1
|
Maschio M, Perversi F, Maialetti A. Brain tumor-related epilepsy: an overview on neuropsychological, behavioral, and quality of life issues and assessment methodology. Front Neurol 2024; 15:1480900. [PMID: 39722690 PMCID: PMC11668670 DOI: 10.3389/fneur.2024.1480900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 11/25/2024] [Indexed: 12/28/2024] Open
Abstract
Brain tumor-related epilepsy (BTRE) is a rare disease in which brain tumor (BT) and epilepsy overlap simultaneously and can have a negative impact on a patient's neuropsychological, behavioral, and quality of life (QoL) spheres. In this review we (a) addressed the main neuropsychological, behavioral, and QoL issues that may occur in BTRE patients, (b) described how BT, BTRE, and their respective treatments can impact these domains, and (c) identified tools and standardized evaluation methodologies specific for BTRE patients. Neuropsychological disorders and behavioral issues can be direct consequences of BTRE and all related treatments, such as surgery, anti-cancer and anti-seizure medication, corticosteroids, etc., which can alter the structure of specific brain areas and networks, and by emotional aspects reactive to BTRE diagnosis, including the possible loss of autonomy, poor prognosis, and fear of death. Unfortunately, it seems there is a lack of uniformity in assessment methodologies, such as the administration of different batteries of neuropsychological tests, different times, frames, and purposes. Further research is needed to establish causality and deepen our understanding of the interplay between all these variables and our intervention in terms of diagnosis, treatment, psychosocial assessment, and their timing. We propose that the care of these patients to rely on the concepts of "BTRE-induced disability" and "biopsychosocial model" of BTRE, to prompt healthcare providers to handle and monitor BTRE-related psychological and social aspects, as to maintain the patient's best possible QoL.
Collapse
Affiliation(s)
- Marta Maschio
- Center for Tumor-Related Epilepsy, UOSD Neuro-oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | | | - Andrea Maialetti
- Center for Tumor-Related Epilepsy, UOSD Neuro-oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| |
Collapse
|
2
|
Poplawska-Domaszewicz K, Limbachiya N, Qamar M, Batzu L, Jones S, Sauerbier A, Rota S, Lau YH, Chaudhuri KR. Addressing the Ethnicity Gap in Catechol O-Methyl Transferase Inhibitor Trials in Parkinson's Disease: A Review of Available Global Data. J Pers Med 2024; 14:939. [PMID: 39338193 PMCID: PMC11433619 DOI: 10.3390/jpm14090939] [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: 07/15/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
Catechol-O-methyltransferase inhibitors (COMT-Is) have significantly improved the quality of life and symptom management for those at advanced stages of Parkinson's Disease (PD). Given that PD is one of the fastest-growing neurodegenerative diseases worldwide, there is a need to establish a clear framework for the systematic distribution of COMT-Is, considering inter-individual and intra-individual variations in patient response. One major barrier to this is the underrepresentation of ethnic minority participants in clinical trials investigating COMT-Is. To investigate this, we performed a narrative review. We searched PubMed for clinical trials investigating COMT-Is in patients with PD and examined the ethnic diversity of cohorts. A total of 63 articles were identified, with 34 trials found to match our inclusion criteria. Among the 34 trials meeting our inclusion criteria, only 8 reported participants' ethnic backgrounds. Our findings reveal a consistent underrepresentation of ethnic minority groups in trials investigating COMT-Is in PD cohorts-a trend that reflects broader concerns across clinical research. In this review, we explore potential reasons for the underrepresentation of ethnic minorities in clinical trials and propose strategies to address this issue.
Collapse
Affiliation(s)
- Karolina Poplawska-Domaszewicz
- Department of Neurology, Poznan University of Medical Sciences, 60-355 Poznan, Poland
- Basic and Clinical Neuroscience Department, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
| | - Naomi Limbachiya
- Basic and Clinical Neuroscience Department, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Mubasher Qamar
- Basic and Clinical Neuroscience Department, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Lucia Batzu
- Basic and Clinical Neuroscience Department, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Shelley Jones
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Anna Sauerbier
- Basic and Clinical Neuroscience Department, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
- Department of Neurology, University Hospital Cologne, Faculty of Medicine, 50937 Cologne, Germany
| | - Silvia Rota
- Basic and Clinical Neuroscience Department, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| | - Yue Hui Lau
- Division of Neurology, Medical Department, Tengku Ampuan Rahimah Hospital, Klang 41200, Malaysia
| | - K Ray Chaudhuri
- Basic and Clinical Neuroscience Department, The Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, 5 Cutcombe Road, London SE5 9RX, UK
- Parkinson's Foundation Centre of Excellence, King's College Hospital, Denmark Hill, London SE5 9RS, UK
| |
Collapse
|
3
|
Ramírez V, González-Palacios P, González-Domenech PJ, Jaimez-Pérez S, Baca MA, Rodrigo L, Álvarez-Cubero MJ, Monteagudo C, Martínez-González LJ, Rivas A. Influence of Genetic Polymorphisms on Cognitive Function According to Dietary Exposure to Bisphenols in a Sample of Spanish Schoolchildren. Nutrients 2024; 16:2639. [PMID: 39203776 PMCID: PMC11357571 DOI: 10.3390/nu16162639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) like intellectual disability (ID) are highly heritable, but the environment plays an important role. For example, endocrine disrupting chemicals (EDCs), including bisphenol A (BPA) and its analogues, have been termed neuroendocrine disruptors. This study aimed to evaluate the influence of different genetic polymorphisms (SNPs) on cognitive function in Spanish schoolchildren according to dietary bisphenol exposure. METHODS A total of 102 children aged 6-12 years old were included. Ten SNPs in genes involved in brain development, synaptic plasticity, and neurotransmission (BDNF, NTRK2, HTR2A, MTHFR, OXTR, SLC6A2, and SNAP25) were genotyped. Then, dietary exposure to bisphenols (BPA plus BPS) was estimated and cognitive functions were assessed using the WISC-V Spanish form. RESULTS BDNF rs11030101-T and SNAP25 rs363039-A allele carriers scored better on the fluid reasoning domain, except for those inheriting the BDNF rs6265-A allele, who had lower scores. Secondly, relevant SNP-bisphenol interactions existed in verbal comprehension (NTRK2 rs10868235 (p-int = 0.043)), working memory (HTR2A rs7997012 (p-int = 0.002), MTHFR rs1801133 (p-int = 0.026), and OXTR rs53576 (p-int = 0.030)) and fluid reasoning (SLC6A2 rs998424 (p-int = 0.004)). CONCLUSIONS Our findings provide the first proof that exploring the synergistic or additive effects between genetic variability and bisphenol exposure on cognitive function could lead to a better understanding of the multifactorial and polygenic aetiology of NDDs.
Collapse
Affiliation(s)
- Viviana Ramírez
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain; (V.R.); (P.G.-P.); (A.R.)
- GENYO Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS Granada—Avenida de la Ilustración, 114, 18016 Granada, Spain;
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Institute of Nutrition and Food Technology “Jose Mataix Verdú”, Biomedical Research Center, Health Sciences Technological Park, University of Granada, 18016 Granada, Spain
| | - Patricia González-Palacios
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain; (V.R.); (P.G.-P.); (A.R.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | | | | | | | - Lourdes Rodrigo
- Department of Legal Medicine, Toxicology and Physical Anthropology, Faculty of Medicine, University of Granada, 18012 Granada, Spain;
| | - María Jesús Álvarez-Cubero
- GENYO Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS Granada—Avenida de la Ilustración, 114, 18016 Granada, Spain;
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Department of Biochemistry and Molecular Biology III, Faculty of Medicine, University of Granada, 18012 Granada, Spain
| | - Celia Monteagudo
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain; (V.R.); (P.G.-P.); (A.R.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
| | - Luis Javier Martínez-González
- GENYO Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS Granada—Avenida de la Ilustración, 114, 18016 Granada, Spain;
- Department of Biochemistry and Molecular Biology III, Faculty of Medicine, University of Granada, 18012 Granada, Spain
| | - Ana Rivas
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain; (V.R.); (P.G.-P.); (A.R.)
- Instituto de Investigación Biosanitaria ibs.GRANADA, 18012 Granada, Spain
- Institute of Nutrition and Food Technology “Jose Mataix Verdú”, Biomedical Research Center, Health Sciences Technological Park, University of Granada, 18016 Granada, Spain
| |
Collapse
|
4
|
Siegel BI, Gust J. How Cancer Harms the Developing Brain: Long-Term Outcomes in Pediatric Cancer Survivors. Pediatr Neurol 2024; 156:91-98. [PMID: 38735088 DOI: 10.1016/j.pediatrneurol.2024.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 02/22/2024] [Accepted: 03/31/2024] [Indexed: 05/14/2024]
Abstract
Survival rates for pediatric cancer are improving, resulting in a rising need to understand and address long-term sequelae. In this narrative review, we summarize the effects of cancer and its treatment on the developing brain, with a focus on neurocognitive function in leukemia and pediatric brain tumor survivors. We then discuss possible mechanisms of brain injury and management considerations.
Collapse
Affiliation(s)
- Benjamin I Siegel
- Brain Tumor Institute, Children's National Hospital, Washington, District of Columbia; Division of Pediatric Hematology and Oncology, Children's National Hospital, Washington, District of Columbia
| | - Juliane Gust
- Department of Neurology, University of Washinton, Seattle, Washington; Seattle Children's Research Institute, Center for Integrative Brain Research, Seattle, Washington.
| |
Collapse
|
5
|
Li J, Liang D, Chen X, Sun W, Shen X. Applications of 3D printing in tumor treatment. BIOMEDICAL TECHNOLOGY 2024; 5:1-13. [DOI: 10.1016/j.bmt.2023.03.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
6
|
Hou R, Hon CC, Huang Y. CamoTSS: analysis of alternative transcription start sites for cellular phenotypes and regulatory patterns from 5' scRNA-seq data. Nat Commun 2023; 14:7240. [PMID: 37945584 PMCID: PMC10636040 DOI: 10.1038/s41467-023-42636-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 10/16/2023] [Indexed: 11/12/2023] Open
Abstract
Five-prime single-cell RNA-seq (scRNA-seq) has been widely employed to profile cellular transcriptomes, however, its power of analysing transcription start sites (TSS) has not been fully utilised. Here, we present a computational method suite, CamoTSS, to precisely identify TSS and quantify its expression by leveraging the cDNA on read 1, which enables effective detection of alternative TSS usage. With various experimental data sets, we have demonstrated that CamoTSS can accurately identify TSS and the detected alternative TSS usages showed strong specificity in different biological processes, including cell types across human organs, the development of human thymus, and cancer conditions. As evidenced in nasopharyngeal cancer, alternative TSS usage can also reveal regulatory patterns including systematic TSS dysregulations.
Collapse
Affiliation(s)
- Ruiyan Hou
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, SAR, China
| | - Chung-Chau Hon
- RIKEN Center for Integrative Medical Sciences, Yokohama City, Kanagawa, 230-0045, Japan
- Graduate School of Integrated Sciences for Life, Hiroshima University, Higashi-Hiroshima, Japan
| | - Yuanhua Huang
- School of Biomedical Sciences, University of Hong Kong, Hong Kong, SAR, China.
- Department of Statistics and Actuarial Science, University of Hong Kong, Hong Kong, AR, China.
- Center for Translational Stem Cell Biology, Hong Kong Science and Technology Park, Hong Kong, SAR, China.
| |
Collapse
|
7
|
Grob ST, Miller KR, Sanford B, Donson AM, Jones K, Griesinger AM, Amani V, Foreman NK, Liu A, Handler M, Hankinson TC, Milgrom S, Levy JMM. Genetic predictors of neurocognitive outcomes in survivors of pediatric brain tumors. J Neurooncol 2023; 165:161-169. [PMID: 37878192 PMCID: PMC10638163 DOI: 10.1007/s11060-023-04472-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/27/2023] [Indexed: 10/26/2023]
Abstract
BACKGROUND Neurocognitive deficits are common in pediatric brain tumor survivors. The use of single nucleotide polymorphism (SNP) analysis in DNA repair genes may identify children treated with radiation therapy for brain tumors at increased risk for treatment toxicity and adverse neurocognitive outcomes. MATERIALS The Human 660W-Quad v1.0 DNA BeadChip analysis (Illumina) was used to evaluate 1048 SNPs from 59 DNA repair genes in 46 subjects. IQ testing was measured by the Wechsler Intelligence Scale for Children. Linear regression was used to identify the 10 SNPs with the strongest association with IQ scores while adjusting for radiation type. RESULTS The low vs high IQ patient cohorts were well matched for time from first treatment to most recent IQ, first treatment age, sex, and treatments received. 5 SNPs on 3 different genes (CYP29, XRCC1, and BRCA1) and on 3 different chromosomes (10, 19, and 17) had the strongest association with most recent IQ score that was not modified by radiation type. Furthermore, 5 SNPs on 4 different genes (WRN, NR3C1, ERCC4, RAD51L1) on 4 different chromosomes (8, 5, 16, 14) had the strongest association with change in IQ independent of radiation type, first IQ, and years between IQ measures. CONCLUSIONS SNPs offer the potential to predict adverse neurocognitive outcomes in pediatric brain tumor survivors. Our results require validation in a larger patient cohort. Improving the ability to identify children at risk of treatment related neurocognitive deficits could allow for better treatment stratification and early cognitive interventions.
