A narrative review of risk factors and interventions for cancer-related cognitive impairment
Review Article

A narrative review of risk factors and interventions for cancer-related cognitive impairment

Lu Bai1,2, Enyan Yu1,2

1Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China;2Institute of Cancer and Basic Medicine, Chinese Academy of Sciences, Beijing, China

Contributions: (I) Conception and design: L Bai; (II) Administrative support: E Yu; (III) Provision of study materials or patients: None; (IV) Collection and assembly of data: L Bai; (V) Data analysis and interpretation: L Bai; (VI) Manuscript writing: All authors; (VII) Final approval of manuscript: All authors.

Correspondence to: Enyan Yu. Department of Clinical Psychology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), East Banshan Rd, Hangzhou, China. Email: yuenyan@aliyun.com.

Abstract: Cancer-related cognitive impairment (CRCI) refers to a series of cognitive impairment symptoms associated with alternations in brain structure and function, caused by a non-central nervous system malignant tumor and its related treatment. CRCI may present as memory loss, impaired concentration, difficulty in multitasking and word retrieval, and reduced comprehension speed. CRCI has become one of the prevalent factors that compromise the quality of life for cancer survivors. Different treatments, including surgery, chemotherapy, radiotherapy, endocrine therapy, and targeted drugs, may contribute to CRCI. Meanwhile, patients’ factors, including emotional challenges and genetic makeup, also contribute to the development of CRCI. The condition can be treated with using stimulants methylphenidate and modafinil, metabolites of nicotine: cotinine, antidepressants of fluoxetine and fluvoxamine, dementia drug of donepezil, and antioxidants ZnSO4, n-acetyl cysteine, propofol, and Chinese herbal of silver leaf medicine. Psychotherapies, including meditation and relaxation, cognitive rehabilitation training, along with physical therapies, including aerobic exercise, resistance training, balance training, yoga, qigong, tai chi electroencephalogram biofeedback, and acupuncture, are also beneficial in alleviating cancer-related cognitive impairment symptoms. In recent years, researchers have focused on factors related to the condition and on the available interventions. However, most research was conducted independently, and no review has yet summarized the latest findings. This review details and discusses the status of related factors and potential treatments for CRCI. We also supply specific recommendations to facilitate future research and integration in this field.

Keywords: Cancer; cognitive impairment; risk factors; treatment


Submitted Jul 31, 2020. Accepted for publication Oct 27, 2020.

doi: 10.21037/atm-20-6443


Introduction

The survival rate of cancer patients is increasing despite the advancements in cancer diagnosis and treatment technology. This has resulted in an increased focus on the quality of life of cancer survivors over recent years. The incidence of cancer-related cognitive impairment (CRCI) ranges from 16% to 75% (1). The number varies because of different cancer type, treatment, duration of follow-up, type of study design and definition of cognitive impairment (2). According to a 2019 web-based survey, the prevalence of CRCI complaints experience is 8.23% for breast cancer, 9.10% for gynecological cancer patients, 8.33% for hematologic cancer patients, and 8.86% for other cancer types (3). Most studies suggest this reversible condition is attributed to chemotherapy and is evident within 18 to 36 months after treatment (4). However, some symptoms have been reported to be observed for up to 20 years (5). Although CRCI is usually mild to moderate in severity, it compromises the patient’s treatment adherence, reduces work productivity, and impacts the quality of daily activities (cooking, driving, etc.) (6). The American Cancer Society indicates that cognitive domains of memory, attention, executive function, and processing speed might be impaired, although research has also found impairment in working memory, new learning, visuospatial skills, and language. One research showed that low IQ score, as well as high fatigue predicted baseline CRCI. IQ score also predicted individual cognitive ability, which were also influenced by age, sex, cancer diagnosis, and an active intervention for hypertension (7). CRCI symptoms are categorized as follows: attention, memory, psychomotor speed, and executive function (8). Memory refers to forgetfulness, including the inability to recall the names of individuals. Attention pertains to insufficient concentration while completing a task, losing track of thoughts, and the inability to grasp the main concepts from written material. Psychomotor speed refers to the sluggish movement, accompanied by an unclever mind. Executive function refers to indecision and the inability to multitask.

When CRCI exits, the condition of the brain and the specific manifestations of the changes is reviewed. DTI Network analyses showed cognitive deficits was related with altered global and local brain networks. Chemotherapy drugs, which is related to hippocampal and frontal lobes dysfunction, and suppression of neurogenesis, cause memory loss, attention, working memory, and strategic learning deficits (9,10). Studies have reported that CRCI patients shows increased level of functional activation and connectivity of brain regions, which might be explained as the cognition have been interpreted as compensatory processes for treatment induced brain injury (11). Several studies have shown widespread reductions in grey matter density or volume and white matter microstructure when there is CRCI (12-15). Furthermore, mitochondrial dysfunction and dysregulation of cytokine activity might be another reason (16). Other possibilities include direct neurotoxicity with damage to neurons or nerve cells from chemotherapy (particularly fluorouracil and methotrexate, which are more likely to cross the blood-brain barrier) or induced hormonal changes from chemotherapy that lead to cognitive change (17). Chemotherapy drug, radiotherapy combined with immunotherapy caused increased levels pro-inflammatory cytokines, such as IL-1b, IL-6, TNF-a, IL-10, which impact on the frontal lobe function (18,19). Multiple studies have linked serum inflammatory markers with cognitive performance. In patients with breast cancer receiving chemotherapy, Williams et al. found an association between increasing concentrations of soluble tumor necrosis factor receptors I and II and decline in short-term visual memory (13). Additionally, CRCI also can be explained by oxidative damage and decreased hypothalamic-pituitary adrenal axis activity, and reduced brain vascularization and blood flow (20).

Neuropsychological assessments usually help to determine the various domains of cognitive in objective methods. According to the International Cognition and Cancer Task Force (ICCTF), the Hopkins Verbal Learning Test-Revised (HVLT-R), Trail Making Test (TMT), and the Controlled Oral Word Association (COWA) of the Multilingual Aphasia Examination are the typica scales to measure cancer patients’ cognitive deficits (21). The ICCTF suggests ≥2 test scores ≤1.5 standard deviations from the normative mean or one test score ≤2.0 standard deviations indicates cognitive impairment. Apart from that, Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment Scale (MOCA), Wechsler Adult Intelligence Scale, California Verbal Learning Test-II, Revised Rey-Osterrieth Complex Figure Test are commonly used in clinical department (22). However, no consensus has been reached for a specific tool to assess CRCI, and therefore a general, precise and effective CRCI scale is in need.

A strong interest in CRCI-related factors and interventions has developed in recent years, with reports linking the incidence of CRCI with the period after diagnosis, surgery, radiotherapy and endocrine therapy, and targeted drug therapy. Other areas of interest in CRCI have included interventions and the individual differences impact on CRCI. However, most of these reports do not collectively present the data in a combined manner. This is the first literature review that summarizes and discusses the latest findings of CRCI, with a focus on related risk factors and interventions. This paper will analyze and discuss the influence of different cancer treatments and individual factors on CRCI, along with the underlying mechanisms. Also, the treatment of CRCI is comprehensively expounded on drug treatment, psychotherapy, and physiotherapy, to supply directions for future research.

We present the following article in accordance with the Narrative Review reporting checklist (available at http://dx.doi.org/10.21037/atm-20-6443).


Methods

We conducted a literature search for papers published up to Oct 2019 on the risk factors and interventions aspects of CRCI using PubMed. The following medical subject heading terms were included for a MEDLINE search: “CRCI”, “cancer cognitive impairment”, “cancer cognitive defecit”, “chemobrain ”, “endocrine therapy cognitive”, “radiotherapy cogntive”, “targeted therapy cogntive”, “drug CRCI”, “psychotherapy CRCI”, “physical therapy CRCI”, “CRCI APOE”, “CRCI TMS”, “CRCI genetic factor”, “CRCI Cognitive rehabilitation”. Qualitative and quantitative data was extracted through interpretation of each article.

Risk factors for CRCI

Treatments of cancer

CRCI, also known as chemobrain and chemofog, mainly refers to cognitive impairment caused by chemotherapy, which may lead to nerve toxicity (23). Longitudinal studies, with neuropsychological assessments, have determined that the incidence of CRCI has been observed in approximately 40%, 75%, and 60% of patients before, during, and months to years after treatment, respectively (24). Findings in recent years support the notion that CRCI exists before chemotherapy. The studies indicated that the development of CRCI originated from cancer and surgery (25-27). Another study found that up to 33% of patients reported cognitive impairment before chemotherapy, including difficulty in expression and slowness in response (28). Also, malignant tumors and surgery were suggested to increase the level of cytokines, causing inflammation and tissue destruction, leading to cognitive decline (29).

