Skip to main content
Journal of Clinical Oncology logoLink to Journal of Clinical Oncology
. 2021 May 27;39(19):2138–2149. doi: 10.1200/JCO.21.00239

Cognitive Function in Older Adults With Cancer: Assessment, Management, and Research Opportunities

Allison Magnuson 1,, Tim Ahles 2, Bihong T Chen 3, Jeanne Mandelblatt 4, Michelle C Janelsins 5
PMCID: PMC8260910  PMID: 34043437

INTRODUCTION

Cancer is primarily a disease of aging, and the majority of new diagnoses of cancer occur in patients ≥ 65 years of age (herein referred to as older).1 At the same time, cancer and its therapies are disease drivers of aging,2 with studies of survivors showing accumulation of multimorbidity, sarcopenia, diminished physical function, and cognitive problems.3 Maintaining cognitive function and independence are important concerns for many patients, and loss of cognitive function is one of the most feared disease side effects.4

CONTEXT

  • Key Objective

  • What are the methods for assessing cognitive function and managing cancer-related cognitive decline (CRCD) in older adults?

  • Knowledge Generated

  • National guidelines recommend validated tools to assess cognitive function and screen older adults with cancer for cognitive problems, including self-report scales, performance-based measures, or a combination of both. Risk factors for the development of CRCD in older adults include low cognitive reserve, comorbidities, and frailty, and a growing body of literature suggests benefit for CRCD interventions, including cognitive rehabilitation approaches and physical activity, but further evidence in the older adult population is needed.

  • Relevance

  • Cognitive function is critical for an older patient's ability to conduct daily activities and maintain independence. Evaluating cognitive function may help inform older patients and their oncologists about the risks of CRCD, provide monitoring for early detection of CRCD, and allow an opportunity to intervene to optimize cognitive function.

Cancer-related cognitive decline (CRCD) is the term that describes subjective and objective changes in cognitive function that occur after a diagnosis of cancer and/or its treatment.5,6 CRCD includes problems with memory, attention, executive function, and speed of processing information.6,7 These symptoms can negatively affect patients' function and quality of life, although are generally more subtle than those that occur in other neurodegenerative disorders of the CNS (eg, dementia and mild cognitive impairment [MCI]).8 CRCD rates vary from 10%-50% depending on the setting, population, measurement, cancer type, and therapy (eg, chemotherapy, endocrine therapies, and radiation).6,7,9-12

Although CRCD mechanisms are not fully understood, CRCD symptoms can be long-lasting, potentially extending for months to years after the completion of therapy for many patients.13,14 Current clinical guidelines15 for older adults with cancer recommend assessment of cognitive function and risks for developing CRCD before initiating therapy to support treatment decision making. Just as the baseline functioning of other organ systems is assessed during an oncology evaluation (eg, renal and cardiac function), an understanding of cognitive function may help inform patients and oncologists about the risk of CRCD and provide an opportunity to monitor cognitive function over time and intervene to preserve cognitive functioning.

This article will highlight clinically relevant factors to consider in the evaluation of cognition when caring for older adults with cancer and discuss when to refer for additional assessment and possible intervention. Finally, we discuss knowledge gaps and future research directions.

EVALUATION OF COGNITIVE FUNCTION IN CLINICAL PRACTICE

Older individuals diagnosed with cancer often have many pre-existing conditions, including neurodegenerative diseases and dementia.16,17 Pre-existing dementia is seen in 3.8%-7% of adults age 65+ years with cancer.18 In older adults without cancer, by 70 years of age, an estimated 13.9% have dementia16 and an additional 22.2% have MCI without overt dementia.17 Additionally, it is possible that cancer and cancer therapy may unmask or exacerbate underlying neurocognitive pathology.19 However, mild deficits are often not apparent in routine clinical encounters.20 Hence, pre-treatment screening can be useful to detect these diseases and consider their impact on treatment decisions.21 Assessing cognitive function can also be useful to establish the patient's baseline before beginning therapy.

The evaluation of cognitive function and the risk for CRCD begins with a detailed patient history of risk factors. There are several known risk factors for the development of CRCD that can be routinely assessed during a clinical history, such as age, sociodemographic factors, comorbid medical conditions, psychologic factors, and host biology (Fig 1).6,7,22 First, older chronologic age can predispose individuals to decline in cognitive function as part of the normative aging process and is a risk factor for the development of CRCD.9,12,19,23 Cognitive aging is associated with gradual changes in memory, attention, and information processing speed with corresponding changes in brain structure and function (reduced gray matter volume and white matter connectivity).6 Cancer treatments mimic these changes, and measurable changes in these elements are observed in imaging studies of older adults with cancer undergoing treatment.24

FIG 1.

FIG 1.

This figure outlines risk factors for CRCD that should be assessed in older adults with cancer, including potentially modifiable risk factors, and associated conditions that can be related to cognitive function. Management considerations for CRCD in older adults with cancer, such as care coordination, referrals to services with cognitive expertise, and behavioral interventions (eg, cognitive rehabilitation and physical activity), are also outlined. aPotentially modifiable factors. APOE, apolipoprotein E; BOMC, Blessed Orientation-Memory-Concentration test; CRCD, cancer-related cognitive decline.

However, the relationship between age and CRCD is not straightforward because aging is heterogeneous and its effects are not fully reflected in chronologic age.25

Cognitive reserve can buffer age-related losses in cognition in a broad array of disorders, and low cognitive reserve is a risk factor for CRCD.11,12 Cognitive reserve refers to the individual differences in innate and acquired cognitive capacity that confers resilience to loss of cognitive function. Advanced education, occupations that require high cognitive challenges, participation in cognitively stimulating activities, and an active lifestyle contribute to higher cognitive reserve, and these factors can be ascertained during a routine personal and social history.26

Other aging-related diseases and conditions (eg, diabetes, cardiovascular disease, and thyroid disease) and frailty are also considered risk factors for cognitive decline.27,28 One study showed that in survivors of breast cancer ≥ 60 years of age, a higher comorbidity burden was associated with worse performance on neuropsychologic assessment pre-treatment.29 Accumulation of comorbidities and functional limitations can lead to frailty, a clinically recognized state of limited reserve. Multiple studies have implicated the co-occurrence of frailty and cognitive decline,9,30,31 which likely represents a patient's systemic loss of reserve and compensatory ability on a broader scale. Patients who are frail and pre-frail (vulnerable) before treatment appear most susceptible to cognitive decline with therapy.30 Therefore, the presence of frailty may signify to the oncologist a limited overall reserve and a heightened risk for CRCD and other adverse outcomes.29

Psychologic distress, anxiety, depression, and other psychologic conditions including those resulting from past trauma can also increase the risk for CRCD.11,32,33 Additionally, symptom clusters including these psychologic conditions along with fatigue and sleep disturbance can contribute to CRCD.34-36 Overall, risk factors for CRCD are routinely collected during a medical history and can help oncology specialists identify patients at risk for cognitive decline during and after therapy.

