Abstract
Chemotherapy-related cognitive impairment (CRCI) can occur during or after chemotherapy and represents a concern for many patients with cancer. Among older patients with cancer, in whom there is little clinical trial evidence examining side effects like CRCI, many unanswered questions remain regarding risk for and resulting adverse outcomes from CRCI. Given the rising incidence of cancer with age, CRCI is of particular concern for older patients with cancer who receive treatment. Therefore, research related to CRCI in older patients with cancers is a high priority. In this manuscript, we discuss current gaps in research highlighting the lack of clinical studies of CRCI in older adults, the complex mechanisms of CRCI, and the challenges in measuring cognitive impairment in older patients with cancer. Although we focus on CRCI, we also discuss cognitive impairment related to cancer itself and other treatment modalities. We highlight several research priorities to improve the study of CRCI in older patients with cancer.
Keywords: Chemotherapy, Cancer treatment, Cognitive impairment, Older patients, Research gaps
1. Introduction
Evidence suggests that cancer treatments can cause cognitive impairment that is subjectively reported or objectively measured using neuropsychological tests.1–4 Patients with cognitive impairment may encounter challenges in daily functioning, decision-making and treatment adherence, leading to decreased quality of life and possibly shorter survival.5–7 Cognitive impairment can also increase caregiver burden. Prevention of cognitive impairment in patients with cancer undergoing treatment is therefore especially important in older patients, given the increasing long-term survival with new treatments and the increasing numbers of older patients living with cancer as a chronic condition.
Cross-sectional and longitudinal studies often do not include enough older patients with cancer to evaluate the interactions that exist among cancer, its treatment, aging and effects on cognition. Assessing cognitive impairment is challenging in the clinical trial setting given lack of routine use of standardized, brief and accurate neuropsychological testing. When cognitive impairment is detected in clinical practice, oncologists are often unprepared to manage the abnormal results. Additionally, there is a lack of evidence-based preventive measures or interventions when cognitive impairment is detected in the cancer population. A study suggested that patients were less likely to accept treatments that may worsen their cognition, but it is unclear how the presence of cognitive impairment in a patient with cancer affects oncologists' decision-making process.8 As a result, controversy exists regarding the benefits of screening patients for cognitive impairment in clinical practice. Therefore, the National Cancer Institute (NCI) has designated chemotherapy-related cognitive impairment (CRCI), often called “chemo brain” or “chemo fog,” as a high-priority area of research. In this manuscript, we discuss current gaps in research and highlight research priorities for the study of CRCI in older patients with cancer, with some mention of cognitive impairment related to cancer itself and also to other treatment modalities given that they frequently overlap.
The research gaps and priorities were initially discussed during a National Institute on Aging (NIA)/NCI sponsored U13 conference. The U13 conference provides a forum for a multidisciplinary team of investigators in geriatrics and oncology to review the present level of evidence in geriatric oncology, identify areas of highest research priority, and develop research approaches to improve clinical care for older adults with cancer. The research gaps were then further refined during monthly calls with the expert group over a 4-month period. The expert group is composed of 6 geriatric oncologists and 2 geriatricians who have expertise in the care of older adults with cognitive impairment and/or have conducted research in the area, 2 researchers with expertise in cognitive effects of cancer treatments and 1 patient advocate. We performed a literature search on PubMed using the keywords “cognition”, “cancer”, “prevalence” and “chemotherapy”. For research gap 1, relevant studies which described prevalence of CRCI and included older adults with cancer were selected from the search results. For research gap 4, studies evaluating treatment and preventive strategies for CRCI were selected if they were randomized trials or pre- and post-intervention in design (keywords “cognition”, “cancer”, “chemotherapy” and “treatment or prevention”). The studies were presented to the group of experts in cognition who selected the ones to be included in the manuscript. The studies were chosen with the purpose of illustrating research gaps rather than providing a comprehensive review of the literature.
2. Research Gaps
2.1. Gap 1: Very Few Studies Focus Exclusively on the Prevalence of CRCI in Older Adults With Cancer
CRCI has been reported in up to 12–75% of patients with cancer and is associated with cancer type, treatment, duration of follow-up, type of study design and definition of cognitive impairment.1,2 Most of these published studies assessed prevalence of CRCI in a heterogeneous group of patients, including both young and old patients, illustrated by first three studies in Table 1.9,10,11 However, since the impact of chemotherapy on cognition in older adults with cancer may be more significant given the higher prevalence of pre-existing cognitive impairment in this age group, studies that include older patients exclusively would provide more relevant information. Little is known about how chemotherapy influences the prevalence of cognitive impairment in older patients with cancer.
Table 1.
Reference | Population | Treatment | Age (years) | Time during which testing was performed |
Tests use/ measures |
Cognitive domains tested | Results |
---|---|---|---|---|---|---|---|
Cruzado et al., 201410 | 81 colon cancer patients |
Chemotherapy | Mean 67.0 (range 38–85) |
Pre-chemotherapy. T2: post-chemotherapy. T3: 6 months from last chemotherapy |
Neuropsychological testsa |
Attention and visual-motor ability, executive function, verbal memory and verbal learning |
Cognitive dysfunction in 37% at baseline, 37% at T2 and 39% at T3 |
Wefel et al., 201011 | 42 T1–3, N0–1, M0 breast cancer patients |
Chemotherapy | Mean 48.8 (range 33–65) |
At baseline: before chemotherapy. T2: during and shortly after chemotherapy [2.9 (SD, 0.59) or 7 (SD, 1.4) months after baseline). T3: 7.7 months (SD, 3.1) after completion of chemotherapy |
Neuropsychological testsa |
Attention, processing speed, learning and memory, executive function |
|
Kohli et al., 20079 | 595 newly diagnosed solid tumor patients |
Chemotherapy or radiation therapy or both |
Mean 56.3 (range 29–92) |
At baseline: within 2 weeks before initiation of chemotherapy. T2: within 2 weeks of completing treatment. T3: 6 months after completing treatment |
Symptom Inventory |
Concentration, memory |
|
Hurria et al., 200659 | 45 stage I–III breast cancer patients; 84 |
Chemotherapy | Mean 70 (range 65–84) |
Before chemotherapy. T2: 6 months after chemotherapy |
Squire Memory Self-rating Questionnaire |
Self-reported learning, working memory and remove learning |
51% perceived decline in memory from baseline to T2. |
Hurria et al., 20063 | 31 stage I–III breast cancer patients |
Chemotherapy | Mean 71 (range 65–84, SD 5) |
Before chemotherapy. T2: 6 months after chemotherapy |
Neuropsychological testsb |
Attention, verbal memory, visual memory; and verbal, spatial, psychomotor and executive functions |
Cognitive dysfunction in 11% at baseline, 29% at T2. |
Impairment defined as:
Scoring 2.0 SDs below published norms on one test or 1.5 SDs below published norms on two tests;
Scoring ≥ 2.0 SDs below published norms on two or more tests.
