchemotherapy or may fail to target the affected cognitivedomains. For example, basic mental status screening testsare probably not sensitive enough for this purpose [31]. Thenumber of neuropsychological measures used may also be acritical determinant of study outcome as the likelihood offinding an impaired score or abnormal decline in a score willgenerally increase in keeping with the size of the test battery[32]. The ICCTF has recommended a core battery of neuro-psychological measures that are well validated, sensitive tothe types of deficits observed in cancer patients, andappropriate for multinational use [19•].

What Do We Know Now?

Is There an Association Between Chemotherapy Exposureand Cognitive Disturbance?

Despite considerable variability in methodology and find-ings, most of the studies done in this area over the past

20 years support the idea that chemotherapy-exposed BCpatients are at increased risk of cognitive dysfunction. Wefeland Schagen [33] recently tabulated results of studiesconducted in BC patients between 1995 and 2012 and foundthat 78 % of cross-sectional studies (n = 23) and 69 % ofprospective longitudinal studies (n = 26) found evidence insupport of CRCI.

Is the Cognitive Disturbance Causedby Chemotherapy-Related Neurotoxicity?

It is now quite clear that many factors contribute to cognitivedisturbances in cancer patients, including anxiety, depression,hormonal fluctuations, fatigue, other treatments, and the dis-ease itself. Recent prospective studies find cognitive dysfunc-tion in a substantial portion of cancer patients even prior tostarting adjuvant therapy [20, 21]. MRI measures of brainstructure and function have also shown pretreatment differ-ences between cancer patients and healthy controls [20,34–36] which cannot be fully accounted for by depression,anxiety, fatigue, or surgical factors, suggesting that they maybe due to the disease itself. In recognition of this, it has beensuggested that terms such as “chemofog” and “chemobrain”be replaced with a more encompassing term such as “cancer-or cancer-therapy- related cognitive change” [37]. At the sametime, there is also mounting evidence that chemotherapyexposure is a significant contributing factor to cognitivesymptoms in cancer survivors.

As recently as 8–10 years ago, a BC patient complainingof cognitive disturbances during chemotherapy was likely tobe prescribed an antidepressant. This belief that cognitivedisturbances were psychogenic was supported by the factthat most studies found poor, if any, correlation betweenpatients’ subjective reports of cognitive disturbance andobjective measures of cognitive performance [26–30]. Sub-jective complaints were found to correlate more stronglywith measures of emotional state, reinforcing this idea thatwhat a patient referred to as chemo fog was a psychologicalreaction to stress. However, recent findings suggest that thepoor correlation between subjective and objective cognitivemeasures may have to do with fundamental differences inmeasurement parameters [29]. It seems likely that patients’self-assessments capture perceived decline from premorbidfunction, whereas, in most cases, the neuropsychologicalscores do not (typically, static scores are used in thesecorrelations). In analyzing our own data, we found that anobjective pretreatment–posttreatment measure of cognitivechange did correlate with subjective reports, whereas a one-time posttreatment objective measure did not (unpublishedobservations). Deprez et al. [38••] also found a significantcorrelation between objective and subjective measures whenusing a cognitive change score. It has also been suggestedthat subjective and objective measures often tap different

262 Curr Oncol Rep (2013) 15:260–269

cognitive domains and that this may contribute to their poorcorrelation. In relating subjective complaints to neuropsy-chological test scores, Deprez et al. also focused on specific,relevant, and compatible cognitive domains. Other investi-gators who have taken this more focused approach [7, 39]have also found significant subjective–objective correla-tions. Moreover, although cognitive complaints do tend tocorrelate with emotional functioning, most studies find thatthis cannot fully account for the objective neuropsycholog-ical abnormalities [2, 23, 27, 33, 40••, 41]. Clearly, there ismore to the story than psychological distress.

Increasingly sophisticated clinical studies, supplementedby animal and brain imaging research, are producing objec-tive evidence that chemotherapy is neurotoxic and can con-tribute to cognitive disturbances. An early study by vanDam et al. [3] hinted at this, by suggesting a dose–responserelationship. They compared cognitive function in high-riskBC patients who had been randomly assigned to receivehigh-dose or standard-dose chemotherapy and in early-stageBC patients receiving no systemic adjuvant treatment. Thisstudy, conducted an average of 2 years after completion ofchemotherapy, revealed a significant difference among thegroups in risk of cognitive impairment, with 32 % of thehigh-dose group showing cognitive impairment comparedwith 17 % of the standard-dose group and 9 % of the controlgroup. In a follow-up to that study, these investigators alsoshowed that their high-dose group was significantly morelikely to show late electrophysiological abnormalities [42,43] than the control patients, with the risks in the standard-dose group falling somewhere in between. This group hascorroborated their findings in a prospective randomizedcontrolled study which showed that BC patients receivinghigh-dose therapy were at a significantly elevated risk ofcognitive decline, whereas the standard-dose group was not[44]. Other cross-sectional studies have identified durationof treatment and number of chemotherapy cycles as riskfactors for cognitive disturbance [2, 26, 45]. Magnetic res-onance spectroscopy studies in BC patients have shownwhite matter abnormalities following high-dose chemother-apy that were not detectable following lower-dose inductionchemotherapy [46–48].