Host biology is also a risk for CRCD, but methods for assessing patients' biologic age remain investigational. Research markers to categorize older patients with increased risk of CRCD have been identified, and candidates with the potential for clinical utility include genotype testing, biomarkers of biologic age, markers of inflammation, and advanced neuroimaging. Most of the literature in this area thus far has reported preclinical research on the effects of chemotherapy on hippocampal neurogenesis, white matter damage, oxidative stress, vascular damage, inflammation, neuroinflammation, and mitochondrial dysfunction.37 Few of these mechanisms have been studied in clinical research, and very few have been studied specifically in older adults, although the mechanisms underlying these research markers are of great relevance to this population.

Several single-nucleotide polymorphisms may affect vulnerability to CRCD including apolipoprotein E (APOE),9,38 catechol-O-methyltransferase,39 DNA methyltransferase 1,40 and brain-derived neurotrophic factor.41 Two studies have identified that CRCD is associated with at least one allele of APOE4, the genetic risk factor for Alzheimer's disease.9,38 One of these studies specifically investigated the role of APOE single-nucleotide polymorphisms in older adults with cancer receiving chemotherapy, showing that those with at least one copy of the E4 allele exhibited worse executive function.9 Further studies, involving multiple cohorts, are needed to validate these findings and test their clinical utility.

There is also growing interest to better characterize biologic age using markers like epigenetic DNA methylation status rather than by the use of chronologic age because these markers capture accumulation of damage and may better reflect biologic-level fitness. Although several studies have investigated epigenetic age on cancer incidence and mortality, this measure has not been thoroughly investigated in the context of CRCD. One study, however, did show that chemotherapy profoundly alters the epigenetic landscape in whole blood from pre- to post-chemotherapy and that methylation changes in epigenetic DNA were associated with perceived cognitive decline from pre- to post-chemotherapy in patients ≥ 50 years of age.42 Additionally, treatment may increase epigenetic age in patients with breast cancer,43 and association between measures of biologic aging and objective measures of cognition has been observed in survivors of breast cancer.44 Another biologic aging marker that has been recently studied is p16 (INK4a), which is a marker of cellular senescence; this has also been shown to be increased by chemotherapy suggestive of accelerated molecular aging.45

Inflammation has long been proposed as a mechanism underlying CRCD. Several cohort and cross-sectional studies have revealed higher levels of inflammatory markers and associations with CRCD before, during, and after chemotherapy.37,46,47 Of those, elevations in serum or plasma interleukin-6 and tumor necrosis factor-α, and tumor necrosis factor receptors are most consistently correlated with CRCD. High levels of inflammation are also seen with cancer itself and several common comorbidities like diabetes. Studies of inflammatory markers specifically in older adults are needed to assess their usefulness in predicting CRCD. Such information could help also inform interventions that target inflammation.

Brain structural and functional changes seen on magnetic resonance imaging scans are often correlated with worse performance on neuropsychologic assessment.48 Data from a pilot neuroimaging study focusing on older adults with breast cancer demonstrated brain structural and cognitive changes including reductions in both temporal lobe volume and oral reading recognition scores among patients receiving a docetaxel and cyclophosphamide regimen.49,50 Additionally, significant gray matter density reductions were observed in several specific brain regions in patients who received chemotherapy.24 Furthermore, there were alterations in brain white matter microstructures showing significant increases in mean diffusivity and radial diffusivity values in the genu of the corpus callosum over time, indicating white matter injury.50 These findings may potentially serve as neuroimaging biomarkers for identifying CRCD in older adults undergoing cancer treatment.

Screening to Evaluate Cognitive Function in Older Adults With Cancer

The ASCO Guideline for Geriatric Oncology recommends that all older adults with cancer receive a screening evaluation of cognitive function to help inform treatment decision making.15 Assessment of cognition with a screening measure is important, as cognitive issues are regularly under-reported and easily overlooked in an oncology setting.51 Brief screening tools such as the Mini-Cog,52 Montreal Cognitive Assessment,53 or the Blessed Orientation-Memory-Concentration54 are recommended to identify patients with cancer and decreased cognitive function who may need additional evaluation (Table 1).15 Many older adults (15%-48%) have an abnormal cognitive screen even before treatment.55-58 Although these tools are not diagnostic, they can identify patients who may benefit from a referral for a more comprehensive evaluation of cognitive function. It is also important to note that these tools were developed to screen for dementia and may not identify those with more subtle cognitive changes seen in CRCD. Thus, patients who self-report cognitive problems should also be considered for evaluation.

TABLE 1.

Exemplar Measures for Cognitive Assessment in Older Adults With Cancer

graphic file with name jco-39-2138-g002.jpg

Although these screening tools are used by some oncologists specializing in geriatrics, they are underutilized in routine oncology care because of lack of time and other resources.21,66 If ancillary staff are available, they could be trained to administer the screening tools. When that is not possible, perceived cognitive functioning could be assessed by patient report using short questionnaires, such as the Functional Assessment of Cancer Therapy-Cognition,59 the Patient-Reported Outcomes Measurement Information System–Cognitive Function,61 or the European Organization for Research and Treatment of Cancer Quality of Life questionnaire-Cognitive Function (EORTC-CF)60 (Table 1). These tools have established thresholds that are suggested to indicate a need for further assessment before treatment decision making.67-69 If there are concerns about cognitive function, the oncology clinician could refer to neuropsychology and other specialty services as further outlined in the Management Considerations section below.

Cognitive Effects During and After Cancer Therapy

It is important to monitor cognition during and after cancer therapy. Monitoring can be conducted using self-reported measures (eg, perceived cognition), performance-based measures (eg, objective cognition), or a combination of both. In this section, we review the cognitive effects of different classes of therapy.

