1
|
Oliva G, Giustiniani A, Danesin L, Burgio F, Arcara G, Conte P. Cognitive impairment following breast cancer treatments: an umbrella review. Oncologist 2024; 29:e848-e863. [PMID: 38723166 PMCID: PMC11224991 DOI: 10.1093/oncolo/oyae090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/16/2024] [Indexed: 07/06/2024] Open
Abstract
OBJECTIVES Cancer-related cognitive impairment (CRCI) refers to a cognitive decline associated with cancer or its treatments. While research into CRCI is expanding, evidence remains scattered due to differences in study designs, methodologies, and definitions. The present umbrella review aims to provide a comprehensive overview of the current evidence regarding the impact of different breast cancer therapies on cognitive functioning, with a particular focus on the interplay among objective cognitive deficits (ie, measured with standardized tests), subjective cognitive concerns, (ie, self-reported), and other mediating psycho-physical factors. METHODS The search was made in Pubmed, Embase, and Scopus for articles published until July 2023, following the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-analysis protocol. RESULTS Chemotherapy and endocrine therapy appear consistently associated with CRCI in patients with breast cancer, primarily affecting memory, attention/concentration, executive functioning, and processing speed. Subjective cognitive concerns were often found weakly or not associated with neuropsychological test results, while overall CRCI seemed consistently associated with psychological distress, fatigue, sleep quality, and inflammatory and biological factors. CONCLUSION Current evidence suggests that CRCI is common after chemotherapy and endocrine therapy for breast cancer. However, heterogeneity in study designs and the scarcity of studies on more recent treatments such as targeted therapies and immunotherapies, highlight the need for more systematic and harmonized studies, possibly taking into account the complex and multifactorial etiology of CRCI. This may provide valuable insights into CRCI's underlying mechanisms and potential new ways to treat it.
Collapse
Affiliation(s)
- Giulia Oliva
- Department of Surgery, Oncology and Gastroenterology (DiSCOG), University of Padua, 35124 Padova, Italy
- IRCCS San Camillo Hospital, 30126 Venice, Italy
| | | | | | | | | | | |
Collapse
|
2
|
Branigan GL, Torrandell-Haro G, Chen S, Shang Y, Perez-Miller S, Mao Z, Padilla-Rodriguez M, Cortes-Flores H, Vitali F, Brinton RD. Breast cancer therapies reduce risk of Alzheimer's disease and promote estrogenic pathways and action in brain. iScience 2023; 26:108316. [PMID: 38026173 PMCID: PMC10663748 DOI: 10.1016/j.isci.2023.108316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 09/08/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Worldwide, an ever-increasing number of women are prescribed estrogen-modulating therapies (EMTs) for the treatment of breast cancer. In parallel, aging of the global population of women will contribute to risk of both breast cancer and Alzheimer's disease. To address the impact of anti-estrogen therapies on risk of Alzheimer's and neural function, we conducted medical informatic and molecular pharmacology analyses to determine the impact of EMTs on risk of Alzheimer's followed by determination of EMT estrogenic mechanisms of action in neurons. Collectively, these data provide both clinical and mechanistic data indicating that select EMTs exert estrogenic agonist action in neural tissue that are associated with reduced risk of Alzheimer's disease while simultaneously acting as effective estrogen receptor antagonists in breast.
Collapse
Affiliation(s)
- Gregory L. Branigan
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
- Department of Pharmacology, University of Arizona College of Medicine; Tucson AZ, USA
- Medical Scientist Training Program, University of Arizona College of Medicine; Tucson AZ, USA
| | - Georgina Torrandell-Haro
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
- Department of Pharmacology, University of Arizona College of Medicine; Tucson AZ, USA
| | - Shuhua Chen
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
| | - Yuan Shang
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
| | | | - Zisu Mao
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
| | | | | | - Francesca Vitali
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
- Center of Bioinformatics and Biostatistics, University of Arizona College of Medicine; Tucson AZ, USA
| | - Roberta Diaz Brinton
- Center for Innovation in Brain Science, University of Arizona; Tucson AZ, USA
- Department of Pharmacology, University of Arizona College of Medicine; Tucson AZ, USA
- Department of Neurology, University of Arizona College of Medicine; Tucson AZ, USA
| |
Collapse
|
3
|
Sharma S, Wright HH. Tamoxifen Effects on Cognition and Language in Women with Breast Cancer. Semin Speech Lang 2023; 44:189-202. [PMID: 37220780 DOI: 10.1055/s-0043-1768135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Cognitive changes following adjuvant treatment for breast cancer (BC) are well documented following chemotherapy. However, limited studies have examined cognitive and/or language functions in chemotherapy-naive women with BC taking tamoxifen (TAM). Using ambulatory cognitive assessment, we investigated the trajectory of cognitive and language changes during early period of adjuvant endocrine treatment (TAM) in women with BC at two time periods (pretreatment and 2 months after treatment began). Four women with BC and 18 cognitively healthy age-matched controls completed three cognitive tasks using smartphones, during a short time period (5 days) and repeated them at two time periods. To determine language ability, language samples were collected at two time periods, where the participants described two stories from two wordless picture books and samples were assessed using core lexicon analyses. Wilcoxon-signed rank tests were computed to identify differences in linguistic and cognitive performances of both the groups at two time periods. No significant within-group or between-group differences were seen on the cognitive and language tasks at the two time periods; however, women with BC performed more poorly compared to the control group. We did see decline in some women with BC and not in others, in cognition and language during initial course of TAM treatment. However, the approach we used to assess these changes is valuable and innovative. This approach will help refine current research paradigms for determining cognitive and linguistic changes and will help determine if women with BC might require language intervention in the future.
Collapse
Affiliation(s)
- Saryu Sharma
- Department of Communication Sciences and Disorders, Idaho State University, Pocatello, Idaho
| | - Heather Harris Wright
- Department of Communication Sciences and Disorders, East Carolina University, Greenville, North Carolina
| |
Collapse
|
4
|
Kara F, Lohse CM, Castillo AM, Tosakulwong N, Lesnick TG, Jack CR, Petersen RC, Olson JE, Couch FJ, Ruddy KJ, Kantarci K, Mielke MM. Association of raloxifene and tamoxifen therapy with cognitive performance, odds of mild cognitive impairment, and brain MRI markers of neurodegeneration. Cancer Med 2022; 12:2805-2817. [PMID: 36040183 PMCID: PMC9939086 DOI: 10.1002/cam4.5175] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 08/09/2022] [Accepted: 08/17/2022] [Indexed: 11/11/2022] Open
Abstract
The aim of this cross-sectional study was to examine whether a history of selective estrogen receptor modifiers (SERMs), tamoxifen and raloxifene, use was associated with cognitive performance, odds of mild cognitive impairment (MCI), or magnetic resonance imaging (MRI) markers of neurodegeneration associated with Alzheimer's disease. We included women with prior history of breast cancer or no prior history of any cancer at enrollment in the Mayo Clinic Study of Aging (MCSA). This information was abstracted using the Rochester Epidemiology Project medical-linkage system. Logistic regression was used to examine associations of SERMs with odds of MCI. Linear regression models were used to examine associations of SERMs with cognitive z-scores (Memory, Executive Function, Language, Visuospatial Skills, Global Cognition), and MRI markers. Among 2840 women aged 50 and older in the MCSA, 151 had a history of breast cancer, and 42 (28%) of these had a history of tamoxifen treatment. A total of 2235 women had no prior history of any cancer, and 76 (3%) of these had a history of raloxifene use. No significant associations between tamoxifen use and cognition, or odds of MCI were observed among women with a history of breast cancer after adjusting for confounders. Similarly, raloxifene use was not significantly associated with cognition, or odds of MCI in women without a history of cancer after adjusting for confounders. We did not find significant associations between the use of either SERM and MRI markers. Use of tamoxifen or raloxifene was not significantly associated with cognition in postmenopausal women.
Collapse
Affiliation(s)
- Firat Kara
- Department of RadiologyMayo ClinicRochesterMinnesotaUSA
| | - Christine M. Lohse
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | - Anna M. Castillo
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | | | - Timothy G. Lesnick
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA
| | | | | | - Janet E. Olson
- Health Sciences ResearchMayo ClinicRochesterMinnesotaUSA
| | - Fergus J. Couch
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | | | | | - Michelle M. Mielke
- Department of Quantitative Health SciencesMayo ClinicRochesterMinnesotaUSA,Department of Epidemiology and PreventionWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
5
|
Flores VA, Pal L, Manson JE. Hormone Therapy in Menopause: Concepts, Controversies, and Approach to Treatment. Endocr Rev 2021; 42:720-752. [PMID: 33858012 DOI: 10.1210/endrev/bnab011] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Indexed: 12/22/2022]
Abstract
Hormone therapy (HT) is an effective treatment for menopausal symptoms, including vasomotor symptoms and genitourinary syndrome of menopause. Randomized trials also demonstrate positive effects on bone health, and age-stratified analyses indicate more favorable effects on coronary heart disease and all-cause mortality in younger women (close proximity to menopause) than in women more than a decade past menopause. In the absence of contraindications or other major comorbidities, recently menopausal women with moderate or severe symptoms are appropriate candidates for HT. The Women's Health Initiative (WHI) hormone therapy trials-estrogen and progestin trial and the estrogen-alone trial-clarified the benefits and risks of HT, including how the results differed by age. A key lesson from the WHI trials, which was unfortunately lost in the posttrial cacophony, was that the risk:benefit ratio and safety profile of HT differed markedly by clinical characteristics of the participants, especially age, time since menopause, and comorbidity status. In the present review of the WHI and other recent HT trials, we aim to provide readers with an improved understanding of the importance of the timing of HT initiation, type and route of administration, and of patient-specific considerations that should be weighed when prescribing HT.
Collapse
Affiliation(s)
- Valerie A Flores
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - Lubna Pal
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale School of Medicine, New Haven, Connecticut, USA
| | - JoAnn E Manson
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
6
|
Jebahi F, Sharma S, Bloss JE, Wright HH. Effects of tamoxifen on cognition and language in women with breast cancer: A systematic search and a scoping review. Psychooncology 2021; 30:1262-1277. [PMID: 33866625 DOI: 10.1002/pon.5696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 12/24/2022]
Abstract
OBJECTIVE Breast cancer treatments bring adverse consequences that interfere with everyday functioning. Importantly, some of these treatments are associated with cognitive and language changes. Tamoxifen is a selective estrogen receptor modulator and is a common endocrine therapy treatment for breast cancer. The current review examines the specific domains of cognition and language affected by the use of tamoxifen in women with breast cancer. METHODS We conducted a systematic search that examined cognitive and/or language functions in chemotherapy-naïve women with breast cancer taking tamoxifen. PubMed, Cochrane CENTRAL, CINAHL Complete, PsycINFO, Scopus, EMBASE, and the Grey Literature Report (greylit.org) were searched. Covidence Systematic Review software (covidence.org) was used to manage the screening process of study titles and abstracts as well as full texts. A total of 17 studies were included in the review. RESULTS A range of cognitive and language domains were reported. These were grouped into seven broad domains: attention, memory, speed, executive functioning, verbal abilities, visual abilities, and language abilities. Results showed that there is compelling evidence that specific domains of memory and speed are negatively affected by the use of tamoxifen. In addition, there was a pattern of change in domains of executive functions and verbal abilities. CONCLUSIONS Tamoxifen affects specific cognitive and language domains. Language domains beyond semantics have not been studied and thus conclusions related to these domains, and language in general, could not be made. Studies exploring the effects of tamoxifen on the different domains of language are recommended.