We capitalized on this notion of “dose–response” rela-tionship as an approach to establishing causality in our latestprospective longitudinal study [40••]. We assessed BC pa-tients after each cycle of chemotherapy and observed alinear decline in cognitive function even after controllingfor baseline performance, practice effects, and changes inmood and fatigue. We submit that this clear dose–responserelationship strongly suggests that chemotherapy is indeed acause of cognitive disturbance.

Compelling evidence of chemotherapy neurotoxicity alsocomes from animal studies, which allow tighter control ofthe factors that confound clinical research. These studies

have shown persistent performance decrements in animalmodels of learning and memory following exposure to che-motherapeutic agents, as well as decreased neurogenesis andcellular proliferation and increased cellular death withinareas of the brain such as the hippocampus and thesubventricular zone [49, 50].

Further causal evidence for the neurotoxicity of chemo-therapy comes from structural and functional neuroimagingstudies, which show differences in cerebral blood flow[51–53] and in the volume of white and grey matter inchemotherapy-treated BC patients compared with controls[38••, 46–48, 54–59]. A landmark neuroimaging study wasthat of McDonald et al. [60••] as it was the first controlledprospective MRI study in the field. They found significantdecreases in gray matter density in bilateral frontal, tempo-ral, and cerebellar regions as well as the right thalamus inBC patients shortly following completion of chemotherapythat were not evident in either a disease or healthy controlgroup. These changes could not be accounted for in terms ofpostsurgical effects, disease stage, psychiatric symptoms,psychotropic medication, or hormonal treatment status,suggesting that they were due to the effects of chemotherapy.New techniques such as diffusion tensor imaging (DTI) arecontributing to a better understanding of the nature and scopeof the underlying structural changes and suggest that whitematter may be particularly vulnerable to the neurotoxic effectsof chemotherapy and that changes in white matter functionmay persist for years after completion of treatment [55].

Functional MRI (fMRI) and positron emission tomogra-phy (PET) studies have also been conducted in cancerpatients. These studies show patterns of brain activation inresponse to a cognitive challenge and provide a “window onthe working brain.” Abnormalities have been observed in anumber of brain areas in response to a variety of activationtasks. For example, increased activation in the prefrontalcortex and the cerebellum has been observed in patientsduring a working memory task [53]. Increased activation hasbeen interpreted as an indication that the brain is having tocompensate for insult by recruiting a wider neural networkthan would normally be required to support the task. Theseaberrations in neural activation patterns can be observed in theabsence of differences in performance on the cognitive activa-tion tasks and may account for patient complaints of cognitivedecline in the absence of actual performance decrements (i.e.,theymay experience their cognitive processing as less efficientand more tiring). Regional decreases in brain activation havealso been observed, and are construed as evidence that thedamage to the brain has surpassed its capacity to compensate.

Neuroimaging studies in this field are restricted by thesame methodological flaws that characterized the early neu-ropsychological studies, including the failure to conductpretreatment baseline assessment. Studies by our group re-vealed significant pretreatment differences in white and grey

Curr Oncol Rep (2013) 15:260–269 263

matter volume (whole brain and regional) between patientsand controls [36], as well as in brain activation patterns onfMRI [34, 35], emphasizing the importance of prospectivelongitudinal designs. We have published one of the first pro-spective longitudinal fMRI studies in BC patients [61], whichshowed abnormal changes in the activation pattern inchemotherapy-treated patients and thus lends further supportto the idea that chemotherapy affects neural function.

Functional imaging studies, although holding great prom-ise, are in their infancy and the significance of the findings isdifficult to interpret. For example, both increases and decreasesin brain activation are construed as evidence of brain dysfunc-tion, and high numbers of comparisons are conducted in smallsamples, raising concerns about inflated type I error rates.Improvement of this situation will require that studies becomemore focused and hypothesis-driven.

The recent study of Deprez et al. [38••] constitutes a sig-nificant advance in the imaging literature and a model forfuture studies. It is one of the first studies to examine relation-ships between changes in neuropsychological and neuroim-aging measures in a well-controlled prospective design.Thirty-five BC patients who received chemotherapy, as wellas a group of cancer patients not treated with chemotherapyand a healthy control group, were repeatedly assessed with acomprehensive neuropsychological battery and DTI. The re-sults showed significant decline in white matter integrity inmultiple tracts involved in cognition, significant decreases onneuropsychological tests of attention and memory, and signif-icant correlations between the neuropsychological and imag-ing results in the chemotherapy group. This cross-validationfrom the neuropsychological and imaging investigations ismuch more compelling than either finding in isolation.

What Is the Frequency/Rate of Occurrence of CRCI?

There is general consensus among researchers that only asubgroup of BC patients develop CRCI; however, the esti-mated rate of impairment ranges from 17 to 78 % [33].Clearly, this variation has to do with the aforementionedmethodological differences. In a recent longitudinal studyconducted by our group that met the recommendations ofthe ICCTF [40••], we found that approximately one third ofpatients showed cognitive decline over the course of theirchemotherapy. This is in close agreement with a previouslongitudinal study conducted by our group [23] and withfigures reported by Jansen et al. [62].

What Cognitive Domains Are Impaired?