Chemotherapy

Multiple studies have demonstrated the development of CRCD in 10% to more than 50% of patients following exposure to chemotherapy, with prevalence estimates varying based upon the population under study and the cognitive assessments used. In one of the largest studies to date, patients with breast cancer receiving chemotherapy were approximately four times more likely to report worsening of perceived cognition from pre-chemotherapy to post-chemotherapy as compared to noncancer controls.10 Although there was slight improvement in perceived cognition 6 months after completion of chemotherapy, patients were still almost three times more likely to report a clinically meaningful decline in perceived cognition (based on 0.5-standard deviation changes). Patients with cancer also demonstrated worse performance on objective measures in multiple cognitive domains, including memory, attention, and executive function up to 6 months post-chemotherapy as compared to noncancer controls.11 In a subset analysis of older patients ≥ 50 years of age from this cohort, longitudinal worsening of cognition was associated with increased frailty.31

In the largest study of cognition in older adults with cancer to date, the Thinking and Living with Cancer study, patients with breast cancer of age 60 years and older who received chemotherapy experienced declines of approximately 0.3 standard deviation in performance-based measures of attention, processing speed, and executive function over 24 months, as compared to those on hormonal therapy or noncancer controls. Patients receiving endocrine therapy, however, also had reduced performance in learning and memory.9

Other smaller studies evaluating longitudinal change in cognition in patients with breast cancer and other cancer types have demonstrated similar findings, although older adults were less well represented on many of these reports.6,13 Generally, the magnitude of decline in cognition is subtle in comparison to degenerative neurologic conditions. Despite this, these declines have negative effects on quality of life.8 The long-term effects of chemotherapy on cognition are less clear; some studies suggesting resolution of cognitive changes over time70 and others demonstrate persistent changes several months to years after completion of therapy.13,14

Nonchemotherapy agents

Multiple studies have been conducted on the cognitive effects of endocrine-based therapies commonly used for prostate cancer and breast cancer,71 malignancies commonly diagnosed in older adults. Several studies have been conducted on older patients with breast cancer receiving tamoxifen or aromatase inhibitor therapy demonstrating mixed results regarding their effects on cognition. Most studies have shown an effect of endocrine therapies on cognitive function in patients with breast cancer,12,72 whereas others have not shown significant change.73 For example, in a study of patients of age 60 years and older receiving aromatase inhibitors compared with age-matched controls, no significant declines in performance-based cognitive assessment were seen over the first 6 months of treatment.73 However, in a separate cohort of postmenopausal women with breast cancer, patients receiving anastrozole (with or without prior chemotherapy) demonstrated worse executive function scores before treatment, as well as at multiple time points up to 18 months after initiation of therapy.72 Declines in working memory and concentration were also observed during the first 6 months of anastrozole therapy, with subsequent improvement; however, patients receiving anastrozole only (no prior chemotherapy) demonstrated later declines in working memory and concentration (up to 18 months after initiation of therapy).

Longitudinal studies of the effects of androgen deprivation therapy (ADT) for the treatment of prostate cancer on cognitive function have generated mixed results. A systematic review and meta-analysis of 14 studies demonstrated that patients treated with ADT performed worse than noncancer controls on performance-based visuomotor tasks, although no significant differences were noted in other cognitive domains.74 Receipt of ADT is also associated with a 21% increase in dementia risk.75

Newer cancer therapies such as immunotherapy with immune checkpoint inhibitors used as monotherapy or in combination with other treatments may also contribute to CRCD.76,77 Other immunotherapy modalities, such as chimeric antigen receptor T-cell therapy targeting cancer-related cell-surface molecules, are frequently associated with cytokine release syndrome and neurotoxicity.78,79 Neurotoxicity associated with chimeric antigen receptor T-cell therapy occurs in more than 40% of patients whose most frequent neurologic symptoms are cognitive disorders with and without language impairment.80 However, it is important to note that the majority of studies evaluating the effects of immune-based therapies on cognition included mainly younger patients, and further research is needed in older adults.

MANAGEMENT CONSIDERATIONS

As the population ages, the number of older patients with cancer who are at risk for CRCD and/or those who have a pre-existing cognitive comorbidity such as MCI or dementia will increase.3 These demographic trends will drive the need for better integration of CRCD management into routine practice.

CRCD Management Issues for Treatment Decision Making

One obvious management issue related to CRCD and pre-existing cognitive problems is that decision-making capacity should be assessed for these patients, particularly for complex decisions about cancer treatment options.81,82 Additionally, understanding of patients' cognitive status may also help to tailor their treatment approach and develop a more comprehensive plan for monitoring and support during treatment and survivorship care.25,28 Studies have shown that worse performance on cognitive screening assessment is associated with adverse cancer-related outcomes, such as chemotherapy toxicity and overall survival.57,83 For example, one such study of patients ≥ 70 years of age receiving chemotherapy demonstrated that a Mini-Mental Status Exam Score < 30 was independently associated with grade 3-5 nonhematologic chemotherapy toxicity.83 In another study of patients ≥ 75 years of age with hematologic malignancies, an abnormal five-word delayed recall screening test was associated with worse overall survival.57 Patients with cognitive impairment may have more difficulties understanding the complex instructions and schedules that are typical of cancer therapies, managing medications independently, and recognizing and reporting side effects and treatment complications in a timely manner.25,84 Thus, ensuring adequate medical and social support for older patients with concurrent cancer and comorbid cognitive impairment is critical, and formal designation of a health care proxy and decisional support should be confirmed. Additionally, caregiver distress and burden are increased among those who care for patients with cognitive impairment, and monitoring of the caregiver's health and their ability to provide the necessary support is important as well.85

For cognitive concerns that arise during and following exposure to therapy, published guidelines and resources for navigating cancer survivorship and managing CRCD exist.15,28,86-88 These include referrals for neuropsychology evaluations, as well as cognitive rehabilitation and integrative oncology care services. These services can further help manage patients with CRCD or those at risk for CRCD. Accessibility of these resources can be variable, and oncologists should have knowledge of available resources in their community before treatment of high-risk patients. Because of the coronavirus pandemic, opportunities for remote neuropsychologic assessments are being explored and if successful, these may be options for research and clinical care in the future.89

Survivorship Care Management and Interventions

More than 10 million older cancer survivors currently reside in the United States.90 Given the prevalence of CRCD in cancer survivors, with up to 35% of patients reporting long-term cognitive symptoms,7 the development of treatments for CRCD is critical. Cancer survivors are very interested in receiving these therapies.91 Although no standard approach to managing CRCD symptoms exists, several interventions have been studied and can be considered.

