Abstract
Over the past few decades, a body of research has emerged confirming what many adult patients with noncentral nervous system cancer have long reported—that cancer and its treatment are frequently associated with cancer-related cognitive impairment (CRCI). The severity of CRCI varies, and symptoms can emerge early or late in the disease course. Nonetheless, CRCI is typically mild to moderate in nature and primarily involves the domains of memory, attention, executive functioning, and processing speed. Animal models and novel neuroimaging techniques have begun to unravel the pathophysiologic mechanisms underlying CRCI, including the role of inflammatory cascades, direct neurotoxic effects, damage to progenitor cells, white matter abnormalities, and reduced functional connectivity, among others. Given the paucity of research on CRCI with other cancer populations, this review synthesizes the current literature with a deliberate focus on CRCI within the context of breast cancer. A hypothetical case-study approach is used to illustrate how CRCI often presents clinically and how current science can inform practice. While the literature regarding intervention for CRCI is nascent, behavioral and pharmacologic approaches are discussed.
Keywords: chemotherapy, psychological/behavioral oncology, breast neoplasms, complications and late effects of therapy
Introduction
Advances in diagnosis and treatment of cancer have greatly improved survival. With reduced mortality, morbidity related to cancer and its treatment has garnered increased attention, and issues surrounding quality of life have become ever more important. Cancer survivors have long reported cognitive dysfunction at various stages of the disease course with associated consequences upon well-being and functional independence. Nonetheless, until relatively recently, cancer-related cognitive impairment (CRCI) in patients with noncentral nervous system (non-CNS) malignancies was largely unacknowledged.1 The prevailing attitude was reinforced by the belief that chemotherapies were unable to cross the blood-brain barrier,2–4 precluding the possibility of a direct neurotoxic effect of cancer therapies. However, since the 1990s, a growing body of literature has verified the existence of CRCI, with recent animal models and neuroimaging studies uncovering pathophysiologic correlates.4–14
CRCI research has largely focused on neurotoxicity associated with chemotherapy, often referred to as “chemobrain” or “chemofog.”5 However, CRCI has also been documented in the absence of chemotherapy, leading to hypothesized associations with cancer itself,15 surgery,15–18 and other adjuvant therapies.19–21 Estimates of the prevalence of CRCI vary widely, although current longitudinal studies suggest that approximately 40% of cancer patients have evidence of CRCI before any treatment, up to 75% may have cognitive decline during treatment, and up to 60% exhibit deterioration in cognition even after completion of therapies.5,22–25 The pattern of CRCI differs across patients and disease course, although severity is typically mild to moderate in nature. Mild cognitive impairments are conventionally considered to be performances that are from −1.5 to −2 standard deviations below population normative means. However, mild to moderate CRCI may also refer to a psychometrically significant decline relative to a patient’s own pretreatment baseline performance (ie, a decline of approximately 1–2 normative standard deviations from baseline scores). Accordingly, these methods of determining impairment yield some difference in absolute impairment levels, depending on the individual patient’s premorbid level of function. Nonetheless, the severity of CRCI is generally milder than the cognitive impairment typical of common neurologic populations, including those with neurodegenerative diseases and stroke. Despite this, encephalopathies involving dementia have been observed in the context of treatment with some cytotostatic agents.26,27 In addition, CRCI can persist for months to years after treatment,28 and even subtle impairments can have profound consequences upon quality of life, including occupational and social functioning.1
To date, the majority of CRCI research in patients with non-CNS cancer has involved women with breast cancer,29,30 who represent approximately 22% of the 14.5 million cancer survivors in the United States alone.31 Investigations have also been conducted in patients with testicular cancer,32,33 lymphoma,28 multiple myeloma,34 colorectal cancer,35 ovarian cancer,36 and prostate cancer,37 among others. However, much of the literature regarding these populations is preliminary, with studies mostly consisting of small sample sizes. Future large, longitudinal, cohort studies are needed to better describe the prevalence and nature of CRCI in these patient populations. In light of the current state of the literature, this review deliberately focuses on the findings from clinical and basic research on CRCI in adult patients with breast cancer or preclinical models thereof, which has rapidly grown over the past few decades. To illustrate this work, the cognitive functioning of a hypothetical breast cancer patient is described at various stages of her disease and treatment course, based upon published reports and our collective clinical experience. These clinical descriptions are supplemented by summaries of associated empirical findings and discussion of pathophysiological underpinnings and potential intervention strategies.
Breast Cancer, Surgery, and Cognitive Functioning
The patient is a 53-year-old, married woman employed as an attorney in a busy corporate law firm. Upon self-examination, she noticed a mass in the outer upper quadrant of her right breast, prompting medical consultation. At the time of presentation, she reported that menopause occurred at age 50 years. She underwent lumpectomy with axillary lymph node dissection, and pathology revealed stage II, infiltrating, estrogen receptor-positive and progesterone receptor-positive (ER+/PR+) ductal carcinoma that was negative for human epidermal growth factor receptor 2 (HER2/neu). Her adjuvant treatment plan was discussed at her follow-up visit, including the role of chemotherapy. She stated that she had read about “chemobrain” and expressed concerns about treatment-related changes in her cognitive functioning. She was referred for formal neuropsychological evaluation by her oncologist to establish a baseline for monitoring her cognition throughout treatment. Her performances on neurocognitive testing were notable for subtle inefficiencies with attention and processing speed, although her endorsements on self-reported measures reflected greater cognitive complaints than objective cognitive impairments identified on neuropsychological testing. Inventories of emotional functioning revealed mild to moderate anxiety and future uncertainty.
In practice, patients rarely present to clinical neuropsychologists before the initiation of adjuvant therapy, although research suggests that the clinical picture described above is not uncommon. On objective testing before the initiation of chemotherapy, the patient evidenced some cognitive weaknesses, which may have been present to some extent even before surgery. While rates vary according to the cognitive instruments and impairment criteria used, studies assessing women with breast cancer before chemotherapy have documented that up to 40% exhibit CRCI before the initiation of therapy.23,38 Some data suggest that CRCI is most commonly observed within the cognitive domains of learning and memory,18 although others report a more nonspecific pattern of impairments.24
In the above example, the patient’s self-reported cognitive complaints exceeded the severity of deficits noted on objective testing. Such discrepancies between objective and subjective measures of cognitive functioning have been documented in patients with various neurological illnesses,39–41 which is also an area of ongoing research in patients with non-CNS CRCI.42–45 While evidence suggests that cognitive complaints do in fact bear some relationship to objective cognitive performance,42 self-reported cognitive symptoms appear to be more strongly associated with affective symptoms (eg, depression and anxiety), coping and adjustment issues, and cancer-related fatigue.43 This highlights the importance of comprehensive assessment of cognitive and emotional functioning using a battery of measures that captures not only objective cognitive performances but also patient-reported symptoms of CRCI and emotional functioning.
Pathophysiological Underpinnings
Pretreatment CRCI identified on objective testing appears to be independent of emotional distress and fatigue,28,46 medical comorbidities,47 and surgical factors,48 although no consistent explanation has been found for CRCI before the initiation of therapies. Hypothesized mechanisms have been proposed at a variety of levels, including the biology of cancer itself (eg, inflammatory responses triggering neurotoxic proinflammatory cytokine cascades) and shared underlying risk factors for the development of cancer and cognitive decline (eg, poor DNA repair mechanisms linked to both cancer and neurodegenerative disorders).4,49 A recent rodent study demonstrated that a non-CNS tumor alone is sufficient to induce hippocampal dysfunction, possibly by reducing the rate of neurogenesis and the levels of BDNF (brain-derived neurotrophic factor) and COX-2 (cyclooxygenase 2), in addition to increasing stress-related parameters and circulating levels of proinflammatory cytokines.50 However, patients participating in pretreatment cognitive studies do not likely represent a random sample of individuals confronted with cancer. Thus, it is possible that these samples are biased toward individuals with impairment. Confirmation of the observation of pretreatment CRCI in an unbiased sample is critical and will help disentangle the possible underlying pathophysiology.
Chemotherapy, Endocrine Therapy, and Cognition
After lumpectomy, the patient received 4 cycles of standard-dose adjuvant chemotherapy with doxorubicin and cyclophosphamide as well as weekly paclitaxel for 12 weeks. This was followed by locoregional radiation therapy. Throughout chemotherapy, the patient reported worsening fatigue and cognitive complaints, including greater difficulty with memory, trouble multitasking, and distractibility. She stated that her symptoms negatively impacted daily functions, as she occasionally forgot doses of medications and missed bill payments. Given the ER+/PR+ subtype of carcinoma and postmenopausal status, chemotherapy was followed by 5 years of endocrine therapy with anastrozole to reduce the risk of recurrence. Worsening in learning and memory was reported after the initiation of endocrine therapy and had an even greater adverse impact on her work functioning. Serial neuropsychological assessments were conducted after chemotherapy and again after 6 months of endocrine therapy. The results demonstrated postchemotherapy decline on measures of attention, processing speed, learning, and memory as well as executive functions, including mental flexibility. During endocrine therapy, performances on testing were largely stable, although slight worsening in learning and memory was noted. Mood assessment evidenced mild depression and continued anxiety.
A common challenge for both clinical assessment and cross-sectional research involves the reliance on normative comparison groups. Without pretreatment neuropsychological assessment for comparison, relative declines in cognitive functioning across the pretreatment to posttreatment interval may be undetected or underestimated. That is, patients may have declined during treatment despite post-treatment performances falling within the “normal” to “low normal” range compared with normative data. This is a particularly important consideration when interpreting the test results of individuals with high levels of preillness functioning (eg, being well-educated), which presumably are indicative of greater cognitive reserve.22 Unlike the situation faced by most neuropsychologists in the clinic, the patient in the above example had repeated neuropsychological evaluations, allowing the direct comparison of her pre-treatment and posttreatment cognitive test performances. With the use of modern statistical approaches (eg, reliable change indices) supplementing clinical analysis and inference, normal variation and treatment-related cognitive change can be distinguished with adequate confidence.5
In the case example, significant postchemotherapy declines were found across several cognitive domains. This phenomenon has been increasingly recognized in both the scientific literature and the clinic. Over the past decade, more than 20 longitudinal studies have been conducted involving both prechemotherapy and postchemotherapy assessment of cognition,5,7,11,13–15,18,23–25,29,32,46,51–67 with nearly 70% of investigations reporting evidence of significant decline after therapy.29 The incidence of CRCI after chemotherapy has been observed in 17% to over 70% of patients in longitudinal studies. Breast cancer patients treated with 5-fluorouracil, doxorubicin, and cyclophosphamide (FAC), a regimen similar to that received by the hypothetical patient, tend to show the most prominent decline in the cognitive domains of learning and memory, attention, and processing speed.52 While CRCI within the context of chemotherapy is often mild to moderate in severity, several agents have been associated with severe encephalopathy and dementia in some patients, including methotrexate, BCNU (bis-chloroethylnitrosurea [carmustine]), fludarabine, cytarabine, 5-fluorouracil, and cisplatin.29
Specific risks for CRCI and patterns of CRCI by regimen are largely unknown. Nonetheless, considerable evidence suggests a dose-response relationship between chemotherapy and CRCI.68–70 Collins and colleagues conducted neuropsychological assessments in 60 women with early stage breast cancer before the initiation of chemotherapy but after surgery, as well as after the completion of each cycle of chemotherapy.70 Compared with matched healthy controls, progressive decline was noted at each subsequent time point, supporting a dose-response relationship in which patients demonstrated a linear worsening of CRCI with each cycle of chemotherapy. In addition, CRCI associated with chemotherapy may emerge at various stages of the disease course. In a longitudinal investigation of 42 patients with breast cancer receiving FAC (with or without paclitaxel), Wefel et al found that 21% of patients exhibited CRCI before the initiation of chemotherapy, 65% exhibited cognitive decline during or shortly after therapy, and 61% showed decline at approximately 1 year after the completion of treatment.25 Of this 61%, a substantial subgroup demonstrated cognitive deterioration that was not previously apparent.
In our case example, slight worsening in cognitive functioning was noted during endocrine therapy. In contrast to the literature regarding chemotherapy and CRCI, few rigorous investigations have been conducted examining the impact of endocrine therapy on cognition. Nonetheless, some research suggests that treatment with selective ER modulators (SERMs) is associated with cognitive deterioration.8,21 For instance, treatment with tamoxifen has been associated with significantly reduced performances on measures of memory, verbal fluency, visuospatial functioning, and processing speed,71 and those receiving both chemotherapy and tamoxifen may exhibit greater impairment than those treated with either alone.8 Aromatase inhibitors, such as anastrozole in the case example, may also contribute to CRCI,72,73 although the literature is inconsistent.20,74 Evidence also suggests that the cognitive effects of aromatase inhibitors may be less than those of SERMs.75 However, it is important to note that patients are often maintained on endocrine therapy for long durations, with some remaining on treatment for up to 10 years per recent American Society of Clinical Oncology guidelines.76 As such, it is possible that CRCI associated with hormonal therapy may emerge over time, although large, well-controlled longitudinal studies are needed.