Collapse
Affiliation(s)
- Fatima Jebahi
- Department of Communication Sciences and Disorders, East Carolina University, Greenville, North Carolina, USA.,Fulbright Association, Washington, District of Columbia, USA
| | - Saryu Sharma
- Department of Communication Sciences and Disorders, East Carolina University, Greenville, North Carolina, USA
| | - Jamie E Bloss
- Laupus Library Liaison, East Carolina University, Greenville, North Carolina, USA
| | - Heather H Wright
- Department of Communication Sciences and Disorders, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
7
|
Liu Y, Wei K, Cao X, Jiang L, Gu N, Feng L, Li C. Development and Validation of a Nomogram Based on Motoric Cognitive Risk Syndrome for Cognitive Impairment. Front Aging Neurosci 2021; 13:618833. [PMID: 33935682 PMCID: PMC8086554 DOI: 10.3389/fnagi.2021.618833] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 02/26/2021] [Indexed: 11/13/2022] Open
Abstract
Objective To develop and validate a prediction nomogram based on motoric cognitive risk syndrome for cognitive impairment in healthy older adults. Methods Using two longitudinal cohorts of participants (aged ≥ 60 years) with 4-year follow-up, we developed (n = 1,177) and validated (n = 2,076) a prediction nomogram. LASSO (least absolute shrinkage and selection operator) regression model and multivariable Cox regression analysis were used for variable selection and for developing the prediction model, respectively. The performance of the nomogram was assessed with respect to its calibration, discrimination, and clinical usefulness. Results The individualized prediction nomogram was assessed based on the following: motoric cognitive risk syndrome, education, gender, baseline cognition, and age. The model showed good discrimination [Harrell’s concordance index (C-index) of 0.814; 95% confidence interval, 0.782–0.835] and good calibration. Comparable results were also seen in the validation cohort, which includes good discrimination (C-index, 0.772; 95% confidence interval, 0.776–0.818) and good calibration. Decision curve analysis demonstrated that the prediction nomogram was clinically useful. Conclusion This prediction nomogram provides a practical tool with all necessary predictors, which are accessible to practitioners. It can be used to estimate the risk of cognitive impairment in healthy older adults.
Collapse
Affiliation(s)
- Yong Liu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Wei
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xinyi Cao
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Clinical Neurocognitive Research Center, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lijuan Jiang
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Nannan Gu
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Feng
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Chunbo Li
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Center for Excellence in Brain Science and Intelligence Technology (CEBSIT), Chinese Academy of Sciences, Shanghai, China.,Institute of Psychology and Behavioral Science, Shanghai Jiao Tong University, Shanghai, China.,Brain Science and Technology Research Center, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
8
|
The short-term effects of estradiol, raloxifene, and a phytoestrogen in women with perimenopausal depression. ACTA ACUST UNITED AC 2021; 28:369-383. [PMID: 33470755 DOI: 10.1097/gme.0000000000001724] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE We examined the short-term efficacies of three estrogen-like compounds under placebo-controlled conditions in women with perimenopause-related depression (PMD). METHODS Women with PMD were randomized in a double-blind parallel design to one of four treatments: transdermal 17-beta estradiol (TE) (100 mcg/d); oral raloxifene (60 mg/d); a proprietary phytoestrogen compound, Rimostil (1,000 mg twice/d); or placebo for 8 weeks. The main outcome measures were the Center for Epidemiology Studies Depression Scale, 17-item Hamilton Rating Scale for Depression (HRSD), and the Beck Depression Inventory completed at each clinic visit. Secondary outcomes included a visual analogue self-rating completed at each clinic visit, and daily self-ratings of hot flush severity. Cognitive tests were performed at pretreatment baseline and at the end of the trial. In the primary analysis, we obtained four repeated measures in each woman in the four treatment arms. Analyses were done with SAS Version 9.4 software (SAS Institute, Inc, Cary, NC), using PROC MIXED (for mixed models). All models included the following four explanatory variables, regardless of whether they were statistically significant: 1) treatment group (TE, raloxifene, Rimostil, placebo); 2) week (W2, W4, W6, W8); 3) treatment group-by-week interaction; and 4) baseline value of the measure being analyzed. The inclusion of additional variables was evaluated individually for each outcome measure. RESULTS Sixty-six women were randomized into the trial, four women dropped out of the trial, and 62 women were included in the final data analysis. No effect of treatment group was observed in either the Center for Epidemiology Studies Depression Scale (P = 0.34) or Beck Depression Inventory (P = 0.27) scores; however, there was a difference in HRSD scores between treatment groups (P = 0.0037) that pair-wise comparisons of the combined weekly scores in each treatment demonstrated TE's beneficial effects on HRSD scores compared with Rimostil (P = 0.0005), and less consistently with placebo (P = 0.099). The average (SD) of the baseline scores for each treatment group on the HRSD was as follows: TE-15.3 (4.5), raloxifene-16.0 (3.7), Rimostil-14.0 (2.7), and placebo-15.2 (3.0). Whereas the HRSD scores after 8 weeks of treatment (least-square means) were TE-5.2(1.1), raloxifene-5.8(1.2), Rimostil-11.2(1.4), and placebo-7.8(1.1). No differences were observed between raloxifene and either TE or placebo in any scale score. HRSD scores in women assigned to TE were improved compared with those on Rimostil during weeks 6 and 8 (P values = 0.0008, 0.0011, respectively). Cognitive testing at week 8 showed that none of the three active treatment groups performed better than placebo. CONCLUSIONS This study did not identify significant therapeutic benefits of TE, Rimostil, or raloxifene compared with placebo in PMD. However, improvements in depression ratings were observed between TE compared with Rimostil. Thus, our findings do not support the role of ERbeta compounds in the treatment of PMD (and indeed could suggest a more important role of ERalpha).
Collapse
|
9
|
Zhang Z, Shi J, Yang T, Liu T, Zhang K. Management of aggressive fibromatosis. Oncol Lett 2021; 21:43. [PMID: 33262835 PMCID: PMC7693298 DOI: 10.3892/ol.2020.12304] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/30/2020] [Indexed: 01/12/2023] Open
Abstract
Aggressive fibromatosis or desmoid tumor is a rare disease resulting from fibroblasts which do not metastasize. However, desmoid tumors belong to low-grade malignant tumors since they have high potential to infiltrate surrounding tissues, causing high local recurrence rates and may affect surrounding organs, threatening life quality and expectancy. Although surgery, watch and wait, radiotherapy, chemotherapy, high intensity focused ultrasound, ablation techniques or several agents have all been frequently investigated for the treatment of this type of disease, none are deemed as standard therapy for high recurrence rates that have been supported by any data. The present review retrieved literature on treatment options for desmoids to summarize the latest treatment modalities and refine their efficacy, as well as their side effects, in order to provide a more comprehensive treatment reference for clinicians.
Collapse
Affiliation(s)
- Zhijun Zhang
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Jian Shi
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Tao Yang
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Tongjun Liu
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| | - Kai Zhang
- Department of Colorectal and Anal Surgery, The Second Hospital of Jilin University, Changchun, Jilin 130012, P.R. China
| |
Collapse
|
10
|
Gurvich C, Hudaib A, Gavrilidis E, Worsley R, Thomas N, Kulkarni J. Raloxifene as a treatment for cognition in women with schizophrenia: the influence of menopause status. Psychoneuroendocrinology 2019; 100:113-119. [PMID: 30299258 DOI: 10.1016/j.psyneuen.2018.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 10/01/2018] [Accepted: 10/01/2018] [Indexed: 02/06/2023]
Abstract
Cognitive impairments cause significant functional issues for people with schizophrenia, often emerging before the onset of hallucinations, delusions and other psychosis symptoms. Current pharmacological treatments do not target cognitive dysfunction. Several lines of evidence support the beneficial effects of estrogens on cognition. Raloxifene hydrochloride, a selective estrogen receptor modulator, has been associated with cognitive improvements in healthy postmenopausal women and in schizophrenia, although findings are inconsistent. Using pooled data from two clinical trials, the aim of the current study was to compare the efficacy of 120 mg/day adjunctive raloxifene to placebo for 12 weeks on cognitive performance in women with schizophrenia who were stratified by menopause status (pre-menopausal; peri-menopausal or post-menopausal). A total of sixty-nine participants with a diagnosis of schizophrenia or schizoaffective disorder were included. Cognition was assessed at baseline and study end using the Repeatable Battery for the Assessment of Neuropsychological Status (RBANS). Results indicated that after stratifying for menopause status (strata) and adjusting for endogenous hormone levels (estrogen, progesterone, follicle stimulating hormone and luteinising hormone), semantic fluency, picture naming and list recognition change from baseline scores for the raloxifene group differed significantly from the placebo group. The findings from the current study highlight the importance of considering menopause status when interpreting the effects of hormonal treatments.
Collapse
Affiliation(s)
- C Gurvich
- Monash Alfred Psychiatry research centre, Monash University and The Alfred Hospital, Central Clinical School, Melbourne, Australia.
| | - A Hudaib
- Monash Alfred Psychiatry research centre, Monash University and The Alfred Hospital, Central Clinical School, Melbourne, Australia
| | - E Gavrilidis
- Monash Alfred Psychiatry research centre, Monash University and The Alfred Hospital, Central Clinical School, Melbourne, Australia
| | - R Worsley
- Monash Alfred Psychiatry research centre, Monash University and The Alfred Hospital, Central Clinical School, Melbourne, Australia
| | - N Thomas
- Monash Alfred Psychiatry research centre, Monash University and The Alfred Hospital, Central Clinical School, Melbourne, Australia
| | - J Kulkarni
- Monash Alfred Psychiatry research centre, Monash University and The Alfred Hospital, Central Clinical School, Melbourne, Australia
| |
Collapse
|
11
|
The role of estradiol in schizophrenia diagnosis and symptoms in postmenopausal women. Schizophr Res 2018; 196:35-38. [PMID: 28587815 DOI: 10.1016/j.schres.2017.05.024] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 05/19/2017] [Accepted: 05/24/2017] [Indexed: 12/17/2022]
Abstract
Schizophrenia is one of the most common mental illnesses in our society, affecting up to 1% of the population. There has been an increase in the number of people who are living longer with schizophrenia and people are being diagnosed later in life, with the majority of those later diagnoses being in women. In addition, there is a spike in diagnoses after women go through menopause, suggesting an important role for gonadal steroids in the disease. This paper examined aspects of aging and schizophrenia in the context of hormonal changes in women. With the rising prevalence rate of schizophrenia and the unique challenges that women face while aging with this disease, the idea of estrogen as a therapeutic agent to reduce symptom severity in postmenopausal women should be considered. In addition, we reviewed literature that suggests that estrogen interacts with the dopaminergic system to affect cognition and this should be studied further in older women with schizophrenia. Positive results in these studies have the potential to drastically improve the aging process for postmenopausal women with schizophrenia.