Behavioral interventions

Behavioral interventions, such as cognitive rehabilitation approaches and physical activity, have been a primary area of investigation for the management of CRCD. Cognitive rehabilitation programs broadly include two treatment types—strategy or educational programs (typically a cognitive behavioral therapy–based approach) and computer-based cognitive training programs—or a combination of the two types. Table 2 highlights select published studies of cognitive rehabilitation interventions.92-106 In a recent systematic review of cognitive rehabilitation programs for CRCD, including 19 studies with 1,124 participants, all studies observed improvements in at least one cognitive measure.107 These studies included mainly younger cancer survivors and only two focused on older cancer survivors.94,95 Of note, these were secondary analyses of outcomes of older cancer survivors included in larger studies of the general population. Additionally, nearly all studies were conducted months to years after cancer therapy was completed. For older adults who may be at greater risk for CRCD and potential subsequent loss of functional independence as a result of cognitive changes, intervening earlier, possibly along with cancer therapy, may help to mitigate the development of CRCD.

TABLE 2.

Cognitive Rehabilitation Interventions for CRCD

graphic file with name jco-39-2138-g003.jpg

A growing body of evidence suggests that physical activity and exercise programs may also play a role in managing CRCD. A systematic review including 29 clinical trials with 3,440 participants evaluated the effect of physical activity and exercise on cognitive function in patients with cancer.108 Overall, the majority of studies were designed to evaluate cognitive function as a secondary outcome. Only three studies evaluated the effect of exercise on CRCD as a primary outcome. Twelve studies observed improvement in perceived cognition, and three studies detected improvement in performance-based cognitive assessment. Again, the majority of patients included in these studies were younger, and less is known about the benefits of physical activity and exercise on cognition in older adults with cancer.

Pharmacologic interventions

There are no established pharmacologic interventions for CRCD. Research on pharmacologic interventions for CRCD has been limited, and results to date have not provided confirmatory results. Pharmacologic intervention studies addressing CRCD have not been conducted specifically on older adults.

Methylphenidate and modafinil are two stimulants that have been studied—both promote wakefulness at least in part by blocking dopamine transporters to increase dopamine levels. For methylphenidate, some trials have shown improvements in cognition, with others not showing any improvement.109-111 Modafinil has been studied as part of a secondary analysis of a phase II randomized trial originally targeting fatigue and it reported improvements in memory and attention in breast cancer survivors.112 A second randomized pilot study also showed improvements in attention and psychomotor speed in adults with advanced cancer.113

Donepezil is an acetylcholinesterase inhibitor that has shown cognitive benefits in patients with brain tumors.114 In a randomized pilot study of breast cancer survivors up to 5 years post-treatment, donepezil improved cognitive function both by both patient report and objective testing.115 However, further studies in older adults with cancer are needed before routine clinical use of donepezil in this setting.

Gaps in Knowledge to Guide Evidence-Based Cognitive Assessment and Care

Although considerable progress has been made in understanding the relationships between cancer, cognition, and aging, several questions remain unanswered. High-priority areas of research include increasing the representation of older adults into CRCD research, particularly those with pre-existing cognitive impairment, advancing cognitive assessment methods and cognitive screening for older adults with cancer, and developing CRCD interventions for older adults.

Older adults remain largely under-represented in CRCD studies, and nearly all studies exclude patients with diagnosed neurodegenerative conditions such as MCI or dementia. Little CRCD research has addressed this population in particular, and more knowledge is needed to better guide treatment decision making and supportive care strategies for this vulnerable group of patients. Additionally, as cancer therapy options evolve, a better understanding of the effects on cognition of targeted therapies and immune-based therapies is needed in older adults.

Cognitive assessment in routine clinical practice remains underutilized21,66 despite national guidelines.15 Mechanisms for implementing cognitive screening tools into oncology care for older adults should be a priority area of research. In the research setting, the International Cognition and Cancer Task Force has recommended a core battery of neuropsychologic assessments for use,5 but it offers no specific considerations for the older adult population. Cognitive evaluation in older adults should include a functional component with measures of cognition, such as Instrumental Activities of Daily Living (eg, medication management). These would allow a better understanding of how cognitive changes influence day-to-day functioning for older adults with cancer and their ability to maintain independence.116 Assessments for more subtle cognitive effects, such as computerized neuroscience-based methods, are considered in detail elsewhere but could also be useful for the study of CRCD in older adults.117

Finally, identification of treatment options for CRCD is a high-priority area of research. Although interventions have been developed, most have been focused on younger patients. Given the unique considerations for CRCD in older adults as outlined above, tailored interventions for this population are needed. CRCD interventions in older adults should evaluate for possible benefit using traditional cognitive measures, but also using measures of functional cognition and independence. Most CRCD interventions have been evaluated in the survivorship setting (months to years after completion of therapy). Given the likelihood of decline in functional cognition and loss of independence in older adults, intervening earlier (eg, concurrent with cancer therapy) should be considered.

In conclusion, cognition, cancer, and aging are dynamically inter-related. A growing body of literature demonstrates that a subset of older patients with cancer will experience CRCD. Like other comorbidity and organ function assessments, cognitive evaluation should be incorporated as part of baseline oncology care to help inform discussions with patients about treatment risks and benefits and guide the development of supportive care strategies where needed. Risk factors for the development of CRCD have been identified. Several of these are highly relevant for the geriatric oncology population, such as cognitive reserve, comorbidities, and advanced age. Emerging data suggest that intervention strategies may be useful in the management of CRCD. However, further studies are needed in this area, particularly for the older adult population. Older patients value independence and cognitive function as well as optimizing cancer outcomes (eg, survival), and we should strive to maintain cognitive function and other aspects of quality of life while also providing goal-concordant cancer therapy individualized to each patient's preferences about treatment.

Michelle C. Janelsins

Consulting or Advisory Role: Charles River Analytics

No other potential conflicts of interest were reported.

SUPPORT

Supported in part by the National Institute of Health (NIH), National Institute on Aging (NIA), Beeson Career Development Award (K76 AG064394 to A.M.), and NIH National Cancer Institute (R01CA129769, R01AG068193, and R35CA197289 to J.M.; R01 CA218496, U54 CA137788, and P30 CA 008748 to T.A., and R01CA231014 and R01CA249467 to M.C.J.).