Collapse
Affiliation(s)
- Sydney T Grob
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, USA
| | - Kristen R Miller
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Bridget Sanford
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Andrew M Donson
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, USA
| | - Kenneth Jones
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Andrea M Griesinger
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, USA
| | - Vladimir Amani
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, USA
| | - Nicholas K Foreman
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, USA
- Department of Neurosurgery, Children's Hospital Colorado, Aurora, CO, USA
| | - Arthur Liu
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, USA
- Department of Radiation Oncology, University of Colorado Anschutz, Aurora, CO, USA
| | - Michael Handler
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, USA
- Department of Neurosurgery, Children's Hospital Colorado, Aurora, CO, USA
| | - Todd C Hankinson
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, USA
- Department of Neurosurgery, Children's Hospital Colorado, Aurora, CO, USA
| | - Sarah Milgrom
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, USA
- Department of Radiation Oncology, University of Colorado Anschutz, Aurora, CO, USA
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Jean M Mulcahy Levy
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Children's Hospital Colorado, Aurora, USA.
- Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO, 80045, USA.
| |
Collapse
|
8
|
Yang M, Baser RE, Khanin R, Autuori I, Li QS, Panageas KS, Orlow I, Mao JJ. COMT Val158Met Affects the Analgesic Response to Acupuncture Among Cancer Survivors With Chronic Pain. THE JOURNAL OF PAIN 2023; 24:1721-1730. [PMID: 37187218 PMCID: PMC11321469 DOI: 10.1016/j.jpain.2023.05.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/30/2023] [Accepted: 05/11/2023] [Indexed: 05/17/2023]
Abstract
Catechol-O-methyltransferase (COMT) is the major enzyme involved in the catabolism of dopamine, a neurotransmitter in the brain's reward system. The common COMT polymorphism Val158Met (rs4680 G>A) modulates pain response to opioids through a reward-motivated mechanism; however, its role in nonpharmacological pain medicine has not been clinically characterized. We genotyped 325 participants from a randomized controlled trial of cancer survivors with chronic musculoskeletal pain. We found that carrying methionine at position 158 (158Met) of COMT, encoded by the A allele, significantly increased the analgesic response to electroacupuncture (74% vs 50%; odds ratio [OR]: 2.79; 95% confidence interval [CI]: 1.31, 6.05; P < .01), but not to auricular acupuncture (68% vs 60%; OR: 1.43; 95% CI: .65, 3.12; P = .37) or usual care (24% vs 18%; OR: 1.46; 95% CI: .38, 7.24; P = .61) compared to Val/Val. These findings raise the possibility that COMT Val158Met might be an important predictor of analgesic response to electroacupuncture, providing novel insights into precision nonpharmacologic pain management tailored to individual genetic backgrounds. PERSPECTIVE: This work suggests the modulating effects of the polymorphism in COMT Val158Met on the response to acupuncture. Further research needs to validate these findings, increase the mechanistic understanding of acupuncture, and guide further development of acupuncture as a precision pain management strategy.
Collapse
Affiliation(s)
- Mingxiao Yang
- Integrative Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Raymond E Baser
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Raya Khanin
- Bioinformatics Core Computational Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Isidora Autuori
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Qing S Li
- Integrative Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Katherine S Panageas
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jun J Mao
- Integrative Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
9
|
Grob ST, Miller KR, Sanford B, Donson AM, Jones K, Griesinger AM, Amani V, Foreman NK, Liu A, Handler M, Hankinson TC, Milgrom S, Levy JM. Genetic Predictors of Neurocognitive Outcomes in Survivors of Pediatric Brain Tumors. RESEARCH SQUARE 2023:rs.3.rs-3225952. [PMID: 37609195 PMCID: PMC10441450 DOI: 10.21203/rs.3.rs-3225952/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Purpose Neurocognitive deficits are common in pediatric brain tumor survivors. The use of single nucleotide polymorphism (SNP) analysis in DNA repair genes may identify children treated with radiation therapy for brain tumors at increased risk for treatment toxicity and adverse neurocognitive outcomes. Methods The Human 660W-Quad v1.0 DNA BeadChip analysis (Illumina) was used to evaluate 1048 SNPs from 59 DNA repair genes in 46 subjects. IQ testing was measured by the Wechsler Intelligence Scale for Children. Linear regression was used to identify the 10 SNPs with the strongest association with IQ scores while adjusting for radiation type. Results The low vs high IQ patient cohorts were well matched for time from first treatment to most recent IQ, first treatment age, gender, and treatments received. 5 SNPs on 3 different genes (CYP29, XRCC1, and BRCA1) and on 3 different chromosomes (10, 19, and 17) had the strongest association with most recent IQ score that was not modified by radiation type. Furthermore, 5 SNPs on 4 different genes (WRN, NR3C1, ERCC4, RAD51L1) on 4 different chromosomes (8, 5, 16, 14) had the strongest association with change in IQ independent of radiation type, first IQ, and years between IQ measures. Conclusions SNP polymorphisms offer potential to predict adverse neurocognitive outcomes in pediatric brain tumor survivors. Our results require validation in a larger patient cohort. Improving the ability to identify children at risk of treatment related neurocognitive deficits could allow for better treatment stratification and early cognitive interventions.
Collapse
|
10
|
Yuksel B, Dogan M, Boyacioglu O, Sahin M, Orenay-Boyacioglu S. Association Between Chronic Tinnitus and Brain-Derived Neurotrophic Factor Antisense RNA Polymorphisms Linked to the Val66Met Polymorphism in BDNF. Gene 2023; 875:147507. [PMID: 37230202 DOI: 10.1016/j.gene.2023.147507] [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: 12/16/2022] [Revised: 04/24/2023] [Accepted: 05/22/2023] [Indexed: 05/27/2023]
Abstract
Tinnitus is the sound heard in the ear or head of a person in the absence of external stimuli. Its etiopathogenesis is still not fully understood and the etiological causes responsible for tinnitus are quite variable. Brain-derived neurotrophic factor (BDNF) is one of the key neurotrophic factors in the growth, differentiation, and survival of neurons and in the developing auditory pathway, including the inner ear sensory epithelium. The regulation of BDNF gene is known to be managed by BDNF antisense (BDNF-AS) gene. BDNF-AS is located downstream of the BDNF gene and transcribes a long non-coding RNA. Inhibition of BDNF-AS upregulates BDNF mRNA, which increases protein levels and stimulates neuronal development and differentiation. Thus, BDNF and BDNF-AS both may play roles in the auditory pathway. Polymorphisms in both genes may have impact on hearing performance. A link was suggested between tinnitus and BDNF Val66Met polymorphism. However, there is no study questioning the relationship of tinnitus with BDNF-AS polymorphisms linked with BDNF Val66Met polymorphism. Therefore, this study aimed to scrutinize the role of BDNF-AS polymorphisms showing linkage with the BDNF Val66Met polymorphism in the course of tinnitus pathophysiology. Six BDNF-AS polymorphisms were analyzed on the tinnitus patients (n=85) and the control subjects (n=60) by Fluidigm Real-Time PCR using the Fluidigm Biomark microfluidic platform. When BDNF-AS polymorphisms were compared between the groups in terms of genotype and gender distribution, statistically significant differences were detected in rs925946, rs1519480, and rs10767658, polymorphisms (p<0.05). When the polymorphisms were compared by the duration of tinnitus, significant differences were found in rs925946, rs1488830, rs1519480, and rs10767658 polymorphisms (p<0.05). According to genetic inheritance model analysis, 2.33 and 1.53-fold risks were found for the rs10767658 polymorphism in the recessive and the additive models, respectively. For the rs1519480 polymorphism, a 2.25 fold risk was observed in the additive model. For the rs925946 polymorphism, 2.44 fold protective effect in dominant model, and 0.62 fold risk was found in the additive model. In conclusion, four of the polymorphisms in BDNF-AS gene (rs955946, rs1488830, rs1519480, and rs10767658) are potential gene loci that may play a role in the auditory pathway and affect auditory performance.
Collapse
Affiliation(s)
- Buse Yuksel
- Department of Molecular Biotechnology, Institute of Health Sciences, Aydın Adnan Menderes University, Aydın 09010, Turkey
| | - Murat Dogan
- Department of Otolaryngology, Faculty of Medicine, Aydın Adnan Menderes University, Aydın 09010, Turkey; Department of Otolaryngology, Medicana International Istanbul-Beylikduzu, Istanbul 34520, Turkey
| | - Olcay Boyacioglu
- Faculty of Engineering, Aydın Adnan Menderes University, Aydın 09010, Turkey; Department of Cancer Biology, Faculty of Medicine, Wake Forest University, Winston Salem, NC, USA
| | - Mustafa Sahin
- Department of Otolaryngology, Faculty of Medicine, Aydın Adnan Menderes University, Aydın 09010, Turkey
| | - Seda Orenay-Boyacioglu
- Department of Medical Genetics, Faculty of Medicine, Aydın Adnan Menderes University, Aydın 09010, Turkey.
| |
Collapse
|
11
|
Reyes-González J, Barajas-Olmos F, García-Ortiz H, Magraner-Pardo L, Pons T, Moreno S, Aguirre-Cruz L, Reyes-Abrahantes A, Martínez-Hernández A, Contreras-Cubas C, Barrios-Payan J, Ruiz-Garcia H, Hernandez-Pando R, Quiñones-Hinojosa A, Orozco L, Abrahantes-Pérez MDC. Brain radiotoxicity-related 15CAcBRT gene expression signature predicts survival prognosis of glioblastoma patients. Neuro Oncol 2022; 25:303-314. [PMID: 35802478 PMCID: PMC9925695 DOI: 10.1093/neuonc/noac171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Glioblastoma is the most common and devastating primary brain cancer. Radiotherapy is standard of care; however, it is associated with brain radiation toxicity (BRT). This study used a multi-omics approach to determine whether BRT-related genes (RGs) harbor survival prognostic value and whether their encoded proteins represent novel therapeutic targets for glioblastoma. METHODS RGs were identified through analysis of single-nucleotide variants associated with BRT (R-SNVs). Functional relationships between RGs were established using Protein-Protein Interaction networks. The influence of RGs and their functional groups on glioblastoma prognosis was evaluated using clinical samples from the Glioblastoma Bio-Discovery Portal database and validated using the Chinese Glioma Genome Atlas dataset. The identification of clusters of radiotoxic and putative pathogenic variants in proteins encoded by RGs was achieved by computational 3D structural analysis. RESULTS We identified the BRT-related 15CAcBRT molecular signature with prognostic value in glioblastoma, by analysis of the COMT and APOE protein functional groups. Its external validation confirmed clinical relevance independent of age, MGMT promoter methylation status, and IDH mutation status. Interestingly, the genes IL6, APOE, and MAOB documented significant gene expression levels alteration, useful for drug repositioning. Biological networks associated with 15CAcBRT signature involved pathways relevant to cancer and neurodegenerative diseases. Analysis of 3D clusters of radiotoxic and putative pathogenic variants in proteins coded by RGs unveiled potential novel therapeutic targets in neuro-oncology. CONCLUSIONS 15CAcBRT is a BRT-related molecular signature with prognostic significance for glioblastoma patients and represents a hub for drug repositioning and development of novel therapies.