To date, there are only a few studies that focused on prevalence of CRCI in older patients with cancer (29–51% in Table 1). The limited number of studies may be because researchers are reluctant to study CRCI in older adults due to challenges in study accrual and high dropout rates compared to their younger counterparts.12,13 Older adults are also more reluctant to participate in clinical trials due to comorbidities, economic constraints, communication issues such as impaired hearing and eyesight, cultural divisions, language barriers, physical immobility with constraints in transportation and lack of social support.14
To overcome the barriers for researchers and patients, a number of strategies can be explored. McMurdo et al. recommended improving recruitment of older adults to research through good practice, including proper planning and engaging older adults in the importance of research.12 Proper planning may include simple, clear and legible reading materials that are appropriate for those with visual, hearing and cognitive impairment; adequate time allocation for appointments and breaks; and optimization of physical environment such as mobility and transportation assistance. To increase engagement of older adults in research, it may be meaningful to include the participants and their carers in the planning stages. McHenry et al. also presented three major themes in their recruitment strategies in older adults: communication and trust-building, providing comfort and security, and expressing gratitude.15
Older patients frequently have multiple barriers to limit their participation in research, and a team of members with different expertise within a multidisciplinary team (MDT) may help address barriers that interfere with the representation of older adults in research studies. The MDT approach is used in the care of elderly patients clinically and it has been shown to improve functional outcomes.16 In the research setting, the MDT team can include the physicians who introduce the studies, researchers and research assistants who provide information on the studies with proper planning, social workers who provide assistance with transportation and financial difficulties and caregivers who provide support in navigating the complex research process. However this approach can be resource and time-consuming, has not been studied in the recruitment of older adults in research, and needs to be investigated further.
2.2. Gap 2: Biologic Drivers of CRCI in Older Patients With Cancer are Unknown
Evidence from clinical and pre-clinical research suggests that many mechanisms play a role in the development of CRCI, including inflammation, hormonal changes, DNA damage, oxidative stress, reduced synaptic plasticity, altered growth factor levels, and impaired hippocampal neurogenesis.17–37 Several neuroimaging studies in patients with cancer show the impact of cancer and chemotherapy on brain structure and function, revealing white and gray matter loss, altered resting state metabolism changes and altered white matter integrity, and brain activation upon cognitive task challenge.38–55 Additionally, alleles in genes APOE and COMT have been linked to CRCI, suggesting that lipid metabolism, neural repair and neurotransmitter signaling also play a role in CRCI.56,57 Other mechanisms that have been proposed including epigenetic effects, and genes involved in longevity and aging.
Due to multi-morbidity and polypharmacy, the investigations of the biological mechanisms of CRCI in older patients are more complex than in younger patients for several reasons: 1) many mechanisms that are involved in CRCI are also involved in comorbid conditions, 2) comorbid conditions can increase vulnerability to CRCI by biological mechanisms similar or dissimilar from the mechanisms causing the condition itself, 3) the use of multiple medications can impact the measurement of many biological factors that may be related to CRCI, and 4) older patients are more likely to need dose- and drug-related changes to treatments which could differentially impact cognition.
In addition to cognitive reserve, education, gender, and anxiety and depression that are often assessed for their contribution to cognitive change in CRCI, careful measurement and assessment of comorbid conditions and medications in older patients are particularly important to consider in biomarker studies due to their increased frequency compared to younger patients. For example, hypertension, hyperlipidemia, diabetes, arthritis, osteoporosis, respiratory conditions, and neurodegenerative disorders are all impacted by inflammatory processes—the same cytokines, chemokines, and cognate receptors implicated in these diseases are also implicated in CRCI.19,20 It is not well-understood how chronic inflammation and resultant oxidative stress across the lifespan impact a new diagnosis of cancer in the older patient. Teasing apart these interactions is challenging and emphasizes the need for control groups of similar age and comorbidity level so that we can better understand the specific impact of cancer and chemotherapy treatments on cognition. It is important to appreciate that some biological mechanisms may already be at play impacting cognition as part of the normal aging process in this population. For example, studies investigating cognition in patients with cancer prior to surgery and chemotherapy suggest that inflammation is associated with cognitive decline.58
While not yet studied, it is likely that other biological, genetic, and epigenetic pathways also play a role in cognitive changes in older patients with cancer. Having a grasp on the biological and cognitive status of the older patient prior to cancer treatment is essential, as is understanding how these change over time in the context of their treatment, disease status, and comorbid conditions. Although not limited to older patients, there are two other ongoing trials that will be of interest: (1) studying changes in size, shape and activity in some brain areas that can occur in women receiving different types of breast cancer therapy (NCT01949376) and (2) studying the pathological changes of the brain using PET in patients with prostate cancer after ADT (NCT00006349).
By understanding the risk factors for cognitive dysfunction and the interaction between cancer treatment and the aging process, we can develop tools to risk-stratify patients for likelihood of cognitive impairment. These tools could help oncologists weigh the benefits relative to the risks of cancer therapy and introduce interventions to reduce the risks of new or worsening cognitive impairment.
2.3. Gap 3: Impact of Treatment on Cognition is not Routinely Measured in Trials Involving Older Adults With Cancer
Traditionally, clinical trials in oncology have focused on cancer-specific clinical endpoints such as overall survival, progression-free survival and response rates. Cognitive function is often not included as an endpoint in intervention trials assessing chemotherapy in most cancer subtypes, despite the fact that patients value cognition as an important outcome.8 Additionally, patients and caregivers want information on how cancer treatment affects cognition.59,60 In a qualitative study of the cancer treatment decision-making process in older patients, most patients accepted treatments only if they felt they were physically and cognitively able to tolerate them.60 Older patients with cancer also based their treatment decisions on anticipated adverse cognitive outcomes, independent from the burden of the treatment such as the length of the hospital stay, extent of testing, and invasiveness of interventions.8 Fried et al. studied 226 adults age ≥ 60 years with a limited life expectancy due to cancer, congestive heart failure, or chronic obstructive pulmonary disease and found that if the treatment burden was low but resulted in survival with severe cognitive impairment, 89% would decline treatment.8 These data highlight the importance of incorporating cognitive function as an endpoint in clinical trials involving older patients with cancer, especially if the interventions are known to have a risk of cognitive impairment.
Understanding how to best measure the impact of cancer and treatment on cognitive function in older patients is a research priority. Cognitive dysfunction and its effects in published studies are generally assessed via patient-reported outcomes through semi-structured interviews or through validated questionnaires or batteries of standardized neuropsychological tools. Examples of some validated questionnaires and neuropsychological tests are listed in Tables 2 and 3. The International Cognition and Cancer Task Force (ICCTF) recommended The Hopkins Verbal Learning Test-Revised (HVLT-R), Trail Making Test (TMT), and the Controlled Oral Word Association (COWA) of the Multilingual Aphasia Examination as cognitive assessments to be used in patients with cancer.61 However, the ICCTF did not make specific recommendations for the optimal assessments to use with older patients with cancer. Prior to the ICCTF recommendations, the Radiation Therapy Oncology Group (RTOG) has adapted a similar cognitive assessment battery in clinical trial of patients with cancer.62,63 However, the administration of these standardized tests by neuropsychologists, trained investigators or research assistants required substantial training. Moreover, administration of these tests is time consuming and fatiguing and therefore can be challenging in older patients due to impaired hearing or vision or low education at baseline.3,64
Table 2.
Cognitive domain | Neuropsychological test |
---|---|
Verbal/visual learning and memory |
Rey Auditory Verbal Learning Test |
Hopkins Verbal Learning Test | |
Wechsler Memory Scale | |
Verbal Memory Subtest of the Barcelona Test | |
Brief Visuospatial Memory Test | |
Luria MemoryWords Test | |
Spanish Adaptation Barcelona Test | |
Executive function | Controlled Oral Word Association Test |
Timed Instrumental Activities of Daily | |
Living Test | |
Stroop Test | |
Interference Score of the Stroop Color and | |
Word Test | |
Trail Making Test B | |
Intelligence | National Adult Reading Test |
Wechsler Adult Intelligence Scale | |
Language Concentration/ Attention |
Boston Naming Test |
Digit Span of the Wechsler Adult | |
Intelligence Scale | |
Trail Making Test | |
D2 Test of Attention | |
Color Trails | |
Fluency | Word Fluency Subtest from the Dutch |
Aphasia Society Test | |
Motor speed | Fepsy Finger Tapping Task |
Table 3.