AUTHOR CONTRIBUTIONS

Conception and design: All authors

Collection and assembly of data: All authors

Data analysis and interpretation: All authors

Manuscript writing: All authors

Final approval of manuscript: All authors

Accountable for all aspects of the work: All authors

AUTHORS' DISCLOSURES OF POTENTIAL CONFLICTS OF INTEREST

Cognitive Function in Older Adults with Cancer: Assessment, Management, and Research Opportunities

The following represents disclosure information provided by the authors of this manuscript. All relationships are considered compensated unless otherwise noted. Relationships are self-held unless noted. I = Immediate Family Member, Inst = My Institution. Relationships may not relate to the subject matter of this manuscript. For more information about ASCO's conflict of interest policy, please refer to www.asco.org/rwc or ascopubs.org/jco/authors/author-center.

Open Payments is a public database containing information reported by companies about payments made to US-licensed physicians (Open Payments).

Michelle C. Janelsins

Consulting or Advisory Role: Charles River Analytics

No other potential conflicts of interest were reported.

REFERENCES

  • 1.Siegel RL, Miller KD, Jemal A.Cancer statistics, 2019 CA Cancer J Clin 697–342019 [DOI] [PubMed] [Google Scholar]
  • 2.Hodes RJ, Sierra F, Austad SN, et al. Disease drivers of aging. Ann N Y Acad Sci. 2016;1386:45. doi: 10.1111/nyas.13299. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Bluethmann SM, Mariotto AB, Rowland JH.Anticipating the “silver tsunami”: Prevalence trajectories and comorbidity burden among older cancer survivors in the United States Cancer Epidemiol Biomarkers Prev; 251029–1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Fried TR, Bradley EH, Towle VR, et al. Understanding the treatment preferences of seriously ill patients N Engl J Med 3461061–10662002 [DOI] [PubMed] [Google Scholar]
  • 5.Wefel JS, Vardy J, Ahles T, et al. International Cognition and Cancer Task Force recommendations to harmonise studies of cognitive function in patients with cancer Lancet Oncol 12703–7082011 [DOI] [PubMed] [Google Scholar]
  • 6.Ahles TA, Root JC.Cognitive effects of cancer and cancer treatments Annu Rev Clin Psychol 14425–4512018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Janelsins MC, Kesler SR, Ahles TA, et al. Prevalence, mechanisms, and management of cancer-related cognitive impairment Int Rev Psychiatry 26102–1132014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Selamat MH, Loh SY, Mackenzie L, et al. Chemobrain experienced by breast cancer survivors: A meta-ethnography study investigating research and care implications. PLoS One. 2014;9:e108002. doi: 10.1371/journal.pone.0108002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Mandelblatt JS, Small BJ, Luta G, et al. Cancer-related cognitive outcomes among older breast cancer survivors in the thinking and living with cancer study J Clin Oncol 363211–32222018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Janelsins MC, Heckler CE, Peppone LJ, et al. Cognitive complaints in survivors of breast cancer after chemotherapy compared with age-matched controls: An analysis from a nationwide, multicenter, prospective longitudinal study J Clin Oncol 35506–5142017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Janelsins MC, Heckler CE, Peppone LJ, et al. Longitudinal trajectory and characterization of cancer-related cognitive impairment in a nationwide cohort study J Clin Oncol 36:3231–32392018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.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 284434–44402010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Collins B, Mackenzie J, Tasca GA, et al. Persistent cognitive changes in breast cancer patients 1 year following completion of chemotherapy J Int Neuropsychol Soc 20370–3792014 [DOI] [PubMed] [Google Scholar]
  • 14.Yamada TH, Denburg NL, Beglinger LJ, et al. Neuropsychological outcomes of older breast cancer survivors: Cognitive features ten or more years after chemotherapy J Neuropsychiatry Clin Neurosci 2248–542010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Mohile SG, Dale W, Somerfield MR, et al. Practical assessment and management of vulnerabilities in older patients receiving chemotherapy: ASCO guideline for geriatric oncology J Clin Oncol 362326–23472018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Plassman BL, Langa KM, Fisher GG, et al. Prevalence of dementia in the United States: The aging, demographics, and memory study Neuroepidemiology 29125–1322007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Plassman BL, Langa KM, Fisher GG, et al. Prevalence of cognitive impairment without dementia in the United States Ann Intern Med 148427–4342008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Raji MA, Kuo YF, Freeman JL, et al. Effect of a dementia diagnosis on survival of older patients after a diagnosis of breast, colon, or prostate cancer: Implications for cancer care Arch Intern Med 1682033–20402008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Mandelblatt JS, Jacobsen PB, Ahles T.Cognitive effects of cancer systemic therapy: Implications for the care of older patients and survivors J Clin Oncol 322617–26262014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kirkhus L, Benth JŠ, Rostoft S, et al. Geriatric assessment is superior to oncologists’ clinical judgement in identifying frailty Br J Cancer 117470–4772017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Mohile SG, Magnuson A, Pandya C, et al. Community oncologists' decision-making for treatment of older patients with cancer J Natl Compr Canc Netw 16301–3092018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Wefel JS, Kesler SR, Noll KR, et al. Clinical characteristics, pathophysiology, and management of noncentral nervous system cancer‐related cognitive impairment in adults CA Cancer J Clin 65123–1382015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Park DC, Reuter-Lorenz P.The adaptive brain: Aging and neurocognitive scaffolding Annu Rev Psychol 60173–1962009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Chen BT, Jin T, Patel SK, et al. Gray matter density reduction associated with adjuvant chemotherapy in older women with breast cancer Breast Cancer Res Treat 172363–3702018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Magnuson A, Mohile S, Janelsins M.Cognition and cognitive impairment in older adults with cancer Curr Geriatr Rep 5213–2192016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Cabeza R, Albert M, Belleville S, et al. Maintenance, reserve and compensation: The cognitive neuroscience of healthy ageing Nat Rev Neurosci 19701–7102018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Hay M, Barnes C, Huentelman M, et al. Hypertension and age-related cognitive impairment: Common risk factors and a role for precision aging. Curr Hypertens Rep. 2020;22:80. doi: 10.1007/s11906-020-01090-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Magnuson A, Sattar S, Nightingale G, et al. A practical guide to geriatric syndromes in older adults with cancer: A focus on falls, cognition, polypharmacy, and depression Am Soc Clin Oncol Ed Book 39e96–e1092019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.