1
|
Devi N, Madaan P, Kandoth N, Lal P, Sahu JK, Bansal D. First-choice hormonal therapies for children with infantile epileptic spasms syndrome in South Asia: A network meta-analysis of randomized controlled trials. Epilepsia Open 2024. [PMID: 39513499 DOI: 10.1002/epi4.13086] [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: 05/08/2024] [Revised: 09/16/2024] [Accepted: 10/11/2024] [Indexed: 11/15/2024] Open
Abstract
Considering the peculiar challenges with infantile epileptic spasms syndrome (IESS) in South Asia and a wide variation in the usage of hormonal therapies, we compared the efficacy and safety of various hormonal therapies for children with IESS in South Asia. We searched PubMed, Embase, Scopus, and Web of Science databases from the inception until April 2024. We included only randomized clinical trials (RCTs) evaluating the efficacy and safety of hormonal therapies for IESS in the South Asian region. Complete cessation of epileptic spasms (ES), electro-clinical response, and time taken to be spasm-free at 2 or 6 weeks of therapy were efficacy outcomes, while the occurrence of adverse events was the safety outcome. Effect estimates were reported as odds ratio (OR) or mean difference (MD) with 95% confidence intervals (CI) and Cochrane risk of bias 2.0 (ROB 2.0) used for quality assessment of each study. The surface under the cumulative ranking curve (SUCRA) was used to rank the different therapies and reported as a p-score ranging from 0 to 1. Of 747 citations, nine RCTs comprising 566 children with IESS were included. After 2-week treatment, dexamethasone (OR: 6.72; 95% CI: 1.47, 30.72), adrenocorticotropic hormone therapy (ACTH) high dose (HD) (OR: 5.30; 95% CI: 1.05, 26.91), and prednisolone HD (OR: 2.41; 95% CI:1.07, 5.46) had shown significantly greater efficacy for cessation of EScompared with ACTH low dose (LD). Similarly, for electroclinical response, dexamethasone (OR: 9.63; 95% CI: 1.99, 46.70) and prednisolone HD (OR: 3.46; 95% CI: 1.38, 8.68) had greater efficacy compared with ACTH LD. Safety outcomes revealed that hypertension was significantly less common with ACTH LD and prednisolone HD as compared with ACTH HD. This study provides quality evidence on preferred first-choice hormonal therapy for managing IESS in South Asia. ACTH HD, dexamethasone, and prednisolone HD are the most effective hormonal therapy options with dose-dependent therapeutic efficacy. PLAIN LANGUAGE SUMMARY: This study provides insights into the selection of first-line hormonal therapies among the various treatments for managing infantile epileptic spasms syndrome (IESS) in South Asia. The study findings suggested that the effectiveness of these therapies is dose-dependent, with high doses of ACTH, dexamethasone, and prednisolone being the most effective for achieving cessation of epileptic spasms.
Collapse
Affiliation(s)
- Nagita Devi
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Mohali, Punjab, India
- Department of Pharmacy Practice, Chandigarh College of Pharmacy, Landran, Punjab, India
| | - Priyanka Madaan
- Department of Pediatric Neurology, Amrita School of Medicine, Amrita Vishwavidyapeetham, Faridabad, India
| | - Nidhun Kandoth
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Mohali, Punjab, India
| | - Parth Lal
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Jitendra Kumar Sahu
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Dipika Bansal
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research, Mohali, Punjab, India
| |
Collapse
|
2
|
Arredondo KH, Jülich K, Roach ES. Tuberous sclerosis complex: Diagnostic features, surveillance, and therapeutic strategies. Semin Pediatr Neurol 2024; 51:101155. [PMID: 39389658 DOI: 10.1016/j.spen.2024.101155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/12/2024]
Abstract
Tuberous sclerosis complex (TSC) is a rare neurocutaneous disorder of mTOR pathway dysregulation resulting from pathogenic variants in the TSC1 or TSC2 genes. Expression of this disorder may involve abnormal tissue growth and dysfunction within the brain, kidneys, heart, lungs, eyes, skin, bones, and teeth. Neurological manifestations can include subependymal giant cell astrocytomas (SEGAs), high rates of infantile spasms, drug-resistant epilepsy, developmental delay, cognitive impairment, autism spectrum disorder, and other neurobehavioral manifestations. Here we review the potential clinical manifestations of TSC by system, recommended diagnostic and surveillance testing, genetic testing, currently available therapeutic options, and considerations for education and social support resources given the unique challenges of this multi-system disorder.