Collapse
Affiliation(s)
| | | | - Humberto García-Ortiz
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
| | | | - Tirso Pons
- Department of Immunology and Oncology, National Center for Biotechnology, Spanish National Research Council (CNB-CSIC), Madrid, Spain
| | - Sergio Moreno
- Radioneurosurgery Unit, National Institute of Neurology and Neurosurgery;Mexico City, Mexico
| | - Lucinda Aguirre-Cruz
- Neuroendocrinology Laboratory, National Institute of Neurology and Neurosurgery; Mexico City, Mexico
| | - Andy Reyes-Abrahantes
- Precision Translational Oncology Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Angélica Martínez-Hernández
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Cecilia Contreras-Cubas
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
| | - Jorge Barrios-Payan
- Department of Pathology, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico
| | - Henry Ruiz-Garcia
- Department of Neurosurgery and Brain Tumor Stem Cell Research Laboratory, Mayo Clinic, Jacksonville, Florida,USA
| | - Rogelio Hernandez-Pando
- Department of Pathology, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico
| | - Alfredo Quiñones-Hinojosa
- Department of Neurosurgery and Brain Tumor Stem Cell Research Laboratory, Mayo Clinic, Jacksonville, Florida,USA
| | - Lorena Orozco
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, Mexico City, Mexico
| | - María del Carmen Abrahantes-Pérez
- Corresponding Author: María del Carmen Abrahantes-Pérez, PhD, Precision Translational Oncology Laboratory, National Institute of Genomic Medicine, Periférico Sur 4809, Tlalpan, Mexico City C.P. 14610, Mexico ()
| |
Collapse
|
12
|
Kline C, Stoller S, Byer L, Samuel D, Lupo JM, Morrison MA, Rauschecker AM, Nedelec P, Faig W, Dubal DB, Fullerton HJ, Mueller S. An Integrated Analysis of Clinical, Genomic, and Imaging Features Reveals Predictors of Neurocognitive Outcomes in a Longitudinal Cohort of Pediatric Cancer Survivors, Enriched with CNS Tumors (Rad ART Pro). Front Oncol 2022; 12:874317. [PMID: 35814456 PMCID: PMC9259981 DOI: 10.3389/fonc.2022.874317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Background Neurocognitive deficits in pediatric cancer survivors occur frequently; however, individual outcomes are unpredictable. We investigate clinical, genetic, and imaging predictors of neurocognition in pediatric cancer survivors, with a focus on survivors of central nervous system (CNS) tumors exposed to radiation. Methods One hundred eighteen patients with benign or malignant cancers (median diagnosis age: 7; 32% embryonal CNS tumors) were selected from an existing multi-institutional cohort (RadART Pro) if they had: 1) neurocognitive evaluation; 2) available DNA; 3) standard imaging. Utilizing RadART Pro, we collected clinical history, genomic sequencing, CNS imaging, and neurocognitive outcomes. We performed single nucleotide polymorphism (SNP) genotyping for candidate genes associated with neurocognition: COMT, BDNF, KIBRA, APOE, KLOTHO. Longitudinal neurocognitive testing were performed using validated computer-based CogState batteries. The imaging cohort was made of patients with available iron-sensitive (n = 28) and/or T2 FLAIR (n = 41) sequences. Cerebral microbleeds (CMB) were identified using a semi-automated algorithm. Volume of T2 FLAIR white matter lesions (WML) was measured using an automated method based on a convolutional neural network. Summary statistics were performed for patient characteristics, neurocognitive assessments, and imaging. Linear mixed effects and hierarchical models assessed patient characteristics and SNP relationship with neurocognition over time. Nested case-control analysis was performed to compare candidate gene carriers to non-carriers. Results CMB presence at baseline correlated with worse performance in 3 of 7 domains, including executive function. Higher baseline WML volumes correlated with worse performance in executive function and verbal learning. No candidate gene reliably predicted neurocognitive outcomes; however, APOE ϵ4 carriers trended toward worse neurocognitive function over time compared to other candidate genes and carried the highest odds of low neurocognitive performance across all domains (odds ratio 2.85, P=0.002). Hydrocephalus and seizures at diagnosis were the clinical characteristics most frequently associated with worse performance in neurocognitive domains (5 of 7 domains). Overall, executive function and verbal learning were the most frequently negatively impacted neurocognitive domains. Conclusion Presence of CMB, APOE ϵ4 carrier status, hydrocephalus, and seizures correlate with worse neurocognitive outcomes in pediatric cancer survivors, enriched with CNS tumors exposed to radiation. Ongoing research is underway to verify trends in larger cohorts.
Collapse
Affiliation(s)
- Cassie Kline
- Division of Oncology, Department of Pediatrics, Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, United States
- Division of Child Neurology, Department of Neurology, University of California, San Francisco, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Schuyler Stoller
- Division of Child Neurology, Department of Neurology, University of California, San Francisco, United States
| | - Lennox Byer
- UCSF School of Medicine, University of California, San Francisco, United States
| | - David Samuel
- Division of Pediatric Hematology/Oncology, Valley Children’s Hospital, Madera, CA, United States
| | - Janine M. Lupo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Melanie A. Morrison
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Andreas M. Rauschecker
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Pierre Nedelec
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, United States
| | - Walter Faig
- Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Dena B. Dubal
- Department of Neurology, University of California, San Francisco, CA, United States
| | - Heather J. Fullerton
- Division of Child Neurology, Department of Neurology, University of California, San Francisco, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
| | - Sabine Mueller
- Division of Child Neurology, Department of Neurology, University of California, San Francisco, United States
- Department of Pediatrics, University of California, San Francisco, San Francisco, CA, United States
- Department of Neurological Surgery, University of California, San Francisco, CA, United States
- *Correspondence: Sabine Mueller,
| |
Collapse
|
13
|
Ng DQ, Chan D, Agrawal P, Zhao W, Xu X, Acharya M, Chan A. EVIDENCE OF BRAIN-DERIVED NEUROTROPHIC FACTOR IN AMELIORATING CANCER-RELATED COGNITIVE IMPAIRMENT: A SYSTEMATIC REVIEW OF HUMAN STUDIES. Crit Rev Oncol Hematol 2022; 176:103748. [PMID: 35718064 DOI: 10.1016/j.critrevonc.2022.103748] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 06/02/2022] [Accepted: 06/13/2022] [Indexed: 12/27/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) plays an essential role in neurogenesis and neuroplasticity and may be a key protein in cancer-related cognitive impairment (CRCI). This systematic review assessed the relationship between BDNF biomarkers and neurocognitive outcomes in cancer patients and survivors. A search in PubMed, Scopus, and PsycINFO yielded 638 articles, of which 26 were eligible. Fourteen (54%)studied BDNF protein levels while 15 (58%) analyzed BDNF rs6265 polymorphism. Of the nine observational studies reporting BDNF plasma/serum levels, five (56%) exhibited a positive association between BDNF and cognitive function. One study reported intra-tumoral BDNF levels that were negatively associated with memory. For rs6265, three (20%) of 15 studies reported an association with cognitive function with inconsistent directions. Among seven neuroimaging studies, three (43%) demonstrated an effect of BDNF on brain function and structure. These results suggest that BDNF is a potential monitoring biomarker and druggable target for CRCI.
Collapse
Affiliation(s)
- Ding Quan Ng
- Department of Clinical Pharmacy Practice, University of California, Irvine, Irvine, California, United States of America; Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, California, United States of America
| | - Daniella Chan
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, California, United States of America
| | - Parisa Agrawal
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, California, United States of America
| | - Weian Zhao
- Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, California, United States of America; Department of Biological Chemistry, University of California, Irvine, Irvine, California, USA; Department of Biomedical Engineering, The Henry Samueli School of Engineering, University of California, Irvine, Irvine, California, USA; Institute for Immunology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA; Edwards Life Sciences Center for Advanced Cardiovascular Technology, University of California, Irvine, Irvine, California, USA; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, California, USA
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California, USA; The Center for Neural Circuit Mapping, University of California Irvine, Irvine, California, USA
| | - Munjal Acharya
- Department of Anatomy and Neurobiology, University of California, Irvine, Irvine, California, USA; Department of Radiation Oncology, School of Medicine, University of California, Irvine, Irvine, California, USA
| | - Alexandre Chan
- Department of Clinical Pharmacy Practice, University of California, Irvine, Irvine, California, United States of America; Department of Pharmaceutical Sciences, School of Pharmacy & Pharmaceutical Sciences, University of California, Irvine, Irvine, California, United States of America; Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, California, USA.
| |
Collapse
|
14
|
Zhao Y, Wu J, Li D, Liu J, Chen W, Hou Z, Liu K, Jiang L, Chen X, Wang L, Hu B, Zong F, Wang Y, Wang Y. Human ESC-derived immunity- and matrix- regulatory cells ameliorated white matter damage and vascular cognitive impairment in rats subjected to chronic cerebral hypoperfusion. Cell Prolif 2022; 55:e13223. [PMID: 35437845 PMCID: PMC9136497 DOI: 10.1111/cpr.13223] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/24/2022] [Accepted: 03/03/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES This study investigated the ability of immunity- and matrix- regulatory cells (IMRCs) to improve cognitive function in a rat model of vascular cognitive impairment. MATERIALS AND METHODS A chronic cerebral hypoperfusion (CCH) model was established in rats via permanent bilateral occlusion of the common carotid arteries (two-vessel occlusion, 2VO). The rats then received intravenous injections of IMRCs or saline. A single injection of different doses of IMRCs (1 × 106 cells/rat, 2 × 106 cells/rat, or 4 × 106 cells/rat) was administered via tail vein 72 h after establishment of the model. To evaluate functional recovery, the rats were subjected to behavioural tests after 30 days of CCH. Imaging, western blotting, immunofluorescence staining, and quantitative real-time PCR were used to analyse neuroinflammation and white matter injury after 14 and 40 days of CCH. RNA sequencing (RNA-seq) was used to profile gene expression changes in copine 1 (CPNE1) in response to IMRCs treatment. RESULTS Intravenous injection of 4 × 106 IMRCs alleviated white matter damage and ameliorated cognitive deficits in rats subjected to CCH. Immunofluorescence staining suggested that activation of microglia and astrocytes was reduced, and RNA sequencing showed that CPNE1 expression was significantly elevated following treatment with IMRCs. CONCLUSIONS Intravenous injection of IMRCs protected against CCH-induced white matter injury and cognitive impairment inhibition of microglial activation and regulation of microglia polarization.
Collapse
|
15
|
Cognitive deficits in adult patients with high-grade glioma: A systematic review. Clin Neurol Neurosurg 2022; 219:107296. [DOI: 10.1016/j.clineuro.2022.107296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 04/04/2022] [Accepted: 05/13/2022] [Indexed: 11/15/2022]
|
16
|
Li W, Zhang Q, Cai Y, Chen T, Cheng H. The COMT Genetic Factor Regulates Chemotherapy-Related Prospective Memory Impairment in Survivors With HER2-/+ Breast Cancer. Front Oncol 2022; 12:816923. [PMID: 35211407 PMCID: PMC8861381 DOI: 10.3389/fonc.2022.816923] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
Background Previous findings indicated that polymorphism in gene catechol-O-methyltransferase (COMT) had been linked to chemotherapy-related cognitive impairment (CRCI). Nevertheless, the motivation of COMT polymorphisms in regulating cognitive impairment in breast cancer survivors with disparate status of human epidermal growth factor receptor 2 (HER2) was still vague. Objective The current research aimed to evaluate the regulation of the risk by COMT genotype on CRCI in breast cancer survivors with disparate status of HER2. Methods Breast cancer survivors (103 with HER2− and 118 with HER2+) underwent neuropsychological tests before and after chemotherapy, containing event- and time-based prospective memory (EBPM and TBPM). Three single-nucleotide polymorphisms (SNPs) were estimated by providing peripheral blood, containing COMT (rs165599, rs737865, and rs4680). Results The EBPM and TBPM performances was lower as compared with these before chemotherapy (z = −7.712, z = −2.403, respectively, p < 0.01). Furthermore, the EBPM and TBPM performances of HER2− group survivors were lower than those of HER2+ group survivors after chemotherapy (z = −7.181, p < 0.01; z = −2.205 p < 0.05, respectively). The survivors with COMT (rs165599) A/A genotype carriers had a meaningfully poorer chance of memory descend [dominant model: adjusted, OR = 2.21, CI (95%) = 1.156–4.225, p = 0.016] and showed better on TBPM test, relative to G/G genotype. Patients with the COMT (rs737865) A/G and G/G genotype showed protective function than the patients with the A/A and performed better on MMSE and TBPM tests. Conclusion The types of HER2 may be correlated to chemotherapy-related prospective memory impairments in breast cancer survivors. Furthermore, the COMT (rs165599, rs737865) polymorphisms were correlated to the risk of TBPM decline scores and possibly be a potential genetic identifying for increasing risk of CRCI in breast cancer patients with disparate status of HER2.