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 321909–19182014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Mandelblatt JS, Zhou X, Small BJ, et al. Deficit accumulation frailty trajectories of older breast cancer survivors and non-cancer controls: The thinking and living with cancer study. J Natl Cancer Inst. doi: 10.1093/jnci/djab003. 10.1093/jnci/djab003 epub ahead of print January 23, 2021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Magnuson A, Lei L, Gilmore N, et al. Longitudinal relationship between frailty and cognition in patients 50 years and older with breast cancer J Am Geriatr Soc 67928–9362019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Hermelink K, Buhner M, Sckopke P, et al. Chemotherapy and post-traumatic stress in the causation of cognitive dysfunction in breast cancer patients. J Natl Cancer Inst. 2017;109:djx057. doi: 10.1093/jnci/djx057. [DOI] [PubMed] [Google Scholar]
  • 33.Kamen C, Scheiber C, Janelsins M, et al. Effects of childhood trauma exposure and cortisol levels on cognitive functioning among breast cancer survivors Child Abuse Negl 72163–1712017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.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 90210–2181998 [DOI] [PubMed] [Google Scholar]
  • 35.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 147445–4552014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Carroll JE, Small BJ, Tometich DB, et al. Sleep disturbance and neurocognitive outcomes in older patients with breast cancer: Interaction with genotype Cancer 1254516–45242019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Castel H, Denouel A, Lange M, et al. Biomarkers associated with cognitive impairment in treated cancer patients: Potential predisposition and risk factors. Front Pharmacol. 2017;8:138. doi: 10.3389/fphar.2017.00138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Ahles TA, Saykin AJ, Noll WW, et al. The relationship of APOE genotype to neuropsychological performance in long‐term cancer survivors treated with standard dose chemotherapy Psychooncology 12612–6192003 [DOI] [PubMed] [Google Scholar]
  • 39.Small BJ, Rawson KS, Walsh E, et al. Catechol-O-methyltransferase genotype modulates cancer treatment-related cognitive deficits in breast cancer survivors Cancer 1171369–13762011 [DOI] [PubMed] [Google Scholar]
  • 40.Chan A, Yeo A, Shwe M, et al. An evaluation of DNA methyltransferase 1 (DNMT1) single nucleotide polymorphisms and chemotherapy-associated cognitive impairment: A prospective, longitudinal study. Sci Rep. 2019;9:14570. doi: 10.1038/s41598-019-51203-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Ng T, Teo SM, Yeo HL, et al. Brain-derived neurotrophic factor genetic polymorphism (rs6265) is protective against chemotherapy-associated cognitive impairment in patients with early-stage breast cancer Neuro Oncol 18244–2512016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Yao S, Hu Q, Kerns S, et al. Impact of chemotherapy for breast cancer on leukocyte DNA methylation landscape and cognitive function: A prospective study. Clin Epigenetics. 2019;11:45. doi: 10.1186/s13148-019-0641-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Sehl ME, Carroll JE, Horvath S, et al. The acute effects of adjuvant radiation and chemotherapy on peripheral blood epigenetic age in early stage breast cancer patients NPJ Breast Cancer 61–52020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Carroll JE, Van Dyk K, Bower JE, et al. Cognitive performance in survivors of breast cancer and markers of biological aging Cancer 125298–3062019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sanoff HK, Deal AM, Krishnamurthy J, et al. Effect of cytotoxic chemotherapy on markers of molecular age in patients with breast cancer. J Natl Cancer Inst. 2014;106:dju057. doi: 10.1093/jnci/dju057. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Toh YL, Wang C, Ho HK, et al. Distinct cytokine profiles across trajectories of self-perceived cognitive impairment among early-stage breast cancer survivors. J Neuroimmunol. 2020;342:577196. doi: 10.1016/j.jneuroim.2020.577196. [DOI] [PubMed] [Google Scholar]
  • 47.Williams AM, Shah R, Shayne M, et al. Associations between inflammatory markers and cognitive function in breast cancer patients receiving chemotherapy J Neuroimmunol 31417–232018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.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 332971–29832012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Chen BT, Sethi SK, Jin T, et al. Assessing brain volume changes in older women with breast cancer receiving adjuvant chemotherapy: A brain magnetic resonance imaging pilot study. Breast Cancer Res. 2018;20:38. doi: 10.1186/s13058-018-0965-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Chen BT, Ye N, Wong CW, et al. Effects of chemotherapy on aging white matter microstructure: A longitudinal diffusion tensor imaging study J Geriatr Oncol 11290–2962020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Amjad H, Roth DL, Sheehan OC, et al. Underdiagnosis of dementia: An observational study of patterns in diagnosis and awareness in US older adults J Gen Intern Med 331131–11382018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Borson S, Scanlan JM, Chen P, et al. The mini-cog as a screen for dementia: Validation in a population-based sample J Am Geriatr Soc 511451–14542003 [DOI] [PubMed] [Google Scholar]
  • 53.Nasreddine ZS, Phillips NA, Bédirian V, et al. The Montreal Cognitive Assessment, MoCA: A brief screening tool for mild cognitive impairment J Am Geriatr Soc 53695–6992005 [DOI] [PubMed] [Google Scholar]
  • 54.Katzman R, Brown T, Fuld P, et al. Validation of a short Orientation-Memory-Concentration Test of cognitive impairment Am J Psychiatry 140734–7391983 [DOI] [PubMed] [Google Scholar]
  • 55.Corre R, Greillier L, Le Caer H, et al. Use of a comprehensive geriatric assessment for the management of elderly patients with advanced non-small-cell lung cancer: The phase III randomized ESOGIA-GFPC-GECP 08-02 study J Clin Oncol 341476–14832016 [DOI] [PubMed] [Google Scholar]