Subjective assessment of cognition | Description | Scores |
---|---|---|
Functional Assessment of Cancer Therapy- Cognitive Function (FACT-Cog)87 |
37-Item questionnaire is divided into six cognitive domains: memory, concentration, mental acuity, verbal fluency, functional interference, and multitasking ability. |
Range from 0 to 148, with higher scores indicating better cognitive functioning |
European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC-QLQ-C30)77,88 |
30-Itemquestionnaire, two items [cognitive functioning scale (EORTC-CF)] that assess the cognitive domains of concentration and memory |
Range from 0 to 100, with higher scores indicating better perceived cognitive function |
Cognitive Failures Questionnaire89 | 25-item, 4 point Likert scale, self-report measure of everyday cognitive lapses such as forgetting appointments or where one has left things (e.g. keys, wallet), lapses in concentration or attention, or word-finding difficulty |
Range from 0 to 100, higher scores on the CFQ are indicative of greater number or severity of cognitive complaints. |
Multiple Abilities Questionnaire (MAQ)90 | Self-report measure of cognitive problems encountered in daily life. Each of the 48 items is rated on a 5-point scale for frequency of cognitive lapses or successes yielding a total score, as well as scores for 6 domains (attention, language, remote memory, verbal memory, visual–spatial memory, and visual–spatial perception). |
Range from 0 to 240, higher scores indicating worst perceived cognitive function |
Self-perceived deficits in attention (FEDA) and for subjectively experienced everyday memory performance (FEAG)73,74 |
27 items in FEDA and 29 items in FEAG, each item is rated on a 5-point scale. FEDA assesses self-rated distractibility and retardation of mental processes, fatigue and retardation in activities of daily living and decrease in motivation. FEAG assesses forgetfulness. |
Range from 27 to 135 in FEDA, 29–145 for FEAG, lower scores indicating worst perceived cognitive function |
Mini-mental State Examination (MMSE)68 | Brief screening test for cognitive deficits, covers a number of domains in orientation, registration, attention and calculation, recall, language, and copying. |
Range from 0 to 30, higher scores indicating better cognitive function |
Montreal Cognitive Assessment (MOCA)91 | Screening test for cognitive deficits, covers a number of domains in short-term memory, visuospatial abilities, executive functions, attention, concentration, and working memory |
Range from 0 to 30, higher scores indicating better cognitive function |
As studies have shown that certain domains are more commonly affected in patients with cancer (such as memory, motor function, attention, processing speed, concentration and executive functioning), one possible solution is to utilize screening tools focused on these domains [e.g. mini–mental state examination (MMSE), Montreal Cognitive Assessment (MOCA) or Blessed Orientation-Memory-Concentration (BOMC)], and if the tests are positive, comprehensive neuropsychological testing can follow.65–67 However, the Mini-mental State Examination (MMSE) has only rarely been used in studies of cognitive function screening in patients with cancer, and now requires a per use fee.68 The MOCA test may be a superior screening test for multi-domains including executive functioning. Nevertheless, these tests have variable sensitivity and specificity in non-cancer populations,69–71 therefore there is a definite need to validate the sensitivity and specificity of these screening tests in patients with cancer prior to wider usage.
Another challenge in studying cognitive impairment among patients with cancer is determining what magnitude of cognitive function change should be considered clinically relevant. The ICCTF recommends defining clinically significant cognitive impairment if the result is ≥2 SDs below the mean for one test, or ≥1.5 SDs below the mean for more than one test to facilitate between study-comparison.61 The ICCTF also encourages investigators to report the frequency of impairment for each test, the number of patients showing impairment on one, two or three tests and so forth, and the most common patterns of impairment. However the task force only defines cognitive impairment using neuropsychological testing, not other clinical measures such decline in activities of daily living (ADL) or instrumental activities of daily living (IADLs). In other studies, impairment was defined as scoring below the lowest quartile on at least four tests, scoring below the tenth percentile on two cognitive domains and scoring below the fifth percentile on at least four tests.72–74 The clinical relevance of these changes is unclear, as a systematic review by Hutchinson et al. found that only 8 of the 24 studies had a significant relationship between objective cognitive impairment measured by neuropsychological testing and patient-reported cognitive impairment.75 A European task force recommends the use of changes in ADL as a co-primary outcome for Alzheimer's trials, but this recommendation has not been widely adopted in cancer research.76 One possible explanation may be that a significant decline in cognitive function is required for a change in ADLs.77 More research is needed to associate changes in measurements of cognitive function with outcomes meaningful to older patients with cancer.
2.4. Gap 4: Few Randomized Clinical Trials Study Treatment and Preventive Strategies for CRCI in Older Patients With Cancer
Clinical trials of CRCI interventions have focused mostly on cancer survivors with cognitive impairment after cancer treatment, and few studies have evaluated how to prevent or decrease cognitive impairment during treatment for cancer.44,78–80 Additionally, most intervention studies have included only a small number of patients. These interventions can be grouped into pharmacological and non-pharmacological categories. Examples of pharmacological agents investigated included donepezil, methylphenidate, modafinil and erythropoietin.78,79,81,82 Non-pharmacological interventions include specific exercise programs and cognitive training.80,83,84 Selected studies are shown in Table 4.
Table 4.