A meta-analysis of 30 studies (838 participants) identified significant changes in executive function and verbal memory in patients that underwent chemotherapy when compared to the control group (30). In another meta-analysis, Falleti et al. reported small to moderate cognitive decline concerning memory, executive function, and attention tests in breast cancer patients, post-chemotherapy (31). Another meta-analysis of 7 studies found that over 300 breast cancer patients showed decreases in short-term memory, verbal and spatial language ability after undergoing chemotherapy (32). As previously mentioned, chemotherapy may cause cognitive decline in four areas: memory, attention, psychomotor speed, and executive function.

Furthermore, the dose of chemotherapy affects the severity of CRCI. One study showed 32% and 17% of patients who received high- and standard-dose chemotherapy, respectively, developed cognitive impairment compared to 9% of the control group in cancer patients post-treatment (33). Another investigation found that patients who received high-dose chemotherapy displayed more severe cognitive deterioration than those who were administered low-dose chemotherapy (34). Collins et al. evaluated the cognitive status of 60 patients with early-stage breast cancer before and upon completion of chemotherapy, along with after surgery. They demonstrated a linear deterioration of CRCI at each subsequent time point when compared to healthy controls (35). However, Schagen’s follow-up study revealed that the cognitive function of patients in both the high- and low-dose groups improved in the following four years, and considered the chemotherapy-induced change in cognitive function as transient (36,37). However, another study examined the effects of the chemotherapy combination CMF group [cyclophosphamide, methotrexate, 5-fluorouracil (5-fu), CMF] on cognitive function. A neuropsychological evaluation indicated that the CMF chemotherapy groups impaired cognitive functions on immediate and delayed memory, psychomotor speed, executive function, and processing speed, which were present for up to 20 years (5).

Another study examined the risk of dementia among 6,932 elderly breast cancer patients after they had received different chemotherapy drugs, including anthracyclines (doxorubicin, mitoxantrone, daorubicin, and epirubicin), CMF, taxane (polyene taxol, paclitaxel), and others. Interestingly, no significant difference was noted between different chemotherapy drugs and the risk of dementia diagnosis (38).

The increase of pro-inflammatory cytokines levels in cancer patients post-chemotherapy is regarded as the mechanism behind CRCI. Increased levels of serum interleukin-6 (IL-6), c-reactive protein (CRP), tumor necrosis factor α (TNF-α), interleukin-1 (IL-1), and other cytokines were associated with cognitive declines, including memory problems (39). A longitudinal study found that higher IL-6 and IL-1 concentrations were associated with increased severity in cognitive impairment, while higher IL-1 levels correlated with slower responses of patients (40). On the contrary, increased IL-4 was associated with faster response rates of breast cancer patients and fewer complaints of cognitive function, indicating it might serve as a protective factor for nerves in cancer patients (40). Also, the destruction of the blood-brain barrier, DNA oxidative stress damage, and shortening of the neutrophils are possible hypotheses of its mechanism (17).

Radiotherapy, endocrine targeted drug treatments may also contribute to the development of CRCI (41-45). The duration of radiotherapy-induced CRCI in cancer of the non-central nervous system has been observed at months to years after treatment (46-48). However, its effect is not as severe as that caused by chemotherapy (27). After seven months of breast-conserving therapy with adjuvant breast radiotherapy, women with breast cancer displayed lower scores on the short-term verbal memory and delayed recall measures, assessed by the Wechsler Memory Scale compared to those without radiotherapy. However, this difference disappeared three years after radiotherapy (49). The short-term memory impairment after radiotherapy is estimated to correlate significantly with elevated plasma IL-6 levels (50). This investigation revealed that memory levels recovered to baseline three years after radiotherapy, along with decreased plasma interleukin IL-6 levels. IL-6 was potentially regarded as the mechanism behind CRCI induced by radiotherapy (51). A longitudinal study of breast-cancer patients demonstrated that immediate and delayed memory cognitive functions improved in patients who received adjuvant breast radiotherapy and those who did not receive radiotherapy at 18 and 36 months after diagnosis (52). In a cross-sectional study, the cognitive abilities of testicular cancer patients were assessed using neuropsychological tests three years after radiation. No significant difference in average scores was observed between patients who underwent surgery alone, and those received adjuvant radiation (53). In conclusion, radiotherapy causes a transient decline in memory function, related to IL-6 levels.

Endocrine therapy using antagonistic estrogen is commonly used to treat breast cancer patients. de Ruiter et al. postulated that endocrine therapy is associated with cognitive impairment in cancer patients (54). Tamoxifen, a hormone therapy combined with chemotherapy, prolonged visual, and verbal memory impairment by one year when compared to chemotherapy alone (55). The combination of tamoxifen and chemotherapy significantly reduced memory, speech fluency, visuospatial function, and processing speed than chemotherapy alone (56). A study with strict control of variables found that patients with androgen deprivation therapy mainly affected the spatial memory and language memory (57).

Endocrine therapy primarily compromises using estrogen receptor modulators and/or aromatase inhibitors. Aromatase inhibitors block the synthesis of estradiol, depriving the brain of its ability to regulate estradiol, resulting in decreased neuroplasticity and impaired cognitive function. Clinical studies have shown that selective estrogen receptor modulator therapy has adverse effects on cognitive function, particularly in elderly breast cancer patients (43). Another study found that testosterone and estradiol levels decreased significantly in prostate cancer patients after treatment with leuproverine for three months, and the related spatial memory function was significantly impaired (58). A preclinical model is used to assess the effects of endocrine therapy on androgen-blocking of the brain. It demonstrated adverse effects in the hippocampus, associated with spatial ability and the ability to memorize new events (59,60). In summary, endocrine therapy has been shown to impair memory function, including spatial and verbal memory, along with psychomotor speed function. The mechanism of impairment was linked to changes in estradiol, testosterone, and the hippocampus.

In recent years, targeted drugs have been recognized as novel cancer treatments (61). Studies have found sunitinib aided in the reversal of cognitive impairment, including memory loss and word retrieval difficulty (62). A longitudinal study found that 31% of renal cancer patients treated with antiangiogenic tyrosine kinase inhibitors displayed a decline in cognitive function compared to baseline performance. This was more pronounced with executive function and episodic memory, suggesting that targeted drugs may elicit a direct neurotoxic effect (63). However, the mechanism behind targeted drug-induced CRCI is still unclear. Investigations have established that vascular endothelial growth factor (VEGF) plays a vital role in this process, and VEGF inhibitors may affect cognitive function by acting on neurogenesis, brain blood flow and the regulation of long-term potential (64,65).

Additionally, immune dysregulation and cytokine release may also be underlying mechanisms (66). Interestingly, everolimus has been found to delay aging and reduce the risk of Alzheimer’s disease in preclinical trials (67). A study found that everolimus increased hippocampal neurogenesis and synaptogenesis to improve cognitive function and prevent cognitive decline (68). Different targeted drugs show various effects on cognitive function, with the underlying mechanism associated with changes in VEGF, immune function, and level of cytokines. The emergence of novel targeted drugs highlights the need for further investigations to elucidate their effects on CRCI development.

Individual factors

Anxiety, depression, fatigue, insomnia

Some biological and psychological variables, including anxiety, depression, and insomnia, may aid in showing the likelihood of cognitive impairment in some patients. Emotional problems, including depression and anxiety, induce cognitive impairment, with a significant and linear correlation observed between emotional problems and cognitive impairment, including attention deficiency (69,70). People with cancer are more likely to experience symptoms of depression and anxiety compared to healthy individuals (71,72). Studies have found an inverse correlation between affective problems and cognitive function (73). Anxiety and depression also show a synergistic effect on executive function in cancer patients. Individuals diagnosed with the combination of depression and anxiety disorders display poorer performance with tasks on executive function than individuals with depression or anxiety disorder alone (74). However, the correlation between the subjective and objective evaluation of cognitive function is weak. Additional studies have revealed a significant correlation between subjective assessments of cognitive function problems and affective problems (75,76).