Collapse
Affiliation(s)
- Kristen H Arredondo
- Department of Neurology, The University of Texas at Austin Dell Medical School, Austin, TX.
| | - Kristina Jülich
- Department of Neurology, The University of Texas at Austin Dell Medical School, Austin, TX.
| | - E Steve Roach
- Department of Neurology, The University of Texas at Austin Dell Medical School, Austin, TX.
| |
Collapse
|
3
|
Aungaroon G, Cooke A, Ritter D, Krueger D, Horn P, Franz DN. Cenobamate's Efficacy for Seizure Treatment in Tuberous Sclerosis Complex. Pediatr Neurol 2024; 161:201-207. [PMID: 39426344 DOI: 10.1016/j.pediatrneurol.2024.09.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 08/16/2024] [Accepted: 09/20/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Epilepsy is prevalent, and seizure control is challenging in patients with tuberous sclerosis complex (TSC). Cenobamate (CBM) has proven efficacy in several studies; however, its benefit in the TSC population is not known. METHODS We performed a retrospective review of patients with TSC who received adjunctive CBM for seizure treatments. We assessed treatment efficacy by comparing seizure frequencies three months before CBM (baseline) and those at 3-, 6-, 12-, and 18- month follow-ups. RESULTS We identified 70 patients with TSC receiving CBM and excluded 16 with insufficient data. Fifty-four patients aged 2 to 39 years, with an average baseline seizure of 66.1 ± 88.9 per month, were analyzed. Treatment retention rates at 3, 6, 12, and 18 months were 94.4%, 79.6%, 66.7%, 44.4%, and responder rates (proportions of patients who remained on treatment and had ≥50% seizure reduction) were 38.1%, 51.7%, 53.1%, and 59.1%, respectively. Seizure-free rates at these respective follow-ups were 7.1%, 13.8%, 6.3%, and 9.1%. For patients experiencing reduced seizures, the mean percentage of change ranged from 61.5% to 74.6%. Side effects were common (64.8%), particularly sedation (42.6%), behavioral disturbance (24.1%), and gastrointestinal disturbance (22.2%). CONCLUSIONS Most patients in this study showed seizure reduction; however, the overall responder and seizure-free rates were lower than the literature, likely due to the unique underlying epileptogenesis in TSC and the challenges of tolerating CBM. The lower treatment retention rates signal areas for improvement in concurrent medication adjustment practices.
Collapse
Affiliation(s)
- Gewalin Aungaroon
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Pediatrics, University of Cincinnati, Cincinnati, Ohio.
| | - Alexander Cooke
- University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - David Ritter
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Darcy Krueger
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - Paul Horn
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| | - David N Franz
- Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio; Division of Pediatrics, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
4
|
Mao D, Li S, Xu Y, Chen H, Liu P, Hu W. Factors influencing efficacy and relapse of adrenocorticotropic hormone in infantile epileptic spasms syndrome. Epilepsy Behav 2024; 161:110055. [PMID: 39312843 DOI: 10.1016/j.yebeh.2024.110055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/22/2024] [Accepted: 09/14/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Infantile epileptic spasms syndrome (IESS) is a severe epileptic condition characterized by persistent uncontrolled seizures, with some children experiencing recurrent seizures despite multiple pharmacological therapies. The prognostic risk factorsassociated with IESS remain unclear. This study aimed to evaluate the factors influencing the efficacy and relapse of adrenocorticotropic hormone (ACTH) treatment for IESS in infants, as well as to assess the correlation between the Burden of Amplitudes and Epileptiform Discharges (BASED) score and clinical outcomes. METHODS A retrospective analysis was conducted on a cohort of 88 pediatric patients diagnosed with IESS who received ACTH therapy at our hospital from February 2016 to August 2023. Patients were categorized into response (n = 47) and non-response (n = 41) groups based on their treatment response at day 28. Responders were further classified into relapse and non-relapse groups. A modified Poisson regression model and receiver operating characteristic (ROC) curves were employed to evaluate the positive predictive values. RESULTS In this study, a total of 47 patients (53.4 %) responded to ACTH treatment. Patients in the response group demonstrated significantly greater reductions in BASED scores by day 14 of ACTH treatment, yielding an area under the curve (AUC) of 0.859 (95 % CI: 0.782-0.937, P<0.05), with a sensitivity of 68.1 % and a specificity of 95.1 %. The optimal cut-off point was established at ≥ 2, corresponding to a Youden index of 0.632. Notably, patientswho were on anti-seizure medications (ASMs) before ACTH treatment and those with developmental delay prior to the onset of spasms exhibited lower short-term response rates (P<0.05), although these factors did not demonstrate predictive value. Among the responders, 20 cases (42.6 %) experienced a relapse, with only those patients showing specific abnormalities on cranial magnetic resonance imaging (MRI) exhibiting a statistically higher proportion of relapse. CONCLUSION Patients receiving ASMs before ACTH treatment and those with developmental delays prior to the onset of spasms may have a less favorable therapeutic response. A reduction in BASED scores of 2 or greater by day 14 of ACTH treatment may signify a potentially positive treatment response. Additionally, patients with IESS who present with specific abnormalities on cranial MRI may have an increased likelihood of relapse following ACTH treatment.