All cognitive and psychomotor domains have been impli-cated in one study or another [26, 40••, 63–67]. Disagree-ment among studies as to the most vulnerable cognitivedomains is primarily due to the multifactorial nature of

neuropsychological tests and the relatively arbitrary assign-ment of a given test to a particular cognitive domain. In ourrecent longitudinal study [40••], we assigned tests to cogni-tive domains on empirical grounds by means of principalcomponents analysis. This resulted in four cognitive factorswhich seemed to correspond to processing speed, workingmemory, visual memory, and verbal memory. We obtained alarge effect size for working memory and processing speed,and significant but small effect sizes for the domains ofvisual memory and verbal memory. This fits well withpatients’ self-reports of diminished cognitive efficiencyand difficulties with multitasking. It is also consistent withresults of recent imaging [38••, 54, 56] and animal [49]studies showing that the toxic effects of chemotherapymay affect white matter. Alternatively, working memoryand processing speed scores may have been more sensitivebecause they were time-dependent and because very subtlecognitive deficits may be better captured by speed than byaccuracy of response. For this reason, computerized testingis attracting increasing interest from researchers in this fieldbecause it allows for accurate measurement of response timein milliseconds.

It could well be that chemotherapy-related neurotoxicityresults in a generalized decrease in mental processing speedand capacity rather than focal cognitive deficit and that thismay be manifested in preexisting areas of weakness that willdiffer from one individual to another. This could partlyaccount for why studies are more likely to find significanteffects if impairment is analyzed at the individual levelindependently of a specific cognitive domain.

What Is the Severity and Duration of CRCI?

Meta-analyses of BC studies [12, 68, 69, 70•, 71] consis-tently yield small to moderate effect sizes (−0.2 to −0.5range), indicating a subtle negative effect of chemotherapyon cognitive functioning. Anderson-Hanley et al. [72] foundlarger effect sizes (approaching 1.0 in the domains of exec-utive function and verbal memory), but their analysis in-cluded studies of various cancer populations and covered abroader range of disease severity.

Retrospective, cross-sectional studies have found evi-dence of impaired cognitive functioning in BC patients aslong as 21 years after treatment [51, 73], and irregularities inbrain structure and function have also been reported in long-term BC survivors 4–21 years after treatment. Functionaldifferences include altered activation patterns on fMRI [51]and PET [53] and lower amplitude and increased latency ofthe P3 component in electrophysiological studies [42,74–76]. Structural studies using various MRI techniques(voxel-based morphometry, spectroscopy, DTI) show reduc-tions in brain volume [55, 59], axonal injury, and decreasedwhite matter integrity [55]

264 Curr Oncol Rep (2013) 15:260–269

More recent prospective longitudinal studies with mid-range to long-range follow-up (9 months to 4 years) gener-ally find that CRCI remits after termination of chemothera-py [26, 63, 77, 78]. However, there appears to be a smallsubgroup showing more intractable cognitive dysfunction[78], and one recent prospective longitudinal study [79]indicated the occurrence of new, delayed cognitive declinein 29 % of patients following treatment completion (wequestion if this might be partly due to postchemotherapycommencement of hormonal therapy). Recent epidemiolog-ical studies have looked at whether or not chemotherapyexposure is a risk factor for the later development of de-mentia. Heflin et al. [80] found that, among 486 monozy-gotic geriatric twin pairs discordant for cancer, long-termcancer survivors were twice as likely to be diagnosed withdementia as their unaffected sibling. In very large cohorts ofelderly BC survivors, Heck et al. [81] found a significantlyhigher risk of dementia in women treated with chemothera-py than in those patients not exposed to chemotherapy,whereas Baxter et al. [82] reported that past chemotherapywas not associated with a greater risk of developingdementia and Du et al. [83] found that dementia risk wassignificantly lower in patients who had received chemo-therapy in the past. We simply do not know yet whether ornot chemotherapy exposure increases the risk of dementialater in life.

What Is the Impact of CRCI in Everyday Life?

In an online survey conducted in 2010 by the CanadianBreast Cancer Network to determine the economic impactof BC, it was determined that chemotherapy had importanteconomic implications for survivors. Among some 450 re-spondents—women with a BC diagnosis in the previous5 years—8 % reported that “chemobrain” was a significantbarrier to returning to work. Those women who had re-ceived chemotherapy had the greatest reduction in house-hold income, had taken more time off work (as had theirfamily members), were more likely to have had to quit theirjobs, and had a greater perception that the financial burdenimposed by their illness would impact their long-termhealth. Chemotherapy exposure also emerged as a signifi-cant predictor of work changes after cancer in a population-based study with a more heterogeneous cancer population[84]. Smaller cohort studies of BC patients that includedneuropsychological testing have reported an association be-tween cognitive functioning and work-related outcomes(perceived ability to work and actual likelihood of returnto work) [22, 85]. Reid-Arndt et al. [86] reported an associ-ation between executive functioning deficits and decreasedproductivity, community involvement, and social role func-tioning in BC patients 1 month after chemotherapy. In sum,evidence is beginning to accrue that, although CRCI may be

subtle, it has important functional implications and canadversely affect quality of life for cancer survivors.

What Are the Risk Factors for CRCI?