Collapse
|
12
|
Affiliation(s)
- Andrea Martoni
- UO Oncologia Medica, Policlinico S Orsola-Malpighi, Bologna, Italy.
| | | | | |
Collapse
|
13
|
Tonelli F, Ficari F, Valanzano R, Brandi ML. Treatment of Desmoids and Mesenteric Fibromatosis in Familial Adenomatous Polyposis with Raloxifene. TUMORI JOURNAL 2018; 89:391-6. [PMID: 14606641 DOI: 10.1177/030089160308900408] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background Among the great variety of extracolonic manifestations of familial adenomatous polyposis, the most serious are desmoids and fibromatosis of the abdominal cavity. These may be a danger to the patient and a concern to the clinician. Pharmacological management of this relentless problem is favored by surgical intervention. At present, however, beneficial actions of medical therapy are not separable from undesirable side effects. Methods We studied the effects of 120 mg daily of raloxifene, a non-steroidal benzothiophene, on progressive desmoid tumors and mesenteric fibromatosis by evaluation of lesion size and symptoms in 13 patients with familial adenomatous polyposis, selected on the basis of intra-abdominal localization of the lesion, on refractoriness to other medical treatments, and on estrogen receptor-α expression. Results The patients had a significant response to raloxifene therapy, with complete remission in 8 cases and partial response in 5 cases, evaluated by regression of symptoms and tumor size. Serum biochemical parameters did not show any significant changes. Side effects were never observed. Conclusions Although the number of patients included in the study is limited and in spite of some limitations, the available results support that, in the evaluation of response, daily therapy with raloxifene decreases desmoid tumor and mesenteric fibromatosis size and symptoms and does not cause side effects. These findings offer a novel option in the pharmacological treatment of desmoids, leading to medical therapy of these neoplastic lesions in familial adenomatous polyposis patients.
Collapse
Affiliation(s)
- Francesco Tonelli
- Department of Clinical Physiopathology, University of Florence, Italy
| | | | | | | |
Collapse
|
14
|
de Boer J, Prikken M, Lei WU, Begemann M, Sommer I. The effect of raloxifene augmentation in men and women with a schizophrenia spectrum disorder: a systematic review and meta-analysis. NPJ SCHIZOPHRENIA 2018; 4:1. [PMID: 29321530 PMCID: PMC5762671 DOI: 10.1038/s41537-017-0043-3] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/18/2017] [Accepted: 12/18/2017] [Indexed: 12/20/2022]
Abstract
Recognizing the robust sex differences in schizophrenia prevalence, the selective estrogen receptor modulator (SERM) raloxifene is a likely candidate for augmentation therapy in this disorder. Therefore, a systematic search was performed using PubMed (Medline), Embase, PsychInfo, and Cochrane Database of Systematic Reviews. Randomized controlled trials investigating the effect of raloxifene in schizophrenia spectrum disorders were included in the quantitative analyses. Outcome measures were psychotic symptom severity, depression, and cognition. Meta-analyses were performed using Comprehensive Meta-Analysis software. A random-effects model was used to compute overall weighted effect sizes in Hedges’ g. Nine studies were included, investigating 561 patients with a schizophrenia spectrum disorder. Raloxifene was superior to placebo in improving total symptom severity (N = 482; Hedge’s g = .57, p = 0.009), as well as positive (N = 561; Hedge’s g = 0.32, p = 0.02), negative (N = 561; Hedge’s g = 0.40, p = 0.02), and general (N = 526; Hedge’s g = 0.46, p = 0.01) subscales, as measured by the Positive and Negative Syndrome Scale. No significant effects were found for comorbid depression and cognitive functioning. Altogether, these results confirm the potential of raloxifene augmentation in the treatment of schizophrenia.
Collapse
Affiliation(s)
- Janna de Boer
- Department of Psychiatry, University Medical Center Utrecht, Utrecht University & Brain Center Rudolf Magnus, Utrecht, The Netherlands.
| | - Merel Prikken
- Department of Psychiatry, University Medical Center Utrecht, Utrecht University & Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Wan U Lei
- Department of Psychiatry, University Medical Center Utrecht, Utrecht University & Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Marieke Begemann
- Department of Psychiatry, University Medical Center Utrecht, Utrecht University & Brain Center Rudolf Magnus, Utrecht, The Netherlands
| | - Iris Sommer
- Department of Neuroscience and Department of Psychiatry, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.,Department of Biological and Medical Psychology, University of Bergen, Bergen, Norway
| |
Collapse
|
15
|
Tai LM, Balu D, Avila-Munoz E, Abdullah L, Thomas R, Collins N, Valencia-Olvera AC, LaDu MJ. EFAD transgenic mice as a human APOE relevant preclinical model of Alzheimer's disease. J Lipid Res 2017; 58:1733-1755. [PMID: 28389477 PMCID: PMC5580905 DOI: 10.1194/jlr.r076315] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Revised: 04/06/2017] [Indexed: 01/12/2023] Open
Abstract
Identified in 1993, APOE4 is the greatest genetic risk factor for sporadic Alzheimer's disease (AD), increasing risk up to 15-fold compared with APOE3, with APOE2 decreasing AD risk. However, the functional effects of APOE4 on AD pathology remain unclear and, in some cases, controversial. In vivo progress to understand how the human (h)-APOE genotypes affect AD pathology has been limited by the lack of a tractable familial AD-transgenic (FAD-Tg) mouse model expressing h-APOE rather than mouse (m)-APOE. The disparity between m- and h-apoE is relevant for virtually every AD-relevant pathway, including amyloid-β (Aβ) deposition and clearance, neuroinflammation, tau pathology, neural plasticity and cerebrovascular deficits. EFAD mice were designed as a temporally useful preclinical FAD-Tg-mouse model expressing the h-APOE genotypes for identifying mechanisms underlying APOE-modulated symptoms of AD pathology. From their first description in 2012, EFAD mice have enabled critical basic and therapeutic research. Here we review insights gleaned from the EFAD mice and summarize future directions.
Collapse
Affiliation(s)
- Leon M Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Evangelina Avila-Munoz
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Riya Thomas
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | - Nicole Collins
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612
| | | | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL 60612.
| |
Collapse
|
16
|
Bender AC, Austin AM, Grodstein F, Bynum JPW. Executive function, episodic memory, and Medicare expenditures. Alzheimers Dement 2017; 13:792-800. [PMID: 28174070 DOI: 10.1016/j.jalz.2016.12.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 11/08/2016] [Accepted: 12/28/2016] [Indexed: 11/24/2022]
Abstract
INTRODUCTION We examined the relationship between health care expenditures and cognition, focusing on differences across cognitive systems defined by global cognition, executive function, or episodic memory. METHODS We used linear regression models to compare annual health expenditures by cognitive status in 8125 Nurses' Health Study participants who completed a cognitive battery and were enrolled in Medicare parts A and B. RESULTS Adjusting for demographics and comorbidity, executive impairment was associated with higher total annual expenditures of $1488 per person (P < .01) compared with those without impairment. No association for episodic memory impairment was found. Expenditures exhibited a linear relationship with executive function, but not episodic memory ($584 higher for every 1 standard deviation decrement in executive function; P < .01). DISCUSSION Impairment in executive function is specifically and linearly associated with higher health care expenditures. Focusing on management strategies that address early losses in executive function may be effective in reducing costly services.
Collapse
Affiliation(s)
- Alex C Bender
- Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Andrea M Austin
- The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Francine Grodstein
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Julie P W Bynum
- The Dartmouth Institute for Health Policy and Clinical Practice, Geisel School of Medicine at Dartmouth, Hanover, NH, USA.
| |
Collapse
|
17
|
Osmanovic-Barilar J, Salkovic-Petrisi M. Evaluating the Role of Hormone Therapy in Postmenopausal Women with Alzheimer’s Disease. Drugs Aging 2016; 33:787-808. [DOI: 10.1007/s40266-016-0407-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
18
|
The Potential of Gonadal Hormone Signalling Pathways as Therapeutics for Dementia. J Mol Neurosci 2016; 60:336-348. [PMID: 27525638 DOI: 10.1007/s12031-016-0813-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/01/2016] [Indexed: 12/17/2022]
Abstract
Dementia is an ever-expanding problem facing an ageing society. Currently, there is a sharp paucity of treatment strategies. It has long been known that sex hormones, namely 17β-estradiol and testosterone, possess neuroprotective- and cognitive-enhancing qualities. However, certain lacunae in the knowledge underlying their molecular mechanisms have delayed their use as treatment strategies in dementia. With recent advancements in pharmacology and molecular biology, especially in the development of safer selective oestrogen receptor modulators and the recent discovery of the small-molecule brain-derived neurotrophic factor receptor agonist, 7,8-dihydroxyflavone, the exploitation of these signalling pathways for clinical use has become possible. This review aims to adumbrate the evidence and hurdles underscoring the use of sex hormones in the treatment of dementia as well as discussing some direction that is required to advance the translation of evidence into practise.
Collapse
|
19
|
Yazğan B, Yazğan Y, Övey İS, Nazıroğlu M. Raloxifene and Tamoxifen Reduce PARP Activity, Cytokine and Oxidative Stress Levels in the Brain and Blood of Ovariectomized Rats. J Mol Neurosci 2016; 60:214-22. [PMID: 27372663 DOI: 10.1007/s12031-016-0785-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 06/21/2016] [Indexed: 01/14/2023]
Abstract
It is well known that 17β-estradiol (E2) has an antioxidant role on neurological systems in the brain. Raloxifene (RLX) and tamoxifen (TMX) are selective estrogen receptor modulators. An E2 deficiency stimulates mitochondrial functions for promoting apoptosis and increasing reactive oxygen species (ROS) production. However, RLX and TMX may reduce the mitochondrial ROS production via their antioxidant properties in the brain and erythrocytes of ovariectomized (OVX) rats. We aimed to investigate the effects of E2, RLX, and TMX on oxidative stress, apoptosis, and cytokine production in the brain and erythrocytes of OVX rats.Forty female rats were divided into five groups. The first group was used as a control group. The second group was the OVX group. The third, fourth, and fifth groups were OVX + E2, OVX + TMX, and OVX + RLX groups, respectively. E2, TMX, and RLX were given subcutaneously to the OVX + E2 and OVX + TMX, OVX + RLX groups for 14 days after the ovariectomy respectively.While brain and erythrocyte lipid peroxidation levels were high in the OVX group, they were low in the OVX + E2, OVX + RLX, and OVX + TMX groups. OVX + E2, OVX + RLX, and OVX + TMX treatments increased the lowered glutathione peroxidase activity in erythrocytes and the brain and reduced glutathione and vitamin E concentrations in the brain. β-carotene and vitamin A concentrations in the brain and TNF-α and interleukin (IL)-1β levels in the plasma of the five groups were not significantly changed by the treatments. However, increased plasma IL-4 levels and Western blot results for brain poly (ADP-ribose) polymerase (PARP) in the OVX groups were decreased by E2, TMX, and RLX treatments, although proapoptotic procaspase 3 and 9 activities were increased by the treatments.In conclusion, we observed that E2, RLX, and TMX administrations were beneficial on oxidative stress, inflammation, and PARP levels in the serum and brain of OVX rats by modulating antioxidant systems, DNA damage, and cytokine production.