The patient we describe above reported some occupational difficulties and distress. While problems performing instrumental activities of daily living (eg, medication and financial management, occupational functioning) are associated with cognitive impairment in neurological populations,77 these relationships are not as well studied in patients who have CRCI associated with non-CNS cancer. Nonetheless, some evidence suggests that acquired executive deficits (eg, difficulty planning and multitasking) contribute to reductions in community involvement and social functioning78 as well as more general degradation of quality of life.79 Those who exhibit postchemotherapy decline in cognition also tend to report greater occupational difficulties than those who remain stable throughout treatment,52 with approximately 13% of cancer survivors ceasing to work within 4 years of diagnosis secondary to “cancer-related reasons,” including CRCI.80
Late Effects of Cancer Therapy
As evidenced by Wefel and colleagues, CRCI can be persistent and may even emerge in the months to years after completion of all therapies.25 Koppelmans et al investigated the neuropsychological functioning of patients with breast cancer who received an average of 6 cycles of chemotherapy with cyclophosphamide, methotrexate, and fluorouracil (CMF) and had a mean of 21 years since completion of treatment.81 Compared with a healthy, population-based comparison group, significantly worse performances were noted in memory, processing speed, and executive functioning, although patients reported less symptoms of depression. However, despite evidence of persistent CRCI in some patients with breast cancer, other subsets of patients have been found to improve in the months to years after cessation of chemotherapy51 and endocrine therapy.13
At present, it is not understood why some patients exhibit decline or persistent CRCI after treatment while others improve or remain stable. Premorbid cognitive capacity may play a role, as greater posttreatment decline in processing speed has been documented in breast cancer patients with lower preillness cognitive capacities.4 Lifestyle and medical comorbidities as well as problems with fatigue and sleep may also represent risk factors for later cognitive decline, although existing studies have not found these to be strong predictors of cognitive decline. Geriatric research suggests that older individuals and carriers of variants of the apolipoprotein E (APOE) gene (eg, APOEε4) are at increased risk of cognitive decline and development of dementia.82,83 Accordingly, it is possible that CRCI may become more pronounced later in life or in individuals with certain genetic vulnerabilities.29 Further research is needed to clarify the risk factors associated with the development and maintenance of CRCI. Greater consistency across studies regarding assessment, criteria for impairment, and analytical methods is required to facilitate the comparison of results across studies and cancer populations. Toward this end, the International Cognition and Cancer Task Force has proposed a useful set of guidelines for the assessment and study of CRCI, providing the groundwork for unification of future investigations.84
Pathophysiological Underpinnings
Preclinical research
Preclinical studies have provided some insights into the pathophysiological mechanisms underlying CRCI associated with nontargeted delivery of chemotherapy.85–88 It has been shown in animal models that cytostatic agents from different classes (eg, antimetabolites, DNA cross-linking agents, and alkylating agents) administered peripherally and in clinically relevant dosages can disrupt various neurobiological processes and induce cognitive impairment.49 A direct toxic effect of chemotherapeutic agents on various cell populations has been proposed as etiology for these neurotoxicities. Specifically, many cytotostatic agents appear to be preferentially toxic to neural progenitor cells (the direct ancestors of all differentiated cell types in the CNS) and postmitotic oligodendrocytes (the myelin-forming cells of the CNS).89,90 Reduced white matter integrity and impaired neurogenesis are also postulated as important mechanisms underlying the typical pattern of CRCI seen in patients.87,89 A recent study in rats demonstrated that cytostatic agents may induce persistent neuroinflammation, which, in turn, is involved in changes in myelination and cognitive dysfunction.91 Other etiologic factors comprise common indirect mechanisms, such as changes in oxidative balance92 (a critical modulator of cellular functions in stem cells and progenitor cells),93 neurotransmitter/monoamine release, and disruption of blood vessel density and blood supply.94–96 While distinguishing between primary and secondary mechanisms is difficult, cytostatic agents likely exert their negative effect on cognition through multiple pathways.
Compared with the rapidly developing literature on the mechanisms of CRCI in the context of chemotherapy, less effort has been directed at understanding the cognitive consequences of endocrine therapies through preclinical experiments and animal models. The results of currently available studies are complex and inconclusive, with large variation across investigations. While preclinical research shows that SERMs like tamoxifen and raloxifene have mainly estrogenic agonistic effects on the serotonergic system after ovariectomy,97,98 they also appear to exhibit neuroprotective functions in the dopamine and acetylcholine systems and to increase plasticity in the hippocampus.98,99 Conversely, rodent studies with aromatase inhibitors like anastrozole and letrozole suggest that impaired estrogen synthesis in the brain is detrimental to the neuroprotective functions of estrogens, leading to impaired long-term potentiation, altered catecholamine levels (depending on the presence or absence of endogenous estrogen), and a decreased number of dendritic spines.100,101 Based on these cellular effects, greater estrogenic effects within the brain would be expected from SERMs, resulting in positive effects upon cognition, including memory. However, preclinical behavioral research shows inconsistent results, ranging from increased anxiety,102 to decreased memory consolidation and retrieval processes,103 or to no effect, and even to improved cognition.104 Interestingly, aromatase inhibitors also facilitate cognitive improvement in rodents, contrary to reports of cognitive decline related to endocrine therapy in patients with breast cancer.101,105
Taken together, there are still large gaps between the preclinical and human data with respect to the effects of endocrine therapies on the brain and cognition. In addition, relatively little attention has been paid to the modeling of individual differences in cognitive vulnerability to cancer and cancer therapies. It is expected that studying the genetic factors that modulate CNS toxicity in preclinical models will be an area of increased interest and challenge in the near future.
Neuroimaging
The final common pathophysiologic pathway is altered neurobiologic status, which results in brain changes and cognitive dysfunction. To date, evidence for this pathway stems primarily from in vivo neuroimaging studies of breast cancer patients and survivors. Volumetric magnetic resonance imaging (MRI) and diffusion tensor imaging (DTI) studies demonstrate widespread reductions in gray matter volume and white matter connectivity.66,106–118 For example, Deprez et al examined white matter connectivity using DTI in 16 patients with breast cancer before chemotherapy and again 3 to 4 months after chemotherapy.110 These patients showed significantly reduced frontal, parietal, and occipital white matter tract integrity over time, whereas patients who did not receive chemotherapy showed no change. The chemotherapy-treated group also demonstrated decreased performance on tests of attention, processing speed, and memory compared with controls, and performance changes were correlated with reductions in white matter integrity.
Altered brain structure, such as changes in gray and/or white matter, reduces the brain network’s ability to stabilize and support the dynamic functional networks that underlie various cognitive processes.119,120 Multiple functional MRI (fMRI) studies have shown altered functional brain activation and connectivity associated with breast cancer and its treatments.16,108,121–134 McDonald and colleagues measured brain activation associated with working memory using fMRI in 16 patients who received chemotherapy, 12 who did not receive chemotherapy, and 15 healthy female controls.132 Participants were evaluated before chemotherapy, 1 month after chemotherapy, and 1 year later or at matched intervals for controls. Compared with controls, patients demonstrated overactivation of frontal regions.132 Overactivation is believed to represent a deficient or inefficient neural response in which additional neural resources are required to perform the task. These frontal regions were also noted to have decreased gray matter from prechemotherapy to postchemotherapy in a previous study by the same authors.117,118 Findings from fMRI studies are supported by results from complementary techniques, including positron emission tomography,134–136 arterial spin labeling,137 and electroencephalography (EEG).138–141 Magnetic resonance spectroscopy imaging has also provided further insight regarding certain pathophysiologic mechanisms, including inflammation and demyelination.109,142,143
Brain injury has been observed in association with several putative CRCI mechanisms, including cancer pathogenesis and/or surgery,16,121,122,129,132,133 chemotherapy,66,109,110,113,115–118,124–126,129,130,132,134,137,144 locoregional radiation therapy,145 endocrine therapy,111,146 menopausal status,123 and inflammatory response.114,135,147 Neuroimaging studies have also demonstrated neurobiologic alterations associated with specific symptoms that are often comorbid with CRCI, including fatigue, depression, and anxiety.131,148–150 However, none of these studies distinguished among the specific effects of various potential CRCI mechanisms. Therefore, it is currently unclear whether certain mechanisms are more or less likely to result in CRCI and/or whether CRCI results from the cumulative effects of various mechanisms.
Previous studies have consistently demonstrated correlations between altered neurobiologic status and atypical neurocognitive testing performance.66,106–108,110,113,114,123,134,137 While neurocognitive testing has shown an inconsistent correlation with patient self-report,151,152 several studies have demonstrated significant associations between various neuroimaging metrics and subjective cognitive complaints.66,118,120,125,129,139,142 Future investigations are needed that directly compare the imaging characteristics of patients with and without significant subjective and/or objective cognitive impairment.
Although neuroimaging research offers significant insight regarding the biologic mechanisms underlying CRCI, neuroimaging is not currently part of the clinical standard of care for primary breast cancer. Given our increasing awareness of the potential adverse effects of many cancer therapies on brain and cognition, methods for predicting which patients are at highest risk are not only feasible but may also help prevent long-term disability. Neuroimaging is increasingly used to aid the prediction of outcomes for various brain-based disorders, including future cognitive decline.153–155 Prediction algorithms have benefitted from the innovative application of machine learning to neuroimaging data. Machine learning is a branch of artificial intelligence involving methods that can effectively detect complex patterns in high-dimensional data and make predictions. This translates into marked potential for the diagnosis of neurodegenerative brain diseases.156–158 Machine learning studies in breast cancer survivors provide support for the use of baseline neuroimaging data to predict which patients will have persistent and/or progressive CRCI.120,128,130 These emerging approaches hold promise for identifying those at risk of CRCI, and the International Cognition and Cancer Task Force is currently working to create recommendations for methods and timing of neuroimaging acquisitions.
Intervention
Given the neurocognitive deficits identified on neuropsychological evaluation and reported difficulties with instrumental activities of daily living, the patient was referred for cognitive rehabilitation after initiation of endocrine therapy. She participated in 60-minute sessions, once per week for 12 weeks. The treatment plan included psychoeducation (eg, presentation of information regarding symptoms and mechanisms of CRCI), training in compensatory strategies (eg, using a smartphone), in vivo training of real-world skills (eg, planning a weekly schedule), and homework assignments (eg, workbook readings and skills practice). Homework also included a curriculum of computerized cognitive training focused on executive functioning and memory. She completed the exercises using her home computer for 20 to 30 minutes per day, 5 days per week, for 12 weeks. The patient was further prescribed a program of physical exercise to help with fatigue, sleep, and cognition. At the recommendation of her neuropsychologist, her employer provided workplace accommodations, including a change within the law firm to a less demanding role.
Behavioral Strategies
Cognitive rehabilitation refers to a clinic-based, therapeutic program aimed at improving cognitive abilities, functional capacity, real-world skills, and/or internal metacognitive strategies (ie, internal plans of action for completing cognitive tasks). Programs can be inpatient or outpatient and involve patients meeting individually and/or in groups with a trained clinician (typically a neuropsychologist, psychologist, speech and language pathologist, or occupational therapist). Thus far, there have been a limited number of studies involving cognitive rehabilitation after breast cancer, but the majority have demonstrated significant improvement in both objective and subjective cognitive performance as well as quality of life (Table 1).159–172 Cognitive domains showing intervention effects include executive functioning, working memory, attention, memory, processing speed, and visual-spatial skills. One study also included a quantitative EEG biomarker of intervention outcome and demonstrated increased global brain activity after cognitive rehabilitation.160 Ferguson et al randomized 40 breast cancer survivors who were at least 18 months postchemotherapy and had subjective complaints of CRCI to a manualized cognitive rehabilitation program focusing on memory and attention or a waitlist control condition.162 The intervention program consisted of 4 biweekly, individual office visits, 30 to 50 minutes in duration, with phone contacts between visits. Intervention participants received psychoeducation regarding memory and attention, as well as training in self-awareness, self-regulation, and cognitive compensatory strategies. Compared with the waitlist control group, the intervention group demonstrated significantly increased memory performance and improved self-reported quality of life. Participants rated the intervention as being most useful for helping them compensate for daily memory difficulties.
TABLE 1.