Collapse
Affiliation(s)
- Wen Li
- Cancer Treatment Center, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qianqian Zhang
- Cancer Treatment Center, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yinlian Cai
- Cancer Treatment Center, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Tingting Chen
- Cancer Treatment Center, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Huaidong Cheng
- Cancer Treatment Center, the Second Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
17
|
Cheng X, Li D, Qi T, Sun J, Zhou T, Zheng WV. Objective to identify and verify the regulatory mechanism of DTNBP1 as a prognostic marker for hepatocellular carcinoma. Sci Rep 2022; 12:211. [PMID: 34997064 PMCID: PMC8742032 DOI: 10.1038/s41598-021-04055-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/09/2021] [Indexed: 11/09/2022] Open
Abstract
Although the overall survival of hepatocellular carcinoma (HCC) patients has been significantly improved, prognostic clinical evaluation remains a substantial problem owing to the heterogeneity and complexity of tumor. A reliable and accurate predictive biomarker may assist physicians in better monitoring of patient treatment outcomes and follow the overall survival of patients. Accumulating evidence has revealed that DTNBP1 plays functional roles in cancer prognosis. Therefore, the expression and function of DTNBP1in HCC was systematically investigated in our study. The expression and prognostic value of DTNBP1 were investigated using the data from Cancer Genome Atlas (TCGA) database, Gene Expression Omnibus (GEO) cohorts and clinical samples. A series of cellular function assays were performed to elucidate the effect of DTNBP1 on cellular proliferation, apoptosis and metastasis. Kyoto Encyclopaedia of Genes and Genomes (KEGG) pathway enrichment and Protein–protein interaction (PPI) network construction were performed to screen the genes with highest interaction scores with DTNBP1. Finally, the underlying mechanism was also analyzed using Gene Set Enrichment Analysis (GSEA) and confirmed using RT-qPCR and western blotting. DTNBP1 was upregulated in many types of cancers, especially in HCC. The DTNBP1 expression levels is associated with clinicopathologic variables and patient survival status. The differential expression of DTNBP1 could be used to determine the risk stratification of patients with HCC. DTNBP1 deficiency inhibited cell proliferation and metastasis, but promoted cell apoptosis. Mechanistically, DTNBP1 regulated the cell cycle progression through affecting the expression of cell cycle-related genes such as CDC25A, CCNE1, CDK2, CDC20, CDC25B, CCNB1, and CDK1. DTNBP1, which regulates the cell cycle progression, may be used as a prognostic marker for HCC.
Collapse
Affiliation(s)
- Xianyi Cheng
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Dezhi Li
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Tiangyang Qi
- Ascentawits Pharmaceuticals, Ltd., Biomedical Innovation Industrial Park, No. 14 Jinhui Road, Jinsha Community, Kengzi Street, Pingshan District, Shenzhen, Guangdong, People's Republic of China
| | - Jia Sun
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China.,ShenZhen Beike Biotechnology Research Institute, Shenzhen, 518057, People's Republic of China
| | - Tao Zhou
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China
| | - Wei V Zheng
- Intervention and Cell Therapy Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, People's Republic of China.
| |
Collapse
|
18
|
Wefel JS, Zhou R, Sulman EP, Boehling NS, Armstrong GN, Tsavachidis S, Liang FW, Etzel CJ, Kahalley LS, Small BJ, Scheurer ME, Bondy ML, Liu Y. Genetic modulation of longitudinal change in neurocognitive function among adult glioma patients. J Neurooncol 2021; 156:185-193. [PMID: 34817796 DOI: 10.1007/s11060-021-03905-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 11/17/2021] [Indexed: 11/24/2022]
Abstract
PURPOSE Impaired neurocognitive function (NCF) is extremely common in patients with higher grade primary brain tumor. We previously reported evidence of genetic variants associated with NCF in glioma patients prior to treatment. However, little is known about the effect of genetic variants on NCF decline after adjuvant therapy. METHODS Patients (N = 102) completed longitudinal NCF assessments that included measures of verbal memory, processing speed, and executive function. Testing was conducted in the postoperative period with an average follow up interval of 1.3 years. We examined polymorphisms in 580 genes related to five pathways (inflammation, DNA repair, metabolism, cognitive, and telomerase). RESULTS Five polymorphisms were associated with longitudinal changes in processing speed and 14 polymorphisms with executive function. Change in processing speed was strongly associated with MCPH1 rs17631450 (P = 2.2 × 10-7) and CCDC26 rs7005206 (P = 9.3 × 10-7) in the telomerase pathway; while change in executive function was more strongly associated with FANCF rs1514084 (P = 2.9 × 10-6) in the DNA repair pathway and DAOA rs12428572 (P = 2.4 × 10-5) in the cognitive pathway. Joint effect analysis found significant genetic-dosage effects for longitudinal changes in processing speed (Ptrend = 1.5 × 10-10) and executive function (Ptrend = 2.1 × 10-11). In multivariable analyses, predictors of NCF decline included progressive disease, lower baseline NCF performance, and more at-risk genetic variants, after adjusting for age, sex, education, tumor location, histology, and disease progression. CONCLUSION Our longitudinal analyses revealed that polymorphisms in telomerase, DNA repair, and cognitive pathways are independent predictors of decline in NCF in glioma patients.
Collapse
Affiliation(s)
- Jeffrey S Wefel
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX, 77030, USA.
| | - Renke Zhou
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA
| | - Erik P Sulman
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Nicholas S Boehling
- Department of Radiation Oncology, The University of Texas M.D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Georgina N Armstrong
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA
| | - Spiridon Tsavachidis
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA
| | - Fu-Wen Liang
- Institute of Public Health, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Carol J Etzel
- Biostatistics, Corrona, LLC, Southborough, MA, 01772, USA
| | - Lisa S Kahalley
- Department of Pediatrics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Brent J Small
- School of Aging Studies, University of South Florida, 4202 E Fowler Avenue, Tampa, FL, 33620, USA
| | - Michael E Scheurer
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA
| | - Melissa L Bondy
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA.
| | - Yanhong Liu
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, One Baylor Plaza, Mailstop BCM305, Houston, TX, 77030, USA.
| |
Collapse
|
19
|
Parsons MW, Peters KB, Floyd SR, Brown P, Wefel JS. Preservation of neurocognitive function in the treatment of brain metastases. Neurooncol Adv 2021; 3:v96-v107. [PMID: 34859237 PMCID: PMC8633744 DOI: 10.1093/noajnl/vdab122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Neurocognitive function (NCF) deficits are common in patients with brain metastases, occurring in up to 90% of cases. NCF deficits may be caused by tumor-related factors and/or treatment for the metastasis, including surgery, radiation therapy, chemotherapy, and immunotherapy. In recent years, strategies to prevent negative impact of treatments and ameliorate cognitive deficits for patients with brain tumors have gained momentum. In this review, we report on research that has established the efficacy of preventative and rehabilitative therapies for NCF deficits in patients with brain metastases. Surgical strategies include the use of laser interstitial thermal therapy and intraoperative mapping. Radiotherapy approaches include focal treatments such as stereotactic radiosurgery and tailored approaches such as hippocampal avoidant whole-brain radiotherapy (WBRT). Pharmacologic options include use of the neuroprotectant memantine to reduce cognitive decline induced by WBRT and incorporation of medications traditionally used for attention and memory problems. Integration of neuropsychology into the care of patients with brain metastases helps characterize cognitive patterns, educate patients and families regarding their management, and guide rehabilitative therapies. These and other strategies will become even more important for long-term survivors of brain metastases as treatment options improve.
Collapse
Affiliation(s)
- Michael W Parsons
- Pappas Center for Neuro-Oncology, Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Katherine B Peters
- Preston Robert Tisch Brain Tumor Center, Department of Neurosurgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Scott R Floyd
- Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Paul Brown
- Department of Radiation Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Jeffrey S Wefel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
20
|
Genovese TJ, Gehrman P, Yang M, Li Y, Garland SN, Orlow I, Mao JJ. Genetic Predictors of Response to Acupuncture or Cognitive Behavioral Therapy for Insomnia in Cancer Survivors: An Exploratory Analysis. J Pain Symptom Manage 2021; 62:e192-e199. [PMID: 33716034 PMCID: PMC9297333 DOI: 10.1016/j.jpainsymman.2021.03.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 02/24/2021] [Accepted: 03/04/2021] [Indexed: 11/30/2022]
Abstract
CONTEXT Insomnia is a common problem affecting cancer survivors. While effective nonpharmacological treatments are available, it is unknown whether individual genetic characteristics influence treatment response. OBJECTIVES We conducted an exploratory analysis of genetic associations with insomnia treatment response in a randomized trial of cognitive behavioral therapy for insomnia (CBT-I) vs. acupuncture in a heterogeneous group of cancer survivors. METHODS We successfully genotyped 136 participants for 11 genetic variants. Successful treatment response was defined as a reduction in Insomnia Severity Index score of at least eight points from baseline to week 8. We used Fisher exact tests to evaluate associations between genotype and treatment success for each treatment arm, for an alpha level of 0.05 with unadjusted and Holm-Bonferroni-adjusted P-values. RESULTS We found that more carriers of COMT rs4680-A alleles responded to acupuncture compared to the GG carriers (63.6% vs. 27.8%, P = 0.013). More carriers of the NFKB2 rs1056890 CC genotype also responded to acupuncture compared to TT or CT carriers (72.2% vs. 38.9%, P = 0.009). There were no significant differences found between any of the tested gene variants and CBT-I response. None of the results remained statistically significant after adjustment for multiple testing. CONCLUSION In cancer survivors, specific variants in the COMT and NFKB2 genes are potentially associated with response to acupuncture but not to CBT-I. Confirming these preliminary results will help inform precision insomnia management for cancer survivors.
Collapse
Affiliation(s)
- Timothy J Genovese
- Harvard T.H. Chan School of Public Health, Boston, MA, USA; The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Philip Gehrman
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - MingXiao Yang
- Integrative Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yuelin Li
- Department of Psychiatry and Behavioral Sciences, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sheila N Garland
- Departments of Psychology and Oncology, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jun J Mao
- Integrative Medicine Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
21
|
Lei C, Chen W, Wang Y, Zhao B, Liu P, Kong Z, Liu D, Dai C, Wang Y, Wang Y, Ma W. Prognostic Prediction Model for Glioblastoma: A Metabolic Gene Signature and Independent External Validation. J Cancer 2021; 12:3796-3808. [PMID: 34093788 PMCID: PMC8176239 DOI: 10.7150/jca.53827] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 04/21/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Glioblastoma (GBM) is the most common primary malignant intracranial tumor and closely related to metabolic alteration. However, few accepted prognostic models are currently available, especially models based on metabolic genes. Methods: The transcriptome data were obtained for all of the patients diagnosed with GBM from the Gene Expression Omnibus (GEO) (training cohort, n=369) and The Cancer Genome Atlas (TCGA) (validation cohort, n=152) with the following variables: age at diagnosis, sex, follow-up and overall survival (OS). Metabolic genes according to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were contracted, and a Lasso regression model was constructed. Survival was assessed by univariate or multivariate Cox proportional hazards regression and Kaplan-Meier analysis, and an independent external validation was also conducted to examine the model. Results: There were 341 metabolic genes showed significant differences between normal brain and GBM tissues in both the training and validation cohorts, among which 56 genes were dramatically correlated to the OS of patients. Lasso regression revealed that the metabolic prognostic model was composed of 18 genes, including COX10, COMT, and GPX2 with protective effects, as well as OCRL and RRM2 with unfavorable effects. Patients classified as high-risk by the risk score from this model had markedly shorter OS than low-risk patients (P<0.0001), and this significant result was also observed in independent external validation (P<0.001). Conclusions: The prognosis of GBM was dramatically related to metabolic pathways, and our metabolic prognostic model had high accuracy and application value in predicting the OS of GBM patients.
Collapse
Affiliation(s)
- Chuxiang Lei
- Department of Vascular Surgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Wenlin Chen
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Yuekun Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Binghao Zhao
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Penghao Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Ziren Kong
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Delin Liu
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Congxin Dai
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Yaning Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Yu Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| | - Wenbin Ma
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, No. 1 Shuaifuyuan, Dongcheng District, Beijing, China
| |
Collapse
|
22
|
Butterbrod E, Sitskoorn M, Bakker M, Jakobs B, Fleischeuer R, Roijers J, Rutten G, Gehring K. The APOE ε4 allele in relation to pre- and postsurgical cognitive functioning of patients with primary brain tumors. Eur J Neurol 2021; 28:1665-1676. [PMID: 33342004 PMCID: PMC8247965 DOI: 10.1111/ene.14693] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 12/03/2020] [Accepted: 12/06/2020] [Indexed: 12/01/2022]
Abstract
BACKGROUND Recent studies suggest a relationship between the APOE ε4 allele and cognitive outcome in patients treated for malignant brain tumors. Still, longitudinal investigations that include a pretreatment cognitive assessment are lacking and APOE's effects in patients with benign tumors are understudied. This study investigated presurgical cognitive performance and postsurgical change in ε4-carrying and non-carrying patients with glioma and meningioma. METHODS Neuropsychological test scores (CNS Vital Signs battery [seven measures], Digit Span Forward/Backward, Letter Fluency test) were obtained as part of a prospective study in which patients with meningioma and glioma underwent cognitive assessment 1 day before (T0, n = 505) and 3 (T3, n = 418) and 12 months after (T12, n = 167) surgery. APOE isoforms were identified retrospectively. ε4 carriers and non-carriers were compared with regard to pretreatment cognitive performance on the group and individual level. Changes in performances over time were compared with longitudinal mixed model analysis in the total sample and the subgroup receiving adjuvant treatment. RESULTS Carriers and non-carriers did not differ with regard to pretreatment performance. No significant main effect of ε4 carrier status or interaction between time (T0-T12) and carrier status was found on any of the tests in the whole sample nor in the sample receiving adjuvant treatment. CONCLUSIONS This study found no evidence of increased vulnerability for pretreatment cognitive dysfunction or cognitive decline within 1 year after surgery in APOE ε4-carrying meningioma and glioma patients. Investigations that include larger samples at longer-term follow-up are recommended to investigate potential late treatment effects.