Study | Design | N | Population | Intervention group | Comparison group | Outcome |
---|---|---|---|---|---|---|
Lawrence et al., 201578 | Randomized, double- blind, placebo controlled |
62 | Patients with invasive breast cancer who completed adjuvant chemotherapy 1 to 5 years previously and who reported significant cognitive symptoms |
Median age 55.8 (range 41–78, 35% ≥ 60) Donepazil 5 mg/day vs. placebo for 6 weeks, if tolerated 10 mg/day for 18 weeks |
Median age 55.8 (range 39.79, 43% ≥ 60) Placebo |
Improvement in recall and discrimination |
O′Shaughnessy et al., 200592 | Randomized, double- blind, placebo controlled |
100 | Women ≥18 years of age diagnosed with stage I, II, or III breast cancer scheduled to receive 4 cycles of anthracycline-based adjuvant or neoadjuvant chemotherapy |
Mean age 53.3 (SD 9.7) 40,000 units of EPO weekly throughout chemotherapy |
Mean age 54.3 (SD 12) Placebo |
Prior to cycle 4 of chemotherapy, executive functioning was improved in EPO-treated group compared to placebo but no difference was noted at 6 months |
Fan HG et al., 200893 | Randomized, double- blind, placebo controlled |
57 | Patients with fully resected early breast cancer undergoing adjuvant chemotherapy |
Median age 50 (range 36–73) d-Methylphenidate 5–10 mg until the end of chemotherapy |
Median age 51 (range 37–74) Placebo |
No change in cognitive function |
Kohli et al., 200979 | Randomized, double- blind, placebo controlled |
82 | Breast cancer patients who completed chemotherapy for a month |
Mean age 22.1 (SD 22.7) Modafinil 200 mg/day one-month after chemotherapy and/or radiation treatment |
Mean age 22.1 (SD 22.7) Placebo |
Improvement in memory and attention skills |
Lundorff et al., 200985 | Randomized, double- blind, cross-over |
28 | Advanced cancer patients treated in palliative care settings |
Median age 62 (range 40–79) Modafinil day 1 then cross-cover to placebo on day 8, placebo day 1 then cross-cover to modafinil on day 8 |
NA | Improvement in executive function and motor speed |
Oh B et al., 201284 | Randomized trial | 81 | Cancer patients who had received or undergoing chemotherapy |
Mean age 64.6 (SD 12.3) Qigong (combination of gentle exercise and meditation) for 10 weeks |
Mean age 61.1 (SD 11.0) Usual care |
Improved cognitive function |
Reid-Arndt et al., 201283 | Before and after intervention |
23 | Cancer patients who had completed chemotherapy at least 12 months |
Mean age 62.3 (SD 10.8) Tai-chi 60-min class twice/week for 10 weeks |
NA | Improvement in memory, verbal fluency and attention |
Kesler et al., 201380 | Randomized, placebo trial |
41 | Breast cancer survivors (stage I–IIIA) who on average completed chemotherapy for 6 years |
Mean age 55 (SD 7) Online executive training program for 12 weeks |
Mean age 56 (SD 6) Waitlist |
Improvement in cognitive flexibility, verbal fluency and processing speed |
Ferguson et al., 201294 | Randomized, double- blind, placebo controlled |
40 | Stage I and II breast cancer who had received adjuvant chemotherapy |
Mean age 51.2 (SD 7.3) Cognitive behavioral therapy (Memory and Attention Adaptation Training) |
Mean age 49.4 (SD 5.1) Waitlist |
Improvement in verbal memory |
Janelsins et al., 201595 | Randomized trial | 328 | Cancer survivors after adjuvant treatment |
Mean age 55.2 (SD 11.0) Standard care and YOCAS©® yoga—a program of breathing, gentle Hatha and restorative postures, and meditation |
Mean age 54.0 (SD 8.7) Standard care |
Reduced patient-reported memory difficulty |
Only three intervention studies included patients with a mean age above 6069,70,72; two of these studies evaluated non-pharmacological interventions. Trials evaluating the safety and efficacy of pharmacological agents for cognitive impairment typically do not include many older patients with cancer. Modafinil is a wake-promoting agent that is effective in the treatment of excessive sleepiness associated with narcolepsy and in persons with shift-work sleep disorder. Lundorff et al. randomized 28 patients with advanced cancer (not specific to CRCI; mean age 62, range 40–79) treated in palliative care settings to modafinil or placebo.85 Compared to placebo, the modafinil group demonstrated improvement in executive function and motor speed.
More data support non-pharmacological interventions such as exercise and cognitive behavioral training in older patients. A randomized study by Oh et al. evaluated the effects of medical Qigong (combination of gentle exercise and meditation) in 81 patients with cancer (mean age 64.6, SD 12.3) who either had received chemotherapy (66%) or were undergoing chemotherapy (34%). The intervention group participated in a 10-week Qigong program.84 Compared to placebo, the intervention group reported significantly better cognitive function. Another study by Reid-Arndt evaluated the use of Tai-chi in 23 women (mean age 62.3, SD 10.8) who had been diagnosed with any cancer and had undergone chemotherapy, with treatment completed at least 12 months prior to the start of the study. Study participants received 60 minute Tai-Chi classes two times/week for 10 weeks.83 At the end of the study, improvements were noted in memory, verbal fluency and attention. A clinical trial is currently ongoing to evaluate the use of acupuncture to prevent “chemo brain” in patients with breast cancer (NCT02457039) but it is not limited to older patients.
None of the aforementioned studies were exclusive to older patients with cancer. Additionally, most of the studies had small sample size, and only included a small number of older adults. Sprod at al. conducted a secondary analysis of a longitudinal study of 408 newly diagnosed older patients with cancer (mean age 73, range 65–92) who underwent chemotherapy and/or any radiation therapy.86 The group found that the oldest patients (≥80 years) who exercised during treatment self-reported less memory loss during treatment.
To date, no studies have looked at strategies to prevent CRCI. Therefore, we need randomized trials to evaluate interventions for prevention of cognitive impairment in patients with cancer undergoing chemotherapy, especially among older patients who have a high risk for cognitive decline with cancer treatments.
3. Conclusion and Research Priorities
CRCI is increasingly recognized but research focused on older patients with cancer is limited, which may be due to the difficulty of studying this group due to underlying medical complexities and the effect of aging as a potential confounder. The development of cognitive impairment is likely multifactorial, with contributing factors including, but not limited to, aging, comorbidities, underlying cancer, cancer treatment, psychoactive medications, and psychosocial, environmental and genetic risk factors. Yet this is an essential area of research given its clinical importance. Effective interventions for patients with cancer with CRCI are also lacking, as are guidelines on how to care for older patients with cancer and CRCI.
Addressing the following research priorities will help close gaps in knowledge by illuminating how to best prevent or improve cognitive outcomes in older patients with cancer. These priorities are: 1) design a longitudinal study to evaluate the prevalence of cognitive impairment exclusively in older patients with cancer; 2) investigate the role of the MDT approach to increase the participation of older adults with cancer in clinical studies; 3) delineation of the mechanisms of injury in older patients with cancer and examine the complex interactions of cognition with cancer, cancer treatments and psychosocial, lifestyle and genetic risk factors in parallel to aging; 4) develop tools to risk-stratify the likelihood of developing cognitive impairment; 5) develop validated cognitive screening and neuropsychological tests as well as patient-reported outcomes that are feasible and meaningful for older patients with cancer; 6) implement randomized controlled trials on the prevention and treatment of cognitive impairment in older patients with cancer. It is crucial that we design studies exclusively for older adults with cancer to fill the gaps illustrated above and to better understand the interplay between CRCI and aging.
Acknowledgments
This work was funded by a U13 AG038151 from the National Institute on Aging. The work was also funded by the American Cancer Society and a Patient-centered Outcomes Research Institute (PCORI) Program contract (4634). The work received support from the James Wilmot Cancer Institute (WCI), the Alliance for Clinical Trials in Oncology (National Cancer Institute of the National Institutes of Health under Award Numbers U10CA18082 and 1UG1CA189823), and UG1 CA189961 from the National Cancer Institute. This work was made possible by the generous donors to the WCI geriatric oncology philanthropy fund. All statements in this report, including its findings and conclusions, are solely those of the authors, do not necessarily represent the official views of the funding agencies, and do not necessarily represent the views of the Patient-centered Outcomes Research Institute (PCORI), its Board of Governors or Methodology Committee.
We wish to acknowledge Dr. Susan Rosenthal for helping us edit this manuscript in order to improve the language and flow.
Footnotes
Disclosures and Conflict of Interest Statements
The authors have no conflict of interest to report.
Author Contributions
Study concept: KP Loh, M Janelsins, S Mohile, H Holmes, T Hsu, S Inouye, M Karuturi, G Kimmick, S Lichtman, A Magnuson, M Whitehead, M Wong, and T Ahles.
Manuscript preparation: KP Loh and M Janelsins.
Manuscript editing: KP Loh, M Janelsins, S Mohile, H Holmes, T Hsu, S Inouye, M Karuturi, G Kimmick, S Lichtman, A Magnuson, M Whitehead, M Wong, and T Ahles.