Insomnia has been shown to contribute to the severity of cognitive impairment in cancer patients (77,78). A study analyzing the relationship between insomnia disorders and cognitive function in breast cancer patients found patients with mild, moderate, and severe insomnia scored lower on cognitive assessments when compared with cancer patients without sleeping disorders. Additionally, a direct correlation was noted between insomnia severity and cognitive impairment (78). These findings highlighted the need for systematic evaluation of areas other than cognition, including emotional states and sleep quality.

Genes

Genetic factors, apolipoprotein E (APOE), catechol-O-methyltransferase (COMT), and brain-derived neurotrophic factor (BDNF), increase the risk of CRCI. The subtype of apolipoprotein E (APOE), APOE 4, is the leading risk factor for Alzheimer’s disease (79). A systematic review revealed three longitude studies and one cross-sectional study, which indicated the APOE 4 alleles was a risk factor for CRCI (80-83). The study found patients with the ApoE 4 alleles were more susceptible to neurotoxicity from chemotherapy than those without this gene, displaying significantly lower scores on language learning and memory, visual memory, spatial memory, and executive function (17,82). Another investigation revealed that testicular cancer patients with APOE 4 were found to be less cognitively competent after chemotherapy than those without this gene. There was no association noted established between APOE 4 and structural brain changes (83). While another six studies reported a correlation between APOE 4 and brain changes, these were reported to be non-significant (84-89).

Other related genes include COMT, DNA repair genes, oxidative stress genes, genes associated with breast cancer phenotypes, and GNB3 genes. COMT regulate dopamine levels in the frontal cortex, which is mediated executive and memory functions. A study found COMT gene polymorphisms were related with higher risk of CRCI in breast cancer survivors after chemotherapy (87). Research has found that patients carry e BDNF Met allele are at lower risk of CRCI, especially in terms of speech fluency and multitasking (90). Other genetic polymorphisms, such as those related to the folic acid pathway, estrogen receptor alpha genetic polymorphism, and DNA methyltransferase 1 polymorphism are also related to cognitive function (91,92). CRCI was not found to be associated with the genes coding IL-6, TNF, IL-1β, and brain-derived neurotrophic factor (BDNF). Except for APOE 4, genetic risk factors were not widely investigated (93). In general, the evidence is limited for possible genetic risk factors for CRCI, but we can reach a consensus that genetic factors are associated with cognitive impairment in cancer patients after treatment.

Risk factors for children

Some might wonder whether risk factors for children is different from that for adults. Risk factors for adult CRCI include cognitive reserve, age, genetic factors, and ethnicity, while risk factors for children include genetic factors, female sex, younger age at diagnosis, chemotherapy dose, and both dose and field size for radiation. Studies have shown 25% and 33% of children have cognitive impairment due to the chemotherapy (94). Boys shows better performance than girls in the aspect of cognitive function (95), and children who have the chemotherapy at younger ages have the worse performance (96). Research has shown that higher dose of chemotherapy, the severer cognitive deficits when the child grow up. One study on the survivors of childhood acute lymphoblastic leukemia showed worsened deficits in attention, as well as executive function and processing speed that higher doses of methotrexate (97).

CRCI interventions

Drug therapy

CRCI interventions have always been a topic of interest in researchers. Some believe stimulants, including methylphenidate and modafinil, play a significant role in treating CRCI. In 2019, a systematic review of six randomized controlled trials was conducted, where methylphenidate was administered in 244 cases across three trials while 146 cases received modafinil in the remaining three studies. The effects of their treatments on cognitive function and quality of life were assessed in all participants. Improvements in the cognitive function were observed in two studies using modafinil and one using methylphenidate. The underlying mechanism behind stimulants refers to neural signal processing by increasing synaptic concentrations of dopamine and norepinephrine in the brain’s prefrontal cortex (98-100).

Cotinine is the primary metabolite of nicotine. A study that examined the effects of cotinine on cognition and depression-like behavior in rats treated with CMF suggested it may contribute to CRCI recovery. The investigation also revealed that cotinine could alleviate the cognitive function side effects caused by chemotherapy (101).

Antidepressants have been shown to contribute to the treatment of CRCI. Studies have demonstrated that fluoxetine ameliorated memory impairment is caused by methotrexate chemotherapy and reversed neurogenic disorders in mice (102,103). The study revealed that 5-fu, a widely used chemotherapy drug, significantly reduced the number of proliferating cells in the dentate gyrus. The administration of fluoxetine, in combination with 5-fu, prevented the reduction of the number of proliferating cells in the sub-granular area of the dentate gyrus. Additionally, recipients of the combination therapy outperformed those who received monotherapy concerning hippocampal-dependent spatial working memory tests. These studies found that 5-fu exerted a negative effect on both cell proliferation and hippocampal-dependent working memory, while fluoxetine contributed to reversing these defects (104). A study on fluvoxamine found fluvoxamine ameliorated the taxol-induced neurotoxicity by partially inducing sigma-1 receptors, providing new evidence for the role of antidepressants in improving the chemical brain (105).

Donepezil, an acetylcholinesterase inhibitor used to treat Alzheimer’s disease, is regarded as a neurotransmitter modulator and has received considerable attention from CRCI investigators. Animal model studies and clinical trials confirmed donepezil was effective in improving cognitive function in cancer patients (106-108). In one study, breast cancer patients with CRCI symptoms were administered donepezil for 24 weeks, at doses of 5 mg daily for the first four weeks, which increased to 10 mg daily for the subsequent 20 weeks, post-chemotherapy. The donepezil group showed a significant improvement in memory compared with the placebo group, suggesting a considerable effect on the hippocampal memory system (108). Another study found donepezil improved hippocampal-dependent memories, including spatial memory (106).

The central oxidative stress response is another suspected mechanism hypothesized for chemotherapy-induced CRCI (17). Studies have shown that antioxidant therapy before and during chemotherapy can prevent oxidative stress and cognitive dysfunction caused by chemotherapy. Treatment with the antioxidant zinc sulfate (ZnSO4) before chemotherapy can prevent short-term memory impairment caused by carmustine. Further, carmustine caused hippocampal cell death and inflammation, which were also reduced in rats treated with ZnSO4 (109). Konat et al. studied the effects of cyclophosphamide and adriamycin on memory in rats. The investigation revealed that the combination of cyclophosphamide and doxorubicin impaired memory function. However, no short-term memory impairment was found when the rats were administered the antioxidant n-acetylcysteine during chemotherapy (110).

Animal studies have shown that acute cisplatin treatment reduces dendritic branching and spinal density, along with the induction of mitochondrial degradation. When compared with the control group, rats receiving long-term cisplatin showed poor performance in conditional situational fear, situational object discrimination, and new object recognition tasks. N-acetylcysteine therapy can prevent free radical production, improve apoptotic cell death and loss of dendritic spines, as well as partially reverse cisplatin-induced cognitive impairment (111).

A series of studies found that cognitive impairment in cancer patients was associated with persistent neuroinflammation (112,113). A study on mice reported that a lower-dose of the anesthetic propofol slowed cognitive impairment caused by the chemotherapeutic drug cisplatin (114).

Propofol has also been reported to play a neuroprotective role by inhibiting inflammatory activity. It reduced oxidative stress and prevented an increase in neuronal mitochondrial swelling in animal models of nerve injuries, including ischemic stroke and traumatic brain injury (115). Previous studies have shown that cisplatin-induced cognitive dysfunction is associated with damage to the synaptic mitochondria of the brain and abnormalities in the mitochondria of the peripheral nervous system (113). Therefore, propofol may reduce cisplatin-induced memory impairment by inhibiting neuroinflammation, reducing oxidation, and preventing mitochondrial damage.

Silver leaf, a Chinese herbal medicine, is used to prevent cognitive decline in elderly individuals. It has been found to have a curative effect on cognitive function (116). A study found the ginkgo Biloba extract 761, a standardized extract compound of ginkgo Biloba, has similarities to antioxidants regarding function. It protects the brain from oxidative stress damage and apoptosis, regulates cerebral blood flow, and interacts with catecholamine to effectively improve executive ability, selective attention, and memory function for verbal and non-verbal material in cancer survivors (117).

In conclusion, the stimulants (methylphenidate and modafinil), metabolites of nicotine(cotinine), antidepressants (fluoxetine and fluvoxamine), dementia drugs (donepezil), antioxidants (ZnSO4, n-acetyl cysteine, propofol and Chinese herbal of the silver leaf) have been shown to contribute to the prevention of cognitive impairment of patients with cancer.