  • 56.Mohile SG, Epstein RM, Hurria A, et al. Communication with older patients with cancer using geriatric assessment: A cluster-randomized clinical trial from the National Cancer Institute community oncology research program JAMA Oncol 6196–2042020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Hshieh TT, Jung WF, Grande LJ, et al. Prevalence of cognitive impairment and association with survival among older patients with hematologic cancers JAMA Oncol 4686–6932018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Magnuson A, Lemelman T, Pandya C, et al. Geriatric assessment with management intervention in older adults with cancer: A randomized pilot study Support Care Cancer 26605–6132018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Wagner LI, Sweet J, Butt Z, et al. Measuring patient self-reported cognitive function: Development of the functional assessment of cancer therapy-cognitive function instrument J Support Oncol 7W32–W392009 [Google Scholar]
  • 60.Aaronson NK, Ahmedzai S, Bergman B, et al. The European Organization for Research and Treatment of Cancer QLQ-C30: A quality-of-life instrument for use in international clinical trials in oncology J Natl Cancer Inst 85365–3761993 [DOI] [PubMed] [Google Scholar]
  • 61.National Institutes of Health: PROMIS Instrument Development and Validation Scientific Standards Version 2.0. https://www.healthmeasures.net/images/PROMIS/PROMISStandards_vers2.0_final.pdf [Google Scholar]
  • 62.Brandt J, Benedict RHB. Hopkins Verbal Learning Test-Rrevised: Professional Manual. Lutz, FL, Psychological Assessment Resources; 2001. [Google Scholar]
  • 63.Reynolds C. Comprehensive Trail-Making Test Professional Manual. Austin, TX: Pro-Ed; 2002. [Google Scholar]
  • 64.Reitan RM.Validity of the Trail Making Test as an indicator of organic brain damage Perceptual Mot Skills 8271–2761958 [Google Scholar]
  • 65.Benton L, Hamsher K, Sivan A. Controlled Oral Word Association Test. Multilingual Aphasia Examination. Iowa City, IA, AJA Associates; 1994. [Google Scholar]
  • 66.Dale W, Williams GR, MacKenzie AR, et al. How is geriatric assessment used in clinical practice for older adults with cancer? A survey of cancer providers by the American Society of Clinical Oncology. JCO Oncol Pract. doi: 10.1200/OP.20.00442. 10.1200/OP.20.00442 epub ahead of print on October 15, 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Dyk KV, Crespi CM, Petersen L, et al. Identifying cancer-related cognitive impairment using the FACT-Cog Perceived Cognitive Impairment. JNCI Cancer Spectr. 2019;4:pkz099. doi: 10.1093/jncics/pkz099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Bell ML, Dhillon HM, Bray VJ, et al. Important differences and meaningful changes for the Functional Assessment of Cancer Therapy-Cognitive Function (FACT-Cog) J Patient Rep Outcomes. 2018;2:48. [Google Scholar]
  • 69.Giesinger JM, Loth FL, Aaronson NK, et al. Thresholds for clinical importance were established to improve interpretation of the EORTC QLQ-C30 in clinical practice and research J Clin Epidemiol 1181–82020 [DOI] [PubMed] [Google Scholar]
  • 70.Koppelmans V, Breteler M, Boogerd W, et al. Neuropsychological performance in survivors of breast cancer more than 20 years after adjuvant chemotherapy J Clin Oncol 301080–10862012 [DOI] [PubMed] [Google Scholar]
  • 71.Ganz PA, Petersen L, Castellon SA, et al. Cognitive function after the initiation of adjuvant endocrine therapy in early-stage breast cancer: An observational cohort study J Clin Oncol 313559–35672014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Bender CM, Merriman JD, Gentry AL, et al. Patterns of change in cognitive function with anastrozole therapy Cancer 1212627–26362015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.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 14132–1402014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.McGinty HL, Phillips KM, Jim HS, et al. Cognitive functioning in men receiving androgen deprivation therapy for prostate cancer: A systematic review and meta-analysis Support Care Cancer 222271–22802014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Cui H, Wang Y, Li F, et al. Quantifying observational evidence for risk of dementia following androgen deprivation therapy for prostate cancer: An updated systematic review and meta-analysis Prostate Cancer Prostatic Dis 2415–232021 [DOI] [PubMed] [Google Scholar]
  • 76.Wang Y, Deng W, Li N, et al. Combining immunotherapy and radiotherapy for cancer treatment: Current challenges and future directions. Front Pharmacol. 2018;9:185. doi: 10.3389/fphar.2018.00185. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Santos JC, Pyter LM. Neuroimmunology of behavioral comorbidities associated with cancer and cancer treatments. Front Immunol. 2018;9:1195. doi: 10.3389/fimmu.2018.01195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Neelapu SS, Tummala S, Kebriaei P, et al. Chimeric antigen receptor T-cell therapy—Assessment and management of toxicities Nat Rev Clin Oncol 1547–622018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Joly F, Castel H, Tron L, et al. Potential effect of immunotherapy agents on cognitive function in cancer patients J Natl Cancer Inst 112123–1272019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Belin C, Devic P, Ayrignac X, et al. Description of neurotoxicity in a series of patients treated with CAR T-cell therapy. Sci Rep. 2020;10:18997. doi: 10.1038/s41598-020-76055-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Appelbaum PS.Assessment of patients' competence to consent to treatment N Engl J Med 3571834–18402007 [DOI] [PubMed] [Google Scholar]
  • 82.Marron JM, Kyi K, Appelbaum PS, et al. Medical decision-making in oncology for patients lacking capacity Am Soc Clin Oncol Ed Book 40e186–e962020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Extermann M, Boler I, Reich RR, et al. Predicting the risk of chemotherapy toxicity in older patients: The Chemotherapy Risk Assessment Scale for High-Age patients (CRASH) score Cancer 1183377–33862012 [DOI] [PubMed] [Google Scholar]
  • 84.Karuturi M, Wong ML, Hsu T, et al. Understanding cognition in older patients with cancer J Geriatr Oncol 7258–2692016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Kadambi S, Loh KP, Dunne R, et al. Older adults with cancer and their caregivers—Current landscape and future directions for clinical care Nat Rev Clin Oncol 17742–7552020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Runowicz CD, Leach CR, Henry NL, et al. American Cancer Society/American Society of Clinical Oncology breast cancer survivorship care guideline J Clin Oncol 34611–6352016 [DOI] [PubMed] [Google Scholar]