AUTHORS | NO. OF PARTICIPANTS | INTERVENTION | FINDINGS |
---|---|---|---|
Cherrier 2013159a | CR, n = 12; NTCs, n = 16; RD; OffTx 4.8 y (≥6 mo) | Seven weekly 1-h group sessions focused on memory aids and skills, mindfulness meditation, homework | Improved working memory, attention, QOL, and SR cognition |
Ercoli 2013160 | CR, n = 27; OffTx ≥18 mo | Five weekly 2-h, manual-guided sessions focused on attention, memory, and executive functioning; homework | Improved attention and executive functioning, SR memory, and cognition; increased global brain EEG activity |
Ferguson 2007161 | CR, n = 29; OffTx ≥3 y | Four monthly 30-min to 50-min, manual-guided sessions focused on attention and memory, CBT, self-awareness, and self-regulation; manual-guided homework; 3 total telephone contacts for support and review | Improved verbal memory, processing speed, response inhibition, executive function, attention, SR cognition, and QOL |
Ferguson 2012162 | CR, n = 19; NTCs, n = 21; RD; OffTx ≥18 mo | Same as above | Improved verbal memory, QOL |
McDougall 2011163a | CR, n = 8; AC, n = 14; NRD; OffTx not specified | Eight 50-min sessions focused on anxiety, depression, health, metamemory, memory self-efficacy; 4 weekly 2-h booster sessions for 1 mo within 3 mo of completing initial training | Improved SR memory |
Popplereuter 2009164 | CR, n = 33; CT, n = 34; NTCs, n = 29; RD to CT or CR (but NTCs not randomized); OffTx 2.1 mo | Four 1-h inpatient sessions per wk for 3–5 wk; CR: attention, memory processes, skills, and compensatory strategies; CT: unspecified computerized program for attention, memory | No intervention effects |
Schuurs and Green 2013165a | CR, n = 23; NTCs (cancer), n = 9; NTCs (noncancer), n = 23; NRD; OffTx 36–48 mo | Weekly 2-h, manual-guided group sessions over 6 wk focused on psychoeducation, thematic group discussion, memory and attention skills training, and compensatory strategies with homework; CBT: emotional adjustment, fatigue, sleep, self-care | Improved immediate memory, visuospatial memory, and delayed memory |
Kesler 2013166 | CT, n = 21; WLCs, n = 20; RD; OffTx 6 y (≥18 mo) | Forty-eight 20-min to 30-min sessions over 12 wk of home-based, computerized Lumosity program for executive function, processing speed, verbal fluency | Improved executive function, verbal fluency, processing speed, SR executive function behaviors |
Von Ah 2012167 | CT memory, n = 26; CT processing speed, n = 27; WLCs, n = 29; RD; OffTx 65.5 mo (≥1 y) | Ten 1-h small group sessions over 6–8 wk; computerized Posit Science program for memory or processing speed | Improved verbal memory, SR cognition, SR memory; anxiety in CT memory group vs WLCs; improved processing speed, memory, depression, anxiety, mental health in CT processing speed group vs WLCs |
Galantino 2012168 | PA, n = 4 (case studies); OffTx, n = 0 (enrolled prechemotherapy) | Iyengar yoga 2 × wk for 12 wk | Improved cognitive speed and accuracy, QOL |
Janelsins 2012169a | PA, n = 168; NTCs, n = 160; RD; OffTx 2–24 mo | Biweekly 75-min sessions for 4 wk; breathing exercises; gentle Hatha and Restorative yoga postures and meditation | Improved SR memory |
Reid-Arndt 2012170a | PA, n = 23; OffTx not specified, but participants were 6.5 y postdiagnosis | Biweekly 1-h Tai Chi classes for 10 wk | Improved verbal memory, attention, verbal fluency, executive function, SR verbal and visual memory |
Oh 2012171a | PA, n = 37; NTCs, n = 44; RD; OffTx not specified, but some patients were still undergoing chemotherapy | Biweekly 90-min sessions for 10 wk of medical Qigong group class | Improved SR cognition, QOL; reduced C-reactive protein levels |
Alvarez 2013172 | Neurofeedback, n = 23; participants served as their own WLCs; OffTx 6–60 mo | Biweekly 30-min sessions for 10 wk of Zengar approach, whole-brain EEG neurofeedback | Improved SR cognition, QOL, fatigue, sleep function, psychological symptoms |
AC indicates active/attention control; CBT, cognitive-behavioral therapy; CR, cognitive rehabilitation; CT, cognitive training; EEG, electroencephalography; NRD, nonrandomized; NTCs, no treatment controls; OffTx, time off-therapy (chemotherapy, radiation; presented as the mean time if available or the minimum requirement as indicated by study methods); PA, physical activity; QOL, quality of life; RD, randomized; SR, self-rated; WLCs, waitlist controls.
References for our literature review of intervention studies were identified via PubMed and Google Scholar searches using combinations of the keywords cognit*, rehabilitation, training, remediation, neuropsycholog*, intervention*, trial, cancer, radiation, chemotherap*, chemobrain, brain, exercise*, “physical exercise,” “physical activity,” neuroplasticity, enhance*, learn*, memory, executive, speed, and attention. Searches were limited to human subjects and articles written in English.
The sample for this study included participants with other cancer diagnoses in addition to breast cancer.
Cognitive rehabilitation can also be advantageous for improving achievement of real-world objectives and managing psychological comorbidities, such as the anxiety and depression experienced by the patient described above. The social aspects of this approach (eg, therapeutic alliance with the clinician and group participants) are believed to contribute significantly to the cognitive and psychological effects. Unfortunately, cognitive rehabilitation is not always feasible because it requires multiple in-person sessions and administration by trained clinical providers. In addition, cognitive rehabilitation is not widely available and has a history of unpredictable coverage by health insurance.173 However, many studies of cognitive rehabilitation in breast cancer survivors involve a manualized treatment approach,160–162,165 which improves intervention standardization and practicality of dissemination.
Cognitive training aims to improve and maintain cognitive skills via distributed, adaptive practice of specific cognitive domains. Unlike cognitive rehabilitation, cognitive training tends to focus on independent cognitive skills practice, without compensatory or metacognitive strategies, and frequently relies heavily on computerized exercises. These exercises are typically game-based, given that games require various degrees of active problem solving and decision making to progress.174 Exercises also involve algorithmic control of difficulty level to optimize the balance between challenge and motivation. Studies of cognitive training for CRCI after breast cancer, although preliminary, suggest that executive function, memory, processing speed, self-rated cognition, anxiety, and depression can improve with as few as 10 hours of distributed training.164,166,167 In 2012, Von Ah et al randomized 82 breast cancer survivors who were at least 1 year postchemotherapy and had subjective complaints of CRCI to memory cognitive rehabilitation, processing speed cognitive training, or waitlist control conditions.167 Memory rehabilitation consisted of training participants in various compensatory strategies, including in vivo practice of these strategies. Processing speed training involved adaptive, computerized exercises focusing on time-order judgment, discrimination, spatial match, forward span, instruction after, and narrative-memory tasks. Each intervention was delivered for a total of 10 hours and was conducted in small groups over 6 to 8 weeks. The memory training group demonstrated significant improvements in immediate and delayed memory performance compared with waitlist controls. The processing speed group demonstrated significantly improved processing speed as well as improved immediate and delayed memory performance compared with waitlist controls. Both intervention groups also demonstrated improved subjective cognition and reduced psychological distress.167
In-clinic167 and home-based166 cognitive training programs appear to show very similar effect sizes in breast cancer survivors, consistent with previous research in healthy older adults.175 Cognitive training programs by Posit Science Corporation (San Francisco, CA; brainhq.com/)167 and Lumosity (Lumos Labs, Inc. San Francisco, CA; lumosity.com)166 have been tested in breast cancer, and both showed similar effects. Cognitive training programs are widely available, relatively inexpensive, and allow for remote administration with improved feasibility and access. Previous studies of healthy adults have claimed that cognitive training is neuroprotective with respect to improvement and conservation of cognitive performance.175,176 These findings may be particularly salient for breast cancer survivors, given that previous research suggests cancer and/or its treatments may accelerate the aging process.12,177,178 However, cognitive training may not be suitable for individuals with more severe cognitive deficits or significant psychological comorbidities.
Some studies of cancer and noncancer groups have shown transfer of benefits to nontrained skills179–181 and improvement in symptoms other than cognitive difficulties, such as sleep quality, psychological distress, and fatigue.166,167,182,183 Given that emotional regulation strategies are not typically included in cognitive training approaches, the improvement of other symptoms may reflect training-induced increase in general brain health. These improvements may also reflect the effects of user-game-experience and positive psychology principles,184,185 which increase internal locus of control, self-efficacy, and distraction from real-world problems. However, in general, transfer effects of cognitive training remain inconsistent.186 In addition, “training to the test” can confound cognitive training effects, as certain training exercises resemble the cognitive tests that are used to evaluate their effectiveness.
Physical Activity
Physical activity (PA) is associated with improved cognitive function in both human and animal studies.188–190 In healthy adults, executive functions show the largest and most consistent exercise-related increases, while improvements in memory and other cognitive skills have been less reliable thus far.190 PA increases neurogenesis188–190 and the levels of neurotransmitters and neurotrophins that promote cognitive function,188,191 it reduces inflammation,192 and it stimulates positive brain vasculature changes.188 Reduction of psychological and chronic medical conditions (eg, depression, sleep disruption, diabetes, obesity) associated with PA193 may also indirectly improve cognitive function. PA is also believed to moderate the signaling pathways involved in neuroprotection,194 such as increasing the expression of neuroprotective genes.191 Importantly, animal studies suggest that PA can improve cognitive function after breast cancer treatment.195
Human studies of PA as an intervention for CRCI in breast cancer (Table 1) have involved Hatha yoga, Iyengar yoga, Qigong, and Tai Chi.168–171 Improvements were noted in processing speed, memory, executive function, and quality of life. In 1 study,169 cognitive effects were observed after as little as 1 month of intervention with PA, which is consistent with results from previous studies in healthy adults.196 Specifically, Janelsins and colleagues randomized 358 cancer survivors (75% breast cancer) to a yoga intervention or a no-treatment condition.169 The intervention consisted of breathing exercises, gentle Hatha and Restorative yoga postures, and meditation twice per week for 75 minutes across 4 weeks. The intervention group demonstrated significantly improved subjective memory functioning and quality of life as well as reduced fatigue compared with the no-treatment group.
Thus far, PA studies in breast CRCI have focused on specific types of nonaerobic interventions, despite national guidelines suggesting that moderate to vigorous intensity aerobic exercise is recommended for brain health.196 PA interventions have also involved relatively short durations (ie, 2–10 weeks), which may not result in lasting benefit among older adults.197 In addition, most of the existing studies did not include objective cognitive tests, and none have adequately evaluated executive function, although this domain is among the most commonly impaired after breast cancer and the most amenable to PA intervention. The relationship between PA and cognitive function, especially executive function, is bidirectional. In healthy adults, executive function has been shown to moderate PA adherence.198–201 In addition, decreased prefrontal cortex activity has been associated with reduced self-regulation of PA behavior in healthy adults.202 This is unsurprising given that core executive functions, including working memory, inhibition, and task switching, are believed to be critical for successful self-regulation.203 Individuals with executive dysfunction often have difficulty changing ideas or behaviors,204,205 and cognitive dysfunction can be a barrier to PA engagement after cancer.206 Accordingly, the combination of PA with cognitive interventions may prove more optimal for some cancer survivors, particularly those with significant CRCI.
Both physical and cognitive exercise increase neuroplasticity but do so through different pathways. PA results in an increased number of new neurons; however, roughly 50% of these die within a few weeks, never reaching full maturity or becoming connected with other neurons.207 Based on emerging animal studies, many of these neurons may be rescued with cognitive training, which enables them to form functional connections within brain networks.207 Therefore, the combination of PA and cognitive training may result in greater cognitive benefit compared with either approach alone. Preliminary human and animal studies provide additional support for this hypothesis.190,207–213
Regular mental activity is known to be neuroprotective by increasing cognitive reserve.214 As such, it is theoretically possible that cognitive interventions conducted before and/ or during cancer therapy could help prevent cognitive difficulties. However, the optimal timing of behavioral interventions for CRCI remains unaddressed within the literature. It is also unknown how natural neural recovery and response to behavioral intervention are affected by different cancer types and treatments. Only 1 study of cognitive rehabilitation and cognitive training in breast cancer conducted thus far demonstrated no intervention effects. In that study, the breast cancer group showed a decline in verbal memory from post-intervention to 6-month follow–up, suggesting that early cognitive rehabilitation/cognitive training intervention may not have stable effects.164 That study was unique in that participants were only off-therapy an average of 2 months. As in other forms of acquired brain injury, cognitive impairment after breast cancer treatment is often characterized by initial deficit occurring within the first 6 months followed by a 1-year to 2-year recovery/stabilization period.62,215 The majority of the other intervention studies reviewed above involved long-term survivors and several years off-therapy, with a minimum time off-therapy (excluding endocrine therapy) ranging from 0.5 to 3 years. The rationale for the minimum time off-therapy, when provided, was consistently to allow for neurologic and medical/health stabilization.
Further research is needed regarding cognitive interventions conducted before treatment and/or during the acute recovery phase (approximately 0–6 months) after adjuvant cancer therapies (ie, chemotherapy and radiation). Cognitive rehabilitation or cognitive training programs may require currently unknown modifications to be effective preventive approaches. It is also possible that exercise interventions may have greater potential for prevention, given that the majority of exercise studies conducted to date occurred during or shortly after cancer therapy.
Neuromodulation Strategies
Cognitive neuroscience-based interventional research offers new, noninvasive approaches for ameliorating cognitive dysfunction. Neurofeedback is a method that involves providing a participant with feedback regarding her brain activity as a means of training her to control the up-regulation and down-regulation of brain activity using metacognitive strategies. Various cognitive-behavioral symptoms are potentially reduced by targeting appropriate brain regions. Neurofeed-back is provided via EEG, functional near-infrared spectroscopy (NIRS), or real-time fMRI. Neurofeedback studies conducted in noncancer populations demonstrate that participants can learn to control the activation of specific brain regions related to motor function, attention, pain response, and emotion regulation, among others.216 One study has been conducted in breast cancer survivors and demonstrated positive effects on subjective cognitive function using EEG neurofeedback (Table 1), suggesting potential for this and similar neurofeedback techniques.172
Repetitive transcranial magnetic stimulation (rTMS) uses magnetic fields to modulate neuronal excitability. During the procedure, a TMS coil is suspended over a specific area of the head to deliver short electromagnetic pulses. The technique is currently approved by the US Food and Drug Administration for the treatment of depression, but it is also being investigated as a means of improving cognitive performance.217,218 Currently, there have been no studies of rTMS for CRCI, although positive effects on memory and attention impairments have been documented in other populations,219,220 making it a promising potential intervention for future studies.
Pharmacotherapy
Very few psychopharmacologic agents have proven effective in reducing or preventing cognitive impairment in non-CNS cancer patients. Psychostimulants like methylphenidate, dexmethylphenidate, and modafinil have produced mixed results; and, at present, the effectiveness of these agents is not well established. Another example of an agent that received attention after initial preclinical findings is donepezil, an acetylcholinesterase inhibitor. However, its effectiveness remains uncertain, as only 2 small studies with conflicting findings have been published so far.221–223 More rigorous clinical trials with sufficient sample sizes are necessary to definitively assess the potential of such pharmacologic agents in the treatment of CRCI.