Collapse
Affiliation(s)
- Elke Butterbrod
- Department of Cognitive NeuropsychologyTilburg UniversityTilburgThe Netherlands
| | - Margriet Sitskoorn
- Department of Cognitive NeuropsychologyTilburg UniversityTilburgThe Netherlands
| | - Marjan Bakker
- Department of Methodology and StatisticsTilburg UniversityTilburgThe Netherlands
| | - Bernadette Jakobs
- Department of Laboratory MedicineElisabeth‐Tweesteden HospitalTilburgThe Netherlands
| | - Ruth Fleischeuer
- Clinical Pathology LaboratoryElisabeth‐Tweesteden HospitalTilburgThe Netherlands
| | - Janine Roijers
- Department of Laboratory MedicineElisabeth‐Tweesteden HospitalTilburgThe Netherlands
| | - Geert‐Jan Rutten
- Department of NeurosurgeryElisabeth‐Tweesteden HospitalTilburgThe Netherlands
| | - Karin Gehring
- Department of Cognitive NeuropsychologyTilburg UniversityTilburgThe Netherlands
- Department of NeurosurgeryElisabeth‐Tweesteden HospitalTilburgThe Netherlands
| |
Collapse
|
23
|
Geng H, Tsang M, Subbaraj L, Cleveland J, Chen L, Lu M, Sharma J, Vigneron DB, Kurhanewicz J, LaFontaine M, Luks T, Barshop BA, Gangoiti J, Villanueva-Meyer JE, Rubenstein JL. Tumor Metabolism and Neurocognition in CNS Lymphoma. Neuro Oncol 2021; 23:1668-1679. [PMID: 33625503 DOI: 10.1093/neuonc/noab045] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The mechanistic basis for neurocognitive deficits in CNS lymphoma and other brain tumors is incompletely understood. We tested the hypothesis that tumor metabolism impairs neurotransmitter pathways and neurocognitive function. METHODS We performed serial cerebrospinal fluid (CSF) metabolomic analyses using liquid chromatography-electrospray tandem mass spectrometry to evaluate changes in the tumor microenvironment in 14 patients with recurrent CNS lymphoma, focusing on 18 metabolites involved in neurotransmission and bioenergetics. These were paired with serial mini-mental state examinations (MMSE) and MRI studies for tumor volumetric analyses. Patients were analyzed in the setting of the phase I trial of lenalidomide/rituximab. Associations were assessed by Pearson and Spearman correlation coefficient. Generalized estimating equation (gee) models were also established, adjusting for within-subject repeated measures. RESULTS Of 18 metabolites, elevated CSF lactate correlated most strongly with lower MMSE score (p<8E-8, rho=-0.67). High lactate was associated with lower GABA, higher glutamate/GABA ratio and dopamine. Conversely, high succinate correlated with higher MMSE score. Serial analysis demonstrated a reproducible, time-dependent, reciprocal correlation between changes in lactate and GABA concentrations. While high lactate and low GABA correlated with tumor contrast enhancing volume, they correlated more significantly with lower MMSE scores than tumor volumes. CONCLUSIONS We provide evidence that lactate production and Warburg metabolism may impact neurotransmitter dysregulation and neurocognition in CNS lymphomas. We identify novel metabolomic biomarkers that may be applied in future studies of neurocognition in CNS lymphomas. Elucidation of mechanistic interactions between lymphoma metabolism, neurotransmitter imbalance and neurocognition may promote interventions that preserve cognitive function.
Collapse
Affiliation(s)
- Huimin Geng
- Laboratory Medicine, University of California, San Francisco (UCSF).,Helen Diller Family Comprehensive Cancer Center, UCSF
| | - Mazie Tsang
- Hematology/Oncology, UCSF.,Department of Medicine, UCSF
| | | | | | - Lingjing Chen
- Hematology/Oncology, UCSF.,Department of Medicine, UCSF
| | - Ming Lu
- Hematology/Oncology, UCSF.,Department of Medicine, UCSF
| | | | - Daniel B Vigneron
- Helen Diller Family Comprehensive Cancer Center, UCSF.,Radiology and Biomedical Imaging
| | - John Kurhanewicz
- Helen Diller Family Comprehensive Cancer Center, UCSF.,Radiology and Biomedical Imaging
| | | | | | - Bruce A Barshop
- Genetics and Pediatrics, University of California, San Diego
| | - Jon Gangoiti
- Genetics and Pediatrics, University of California, San Diego
| | | | - James L Rubenstein
- Helen Diller Family Comprehensive Cancer Center, UCSF.,Hematology/Oncology, UCSF
| |
Collapse
|
24
|
Krishna S, Kakaizada S, Almeida N, Brang D, Hervey-Jumper S. Central Nervous System Plasticity Influences Language and Cognitive Recovery in Adult Glioma. Neurosurgery 2021; 89:539-548. [PMID: 33476391 DOI: 10.1093/neuros/nyaa456] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 08/05/2020] [Indexed: 01/01/2023] Open
Abstract
Gliomas exist within the framework of complex neuronal circuitry in which network dynamics influence both tumor biology and cognition. The generalized impairment of cognition or loss of language function is a common occurrence for glioma patients. The interface between intrinsic brain tumors such as gliomas and functional cognitive networks are poorly understood. The ability to communicate effectively is critically important for receiving oncological therapies and maintaining a high quality of life. Although the propensity of gliomas to infiltrate cortical and subcortical structures and disrupt key anatomic language pathways is well documented, there is new evidence offering insight into the network and cellular mechanisms underpinning glioma-related aphasia and aphasia recovery. In this review, we will outline the current understanding of the mechanisms of cognitive dysfunction and recovery, using aphasia as an illustrative model.
Collapse
Affiliation(s)
- Saritha Krishna
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Sofia Kakaizada
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Nyle Almeida
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - David Brang
- Department of Psychology, University of Michigan, Ann Arbor, Michigan
| | - Shawn Hervey-Jumper
- Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| |
Collapse
|
25
|
Correa DD, Satagopan J, Martin A, Braun E, Kryza-Lacombe M, Cheung K, Sharma A, Dimitriadoy S, O'Connell K, Leong S, Karimi S, Lyo J, DeAngelis LM, Orlow I. Genetic variants and cognitive functions in patients with brain tumors. Neuro Oncol 2020; 21:1297-1309. [PMID: 31123752 PMCID: PMC6784270 DOI: 10.1093/neuonc/noz094] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Patients with brain tumors treated with radiotherapy (RT) and chemotherapy (CT) often experience cognitive dysfunction. We reported that single nucleotide polymorphisms (SNPs) in the APOE, COMT, and BDNF genes may influence cognition in brain tumor patients. In this study, we assessed whether genes associated with late-onset Alzheimer's disease (LOAD), inflammation, cholesterol transport, dopamine and myelin regulation, and DNA repair may influence cognitive outcome in this population. METHODS One hundred and fifty brain tumor patients treated with RT ± CT or CT alone completed a neurocognitive assessment and provided a blood sample for genotyping. We genotyped genes/SNPs in these pathways: (i) LOAD risk/inflammation/cholesterol transport, (ii) dopamine regulation, (iii) myelin regulation, (iv) DNA repair, (v) blood-brain barrier disruption, (vi) cell cycle regulation, and (vii) response to oxidative stress. White matter (WM) abnormalities were rated on brain MRIs. RESULTS Multivariable linear regression analysis with Bayesian shrinkage estimation of SNP effects, adjusting for relevant demographic, disease, and treatment variables, indicated strong associations (posterior association summary [PAS] ≥ 0.95) among tests of attention, executive functions, and memory and 33 SNPs in genes involved in: LOAD/inflammation/cholesterol transport (eg, PDE7A, IL-6), dopamine regulation (eg, DRD1, COMT), myelin repair (eg, TCF4), DNA repair (eg, RAD51), cell cycle regulation (eg, SESN1), and response to oxidative stress (eg, GSTP1). The SNPs were not significantly associated with WM abnormalities. CONCLUSION This novel study suggests that polymorphisms in genes involved in aging and inflammation, dopamine, myelin and cell cycle regulation, and DNA repair and response to oxidative stress may be associated with cognitive outcome in patients with brain tumors.
Collapse
Affiliation(s)
- Denise D Correa
- Department of Neurology and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Neurology, Weill Cornell Medical College, New York, New York
| | - Jaya Satagopan
- Department of Epidemiology and Biostatistics and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Axel Martin
- Department of Epidemiology and Biostatistics and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Erica Braun
- Department of Neurology and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria Kryza-Lacombe
- San Diego State University/University of California, San Diego Joint Doctoral Program in Clinical Psychology, San Diego, California
| | - Kenneth Cheung
- Department of Epidemiology and Biostatistics and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ajay Sharma
- Department of Epidemiology and Biostatistics and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sofia Dimitriadoy
- Department of Epidemiology and Biostatistics and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kelli O'Connell
- Department of Epidemiology and Biostatistics and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Siok Leong
- Department of Epidemiology and Biostatistics and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sasan Karimi
- Department of Neurology and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - John Lyo
- Department of Neurology and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Lisa M DeAngelis
- Department of Neurology and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Neurology, Weill Cornell Medical College, New York, New York
| | - Irene Orlow
- Department of Epidemiology and Biostatistics and Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
26
|
Liou KT, Baser R, Romero SA, Green J, Li QS, Orlow I, Panageas KS, Mao JJ. Personalized electro-acupuncture versus auricular-acupuncture comparative effectiveness (PEACE): A protocol of a randomized controlled trial for chronic musculoskeletal pain in cancer survivors. Medicine (Baltimore) 2020; 99:e20085. [PMID: 32481275 PMCID: PMC7249872 DOI: 10.1097/md.0000000000020085] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/01/2020] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION Chronic pain is a leading cause of disability and remains under-treated in nearly half of patients with cancer. The opioid crisis has highlighted an urgent public health need for effective nonpharmacological pain management. Electroacupuncture (EA) and Battlefield Acupuncture (BFA) represent nonpharmacological modalities used in clinical practice to manage pain; however, their effectiveness has not been rigorously evaluated in oncology settings. METHODS We describe the design of a 3-arm, parallel, single-center, multisite randomized controlled trial that investigates EA and BFA versus usual-care wait-list control (WLC) for chronic musculoskeletal pain among 360 patients with diverse cancer types across various stages. The primary aim is to compare effects of EA and BFA versus WLC on pain, physical function, and co-morbid symptoms. The secondary aim is to examine the interaction between patient outcome expectancy and acupuncture modality (EA vs BFA) on pain reduction. The tertiary aim is to evaluate the association between genetic polymorphisms and responses to acupuncture. Patients will be randomized in a 2:2:1 ratio to EA:BFA:WLC. Acupuncture groups will receive weekly treatments over 10 weeks. WLC will receive usual care over the same evaluation period as the acupuncture groups. The primary endpoint will be the change in average pain intensity score from baseline to week 12. We will collect validated patient-reported outcomes and blood/saliva samples at multiple timepoints over 24 weeks. DISCUSSION Our findings will advance nonpharmacological pain management in oncology and inform personalized treatment approaches that integrate individuals' expectations and genetic biomarkers to deliver "precision" acupuncture to cancer patients with chronic pain. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02979574.