Collapse
Affiliation(s)
- Betül Yazğan
- Department of Physiology, Medical Faculty, Adıyaman University, Adıyaman, Turkey
| | - Yener Yazğan
- Department of Biophysics, Medical Faculty, Suleyman Demirel University, Isparta, Turkey
| | - İshak Suat Övey
- Department of Biophysics, Medical Faculty, Suleyman Demirel University, Isparta, Turkey
| | - Mustafa Nazıroğlu
- Department of Biophysics, Medical Faculty, Suleyman Demirel University, Isparta, Turkey. .,Neuroscience Research Center, Suleyman Demirel University, Isparta, Turkey.
| |
Collapse
|
20
|
Baber RJ, Panay N, Fenton A. 2016 IMS Recommendations on women’s midlife health and menopause hormone therapy. Climacteric 2016; 19:109-50. [DOI: 10.3109/13697137.2015.1129166] [Citation(s) in RCA: 520] [Impact Index Per Article: 65.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
21
|
Henderson VW, Ala T, Sainani KL, Bernstein AL, Stephenson BS, Rosen AC, Farlow MR. Raloxifene for women with Alzheimer disease: A randomized controlled pilot trial. Neurology 2015; 85:1937-44. [PMID: 26537053 DOI: 10.1212/wnl.0000000000002171] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2015] [Accepted: 08/06/2015] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To determine whether raloxifene, a selective estrogen receptor modulator, improves cognitive function compared with placebo in women with Alzheimer disease (AD) and to provide an estimate of cognitive effect. METHODS This pilot study was conducted as a randomized, double-blind, placebo-controlled trial, with a planned treatment of 12 months. Women with late-onset AD of mild to moderate severity were randomly allocated to high-dose (120 mg) oral raloxifene or identical placebo provided once daily. The primary outcome compared between treatment groups at 12 months was change in the Alzheimer's Disease Assessment Scale, cognitive subscale (ADAS-cog). RESULTS Forty-two women randomized to raloxifene or placebo were included in intent-to-treat analyses (mean age 76 years, range 68-84), and 39 women contributed 12-month outcomes. ADAS-cog change scores at 12 months did not differ significantly between treatment groups (standardized difference 0.03, 95% confidence interval -0.39 to 0.44, 2-tailed p = 0.89). Raloxifene and placebo groups did not differ significantly on secondary analyses of dementia rating, activities of daily living, behavior, or a global cognition composite score. Caregiver burden and caregiver distress were similar in both groups. CONCLUSIONS Results on the primary outcome showed no cognitive benefits in the raloxifene-treated group. CLASSIFICATION OF EVIDENCE This study provides Class I evidence that for women with AD, raloxifene does not have a significant cognitive effect. The study lacked the precision to exclude a small effect.
Collapse
Affiliation(s)
- Victor W Henderson
- From the Departments of Health Research and Policy (Epidemiology) (V.W.H., K.L.S.), Neurology and Neurological Sciences (V.W.H.), and Psychiatry and Behavioral Sciences (A.C.R.), Stanford University, CA; Department of Neurology (T.A.), Southern Illinois University School of Medicine, Springfield; Neurology (A.L.B.) and Family Medicine (B.S.S.) Departments, Kaiser Permanente Santa Rosa Medical Center, CA; Palo Alto Veterans Affairs Health Care System (A.C.R.), CA; and Department of Neurology (M.R.F.), Indiana University School of Medicine, Indianapolis. A.L.B. is currently affiliated with Sonoma West Medical Center, Sebastopol, CA.
| | - Tom Ala
- From the Departments of Health Research and Policy (Epidemiology) (V.W.H., K.L.S.), Neurology and Neurological Sciences (V.W.H.), and Psychiatry and Behavioral Sciences (A.C.R.), Stanford University, CA; Department of Neurology (T.A.), Southern Illinois University School of Medicine, Springfield; Neurology (A.L.B.) and Family Medicine (B.S.S.) Departments, Kaiser Permanente Santa Rosa Medical Center, CA; Palo Alto Veterans Affairs Health Care System (A.C.R.), CA; and Department of Neurology (M.R.F.), Indiana University School of Medicine, Indianapolis. A.L.B. is currently affiliated with Sonoma West Medical Center, Sebastopol, CA
| | - Kristin L Sainani
- From the Departments of Health Research and Policy (Epidemiology) (V.W.H., K.L.S.), Neurology and Neurological Sciences (V.W.H.), and Psychiatry and Behavioral Sciences (A.C.R.), Stanford University, CA; Department of Neurology (T.A.), Southern Illinois University School of Medicine, Springfield; Neurology (A.L.B.) and Family Medicine (B.S.S.) Departments, Kaiser Permanente Santa Rosa Medical Center, CA; Palo Alto Veterans Affairs Health Care System (A.C.R.), CA; and Department of Neurology (M.R.F.), Indiana University School of Medicine, Indianapolis. A.L.B. is currently affiliated with Sonoma West Medical Center, Sebastopol, CA
| | - Allan L Bernstein
- From the Departments of Health Research and Policy (Epidemiology) (V.W.H., K.L.S.), Neurology and Neurological Sciences (V.W.H.), and Psychiatry and Behavioral Sciences (A.C.R.), Stanford University, CA; Department of Neurology (T.A.), Southern Illinois University School of Medicine, Springfield; Neurology (A.L.B.) and Family Medicine (B.S.S.) Departments, Kaiser Permanente Santa Rosa Medical Center, CA; Palo Alto Veterans Affairs Health Care System (A.C.R.), CA; and Department of Neurology (M.R.F.), Indiana University School of Medicine, Indianapolis. A.L.B. is currently affiliated with Sonoma West Medical Center, Sebastopol, CA
| | - B Sue Stephenson
- From the Departments of Health Research and Policy (Epidemiology) (V.W.H., K.L.S.), Neurology and Neurological Sciences (V.W.H.), and Psychiatry and Behavioral Sciences (A.C.R.), Stanford University, CA; Department of Neurology (T.A.), Southern Illinois University School of Medicine, Springfield; Neurology (A.L.B.) and Family Medicine (B.S.S.) Departments, Kaiser Permanente Santa Rosa Medical Center, CA; Palo Alto Veterans Affairs Health Care System (A.C.R.), CA; and Department of Neurology (M.R.F.), Indiana University School of Medicine, Indianapolis. A.L.B. is currently affiliated with Sonoma West Medical Center, Sebastopol, CA
| | - Allyson C Rosen
- From the Departments of Health Research and Policy (Epidemiology) (V.W.H., K.L.S.), Neurology and Neurological Sciences (V.W.H.), and Psychiatry and Behavioral Sciences (A.C.R.), Stanford University, CA; Department of Neurology (T.A.), Southern Illinois University School of Medicine, Springfield; Neurology (A.L.B.) and Family Medicine (B.S.S.) Departments, Kaiser Permanente Santa Rosa Medical Center, CA; Palo Alto Veterans Affairs Health Care System (A.C.R.), CA; and Department of Neurology (M.R.F.), Indiana University School of Medicine, Indianapolis. A.L.B. is currently affiliated with Sonoma West Medical Center, Sebastopol, CA
| | - Martin R Farlow
- From the Departments of Health Research and Policy (Epidemiology) (V.W.H., K.L.S.), Neurology and Neurological Sciences (V.W.H.), and Psychiatry and Behavioral Sciences (A.C.R.), Stanford University, CA; Department of Neurology (T.A.), Southern Illinois University School of Medicine, Springfield; Neurology (A.L.B.) and Family Medicine (B.S.S.) Departments, Kaiser Permanente Santa Rosa Medical Center, CA; Palo Alto Veterans Affairs Health Care System (A.C.R.), CA; and Department of Neurology (M.R.F.), Indiana University School of Medicine, Indianapolis. A.L.B. is currently affiliated with Sonoma West Medical Center, Sebastopol, CA
| |
Collapse
|
22
|
Kindler J, Weickert CS, Skilleter AJ, Catts SV, Lenroot R, Weickert TW. Selective Estrogen Receptor Modulation Increases Hippocampal Activity during Probabilistic Association Learning in Schizophrenia. Neuropsychopharmacology 2015; 40:2388-97. [PMID: 25829142 PMCID: PMC4538353 DOI: 10.1038/npp.2015.88] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 02/14/2015] [Accepted: 03/09/2015] [Indexed: 02/06/2023]
Abstract
People with schizophrenia show probabilistic association learning impairment in conjunction with abnormal neural activity. The selective estrogen receptor modulator (SERM) raloxifene preserves neural activity during memory in healthy older men and improves memory in schizophrenia. Here, we tested the extent to which raloxifene modifies neural activity during learning in schizophrenia. Nineteen people with schizophrenia participated in a twelve-week randomized, double-blind, placebo-controlled, cross-over adjunctive treatment trial of the SERM raloxifene administered orally at 120 mg daily to assess brain activity during probabilistic association learning using functional magnetic resonance imaging (fMRI). Raloxifene improved probabilistic association learning and significantly increased fMRI BOLD activity in the hippocampus and parahippocampal gyrus relative to placebo. A separate region of interest confirmatory analysis in 21 patients vs 36 healthy controls showed a positive association between parahippocampal neural activity and learning in patients, but no such relationship in the parahippocampal gyrus of healthy controls. Thus, selective estrogen receptor modulation by raloxifene concurrently increases activity in the parahippocampal gyrus and improves probabilistic association learning in schizophrenia. These results support a role for estrogen receptor modulation of mesial temporal lobe neural activity in the remediation of learning disabilities in both men and women with schizophrenia.