Manuscript review: KP Loh, M Janelsins, S Mohile, H Holmes, T Hsu, S Inouye, M Karuturi, G Kimmick, S Lichtman, A Magnuson, M Whitehead, M Wong, and T Ahles.
REFERENCES
- 1.Koppelmans V, Breteler MMB, 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 Off J Am Soc Clin Oncol. 2012 Apr 1;30(10):1080–1086. doi: 10.1200/JCO.2011.37.0189. [DOI] [PubMed] [Google Scholar]
- 2.Janelsins MC, Kesler SR, Ahles TA, Morrow GR. Prevalence, mechanisms, and management of cancer-related cognitive impairment. Int Rev Psychiatry Abingdon Engl. 2014 Feb;26(1):102–113. doi: 10.3109/09540261.2013.864260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Hurria A, Rosen C, Hudis C, Zuckerman E, Panageas KS, Lachs MS, et al. Cognitive function of older patients receiving adjuvant chemotherapy for breast cancer: a pilot prospective longitudinal study. J Am Geriatr Soc. 2006 Jun;54(6):925–931. doi: 10.1111/j.1532-5415.2006.00732.x. [DOI] [PubMed] [Google Scholar]
- 4.Ahles TA, Saykin AJ, McDonald BC, Li Y, Furstenberg CT, Hanscom BS, et al. Longitudinal assessment of cognitive changes associated with adjuvant treatment for breast cancer: impact of age and cognitive reserve. J Clin Oncol Off J Am Soc Clin Oncol. 2010 Oct 10;28(29):4434–4440. doi: 10.1200/JCO.2009.27.0827. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Stilley CS, Bender CM, Dunbar-Jacob J, Sereika S, Ryan CM. The impact of cognitive function on medication management: three studies. Health Psychol Off J Div Health Psychol Am Psychol Assoc. 2010 Jan;29(1):50–55. doi: 10.1037/a0016940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Bárrios H, Narciso S, Guerreiro M, Maroco J, Logsdon R, de Mendonça A. Quality of life in patients with mild cognitive impairment. Aging Ment Health. 2013;17(3):287–292. doi: 10.1080/13607863.2012.747083. [DOI] [PubMed] [Google Scholar]
- 7.Robb C, Boulware D, Overcash J, Extermann M. Patterns of care and survival in cancer patients with cognitive impairment. Crit Rev Oncol Hematol. 2010 Jun;74(3):218–224. doi: 10.1016/j.critrevonc.2009.07.002. [DOI] [PubMed] [Google Scholar]
- 8.Fried TR, Bradley EH, Towle VR, Allore H. Understanding the treatment preferences of seriously ill patients. N Engl J Med. 2002 Apr 4;346(14):1061–1066. doi: 10.1056/NEJMsa012528. [DOI] [PubMed] [Google Scholar]
- 9.Kohli S, Griggs JJ, Roscoe JA, Jean-Pierre P, Bole C, Mustian KM, et al. Self-reported cognitive impairment in patients with cancer. J Oncol Pract Am Soc Clin Oncol. 2007 Mar;3(2):54–59. doi: 10.1200/JOP.0722001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Cruzado JA, López-Santiago S, Martínez-Marín V, José-Moreno G, Custodio AB, Feliu J. Longitudinal study of cognitive dysfunctions induced by adjuvant chemotherapy in colon cancer patients. Support Care Cancer Off J Multinatl Assoc Support Care Cancer. 2014 Jul;22(7):1815–1823. doi: 10.1007/s00520-014-2147-x. [DOI] [PubMed] [Google Scholar]
- 11.Wefel JS, Saleeba AK, Buzdar AU, Meyers CA. Acute and late onset cognitive dysfunction associated with chemotherapy in women with breast cancer. Cancer. 2010 Jul 15;116(14):3348–3356. doi: 10.1002/cncr.25098. [DOI] [PubMed] [Google Scholar]
- 12.McMurdo MET, Roberts H, Parker S, Wyatt N, May H, Goodman C, et al. Improving recruitment of older people to research through good practice. Age Ageing. 2011 Nov 1;40(6):659–665. doi: 10.1093/ageing/afr115. [DOI] [PubMed] [Google Scholar]
- 13.Mody L, Miller DK, McGloin JM, Div M, Freeman M, Marcantonio ER, et al. Recruitment and retention of older adults in aging research. J Am Geriatr Soc. 2008 Dec;56(12):2340–2348. doi: 10.1111/j.1532-5415.2008.02015.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Herrera AP, Snipes SA, King DW, Torres-Vigil I, Goldberg DS, Weinberg AD. Disparate inclusion of older adults in clinical trials: priorities and opportunities for policy and practice change. Am J Public Health. 2010 Apr;100(Suppl 1):S105–S112. doi: 10.2105/AJPH.2009.162982. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.McHenry JC, Insel KC, Einstein GO, Vidrine AN, Koerner KM, Morrow DG. Recruitment of older adults: success may be in the details. Gerontologist. 2015 Oct;55(5):845–853. doi: 10.1093/geront/gns079. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Tanaka M. Multidisciplinary team approach for elderly patients. Geriatr Gerontol Int. 2003 Jun 1;3(2):69–72. [Google Scholar]
- 17.Rego SL, Helms RS, Dréau D. Tumor necrosis factor-alpha-converting enzyme activities and tumor-associated macrophages in breast cancer. Immunol Res. 2014 Jan;58(1):87–100. doi: 10.1007/s12026-013-8434-7. [DOI] [PubMed] [Google Scholar]
- 18.Korkaya H, Liu S, Wicha MS. Breast cancer stem cells, cytokine networks, and the tumor microenvironment. J Clin Invest. 2011 Oct;121(10):3804–3809. doi: 10.1172/JCI57099. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Janelsins MC, Mustian KM, Palesh OG, Mohile SG, Peppone LJ, Sprod LK, et al. Differential expression of cytokines in breast cancer patients receiving different chemotherapies: implications for cognitive impairment research. Support Care Cancer Off J Multinatl Assoc Support Care Cancer. 2012 Apr;20(4):831–839. doi: 10.1007/s00520-011-1158-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Ahles TA, Saykin AJ. Candidate mechanisms for chemotherapy-induced cognitive changes. Nat Rev Cancer. 2007 Mar;7(3):192–201. doi: 10.1038/nrc2073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Villani F, Busia A, Villani M, Vismara C, Viviani S, Bonfante V. Serum cytokine in response to chemo-radiotherapy for Hodgkin’s disease. Tumori. 2008 Dec;94(6):803–808. doi: 10.1177/030089160809400605. [DOI] [PubMed] [Google Scholar]
- 22.Meyers CA, Albitar M, Estey E. Cognitive impairment, fatigue, and cytokine levels in patients with acute myelogenous leukemia or myelodysplastic syndrome. Cancer. 2005 Aug 15;104(4):788–793. doi: 10.1002/cncr.21234. [DOI] [PubMed] [Google Scholar]
- 23.Cleeland CS, Bennett GJ, Dantzer R, Dougherty PM, Dunn AJ, Meyers CA, et al. Are the symptoms of cancer and cancer treatment due to a shared biologic mechanism? A cytokine-immunologic model of cancer symptoms. Cancer. 2003 Jun 1;97(11):2919–2925. doi: 10.1002/cncr.11382. [DOI] [PubMed] [Google Scholar]