Collapse
Affiliation(s)
- Kevin T. Liou
- Integrative Medicine Service, Department of Medicine
| | - Ray Baser
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Sally A.D. Romero
- Department of Family Medicine and Public Health, UC San Diego School of Medicine, San Diego, CA
| | - Jamie Green
- Integrative Medicine Service, Department of Medicine
| | - Q. Susan Li
- Integrative Medicine Service, Department of Medicine
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Katherine S. Panageas
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jun J. Mao
- Integrative Medicine Service, Department of Medicine
| |
Collapse
|
27
|
Abstract
Increased life expectancy in brain tumour patients had led to the need for strategies that preserve and improve cognitive functioning, as many patients suffer from cognitive deficits. The tumour itself, as well as antitumor treatment including surgery, radiotherapy and chemotherapy, supportive treatment and individual patient factors are associated with cognitive problems. Here, we review the recent literature on approaches that preserve and improve cognitive functioning, including pharmacological agents and rehabilitation programs.
Collapse
|
28
|
Sharafeldin N, Richman J, Bosworth A, Chen Y, Singh P, Patel SK, Wang X, Francisco L, Forman SJ, Wong FL, Bhatia S. Clinical and Genetic Risk Prediction of Cognitive Impairment After Blood or Marrow Transplantation for Hematologic Malignancy. J Clin Oncol 2020; 38:1312-1321. [PMID: 32083992 DOI: 10.1200/jco.19.01085] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
PURPOSE Using a candidate gene approach, we tested the hypothesis that individual single nucleotide polymorphisms (SNPs) and gene-level variants are associated with cognitive impairment in patients with hematologic malignancies treated with blood or marrow transplantation (BMT) and that inclusion of these SNPs improves risk prediction beyond that offered by clinical and demographic characteristics. PATIENTS AND METHODS In the discovery cohort, BMT recipients underwent a standardized battery of neuropsychological tests pre-BMT and at 6 months, 1 year, 2 years, and 3 years post-BMT. Associations between 68 candidate genes and cognitive impairment were assessed using generalized estimating equation models. Elastic-Net regression was used to build Base (sociodemographic), Clinical, and Combined (Base plus Clinical plus genetic) risk prediction models of post-BMT impairment. An independent nonoverlapping cohort from the BMT Survivor Study with self-report of learning/memory problems (as identified by their health care provider) was used for model replication. RESULTS The discovery cohort included 277 participants (58.5% males; 68.6% non-Hispanic whites; and 46.6% allogeneic BMT recipients). Adjusting for BMT type, age at BMT, sex, race/ethnicity, and cognitive reserve, SNPs in the blood-brain barrier, telomere homeostasis, and DNA repair genes were significantly associated with cognitive impairment. Compared with the Clinical Model, the Combined Model had higher predictive power in both the discovery cohort (mean area under the receiver operating characteristic curve [AUC], 0.89; 95% CI, 0.85 to 0.93 v 0.77; 95% CI, 0.71 to 0.83; P = 1.24 × 10-9) and the replication cohort (AUC, 0.71; 95% CI, 0.66 to 0.76 v 0.63; 95% CI, 0.57 to 0.68; P = .004). CONCLUSION Inclusion of candidate genetic variants enhanced the prediction of risk of post-BMT cognitive impairment beyond that offered by demographic/clinical characteristics and represents a step toward a personalized approach to managing patients at high risk for cognitive impairment after BMT.
Collapse
Affiliation(s)
- Noha Sharafeldin
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Joshua Richman
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | | | - Yanjun Chen
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Purnima Singh
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | | | - Xuexia Wang
- Department of Mathematics, University of North Texas, Denton, TX
| | - Liton Francisco
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| | - Stephen J Forman
- Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA
| | | | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
29
|
Waddington JL, Zhen X, O'Tuathaigh CMP. Developmental Genes and Regulatory Proteins, Domains of Cognitive Impairment in Schizophrenia Spectrum Psychosis and Implications for Antipsychotic Drug Discovery: The Example of Dysbindin-1 Isoforms and Beyond. Front Pharmacol 2020; 10:1638. [PMID: 32063853 PMCID: PMC7000454 DOI: 10.3389/fphar.2019.01638] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 12/16/2019] [Indexed: 12/19/2022] Open
Abstract
Alongside positive and negative symptomatology, deficits in working memory, attention, selective learning processes, and executive function have been widely documented in schizophrenia spectrum psychosis. These cognitive abnormalities are strongly associated with impairment across multiple function domains and are generally treatment-resistant. The DTNBP1 (dystrobrevin-binding protein-1) gene, encoding dysbindin, is considered a risk factor for schizophrenia and is associated with variation in cognitive function in both clinical and nonclinical samples. Downregulation of DTNBP1 expression in dorsolateral prefrontal cortex and hippocampal formation of patients with schizophrenia has been suggested to serve as a primary pathophysiological process. Described as a "hub," dysbindin is an important regulatory protein that is linked with multiple complexes in the brain and is involved in a wide variety of functions implicated in neurodevelopment and neuroplasticity. The expression pattern of the various dysbindin isoforms (-1A, -1B, -1C) changes depending upon stage of brain development, tissue areas and subcellular localizations, and can involve interaction with different protein partners. We review evidence describing how sequence variation in DTNBP1 isoforms has been differentially associated with schizophrenia-associated symptoms. We discuss results linking these isoform proteins, and their interacting molecular partners, with cognitive dysfunction in schizophrenia, including evidence from drosophila through to genetic mouse models of dysbindin function. Finally, we discuss preclinical evidence investigating the antipsychotic potential of molecules that influence dysbindin expression and functionality. These studies, and other recent work that has extended this approach to other developmental regulators, may facilitate identification of novel molecular pathways leading to improved antipsychotic treatments.
Collapse
Affiliation(s)
- John L Waddington
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,Jiangsu Key Laboratory of Translational Research & Therapy for Neuro-Psychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Xuechu Zhen
- Jiangsu Key Laboratory of Translational Research & Therapy for Neuro-Psychiatric Disorders and Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Colm M P O'Tuathaigh
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland.,Medical Education Unit, School of Medicine, Brookfield Health Sciences Complex, University College Cork, Cork, Ireland
| |
Collapse
|
30
|
Jalali R, Dutta D. Impact of systemic therapies on cognition in patients with primary brain tumors. CANCER RESEARCH, STATISTICS, AND TREATMENT 2020. [DOI: 10.4103/crst.crst_263_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
31
|
Altshuler DB, Wang L, Zhao L, Miklja Z, Linzey J, Brezzell A, Kakaizada S, Krishna S, Orringer DA, Briceño EM, Gabel N, Hervey-Jumper SL. BDNF, COMT, and DRD2 polymorphisms and ability to return to work in adult patients with low- and high-grade glioma. Neurooncol Pract 2019; 6:375-385. [PMID: 31555452 PMCID: PMC6753359 DOI: 10.1093/nop/npy059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Cognitive and language dysfunction is common among patients with glioma and has a significant impact on survival and health-related quality of life (HRQOL). Little is known about the factors that make individual patients more or less susceptible to the cognitive sequelae of the disease. A better understanding of the individual and population characteristics related to cognitive function in glioma patients is required to appropriately stratify patients, prognosticate, and develop more efficacious treatment regimens. There is evidence that allelic variation among genes involved in neurotransmission and synaptic plasticity are related to neurocognitive performance in states of health and neurologic disease. METHODS We studied the association of single-nucleotide polymorphism variations in brain-derived neurotrophic factor (BDNF, rs6265), dopamine receptor 2 (DRD2, rs1076560), and catechol-O-methyltransferase (COMT, rs4680) with neurocognitive function and ability to return to work in glioma patients at diagnosis and at 3 months. We developed a functional score based on the number of high-performance alleles that correlates with the capacity for patients to return to work. RESULTS Patients with higher-performing alleles have better scores on neurocognitive testing with the Repeatable Battery for the Assessment of Neuropsychological Status and Stroop test, but not the Trail Making Test. CONCLUSIONS A better understanding of the genetic contributors to neurocognitive performance in glioma patients and capacity for functional recovery is necessary to develop improved treatment strategies based on patient-specific factors.
Collapse
Affiliation(s)
| | - Lin Wang
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
| | - Lili Zhao
- Department of Biostatistics, University of Michigan, Ann Arbor, USA
| | - Zachary Miklja
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
| | - Joey Linzey
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
| | - Amanda Brezzell
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
| | - Sofia Kakaizada
- Department of Neurosurgery, University of California San Francisco, USA
| | - Saritha Krishna
- Department of Neurosurgery, University of California San Francisco, USA
| | - Daniel A Orringer
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, USA
| | - Emily M Briceño
- Department of Neurosurgery, Physical Medicine and Rehabilitation, University of Michigan, Ann Arbor, USA
| | - Nicolette Gabel
- Department of Neurosurgery, Physical Medicine and Rehabilitation, University of Michigan, Ann Arbor, USA
| | - Shawn L Hervey-Jumper
- Department of Neurosurgery, University of Michigan, Ann Arbor, USA
- Department of Neurosurgery, University of California San Francisco, USA
| |
Collapse
|
32
|
Siegel BI, King TZ, Rupji M, Dwivedi B, Carter AB, Kowalski J, MacDonald TJ. Host Genome Variation is Associated with Neurocognitive Outcome in Survivors of Pediatric Medulloblastoma. Transl Oncol 2019; 12:908-916. [PMID: 31078964 PMCID: PMC6515414 DOI: 10.1016/j.tranon.2019.03.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 03/20/2019] [Accepted: 03/26/2019] [Indexed: 12/23/2022] Open
Abstract
Host genome analysis is a promising source of predictive information for long-term morbidity in cancer survivors. However, studies on genetic predictors of long-term outcome, particularly neurocognitive function following chemoradiation in pediatric oncology, are limited. Here, we evaluated variation in host genome of long-term survivors of medulloblastoma and its association with neurocognitive outcome. Whole-genome sequencing was conducted on peripheral blood of long-term survivors of pediatric medulloblastoma who also completed neuropsychological testing. Cognitively impaired and less impaired survivors did not differ in exposure to chemoradiation therapy or age at treatment. Unsupervised consensus clustering yielded two distinct variant clusters that were significantly associated with neurocognitive outcome. Interestingly, 34 of the 36 significant variants were found in noncoding DNA regions with unknown regulatory function. A separate unsupervised cluster analysis of variants within DNA repair genes identified discrete variant groups that were not associated with neurocognitive outcome, suggesting that variations in genes corresponding to a single functional group may be insufficient to predict long-term outcome alone. These findings are supportive of the presence of a genetic diathesis for treatment-related neurocognitive morbidity in medulloblastoma that may be driven by variation in noncoding regulatory elements.
Collapse
Affiliation(s)
- Benjamin I Siegel
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA
| | - Tricia Z King
- Department of Psychology and Neuroscience Institute, Georgia State University, Atlanta, GA.
| | - Manali Rupji
- Winship Cancer Institute of Emory University, Atlanta, GA
| | - Bhakti Dwivedi
- Winship Cancer Institute of Emory University, Atlanta, GA
| | - Alexis B Carter
- Department of Pathology and Laboratory Medicine, Children's Healthcare of Atlanta, Atlanta, GA
| | - Jeanne Kowalski
- Winship Cancer Institute of Emory University, Atlanta, GA; Department of Biostatistics and Bioinformatics, Emory University Rollins School of Public Health, Atlanta, GA
| | - Tobey J MacDonald
- Department of Pediatrics, Emory University School of Medicine, Atlanta, GA; Aflac Cancer & Blood Disorders Center, Children's Healthcare of Atlanta, Atlanta, GA
| |
Collapse
|
33
|
Noll KR, Bradshaw ME, Parsons MW, Dawson EL, Rexer J, Wefel JS. Monitoring of Neurocognitive Function in the Care of Patients with Brain Tumors. Curr Treat Options Neurol 2019; 21:33. [PMID: 31250277 DOI: 10.1007/s11940-019-0573-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE OF REVIEW A detailed characterization of the nature of neurocognitive impairment in patients with brain tumors is provided, as well as considerations for clinical practice regarding neuropsychological assessment throughout the disease course. RECENT FINDINGS Neurocognitive impairment is common in patients with brain tumors and may result from the tumor itself, as a consequence of treatment, including surgery, chemotherapy, and radiation, or in association with supportive care medications (e.g., anticonvulsant and pain medications). Serial surveillance of neurocognitive functioning in this population can facilitate medical decision-making and inform recommendations to improve patient daily functioning and quality of life. Neuropsychological assessment is increasingly recognized as a critical component of the multidisciplinary care of patients with brain tumors and has already had practice-changing effects. Further understanding of genetic risk factors for neurocognitive decline along with the development of novel assessment and intervention strategies may further enhance functioning and general well-being in this patient population.