Psychotherapy

In 2019, Fernandes et al. conducted a systematic review and revealed the beneficial effects of rehabilitation training in cognitive function improvement, including memory, attention, executive function, and processing speed (118). Two types of cognitive rehabilitation training have been identified: computer and strategy training. Computer training refers to the performance of cognitive training tasks using a computer. Strategy training refers to cognitive behavior training and behavior-oriented practice. The training, which aims to rehabilitate impaired cognitive and compensation functions through psychological education and training, consists of 20 minutes to 2-hour sessions over 4–12 weeks in the group and individual settings (119,120). Most studies applying cognitive rehabilitation after breast cancer indicated that profound improvement in cognitive tests and quality of life. The areas consist of executive functioning, working memory, attention, memory, processing speed, and visual-spatial skills were found to be improved due to the intervention (121,122). One study selected quantitative EEG biomarker and found that cognitive rehabilitation contributed to increased global brain activity. In the view of participants, the cognitive rehabilitation intervention was helpful to compensate for memory problems (123). Additionally, this intervention is also helpful in treating emotional problems, such as the anxiety and depression (124).

Meditation, including breathing relaxation, also contributes to recovery from CRCI. Studies have shown that meditation relieves stress and pain, reduces fatigue, slows cognitive decline, and improves the quality of life (22). Some meditation relaxation exercises reduce stress, which directly or indirectly strengthens the immune system and regulates cytokines (125). Studies have confirmed mindfulness-based stress-reduction courses aid in the cognitive rehabilitation of cancer patients (126,127).

Physical therapy

Exercise therapies, including aerobic exercise, resistance training, balance training, yoga, qigong, tai chi, are believed to be effective in ameliorating CRCI and improving patients’ quality of life (128-132). Aerobic exercise leads to an increase in the expression of neurotrophic and nerve protection factors, including BDNF and VEGF. An increased level of these factors is related explicitly to the hippocampus neurogenesis, which is crucial for spatial memory. Additionally, many studies have shown that exercise induces neurogenesis, increases the hippocampus volume, and improves hippocampus-dependent cognitive function (133-135). In a study of sedentary breast-cancer survivors, the group of individuals who participated in a 12-week exercise program showed improved processing speed and reduced cognitive impaired symptoms within two years of diagnosis compared to the control group (136). Recently, treatment programs use high-intensity endurance training with set intervals and assign an exercise physiotherapist to each breast-cancer patient to closely monitor the amount of daily exercise. Studies have suggested that these types of programs significantly improve cognitive ability by affecting markers, including TNF-α, IL-6, IL-1, CRP, BDNF, and VEGF (137).

Electroencephalogram (EEG) biofeedback, otherwise known as neural feedback, have also been incorporated into treatment programs for breast cancer patients at 6 to 60 months post-chemotherapy. This program consisted of a total of 20 sessions over ten weeks and was found to mitigate fatigue, sleep disorders, and emotional problems. Also, improvements were shown in cognitive function performance. Furthermore, all participants completed the program, which suggests the EEG biofeedback treatment is a safe and effective form of CRCI therapy (138).

A prospective cohort study of post-chemotherapy cancer patients found patients treated with acupuncture scores higher on a neurocognitive assessment and outperformed the control group concerning cognitive function after adjusting for age, menopause, cancer stage, and a dose of a chemotherapy regimen. Magnetic resonance spectroscopy analysis revealed that the relative concentration of n-acetyl aspartic acid in the left hippocampus, an area associated with impaired working memory, was significantly reduced in the group treated with acupuncture. The findings of the study led to the proposal that acupuncture improved CRCI through reducing demyelination and neuronal activity in the white matter of the hippocampus (139). In summary, exercise, EEG biofeedback therapy, and acupuncture can improve memory, processing speed, and other cognitive functions.

Currently, drug treatment is regarded as a rapid, effective, and resource-efficient way to treat CRCI. However, the side effects associated with drug therapy still poses to be a disadvantage. Psychological therapy and physical therapy are less likely to have side effects but display less efficacy compared to drug therapy (125).


Summary and prospect

CRCI is a significant problem for cancer survivors and originates from cancer itself, surgery, chemotherapy, radiotherapy, endocrine treatments, and targeted drug treatments. Individual factors, including genetic makeup and emotional state, also contribute to CRCI development.

Specifically, chemotherapy is associated with memory impairment, attention, psychomotor speed, and executive functions. The severity of cognitive impairment was directly correlated with the chemotherapy dose. However, the long-term effects of different chemotherapy methods on cognitive function did not differ significantly different. Radiotherapy, endocrine therapy, and targeted drugs can also cause cognitive decline. However, side effects are often temporary for radiotherapy, controversial for chemotherapy, and unclear for endocrine and targeted drug therapy, which all require further examination.

Depression and anxiety problems often accompany cancer, which may cause cognitive disorders. Further longitudinal studies are needed to confirm the mechanism behind the effect of antidepressants on CRCI and the mediating role of emotional problems. Other antidepressants, including vortioxetine, escitalopram, and Zoloft, along with venlafaxine and duloxetine, have not yet been examined and require further investigations.

At present, the relationship between ApoE 4 and CRCI is still unclear. Furthermore, the contribution of specific genes to CRCI, including COMT, DNA repair genes, are not widely investigated. Current literature does not supply robust conclusions about the role of genetic factors in CRCI development. This highlights the requirement for a comprehensive genetic study across a large population to investigate the effect of genetic variations and its contribution to CRCI development. The underlying mechanism is still controversial and requires further research with more robust and systematic experimental designs across large patient populations. The beneficial effect of anti-dementia drugs for CRCI, donepezil, has been confirmed by animal experiments and clinical trials. Moreover, memantine, an n-methyl-d-aspartic acid receptor antagonist, is an anti-dementia drug that improves glutamate neurotransmission. It is useful for the treatment of many dementia types, including Alzheimer’s disease and Lewy body dementia. Currently, memantine is found to be effective in improving cognitive function in patients with central-nervous-system cancer. However, clinical studies investigating its efficacy in non-CNS cancer patients have not been performed, highlighting a need for this area to be studied (140).

Additionally, there is no consensus on the relationship between CRCI and dementia. A large retrospective study involving 18,360 patients found that CRCI increased the risk of Alzheimer’s disease (141). However, a recent study found a negative association between Alzheimer’s disease and cancer. This study reported a lower risk of developing Alzheimer’s disease in cancer patients compared to healthy people. Also, Alzheimer’s patients displayed a lower risk of developing cancer, which might be explained by cancer prevents neurodegeneration (142). In general, the relationships among ApoE 4, anti-dementia drugs, dementia, and CRCI should be confirmed in future studies. Additionally, brain function modifications need to be examined to explore the mechanism behind the changes.

Preclinical studies suggest that antioxidants improved cognitive impairment. The effects of antioxidants ZnSO4, n-acetylcysteine, and propofol on CRCI need to be confirmed by clinical trials with longitudinal design across large patient samples.

Traditional Chinese medicine ginkgo and physical therapy acupuncture may help treat CRCI in cancer patients. However, the sample sizes of these trials were small. Also, the number of prospective studies was limiting. A prospective clinical design with large patient samples is needed, along with the identification of underlying mechanisms requiring future studies. Furthermore, the anti-dementia drug, fufanghaishe, has been proven to be effective in treating Alzheimer’s disease (143). In this regard, the efficacy and pathological mechanism of fufanghaishe may also be a potential avenue of future research in treating CRCI.

Psychological therapy, including cognitive rehabilitation training and meditation, can alleviate CRCI. The effects of other psychotherapy methods, including music therapy and painting, on CRCI requires further investigations.

Exercise, EEG biofeedback therapy, and acupuncture were found to have a positive effect on CRCI. Other physical therapy methods should also be examined.

Repeated transcranial magnetic stimulation (rTMS) is applied by passing unattenuated repeated stimulation of a magnetic signal and stimulates the nerves in the cortex through the skull. Studies have found that rTMS contributes to improving work executive function and objective memory function in people diagnosed with depression. High-frequency rTMS improves working memory, cognitive flexibility, and verbal fluency in the same patient population (144). At present, no research of rTMS has been conducted on the cognitive function of cancer patients, which still has yet to be verified by subsequent studies.


Acknowledgments

Funding: This pilot study was funded by the National Natural Science Foundation of China (No. 81771158).