Unraveling the precise mechanisms underlying CRCI may facilitate the identification of novel pharmacologic treatment strategies. Several animal studies focusing on the impact of chemotherapy on cognition have suggested promising interventions, such as preventing oxidative stress associated with chemotherapy using 2-Mercaptoethane sulfonate,224 N-acetylcysteine, or melatonin225; stimulating neurogenesis with insulin-like growth factor-1,226 fluoxetine, or glucose227,228; and treatments with glutamate receptor antagonists, such as dextromethorphan229 or memantine,230 that ameliorate chemotherapy-induced CRCI. The arrival of new targeted agents and immunotherapies that may influence cognition, either alone or in combination with traditional anticancer agents, will necessitate further preclinical work to understand and potentially improve adverse side effects on cognition. In addition, tumor-bearing model systems will be essential in the evaluation of potential adverse effects of promising agents on antitumor efficacy.
Summary
CRCI is a frequent adverse effect of non-CNS cancer and systemic anticancer therapies in adults. While predominantly investigated in women with breast cancer, these adverse outcomes are common to many other cancer patient populations (eg, colorectal carcinoma, leukemia, lymphoma, multiple myeloma, ovarian carcinoma, prostate carcinoma, testicular cancer). Clinical observational studies have characterized the most typical pattern of cognitive dysfunction associated with CRCI (ie, memory loss, slowed processing speed, and executive dysfunction), have provided some early estimates of incidence (ranging from17% to over 70%), and have identified potential clinical and patient factors that modify risk (eg, chemotherapy dose, cognitive reserve, presence of an APOEε4 allele). It has been demonstrated that this neurotoxicity adversely impacts patients’ and survivors’ quality of life, including occupational and social functioning, and results in increased health care and societal costs.
More recently, various preclinical and neuroimaging studies have begun to shed light on the pathophysiologic mechanisms that may underlie CRCI. It is anticipated that continued research in these areas will create translational science opportunities to develop and test pharmacologic and behavioral interventions to prevent, reduce, or eliminate this untoward neurotoxicity. Currently, few intervention studies involving CRCI after breast cancer have been conducted, and many of these are limited by methodological concerns, including small sample sizes, lack of objective cognitive testing, inclusion of other cancer types, limited public health dissemination potential, and/or lack of proper control conditions. In addition, no effective prevention strategies have been identified to date. However, many promising lines of research are emerging that offer hope and promise to future cancer patients and the clinicians who care for them.
Acknowledgments
DISCLOSURES: Supported by the National Institute of Nursing Research of the National Institutes of Health under Award Number R01NR014195 (Dr. Wefel and Dr. Kesler); the National Cancer Institute under Award Number R01CA172145 (Dr. Kesler); and the Dutch Cancer Society under Award Numbers 2009-4284, 2010-4829, and 2012-5495 (Dr. Schagen). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health or the Dutch Cancer Society. Dr. Kesler reports grants from the National Institutes of Health and personal fees for lectures at academic institutions.
Footnotes
To earn free CME credit or nursing contact hours for successfully completing the online quiz based on this article, go to acsjournals.com/ce.
The remaining authors report no conflicts of interest.
References
- 1.Boykoff N, Moieni M, Subramanian SK. Confronting chemobrain: an in-depth look at survivors’ reports of impact on work, social networks, and health care response. J Cancer Surviv. 2009;3:223–232. doi: 10.1007/s11764-009-0098-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Nelson WL, Suls J. New approaches to understand cognitive changes associated with chemotherapy for non-central nervous system tumors. J Pain Symptom Manage. 2013;46:707–721. doi: 10.1016/j.jpainsymman.2012.11.005. [DOI] [PubMed] [Google Scholar]
- 3.Weiss HD, Walker MD, Wiernik PH. Neurotoxicity of commonly used antineoplastic agents (first of two parts) N Engl J Med. 1974;291:75–81. doi: 10.1056/NEJM197407112910205. [DOI] [PubMed] [Google Scholar]
- 4.Ahles TA, Saykin AJ. Candidate mechanisms for chemotherapy-induced cognitive changes. Nat Rev Cancer. 2007;7:192–201. doi: 10.1038/nrc2073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Ouimet LA, Stewart A, Collins B, Schindler D, Bielajew C. Measuring neuropsychological change following breast cancer treatment: an analysis of statistical models. J Clin Exp Neuropsychol. 2009;31:73–89. doi: 10.1080/13803390801992725. [DOI] [PubMed] [Google Scholar]
- 6.Ahles TA, Saykin AJ. Breast cancer chemotherapy-related cognitive dysfunction. Clin Breast Cancer. 2002;3(suppl 3):S84–S90. doi: 10.3816/cbc.2002.s.018. [DOI] [PubMed] [Google Scholar]
- 7.Bender CM, Sereika SM, Berga SL, et al. Cognitive impairment associated with adjuvant therapy in breast cancer. Psychooncology. 2006;15:422–430. doi: 10.1002/pon.964. [DOI] [PubMed] [Google Scholar]
- 8.Castellon SA, Ganz PA, Bower JE, Petersen L, Abraham L, Greendale GA. Neurocognitive performance in breast cancer survivors exposed to adjuvant chemotherapy and tamoxifen. J Clin Exp Neuropsychol. 2004;26:955–969. doi: 10.1080/13803390490510905. [DOI] [PubMed] [Google Scholar]
- 9.Donovan KA, Small BJ, Andrykowski MA, Schmitt FA, Munster P, Jacobsen PB. Cognitive functioning after adjuvant chemotherapy and/or radiotherapy for early stage breast carcinoma. Cancer. 2005;104:2499–2507. doi: 10.1002/cncr.21482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Gottschalk LA, Holcombe RF, Jackson D, Bechtel RJ. The effects of anticancer chemotherapeutic drugs on cognitive function and other neuropsychiatric dimensions in breast cancer patients. Methods Find Exp Clin Pharmacol. 2003;25:117–122. doi: 10.1358/mf.2003.25.2.723685. [DOI] [PubMed] [Google Scholar]
- 11.Hermelink K, Untch M, Lux MP, et al. Cognitive function during neoadjuvant chemotherapy for breast cancer: results of a prospective, multicenter, longitudinal study. Cancer. 2007;109:1905–1913. doi: 10.1002/cncr.22610. [DOI] [PubMed] [Google Scholar]
- 12.Mandelblatt JS, Hurria A, McDonald BC, et al. Cognitive effects of cancer and its treatments at the intersection of aging: what do we know; what do we need to know? Semin Oncol. 2013;40:709–725. doi: 10.1053/j.seminoncol.2013.09.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Jenkins V, Shilling V, Deutsch G, et al. A 3-year prospective study of the effects of adjuvant treatments on cognition in women with early stage breast cancer. Br J Cancer. 2006;94:828–834. doi: 10.1038/sj.bjc.6603029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Stewart A, Collins B, Mackenzie J, Tomiak E, Verma S, Bielajew C. The cognitive effects of adjuvant chemotherapy in early stage breast cancer: a prospective study. Psychooncology. 2008;17:122–130. doi: 10.1002/pon.1210. [DOI] [PubMed] [Google Scholar]
- 15.Debess J, Riis JO, Pedersen L, Ewertz M. Cognitive function and quality of life after surgery for early breast cancer in North Jutland, Denmark. Acta Oncol. 2009;48:532–540. doi: 10.1080/02841860802600755. [DOI] [PubMed] [Google Scholar]
- 16.Scherling C, Collins B, Mackenzie J, Bielajew C, Smith A. Pre-chemotherapy differences in visuospatial working memory in breast cancer patients compared to controls: an FMRI study [serial online] Front Hum Neurosci. 2011;5:122. doi: 10.3389/fnhum.2011.00122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Chen ML, Miaskowski C, Liu LN, Chen SC. Changes in perceived attentional function in women following breast cancer surgery. Breast Cancer Res Treat. 2012;131:599–606. doi: 10.1007/s10549-011-1760-3. [DOI] [PubMed] [Google Scholar]
- 18.Wefel JS, Lenzi R, Theriault R, Buzdar AU, Cruickshank S, Meyers CA. ‘Chemobrain’ in breast carcinoma?: a prologue. Cancer. 2004;101:466–475. doi: 10.1002/cncr.20393. [DOI] [PubMed] [Google Scholar]
- 19.Ernst T, Chang L, Cooray D, et al. The effects of tamoxifen and estrogen on brain metabolism in elderly women. J Natl Cancer Inst. 2002;94:592–597. doi: 10.1093/jnci/94.8.592. [DOI] [PubMed] [Google Scholar]
- 20.Schilder CM, Seynaeve C, Beex LV, et al. Effects of tamoxifen and exemestane on cognitive functioning of postmenopausal patients with breast cancer: results from the neuropsychological side study of the tamoxifen and exemestane adjuvant multinational trial. J Clin Oncol. 2010;28:1294–1300. doi: 10.1200/JCO.2008.21.3553. [DOI] [PubMed] [Google Scholar]
- 21.Shilling V, Jenkins V, Fallowfield L, Howell T. The effects of hormone therapy on cognition in breast cancer. J Steroid Biochem Mol Biol. 2003;86:405–412. doi: 10.1016/j.jsbmb.2003.07.001. [DOI] [PubMed] [Google Scholar]
- 22.Janelsins MC, Kesler SR, Ahles TA, Morrow GR. Prevalence, mechanisms, and management of cancer-related cognitive impairment. Int Rev Psychiatry. 2014;26:102–113. doi: 10.3109/09540261.2013.864260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Ahles TA, Saykin AJ, McDonald BC, et al. Cognitive function in breast cancer patients prior to adjuvant treatment. Breast Cancer Res Treat. 2008;110:143–152. doi: 10.1007/s10549-007-9686-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Jansen CE, Cooper BA, Dodd MJ, Miaskowski CA. A prospective longitudinal study of chemotherapy-induced cognitive changes in breast cancer patients. Support Care Cancer. 2011;19:1647–1656. doi: 10.1007/s00520-010-0997-4. [DOI] [PubMed] [Google Scholar]
- 25.Wefel JS, Saleeba AK, Buzdar AU, Meyers CA. Acute and late onset cognitive dysfunction associated with chemotherapy in women with breast cancer. Cancer. 2010;116:3348–3356. doi: 10.1002/cncr.25098. [DOI] [PubMed] [Google Scholar]
- 26.Perry A, Schmidt RE. Cancer therapy-associated CNS neuropathology: an update and review of the literature. Acta Neuropathol. 2006;111:197–212. doi: 10.1007/s00401-005-0023-y. [DOI] [PubMed] [Google Scholar]
- 27.Videnovic A, Semenov I, Chua-Adajar R, et al. Capecitabine-induced multifocal leukoencephalopathy: a report of five cases. Neurology. 2005;65:1792–1794. doi: 10.1212/01.wnl.0000187313.83515.7e. discussion 1685. [DOI] [PubMed] [Google Scholar]
- 28.Ahles TA, Saykin AJ, Furstenberg CT, et al. Neuropsychologic impact of standard-dose systemic chemotherapy in long-term survivors of breast cancer and lymphoma. J Clin Oncol. 2002;20:485–493. doi: 10.1200/JCO.2002.20.2.485. [DOI] [PubMed] [Google Scholar]
- 29.Wefel JS, Schagen SB. Chemotherapy-related cognitive dysfunction. Curr Neurol Neurosci Rep. 2012;12:267–275. doi: 10.1007/s11910-012-0264-9. [DOI] [PubMed] [Google Scholar]
- 30.Janelsins MC, Kohli S, Mohile SG, Usuki K, Ahles TA, Morrow GR. An update on cancer- and chemotherapy-related cognitive dysfunction: current status. Semin Oncol. 2011;38:431–438. doi: 10.1053/j.seminoncol.2011.03.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.DeSantis CE, Lin CC, Mariotto AB, et al. Cancer treatment and survivorship statistics, 2014. CA Cancer J Clin. 2014;64:252–271. doi: 10.3322/caac.21235. [DOI] [PubMed] [Google Scholar]
- 32.Pedersen AD, Rossen P, Mehlsen MY, Pedersen CG, Zachariae R, von der Maase H. Long-term cognitive function following chemotherapy in patients with testicular cancer. J Int Neuropsychol Soc. 2009;15:296–301. doi: 10.1017/S1355617709090316. [DOI] [PubMed] [Google Scholar]
- 33.Wefel JS, Vidrine DJ, Marani SK, et al. A prospective study of cognitive function in men with non-seminomatous germ cell tumors. Psychooncology. 2014;23:626–633. doi: 10.1002/pon.3453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Jones D, Vichaya EG, Wang XS, Sailors MH, Cleeland CS, Wefel JS. Acute cognitive impairment in patients with multiple myeloma undergoing autologous hematopoietic stem cell transplant. Cancer. 2013;119:4188–4195. doi: 10.1002/cncr.28323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Vardy JL, Booth C, Pond GR, et al. Cytokine levels in patients (pts) with colorectal cancer and breast cancer and their relationship to fatigue and cognitive function [abstract] J Clin Oncol. 2007;25(18S):Abstract 9070. [Google Scholar]
- 36.Correa DD, Hess LM. Cognitive function and quality of life in ovarian cancer. Gynecol Oncol. 2012;124:404–409. doi: 10.1016/j.ygyno.2011.11.005. [DOI] [PubMed] [Google Scholar]