Collapse
Affiliation(s)
- Jochen Kindler
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia,Neuroscience Research Australia, Randwick, NSW, Australia,Department of Psychiatric Neurophysiology, University of Bern, Bern, Switzerland
| | - Cynthia Shannon Weickert
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia,Neuroscience Research Australia, Randwick, NSW, Australia,Schizophrenia Research Institute, Darlinghurst, NSW, Australia
| | - Ashley J Skilleter
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia,Neuroscience Research Australia, Randwick, NSW, Australia,Schizophrenia Research Institute, Darlinghurst, NSW, Australia
| | - Stanley V Catts
- School of Medical Science, University of Queensland, Brisbane, QLD, Australia
| | - Rhoshel Lenroot
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia,Neuroscience Research Australia, Randwick, NSW, Australia,Schizophrenia Research Institute, Darlinghurst, NSW, Australia
| | - Thomas W Weickert
- School of Psychiatry, University of New South Wales, Randwick, NSW, Australia,Neuroscience Research Australia, Randwick, NSW, Australia,Schizophrenia Research Institute, Darlinghurst, NSW, Australia,School of Psychiatry, University of New South Wales, Neuroscience Research Australia, Barker Street, Randwick, NSW 2031, Australia, Tel: +61 2 9399 1730, Fax: +61 2 9399 1034, E-mail:
| |
Collapse
|
23
|
Effects of raloxifene on cognition in postmenopausal women with schizophrenia: a double-blind, randomized, placebo-controlled trial. Eur Neuropsychopharmacol 2014; 24:223-31. [PMID: 24342775 DOI: 10.1016/j.euroneuro.2013.11.012] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 11/19/2013] [Accepted: 11/22/2013] [Indexed: 10/25/2022]
Abstract
Studies of estrogen therapy in postmenopausal women provide evidence of an effect of sex hormones on cognitive function. Estrogen has demonstrated some utility in the prevention of normal, age-related decline in cognitive functions, especially in memory. The potential therapeutic utility of estrogens in schizophrenia is increasingly being recognized. Raloxifene, a selective estrogen receptor modulator (SERM), appears to act similarly to conjugated estrogens on dopamine and serotonin brain systems, and may be a better option since it lacks the possible negative effects of estrogen on breast and uterine tissue. We assessed the utility of raloxifene as an adjuvant treatment for cognitive symptoms in postmenopausal women with schizophrenia in a 12-week, double-blind, randomized, placebo-controlled study. Patients were recruited from both the inpatient and outpatient departments. Thirty-three postmenopausal women with schizophrenia (DSM-IV) were randomized to receive either adjuvant raloxifene (16 women) or adjuvant placebo (17 women) for three months. The main outcome measures were: Memory, attention and executive functions. Assessment was conducted at baseline and week 12. The total sample is homogenous with respect to: age, years of schooling, illness duration, baseline symptomatology and pharmacological treatment. The addition of raloxifene (60 mg) to regular antipsychotic treatment showed: we found significant differences in some aspects of memory and executive function in patients treated with raloxifene. This improvement does not correlate with clinical improvement. The use of raloxifene as an adjuvant treatment in postmenopausal women with schizophrenia seems to be useful in improving cognitive symptoms.
Collapse
|
24
|
Abstract
Raloxifene (Evista, Eli Lilly), a selective estrogen receptor modulator (SERM) and ligand for the estrogen receptor (ER), competes with endogenous estrogen for ER binding. Raloxifene is approved for the prevention and treatment of osteoporosis, and shows promise as a breast cancer prevention drug. Raloxifene may be a preferred agent over tamoxifen due to its side-effect profile; in particular, it does not stimulate the endometrium and is not associated with endometrial cancer. The mechanisms for the differential tissue effects of raloxifene compared with other SERMs are not completely understood; the roles of ERalpha and -beta, classic and alternative signaling pathways, and drug conformation are discussed in this review. The utility of raloxifene will depend on the outcome of trials that are now underway, as well as acceptance by high-risk women and their healthcare practitioners.
Collapse
Affiliation(s)
- Jennifer Eng-Wong
- National Cancer Institute, Medical Oncology Clinical Research Unit, Bldg 10, Rm 12N226, Bethesda, MD 20892, USA.
| | | |
Collapse
|
25
|
Hurria A, Patel SK, Mortimer J, Luu T, Somlo G, Katheria V, Ramani R, Hansen K, Feng T, Chuang C, Geist CL, Silverman DHS. The effect of aromatase inhibition on the cognitive function of older patients with breast cancer. Clin Breast Cancer 2013; 14:132-40. [PMID: 24291380 DOI: 10.1016/j.clbc.2013.10.010] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 10/16/2013] [Accepted: 10/23/2013] [Indexed: 11/30/2022]
Abstract
INTRODUCTION This study evaluated the association between aromatase inhibitor (AI) therapy and cognitive function (over a 6-month period) in a cohort of patients aged ≥ 60 years compared with an age-matched healthy control group, and it evaluated changes in regional cerebral metabolism as measured by positron emission tomography (PET) scans of the brain done in a subset of the patient cohort. PATIENTS AND METHODS Thirty-five patients (32 evaluable) and 35 healthy controls were recruited to this study. Patients with breast cancer completed a neuropsychological battery, self-reported memory questionnaire, and geriatric assessment before initiation of AI therapy and again 6 months later. Age-matched healthy control participants completed the same assessments at the same time points as the patient group. RESULTS No significant decline in cognitive function was seen among individuals receiving an AI from pretreatment to 6 months later compared with healthy controls. In the PET cohort over the same period, both standardized volume of interest and statistical parametric mapping analyses detected specific changes in metabolic activity between baseline and follow-up uniquely in the AI patients, most significantly in the medial temporal lobes. CONCLUSION Although patients undergoing AI treatment had few changes in neuropsychological performance compared with healthy controls over a 6-month period, regionally specific changes in cerebral metabolic activity were identified during this interval in the patient group. Additional longitudinal follow-up is needed to understand the potential clinical implications of these findings.
Collapse
Affiliation(s)
- Arti Hurria
- City of Hope National Medical Center, Duarte, CA.
| | | | | | - Thehang Luu
- City of Hope National Medical Center, Duarte, CA
| | - George Somlo
- City of Hope National Medical Center, Duarte, CA
| | | | - Rupal Ramani
- City of Hope National Medical Center, Duarte, CA
| | - Kurt Hansen
- City of Hope National Medical Center, Duarte, CA
| | - Tao Feng
- City of Hope National Medical Center, Duarte, CA
| | | | | | | |
Collapse
|
26
|
VandeVrede L, Abdelhamid R, Qin Z, Choi J, Piyankarage S, Luo J, Larson J, Bennett BM, Thatcher GRJ. An NO donor approach to neuroprotective and procognitive estrogen therapy overcomes loss of NO synthase function and potentially thrombotic risk. PLoS One 2013; 8:e70740. [PMID: 23976955 PMCID: PMC3745399 DOI: 10.1371/journal.pone.0070740] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Accepted: 06/28/2013] [Indexed: 01/16/2023] Open
Abstract
Selective estrogen receptor modulators (SERMs) are effective therapeutics that preserve favorable actions of estrogens on bone and act as antiestrogens in breast tissue, decreasing the risk of vertebral fractures and breast cancer, but their potential in neuroprotective and procognitive therapy is limited by: 1) an increased lifetime risk of thrombotic events; and 2) an attenuated response to estrogens with age, sometimes linked to endothelial nitric oxide synthase (eNOS) dysfunction. Herein, three 3(rd) generation SERMs with similar high affinity for estrogen receptors (ERα, ERβ) were studied: desmethylarzoxifene (DMA), FDMA, and a novel NO-donating SERM (NO-DMA). Neuroprotection was studied in primary rat neurons exposed to oxygen glucose deprivation; reversal of cholinergic cognitive deficit was studied in mice in a behavioral model of memory; long term potentiation (LTP), underlying cognition, was measured in hippocampal slices from older 3×Tg Alzheimer's transgenic mice; vasodilation was measured in rat aortic strips; and anticoagulant activity was compared. Pharmacologic blockade of GPR30 and NOS; denudation of endothelium; measurement of NO; and genetic knockout of eNOS were used to probe mechanism. Comparison of the three chemical probes indicates key roles for GPR30 and eNOS in mediating therapeutic activity. Procognitive, vasodilator and anticoagulant activities of DMA were found to be eNOS dependent, while neuroprotection and restoration of LTP were both shown to be dependent upon GPR30, a G-protein coupled receptor mediating estrogenic function. Finally, the observation that an NO-SERM shows enhanced vasodilation and anticoagulant activity, while retaining the positive attributes of SERMs even in the presence of NOS dysfunction, indicates a potential therapeutic approach without the increased risk of thrombotic events.
Collapse
Affiliation(s)
- Lawren VandeVrede
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Ramy Abdelhamid
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Zhihui Qin
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jaewoo Choi
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Sujeewa Piyankarage
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Jia Luo
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - John Larson
- Department of Psychiatry, Neuropsychiatric Institute, University of Illinois at Chicago, Chicago, Illinois, United States of America
| | - Brian M. Bennett
- Department of Biomedical and Molecular Sciences, Faculty of Health Sciences, Queen's University, Kingston, Ontario, Canada
| | - Gregory R. J. Thatcher
- Department of Medicinal Chemistry and Pharmacognosy, University of Illinois College of Pharmacy, University of Illinois at Chicago, Chicago, Illinois, United States of America
| |
Collapse
|
27
|
Winkler JM, Fox HS. Transcriptome meta-analysis reveals a central role for sex steroids in the degeneration of hippocampal neurons in Alzheimer's disease. BMC SYSTEMS BIOLOGY 2013; 7:51. [PMID: 23803348 PMCID: PMC3702487 DOI: 10.1186/1752-0509-7-51] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2012] [Accepted: 06/19/2013] [Indexed: 11/12/2022]
Abstract
Background Alzheimer’s disease is the most prevalent form of dementia. While a number of transcriptomic studies have been performed on the brains of Alzheimer’s specimens, no clear picture has emerged on the basis of neuronal transcriptional alterations linked to the disease. Therefore we performed a meta-analysis of studies comparing hippocampal neurons in Alzheimer’s disease to controls. Results Homeostatic processes, encompassing control of gene expression, apoptosis, and protein synthesis, were identified as disrupted during Alzheimer’s disease. Focusing on the genes carrying out these functions, a protein-protein interaction network was produced for graph theory and cluster exploration. This approach identified the androgen and estrogen receptors as key components and regulators of the disrupted homeostatic processes. Conclusions Our systems biology approach was able to identify the importance of the androgen and estrogen receptors in not only homeostatic cellular processes but also the role of other highly central genes in Alzheimer’s neuronal dysfunction. This is important due to the controversies and current work concerning hormone replacement therapy in postmenopausal women, and possibly men, as preventative approaches to ward off this neurodegenerative disorder.