Footnote

Reporting Checklist: The authors have completed the Narrative Review reporting checklist. Available at http://dx.doi.org/10.21037/atm-20-6443

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at http://dx.doi.org/10.21037/atm-20-6443). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work and are responsible for addressing questions related to the accuracy or integrity of any part of the work until they are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Argyriou AAMDP, Assimakopoulos KMDP, Iconomou GP, et al. Either Called “Chemobrain” or “Chemofog,” the Long-Term Chemotherapy-Induced Cognitive Decline in Cancer Survivors Is Real. J Pain Symptom Manage 2011;41:126-39. [Crossref] [PubMed]
  2. Koppelmans V, Breteler MM, Boogerd W, et al. Neuropsychological performance in survivors of breast cancer more than 20 years after adjuvant chemotherapy. J Clin Oncol 2012;30:1080-6. [Crossref] [PubMed]
  3. 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 2019;8:2654-63. [Crossref] [PubMed]
  4. Dietrich J, Prust M, Kaiser J. Chemotherapy, cognitive impairment and hippocampal toxicity. Neuroscience 2015;309:224-32. [Crossref] [PubMed]
  5. Koppelmans V, Breteler MMB, Boogerd W, et al. Neuropsychological Performance in Survivors of Breast Cancer More Than 20 Years After Adjuvant Chemotherapy. J Clin Oncol 2012;30:1080-6. [Crossref] [PubMed]
  6. Li J, Pang H, Sun Z, et al. Health status of middle-aged and older cancer survivors: a nationwide cross-sectional study from the China Health and Retirement Longitudinal Study (CHARLS). Ann Transl Med 2020;8:183. [Crossref] [PubMed]
  7. Lycke M, Pottel L, Pottel H, et al. Predictors of baseline cancer-related cognitive impairment in cancer patients scheduled for a curative treatment. Psychooncology 2017;26:632-9. [Crossref] [PubMed]
  8. Hodgson KD, Hutchinson AD, Wilson CJ, et al. A meta-analysis of the effects of chemotherapy on cognition in patients with cancer. Cancer Treat Rev 2013;39:297-304. [Crossref] [PubMed]
  9. Dubois M, Lapinte N, Villier V, et al. Chemotherapy-induced long-term alteration of executive functions and hippocampal cell proliferation: role of glucose as adjuvant. Neuropharmacology 2014;79:234-48. [Crossref] [PubMed]
  10. Winocur G, Johnston I, Castel H. Chemotherapy and cognition: International cognition and cancer task force recommendations for harmonising preclinical research. Cancer Treat Rev 2018;69:72-83. [Crossref] [PubMed]
  11. Apple AC, Schroeder MP, Ryals AJ, et al. Hippocampal functional connectivity is related to self-reported cognitive concerns in breast cancer patients undergoing adjuvant therapy. Neuroimage Clin 2018;20:110-8. [Crossref] [PubMed]
  12. Deprez S, Kesler SR, Saykin AJ, et al. International Cognition and Cancer Task Force Recommendations for Neuroimaging Methods in the Study of Cognitive Impairment in Non-CNS Cancer Patients. J Natl Cancer Inst 2018;110:223-31. [Crossref] [PubMed]
  13. Williams AM, Shah R, Shayne M, et al. Associations between inflammatory markers and cognitive function in breast cancer patients receiving chemotherapy. J Neuroimmunol 2018;314:17-23. [Crossref] [PubMed]
  14. Koppelmans V, de Ruiter MB, van der Lijn F, et al. Global and focal brain volume in long-term breast cancer survivors exposed to adjuvant chemotherapy. Breast Cancer Res Treat 2012;132:1099-106. [Crossref] [PubMed]
  15. Deprez S, Amant F, Smeets A, et al. Longitudinal assessment of chemotherapy-induced structural changes in cerebral white matter and its correlation with impaired cognitive functioning. J Clin Oncol 2012;30:274-81. [Crossref] [PubMed]
  16. Lomeli N, Di K, Czerniawski J, et al. Cisplatin-induced mitochondrial dysfunction is associated with impaired cognitive function in rats. Free Radic Biol Med 2017;102:274-86. [Crossref] [PubMed]
  17. Ahles TA, Saykin AJ. Candidate mechanisms for chemotherapy-induced cognitive changes. Nat Rev Cancer 2007;7:192-201. [Crossref] [PubMed]
  18. Pomykala KL, Ganz PA, Bower JE, et al. The association between pro-inflammatory cytokines, regional cerebral metabolism, and cognitive complaints following adjuvant chemotherapy for breast cancer. Brain Imaging Behav 2013;7:511-23. [Crossref] [PubMed]
  19. McGinnis GJ, Friedman D, Young KH, et al. Neuroinflammatory and cognitive consequences of combined radiation and immunotherapy in a novel preclinical model. Oncotarget 2017;8:9155-73. [Crossref] [PubMed]
  20. Seigers R, Fardell JE. Neurobiological basis of chemotherapy-induced cognitive impairment: a review of rodent research. Neurosci Biobehav Rev 2011;35:729-41. [Crossref] [PubMed]
  21. 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 2011;12:703-8. [Crossref] [PubMed]
  22. Craig CD, Monk BJ, Farley JH, et al. Cognitive impairment in gynecologic cancers: a systematic review of current approaches to diagnosis and treatment. Support Care Cancer 2014;22:279-87. [Crossref] [PubMed]
  23. Ouimet LA, Stewart A, Collins B, et al. Measuring neuropsychological change following breast cancer treatment: an analysis of statistical models. J Clin Exp Neuropsychol 2009;31:73-89. [Crossref] [PubMed]
  24. Jansen CE, Cooper BA, Dodd MJ, et al. A prospective longitudinal study of chemotherapy-induced cognitive changes in breast cancer patients. Support Care Cancer 2011;19:1647-56. [Crossref] [PubMed]
  25. Scherling C, Collins B, Mackenzie J, et al. Pre-chemotherapy differences in visuospatial working memory in breast cancer patients compared to controls: an FMRI study. Front Hum Neurosci 2011;5:122. [Crossref] [PubMed]
  26. Depboylu B.. Treatment and patient related quality of life issues in elderly and very elderly breast cancer patients. Transl Cancer Res 2020;9:S146-53. [Crossref]
  27. Kohli S, Griggs JJ, Roscoe JA, et al. Self-reported cognitive impairment in patients with cancer. J Oncol Pract 2007;3:54-9. [Crossref] [PubMed]
  28. Ahles TA, Ahles TA, Saykin AJ, et al. Cognitive function in breast cancer patients prior to adjuvant treatment. Breast Cancer Res Treat 2008;110:143-52. [Crossref] [PubMed]
  29. Monk TG, Weldon BC, Garvan CW, et al. Predictors of cognitive dysfunction after major noncardiac surgery. Anesthesiology 2008;108:18-30. [Crossref] [PubMed]
  30. Anderson-Hanley C, Sherman ML, Riggs R, et al. Neuropsychological effects of treatments for adults with cancer: a meta-analysis and review of the literature. J Int Neuropsychol Soc 2003;9:967-82. [Crossref] [PubMed]
  31. Falleti MG, Sanfilippo A, Maruff P, et al. The nature and severity of cognitive impairment associated with adjuvant chemotherapy in women with breast cancer: a meta-analysis of the current literature. Brain Cogn 2005;59:60-70. [Crossref] [PubMed]
  32. Stewart A, Bielajew C, Collins B, et al. A meta-analysis of the neuropsychological effects of adjuvant chemotherapy treatment in women treated for breast cancer. Clin Neuropsychol 2006;20:76-89. [Crossref] [PubMed]
  33. van Dam FS, Schagen SB, Muller MJ, et al. Impairment of cognitive function in women receiving adjuvant treatment for high-risk breast cancer: high-dose versus standard-dose chemotherapy. J Natl Cancer Inst 1998;90:210-8. [Crossref] [PubMed]
  34. Schagen SB, van Dam FS, Muller MJ, et al. Cognitive deficits after postoperative adjuvant chemotherapy for breast carcinoma. Cancer 1999;85:640-50. [Crossref] [PubMed]
  35. Collins B, MacKenzie J, Tasca GA, et al. Cognitive effects of chemotherapy in breast cancer patients: a dose-response study. Psychooncology 2013;22:1517-27. [Crossref] [PubMed]