- 37.Chao HH, Uchio E, Zhang S, et al. Effects of androgen deprivation on brain function in prostate cancer patients—a prospective observational cohort analysis [serial online] BMC Cancer. 2012;12:371. doi: 10.1186/1471-2407-12-371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lange M, Giffard B, Noal S, et al. Baseline cognitive functions among elderly patients with localised breast cancer. Eur J Cancer. 2014;50:2181–2189. doi: 10.1016/j.ejca.2014.05.026. [DOI] [PubMed] [Google Scholar]
- 39.Kinsinger SW, Lattie E, Mohr DC. Relationship between depression, fatigue, subjective cognitive impairment, and objective neuropsychological functioning in patients with multiple sclerosis. Neuropsychology. 2010;24:573–580. doi: 10.1037/a0019222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Slavin MJ, Brodaty H, Kochan NA, et al. Prevalence and predictors of “subjective cognitive complaints” in the Sydney Memory and Ageing Study. Am J Geriatr Psychiatry. 2010;18:701–710. doi: 10.1097/jgp.0b013e3181df49fb. [DOI] [PubMed] [Google Scholar]
- 41.Cicerone KD, Kalmar K. Persistent post-concussion syndrome: the structure of subjective complaints after mild traumatic brain injury. J Head Trauma Rehabil. 1995;10:1–17. [Google Scholar]
- 42.Ganz PA, Kwan L, Castellon SA, et al. Cognitive complaints after breast cancer treatments: examining the relationship with neuropsychological test performance. J Natl Cancer Inst. 2013;105:791–801. doi: 10.1093/jnci/djt073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Shilling V, Jenkins V. Self-reported cognitive problems in women receiving adjuvant therapy for breast cancer. Eur J Oncol Nurs. 2007;11:6–15. doi: 10.1016/j.ejon.2006.02.005. [DOI] [PubMed] [Google Scholar]
- 44.Pullens MJ, De Vries J, Roukema JA. Subjective cognitive dysfunction in breast cancer patients: a systematic review. Psychooncology. 2010;19:1127–1138. doi: 10.1002/pon.1673. [DOI] [PubMed] [Google Scholar]
- 45.Pullens MJ, De Vries J, Van Warmerdam LJ, Van De Wal MA, Roukema JA. Chemotherapy and cognitive complaints in women with breast cancer. Psychooncology. 2013;22:1783–1789. doi: 10.1002/pon.3214. [DOI] [PubMed] [Google Scholar]
- 46.Cimprich B, Ronis DL, Trask C. Pre-treatment factors related to cognitive functioning in women newly diagnosed with breast cancer. Psychooncology. 2005;14:70–78. doi: 10.1002/pon.821. [DOI] [PubMed] [Google Scholar]
- 47.Mandelblatt JS, Stern RA, Luta G, et al. Cognitive impairment in older patients with breast cancer before systemic therapy: is there an interaction between cancer and comorbidity? J Clin Oncol. 2014;32:1909–1918. doi: 10.1200/JCO.2013.54.2050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Evered L, Scott DA, Silbert B, Maruff P. Postoperative cognitive dysfunction is independent of type of surgery and anesthetic. Anesth Analg. 2011;112:1179–1185. doi: 10.1213/ANE.0b013e318215217e. [DOI] [PubMed] [Google Scholar]
- 49.Seigers R, Fardell JE. Neurobiological basis of chemotherapy-induced cognitive impairment: a review of rodent research. Neurosci Biobehav Rev. 2011;35:729–741. doi: 10.1016/j.neubiorev.2010.09.006. [DOI] [PubMed] [Google Scholar]
- 50.Yang M, Kim J, Kim J-S, et al. Hippocampal dysfunctions in tumor-bearing mice. Brain Behav Immun. 2014;36:147–155. doi: 10.1016/j.bbi.2013.10.022. [DOI] [PubMed] [Google Scholar]
- 51.Schagen SB, Muller MJ, Boogerd W, et al. Late effects of adjuvant chemotherapy on cognitive function: a follow-up study in breast cancer patients. Ann Oncol. 2002;13:1387–1397. doi: 10.1093/annonc/mdf241. [DOI] [PubMed] [Google Scholar]
- 52.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. 2004;100:2292–2299. doi: 10.1002/cncr.20272. [DOI] [PubMed] [Google Scholar]
- 53.Mar Fan HG, Houede-Tchen N, Yi Q-L, et al. Fatigue, menopausal symptoms, and cognitive function in women following adjuvant chemotherapy for breast cancer with or without subsequent tamoxifen: one and two year follow-up of a prospective controlled study. Breast Cancer Res Treat. 2004;88:S138–S138. [Google Scholar]
- 54.Shilling V, Jenkins V, Morris R, Deutsch G, Bloomfield D. The effects of adjuvant chemotherapy on cognition in women with breast cancer—preliminary results of an observational longitudinal study. Breast. 2005;14:142–150. doi: 10.1016/j.breast.2004.10.004. [DOI] [PubMed] [Google Scholar]
- 55.Hurria A, Rosen C, Hudis C, et al. Cognitive function of older patients receiving adjuvant chemotherapy for breast cancer: a pilot prospective longitudinal study. J Am Geriatr Soc. 2006;54:925–931. doi: 10.1111/j.1532-5415.2006.00732.x. [DOI] [PubMed] [Google Scholar]
- 56.Schagen SB, Muller MJ, Boogerd W, Mellenbergh GJ, van Dam FS. Change in cognitive function after chemotherapy: a prospective longitudinal study in breast cancer patients. J Natl Cancer Inst. 2006;98:1742–1745. doi: 10.1093/jnci/djj470. [DOI] [PubMed] [Google Scholar]
- 57.Minisini AM, De Faccio S, Ermacora P, et al. Cognitive functions and elderly anti-cancer treatment: a cancer patients receiving prospective study. Crit Rev Oncol Hematol. 2008;67:71–79. doi: 10.1016/j.critrevonc.2008.02.004. [DOI] [PubMed] [Google Scholar]
- 58.Jansen CE, Dodd MJ, Miaskowski CA, Dowling GA, Kramer J. Preliminary results of a longitudinal study of changes in cognitive function in breast cancer patients undergoing chemotherapy with doxorubicin and cyclophosphamide. Psychooncology. 2008;17:1189–1195. doi: 10.1002/pon.1342. [DOI] [PubMed] [Google Scholar]
- 59.Collins B, Mackenzie J, Tasca GA, Scherling C, Smith A. Persistent cognitive changes in breast cancer patients 1 year following completion of chemotherapy. J Int Neuropsychol Soc. 2014;20:370–379. doi: 10.1017/S1355617713001215. [DOI] [PubMed] [Google Scholar]
- 60.Quesnel C, Savard J, Ivers H. Cognitive impairments associated with breast cancer treatments: results from a longitudinal study. Breast Cancer Res Treat. 2009;116:113–123. doi: 10.1007/s10549-008-0114-2. [DOI] [PubMed] [Google Scholar]
- 61.Vearncombe KJ, Rolfe M, Wright M, Pachana NA, Andrew B, Beadle G. Predictors of cognitive decline after chemotherapy in breast cancer patients. J Int Neuropsychol Soc. 2009;15:951–962. doi: 10.1017/S1355617709990567. [DOI] [PubMed] [Google Scholar]
- 62.Ahles TA, Saykin AJ, McDonald BC, et al. Longitudinal assessment of cognitive changes associated with adjuvant treatment for breast cancer: impact of age and cognitive reserve. J Clin Oncol. 2010;28:4434–4440. doi: 10.1200/JCO.2009.27.0827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Kvale EA, Clay OJ, Ross-Meadows LA, et al. Cognitive speed of processing and functional declines in older cancer survivors: an analysis of data from the ACTIVE trial. Eur J Cancer Care. 2010;19:110–117. doi: 10.1111/j.1365-2354.2008.01018.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Reid-Arndt SA, Hsieh C, Perry MC. Neuropsychological functioning and quality of life during the first year after completing chemotherapy for breast cancer. Psychooncology. 2010;19:535–544. doi: 10.1002/pon.1581. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Tager FA, McKinley PS, Schnabel FR, et al. The cognitive effects of chemotherapy in post-menopausal breast cancer patients: a controlled longitudinal study. Breast Cancer Res Treat. 2010;123:25–34. doi: 10.1007/s10549-009-0606-8. [DOI] [PubMed] [Google Scholar]
- 66.Deprez S, Amant F, Yigit R, et al. Chemotherapy-induced structural changes in cerebral white matter and its correlation with impaired cognitive functioning in breast cancer patients. Hum Brain Mapp. 2011;32:480–493. doi: 10.1002/hbm.21033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Biglia N, Bounous VE, Malabaila A, et al. Objective and self-reported cognitive dysfunction in breast cancer women treated with chemotherapy: a prospective study. Eur J Cancer Care. 2012;21:485–492. doi: 10.1111/j.1365-2354.2011.01320.x. [DOI] [PubMed] [Google Scholar]
- 68.Wieneke MH, Dienst ER. Neuropsychological assessment of cognitive-functioning following chemotherapy for breast-cancer. Psychooncology. 1995;4:61–66. [Google Scholar]
- 69.van Dam FS, Schagen SB, Muller MJ, et al. Impairment of cognitive function in women receiving adjuvant treatment for high-risk breast cancer: high-dose versus standard-dose chemotherapy. J Natl Cancer Inst. 1998;90:210–218. doi: 10.1093/jnci/90.3.210. [DOI] [PubMed] [Google Scholar]
- 70.Collins B, MacKenzie J, Tasca GA, Scherling C, Smith A. Cognitive effects of chemotherapy in breast cancer patients: a dose-response study. Psychooncology. 2013;22:1517–1527. doi: 10.1002/pon.3163. [DOI] [PubMed] [Google Scholar]
- 71.Palmer JL, Trotter T, Joy AA, Carlson LE. Cognitive effects of tamoxifen in pre-menopausal women with breast cancer compared to healthy controls. J Cancer Surviv. 2008;2:275–282. doi: 10.1007/s11764-008-0070-1. [DOI] [PubMed] [Google Scholar]
- 72.Batalo M, Nagaiah G, Abraham J. Cognitive dysfunction in postmenopausal breast cancer patients on aromatase inhibitors. Expert Rev Anticancer Ther. 2011;11:1277–1282. doi: 10.1586/era.11.112. [DOI] [PubMed] [Google Scholar]
- 73.Jenkins V, Shilling V, Fallowfield L, Howell A, Hutton S. Does hormone therapy for the treatment of breast cancer have a detrimental effect on memory and cognition? A pilot study. Psychooncology. 2004;13:61–66. doi: 10.1002/pon.709. [DOI] [PubMed] [Google Scholar]
- 74.Jenkins VA, Ambroisine LM, Atkins L, Cuzick J, Howell A, Fallowfield LJ. Effects of anastrozole on cognitive performance in postmenopausal women: a randomised, double-blind chemoprevention trial (IBIS II) Lancet Oncol. 2008;9:953–961. doi: 10.1016/S1470-2045(08)70207-9. [DOI] [PubMed] [Google Scholar]
- 75.Phillips KA, Ribi K, Sun ZX, et al. Cognitive function in postmenopausal women receiving adjuvant letrozole or tamoxifen for breast cancer in the BIG 1–98 randomized trial. Breast. 2010;19:388–395. doi: 10.1016/j.breast.2010.03.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Burstein HJ, Temin S, Anderson H, et al. Adjuvant endocrine therapy for women with hormone receptor-positive breast cancer: American Society of Clinical Oncology clinical practice guideline focused update. J Clin Oncol. 2014;32:2255–2269. doi: 10.1200/JCO.2013.54.2258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kierkegaard M, Einarsson U, Gottberg K, von Koch L, Holmqvist LW. The relationship between walking, manual dexterity, cognition and activity/participation in persons with multiple sclerosis. Mult Scler. 2012;18:639–646. doi: 10.1177/1352458511426736. [DOI] [PubMed] [Google Scholar]
- 78.Reid-Arndt SA, Yee A, Perry MC, Hsieh C. Cognitive and psychological factors associated with early posttreatment functional outcomes in breast cancer survivors. J Psychosoc Oncol. 2009;27:415–434. doi: 10.1080/07347330903183117. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Myers JS. Chemotherapy-related cognitive impairment: the breast cancer experience. Oncol Nurs Forum. 2012;39:E31–E40. doi: 10.1188/12.ONF.E31-E40. [DOI] [PubMed] [Google Scholar]
- 80.Short PF, Vasey JJ, Tunceli K. Employment pathways in a large cohort of adult cancer survivors. Cancer. 2005;103:1292–1301. doi: 10.1002/cncr.20912. [DOI] [PubMed] [Google Scholar]
- 81.Koppelmans V, Breteler MM, Boogerd W, Seynaeve C, Gundy C, Schagen SB. Neuropsychological performance in survivors of breast cancer more than 20 years after adjuvant chemotherapy. J Clin Oncol. 2012;30:1080–1086. doi: 10.1200/JCO.2011.37.0189. [DOI] [PubMed] [Google Scholar]
- 82.Salthouse TA. Neuroanatomical substrates of age-related cognitive decline [serial online] Psychol Bull. 2011;137:753. doi: 10.1037/a0023262. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Nicoll J, Savva G, Stewart J, Matthews F, Brayne C, Ince P. Association between APOE genotype, neuropathology and dementia in the older population of England and Wales. Neuropathol Appl Neurobiol. 2011;37:285–294. doi: 10.1111/j.1365-2990.2010.01130.x. [DOI] [PubMed] [Google Scholar]
- 84.Wefel JS, Vardy J, Ahles T, Schagen SB. International Cognition and Cancer Task Force recommendations to harmonise studies of cognitive function in patients with cancer. Lancet Oncol. 2011;12:703–708. doi: 10.1016/S1470-2045(10)70294-1. [DOI] [PubMed] [Google Scholar]
- 85.Kaiser J, Dietrich J. Challenges in research on the neural basis of “chemobrain”. Translational Neurosci. 2014;5:222–225. [Google Scholar]