Collapse
Affiliation(s)
- Jessica M Winkler
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | |
Collapse
|
28
|
Espeland MA, Shumaker SA, Limacher M, Rapp SR, Bevers TB, Barad DH, Coker LH, Gaussoin SA, Stefanick ML, Lane DS, Maki PM, Resnick SM. Relative effects of tamoxifen, raloxifene, and conjugated equine estrogens on cognition. J Womens Health (Larchmt) 2013; 19:371-9. [PMID: 20136553 DOI: 10.1089/jwh.2009.1605] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To compare the relative effects of conjugated equine estrogens (CEE), raloxifene, and tamoxifen therapies on cognition among women aged > or =65 years. METHODS Annual Modified Mini-Mental State (3MS) examinations were used to assess global cognitive function in the two randomized placebo-controlled clinical trials of CEE therapies of the Women's Health Initiative Memory Study (WHIMS) and the Cognition in the Study of Tamoxifen and Raloxifene (CoSTAR). Analyses were limited to women who had 3MS testing at baseline and the first 3 years of follow-up and, because of potential ethnic-related differences between studies, to Caucasian women (WHIMS n = 6211, CoSTAR n = 250). Covariate adjustment was used to compare the postrandomization mean 3MS scores among the three active therapies with placebo therapy while controlling for differences between groups with respect to dementia risk factors. RESULTS At baseline, the average (SD) 3MS scores by group were 95.24 (4.28) for placebo, 95.19 (4.33) for CEE, 94.60 (4.76) for raloxifene, and 95.02 (4.03) for tamoxifen. Compared with placebo, each active therapy was associated with a small mean relative deficit in 3MS scores of < or =0.5 units, which was fairly consistent between women with and without prior hysterectomy. Relative deficits were slightly greater for tamoxifen (p = 0.001) and less marked for raloxifene (p = 0.06) and CEE (p = 0.02) therapies. Relative deficits appeared to be greater among women with lower baseline 3MS scores: p = 0.009 (tamoxifen), p = 0.08 (raloxifene), and p = 0.03 (CEE). CONCLUSIONS Although unmeasured differences between trials may have confounded analyses, these findings raise the possibility that both tamoxifen and raloxifene adversely affect cognitive function in older women; however, the magnitude of the effect is small, and the long-term consequences are unknown.
Collapse
Affiliation(s)
- Mark A Espeland
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, North Carolina 27157, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Buwalda B, Schagen SB. Is basic research providing answers if adjuvant anti-estrogen treatment of breast cancer can induce cognitive impairment? Life Sci 2013; 93:581-8. [PMID: 23353876 DOI: 10.1016/j.lfs.2012.12.012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2012] [Revised: 11/27/2012] [Accepted: 12/21/2012] [Indexed: 12/23/2022]
Abstract
Adjuvant treatment of cancer by chemotherapy is associated with cognitive impairment in some cancer survivors. Breast cancer patients are frequently also receiving endocrine therapy with selective estrogen receptor modulators (SERMs) and/or aromatase inhibitors (AIs) to suppress the growth of estradiol sensitive breast tumors. Estrogens are well-known, however, to target brain areas involved in the regulation of cognitive behavior. In this review clinical and basic preclinical research is reviewed on the actions of estradiol, SERMs and AIs on brain and cognitive functioning to see if endocrine therapy potentially induces cognitive impairment and in that respect may contribute to the detrimental effects of chemotherapy on cognitive performance in breast cancer patients. Although many clinical studies may be underpowered to detect changes in cognitive function, current basic and clinical reports suggest that there is little evidence that AIs may have a lasting detrimental effect on cognitive performance in breast cancer patients. The clinical data on SERMs are not conclusive, but some studies do suggest that tamoxifen administration may form a risk for cognitive functioning particularly in older women. An explanation may come from basic preclinical research which indicates that tamoxifen often acts agonistic in the absence of estradiol but antagonistic in the presence of endogenous estradiol. It could be hypothesized that the negative effects of tamoxifen in older women is related to the so-called window of opportunity for estrogen. Administration of SERMs beyond this so-called window of opportunity may not be effective or might even have detrimental effects similar to estradiol.
Collapse
Affiliation(s)
- Bauke Buwalda
- Behavioral Physiology, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands.
| | | |
Collapse
|
30
|
Acevedo SF. Why, After Chemotherapy, is it Necessary to Assess Memory Using Translational Testing? BREAST CANCER-BASIC AND CLINICAL RESEARCH 2012; 6:181-90. [PMID: 23226022 PMCID: PMC3512448 DOI: 10.4137/bcbcr.s10293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
As the number of cancer survivors rises, so does the importance of understanding what happens post-chemotherapy. The evidence is clear that chemotherapy affects not only cancer cells, but also healthy cells including neurons, leading to long-term cognitive dysfunction in a large portion of survivors. In order to understand the mechanism of action and in the hope of reducing the potential neurocognitive side effects of chemotherapy, pre-clinical testing should be used more effectively. However, the field is lacking translation from clinical studies to animal models. Spatial learning and memory paradigms based on the water maze, the most commonly used rodent model, are available for translational testing in humans and could overcome this weakness. There is an overwhelming need in the field to understand whether the water maze is an adequate model for post-chemotherapy impairments or whether other paradigms should be used. This is of great importance for the understanding of the mechanisms, side effects of new drugs, appropriate pharmacotherapy, and confounding factors related to chemotherapy treatment regiments. This review is very important to both basic scientists and clinicians determining how translational paradigms are critical to future cancer research, as well as what type of paradigms are appropriate in our technically advancing society.
Collapse
Affiliation(s)
- Summer F Acevedo
- Department of Physiology, Pharmacology, and Toxicology, Psychology Program, Ponce School of Medicine and Health Sciences, Ponce, Puerto Rico
| |
Collapse
|
31
|
Deal CL, Draper MW. Raloxifene: a selective estrogen-receptor modulator for postmenopausal osteoporosis - a clinical update on efficacy and safety. ACTA ACUST UNITED AC 2012; 2:199-210. [PMID: 19803890 DOI: 10.2217/17455057.2.2.199] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Selective estrogen-receptor modulators are molecules with specific estrogen-receptor binding affinity. Each selective estrogen-receptor modulator induces a unique conformation in the ligand-receptor complex, which leads to transcriptional activation and/or inhibition. Raloxifene 60 mg/day, a benzothiophene selective estrogen-receptor modulator, is approved for the prevention and treatment of postmenopausal osteoporosis. This article provides an update on new studies and further analyses of clinical trial data for raloxifene. The Multiple Outcomes of Raloxifene Evaluation (MORE) trial of women with osteoporosis has described the efficacy of raloxifene in decreasing vertebral fracture risk over 4 years. The Continuing Outcomes Relevant to Evista((R)) (CORE) trial, designed to assess the effects of raloxifene on breast cancer prevention, is a 4-year continuation of MORE. The skeletal and cardiovascular effects of raloxifene in the CORE study were similar to those observed in MORE. The relative risk of developing breast cancer was significantly decreased in women treated with raloxifene, compared with placebo, after 4 years in MORE and 8 years in the CORE trial. The incidence of uterine bleeding, endometrial hyperplasia and endometrial cancer was similar between raloxifene and placebo after 8 years of treatment. Raloxifene use is associated with a higher incidence of hot flashes and leg cramps, and an increased risk of venous thromboembolic events.
Collapse
Affiliation(s)
- Chad L Deal
- Center for Osteoporosis and Metabolic Bone Disease, Department of Rheumatic and Immunology Diseases/A50, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA.
| | | |
Collapse
|
32
|
Maki PM, Henderson VW. Hormone therapy, dementia, and cognition: the Women's Health Initiative 10 years on. Climacteric 2012; 15:256-62. [PMID: 22612612 DOI: 10.3109/13697137.2012.660613] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Principal findings on dementia from the Women's Health Initiative Memory Study (WHIMS) showed that conjugated equine estrogens plus medroxyprogesterone acetate (CEE/MPA) increase dementia risk in women aged 65 years and above, but not risk of mild cognitive impairment. The dementia finding was unexpected, given consistent observational evidence that associates use of estrogen-containing hormone therapy with reduced risk of Alzheimer's disease. It remains controversial whether hormone use by younger postmenopausal women near the time of menopause reduces dementia risk or whether WHIMS findings should be generalized to younger women. Given the challenges of conducting a primary prevention trial to address that question, it is helpful to consider the impact of hormone therapy on cognitive test performance, particularly verbal memory, for its own sake and as a proxy for dementia risk. The WHI Study of Cognitive Aging (WHISCA) showed that CEE/MPA worsened verbal memory, whereas CEE alone had no influence on cognition. These findings have been replicated in several randomized, clinical trials. The apparent negative effect of CEE/MPA on verbal memory does not appear to be age-dependent. Additional investigations are needed to understand the impact of other hormonally active compounds on dementia and cognitive outcomes.
Collapse
Affiliation(s)
- P M Maki
- Department of Psychiatry, University of Illinois at Chicago, Chicago, Illinois, USA
| | | |
Collapse
|
33
|
Okereke OI, Rosner BA, Kim DH, Kang JH, Cook NR, Manson JE, Buring JE, Willett WC, Grodstein F. Dietary fat types and 4-year cognitive change in community-dwelling older women. Ann Neurol 2012; 72:124-34. [PMID: 22605573 PMCID: PMC3405188 DOI: 10.1002/ana.23593] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 02/15/2012] [Accepted: 03/15/2012] [Indexed: 11/06/2022]
Abstract
OBJECTIVE A study was undertaken to relate dietary fat types to cognitive change in healthy community-based elders. METHODS Among 6,183 older participants in the Women's Health Study, we related intake of major fatty acids (saturated [SFA], monounsaturated [MUFA], total polyunsaturated [PUFA], trans-unsaturated) to late-life cognitive trajectory. Serial cognitive testing, conducted over 4 years, began 5 years after dietary assessment. Primary outcomes were global cognition (averaging tests of general cognition, verbal memory, and semantic fluency) and verbal memory (averaging tests of recall). We used analyses of response profiles and logistic regression to estimate multivariate-adjusted differences in cognitive trajectory and risk of worst cognitive change (worst 10%) by fat intake. RESULTS Higher SFA intake was associated with worse global cognitive (p for linear trend = 0.008) and verbal memory (p for linear trend = 0.01) trajectories. There was a higher risk of worst cognitive change, comparing highest versus lowest SFA quintiles; the multivariate-adjusted odds ratio (OR) with 95% confidence interval (CI) was 1.64 (1.04-2.58) for global cognition and 1.65 (1.04-2.61) for verbal memory. By contrast, higher MUFA intake was related to better global cognitive (p for linear trend < 0.001) and verbal memory (p for linear trend = 0.009) trajectories, and lower OR (95% CI) of worst cognitive change in global cognition (0.52 [0.31-0.88]) and verbal memory (0.56 [0.34-0.94]). Total fat, PUFA, and trans-fat intakes were not associated with cognitive trajectory. INTERPRETATION Higher SFA intake was associated with worse global cognitive and verbal memory trajectories, whereas higher MUFA intake was related to better trajectories. Thus, different consumption levels of the major specific fat types, rather than total fat intake itself, appeared to influence cognitive aging.