  36. Schagen SB, Muller MJ, Boogerd W, et al. Late effects of adjuvant chemotherapy on cognitive function: a follow-up study in breast cancer patients. Ann Oncol 2002;13:1387-97. [Crossref] [PubMed]
  37. Schagen SB, Muller MJ, Boogerd W, et al. Change in cognitive function after chemotherapy: a prospective longitudinal study in breast cancer patients. J Natl Cancer Inst 2006;98:1742-5. [Crossref] [PubMed]
  38. Raji MA, Tamborello LP, Kuo YF, et al. Risk of subsequent dementia diagnoses does not vary by types of adjuvant chemotherapy in older women with breast cancer. Med Oncol 2009;26:452-9. [Crossref] [PubMed]
  39. Briones TL, Woods J. Dysregulation in myelination mediated by persistent neuroinflammation: Possible mechanisms in chemotherapy-related cognitive impairment. Brain Behav Immun 2014;35:23-32. [Crossref] [PubMed]
  40. Cheung YT, Ng T, Shwe M, et al. Association of proinflammatory cytokines and chemotherapy-associated cognitive impairment in breast cancer patients: A multi-centered, prospective, cohort study. Ann Oncol 2015;26:1446-51. [Crossref] [PubMed]
  41. Ernst T, Chang L, Cooray D, et al. The effects of tamoxifen and estrogen on brain metabolism in elderly women. J Natl Cancer Inst 2002;94:592-7. [Crossref] [PubMed]
  42. Castellon SA, Ganz PA, Bower JE, et al. Neurocognitive performance in breast cancer survivors exposed to adjuvant chemotherapy and tamoxifen. J Clin Exp Neuropsychol 2004;26:955-69. [Crossref] [PubMed]
  43. Schilder CM, Seynaeve C, Beex LV, et al. Effects of tamoxifen and exemestane on cognitive functioning of postmenopausal patients with breast cancer: results from the neuropsychological side study of the tamoxifen and exemestane adjuvant multinational trial. J Clin Oncol 2010;28:1294-300. [Crossref] [PubMed]
  44. Jenkins V, Shilling V, Fallowfield L, et al. Does hormone therapy for the treatment of breast cancer have a detrimental effect on memory and cognition? A pilot study. Psychooncology 2004;13:61-6. [Crossref] [PubMed]
  45. Ahles TA, Saykin AJ, McDonald BC, et al. Longitudinal assessment of cognitive changes associated with adjuvant treatment for breast cancer: impact of age and cognitive reserve. J Clin Oncol 2010;28:4434-40. [Crossref] [PubMed]
  46. Janaki MG, Kadam AR, Mukesh S, et al. Magnitude of fatigue in cancer patients receiving radiotherapy and its short term effect on quality of life. J Cancer Res Ther 2010;6:22-6. [Crossref] [PubMed]
  47. Noal S, Levy C, Hardouin A, et al. One-year longitudinal study of fatigue, cognitive functions, and quality of life after adjuvant radiotherapy for breast cancer. Int J Radiat Oncol Biol Phys 2011;81:795-803. [Crossref] [PubMed]
  48. Phillips KM, Jim HS, Small BJ, et al. Cognitive functioning after cancer treatment: a 3-year longitudinal comparison of breast cancer survivors treated with chemotherapy or radiation and noncancer controls. Cancer 2012;118:1925-32. [Crossref] [PubMed]
  49. Omura K, Sugishita M. Simultaneous Confirmatory Factor Analysis of the Wechsler Memory Scale - Revised for Two Standardization Samples: A Comparison of Groups from Japan and the United States. J Clin Exp Neuropsychol 2004;26:645-52. [PubMed]
  50. Shibayama O, Yoshiuchi K, Inagaki M, et al. Association between adjuvant regional radiotherapy and cognitive function in breast cancer patients treated with conservation therapy. Cancer Med 2014;3:702-9. [Crossref] [PubMed]
  51. Nguyen CM, Yamada TH, Beglinger LJ, et al. Cognitive features 10 or more years after successful breast cancer survival: comparisons across types of cancer interventions: Cognitive features of older breast cancer survivors. Psycho-Oncology 2013;22:862-8. [Crossref] [PubMed]
  52. Zheng Y, Luo J, Bao P, et al. Long-term cognitive function change among breast cancer survivors. Breast Cancer Res Treat 2014;146:599-609. [Crossref] [PubMed]
  53. Schagen SB, Boogerd W, Muller MJ, et al. Cognitive complaints and cognitive impairment following BEP chemotherapy in patients with testicular cancer. Acta Oncol 2008;47:63-70. [Crossref] [PubMed]
  54. de Ruiter MB, Reneman L, Boogerd W, et al. Late effects of high-dose adjuvant chemotherapy on white and gray matter in breast cancer survivors: converging results from multimodal magnetic resonance imaging. Hum Brain Mapp 2012;33:2971-83. [Crossref] [PubMed]
  55. Bender CM, Sereika SM, Berga SL, et al. Cognitive impairment associated with adjuvant therapy in breast cancer. Psychooncology 2006;15:422-30. [Crossref] [PubMed]
  56. Palmer JL, Trotter T, Joy AA, et al. Cognitive effects of Tamoxifen in pre-menopausal women with breast cancer compared to healthy controls. J Cancer Surviv 2008;2:275-82. [Crossref] [PubMed]
  57. Jamadar RJ, Winters MJ, Maki PM. Cognitive changes associated with ADT: a review of the literature. Asian J Androl 2012;14:232-8. [Crossref] [PubMed]
  58. Jenkins VA, Bloomfield DJ, Shilling VM, et al. Does neoadjuvant hormone therapy for early prostate cancer affect cognition? Results from a pilot study. BJU Int 2005;96:48-53. [Crossref] [PubMed]
  59. Alibhai SM, Breunis H, Timilshina N, et al. Impact of androgen-deprivation therapy on cognitive function in men with nonmetastatic prostate cancer. J Clin Oncol 2010;28:5030-7. [Crossref] [PubMed]
  60. Janowsky JS. The role of androgens in cognition and brain aging in men. Neuroscience 2006;138:1015-20. [Crossref] [PubMed]
  61. Metzger-Filho O, Moulin C, Awada A. Molecular targeted therapy in prevalent tumors: learning from the past and future perspectives. Curr Clin Pharmacol 2010;5:166-77. [Crossref] [PubMed]
  62. Patel P, Srinivas S. Toxicities of targeted agents in advanced renal cell carcinoma. Curr Clin Pharmacol 2011;6:181-8. [Crossref] [PubMed]
  63. Mulder SF, Bertens D, Desar IM, et al. Impairment of cognitive functioning during Sunitinib or Sorafenib treatment in cancer patients: a cross sectional study. BMC Cancer 2014;14:219. [Crossref] [PubMed]
  64. Licht T, Goshen I, Avital A, et al. Reversible modulations of neuronal plasticity by VEGF. Proc Natl Acad Sci U S A 2011;108:5081-6. [Crossref] [PubMed]
  65. Licht T, Keshet E. Delineating multiple functions of VEGF-A in the adult brain. Cell Mol Life Sci 2013;70:1727-37. [Crossref] [PubMed]
  66. Ng T, Cheung YT, Ng QS, et al. Vascular endothelial growth factor inhibitors and cognitive impairment: evidence and controversies. Expert Opin Drug Saf 2014;13:83-92. [Crossref] [PubMed]
  67. Wilkinson JE, Burmeister L, Brooks SV, et al. Rapamycin slows aging in mice. Aging Cell 2012;11:675-82. [Crossref] [PubMed]
  68. Russo E, Leo A, Crupi R, et al. Everolimus improves memory and learning while worsening depressive- and anxiety-like behavior in an animal model of depression. J Psychiatr Res 2016;78:1-10. [Crossref] [PubMed]
  69. Biringer E, Lundervold A, Stordal K, et al. Executive function improvement upon remission of recurrent unipolar depression. Eur Arch Psychiatry Clin Neurosci 2005;255:373-80. [Crossref] [PubMed]
  70. Schagen SB, van Dam FSAM, Muller MJ, et al. Cognitive deficits after postoperative adjuvant chemotherapy for breast carcinoma. Cancer 1999;85:640-50. [Crossref] [PubMed]
  71. Nordin K, Berglund G, Glimelius B, et al. Predicting anxiety and depression among cancer patients: a clinical model. Eur J Cancer 2001;37:376-84. [Crossref] [PubMed]