- 24.Raffa R. A proposed mechanism for chemotherapy-related cognitive impairment (“chemo-fog”) J Clin Pharm Ther. 2011 Jun;36(3):257–259. doi: 10.1111/j.1365-2710.2010.01188.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kesler S, Janelsins M, Koovakkattu D, Palesh O, Mustian K, Morrow G, 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 Mar;30(Suppl):S109–S116. doi: 10.1016/j.bbi.2012.05.017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Pusztai L, Mendoza TR, Reuben JM, Martinez MM, Willey JS, Lara J, et al. Changes in plasma levels of inflammatory cytokines in response to paclitaxel chemotherapy. Cytokine. 2004 Feb 7;25(3):94–102. doi: 10.1016/j.cyto.2003.10.004. [DOI] [PubMed] [Google Scholar]
- 27.Mills PJ, Ancoli-Israel S, Parker B, Natarajan L, Hong S, Jain S, et al. Predictors of inflammation in response to anthracycline-based chemotherapy for breast cancer. Brain Behav Immun. 2008 Jan;22(1):98–104. doi: 10.1016/j.bbi.2007.07.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Vardy J, Rourke S, Galica J, Pond GR, Park A, Zhang H, et al. Cytokine levels in patients (pts) with localized colorectal cancer (CRC) after surgery and their relationship to fatigue and cognitive function. ASCO Meet Abstr. 2006 Jun 20;24(18_suppl):3623. [Google Scholar]
- 29.Vardy J, Dhillon H, Pond GR, et al. Cognitive function in colorectal cancer patients: interim analysis of a longitudinal prospective study. J Clin Oncol Suppl Abstr. 2012;9021 [Abstract] [Google Scholar]
- 30.Ganz PA, Bower JE, Kwan L, Castellon SA, Silverman DHS, Geist C, et al. Does tumor necrosis factor-alpha (TNF-α) play a role in post-chemotherapy cerebral dysfunction? Brain Behav Immun. 2013 Mar;30(Suppl):S99–S108. doi: 10.1016/j.bbi.2012.07.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Pomykala KL, Ganz PA, Bower JE, Kwan L, Castellon SA, Mallam S, et al. The association between pro-inflammatory cytokines, regional cerebral metabolism, and cognitive complaints following adjuvant chemotherapy for breast cancer. Brain Imaging Behav. 2013 Dec;7(4):511–523. doi: 10.1007/s11682-013-9243-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Dietrich J, Han R, Yang Y, Mayer-Pröschel M, Noble M. CNS progenitor cells and oligodendrocytes are targets of chemotherapeutic agents in vitro and in vivo. J Biol. 2006;5(7):22. doi: 10.1186/jbiol50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Systemic 5-fluorouracil treatment causes a syndrome of delayed myelin destruction in the central nervous system. — PubMed — NCBI [internet] [cited 2015 Oct 27]; doi: 10.1186/jbiol69. Available from, http://www.ncbi.nlm.nih.gov/pubmed/18430259. [DOI] [PMC free article] [PubMed]
- 34.Aluise CD, St Clair D, Vore M, Butterfield DA. In vivo amelioration of adriamycin induced oxidative stress in plasma by gamma-glutamylcysteine ethyl ester (GCEE) Cancer Lett. 2009 Sep 8;282(1):25–29. doi: 10.1016/j.canlet.2009.02.047. [DOI] [PubMed] [Google Scholar]
- 35.Redox proteomic identification of oxidized cardiac proteins in adriamycin-treated mice [internet] [cited 2015 Oct 27]; doi: 10.1016/j.freeradbiomed.2006.08.006. Available from:, http://www.sciencedirect.com/science/article/pii/S0891584906005119. [DOI] [PubMed] [Google Scholar]
- 36.Tangpong J, Cole MP, Sultana R, Joshi G, Estus S, Vore M, et al. Adriamycin-induced, TNF-alpha-mediated central nervous system toxicity. Neurobiol Dis. 2006 Jul;23(1):127–139. doi: 10.1016/j.nbd.2006.02.013. [DOI] [PubMed] [Google Scholar]
- 37.Janelsins MC, Roscoe JA, Berg MJ, Thompson BD, Gallagher MJ, Morrow GR, et al. IGF-1 partially restores chemotherapy-induced reductions in neural cell proliferation in adult C57BL/6 mice. Cancer Invest. 2010 Jun;28(5):544–553. doi: 10.3109/07357900903405942. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Inagaki M, Yoshikawa E, Matsuoka Y, Sugawara Y, Nakano T, Akechi T, et al. Smaller regional volumes of brain gray and white matter demonstrated in breast cancer survivors exposed to adjuvant chemotherapy. Cancer. 2007 Jan 1;109(1):146–156. doi: 10.1002/cncr.22368. [DOI] [PubMed] [Google Scholar]
- 39.Abraham J, Haut MW, Moran MT, 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 Feb;8(1):88–91. doi: 10.3816/CBC.2008.n.007. [DOI] [PubMed] [Google Scholar]
- 40.Swayampakula AK, Alkhouri N, Haut MW, Abraham J. Cognitive impairment with significant brain parenchymal volume loss following standard adjuvant chemotherapy in a patient with breast cancer. Clin Adv Hematol Oncol HO. 2007 Dec;5(12):985–987. discussion 987–8. [PubMed] [Google Scholar]
- 41.de Ruiter MB, Reneman L, Boogerd W, Veltman DJ, Caan M, Douaud G, 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 Dec;33(12):2971–2983. doi: 10.1002/hbm.21422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Deprez S, Amant F, Yigit R, Porke K, Verhoeven J, Van den Stock J, 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 Mar;32(3):480–493. doi: 10.1002/hbm.21033. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Hosseini SMH, Kesler SR. Multivariate pattern analysis of FMRI in breast cancer survivors and healthy women. J Int Neuropsychol Soc. 2014 Apr;20(4):391–401. doi: 10.1017/S1355617713001173. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Ferguson RJ, Ahles TA, Saykin AJ, McDonald BC, Furstenberg CT, Cole BF, et al. Cognitive–behavioral management of chemotherapy-related cognitive change. Psychooncology. 2007 Aug;16(8):772–777. doi: 10.1002/pon.1133. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.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 Off J Am Soc Clin Oncol. 2012 Jul 10;30(20):2500–2508. doi: 10.1200/JCO.2011.38.5674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.de Ruiter MB, Reneman L, Boogerd W, Veltman DJ, van Dam FSAM, Nederveen AJ, et al. Cerebral hyporesponsiveness and cognitive impairment 10 years after chemotherapy for breast cancer. Hum Brain Mapp. 2011 Aug;32(8):1206–1219. doi: 10.1002/hbm.21102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kesler SR, Kent JS, O’Hara R. Prefrontal cortex and executive function impairments in primary breast cancer. Arch Neurol. 2011 Nov;68(11):1447–1453. doi: 10.1001/archneurol.2011.245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Kesler SR, Bennett FC, Mahaffey ML, Spiegel D. Regional brain activation during verbal declarative memory in metastatic breast cancer. Clin Cancer Res Off J Am Assoc Cancer Res. 2009 Nov 1;15(21):6665–6673. doi: 10.1158/1078-0432.CCR-09-1227. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Kesler SR, Watson C, Koovakkattu D, Lee C, O’Hara R, Mahaffey ML, et al. Elevated prefrontal myo-inositol and choline following breast cancer chemotherapy. [cited 2015 Oct 9];Brain Imaging Behav [internet] 2013 Dec;7(4) doi: 10.1007/s11682-013-9228-1. Available from, http://www.ncbi.nlm.nih.gov/pmc/articles/PMC3731420/ [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.