- 86.Monje M, Dietrich J. Cognitive side effects of cancer therapy demonstrate a functional role for adult neurogenesis. Behav Brain Res. 2012;227:376–379. doi: 10.1016/j.bbr.2011.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Seigers R, Schagen SB, Van Tellingen O, Dietrich J. Chemotherapy-related cognitive dysfunction: current animal studies and future directions. Brain Imaging Behav. 2013;7:453–459. doi: 10.1007/s11682-013-9250-3. [DOI] [PubMed] [Google Scholar]
- 88.Pereira Dias G, Hollywood R, Bevilaqua MC, et al. Consequences of cancer treatments on adult hippocampal neurogenesis: implications for cognitive function and depressive symptoms. Neuro Oncol. 2014;16:476–492. doi: 10.1093/neuonc/not321. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Han R, Yang YM, Dietrich J, Luebke A, Mayer-Proschel M, Noble M. Systemic 5-fluorouracil treatment causes a syndrome of delayed myelin destruction in the central nervous system [serial online] J Biol. 2008;7:12. doi: 10.1186/jbiol69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Dietrich J, Han R, Yang Y, Mayer-Proschel M, Noble M. CNS progenitor cells and oligodendrocytes are targets of chemotherapeutic agents in vitro and in vivo [serial online] J Biol. 2006;5:22. doi: 10.1186/jbiol50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Briones T, Woods J, Wadowska M. Chronic neuroinflammation and cognitive impairment following transient global cerebral ischemia: role of fractalkine/ CX3CR1 signaling [serial online] J Neuroinflammation. 2014;11:13. doi: 10.1186/1742-2094-11-13. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 92.Kaya E, Keskin L, Aydogdu I, Kuku I, Bayraktar N, Erkut M. Oxidant/antioxidant parameters and their relationship with chemotherapy in Hodgkin’s lymphoma. J Int Med Res. 2005;33:687–692. doi: 10.1177/147323000503300611. [DOI] [PubMed] [Google Scholar]
- 93.Smith J, Ladi E, Mayer-Proschel M, Noble M. Redox state is a central modulator of the balance between self-renewal and differentiation in a dividing glial precursor cell. Proc Natl Acad Sci U S A. 2000;97:10032–10037. doi: 10.1073/pnas.170209797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Seigers R, Timmermans J, van der Horn HJ, et al. Methotrexate reduces hippocampal blood vessel density and activates microglia in rats but does not elevate central cytokine release. Behav Brain Res. 2010;207:265–272. doi: 10.1016/j.bbr.2009.10.009. [DOI] [PubMed] [Google Scholar]
- 95.Helal GK, Aleisa AM, Helal OK, et al. Metallothionein induction reduces caspase-3 activity and TNFalpha levels with preservation of cognitive function and intact hippocampal neurons in carmustine-treated rats. Oxid Med Cell Longev. 2009;2:26–35. doi: 10.4161/oxim.2.1.7901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Madhyastha S, Somayaji S, Rao M, Nalini K, Bairy KL. Hippocampal brain amines in methotrexate-induced learning and memory deficit. Can J Physiol Pharmacol. 2002;80:1076–1084. doi: 10.1139/y02-135. [DOI] [PubMed] [Google Scholar]
- 97.Landry M, Di Paolo T. Effect of chronic estradiol, tamoxifen or raloxifene treatment on serotonin 5-HT1A receptor. Brain Res Mol Brain Res. 2003;112:82–89. doi: 10.1016/s0169-328x(03)00049-4. [DOI] [PubMed] [Google Scholar]
- 98.Bethea CL, Mirkes SJ, Su A, Michelson D. Effects of oral estrogen, raloxifene and arzoxifene on gene expression in serotonin neurons of macaques. Psychoneuroendocrinology. 2002;27:431–445. doi: 10.1016/s0306-4530(01)00054-3. [DOI] [PubMed] [Google Scholar]
- 99.Cyr M, Landry M, Di Paolo T. Modulation by estrogen-receptor directed drugs of 5-hydroxytryptamine-2A receptors in rat brain. Neuropsychopharmacology. 2000;23:69–78. doi: 10.1016/S0893-133X(00)00085-3. [DOI] [PubMed] [Google Scholar]
- 100.Grassi S, Tozzi A, Costa C, et al. Neural 17β-estradiol facilitates long-term potentiation in the hippocampal CA1 region. Neuroscience. 2011;192:67–73. doi: 10.1016/j.neuroscience.2011.06.078. [DOI] [PubMed] [Google Scholar]
- 101.Aydin M, Yilmaz B, Alcin E, Nedzvetsky VS, Sahin Z, Tuzcu M. Effects of letrozole on hippocampal and cortical catecholaminergic neurotransmitter levels, neural cell adhesion molecule expression and spatial learning and memory in female rats. Neuroscience. 2008;151:186–194. doi: 10.1016/j.neuroscience.2007.09.005. [DOI] [PubMed] [Google Scholar]
- 102.Mook D, Felger J, Graves F, Wallen K, Wilson ME. Tamoxifen fails to affect central serotonergic tone but increases indices of anxiety in female rhesus macaques. Psychoneuroendocrinology. 2005;30:273–283. doi: 10.1016/j.psyneuen.2004.08.005. [DOI] [PubMed] [Google Scholar]
- 103.Chen D, Wu CF, Shi B, Xu YM. Tamoxifen and toremifene cause impairment of learning and memory function in mice. Pharmacol Biochem Behav. 2002;71:269–276. doi: 10.1016/s0091-3057(01)00656-6. [DOI] [PubMed] [Google Scholar]
- 104.Kokiko ON, Murashov AK, Hoane MR. Administration of raloxifene reduces sensorimotor and working memory deficits following traumatic brain injury. Behav Brain Res. 2006;170:233–240. doi: 10.1016/j.bbr.2006.02.026. [DOI] [PubMed] [Google Scholar]
- 105.Moradpour F, Naghdi N, Fathollahi Y. Anastrozole improved testosterone-induced impairment acquisition of spatial learning and memory in the hippocampal CA1 region in adult male rats. Behav Brain Res. 2006;175:223–232. doi: 10.1016/j.bbr.2006.08.037. [DOI] [PubMed] [Google Scholar]
- 106.Abraham J, Haut M, Moran M, Filburn S, Lemiuex S, Kuwabara H. Adjuvant chemotherapy for breast cancer: effects on cerebral white matter seen in diffusion tensor imaging. Clin Breast Cancer. 2008;8:88–91. doi: 10.3816/CBC.2008.n.007. [DOI] [PubMed] [Google Scholar]
- 107.Bergouignan L, Lefranc JP, Chupin M, Morel N, Spano JP, Fossati P. Breast cancer affects both the hippocampus volume and the episodic autobiographical memory retrieval [serial online] PLoS One. 2011;6:e25349. doi: 10.1371/journal.pone.0025349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Conroy SK, McDonald BC, Smith DJ, et al. Alterations in brain structure and function in breast cancer survivors: effect of post-chemotherapy interval and relation to oxidative DNA damage. Breast Cancer Res Treat. 2013;137:493–502. doi: 10.1007/s10549-012-2385-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.de Ruiter MB, Reneman L, Boogerd W, et al. Late effects of high-dose adjuvant chemotherapy on white and gray matter in breast cancer survivors: converging results from multimodal magnetic resonance imaging. Hum Brain Mapp. 2012;33:2971–2983. doi: 10.1002/hbm.21422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Deprez S, Amant F, Smeets A, et al. Longitudinal assessment of chemotherapy-induced structural changes in cerebral white matter and its correlation with impaired cognitive functioning. J Clin Oncol. 2012;30:274–281. doi: 10.1200/JCO.2011.36.8571. [DOI] [PubMed] [Google Scholar]
- 111.Eberling JL, Wu C, Tong-Turnbeaugh R, Jagust WJ. Estrogen- and tamoxifen-associated effects on brain structure and function. Neuroimage. 2004;21:364–371. doi: 10.1016/j.neuroimage.2003.08.037. [DOI] [PubMed] [Google Scholar]
- 112.Hosseini SM, Koovakkattu D, Kesler SR. Altered small-world properties of gray matter networks in breast cancer [serial online] BMC Neurol. 2012;12:28. doi: 10.1186/1471-2377-12-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Inagaki M, Yoshikawa E, Matsuoka Y, et al. Smaller regional volumes of brain gray and white matter demonstrated in breast cancer survivors exposed to adjuvant chemotherapy. Cancer. 2007;109:146–156. doi: 10.1002/cncr.22368. [DOI] [PubMed] [Google Scholar]
- 114.Kesler S, Janelsins M, Koovakkattu D, et al. Reduced hippocampal volume and verbal memory performance associated with interleukin-6 and tumor necrosis factor-alpha levels in chemotherapy-treated breast cancer survivors. Brain Behav Immun. 2013;30(suppl):S109–S116. doi: 10.1016/j.bbi.2012.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Koppelmans V, de Groot M, de Ruiter MB, et al. Global and focal white matter integrity in breast cancer survivors 20 years after adjuvant chemotherapy. Hum Brain Mapp. 2014;35:889–899. doi: 10.1002/hbm.22221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Koppelmans V, de Ruiter MB, van der Lijn F, et al. Global and focal brain volume in long-term breast cancer survivors exposed to adjuvant chemotherapy. Breast Cancer Res Treat. 2012;132:1099–1106. doi: 10.1007/s10549-011-1888-1. [DOI] [PubMed] [Google Scholar]
- 117.McDonald BC, Conroy SK, Ahles TA, West JD, Saykin AJ. Gray matter reduction associated with systemic chemotherapy for breast cancer: a prospective MRI study. Breast Cancer Res Treat. 2010;123:819–828. doi: 10.1007/s10549-010-1088-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.McDonald BC, Conroy SK, Smith DJ, West JD, Saykin AJ. Frontal gray matter reduction after breast cancer chemotherapy and association with executive symptoms: a replication and extension study. Brain Behav Immun. 2013;30(suppl):S117–S125. doi: 10.1016/j.bbi.2012.05.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Sporns O. The human connectome: a complex network. Ann NY Acad Sci. 2011;1224:109–125. doi: 10.1111/j.1749-6632.2010.05888.x. [DOI] [PubMed] [Google Scholar]
- 120.Kesler SR. Default mode network as a potential biomarker of chemotherapy-related brain injury. Neurobiol Aging. 2014;35(suppl 2):S11–S19. doi: 10.1016/j.neurobiolaging.2014.03.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Bruno J, Hosseini SM, Kesler S. Altered resting state functional brain network topology in chemotherapy-treated breast cancer survivors. Neurobiol Dis. 2012;48:329–338. doi: 10.1016/j.nbd.2012.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Cimprich B, Reuter-Lorenz P, Nelson J, et al. Prechemotherapy alterations in brain function in women with breast cancer. J Clin Exp Neuropsychol. 2010;32:324–331. doi: 10.1080/13803390903032537. [DOI] [PubMed] [Google Scholar]
- 123.Conroy SK, McDonald BC, Ahles TA, West JD, Saykin AJ. Chemotherapy-induced amenorrhea: a prospective study of brain activation changes and neurocognitive correlates. Brain Imaging Behav. 2013;7:491–500. doi: 10.1007/s11682-013-9240-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.de Ruiter MB, Reneman L, Boogerd W, et al. Cerebral hyporesponsiveness and cognitive impairment 10 years after chemotherapy for breast cancer. Hum Brain Mapp. 2011;32:1206–1219. doi: 10.1002/hbm.21102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Deprez S, Vandenbulcke M, Peeters R, et al. Longitudinal assessment of chemotherapy-induced alterations in brain activation during multitasking and its relation with cognitive complaints. J Clin Oncol. 2014;32:2031–2038. doi: 10.1200/JCO.2013.53.6219. [DOI] [PubMed] [Google Scholar]
- 126.Dumas JA, Makarewicz J, Schaubhut GJ, et al. Chemotherapy altered brain functional connectivity in women with breast cancer: a pilot study. Brain Imaging Behav. 2013;7:524–532. doi: 10.1007/s11682-013-9244-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Ferguson RJ, McDonald BC, Saykin AJ, Ahles TA. Brain structure and function differences in monozygotic twins: possible effects of breast cancer chemotherapy. J Clin Oncol. 2007;25:3866–3870. doi: 10.1200/JCO.2007.10.8639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Hosseini SM, Kesler SR. Multivariate pattern analysis of fMRI in breast cancer survivors and healthy women. J Int Neuropsychol Soc. 2013;19:1–11. doi: 10.1017/S1355617713001173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Kesler SR, Kent JS, O’Hara R. Prefrontal cortex and executive function impairments in primary breast cancer. Arch Neurol. 2011;68:1447–1453. doi: 10.1001/archneurol.2011.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Kesler SR, Wefel JS, Hosseini SM, Cheung M, Watson CL, Hoeft F. Default mode network connectivity distinguishes chemotherapy-treated breast cancer survivors from controls. Proc Natl Acad Sci. 2013;110:11600–11605. doi: 10.1073/pnas.1214551110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lopez Zunini RA, Scherling C, Wallis N, et al. Differences in verbal memory retrieval in breast cancer chemotherapy patients compared to healthy controls: a prospective fMRI study. Brain Imaging Behav. 2012;7:460–477. doi: 10.1007/s11682-012-9213-0. [DOI] [PubMed] [Google Scholar]
- 132.McDonald BC, Conroy SK, Ahles TA, West JD, Saykin AJ. Alterations in brain activation during working memory processing associated with breast cancer and treatment: a prospective functional magnetic resonance imaging study. J Clin Oncol. 2012;30:2500–2508. doi: 10.1200/JCO.2011.38.5674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Scherling C, Collins B, Mackenzie J, Bielajew C, Smith A. Prechemotherapy differences in response inhibition in breast cancer patients compared to controls: a functional magnetic resonance imaging study. J Clin Exp Neuropsychol. 2012;34:543–560. doi: 10.1080/13803395.2012.666227. [DOI] [PubMed] [Google Scholar]