Collapse
Affiliation(s)
- Dandan Mao
- Pediatric Neurology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
| | - Sixiu Li
- Pediatric Neurology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
| | - Yang Xu
- Pediatric Neurology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
| | - Hui Chen
- Pediatric Neurology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
| | - Ping Liu
- Pediatric Neurology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China
| | - Wenguang Hu
- Pediatric Neurology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan 611731, China.
| |
Collapse
|
5
|
He W, Wang QH, Li JW, Wang YY, Luo XM, Wan L, Wang J, Shi XY, Zhang WH, Fang F, Zou LP. Adrenocorticotropic hormone combined with magnesium sulfate therapy for infantile epileptic spasms syndrome: a real-world study. World J Pediatr 2024; 20:834-847. [PMID: 38070098 PMCID: PMC11402841 DOI: 10.1007/s12519-023-00771-1] [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: 06/14/2023] [Accepted: 10/08/2023] [Indexed: 09/16/2024]
Abstract
BACKGROUND Infantile epileptic spasms syndrome (IESS) is a serious disease in infants, and it usually evolves to other epilepsy types or syndromes, especially refractory or super-refractory focal epilepsies. Although adrenocorticotropic hormone (ACTH) is one of the first-line and effective treatment plans for IESS, it has serious side effects and is not sufficiently effective. METHODS A retrospective study of the clinical outcomes of ACTH combined with magnesium sulfate (MgSO4) therapy for IESS in two hospital centers was conducted. The major outcome of the single and combined treatment was evaluated by changes in seizure frequency and improvements in hypsarrhythmia electroencephalography (EEG). To reduce the confounding bias between the two groups, we used SPSS for the propensity score matching (PSM) analysis. RESULTS We initially recruited 1205 IESS patients from two Chinese hospitals and treated them with ACTH combined with MgSO4 and ACTH alone. Only 1005 patients were enrolled in the treatment (ACTH combined with MgSO4: 744, ACTH: 261), and both treatment plans had a more than 55% response rate. However, compared to patients treated with ACTH alone, those patients treated with ACTH combined with MgSO4 had better performance in terms of the seizure frequency and hypsarrhythmia EEG. After PSM, the two groups also showed significant differences in responder rate [70.8% (95% confidence interval, CI) = 66.7%-74.8%) vs. 53.8% (95% CI = 47.4%-60.2%), P < 0.001], seizure frequency (P < 0.001) and hypsarrhythmia EEG resolution (P < 0.001). Notably, multivariate analysis revealed that the lead time to treatment and the number of antiseizure medications taken before treatment were two factors that may affect the clinical outcome. Patients with less than 3 months of lead time responded to the treatment much better than those with > 3 months (P < 0.05). In addition, the overall incidence of adverse reactions in the ACTH combined with MgSO4 group was much lower than that in the ACTH group (31.4% vs. 63.1%, P < 0.001). During the treatment, only infection (P = 0.045) and hypertension (P = 0.025) were significantly different between the two groups, and no baby died. CONCLUSION Our findings support that ACTH combined with MgSO4 is a more effective short-term treatment protocol for patients with IESS than ACTH alone, especially for those patients with short lead times to treatment. Video Abstract (MP4 533623 KB).
Collapse
Affiliation(s)
- Wen He
- Senior Department of Pediatrics, the Seventh Medical Center of Chinese PLA General Hospital, Department of Pediatrics, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Qiu-Hong Wang
- Senior Department of Pediatrics, the Seventh Medical Center of Chinese PLA General Hospital, Department of Pediatrics, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Jiu-Wei Li
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yang-Yang Wang
- Senior Department of Pediatrics, the Seventh Medical Center of Chinese PLA General Hospital, Department of Pediatrics, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xiao-Mei Luo
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing 100069, China
| | - Lin Wan
- Senior Department of Pediatrics, the Seventh Medical Center of Chinese PLA General Hospital, Department of Pediatrics, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Jing Wang
- Senior Department of Pediatrics, the Seventh Medical Center of Chinese PLA General Hospital, Department of Pediatrics, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Xiu-Yu Shi
- Senior Department of Pediatrics, the Seventh Medical Center of Chinese PLA General Hospital, Department of Pediatrics, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China
| | - Wei-Hua Zhang
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Fang Fang
- Department of Neurology, Beijing Children's Hospital, Capital Medical University, Beijing, China.
| | - Li-Ping Zou
- Senior Department of Pediatrics, the Seventh Medical Center of Chinese PLA General Hospital, Department of Pediatrics, the First Medical Center of Chinese PLA General Hospital, Beijing 100853, China.