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 Off J Am Soc Clin Oncol. 2007 Sep 1;25(25):3866–3870. doi: 10.1200/JCO.2007.10.8639. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Bruno J, Hosseini SMH, Kesler S. Altered resting state functional brain network topology in chemotherapy-treated breast cancer survivors. Neurobiol Dis. 2012 Dec;48(3):329–338. doi: 10.1016/j.nbd.2012.07.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Kesler SR. Default mode network as a potential biomarker of chemotherapy-related brain injury. Neurobiol Aging. 2014 Sep;35(Supplement 2):S11–S19. doi: 10.1016/j.neurobiolaging.2014.03.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Dumas JA, Makarewicz J, Schaubhut GJ, Devins R, Albert K, Dittus K, et al. Chemotherapy altered brain functional connectivity in women with breast cancer: a pilot study. Brain Imaging Behav. 2013 Dec;7(4):524–532. doi: 10.1007/s11682-013-9244-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hosseini SMH, Koovakkattu D, Kesler SR. Altered small-world properties of gray matter networks in breast cancer. BMC Neurol. 2012;12:28. doi: 10.1186/1471-2377-12-28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Kesler SR, Wefel JS, Hosseini SMH, Cheung M, Watson CL, Hoeft F. Default mode network connectivity distinguishes chemotherapy-treated breast cancer survivors from controls. Proc Natl Acad Sci U S A. 2013 Jul 9;110(28):11600–11605. doi: 10.1073/pnas.1214551110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Ahles TA, Saykin AJ, Noll WW, Furstenberg CT, Guerin S, Cole B, et al. The relationship of APOE genotype to neuropsychological performance in long-term cancer survivors treated with standard dose chemotherapy. Psychooncology. 2003 Sep;12(6):612–619. doi: 10.1002/pon.742. [DOI] [PubMed] [Google Scholar]
- 57.Small BJ, Rawson KS, Walsh E, Jim HSL, Hughes TF, Iser L, et al. Catechol-O-methyltransferase genotype modulates cancer treatment-related cognitive deficits in breast cancer survivors. Cancer. 2011 Apr 1;117(7):1369–1376. doi: 10.1002/cncr.25685. [DOI] [PubMed] [Google Scholar]
- 58.Patel SK, Wong AL, Wong FL, Breen EC, Hurria A, Smith M, et al. Inflammatory biomarkers, comorbidity, and neurocognition in women with newly diagnosed breast cancer. J Natl Cancer Inst. 2015 Aug;107(8) doi: 10.1093/jnci/djv131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Hurria A, Goldfarb S, Rosen C, Holland J, Zuckerman E, Lachs MS, et al. Effect of adjuvant breast cancer chemotherapy on cognitive function from the older patient’s perspective. Breast Cancer Res Treat. 2006 Aug;98(3):343–348. doi: 10.1007/s10549-006-9171-6. [DOI] [PubMed] [Google Scholar]
- 60.Chouliara Z, Miller M, Stott D, Molassiotis A, Twelves C, Kearney N. Older people with cancer: perceptions and feelings about information, decision-making and treatment—a pilot study. Eur J Oncol Nurs. 2004 Sep 1;8(3):257–261. doi: 10.1016/j.ejon.2003.12.010. [DOI] [PubMed] [Google Scholar]
- 61.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 Jul;12(7):703–708. doi: 10.1016/S1470-2045(10)70294-1. [DOI] [PubMed] [Google Scholar]
- 62.Brown PD, Pugh S, Laack NN, Wefel JS, Khuntia D, Meyers C, et al. Memantine for the prevention of cognitive dysfunction in patients receiving whole-brain radiotherapy: a randomized, double-blind, placebo-controlled trial. Neuro Oncol. 2013 Oct;15(10):1429–1437. doi: 10.1093/neuonc/not114. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Meyers CA, Brown PD. Role and relevance of neurocognitive assessment in clinical trials of patients with CNS tumors. J Clin Oncol Off J Am Soc Clin Oncol. 2006 Mar 10;24(8):1305–1309. doi: 10.1200/JCO.2005.04.6086. [DOI] [PubMed] [Google Scholar]
- 64.Mandelblatt JS, Stern RA, Luta G, McGuckin M, Clapp JD, Hurria A, 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 Jun 20;32(18):1909–1918. doi: 10.1200/JCO.2013.54.2050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Anderson-Hanley C, Sherman ML, Riggs R, Agocha VB, Compas BE. Neuropsychological effects of treatments for adults with cancer: a meta-analysis and review of the literature. J Int Neuropsychol Soc. 2003 Nov;9(7):967–982. doi: 10.1017/S1355617703970019. [DOI] [PubMed] [Google Scholar]
- 66.Ono M, Ogilvie JM, Wilson JS, Green HJ, Chambers SK, Ownsworth T, et al. A meta-analysis of cognitive impairment and decline associated with adjuvant chemotherapy in women with breast cancer. Front Oncol. 2015;5:59. doi: 10.3389/fonc.2015.00059. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Davis J, Ahlberg FM, Berk M, Ashley DM, Khasraw M. Emerging pharmacotherapy for cancer patients with cognitive dysfunction. BMC Neurol. 2013 Oct 24;13(1):153. doi: 10.1186/1471-2377-13-153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Iconomou G, Mega V, Koutras A, Iconomou AV, Kalofonos HP. Prospective assessment of emotional distress, cognitive function, and quality of life in patients with cancer treated with chemotherapy. Cancer. 2004 Jul 15;101(2):404–411. doi: 10.1002/cncr.20385. [DOI] [PubMed] [Google Scholar]
- 69.Smith T, Gildeh N, Holmes C. The Montreal cognitive assessment: validity and utility in a memory clinic setting. Can J Psychiatr Rev Can Psychiatr. 2007 May;52(5):329–332. doi: 10.1177/070674370705200508. [DOI] [PubMed] [Google Scholar]
- 70.Godefroy O, Fickl A, Roussel M, Auribault C, Bugnicourt JM, Lamy C, et al. Is the Montreal cognitive assessment superior to the mini-mental state examination to detect poststroke cognitive impairment? A study with neuropsychological evaluation. Stroke J Cereb Circ. 2011 Jun;42(6):1712–1716. doi: 10.1161/STROKEAHA.110.606277. [DOI] [PubMed] [Google Scholar]
- 71.Pendlebury ST, Mariz J, Bull L, Mehta Z, Rothwell PM. MoCA, ACE-R, and MMSE versus the National Institute of Neurological Disorders and Stroke–Canadian Stroke Network Vascular Cognitive Impairment Harmonization Standards Neuropsychological Battery after TIA and stroke. Stroke J Cereb Circ. 2012 Feb;43(2):464–469. doi: 10.1161/STROKEAHA.111.633586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Ahles TA, Saykin AJ, Furstenberg CT, Cole B, Mott LA, Skalla K, et al. Neuropsychologic impact of standard-dose systemic chemotherapy in long-term survivors of breast cancer and lymphoma. J Clin Oncol Off J Am Soc Clin Oncol. 2002 Jan 15;20(2):485–493. doi: 10.1200/JCO.2002.20.2.485. [DOI] [PubMed] [Google Scholar]