In their recent prospective longitudinal study, Ahles et al.[63] found that older age and lower cognitive reserve wererisk factors for short-term treatment-related reductions inprocessing speed, and that additional treatment with hor-monal therapy was a risk factor for more persistent declinein verbal ability. The results of our initial study were com-patible with this, likewise indicating that lower educationallevel (considered an index of cognitive reserve) was a riskfactor for short-term decline [23] and that patients who wenton to receive hormonal therapy after chemotherapy weremore likely to show persistent decline at the 1-year follow-up [77]. Wefel et al. [79] found that the delayed declineobserved in almost one third of their participants was relatedto baseline performance, further hinting at neural reserve asa risk factor for long-term deficits. At the same time, otherstudies have failed to find any correlation between CRCIand age or cognitive reserve (at least as reflected in educa-tion) [33]. There may also be genetic risk factors for CRCI.Ahles et al. [87] found that, among long-term survivors ofBC and lymphoma treated with chemotherapy, those whocarried an ε4 allele of the apolipoprotein E gene (which is agenetic risk factor for other cognitive disorders, includingAlzheimer’s disease) scored lower on cognitive tests thanthose without an ε4 allele. However, to our knowledge,these results have not been replicated. Higher dose of che-motherapy, whether we are talking about dose intensity [3]or cumulative dose [2, 40••] is a well-established risk factor.Most prospective studies find no association between cog-nitive and menopausal status, although Jenkins et al. [88]reported that BC patients who experienced a chemotherapy-induced menopause were more likely to show cognitivedecline over the course of treatment than those who didnot. As noted, most studies fail to find a correlation betweencognitive change and psychological factors, such as depres-sion, anxiety, and fatigue [26, 33]. The differential riskassociated with specific chemotherapeutic agents and regi-mens is not well characterized as yet. This awaits largermultisite studies with sufficient sample sizes to provideadequate power for this type of subgroup analysis.

Conclusions and Future Directions

Although complaints of “chemo fog” have existed since theinstitution of systemic adjuvant chemotherapy, it is only inthe last 15–20 years, with increasing use of adjuvant che-motherapy and a growing cohort of cancer survivors with afocus on quality of life, that we have seen serious systematic

Curr Oncol Rep (2013) 15:260–269 265

study of this problem. It has proved to be a rather elusivephenomenon—it seems that only a subgroup of patients areaffected and the cognitive changes are generally subtle. As aresult, methodological variations from study to study cangreatly influence whether or not cognitive changes aredetected. Furthermore, there are a host of other factors thatcan influence cognition in cancer patients—anxiety, mooddisturbance, other medications, and biological responses tothe disease itself—and confound the study of CRCI. In spiteof this, with increasing collaboration among researchers andresulting refinement and harmonization of study methods,the “fog” surrounding chemo fog is beginning to clear.

The preponderance of studies shows that cancer patientsare at increased risk of mild cognitive impairment. In manyinstances, abnormalities can be detected even prior to com-mencement of systemic adjuvant treatment, suggesting thatfactors other than chemotherapy contribute to these cogni-tive changes. However, a prospective study that controlledfor practice effects indicated that about one third of BCpatients, the most widely studied population, show signifi-cant cognitive decline over the course of treatment. Al-though many of these women still score within normallimits on neuropsychological testing after chemotherapy,there is now some evidence to suggest that the decline issufficient to affect quality of life (e.g., return to work) forsome individuals. Given the impossibility of doing random-ized controlled trials in this field, the type and duration oftreatment are inextricably confounded with host and diseasecharacteristics, making it very difficult to establish whetherchemotherapy is truly a causative factor. However, datafrom animal studies and from imaging studies, along withevidence of a dose–response relationship between chemo-therapy and cognitive disturbance, strongly indicate thatsystemic chemotherapy can be neurotoxic and that its effectson the central nervous system do contribute to the cognitivechanges. Psychological distress—long thought to be respon-sible for the cognitive changes in cancer patients—does notprove to be significantly related to the neuropsychologicalchanges. Longitudinal studies that extend beyond the end oftreatment suggest that, in most cases, CRCI abates over time[77, 89]. However, both cross-sectional and longitudinalstudies indicate that there may be a small subgroup ofpatients who experience more chronic, perhaps even perma-nent, cognitive changes. The risk factors for this are still notwell understood but may include chemotherapy dose andneural reserve. At this time, there is no evidence to suggestthat chemotherapy exposure increases the risk of developingdementia later in life.

An understanding of the potential cognitive side effectsof cancer treatment is critical to making informed decisionsabout treatment, particularly for patients with a good prog-nosis for whom adjuvant chemotherapy minimally reducesthe risk of disease-free survival. It is, moreover, important

that we screen for CRCI in our patients so that we canprovide education, support, reassurance, and interventionfor those patients who are affected. Future studies shouldaim to provide a better understanding of the relative neuro-toxicity of specific chemotherapeutic agents and regimensused in the adjuvant setting as well as host factors whichplace a given individual at risk of developing cognitive sideeffects, as such information can be used to personalizetreatment recommendations. Further study of the mecha-nisms of neurotoxicity is also a priority for future research,as this will guide the development of neuroprotective andpreventive strategies. Ultimately, we will seek to developnew chemotherapeutic drugs with minimal neurotoxicityand, towards this aim, we should consider routinely includ-ing cognitive testing as a secondary outcome in chemotherapydrug trials.

Conflict of Interest Erin O’Farrell declares no conflict of interest.Joyce MacKenzie declares no conflict of interest.Barbara Collins declares no conflict of interest.