- 134.Silverman DH, Dy CJ, Castellon SA, et al. Altered frontocortical, cerebellar, and basal ganglia activity in adjuvant-treated breast cancer survivors 5–10 years after chemotherapy. Breast Cancer Res Treat. 2007;103:303–311. doi: 10.1007/s10549-006-9380-z. [DOI] [PubMed] [Google Scholar]
- 135.Pomykala KL, Ganz PA, Bower JE, et al. The association between pro-inflammatory cytokines, regional cerebral metabolism, and cognitive complaints following adjuvant chemotherapy for breast cancer. Brain Imaging Behav. 2013;7:511–523. doi: 10.1007/s11682-013-9243-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Ponto LL, Menda Y, Magnotta VA, Yamada TH, Denburg NL, Schultz SK. Frontal hypometabolism in elderly breast cancer survivors determined by [18 F]fluorodeoxyglucose (FDG) positron emission tomography (PET): a pilot study [published online ahead of print September 1, 2014] Int J Geriatr Psychiatry. 2014 doi: 10.1002/gps.4189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Nudelman KN, Wang Y, McDonald BC, et al. Altered cerebral blood flow one month after systemic chemotherapy for breast cancer: a prospective study using pulsed arterial spin labeling MRI perfusion [serial online] PLoS One. 2014;9:e96713. doi: 10.1371/journal.pone.0096713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Mathan S, Smart A, Ververs T, Feuerstein M. Towards an index of cognitive efficacy EEG-based estimation of cognitive load among individuals experiencing cancer-related cognitive decline. Conf Proc IEEE Eng Med Bio Soc. 2010;2010:6595–6598. doi: 10.1109/IEMBS.2010.5627126. [DOI] [PubMed] [Google Scholar]
- 139.Hunter AM, Kwan L, Ercoli LM, et al. Quantitative electroencephalography biomarkers of cognitive complaints after adjuvant therapy in breast cancer survivors: a pilot study. Psychooncology. 2014;23:713–715. doi: 10.1002/pon.3487. [DOI] [PubMed] [Google Scholar]
- 140.Kreukels BP, Hamburger HL, de Ruiter MB, et al. ERP amplitude and latency in breast cancer survivors treated with adjuvant chemotherapy. Clin Neurophysiol. 2008;119:533–541. doi: 10.1016/j.clinph.2007.11.011. [DOI] [PubMed] [Google Scholar]
- 141.Kreukels BP, Schagen SB, Ridderinkhof KR, Boogerd W, Hamburger HL, van Dam FS. Electrophysiological correlates of information processing in breast-cancer patients treated with adjuvant chemotherapy. Breast Cancer Res Treat. 2005;94:53–61. doi: 10.1007/s10549-005-7093-3. [DOI] [PubMed] [Google Scholar]
- 142.Kesler SR, Watson C, Koovakkattu D, et al. Elevated prefrontal myoinositol and choline following breast cancer chemotherapy. Brain Imaging Behav. 2013;7:501–510. doi: 10.1007/s11682-013-9228-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Brown MS, Stemmer SM, Simon JH, et al. White matter disease induced by high-dose chemotherapy: longitudinal study with MR imaging and proton spectroscopy. Am J Neuroradiol. 1998;19:217–221. [PMC free article] [PubMed] [Google Scholar]
- 144.Koppelmans V, Breteler MM, Boogerd W, Seynaeve C, Schagen SB. Late effects of adjuvant chemotherapy for adult onset non-CNS cancer; cognitive impairment, brain structure and risk of dementia. Crit Rev Oncol Hematol. 2013;88:87–101. doi: 10.1016/j.critrevonc.2013.04.002. [DOI] [PubMed] [Google Scholar]
- 145.Stouten-Kemperman MM, de Ruiter MB, Koppelmans V, Boogerd W, Reneman L, Schagen SB. Neurotoxicity in breast cancer survivors ≥10 years post-treatment is dependent on treatment type [published online ahead of print May 25, 2014] Brain Imaging Behav. 2014 doi: 10.1007/s11682-014-9305-0. [DOI] [PubMed] [Google Scholar]
- 146.Hurria A, Patel SK, Mortimer J, et al. The effect of aromatase inhibition on the cognitive function of older patients with breast cancer. Clin Breast Cancer. 2014;14:132–140. doi: 10.1016/j.clbc.2013.10.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Ganz PA, Bower JE, Kwan L, et al. Does tumor necrosis factor-alpha (TNF-α) play a role in post-chemotherapy cerebral dysfunction? Brain Behav Immun. 2013;30:S99–S108. doi: 10.1016/j.bbi.2012.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Zick SM, Zwickey H, Wood L, et al. Preliminary differences in peripheral immune markers and brain metabolites between fatigued and non-fatigued breast cancer survivors: a pilot study [published online ahead of print November 13, 2013] Brain Imaging Behav. 2013 doi: 10.1007/s11682-013-9270-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Askren MK, Jung M, Berman MG, et al. Neuromarkers of fatigue and cognitive complaints following chemotherapy for breast cancer: a prospective fMRI investigation. Breast Cancer Res Treat. 2014;147:445–455. doi: 10.1007/s10549-014-3092-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Kim LS, Hwang HS, Jon DI, Ham BJ, Seok JH. Dysfunction of the neural network associated with sustained attention in cancer patients with clinically significant depressive symptoms. Neurosci Lett. 2008;447:1–6. doi: 10.1016/j.neulet.2008.09.077. [DOI] [PubMed] [Google Scholar]
- 151.Vardy J. Cognitive function in breast cancer survivors. Cancer Treat Res. 2009;151:387–419. doi: 10.1007/978-0-387-75115-3_24. [DOI] [PubMed] [Google Scholar]
- 152.Castellon S, Ganz PA. Neuropsychological studies in breast cancer: in search of chemobrain. Breast Cancer Res Treat. 2009;116:125–127. doi: 10.1007/s10549-008-0211-2. [DOI] [PubMed] [Google Scholar]
- 153.Zhang D, Shen D. Multi-modal multi-task learning for joint prediction of multiple regression and classification variables in Alzheimer’s disease. Neuroimage. 2012;59:895–907. doi: 10.1016/j.neuroimage.2011.09.069. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Young J, Modat M, Cardoso MJ, et al. Accurate multimodal probabilistic prediction of conversion to Alzheimer’s disease in patients with mild cognitive impairment. Neuroimage Clin. 2013;2:735–745. doi: 10.1016/j.nicl.2013.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Chincarini A, Bosco P, Calvini P, et al. Local MRI analysis approach in the diagnosis of early and prodromal Alzheimer’s disease. Neuroimage. 2011;58:469–480. doi: 10.1016/j.neuroimage.2011.05.083. [DOI] [PubMed] [Google Scholar]
- 156.Mahmoudi A, Takerkart S, Regragui F, Boussaoud D, Brovelli A. Multivoxel pattern analysis for FMRI data: a review [serial online] Comput Math Methods Med. 2012;2012:961257. doi: 10.1155/2012/961257. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.O’Toole AJ, Jiang F, Abdi H, Penard N, Dunlop JP, Parent MA. Theoretical, statistical, and practical perspectives on pattern-based classification approaches to the analysis of functional neuroimaging data. J Cogn Neurosci. 2007;19:1735–1752. doi: 10.1162/jocn.2007.19.11.1735. [DOI] [PubMed] [Google Scholar]
- 158.Orru G, Pettersson-Yeo W, Marquand AF, Sartori G, Mechelli A. Using Support Vector Machine to identify imaging biomarkers of neurological and psychiatric disease: a critical review. Neurosci Biobehav Rev. 2012;36:1140–1152. doi: 10.1016/j.neubiorev.2012.01.004. [DOI] [PubMed] [Google Scholar]
- 159.Cherrier MM, Anderson K, David D, et al. A randomized trial of cognitive rehabilitation in cancer survivors. Life Sci. 2013;93:617–622. doi: 10.1016/j.lfs.2013.08.011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Ercoli LM, Castellon SA, Hunter AM, et al. Assessment of the feasibility of a rehabilitation intervention program for breast cancer survivors with cognitive complaints. Brain Imaging Behav. 2013;7:543–553. doi: 10.1007/s11682-013-9237-0. [DOI] [PubMed] [Google Scholar]
- 161.Ferguson RJ, Ahles TA, Saykin AJ, et al. Cognitive-behavioral management of chemotherapy-related cognitive change. Psychooncology. 2007;16:772–777. doi: 10.1002/pon.1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Ferguson RJ, McDonald BC, Rocque MA, et al. Development of CBT for chemotherapy-related cognitive change: results of a waitlist control trial. Psychooncology. 2012;21:176–186. doi: 10.1002/pon.1878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.McDougall GJ, Becker H, Acee TW, Vaughan PW, Delville CL. Symptom management of affective and cognitive disturbance with a group of cancer survivors. Arch Psychiatr Nurs. 2011;25:24–35. doi: 10.1016/j.apnu.2010.05.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Poppelreuter M, Weis J, Bartsch HH. Effects of specific neuropsychological training programs for breast cancer patients after adjuvant chemotherapy. J Psychosoc Oncol. 2009;27:274–296. doi: 10.1080/07347330902776044. [DOI] [PubMed] [Google Scholar]
- 165.Schuurs A, Green HJ. A feasibility study of group cognitive rehabilitation for cancer survivors: enhancing cognitive function and quality of life. Psychooncology. 2013;22:1043–1049. doi: 10.1002/pon.3102. [DOI] [PubMed] [Google Scholar]
- 166.Kesler S, Hosseini SMH, Heckler C, et al. Cognitive training for improving executive function in chemotherapy-treated breast cancer survivors. Clin Breast Cancer. 2013;13:299–306. doi: 10.1016/j.clbc.2013.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Von Ah D, Carpenter JS, Saykin A, et al. Advanced cognitive training for breast cancer survivors: a randomized controlled trial. Breast Cancer Res Treat. 2012;135:799–809. doi: 10.1007/s10549-012-2210-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Galantino ML, Greene L, Daniels L, Dooley B, Muscatello L, O’Donnell L. Longitudinal impact of yoga on chemotherapy-related cognitive impairment and quality of life in women with early stage breast cancer: a case series. Explore. 2012;8:127–135. doi: 10.1016/j.explore.2011.12.001. [DOI] [PubMed] [Google Scholar]
- 169.Janelsins M, Peppone L, Heckler C, et al. YOCAS yoga, fatigue, memory difficulty, and quality of life: results from a URCC CCOP randomized, controlled clinical trial among 358 cancer survivors [abstract] J Clin Oncol. 2012;30(suppl):Abstract 9142. [Google Scholar]
- 170.Reid-Arndt SA, Matsuda S, Cox CR. Tai Chi effects on neuropsychological, emotional, and physical functioning following cancer treatment: a pilot study. Complement Ther Clin Pract. 2012;18:26–30. doi: 10.1016/j.ctcp.2011.02.005. [DOI] [PubMed] [Google Scholar]
- 171.Oh B, Butow PN, Mullan BA, et al. Effect of medical Qigong on cognitive function, quality of life, and a biomarker of inflammation in cancer patients: a randomized controlled trial. Support Care Cancer. 2012;20:1235–1242. doi: 10.1007/s00520-011-1209-6. [DOI] [PubMed] [Google Scholar]
- 172.Alvarez J, Meyer FL, Granoff DL, Lundy A. The effect of EEG biofeedback on reducing postcancer cognitive impairment. Integr Cancer Ther. 2013;12:475–487. doi: 10.1177/1534735413477192. [DOI] [PubMed] [Google Scholar]
- 173.Cajigal S. Health insurers deny coverage for cognitive rehabilitation. Neurol Today. 2007;7:38–41. [Google Scholar]
- 174.Steinkuehler C, Squire K, Barab S. Games, Learning and Society. New York, NY: Cambridge University Press; 2012. [Google Scholar]
- 175.Wolinsky FD, Vander Weg MW, Howren MB, Jones MP, Dotson MM. A randomized controlled trial of cognitive training using a visual speed of processing intervention in middle aged and older adults [serial online] PLoS One. 2013;8:e61624. doi: 10.1371/journal.pone.0061624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Valenzuela M, Sachdev P. Can cognitive exercise prevent the onset of dementia? Systematic review of randomized clinical trials with longitudinal follow-up. Am J Geriatr Psychiatry. 2009;17:179–187. doi: 10.1097/JGP.0b013e3181953b57. [DOI] [PubMed] [Google Scholar]
- 177.Sanoff HK, Deal AM, Krishnamurthy J, et al. Effect of cytotoxic chemotherapy on markers of molecular age in patients with breast cancer [serial online] J Natl Cancer Inst. 2014;106:dju057. doi: 10.1093/jnci/dju057. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Ahles TA. Brain vulnerability to chemotherapy toxicities. Psychooncology. 2012;21:1141–1148. doi: 10.1002/pon.3196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Anguera JA, Boccanfuso J, Rintoul JL, et al. Video game training enhances cognitive control in older adults. Nature. 2013;501:97–101. doi: 10.1038/nature12486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Jaeggi SM, Buschkuehl M, Shah P, Jonides J. The role of individual differences in cognitive training and transfer. Mem Cognit. 2013;42:464–480. doi: 10.3758/s13421-013-0364-z. [DOI] [PubMed] [Google Scholar]