Collapse
Affiliation(s)
- Olivia I Okereke
- Channing Laboratory, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Frick KM. Building a better hormone therapy? How understanding the rapid effects of sex steroid hormones could lead to new therapeutics for age-related memory decline. Behav Neurosci 2012; 126:29-53. [PMID: 22289043 DOI: 10.1037/a0026660] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A wealth of data collected in recent decades has demonstrated that ovarian sex-steroid hormones, particularly 17β-estradiol (E2), are important trophic factors that regulate the function of cognitive regions of the brain such as the hippocampus. The loss of hormone cycling at menopause is associated with cognitive decline and dementia in women, and the onset of memory decline in animal models. However, hormone therapy is not currently recommended to prevent or treat cognitive decline, in part because of its detrimental side effects. In this article, it is proposed that investigations of the rapid effects of E2 on hippocampal function be used to further the design of new drugs that mimic the beneficial effects of E2 on memory without the side effects of current therapies. A conceptual model is presented for elucidating the molecular and biochemical mechanisms through which sex-steroid hormones modulate memory, and a specific hypothesis is proposed to account for the rapid memory-enhancing effects of E2. Empirical support for this hypothesis is discussed as a means of stimulating the consideration of new directions for the development of hormone-based therapies to preserve memory function in menopausal women.
Collapse
Affiliation(s)
- Karyn M Frick
- Department of Psychology, University of Wisconsin-Milwaukee, 2441 East Hartford Avenue, Milwaukee, WI 53211, USA.
| |
Collapse
|
35
|
Amariglio RE, Townsend MK, Grodstein F, Sperling RA, Rentz DM. Specific subjective memory complaints in older persons may indicate poor cognitive function. J Am Geriatr Soc 2011; 59:1612-7. [PMID: 21919893 DOI: 10.1111/j.1532-5415.2011.03543.x] [Citation(s) in RCA: 147] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES To examine the association between the type and number of subjective memory complaints (SMCs) and performance on objective cognitive tests. DESIGN Cross-sectional. SETTING Nurses' Health Study. PARTICIPANTS Sixteen thousand nine hundred sixty-four women (mean age 74) who provided information on SMCs. MEASUREMENTS Telephone cognitive assessments and seven questions regarding SMCs were administered. Cognitive impairment was defined as a score of less than 31 on the Telephone Interview for Cognitive Status (TICS) and below the 10th percentile on other cognitive measures. To assess associations with SMCs, multivariable logistic regression was used to calculate odds ratios for cognitive impairment and multivariable linear regression to calculate mean differences in cognitive test scores, adjusting for age and depressive symptoms. RESULTS Some SMCs, such as trouble following a group conversation or finding one's way around familiar streets, were more highly associated than others with odds of cognitive impairment. The complaint of forgetting things from one second to the next, generally considered part of normal aging, was not associated with cognitive impairment. In addition, there were strong, linear trends of increasingly worse scores on cognitive tests with increasing numbers of memory complaints. For each additional SMC endorsed, the odds of cognitive impairment increased approximately 20% when each SMC was weighted equally. CONCLUSION SMCs are associated with objective cognitive status and may be considered by primary care physicians in determining whether follow-up is warranted.
Collapse
Affiliation(s)
- Rebecca England Amariglio
- Department of Neurology, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
36
|
Park MK, Satoh N, Kumashiro M. Effects of menopausal hot flashes on mental workload. INDUSTRIAL HEALTH 2011; 49:566-574. [PMID: 21804271 DOI: 10.2486/indhealth.ms1222] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
This study examined the differences in psychophysiological responses during mental task performance between women with (Group S) and without (Group A) menopausal hot flashes. Twelve women who reported experiencing daily moderate or severe menopausal hot flashes (Group S) and twelve women who reported having no hot flashes (Group A) participated in a mental arithmetic (Task) and a control (Non-task) experiment that occurred twice during 30 min. Although Group S experienced frequent hot flashes during mental arithmetic task, no significant differences between the two groups emerged for the percentage of correct responses and reaction time, and cardiovascular and thermoregulatory responses. However, the STAI state anxiety score was significantly higher in Group S than in Group A during both rest and mental tasks, and stress-related cortisol secretion showed a tendency to increase in Group S compared with Group A after task. The present study indicates that there were no significant differences in physiological responses and cognitive performance between women with and without menopausal hot flashes during mental arithmetic, but women with menopausal hot flashes might perceive higher psychological stress during rest and mental arithmetic tasks than asymptomatic women.
Collapse
Affiliation(s)
- Mi Kyong Park
- Division of Management Systems Engineering, Faculty of System Design, Tokyo Metropolitan University, Hino, Japan.
| | | | | |
Collapse
|
37
|
Loibl S, Lintermans A, Dieudonné A, Neven P. Management of menopausal symptoms in breast cancer patients. Maturitas 2011; 68:148-54. [DOI: 10.1016/j.maturitas.2010.11.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2010] [Revised: 11/08/2010] [Accepted: 11/09/2010] [Indexed: 10/18/2022]
|
38
|
Abstract
Bone loss with aging places postmenopausal women at a higher risk for osteoporosis and its consequences such as fractures, pain, disability, and increased morbidity and mortality. Approximately 200 million patients worldwide are affected. The Third National Health and Nutrition Examination Survey (NHANES III) estimated that up to 18% of US women aged 50 and older have osteoporosis and up to 50% have osteopenia. Greater than 2 million osteoporotic related fractures occurred in the United States with direct healthcare costs exceeding $17 billion. Hormone Replacement Therapy (HRT) was a popular option for postmenopausal women before the Women's Health Initiative (WHI). Several agents are available in the U.S., including bisphosphonates, hormone therapy, calcitonin, parathyroid hormone and the selective estrogen receptor modulator (SERM) raloxifene. There are concerns about long term safety and compliance. Therefore, other agents are under investigation. SERMs are a diverse group of agents that bind to the estrogen receptor and each SERM appears to have a unique set of clinical responses, which are not always consistent with the typical responses seen with other SERMs. This article will discuss the SERMs approved in the United States, tamoxifene and raloxifene, and investigational SERMs. The ideal SERM would include the beneficial effects of estrogen in bone, heart and the central nervous system, with neutral or antagonistic effects in tissues where estrogen effects are undesirable(breast and endometrium). A new target in treating postmenopausal osteoporosis is the tissue estrogen complex or the pairing of a SERM with a conjugated estrogen known as a tissue selective estrogen complex (TSEC). This novel approach is currently being evaluated with bazodoxifene which could yield the beneficial effects of estrogens and SERMS, while potentially being more tolerable and safer than either therapy alone.
Collapse
|
39
|
Tirona MT, Sehgal R, Ballester O. Prevention of breast cancer (Part II): risk reduction strategies. Cancer Invest 2010; 28:1070-7. [PMID: 20932221 DOI: 10.3109/07357907.2010.512593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Until recently, the best protection against breast cancer mortality was early diagnosis through mammographic screening. However, the possibility that breast cancer in some cases can be prevented has come to light over the past 30 years. Various risk reduction strategies of breast cancer have been explored including lifestyle modification, prophylactic surgeries, and the use of chemopreventive agents. This article is the second portion of a two-part series on breast cancer prevention, and will focus its discussion on the available risk reduction interventions that have been shown to prevent breast cancer in women considered high risk for the disease. (See Part I in Cancer Investigation, 28:743–750, 2010)
Collapse
Affiliation(s)
- Maria Tria Tirona
- Edwards Comprehensive Cancer Center and Joan C. Edwards School of Medicine at Marshall University, Huntington, West Virginia 25701, USA.
| | | | | |
Collapse
|
40
|
Vodermaier A. Breast cancer treatment and cognitive function: the current state of evidence, underlying mechanisms and potential treatments. ACTA ACUST UNITED AC 2010; 5:503-16. [PMID: 19702450 DOI: 10.2217/whe.09.36] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Within the last decade, several studies have investigated whether adjuvant treatment of breast cancer affects cognitive function. A number of prospective studies have demonstrated inconsistent results regarding whether chemotherapy affects cognitive function. Approximately half of the studies demonstrated subtle cognitive decline in a wide range of domains among some breast cancer patients following chemotherapy, and half did not. Concomitant changes in brain structure and function have been identified in neuroimaging and neurophysiologic studies. Estrogenic therapy has been specifically associated with deterioration in verbal memory and processing speed. However, evidence is mostly based on smaller studies with cross-sectional data. Breast cancer patients who underwent both chemotherapy and estrogenic therapy showed the most deterioration and the most persistent decline in cognitive function. Since cognitive impairment is subtle, if evident at all, discrepant findings are due to hormonal, physiological, psychological or temporal confounding variables and differences in study design. Neuropsychological training has been demonstrated to improve cognitive dysfunction experienced by breast cancer patients after chemotherapy. Future research may examine the unique impact of endocrine therapy on cognitive function with prospective, controlled trials, as well as the role of further confounding variables (e.g., menopausal status, cytokine deregulation, cortisol and concurrent medication).
Collapse
Affiliation(s)
- Andrea Vodermaier
- Department of Psychology, University of British Columbia, 2136 West Mall, Vancouver, BC, Canada.
| |
Collapse
|
41
|
Cognitive changes associated with endocrine therapy for breast cancer. Maturitas 2010; 67:209-14. [PMID: 20688441 DOI: 10.1016/j.maturitas.2010.07.004] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2010] [Revised: 07/09/2010] [Accepted: 07/09/2010] [Indexed: 01/22/2023]
Abstract
Endocrine therapy in the setting of breast cancer has undoubtedly advanced clinical outcomes in this disease, but treatment with endocrine therapy is accompanied by a wide spectrum of side effects. It is of prime importance to understand and characterize these toxicities to facilitate clinical decision-making. Somewhat surprisingly, there is a relative paucity of data pertaining to cognitive changes associated with endocrine therapy. In this article we review cognitive associated with two classes of endocrine therapy: (1) selective estrogen receptor modulators (SERMs; tamoxifen and raloxifene) and (2) aromatase inhibitors (AIs; anastrozole, letrozole, and exemestane). Companion studies to the Multiple Outcome of Raloxifene Evaluation (MORE), the Study of Tamoxifen and Raloxifene (STAR) and National Surgical Adjuvant Breast and Bowel Project (NSABP) P-1 trials provide relevant data to understand the effect of SERMs on cognition. In contrast, substudies of the Arimidex, Tamoxifen Alone or in Combination (ATAC), Tamoxifen and Exemestane Adjuvant Multinational (TEAM) and Breast International Group (BIG) 1-98 trials juxtapose cognitive effects of AIs against those of tamoxifen. These and other studies are examined herein to provide a comprehensive overview of the effect of endocrine therapy on cognition.