Collapse
Affiliation(s)
- Kyle R Noll
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX, 77030, USA
| | - Mariana E Bradshaw
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX, 77030, USA
| | - Michael W Parsons
- Department of Neuro-Oncology, Psychology Assessment Center, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Erica L Dawson
- Department of Psychiatry and Behavioral Health, The Ohio State University, Columbus, OH, 43210, USA
| | - Jennie Rexer
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX, 77030, USA
| | - Jeffrey S Wefel
- Section of Neuropsychology, Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Unit 431, Houston, TX, 77030, USA. .,Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
34
|
Sleurs C, Madoe A, Lagae L, Jacobs S, Deprez S, Lemiere J, Uyttebroeck A. Genetic Modulation of Neurocognitive Development in Cancer Patients throughout the Lifespan: a Systematic Review. Neuropsychol Rev 2019; 29:190-219. [DOI: 10.1007/s11065-019-09399-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 02/19/2019] [Indexed: 12/14/2022]
|
35
|
Brooks JD, Comen EA, Reiner AS, Orlow I, Leong SF, Liang X, Mellemkjær L, Knight JA, Lynch CF, John EM, Bernstein L, Woods M, Doody DR, Malone KE, Bernstein JL. CYP2D6 phenotype, tamoxifen, and risk of contralateral breast cancer in the WECARE Study. Breast Cancer Res 2018; 20:149. [PMID: 30526633 PMCID: PMC6288916 DOI: 10.1186/s13058-018-1083-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Tamoxifen treatment greatly reduces a woman's risk of developing a second primary breast cancer. There is, however, substantial variability in treatment response, some of which may be attributed to germline genetic variation. CYP2D6 is a key enzyme in the metabolism of tamoxifen to its active metabolites, and variants in this gene have been associated with reduced tamoxifen metabolism. The impact of variation on risk of contralateral breast cancer (CBC) is unknown. METHODS Germline DNA from 1514 CBC cases and 2203 unilateral breast cancer controls was genotyped for seven single nucleotide polymorphisms, one three-nucleotide insertion-deletion, and a full gene deletion. Each variant has an expected impact on enzyme activity, which in combination allows for the classification of women as extensive, intermediate, and poor metabolizers (EM, IM, and PM respectively). Each woman was assigned one of six possible diplotypes and a corresponding CYP2D6 activity score (AS): EM/EM (AS = 2), EM/IM (AS = 1.5), EM/PM (AS = 1), IM/IM (AS = 0.75), IM/PM (AS = 0.5), and PM/PM (AS = 0). We also collapsed categories of the AS to generate an overall phenotype (EM, AS ≥ 1; IM, AS = 0.5-0.75; PM, AS = 0). Rate ratios (RRs) and 95% confidence intervals (CIs) for the association between tamoxifen treatment and risk of CBC in our study population were estimated using conditional logistic regression, stratified by AS. RESULTS Among women with AS ≥ 1 (i.e., EM), tamoxifen treatment was associated with a 20-55% reduced RR of CBC (AS = 2, RR = - 0.81, 95% CI 0.62-1.06; AS = 1.5, RR = 0.45, 95% CI 0.30-0.68; and AS = 1, RR = 0.55, 95% CI 0.40-0.74). Among women with no EM alleles and at least one PM allele (i.e., IM and PM), tamoxifen did not appear to impact the RR of CBC in this population (AS = 0.5, RR = 1.08, 95% CI 0.59-1.96; and AS = 0, RR = 1.17, 95% CI 0.58-2.35) (p for homogeneity = - 0.02). CONCLUSION This study suggests that the CYP2D6 phenotype may contribute to some of the observed variability in the impact of tamoxifen treatment for a first breast cancer on risk of developing CBC.
Collapse
Affiliation(s)
- Jennifer D. Brooks
- University of Toronto, Dalla Lana School of Public Health Sciences, 155 College St. HSB 676, Toronto, ON M5T 3M7 Canada
| | | | - Anne S. Reiner
- Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Irene Orlow
- Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Siok F. Leong
- Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Xiaolin Liang
- Memorial Sloan Kettering Cancer Center, New York, NY USA
| | | | - Julia A. Knight
- University of Toronto, Dalla Lana School of Public Health Sciences, 155 College St. HSB 676, Toronto, ON M5T 3M7 Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, Canada
| | | | - Esther M. John
- Department of Medicine and Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA USA
| | - Leslie Bernstein
- Beckman Research Institute, City of Hope National Medical Centre, Duarte, CA USA
| | - Meghan Woods
- Memorial Sloan Kettering Cancer Center, New York, NY USA
| | | | | | | |
Collapse
|
36
|
Abstract
OBJECTIVES To address the estimated rates of incidence, potential underlying etiologies, and cognitive domains affected from diagnosis and treatment. To describe potential cognitive function interventions. DATA SOURCES PubMed. CONCLUSION Adults with gliomas report that the most distressing, persistent, and greatest negative impact on their lives relates to the cognitive impairment they experience. However, there are several potential interventions that may prevent cognitive decline during treatment or maintain cognitive function long term. IMPLICATIONS FOR NURSING PRACTICE Awareness of cognitive sequela that adults with gliomas face can lead to early identification, full neurocognitive profiling, and implementation of evidence-based interventions for those experiencing cognitive impairments following cancer treatment.
Collapse
|
37
|
Abstract
Survival alone is no longer an adequate outcome for persons with brain tumors; the quality of the survivorship experience should be viewed with equal importance. Symptom management is a significant component of quality survivorship care. Regardless of their histology, brain tumors and therapies used to treat them produce symptoms that affect an individual's ability to function in everyday life. Common symptoms include fatigue, cognitive impairment, distress, and sleep disturbance. Symptom-based interventions for persons with brain tumors focus on prevention, self-management, and prescriptive interventions targeted to these problems. Unfortunately, little evidence exists to support many interventions, making it challenging for clinicians to provide concrete recommendations. Research is needed to provide evidence in support of symptom-based interventions while novel approaches to these challenging problems are developed.
Collapse
Affiliation(s)
- Christina Amidei
- Northwestern Medicine, Neurological Surgery, Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| |
Collapse
|
38
|
Tan CJ, Lim SWT, Toh YL, Ng T, Yeo A, Shwe M, Foo KM, Chu P, Jain A, Koo SL, Dent RA, Ng RCH, Yap YS, Lim EH, Loh KWJ, Chay WY, Lee GE, Tan TJY, Beh SY, Wong M, Chan JJ, Khor CC, Ho HK, Chan A. Replication and Meta-analysis of the Association between BDNF Val66Met Polymorphism and Cognitive Impairment in Patients Receiving Chemotherapy. Mol Neurobiol 2018; 56:4741-4750. [PMID: 30382534 PMCID: PMC6647505 DOI: 10.1007/s12035-018-1410-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 10/24/2018] [Indexed: 01/23/2023]
Abstract
Cancer-related cognitive impairment (CRCI) adversely affects cancer patients. We had previously demonstrated that the BDNF Val66Met genetic polymorphism is associated with lower odds of subjective CRCI in the multitasking and verbal ability domains among breast cancer patients receiving chemotherapy. To further assess our previous findings, we evaluated the association of BDNF Val66Met polymorphism with subjective and objective CRCI in a temporally separate cohort of patients and pooled findings from both the original (n = 145) and current (n = 193) cohorts in a meta-analysis. Subjective CRCI was assessed using FACT-Cog. Objective CRCI was evaluated using computerized neuropsychological tests. Genotyping was carried out using Sanger sequencing. The association of BDNF Val66Met genotypes and CRCI was examined with logistic regression. A fixed-effect meta-analysis was conducted using the inverse variance method. In the meta-analysis (n = 338), significantly lower odds of CRCI were associated with Met allele carriers based on the global FACT-Cog score (OR = 0.52, 95% CI 0.29–0.94). Furthermore, Met allele carriers were at lower odds of developing impairment in the domains of memory (OR = 0.34, 95% CI: 0.17–0.70), multitasking (OR = 0.33, 95% CI: 0.18–0.59), and verbal ability (OR = 0.46, 95% CI: 0.24–0.88). Consistent with the previous study, lower odds of subjective CRCI among patients with the BDNF Met allele was observed after adjusting for potential confounders in the multitasking (OR = 0.30, 95% CI: 0.14–0.67) domain. In conclusion, carriers of the BDNF Met allele were protected against global subjective CRCI, particularly in the domains of memory, multitasking, and verbal ability. Our findings further contribute to the understanding of CRCI pathophysiology.
Collapse
Affiliation(s)
- Chia Jie Tan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Sheree Wan Ting Lim
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Yi Long Toh
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Terence Ng
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Angie Yeo
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Maung Shwe
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Koon Mian Foo
- Department of Pharmacy, K.K. Women's and Children's Hospital, Singapore, Singapore
| | - Pat Chu
- Singapore Cord Blood Bank, Singapore, Singapore
| | - Amit Jain
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Si-Lin Koo
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Rebecca A Dent
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | | | - Yoon Sim Yap
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Elaine H Lim
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Kiley Wei-Jen Loh
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Wen Yee Chay
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Guek Eng Lee
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Tira Jing Ying Tan
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Sok Yuen Beh
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Mabel Wong
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Jack Junjie Chan
- Division of Medical Oncology, National Cancer Centre, Singapore, Singapore
| | - Chiea Chuen Khor
- Human Genetics, Genome Institute of Singapore, Singapore, Singapore.,Glaucoma Research Group, Singapore Eye Research Institute, Singapore, Singapore
| | - Han Kiat Ho
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Alexandre Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Block S4, 18 Science Drive 4, Singapore, 117543, Singapore. .,Department of Pharmacy, National Cancer Centre, Singapore, Singapore. .,Duke-NUS Graduate Medical School, Singapore, Singapore.
| |
Collapse
|
39
|
Brain structure and function in patients with ovarian cancer treated with first-line chemotherapy: a pilot study. Brain Imaging Behav 2018; 11:1652-1663. [PMID: 27766586 DOI: 10.1007/s11682-016-9608-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Women with ovarian cancer often undergo chemotherapy involving multiple agents. However, little is known about treatment-related central neurotoxicity in this population. The goal of this cross-sectional study was to assess brain structure and function and neurocognitive abilities in patients with ovarian cancer following first-line chemotherapy. Eighteen patients with ovarian, peritoneal and fallopian tube cancer and eighteen healthy controls matched for gender, age and education participated in the study. The patients were evaluated 1-4 months following completion of first-line taxane/platinum chemotherapy. All participants underwent structural and functional magnetic resonance imaging (MRI), and completed neuropsychological tests of attention, memory and executive functions. Neuroimaging assessments included voxel-based morphometry (VBM) for measuring gray matter (GM) volume, and functional MRI (fMRI) during the N-back working memory task. The results of VBM showed that patients had significantly reduced GM volume compared to healthy controls in the right middle/superior frontal gyrus, and in the left supramarginal gyrus and left inferior parietal lobule. fMRI results indicated significantly decreased activation in patients relative to healthy controls in the left middle frontal gyrus and left inferior parietal lobule during the N-back task (1/2/3-back >0-back). There were no statistically significant differences between the two groups on the neuropsychological tests. This is the first study showing structural and functional alterations involving frontal and parietal regions in patients with ovarian cancer treated with first-line chemotherapy. These findings are congruent with studies involving women with breast cancer, and provide additional supporting evidence for central neurotoxicity associated with taxane/platinum chemotherapy.
Collapse
|
40
|
Matsos A, Loomes M, Zhou I, Macmillan E, Sabel I, Rotziokos E, Beckwith W, Johnston I. Chemotherapy-induced cognitive impairments: White matter pathologies. Cancer Treat Rev 2017; 61:6-14. [DOI: 10.1016/j.ctrv.2017.09.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/27/2017] [Accepted: 09/28/2017] [Indexed: 10/18/2022]
|
41
|
Correa DD, Kryza-Lacombe M, Zhou X, Baser RE, Beattie BJ, Beiene Z, Humm J, DeAngelis LM, Orlow I, Weber W, Osborne J. A pilot study of neuropsychological functions, APOE and amyloid imaging in patients with gliomas. J Neurooncol 2017; 136:613-622. [PMID: 29168082 PMCID: PMC5807139 DOI: 10.1007/s11060-017-2692-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Accepted: 11/18/2017] [Indexed: 12/27/2022]
Abstract
Brain tumor patients treated with radiotherapy (RT) often develop cognitive dysfunction, and recent studies suggest that the APOE ε-4 allele may influence cognitive outcome. The ε-4 allele is known to promote beta (β) amyloid deposition in the cortex, and preliminary evidence suggests that RT may be associated with this process. However, it is unknown whether β-amyloid accumulation contributes to treatment neurotoxicity. In this pilot study, we assessed neuropsychological functions and β-amyloid retention using 18F-florbetaben (FBB) PET in a subset of brain tumor patients who participated in our study of APOE polymorphisms and cognitive functions. Twenty glioma patients treated with conformal RT ± chemotherapy participated in the study: 6 were APOE ε-4 carriers and 14 were non-ε-4 carriers. Patients completed a neuropsychological re-evaluation (mean time interval = 5 years, SD = 0.83) and brain MRI and FBB PET scans. Wilcoxon signed-rank test comparisons between prior and current neuropsychological assessments showed a significant decline in attention (Brief Test of Attention, p = 0.018), and a near significant decline in verbal learning (Hopkins Verbal learning Test-Learning, p = 0.07). Comparisons by APOE status showed significant differences over time in attention/working memory (WAIS-III digits forward, p = 0.028 and digits backward, p = 0.032), with a decline among APOE ε-4 carriers. There were no significant differences in any of the FBB PET analyses between APOE ε-4 carriers and non-ε-4 carriers. The findings suggest that glioma patients may experience worsening in attention and executive functions several years after treatment, and that the APOE ε-4 allele may modulate cognitive decline, but independent of increased β-amyloid deposition.