  72. Iconomou G, Mega V, Koutras A, et al. Prospective assessment of emotional distress, cognitive function, and quality of life in patients with cancer treated with chemotherapy. Cancer 2004;101:404-11. [Crossref] [PubMed]
  73. Biglia N, Bounous VE, Malabaila A, et al. Objective and self-reported cognitive dysfunction in breast cancer women treated with chemotherapy: a prospective study. Eur J Cancer Care (Engl) 2012;21:485-92. [Crossref] [PubMed]
  74. Snyder HR. Major depressive disorder is associated with broad impairments on neuropsychological measures of executive function: A meta-analysis and review. Psychological Bulletin 2013;139:81-132. [Crossref] [PubMed]
  75. Wefel JS, Schagen SB. Chemotherapy-related cognitive dysfunction. Curr Neurol Neurosci Rep 2012;12:267-75. [Crossref] [PubMed]
  76. O’Farrell E, MacKenzie J, Collins B. Clearing the Air: A Review of Our Current Understanding of “Chemo Fog”. Curr Oncol Rep 2013;15:260-9. [Crossref] [PubMed]
  77. Menning S, de Ruiter MB, Veltman DJ, et al. Multimodal MRI and cognitive function in patients with breast cancer prior to adjuvant treatment--the role of fatigue. Neuroimage Clin 2015;7:547-54. [Crossref] [PubMed]
  78. Liou KT, Ahles TA, Garland SN, et al. The Relationship Between Insomnia and Cognitive Impairment in Breast Cancer Survivors. JNCI Cancer Spectrum 2019;3:pkz041. [Crossref] [PubMed]
  79. Liu CC, Liu CC, Kanekiyo T, et al. Apolipoprotein E and Alzheimer disease: risk, mechanisms and therapy. Nat Rev Neurol 2013;9:106-18. [Crossref] [PubMed]
  80. Ahles TA, Li Y, McDonald BC, et al. Longitudinal assessment of cognitive changes associated with adjuvant treatment for breast cancer: the impact of APOE and smoking. Psychooncology 2014;23:1382-90. [Crossref] [PubMed]
  81. Koleck TA, Bender CM, Sereika SM, et al. Apolipoprotein E genotype and cognitive function in postmenopausal women with early-stage breast cancer. Oncol Nurs Forum 2014;41:E313-25. [Crossref] [PubMed]
  82. 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 2003;12:612-9. [Crossref] [PubMed]
  83. Amidi A, Agerbæk M, Wu LM, et al. Changes in cognitive functions and cerebral grey matter and their associations with inflammatory markers, endocrine markers, and APOE genotypes in testicular cancer patients undergoing treatment. Brain Imaging Behavior 2017;11:769-83. [Crossref] [PubMed]
  84. Mandelblatt JS, Stern RA, Luta G, et al. Cognitive impairment in older patients with breast cancer before systemic therapy: Is there an interaction between cancer and comorbidity? J Clin Oncol 2014;32:1909-18. [Crossref] [PubMed]
  85. Vardy JL, Stouten-Kemperman MM, Pond G, et al. A mechanistic cohort study evaluating cognitive impairment in women treated for breast cancer. Brain Imaging Behav 2019;13:15-26. [Crossref] [PubMed]
  86. Bender CM, Merriman JD, Sereika SM, et al. Trajectories of Cognitive Function and Associated Phenotypic and Genotypic Factors in Breast Cancer. Oncol Nurs Forum 2018;45:308-26. [Crossref] [PubMed]
  87. Cheng H, Li W, Gan C, et al. The COMT (rs165599) gene polymorphism contributes to chemotherapy-induced cognitive impairment in breast cancer patients. Am J Transl Res 2016;8:5087-97. [PubMed]
  88. Vardy JL, Dhillon HM, Pond GR, et al. Cognitive function in patients with colorectal cancer who do and do not receive chemotherapy: A prospective, longitudinal, controlled study. J Clin Oncol 2015;33:4085-92. [Crossref] [PubMed]
  89. Vardy J, Dhillon HM, Pond GR, et al. Cognitive function and fatigue after diagnosis of colorectal cancer. Ann Oncol 2014;25:2404-12. [Crossref] [PubMed]
  90. 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 2016;18:244-51. [Crossref] [PubMed]
  91. Kamdar KY, Krull KR, El-Zein RA, et al. Folate pathway polymorphisms predict deficits in attention and processing speed after childhood leukemia therapy. Pediatr Blood Cancer 2011;57:454-60. [Crossref] [PubMed]
  92. Terence NG, Chian MT, Angie Yeo HL, et al. DNA methyltransferase 1 (DNMT1) polymorphism and chemotherapy-associated cognitive impairment in early-stage breast cancer patients (ESBC): a prospective, longitudinal study. J Clin Oncol 2016;34:15.
  93. Buskbjerg CDR, Amidi A, Demontis D, et al. Genetic risk factors for cancer-related cognitive impairment: a systematic review. Acta Oncologica 2019;58:537-47. [Crossref] [PubMed]
  94. Mulhern RK, Wasserman AL, Fairclough D, et al. Memory function in disease-free survivors of childhood acute lymphocytic leukemia given CNS prophylaxis with or without 1,800 cGy cranial irradiation. J Clin Oncol 1988;6:315-20. [Crossref] [PubMed]
  95. Brown RT, Madan-Swain A, Walco GA, et al. Cognitive and academic late effects among children previously treated for acute lymphocytic leukemia receiving chemotherapy as CNS prophylaxis. J Pediatr Psychol 1998;23:333-40. [Crossref] [PubMed]
  96. Hill DE, Ciesielski KT, Sethre-Hofstad L, et al. Visual and verbal short-term memory deficits in childhood leukemia survivors after intrathecal chemotherapy. J Pediatr Psychol 1997;22:861-70. [Crossref] [PubMed]
  97. Espy KA, Moore IM, Kaufmann PM, et al. Chemotherapeutic CNS prophylaxis and neuropsychologic change in children with acute lymphoblastic leukemia: a prospective study. J Pediatr Psychol 2001;26:1-9. [Crossref] [PubMed]
  98. Berridge CW, Devilbiss DM. Psychostimulants as Cognitive Enhancers: The Prefrontal Cortex, Catecholamines, and Attention-Deficit/Hyperactivity Disorder. Biological Psychiatry 2011;69:e101-11. [Crossref] [PubMed]
  99. Volkow ND, Fowler JS, Wang G, et al. Mechanism of action of methylphenidate: Insights from PET imaging studies. J Atten Disord 2002;6:S31-43. [Crossref] [PubMed]
  100. Hannestad J, Gallezot JD, Planeta-Wilson B, et al. Clinically relevant doses of methylphenidate significantly occupy norepinephrine transporters in humans in vivo. Biol Psychiatry 2010;68:854-60. [Crossref] [PubMed]
  101. Iarkov A, Appunn D, Echeverria V. Post-treatment with cotinine improved memory and decreased depressive-like behavior after chemotherapy in rats. Cancer Chemother Pharmacol 2016;78:1033-9. [Crossref] [PubMed]
  102. Lyons L, Elbeltagy M, Bennett G, et al. Fluoxetine counteracts the cognitive and cellular effects of 5-Fluorouracil in the rat hippocampus by a mechanism of prevention rather than recovery. PLoS One 2012;7:e30010. [Crossref] [PubMed]
  103. Lyons L, Lyons L, ElBeltagy M, et al. Fluoxetine reverses the memory impairment and reduction in proliferation and survival of hippocampal cells caused by methotrexate chemotherapy. Psychopharmacology 2011;215:105-15. [Crossref] [PubMed]
  104. ElBeltagy M, Mustafa S, Umka J, et al. Fluoxetine improves the memory deficits caused by the chemotherapy agent 5-fluorouracil. Behav Brain Res 2010;208:112-7. [Crossref] [PubMed]
  105. Tanimukai H, Kudo T. Fluvoxamine alleviates paclitaxel-induced neurotoxicity. Biochem Biophys Rep 2015;4:202-6. [Crossref] [PubMed]
  106. Winocur G, Binns MA, Tannock I. Donepezil reduces cognitive impairment associated with anti-cancer drugs in a mouse model. Neuropharmacology 2011;61:1222-8. [Crossref] [PubMed]
  107. Lim I, Joung HY, Yu AR, et al. PET Evidence of the Effect of Donepezil on Cognitive Performance in an Animal Model of Chemobrain. Biomed Res Int 2016;2016:6945415. [Crossref] [PubMed]
  108. 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 2016;10:176-84. [Crossref] [PubMed]
  109. Helal GK, Aleisa AM, Helal OK, et al. Metallothionein induction reduces caspase-3 activity and TNFalpha levels with preservation of cognitive function and intact hippocampal neurons in carmustine-treated rats. Oxid Med Cell Longev 2009;2:26-35. [Crossref] [PubMed]