  • 87.Denlinger CS, Ligibel JA, Are M, et al. Survivorship: Cognitive function, version 1.2014 J Natl Compr Cancer Netw 12976–9862014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Mohile SG, Velarde C, Hurria A, et al. Geriatric assessment-guided care processes for older adults: A Delphi consensus of geriatric oncology experts J Natl Compr Cancer Netw 131120–11302015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Feenstra HEM, Murre JMJ, Vermeulen IE, et al. Reliability and validity of a self-administered tool for online neuropsychological testing: The Amsterdam Cognition Scan J Clin Exp Neuropsychol 40253–2732018 [DOI] [PubMed] [Google Scholar]
  • 90.American Cancer Society . Cancer Treatment & Survivorship Facts & Figures 2019-2021. Atlanta, GA: American Cancer Society; 2019. [Google Scholar]
  • 91.Lange M, Licaj I, Clarisse B, et al. Cognitive complaints in cancer survivors and expectations for support: Results from a web–based survey Cancer Med 82654–26632019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Cherrier M, Anderson K, David D, et al. A randomized trial of cognitive rehabilitation in cancer survivors Life Sci 93617–6222013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.King S, Green HJ. Psychological intervention for improving cognitive function in cancer survivors: A literature review and randomized controlled trial. Front Oncol. 2015;5:72. doi: 10.3389/fonc.2015.00072. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.McDougall GJ., JrMemory improvement program for elderly cancer survivors Geriatr Nurs 22185–1902001 [DOI] [PubMed] [Google Scholar]
  • 95.McDougall GJ, Becker H, Acee TW, et al. Symptom management of affective and cognitive disturbance with a group of cancer survivors Arch Psychiatr Nurs 2524–352011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Ercoli L, Petersen L, Hunter A, et al. Cognitive rehabilitation group intervention for breast cancer survivors: Results of a randomized clinical trial Psychooncology 241360–13672015 [DOI] [PubMed] [Google Scholar]
  • 97.Schuurs A, Green HJ.A feasibility study of group cognitive rehabilitation for cancer survivors: Enhancing cognitive function and quality of life Psychooncology 221043–10492013 [DOI] [PubMed] [Google Scholar]
  • 98.Ferguson RJ, Sigmon ST, Pritchard AJ, et al. A randomized trial of videoconference‐delivered cognitive behavioral therapy for survivors of breast cancer with self‐reported cognitive dysfunction Cancer 1221782–17912016 [DOI] [PubMed] [Google Scholar]
  • 99.Park J-H, Jung YS, Kim KS, et al. Effects of compensatory cognitive training intervention for breast cancer patients undergoing chemotherapy: A pilot study Support Care Cancer 251887–18962017 [DOI] [PubMed] [Google Scholar]
  • 100.Dos Santos M, Hardy‐Léger I, Rigal O, et al. Cognitive rehabilitation program to improve cognition of cancer patients treated with chemotherapy: A 3‐arm randomized trial Cancer 1265328–53362020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Damholdt M, Mehlsen M, O'Toole M, et al. Web‐based cognitive training for breast cancer survivors with cognitive complaints—a randomized controlled trial Psychooncology 251293–13002016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Bray VJ, Dhillon HM, Bell ML, et al. Evaluation of a web-based cognitive rehabilitation program in cancer survivors reporting cognitive symptoms after chemotherapy J Clin Oncol 35217–2252017 [DOI] [PubMed] [Google Scholar]
  • 103.Kesler S, Hosseini SH, Heckler C, et al. Cognitive training for improving executive function in chemotherapy-treated breast cancer survivors Clin Breast Cancer 13299–3062013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Mihuta M, Green H, Shum D. Efficacy of a web‐based cognitive rehabilitation intervention for adult cancer survivors: A pilot study. Eur J Cancer Care. 2018;27:e12805. doi: 10.1111/ecc.12805. [DOI] [PubMed] [Google Scholar]
  • 105.Von Ah D, Carpenter JS, Saykin A, et al. Advanced cognitive training for breast cancer survivors: A randomized controlled trial Breast Cancer Res Treat 135799–8092012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Becker H, Henneghan AM, Volker DL, et al. A pilot study of a cognitive-behavioral intervention for breast cancer survivors Oncol Nurs Forum 44255–2642017 [DOI] [PubMed] [Google Scholar]
  • 107.Fernandes HA, Richard NM, Edelstein K.Cognitive rehabilitation for cancer-related cognitive dysfunction: A systematic review Support Care Cancer 1–272019 [DOI] [PubMed] [Google Scholar]
  • 108.Campbell KL, Zadravec K, Bland KA, et al. The effect of exercise on cancer-related cognitive impairment and applications for physical therapy: Systematic review of randomized controlled trials Phys Ther 100523–5422020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Lower EE, Fleishman S, Cooper A, et al. Efficacy of dexmethylphenidate for the treatment of fatigue after cancer chemotherapy: A randomized clinical trial J Pain Symptom Manag 38650–6622009 [DOI] [PubMed] [Google Scholar]
  • 110.Mar Fan HG, Clemons M, Xu W, et al. A randomised, placebo-controlled, double-blind trial of the effects of d-methylphenidate on fatigue and cognitive dysfunction in women undergoing adjuvant chemotherapy for breast cancer Support Care Cancer 16577–5832008 [DOI] [PubMed] [Google Scholar]
  • 111.Escalante CP, Meyers C, Reuben JM, et al. A randomized, double-blind, 2-period, placebo-controlled crossover trial of a sustained-release methylphenidate in the treatment of fatigue in cancer patients Cancer J 208–142014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Kohli S, Fisher SG, Tra Y, et al. The effect of modafinil on cognitive function in breast cancer survivors Cancer 1152605–26162009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Lundorff LE, Jonsson BH, Sjogren P.Modafinil for attentional and psychomotor dysfunction in advanced cancer: A double-blind, randomised, cross-over trial Palliat Med 23731–7382009 [DOI] [PubMed] [Google Scholar]
  • 114.Rapp SR, Case LD, Peiffer A, et al. Donepezil for irradiated brain tumor survivors: A Phase III randomized placebo-controlled clinical trial J Clin Oncol 331653–16592015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Lawrence JA, Griffin L, Balcueva EP, 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 10176–1842016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Lawton MP, Brody EM.Assessment of older people: Self-maintaining and instrumental activities of daily living Gerontologist 9179–1861969 [PubMed] [Google Scholar]
  • 117.Horowitz TS, Suls J, Trevino M.A call for a neuroscience approach to cancer-related cognitive impairment Trends Neurosci 41493–4962018 [DOI] [PubMed] [Google Scholar]

Articles from Journal of Clinical Oncology are provided here courtesy of American Society of Clinical Oncology

RESOURCES