Collapse
Affiliation(s)
- D D Correa
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10021, USA. .,Department of Neurology, Weill Cornell Medical College, New York, NY, USA.
| | - M Kryza-Lacombe
- San Diego Joint Doctoral Program in Clinical Psychology, San Diego State University/University of California, San Diego, CA, USA
| | - X Zhou
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - R E Baser
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - B J Beattie
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Z Beiene
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - J Humm
- Department of Medical Physics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - L M DeAngelis
- Department of Neurology, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY, 10021, USA.,Department of Neurology, Weill Cornell Medical College, New York, NY, USA
| | - I Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - W Weber
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| | - J Osborne
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA.,Department of Radiology, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
42
|
van Kessel E, Baumfalk AE, van Zandvoort MJE, Robe PA, Snijders TJ. Tumor-related neurocognitive dysfunction in patients with diffuse glioma: a systematic review of neurocognitive functioning prior to anti-tumor treatment. J Neurooncol 2017; 134:9-18. [PMID: 28567586 PMCID: PMC5543199 DOI: 10.1007/s11060-017-2503-z] [Citation(s) in RCA: 126] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 05/20/2017] [Indexed: 11/25/2022]
Abstract
Deficits in neurocognitive functioning (NCF) frequently occur in glioma patients. Both treatment and the tumor itself contribute to these deficits. Data about the role of the tumor are scarce, because NCF has mostly been studied postoperatively. We aimed to summarize data on pre-treatment NCF in glioma patients and to determine the overall and domain-specific prevalence of neurocognitive dysfunction. We searched PubMed and Embase according to PRISMA-P protocol for studies that evaluated pre-treatment NCF in glioma patients (1995-November 2016) and extracted information about NCF. We performed analysis of data for two main outcome measures; mean cognitive functioning of the study sample (at group level) and the percentage of impaired patients (at individual level). We included 23 studies. Most studies were small observational prospective cohort studies. In 11 (47.5%) studies, patient selection was based on tumor location. NCF was analyzed at the group level in 14 studies, of which 13 (92.9%) found decreased NCF at group level, compared to normative data or matched controls. The proportion of individuals with decreased NCF was reported in 15 studies. NCF was impaired (in any domain) in 62.6% of the individuals (median; interquartile range 31.0–79.0). Cognitive impairments were more common in patients with high-grade glioma than with low-grade glioma (OR 2.50; 95% CI 1.71–3.66). Cognitive impairment occurs in the majority of treatment-naive glioma patients, suggesting that neurocognitive dysfunction is related to the tumor. However, the literature about pre-treatment NCF in glioma patients is characterized by small-scale studies and strong heterogeneity in patient selection, resulting in high risk of bias.
Collapse
Affiliation(s)
- Emma van Kessel
- Department of Neurology & Neurosurgery, University Medical Center Utrecht/Brain Center Rudolf Magnus, G03.232, PO Box 85500, 3508 XC, Utrecht, The Netherlands.
| | - Anniek E Baumfalk
- Department of Neurology & Neurosurgery, University Medical Center Utrecht/Brain Center Rudolf Magnus, G03.232, PO Box 85500, 3508 XC, Utrecht, The Netherlands
| | - Martine J E van Zandvoort
- Department of Neurology & Neurosurgery, University Medical Center Utrecht/Brain Center Rudolf Magnus, G03.232, PO Box 85500, 3508 XC, Utrecht, The Netherlands
- Helmhotz Institute, Utrecht University, Room 1715, Heidelberglaan 1, 3584 CS, Utrecht, The Netherlands
| | - Pierre A Robe
- Department of Neurology & Neurosurgery, University Medical Center Utrecht/Brain Center Rudolf Magnus, G03.232, PO Box 85500, 3508 XC, Utrecht, The Netherlands
| | - Tom J Snijders
- Department of Neurology & Neurosurgery, University Medical Center Utrecht/Brain Center Rudolf Magnus, G03.232, PO Box 85500, 3508 XC, Utrecht, The Netherlands
| |
Collapse
|
43
|
Klumpers MJ, Coenen MJ, Gidding CE, Te Loo DMW. The role of germline variants in chemotherapy outcome in brain tumors: a systematic review of pharmacogenetic studies. Pharmacogenomics 2017; 18:501-513. [PMID: 28346057 DOI: 10.2217/pgs-2016-0189] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM This systematic review provides an overview of publications concerning pharmacogenetic research in pediatric patients with medulloblastoma and low-grade glioma. MATERIALS & METHODS Three electronic databases searches including a manual search were performed to identify studies investigating potential interactions between germline variants and chemotherapy efficacy and toxicity. RESULTS Out of 3570 citations, 21 studies were included. Outcomes include overall survival, progression-free survival and treatment-related adverse events (n = 5), cisplatin-induced ototoxicity (n = 13) and vincristine-induced neurotoxicity (n = 3). CONCLUSION This review shows that the number of pharmacogenetic studies in well-defined pediatric brain tumor cohorts is poor and studies often report conflicting results. Large-scale international collaborations allowing analysis of sufficiently sized cohorts are therefore very important for the future of personalized medicine in brain tumors.
Collapse
Affiliation(s)
- Marije J Klumpers
- Department of Pediatric Oncology, Department of Pediatrics, Radboud university medical center, Nijmegen, The Netherlands
| | - Marieke Jh Coenen
- Department of Human Genetics, Radboud Institute for Health Sciences, Radboud university medical center, Nijmegen, The Netherlands
| | - Corrie Em Gidding
- Department of Pediatric Oncology, Department of Pediatrics, Radboud university medical center, Nijmegen, The Netherlands
| | - D Maroeska Wm Te Loo
- Department of Pediatric Oncology, Department of Pediatrics, Radboud university medical center, Nijmegen, The Netherlands
| |
Collapse
|
44
|
BDNF Variants May Modulate Long-Term Visual Memory Performance in a Healthy Cohort. Int J Mol Sci 2017; 18:ijms18030655. [PMID: 28304362 PMCID: PMC5372667 DOI: 10.3390/ijms18030655] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 03/06/2017] [Accepted: 03/13/2017] [Indexed: 12/31/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is involved in numerous cognitive functions including learning and memory. BDNF plays an important role in synaptic plasticity in humans and rats with BDNF shown to be essential for the formation of long-term memories. We previously identified a significant association between the BDNF Val66Met polymorphism (rs6265) and long-term visual memory (p-value = 0.003) in a small cohort (n = 181) comprised of healthy individuals who had been phenotyped for various aspects of memory function. In this study, we have extended the cohort to 597 individuals and examined multiple genetic variants across both the BDNF and BDNF-AS genes for association with visual memory performance as assessed by the Wechsler Memory Scale—Fourth Edition subtests Visual Reproduction I and II (VR I and II). VR I assesses immediate visual memory, whereas VR II assesses long-term visual memory. Genetic association analyses were performed for 34 single nucleotide polymorphisms genotyped on Illumina OmniExpress BeadChip arrays with the immediate and long-term visual memory phenotypes. While none of the BDNF and BDNF-AS variants were shown to be significant for immediate visual memory, we found 10 variants (including the Val66Met polymorphism (p-value = 0.006)) that were nominally associated, and three variants (two variants in BDNF and one variant in the BDNF-AS locus) that were significantly associated with long-term visual memory. Our data therefore suggests a potential role for BDNF, and its anti-sense transcript BDNF-AS, in long-term visual memory performance.
Collapse
|
45
|
Sui J, Li YH, Zhang YQ, Li CY, Shen X, Yao WZ, Peng H, Hong WW, Yin LH, Pu YP, Liang GY. Integrated analysis of long non-coding RNA‑associated ceRNA network reveals potential lncRNA biomarkers in human lung adenocarcinoma. Int J Oncol 2016; 49:2023-2036. [PMID: 27826625 DOI: 10.3892/ijo.2016.3716] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Accepted: 09/27/2016] [Indexed: 11/06/2022] Open
Abstract
Accumulating evidence has highlighted the important roles of long non-coding RNAs (lncRNAs) acting as competing endogenous RNAs (ceRNAs) in tumor biology. However, the roles of cancer specific lncRNAs in lncRNA-related ceRNA network of lung adenocarcinoma (LUAD) are still unclear. In the present study, the 465 RNA sequencing profiles in LUAD patients were obtained from the cancer genome atlas (TCGA) database, which provides large sample RNA sequencing data free of charge, and 41 cancer specific lncRNAs, 25 miRNAs and 1053 mRNAs (fold change >2, p<0.05) were identified. Then, the lncRNA-miRNA-mRNA ceRNA network of LUAD was constructed with 29 key lncRNAs, 24 miRNAs and 72 mRNAs. Subsequently, we selected these 29 key lncRNAs to analyze their correlation with clinical features, and 21 of them were aberrantly expressed with tumor pathological stage, TNM staging system, lymph node metastasis and patient outcome assessment, respectively. Furthermore, there were 5 lncRNAs (BCRP3, LINC00472, CHIAP2, BMS1P20 and UNQ6494) positively correlated with overall survival (OS, log-rank p<0.05). Finally, 7 cancer specific lncRNAs were randomly selected to verify the expression in 53 newly diagnosed LUAD patients using qRT-PCR. The expression results between TCGA and qRT-PCR were 100% in agreement. The correlation between AFAP1-AS1 and LINC00472 and clinical features were also confirmed. Thus, our results showed the lncRNA expression profiles and we constructed an lncRNA-miRNA-mRNA ceRNA network in LUAD. The present study provides novel insight for better understanding of lncRNA-related ceRNA network in LUAD and facilitates the identification of potential biomarkers for diagnosis and prognosis.
Collapse
Affiliation(s)
- Jing Sui
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yun-Hui Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yan-Qiu Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Cheng-Yun Li
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Xian Shen
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Wen-Zhuo Yao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Hui Peng
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Wei-Wei Hong
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Li-Hong Yin
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Yue-Pu Pu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| | - Ge-Yu Liang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, Jiangsu 210009, P.R. China
| |
Collapse
|
46
|
Bergo E, Lombardi G, Pambuku A, Della Puppa A, Bellu L, D'Avella D, Zagonel V. Cognitive Rehabilitation in Patients with Gliomas and Other Brain Tumors: State of the Art. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3041824. [PMID: 27493954 PMCID: PMC4963561 DOI: 10.1155/2016/3041824] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Revised: 05/26/2016] [Accepted: 06/02/2016] [Indexed: 01/01/2023]
Abstract
Disease prognosis is very poor in patients with brain tumors. Cognitive deficits due to disease or due to its treatment have an important weight on the quality of life of patients and caregivers. Studies often take into account quality of life as a fundamental element in the management of disease and interventions have been developed for cognitive rehabilitation of neuropsychological deficits with the aim of improving the quality of life and daily-life autonomy of patients. In this literature review, we will consider the published studies of cognitive rehabilitation over the past 20 years.
Collapse
Affiliation(s)
- E. Bergo
- Department of Clinical and Experimental Oncology, Medical Oncology 1 Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
| | - G. Lombardi
- Department of Clinical and Experimental Oncology, Medical Oncology 1 Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
| | - A. Pambuku
- Department of Clinical and Experimental Oncology, Medical Oncology 1 Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
| | - A. Della Puppa
- Neurosurgery Department, Azienda Ospedaliera di Padova, Padua, Italy
| | - L. Bellu
- Department of Clinical and Experimental Oncology, Medical Oncology 1 Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
| | - D. D'Avella
- Neurosurgery Department, University of Padua, Padua, Italy
| | - V. Zagonel
- Department of Clinical and Experimental Oncology, Medical Oncology 1 Unit, Veneto Institute of Oncology IOV-IRCCS, 35128 Padua, Italy
| |
Collapse
|