- 73.Mehnert A, Scherwath A, Schirmer L, Schleimer B, Petersen C, Schulz-Kindermann F, et al. The association between neuropsychological impairment, self-perceived cognitive deficits, fatigue and health related quality of life in breast cancer survivors following standard adjuvant versus high-dose chemotherapy. Patient Educ Couns. 2007 Apr;66(1):108–118. doi: 10.1016/j.pec.2006.11.005. [DOI] [PubMed] [Google Scholar]
- 74.Poppelreuter M, Weis J, Külz AK, Tucha O, Lange KW, Bartsch HH. Cognitive dysfunction and subjective complaints of cancer patients. A cross-sectional study in a cancer rehabilitation centre. Eur J Cancer Oxf Engl 1990. 2004 Jan;40(1):43–49. doi: 10.1016/j.ejca.2003.08.001. [DOI] [PubMed] [Google Scholar]
- 75.Hutchinson AD, Hosking JR, Kichenadasse G, Mattiske JK, Wilson C. Objective and subjective cognitive impairment following chemotherapy for cancer: a systematic review. Cancer Treat Rev. 2012 Nov;38(7):926–934. doi: 10.1016/j.ctrv.2012.05.002. [DOI] [PubMed] [Google Scholar]
- 76.Vellas B, Andrieu S, Sampaio C, Coley N, Wilcock G European Task Force Group. Endpoints for trials in Alzheimer’s disease: a European task force consensus. Lancet Neurol. 2008 May;7(5):436–450. doi: 10.1016/S1474-4422(08)70087-5. [DOI] [PubMed] [Google Scholar]
- 77.Liu-Seifert H, Siemers E, Sundell K, Price K, Han B, Selzler K, et al. Cognitive and functional decline and their relationship in patients with mild Alzheimer’s dementia. J Alzheimers Dis. 2015;43(3):949–955. doi: 10.3233/JAD-140792. [DOI] [PubMed] [Google Scholar]
- 78.Lawrence JA, Griffin L, Balcueva EP, Groteluschen DL, Samuel TA, Lesser GJ, et al. A study of donepezil in female breast cancer survivors with self-reported cognitive dysfunction 1 to 5 years following adjuvant chemotherapy. J Cancer Surviv. 2015 Jul;1:1–9. doi: 10.1007/s11764-015-0463-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kohli S, Fisher SG, Tra Y, Adams MJ, Mapstone ME, Wesnes KA, et al. The effect of modafinil on cognitive function in breast cancer survivors. Cancer. 2009 Jun 15;115(12):2605–2616. doi: 10.1002/cncr.24287. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Kesler S, Hadi Hosseini SM, Heckler C, Janelsins M, Palesh O, Mustian K, et al. Cognitive training for improving executive function in chemotherapy-treated breast cancer survivors. Clin Breast Cancer. 2013 Aug;13(4):299–306. doi: 10.1016/j.clbc.2013.02.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Meyers CA, Weitzner MA, Valentine AD, Levin VA. Methylphenidate therapy improves cognition, mood, and function of brain tumor patients. J Clin Oncol Off J Am Soc Clin Oncol. 1998 Jul;16(7):2522–2527. doi: 10.1200/JCO.1998.16.7.2522. [DOI] [PubMed] [Google Scholar]
- 82.Weiss MJ. New insights into erythropoietin and epoetin alfa: mechanisms of action, target tissues, and clinical applications. Oncologist. 2003 Dec 1;8(Supplement 3):18–29. doi: 10.1634/theoncologist.8-suppl_3-18. [DOI] [PubMed] [Google Scholar]
- 83.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 Feb;18(1):26–30. doi: 10.1016/j.ctcp.2011.02.005. [DOI] [PubMed] [Google Scholar]
- 84.Oh B, Butow PN, Mullan BA, Clarke SJ, Beale PJ, Pavlakis N, 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 Off J Multinatl Assoc Support Care Cancer. 2012 Jun;20(6):1235–1242. doi: 10.1007/s00520-011-1209-6. [DOI] [PubMed] [Google Scholar]
- 85.Lundorff LE, Jønsson BH, Sjøgren P. Modafinil for attentional and psychomotor dysfunction in advanced cancer: a double-blind, randomised, cross-over trial. Palliat Med. 2009 Dec;23(8):731–738. doi: 10.1177/0269216309106872. [DOI] [PubMed] [Google Scholar]
- 86.Sprod LK, Mohile SG, Demark-Wahnefried W, Janelsins MC, Peppone LJ, Morrow GR, et al. Exercise and cancer treatment symptoms in 408 newly diagnosed older cancer patients. J Geriatr Oncol. 2012 Apr 1;3(2):90–97. doi: 10.1016/j.jgo.2012.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Cheung YT, Foo YL, Shwe M, Tan YP, Fan G, Yong WS, et al. Minimal clinically important difference (MCID) for the functional assessment of cancer therapy: cognitive function (FACT-Cog) in breast cancer patients. J Clin Epidemiol. 2014 Jul;67(7):811–820. doi: 10.1016/j.jclinepi.2013.12.011. [DOI] [PubMed] [Google Scholar]
- 88.van Dam FS, Schagen SB, Muller MJ, Boogerd W, Vd Wall E, ME DF, 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 Feb 4;90(3):210–218. doi: 10.1093/jnci/90.3.210. [DOI] [PubMed] [Google Scholar]
- 89.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 Oct;26(7):955–969. doi: 10.1080/13803390490510905. [DOI] [PubMed] [Google Scholar]
- 90.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 Dec 1;104(11):2499–2507. doi: 10.1002/cncr.21482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Nasreddine ZS, Phillips NA, Bédirian V, Charbonneau S, Whitehead V, Collin I, et al. The Montreal Cognitive Assessment, MoCA: a brief screening tool for mild cognitive impairment. J Am Geriatr Soc. 2005 Apr 1;53(4):695–699. doi: 10.1111/j.1532-5415.2005.53221.x. [DOI] [PubMed] [Google Scholar]
- 92.O’Shaughnessy JA, Vukelja SJ, Holmes FA, Savin M, Jones M, Royall D, et al. Feasibility of quantifying the effects of epoetin alfa therapy on cognitive function in women with breast cancer undergoing adjuvant or neoadjuvant chemotherapy. Clin Breast Cancer. 2005 Feb;5(6):439–446. doi: 10.3816/cbc.2005.n.002. [DOI] [PubMed] [Google Scholar]
- 93.Fan HGM, Park A, Xu W, Yi Q-L, Braganza S, Chang J, et al. The influence of erythropoietin on cognitive function in women following chemotherapy for breast cancer. Psychooncology. 2009 Feb;18(2):156–161. doi: 10.1002/pon.1372. [DOI] [PubMed] [Google Scholar]
- 94.Ferguson RJ, McDonald BC, Rocque MA, Furstenberg CT, Horrigan S, Ahles TA, et al. Development of CBT for chemotherapy-related cognitive change: results of a waitlist control trial. Psychooncology. 2012 Feb;21(2):176–186. doi: 10.1002/pon.1878. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Janelsins MC, Peppone LJ, Heckler CE, Kesler S, Sprod LK, Atkins J, Melnik M, Kamen C, Giguere J, Messino MJ, Mohile SG, Mustian KM. YOCAS©® yoga reduces self-reported memory difficulty in cancer survivors in a nationwide randomized clinical trial: investigating relationships between memory and sleep. Integr Cancer Ther. 2015 doi: 10.1177/1534735415617021. [in press] [DOI] [PMC free article] [PubMed] [Google Scholar]