References

Papers of particular interest, published recently have beenhighlighted as:• Of importance•• Of major importance

1. Boykoff N, Moieni M, Subramanian SK. Confronting chemobrain:an in-depth look at survivors’ reports of impact on work, socialnetworks, and health care response. J Cancer Surviv. 2009;3:323–32.

2. Wieneke MH, Dienst ER. Neuropsychological assessment of cog-nitive functioning following chemotherapy for breast cancer.Psychooncology. 1995;4:61–6.

3. van Dam FS, Schagen SB, Muller MJ, et al. Impairment of cogni-tive function in women receiving adjuvant treatment for high-riskbreast cancer: high-dose versus standard-dose chemotherapy. JNatl Cancer Inst. 1998;90:210–8.

4. Early Breast Cancer Trialists’ Collaborative Group (EBCTCG).Effects of chemotherapy and hormonal therapy for early breastcancer on recurrence and 15-year survival: an overview of therandomised trials. Lancet. 2005;365:1687–717.

5. Maki P, Hogervorst E. HRT and cognitive decline. Best Pract ResClin Endocrinol Metab. 2003;17:105–22.

6. Sherwin BB. Estrogen and cognitive functioning in women: les-sons we have learned. Behav Neurosci. 2012;126:123–7.

7. Bender CM, Sereika SM, Berga SL, et al. Cognitive impairmentassociated with adjuvant therapy in breast cancer. Psychooncology.2006;15:422–30. doi:10.1002/pon.964.

8. Bender CM, Sereika SM, Brufsky AM, et al. Memory impairmentswith adjuvant anastrozole versus tamoxifen in women with early-stage breast cancer. Menopause. 2007;14:995–8.

9. Castellon SA, Ganz PA, Bower JE, et al. Neurocognitive perfor-mance in breast cancer survivors exposed to adjuvant chemother-apy and tamoxifen. J Clin Exp Neuropsychol. 2004;26:955–69.

266 Curr Oncol Rep (2013) 15:260–269

10. Collins B, MacKenzie J, Stewart A, et al. Cognitive effects ofhormonal therapy in early-stage breast cancer patients: a prospec-tive study. Psychooncology. 2009;18:811–21.

11. Eberling JL, Wu C, Tong-Turnbeaugh R, et al. Estrogen- andtamoxifen-associated effects on brain structure and function.NeuroImage. 2004;21:364–71.

12. Falleti MG, Sanfilippo A, Maruff P, et al. The nature and severityof cognitive impairment associated with adjuvant chemotherapy inwomen with breast cancer: a meta-analysis of the current literature.Brain Cogn. 2005;59:60–70.

13. Jenkins V, Shilling V, Fallowfield L, et al. Does hormone therapyfor the treatment of breast cancer have a detrimental effect onmemory and cognition? A pilot study. Psychooncology. 2004;13:61–6.

14. Palmer JL, Trotter T, Joy AA, et al. Cognitive effects of tamoxifenin premenopausal women with breast cancer compared to healthycontrols. J Cancer Surviv. 2008;2:275–82.

15. Phillips KA, Aldridge J, Ribi K, et al. Cognitive function in post-menopausal breast cancer patients one year after completing adju-vant endocrine therapy with letrozole and/or tamoxifen in the BIG1-98 trial. Breast Cancer Res Treat. 2011;126:221–6.

16. Schilder CM, Seynaeve C, Beex LV, et al. Effects of tamoxifen andexesestance on cognitive functioning of post-menopausal patientswith breast cancer: results from the neuropsychological side studyof the Tamoxifen and Exesestance Adjuvant Multinational Trial. JClin Oncol. 2010;28:1294–300.

17. Shilling V, Jenkins V, Fallowfield L, et al. The effects of hormonetherapy on cognition in breast cancer. J Steroid Biochem Mol Biol.2003;86:405–12.

18. Vardy J, Wefel JS, Ahles T, et al. Cancer and cancer-relatedcognitive dysfunction: an international perspective from the Ven-ice cognitive workshop. Ann Oncol. 2008;19:623–9.

19. • Wefel JS, Vardy J, Ahles T, et al. International Cognition andCancer Task Force recommendations to harmonise studies of cogni-tive function in patients with cancer. Lancet Oncol. 2011;12:703–8.An international task force was formed to establish guidelines forresearch in the area of cognition and cancer.

20. Cimprich B, Reuter-Lorenz P, Nelson J, et al. Pre-chemotherapyalterations in brain function in women with breast cancer. J ClinExp Neuropsychol. 2010;32:324–31.

21. Schilder CM, Seynaeve C, Linn SC, et al. The impact of differentdefinitions and reference groups on the prevalence of cognitiveimpairment: a study in postmenopausal breast cancer patientsbefore the start of adjuvant systemic therapy. Psychooncology.2010;19:415–22.

22. Wefel JS, Lenzi R, Theriault R, et al. “Chemobrain” in breastcarcinoma?A prologue. Cancer. 2004;101:466–75.

23. Stewart A, Collins B, Mackenzie J, et al. The cognitive effects ofadjuvant chemotherapy in early stage breast cancer: a prospectivestudy. Psychooncology. 2008;17:122–30.