  110. Konat GW, Kraszpulski M, James I, et al. Cognitive dysfunction induced by chronic administration of common cancer chemotherapeutics in rats. Metab Brain Dis 2008;23:325-33. [Crossref] [PubMed]
  111. Lomeli N, Di K, Czerniawski J, et al. Cisplatin-induced mitochondrial dysfunction is associated with impaired cognitive function in rats. Free Radic Biol Med 2017;102:274-86. [Crossref] [PubMed]
  112. Briones TL, Woods J. Dysregulation in myelination mediated by persistent neuroinflammation: possible mechanisms in chemotherapy-related cognitive impairment. Brain Behav Immun 2014;35:23-32. [Crossref] [PubMed]
  113. Winocur G, Wojtowicz JM, Tannock IF. Memory loss in chemotherapy-treated rats is exacerbated in high-interference conditions and related to suppression of hippocampal neurogenesis. Behav Brain Res 2015;281:239-44. [Crossref] [PubMed]
  114. Zhou Y, Qiu Y. Propofol alleviates cisplatin-related cognitive impairment. Neurol Sci 2019;40:1645-9. [Crossref] [PubMed]
  115. Zheng YY, Lan YP, Tang HF, et al. Propofol pretreatment attenuates aquaporin-4 over-expression and alleviates cerebral edema after transient focal brain ischemia reperfusion in rats. Anesth Analg 2008;107:2009-16. [Crossref] [PubMed]
  116. Attia A, Rapp SR, Case LD, et al. Phase II study of Ginkgo biloba in irradiated brain tumor patients: effect on cognitive function, quality of life, and mood. J Neurooncol 2012;109:357-63. [Crossref] [PubMed]
  117. Kaschel R. Ginkgo biloba: specificity of neuropsychological improvement--a selective review in search of differential effects. Hum Psychopharmacol 2009;24:345-70. [Crossref] [PubMed]
  118. Fernandes HA, Richard NM, Edelstein K. Cognitive rehabilitation for cancer-related cognitive dysfunction: a systematic review. Support Care Cancer 2019;27:3253-79. [Crossref] [PubMed]
  119. Wilson BA. Towards a comprehensive model of cognitive rehabilitation. Neuropsychol Rehabilitation 2002;12:97-110. [Crossref]
  120. 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. [Crossref] [PubMed]
  121. Kesler S, Hadi Hosseini SM, Heckler C, et al. Cognitive training for improving executive function in chemotherapy-treated breast cancer survivors. Clin Breast Cancer 2013;13:299-306. [Crossref] [PubMed]
  122. Schuurs A, Green HJ. A feasibility study of group cognitive rehabilitation for cancer survivors: enhancing cognitive function and quality of life. Psycho-Oncology 2013;22:1043-9. [Crossref] [PubMed]
  123. Ferguson RJ, McDonald BC, Rocque MA, et al. Development of CBT for chemotherapy-related cognitive change: results of a waitlist control trial. Psychooncology 2012;21:176-86. [Crossref] [PubMed]
  124. Von Ah D, Carpenter JS, Saykin A, et al. Advanced cognitive training for breast cancer survivors: a randomized controlled trial. Breast Cancer Res Treat 2012;135:799-809. [Crossref] [PubMed]
  125. Gehring K, Roukema JA, Sitskoorn MM. Review of recent studies on interventions for cognitive deficits in patients with cancer. Expert Rev Anticancer Ther 2012;12:255-69. [Crossref] [PubMed]
  126. Johns SA, Von Ah D, Brown LF, et al. Randomized controlled pilot trial of mindfulness-based stress reduction for breast and colorectal cancer survivors: effects on cancer-related cognitive impairment. J Cancer Surviv 2016;10:437-48. [Crossref] [PubMed]
  127. Johnston MF, Hays RD, Subramanian SK, et al. Patient education integrated with acupuncture for relief of cancer-related fatigue randomized controlled feasibility study. BMC Complement Altern Med 2011;11:49. [Crossref] [PubMed]
  128. Oh B, Butow PN, Mullan BA, et al. Effect of medical Qigong on cognitive function, quality of life, and a biomarker of inflammation in cancer patients: a randomized controlled trial. Support Care Cancer 2012;20:1235-42. [Crossref] [PubMed]
  129. Mishra SI, Mishra SI, Scherer RW, et al. Exercise interventions on health-related quality of life for cancer survivors. Cochrane Database Syst Rev 2012;8:CD007566. [PubMed]
  130. Zimmer P, Baumann FT, Oberste M, et al. Effects of exercise interventions and physical activity behavior on cancer related cognitive impairments: A systematic review. BioMed Res Int 2016;2016:1820954-13. [Crossref] [PubMed]
  131. Janelsins MC, Peppone LJ, Heckler CE, et al. 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 2016;15:263-71. [Crossref] [PubMed]
  132. Wayne PM, Lee MS, Novakowski J, et al. Tai Chi and Qigong for cancer-related symptoms and quality of life: a systematic review and meta-analysis. J Cancer Surviv 2018;12:256-67. [Crossref] [PubMed]
  133. Szuhany KL, Bugatti M, Otto MW. A meta-analytic review of the effects of exercise on brain-derived neurotrophic factor. J Psychiatr Res 2015;60:56-64. [Crossref] [PubMed]
  134. Skriver K, Roig M, Lundbye-Jensen J, et al. Acute exercise improves motor memory: exploring potential biomarkers. Neurobiol Learn Mem 2014;116:46-58. [Crossref] [PubMed]
  135. Erickson KI, Leckie RL, Weinstein AM. Physical activity, fitness, and gray matter volume. Neurobiol Aging 2014;35:S20-8. [Crossref] [PubMed]
  136. Hartman SJ, Nelson SH, Myers E, et al. Randomized controlled trial of increasing physical activity on objectively measured and self-reported cognitive functioning among breast cancer survivors: The memory & motion study. Cancer 2018;124:192-202. [Crossref] [PubMed]
  137. Oberste M, Schaffrath N, Schmidt K, et al. Protocol for the "Chemobrain in Motion - study" (CIM - study): a randomized placebo-controlled trial of the impact of a high-intensity interval endurance training on cancer related cognitive impairments in women with breast cancer receiving first-line chemotherapy. BMC Cancer 2018;18:1071-14. [Crossref] [PubMed]
  138. Alvarez J, Meyer FL, Granoff DL, et al. The Effect of EEG Biofeedback on Reducing Postcancer Cognitive Impairment. Integr Cancer Ther 2013;12:475-87. [Crossref] [PubMed]
  139. Zeng Y, Cheng ASK, Song T, et al. Effects of Acupuncture on Cancer-Related Cognitive Impairment in Chinese Gynecological Cancer Patients: A Pilot Cohort Study. Integr Cancer Ther 2018;17:737-46. [Crossref] [PubMed]
  140. Wong P, Leppert IR, Roberge D, et al. A pilot study using dynamic contrast enhanced-MRI as a response biomarker of the radioprotective effect of memantine in patients receiving whole brain radiotherapy. Oncotarget 2016;7:50986-96. [Crossref] [PubMed]
  141. Heck JE, Albert SM, Franco R, et al. Patterns of dementia diagnosis in surveillance, epidemiology, and end results breast cancer survivors who use chemotherapy. J Am Geriatr Soc 2008;56:1687-92. [Crossref] [PubMed]
  142. Driver JA, Beiser A, Au R, et al. Inverse association between cancer and Alzheimer's disease: results from the Framingham Heart Study. BMJ 2012;344:e1442. [Crossref] [PubMed]
  143. Yu E, Liao Z, Tan Y, et al. The effect of fufanghaishe capsule on preventing amnestic mild cognitive impairment developing into Alzheimer Disease. Chinese Journal of Geriatrics 2017;36:278-81.
  144. Guse B, Falkai P, Wobrock T. Cognitive effects of high-frequency repetitive transcranial magnetic stimulation: a systematic review. J Neural Transm (Vienna) 2010;117:105-22. [Crossref] [PubMed]
Cite this article as: Bai L, Yu E. A narrative review of risk factors and interventions for cancer-related cognitive impairment. Ann Transl Med 2021;9(1):72. doi: 10.21037/atm-20-6443

Download Citation