Collapse
|
42
|
Kadan-Lottick NS, Zeltzer LK, Liu Q, Yasui Y, Ellenberg L, Gioia G, Robison LL, Krull KR. Neurocognitive functioning in adult survivors of childhood non-central nervous system cancers. J Natl Cancer Inst 2010; 102:881-93. [PMID: 20458059 DOI: 10.1093/jnci/djq156] [Citation(s) in RCA: 145] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND We sought to measure self-reported neurocognitive functioning among survivors of non-central nervous system (CNS) childhood cancers, overall and compared with a sibling cohort, and to identify factors associated with worse functioning. METHODS In a retrospective cohort study, 5937 adult survivors of non-CNS cancers and 382 siblings completed a validated neuropsychological instrument with subscales in task efficiency, emotional regulation, organization, and memory. Scores were converted to T scores; scores in the worst 10% of siblings' scores (ie, T score > or =63) were defined as impaired. Non-CNS cancer survivors and siblings were compared with multivariable linear regression and log-binomial regression. Among survivors, log-binomial models assessed the association of patient and treatment factors with neurocognitive dysfunction. All statistical tests were two-sided. RESULTS Non-CNS cancer survivors had similar or slightly worse (<0.5 standard deviation) mean test scores for all four subscales than siblings. However, frequencies of impaired survivors were approximately 50% higher than siblings in task efficiency (13.0% of survivors vs 7.3% of siblings), memory (12.5% vs 7.6%), and emotional regulation (21.2% vs 14.4%). Impaired task efficiency was most often identified in patients with acute lymphoblastic leukemia who received cranial radiation therapy (18.1% with impairment), myeloid leukemia who received cranial radiation therapy (21.2%), and non-Hodgkin lymphoma (13.9%). In adjusted analysis, diagnosis age of younger than 6 years, female sex, cranial radiation therapy, and hearing impairment were associated with impairment. CONCLUSION A statistically and clinically significantly higher percentage of self-reported neurocognitive impairment was found among survivors of non-CNS cancers than among siblings.
Collapse
Affiliation(s)
- Nina S Kadan-Lottick
- MSPH, Section of Pediatric Hematology-Oncology, Yale University School of Medicine, 333 Cedar St, LMP 2073, PO Box 208064, New Haven, CT 06525, USA.
| | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Cognitive performance test research and clinical perils. Menopause 2010; 17:239-41. [DOI: 10.1097/gme.0b013e3181cefd1c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
Abstract
OBJECTIVE To review the relation in midlife and beyond between estrogen exposures and episodic memory in women. BACKGROUND Episodic memory performance declines with usual aging, and impairments in episodic memory often portend the development of Alzheimer disease. In the laboratory, estradiol influences hippocampal function and animal learning. However, it is controversial whether estrogens affect memory after a woman's reproductive years. METHOD Focused literature review, including a summary of a systematic search of clinical trials of estrogens in which outcomes included an objective measure of episodic memory. RESULTS The natural menopause transition is not associated with the objective changes in episodic memory. Strong clinical trial evidence indicates that initiating estrogen-containing hormone therapy after the age of about 60 years does not benefit episodic memory. Clinical trial findings in middle-aged women before the age of 60 years are limited by smaller sample sizes and shorter treatment durations, but these also do not indicate substantial memory effects. Limited short-term evidence, however, suggests that estrogens may improve verbal memory after surgical menopause. Although hormone therapy initiation in old age increases dementia risk, observational studies raise the question of an early critical window during which midlife estrogen therapy reduces late-life Alzheimer disease. However, almost no data address whether midlife estrogen therapy affects episodic memory in old age. CONCLUSIONS Episodic memory is not substantially impacted by the natural menopause transition or improved by the use of estrogen-containing hormone therapy after the age of 60 years. Further research is needed to determine whether outcomes differ after surgical menopause or whether episodic memory later in life is modified by midlife estrogenic exposures.
Collapse
Affiliation(s)
- Victor W Henderson
- Departments of Health Research and Policy (Epidemiology), and of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305-5405, USA.
| |
Collapse
|
46
|
Legault C, Maki PM, Resnick SM, Coker L, Hogan P, Bevers TB, Shumaker SA. Effects of tamoxifen and raloxifene on memory and other cognitive abilities: cognition in the study of tamoxifen and raloxifene. J Clin Oncol 2009; 27:5144-52. [PMID: 19770382 DOI: 10.1200/jco.2008.21.0716] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To compare the effects of two selective estrogen receptor modulators, tamoxifen and raloxifene, on global and domain-specific cognitive function. PATIENTS AND METHODS The National Surgical Adjuvant Breast and Bowel Project's Study of Tamoxifen and Raloxifene (STAR) study was a randomized clinical trial of tamoxifen 20 mg/d or raloxifene 60 mg/d in healthy postmenopausal women at increased risk of breast cancer. The 1,498 women who were randomly assigned in STAR were age 65 years and older, were not diagnosed with dementia, and were enrolled onto the Cognition in the Study of Tamoxifen and Raloxifene (Co-STAR) trial, beginning 18 months after STAR enrollment started. A cognitive test battery modeled after the one used in the Women's Health Initiative Study of Cognitive Aging (WHISCA) was administered. Technicians were centrally trained to administer the battery and recertified every 6 months. Analyses were conducted on all participants and on 273 women who completed the first cognitive battery before they started taking their medications. RESULTS Overall, there were no significant differences in adjusted mean cognitive scores between the two treatment groups across visits. There were significant time effects across the three visits for some of the cognitive measures. Similar results were obtained for the subset of women with true baseline measures. CONCLUSION Tamoxifen and raloxifene are associated with similar patterns of cognitive function in postmenopausal women at increased risk of breast cancer. Future comparisons between these findings and patterns of cognitive function in hormone therapy and placebo groups in WHISCA should provide additional insights into the effects of tamoxifen and raloxifene on cognitive function in older women.
Collapse
Affiliation(s)
- Claudine Legault
- Division of Public Health Sciences, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| | | | | | | | | | | | | |
Collapse
|
47
|
Compton J, Murphy D. Imaging the brain in healthy postmenopausal users and non-users of hormone replacement therapy. Climacteric 2009. [DOI: 10.1080/cmt.6.3.180.183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
48
|
Pike CJ, Carroll JC, Rosario ER, Barron AM. Protective actions of sex steroid hormones in Alzheimer's disease. Front Neuroendocrinol 2009; 30:239-58. [PMID: 19427328 PMCID: PMC2728624 DOI: 10.1016/j.yfrne.2009.04.015] [Citation(s) in RCA: 373] [Impact Index Per Article: 24.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Revised: 04/25/2009] [Accepted: 04/28/2009] [Indexed: 12/19/2022]
Abstract
Risk for Alzheimer's disease (AD) is associated with age-related loss of sex steroid hormones in both women and men. In post-menopausal women, the precipitous depletion of estrogens and progestogens is hypothesized to increase susceptibility to AD pathogenesis, a concept largely supported by epidemiological evidence but refuted by some clinical findings. Experimental evidence suggests that estrogens have numerous neuroprotective actions relevant to prevention of AD, in particular promotion of neuron viability and reduction of beta-amyloid accumulation, a critical factor in the initiation and progression of AD. Recent findings suggest neural responsiveness to estrogen can diminish with age, reducing neuroprotective actions of estrogen and, consequently, potentially limiting the utility of hormone therapies in aged women. In addition, estrogen neuroprotective actions are also modulated by progestogens. Specifically, continuous progestogen exposure is associated with inhibition of estrogen actions whereas cyclic delivery of progestogens may enhance neural benefits of estrogen. In recent years, emerging literature has begun to elucidate a parallel relationship of sex steroid hormones and AD risk in men. Normal age-related testosterone loss in men is associated with increased risk to several diseases including AD. Like estrogen, testosterone has been established as an endogenous neuroprotective factor that not only increases neuronal resilience against AD-related insults, but also reduces beta-amyloid accumulation. Androgen neuroprotective effects are mediated both directly by activation of androgen pathways and indirectly by aromatization to estradiol and initiation of protective estrogen signaling mechanisms. The successful use of hormone therapies in aging men and women to delay, prevent, and or treat AD will require additional research to optimize key parameters of hormone therapy and may benefit from the continuing development of selective estrogen and androgen receptor modulators.
Collapse
Affiliation(s)
- Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| | | | | | | |
Collapse
|
49
|
Visvanathan K, Chlebowski RT, Hurley P, Col NF, Ropka M, Collyar D, Morrow M, Runowicz C, Pritchard KI, Hagerty K, Arun B, Garber J, Vogel VG, Wade JL, Brown P, Cuzick J, Kramer BS, Lippman SM. American society of clinical oncology clinical practice guideline update on the use of pharmacologic interventions including tamoxifen, raloxifene, and aromatase inhibition for breast cancer risk reduction. J Clin Oncol 2009; 27:3235-58. [PMID: 19470930 DOI: 10.1200/jco.2008.20.5179] [Citation(s) in RCA: 220] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PURPOSE To update the 2002 American Society of Clinical Oncology guideline on pharmacologic interventions for breast cancer (BC) risk reduction. METHODS A literature search identified relevant randomized trials published since 2002. Primary outcome of interest was BC incidence (invasive and noninvasive). Secondary outcomes included BC mortality, adverse events, and net health benefits. An expert panel reviewed the literature and developed updated consensus guidelines. Results Seventeen articles met inclusion criteria. In premenopausal women, tamoxifen for 5 years reduces the risk of BC for at least 10 years, particularly estrogen receptor (ER) -positive invasive tumors. Women < or = 50 years of age experience fewer serious side effects. Vascular and vasomotor events do not persist post-treatment across all ages. In postmenopausal women, raloxifene and tamoxifen reduce the risk of ER-positive invasive BC with equal efficacy. Raloxifene is associated with a lower risk of thromboembolic disease, benign uterine conditions, and cataracts than tamoxifen in postmenopausal women. No evidence exists establishing whether a reduction in BC risk from either agent translates into reduced BC mortality. Recommendations In women at increased risk for BC, tamoxifen (20 mg/d for 5 years) may be offered to reduce the risk of invasive ER-positive BC, with benefits for at least 10 years. In postmenopausal women, raloxifene (60 mg/d for 5 years) may also be considered. Use of aromatase inhibitors, fenretinide, or other selective estrogen receptor modulators to lower BC risk is not recommended outside of a clinical trial. Discussion of risks and benefits of preventive agents by health providers is critical to patient decision making.
Collapse
Affiliation(s)
- Kala Visvanathan
- Cancer Policy and Clinical Affairs, 2318 Mill Rd, Suite 800, Alexandria, VA 22314, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Estrogen-containing hormone therapy initiated during late postmenopause does not improve episodic memory (an important early symptom of Alzheimer's disease), and it increases dementia risk. Cognitive consequences of exogenous estrogen exposures during midlife are less certain. Observational evidence implies that use of hormone therapy at a younger age close to the time of menopause may reduce risk of Alzheimer's disease later in life. However, there are concerns that observational findings may be systematically biased. Partial insight on this critical issue may be gleaned from results of ongoing clinical trials involving midlife postmenopausal women (Early versus Late Intervention Trial with Estrogen; Kronos Early Estrogen Prevention Study). The effects of exogenous midlife estrogen exposures and Alzheimer risk can also be approached through better animal models, through carefully designed cohort studies, and through use of surrogate outcomes in randomized controlled trials in midlife women. Selective estrogen receptor modulators have the potential to affect cognitive outcomes and also merit additional study.
Collapse
Affiliation(s)
- Victor W Henderson
- Departments of Health Research & Policy (Epidemiology) and of Neurology & Neurological Sciences, Stanford University, Stanford, California 94305-5405, USA.
| |
Collapse
|