24. Shilling V, Jenkins V, Trapala IS. The (mis)classification ofchemo-fog – methodological inconsistencies in the investigationof cognitive impairment after chemotherapy. Breast Cancer ResTreat. 2006;95:125–9.

25. Ouimet LA, Stewart A, Collins B, et al. Measuring neuropsycho-logical change following breast cancer treatment: an analysis ofstatistical models. J Clin Exp Neuropsychol. 2009;31:73–89.

26. Jansen CE, Cooper BA, Dodd MJ, et al. A prospective longitudinalstudy of chemotherapy-induced cognitive changes in breast cancerpatients. Support Care Cancer. 2011;19:1647–56.

27. Biglia N, Bounous VE, Malabaila A, et al. Objective and self-reported cognitive dysfunction in breast cancer women treatedwith chemotherapy: a prospective study. Eur J Cancer Care.2012;21:485–92.

28. Cheung YT, Tan EH, Chan A. An evaluation on the neuropsycho-logical tests used in the assessment of postchemotherapy cognitive

changes in breast cancer survivors. Support Care Cancer.2012;20:1361–75.

29. Hutchinson AD, Hosking JR, Kichenadasse G, et al. Objectiveand subjective cognitive impairment following chemotherapyfor cancer: a systematic review. Cancer Treat Rev. 2012;38:926–34.

30. Pullens MJJ, DeVries J, Roukema JA. Subjective cognitive dysfunc-tion in breast cancer patients: a systematic review. Psychooncology.2010;19:1127–38.

31. Iconomou G, Mega V, Koutras A, et al. Prospective assessment ofemotional distress, cognitive function and quality of life in patientswith cancer treated with chemotherapy. J Cancer. 2004;101:404–11.

32. Ingraham LJ, Aiken CB. An empirical approach to determiningcriteria for abnormality in test batteries with multiple measures.Neuropsychology. 1996;10:120–4.

33. Wefel JS, Schagen SB. Chemotherapy-related cognitive dysfunc-tion. Curr Neurol Neurosci Rep. 2012;12:267–75.

34. Scherling C, Collins B, MacKenzie J, et al. Pre-chemotherapydifferences in visuo-spatial working memory in breast cancerpatients compared to controls: an fMRI study. Front HumNeurosci. 2011;5:122. doi:10.3389/fnhum.2011.00122.

35. Scherling C, Collins B, MacKenzie J, et al. Pre-chemotherapydifferences in response inhibition in breast cancer patients com-pared to controls: a functional magnetic resonance imaging study. JClin Exp Neuropsychol. 2012;34:543–60.

36. Scherling C, Collins B, MacKenzie J, et al. Structural brain differ-ences in breast cancer patients compared to matched controls priorto chemotherapy. Int J Biol. 2012;4:3–25.

37. Hurria A, Somlo G, Ahles T. Renaming “chemobrain”. CancerInvest. 2007;25:373–7.

38. •• Deprez S, Amant F, Smeets A, et al. Longitudinal assessment ofchemotherapy-induced structural changes in cerebral white matterand its correlation with impaired cognitive functioning. J ClinOncol. 2012;30:274–81. This longitudinal study examined therelationship between changes in neuropsychological functioningand brain structure following chemotherapy and found significantcorrelations between these two measures.

39. Ganz PA. Doctor, will the treatment you are recommending causechemobrain? J Clin Oncol. 2012;30:229–31.

40. •• Collins B, MacKenzie J, Tasca GA, et al. Cognitive effects ofchemotherapy in breast cancer patients: a dose-response study.Psychooncology. 2012. doi:10.1002/pon.3163. This is a prospec-tive longitudinal study of cognitive functioning after cumulativeexposure to chemotherapy. Patients were assessed at the baselineand following each chemotherapy cycle. A linear decline in cog-nitive functioning was observed, supporting a dose–response rela-tionship and suggesting that chemotherapy is a cause of cognitivedisturbance.

41. Hedayati E, Alinaghizadeh H, Schedin A, et al. Effects of adjuvanttreatment on cognitive function in women with early breast cancer.Eur J Oncol Nurs. 2012;16:315–22.

42. Kreukels BPC, Schagen SB, Ridderinkhof KR, et al. Electrophys-iological correlates of information processing in breast-cancerpatients treated with adjuvant chemotherapy. Breast Cancer ResTreat. 2005;94:53–61. doi:10.1007/s10549-005-7093-3.

43. Schagen SB, Hamburger HL, Muller MJ, et al. Neurophysiologicalevaluation of late effects of adjuvant high-dose chemotherapy oncognitive function. J Neurooncol. 2001;51:159–65.

44. Schagen SB, Muller MJ, Boogerd W, et al. Change in cognitivefunction after chemotherapy: a prospective longitudinal study inbreast cancer patients. J Natl Cancer Inst. 2006;98:1742–5.doi:10.1093/jnci/djj470.

45. Ahles TA, Saykin AJ, Furstenberg T, et al. Neuropsychologic impactof standard-dose systemic chemotherapy in long-term survivors ofbreast cancer and lymphoma. J Clin Oncol. 2002;20:485–93.doi:10.1200/JCO.20.2.485.

Curr Oncol Rep (2013) 15:260–269 267

46. Brown MS, Simon JH, Stemmer SM, et al. MR and proton spec-troscopy of white matter disease induced by high-dose chemother-apy with bone marrow transplant in advanced breast carcinoma.Am J Neuroradiol. 1995;16:2013–20.