- 181.Latham AJ, Patston LL, Tippett LJ. The virtual brain: 30 years of video-game play and cognitive abilities [serial online] Front Psychol. 2013;4:629. doi: 10.3389/fpsyg.2013.00629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Haimov I, Shatil E. Cognitive training improves sleep quality and cognitive function among older adults with insomnia [serial online] PLoS One. 2013;8:e61390. doi: 10.1371/journal.pone.0061390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Åkerlund E, Esbjornsson E, Sunnerhagen KS, Bjorkdahl A. Can computerized working memory training improve impaired working memory, cognition and psychological health? Brain Inj. 2013;27:1649–1657. doi: 10.3109/02699052.2013.830195. [DOI] [PubMed] [Google Scholar]
- 184.Niman NB. The allure of games: toward an updated theory of the leisure class. Games Culture. 2013;8:25–42. [Google Scholar]
- 185.Abuhamdeh S, Csikszentmihalyi M. The importance of challenge for the enjoyment of intrinsically motivated, goal-directed activities. Pers Soc Psychol Bull. 2012;38:317–330. doi: 10.1177/0146167211427147. [DOI] [PubMed] [Google Scholar]
- 186.Park DC, Bischof GN. The aging mind: neuroplasticity in response to cognitive training. Dialogues Clin Neurosci. 2013;15:109–119. doi: 10.31887/DCNS.2013.15.1/dpark. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187.Campbell KL, Neil SE, Winters-Stone KM. Review of exercise studies in breast cancer survivors: attention to principles of exercise training. Br J Sports Med. 2012;46:909–916. doi: 10.1136/bjsports-2010-082719. [DOI] [PubMed] [Google Scholar]
- 188.Voss MW, Vivar C, Kramer AF, van Praag H. Bridging animal and human models of exercise-induced brain plasticity. Trends Cogn Sci. 2013;17:525–544. doi: 10.1016/j.tics.2013.08.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Hayes SM, Hayes JP, Cadden M, Verfaellie M. A review of cardiorespiratory fitness-related neuroplasticity in the aging brain [serial online] Front Aging Neurosci. 2013;5:31. doi: 10.3389/fnagi.2013.00031. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Hotting K, Roder B. Beneficial effects of physical exercise on neuroplasticity and cognition. Neurosci Biobehav Rev. 2013;37:2243–2257. doi: 10.1016/j.neubiorev.2013.04.005. [DOI] [PubMed] [Google Scholar]
- 191.Kramer AF, Erickson KI, Colcombe SJ. Exercise, cognition, and the aging brain. J Appl Physiol. 2006;101:1237–1242. doi: 10.1152/japplphysiol.00500.2006. [DOI] [PubMed] [Google Scholar]
- 192.Speisman RB, Kumar A, Rani A, Foster TC, Ormerod BK. Daily exercise improves memory, stimulates hippocampal neurogenesis and modulates immune and neuroimmune cytokines in aging rats. Brain Behav Immun. 2013;28:25–43. doi: 10.1016/j.bbi.2012.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Blake H. Physical activity and exercise in the treatment of depression [serial online] Front Psychiatry. 2012;3:106. doi: 10.3389/fpsyt.2012.00106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Marques-Aleixo I, Oliveira PJ, Moreira PI, Magalhaes J, Ascensao A. Physical exercise as a possible strategy for brain protection: evidence from mitochondrial-mediated mechanisms. Prog Neurobiol. 2012;99:149–162. doi: 10.1016/j.pneurobio.2012.08.002. [DOI] [PubMed] [Google Scholar]
- 195.Fardell JE, Vardy J, Shah JD, Johnston IN. Cognitive impairments caused by oxaliplatin and 5-fluorouracil chemotherapy are ameliorated by physical activity. Psychopharmacology. 2012;220:183–193. doi: 10.1007/s00213-011-2466-2. [DOI] [PubMed] [Google Scholar]
- 196.Ahlskog JE, Geda YE, Graff-Radford NR, Petersen RC. Physical exercise as a preventive or disease-modifying treatment of dementia and brain aging. Mayo Clin Proc. 2011;86:876–884. doi: 10.4065/mcp.2011.0252. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197.Snowden M, Steinman L, Mochan K, et al. Effect of exercise on cognitive performance in community-dwelling older adults: review of intervention trials and recommendations for public health practice and research. J Am Geriatr Soc. 2011;59:704–716. doi: 10.1111/j.1532-5415.2011.03323.x. [DOI] [PubMed] [Google Scholar]
- 198.Hall PA, Fong G, Epp L, Elias LJ. Executive function moderates the intention-behavior link for physical activity and dietary behavior. Psychol Health. 2008;23:309–326. doi: 10.1080/14768320701212099. [DOI] [PubMed] [Google Scholar]
- 199.Hall PA, Zehr C, Paulitzki J, Rhodes R. Implementation intentions for physical activity behavior in older adult women: an examination of executive function as a moderator of treatment effects. Ann Behav Med. 2014;48:130–136. doi: 10.1007/s12160-013-9582-7. [DOI] [PubMed] [Google Scholar]
- 200.McAuley E, Mullen SP, Szabo AN, et al. Self-regulatory processes and exercise adherence in older adults: executive function and self-efficacy effects. Am J Prev Med. 2011;41:284–290. doi: 10.1016/j.amepre.2011.04.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201.Best JR, Nagamatsu LS, Liu-Ambrose T. Improvements to executive function during exercise training predict maintenance of physical activity over the following year [serial online] Front Hum Neurosci. 2014;8:353. doi: 10.3389/fnhum.2014.00353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Hall PA, Elias LJ, Fong GT, Harrison AH, Borowsky R, Sarty GE. A social neuroscience perspective on physical activity. J Sport Exerc Psychol. 2008;30:432–449. doi: 10.1123/jsep.30.4.432. [DOI] [PubMed] [Google Scholar]
- 203.Hofmann W, Schmeichel BJ, Baddeley AD. Executive functions and self-regulation. Trends Cogn Sci. 2012;16:174–180. doi: 10.1016/j.tics.2012.01.006. [DOI] [PubMed] [Google Scholar]
- 204.Hong D, Scaletta Kent J, Kesler S. Cognitive profile of Turner syndrome. Dev Disabil Res Rev. 2009;15:270–278. doi: 10.1002/ddrr.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205.Elliott R. Executive functions and their disorders. Br Med Bull. 2003;65:49–59. doi: 10.1093/bmb/65.1.49. [DOI] [PubMed] [Google Scholar]
- 206.Krull KR, Annett RD, Pan Z, et al. Neurocognitive functioning and health-related behaviours in adult survivors of childhood cancer: a report from the Childhood Cancer Survivor Study. Eur J Cancer. 2011;47:1380–1388. doi: 10.1016/j.ejca.2011.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.Curlik DM, 2nd, Shors TJ. Training your brain: do mental and physical (MAP) training enhance cognition through the process of neurogenesis in the hippocampus? Neuropharmacology. 2013;64:506–514. doi: 10.1016/j.neuropharm.2012.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Karr JE, Areshenkoff CN, Rast P, Garcia-Barrera MA. An empirical comparison of the therapeutic benefits of physical exercise and cognitive training on the executive functions of older adults: a meta-analysis of controlled trials [published online ahead of print June 16, 2014] Neuropsychology. 2014 doi: 10.1037/neu0000101. [DOI] [PubMed] [Google Scholar]
- 209.Kempermann G, Fabel K, Ehninger D, et al. Why and how physical activity promotes experience-induced brain plasticity [serial online] Front Neurosci. 2010;4:189. doi: 10.3389/fnins.2010.00189. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Fabel K, Wolf SA, Ehninger D, Babu H, Leal-Galicia P, Kempermann G. Additive effects of physical exercise and environmental enrichment on adult hippocampal neurogenesis in mice [serial online] Front Neurosci. 2009;3:50. doi: 10.3389/neuro.22.002.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 211.Fabre C, Chamari K, Mucci P, Masse-Biron J, Prefaut C. Improvement of cognitive function by mental and/or individualized aerobic training in healthy elderly subjects. Int J Sports Med. 2002;23:415–421. doi: 10.1055/s-2002-33735. [DOI] [PubMed] [Google Scholar]
- 212.Holzschneider K, Wolbers T, Roder B, Hotting K. Cardiovascular fitness modulates brain activation associated with spatial learning. Neuroimage. 2012;59:3003–3014. doi: 10.1016/j.neuroimage.2011.10.021. [DOI] [PubMed] [Google Scholar]
- 213.Gonzalez-Palau F, Franco M, Bamidis P, et al. The effects of a computer-based cognitive and physical training program in a healthy and mildly cognitive impaired aging sample. Aging Ment Health. 2014;18:838–846. doi: 10.1080/13607863.2014.899972. [DOI] [PubMed] [Google Scholar]
- 214.Stern Y. Cognitive reserve. Neuropsychologia. 2009;47:2015–2028. doi: 10.1016/j.neuropsychologia.2009.03.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215.Correa DD, Ahles TA. Neurocognitive changes in cancer survivors. Cancer J. 2008;14:396–400. doi: 10.1097/PPO.0b013e31818d8769. [DOI] [PubMed] [Google Scholar]
- 216.Sulzer J, Haller S, Scharnowski F, et al. Real-time fMRI neurofeedback: progress and challenges. Neuroimage. 2013;76:386–399. doi: 10.1016/j.neuroimage.2013.03.033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Luber B, Lisanby SH. Enhancement of human cognitive performance using transcranial magnetic stimulation (TMS) Neuroimage. 2014;85:961–970. doi: 10.1016/j.neuroimage.2013.06.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Demirtas-Tatlidede A, Vahabzadeh-Hagh AM, Pascual-Leone A. Can noninvasive brain stimulation enhance cognition in neuropsychiatric disorders? Neuropharmacology. 2013;64:566–578. doi: 10.1016/j.neuropharm.2012.06.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Boggio PS, Valasek CA, Campanha C, et al. Non-invasive brain stimulation to assess and modulate neuroplasticity in Alzheimer’s disease. Neuropsychol Rehabil. 2011;21:703–716. doi: 10.1080/09602011.2011.617943. [DOI] [PubMed] [Google Scholar]
- 220.Bloch Y, Harel EV, Aviram S, Govezensky J, Ratzoni G, Levkovitz Y. Positive effects of repetitive transcranial magnetic stimulation on attention in ADHD subjects: a randomized controlled pilot study. World J Biol Psychiatry. 2010;11:755–758. doi: 10.3109/15622975.2010.484466. [DOI] [PubMed] [Google Scholar]
- 221.Fardell J, Vardy J, Johnston I, Winocur G. Chemotherapy and cognitive impairment: treatment options. Clin Pharmacol Ther. 2011;90:366–376. doi: 10.1038/clpt.2011.112. [DOI] [PubMed] [Google Scholar]
- 222.Gehring K, Roukema JA, Sitskoorn MM. Review of recent studies on interventions for cognitive deficits in patients with cancer. Expert Rev Anticancer Ther. 2012;12:255–269. doi: 10.1586/era.11.202. [DOI] [PubMed] [Google Scholar]
- 223.Von Ah D, Storey S, Jansen CE, Allen DH. Coping strategies and interventions for cognitive changes in patients with cancer. Semin Oncol Nurs. 2013;29:288–299. doi: 10.1016/j.soncn.2013.08.009. [DOI] [PubMed] [Google Scholar]
- 224.Aluise CD, Miriyala S, Noel T, et al. 2-Mer-captoethane sulfonate prevents doxorubicin-induced plasma protein oxidation and TNF-alpha release: implications for the reactive oxygen species-mediated mechanisms of chemobrain. Free Radic Biol Med. 2011;50:1630–1638. doi: 10.1016/j.freeradbiomed.2011.03.009. [DOI] [PubMed] [Google Scholar]
- 225.Konat GW, Kraszpulski M, James I, Zhang HT, Abraham J. Cognitive dysfunction induced by chronic administration of common cancer chemotherapeutics in rats. Metab Brain Dis. 2008;23:325–333. doi: 10.1007/s11011-008-9100-y. [DOI] [PubMed] [Google Scholar]
- 226.Briones TL, Woods J, Wadowska M. Involvement of insulin-like growth factor-1 in chemotherapy-related cognitive impairment [published online ahead of print March 12, 2014] Behav Brain Res. 2014 doi: 10.1016/j.bbr.2014.02.052. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 227.El Beltagy M, Mustafa S, Umka J, et al. Fluoxetine improves the memory deficits caused by the chemotherapy agent 5-fluo-rouracil. Behav Brain Res. 2010;208:112–117. doi: 10.1016/j.bbr.2009.11.017. [DOI] [PubMed] [Google Scholar]
- 228.Dubois M, Lapinte N, Villier V, et al. Chemotherapy-induced long-term alteration of executive functions and hippocampal cell proliferation: role of glucose as adjuvant. Neuropharmacology. 2014;79:234–248. doi: 10.1016/j.neuropharm.2013.11.012. [DOI] [PubMed] [Google Scholar]
- 229.Vijayanathan V, Gulinello M, Ali N, Cole PD. Persistent cognitive deficits, induced by intrathecal methotrexate, are associated with elevated CSF concentrations of excitotoxic glutamate analogs and can be reversed by an NMDA antagonist. Behav Brain Res. 2011;225:491–497. doi: 10.1016/j.bbr.2011.08.006. [DOI] [PubMed] [Google Scholar]
- 230.Cole PD, Vijayanathan V, Ali NF, et al. Memantine protects rats treated with intrathecal methotrexate from developing spatial memory deficits. Clin Cancer Res. 2013;19:4446–4454. doi: 10.1158/1078-0432.CCR-13-1179. [DOI] [PMC free article] [PubMed] [Google Scholar]