47. Brown MS, Stemmer SM, Simon JH, et al. White matter diseaseinduced by high-dose chemotherapy: longitudinal study withMR imaging and proton spectroscopy. Am J Neuroradiol.1998;19:217–21.

48. Stemmer SM, Stears JC, Burton BS, et al. White matter changes inpatients with breast cancer treated with high-dose chemotherapyand autologous bone marrow support. Am J Neuroradiol.1994;15:1267–73.

49. Seigers R, Fardell JE. Neurobiological basis of chemotherapy-induced cognitive impairment: a review of rodent research.Neurosci Biobehav Rev. 2011;35:729–41.

50. Winocur G, Henkelman M, Wojtowicz JM, et al. The effects ofchemotherapy on cognitive function in a mouse model: a prospec-tive study. Clin Cancer Res. 2012;18:3112–21.

51. de Ruiter MB, Reneman L, Boogerd W, et al. Cerebralhyporesponsiveness and cognitive impairment 10 years after che-motherapy for breast cancer. Hum Brain Mapp. 2011;32:1206–19.doi:10.1002/hbm.21102.

52. Kesler SR, Kent JS, O’Hara R. Prefrontal cortex and executivefunction impairments in primary breast cancer. Arch Neurol.2011;68:1447–53.

53. Silverman DH, Dy CJ, Castellon SA, et al. Altered fronto-cortical,cerebellar and basal ganglia activity in adjuvant-treated breastcancer survivors 5–10 years after chemotherapy. Breast CancerRes Treat. 2007;103:303–11.

54. Abraham J, Haut MW, Moran MT, et al. Adjuvant chemotherapyfor breast cancer: effects on cerebral white matter seen in diffusiontensor imaging. Clin Breast Cancer. 2008;8:88–91.

55. de Ruiter MB, Reneman L, Boogerd W, et al. Late effects ofhigh-dose adjuvant chemotherapy on white and gray matter inbreast cancer survivors: converging results from multimodal magnet-ic resonance imaging. Hum Brain Mapp. 2012;33:2971–83.doi:10.1002/hbm.21422.

56. Deprez S, Amant F, Yigit R, et al. Chemotherapy-induced struc-tural changes in cerebral white matter and its correlation withimpaired cognitive functioning in breast cancer patients. HumBrain Mapp. 2011;32:480–93.

57. Ferguson RJ, McDonald BC, Saykin AJ, et al. Brain structure andfunction differences in monozygotic twins: possible effects ofbreast cancer chemotherapy. J Clin Oncol. 2007;25:3866–70.

58. Inagaki M, Yoshikawa E, Matsuoka Y, et al. Smaller regionalvolumes of brain gray and white matter demonstrated in breastcancer survivors exposed to adjuvant chemotherapy. Cancer.2007;109:146–56.

59. Koppelmans V, de Ruiter MB, van der Lijn F, et al. Global andfocal brain volume in long-term breast cancer survivors exposed toadjuvant chemotherapy. Breast Cancer Res Treat. 2012;132:1099–106.

60. •• McDonald BC, Conroy SK, Ahles TA, et al. Gray matterreduction associated with systemic chemotherapy for breast can-cer: a prospective MRI study. Breast Cancer Res Treat.2010;123:819–28. This was the first controlled prospective MRIstudy in the field of CRCI.

61. Lopez-Zunini RA, Scherling C, Wallis N, et al. Differences inverbal memory retrieval in breast cancer chemotherapy patientscompared to healthy controls: a prospective fMRI study. BrainImaging Behav 2012. doi:10.1007/s11682-012-9213-0.

62. Jansen CE, Dodd MJ, Miaskowski CA, et al. Preliminary results ofa longitudinal study of changes in cognitive function in breastcancer patients undergoing chemotherapy with doxorubicin andcyclophosphamide. Psychooncology. 2008;17:1189–95. doi:10.1002/pon.1342.

63. Ahles TA, Saykin AJ, McDonald BC, et al. Longitudinal assess-ment of cognitive changes associated with adjuvant treatment forbreast cancer: impact of age and cognitive reserve. J Clin Oncol.2010;28:4434–40. doi:10.1200/JCO.2009.27.0827.

64. Kvale EA, Clay OJ, Ross-Meadows LA, et al. Cognitive speed ofprocessing and functional declines in older cancer survivors: ananalysis of data from the ACTIVE trial. Eur J Cancer Care.2010;19:110–7. doi:10.1111/j.1365-2354.2008.01018.x.

65. Quesnel C, Savard J, Ivers H. Cognitive impairments associatedwith breast cancer treatments: results from a longitudinal study.Breast Cancer Res Treat. 2009;116:113–23. doi:10.1007/s10549-008-0114-2.

66. Tager FA, McKinley PS, Schnabel FR, et al. The cognitiveeffects of chemotherapy in post-menopausal breast cancerpatients: a controlled longitudinal study. Breast Cancer ResTreat. 2010;123:25–34.

67. Vearncombe KJ, Rolfe M, Wright M, et al. Predictors ofcognitive decline after chemotherapy in breast cancer patients.J Int Neuropsychol Soc. 2009;15:951–62. doi:10.1017/S1355617709990567.

68. Jansen CE, Miaskowski C, Dodd M, et al. A meta-analysis ofstudies of the effects of cancer chemotherapy on various domainsof cognitive function. Cancer. 2005;104:2222–33.

69. Jansen CE, Miaskowski CA, Dodd MJ, et al. A meta-analysis ofthe sensitivity of various neuropsychological tests used to detectchemotherapy-induced cognitive impairments in patients withbreast cancer. Oncol Nurs Forum. 2007;34:997–1005.

70. • Jim HSL, Phillips KM, Chait S, et al. Meta-analysis of cognitivefunctioning in breast cancer survivors previously treated withstandard-dose chemotherapy. J Clin Oncol. 2012;30:3578–87. Thisis a meta-analysis of 17 studies examining the long-term effects ofchemotherapy on cognitive functioning. The results indicate cog-nitive deficits of small effect size in patients treated with chemo-therapy as compared with healthy controls and patients not treatedwith chemotherapy.

71. Stewart A, Bielajew C, Collins B, et al. A meta-analysis of theneuropsychoogical effects of adjuvant chemotherapy treatment inwomen treated for breast cancer. Clin Neuropsychol. 2006;20:76–89.

72. Anderson-Hanley C, Sherman ML, Riggs R, et al. Neuropsycho-logical effects of treatments for adults with cancer: a meta-analysisand review of the literature. J Int Neuropsychol Soc. 2003;9:967–82.

73. Kopplemans V, Breteler MMB, Boogerd W, et al. Neuropsycho-logical performance in survivors of breast cancer more than20 years after adjuvant chemotherapy. J Clin Oncol. 2012;30:1080–6.

74. Kreukels BPC, Schagen SB, Ridderinkhof KR, et al. Effects ofhigh-dose and conventional-dose adjuvant chemotherapy on long-term cognitive sequelae in patients with breast cancer: an electro-physiologic study. Clin Breast Cancer. 2006;7:67–78.

75. Kreukels BPC, van Dam FSAM, Ridderinkhof KR, et al. Persistentneurocognitive problems after adjuvant chemotherapy for breastcancer. Clin Breast Cancer. 2008;8:80–7.

76. Kreukels BPC, Hamburger HL, de Ruiter MB, et al. ERP ampli-tude and latency in breast cancer survivors treated with adjuvantchemotherapy. Clin Neurophysiol. 2008;119:533–41.

77. Collins B, Mackenzie J, Stewart A, et al. Cognitive effects ofchemotherapy in post-menopausal breast cancer patients 1 yearafter treatment. Psychooncology. 2009;18:134–43.

78. Weis J, Poppelreuter M, Bartsch HH. Cognitive deficits as long-term side-effects of adjuvant therapy in breast cancer patients:subjective complaints and objective neuropsychological test re-sults. Psychooncology. 2009;18:775–82. doi:10.1002/pon.1472.

79. Wefel JS, Saleeba AK, Buzdar AU, et al. Acute and late-onsetcognitive dysfunction associated with chemotherapy in womenwith breast cancer. Cancer. 2010;116:3348–56. doi:10.1002/cncr.25098.

268 Curr Oncol Rep (2013) 15:260–269

80. Heflin LH, Meyerowitz BE, Hall P, et al. Cancer as a risk factor forlong-term cognitive deficits and dementia. J Natl Cancer Inst.2005;97:854–6.

81. Heck JE, Albert SM, Franco R, et al. Patterns of dementiadiagnosis in surveillance, epidemiology, and end results breastcancer survivors who use chemotherapy. J Am Geriatr Soc.2008;56:1687–92.

82. Baxter NN, Durham SB, Phillips KA, et al. Risk of dementia inolder breast cancer survivors: a population-based cohort study ofthe association with adjuvant chemotherapy. J Am Geriatr Soc.2009;57:403–11.

83. Du XL, Xia R, Hardy D. Relationship between chemotherapy useand cognitive impairments in older women with breast cancer. AmJ Clin Oncol. 2010;33:533–43.

84. Mols F, Thong MSY, Vreugdenhil G, et al. Long-term cancersurvivors experience work changes after diagnosis: results of apopulation-based study. Psychooncology. 2009;18:1252–60.

85. Nieuwenhuijsen K, de Boer A, Spelten E, et al. The role ofneuropsychological functioning in cancer survivors’ return to workone year after diagnosis. Psychooncology. 2009;18:589–97.

86. Reid-Arndt SA, Yee A, Perry MC, et al. Cognitive andpsychological factors associated with early post-treatmentfunctional outcomes in breast cancer survivors. J PsychosocOncol. 2009;27:415–34.

87. Ahles TA, Saykin AJ, Noll WW, et al. The relationship of ApoEgenotype to neuropsychological performance in long-term cancersurvivors treated with standard dose chemotherapy. Psychooncology.2003;12:612–9.

88. Jenkins V, Shilling V, Deutsch G, et al. A 3-year prospective studyof the effects of adjuvant treatments on cognition in women withearly stage breast cancer. Br J Cancer. 2006;94:828–34.

89. Schagen SB, Muller MJ, Boogerd W, et al. Late effects of adjuvantchemotherapy on cognitive function: a follow-up study in breastcancer patients. Ann Oncol. 2002;13:1387–97.

Curr Oncol Rep (2013) 15:260–269 269