- Center of Epilepsy, Beijing Institute for Brain Disorders, Beijing 100069, China.
| |
Collapse
|
6
|
Winther CCH, Klein-Petersen AW, Preel M, Kofoed IR, Bo Nissen I, Axelgaard S, Green J, Miranda MJ, Hoei-Hansen CE. Epidemiology and outcome of infantile spasms in Denmark in 1996-2019. Seizure 2024; 120:173-179. [PMID: 39029407 DOI: 10.1016/j.seizure.2024.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/09/2024] [Accepted: 07/09/2024] [Indexed: 07/21/2024] Open
Abstract
PURPOSE To investigate the treatment of infantile epileptic spasm syndrome (IESS) in Denmark. METHODS National retrospective cohort study of all patients born 1996-2019 who had a diagnosis of IESS in the National Patient Registry. Medical records were reviewed to evaluate the diagnosis. Patients were included if semiology was compatible with IESS, or if unclear semiology if there was an abnormal EEG or EEG with hypsarrhythmia. RESULTS Number of cases with a register based IESS diagnosis was 538. Medical records were unavailable in 48 and 164 did not fulfil the inclusion criteria. Thereby the cohort consisted of 326 children. Mean age at onset of IESS was 5.9 months and mean lead time to treatment was 26.6 days (SD= 63.5). Consistent with the Danish treatment guidelines most patients received vigabatrin as first treatment. In the cohort 44.7 % of patients solely received vigabatrin, whereas combined vigabatrin and corticosteroid was given to 28.3 % (either hydrocortisone or prednisolone). Other anti-seizure medication was given to 28.4 % within 90 days of IESS onset. Aetiology was prenatal (40.3 %), perinatal (10.5 %), postnatal (3.7 %), with unknown timing (10.2 %) or with unknown aetiology (33.5 %). The cohort was followed to a mean age of 8.2 years. At latest follow-up severe neurodevelopmental outcome was seen in 44.2 % and 76.4 % still had epilepsy. The incidence of IESS was 22 per 100.000 live births. CONCLUSION In Denmark treatment algorithm is based on start of treatment with vigabatrin. A total of 44.7 % became seizure free by vigabatrin. Neurodevelopmental outcome was severe. A national incidence could be established.
Collapse
Affiliation(s)
- Camille Caroline Højer Winther
- Department of Paediatric and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark; Department Paediatric and Adolescent Medicine, University Hospital of Herlev and Gentofte, Herlev, Denmark.
| | | | - Marie Preel
- Department of Paediatric and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark
| | - Inge Ring Kofoed
- Department of Paediatrics, University Hospital of Aalborg, Aalborg, Denmark
| | - Ida Bo Nissen
- Department of Paediatrics, Kolding University Hospital, Kolding, Denmark
| | - Sofie Axelgaard
- Department of Paediatrics, Herning Hospital, Herning, Denmark
| | - Julie Green
- Department of Paediatrics, University Hospital of Aarhus, Aarhus, Denmark
| | - Maria J Miranda
- Department Paediatric and Adolescent Medicine, University Hospital of Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark
| | - Christina Engel Hoei-Hansen
- Department of Paediatric and Adolescent Medicine, University Hospital Rigshospitalet, Copenhagen, Denmark; Department of Clinical Medicine, University of Copenhagen, Denmark
| |
Collapse
|
7
|
Wang X, Huang Q, Wu L, Yang Y, Ye X, Yang B. Application of peripheral blood cytokine and immunoglobulin detection in ACTH therapy for the treatment of infantile spasms. Front Pediatr 2024; 12:1365917. [PMID: 39055621 PMCID: PMC11270505 DOI: 10.3389/fped.2024.1365917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 06/10/2024] [Indexed: 07/27/2024] Open
Abstract
Objective This research aims to investigate the levels of lymphocytes, immunoglobulins, and cytokines in children with infantile spasms (IS) before and after adrenocorticotropic hormone (ACTH) therapy and to explore the application of these markers in evaluating the therapeutic effects of ACTH on infantile spasms. Methods From May to November 2022, 35 children initially diagnosed with IS and treated at our hospital were regarded as the observation group, and 35 healthy children who underwent physical examination at our hospital during the same period were regarded as the control group. Children in the observation group received intramuscular injections of ACTH for 2 weeks. Fasting venous blood was collected from the control group and the observation group before and after ACTH therapy. Serum levels of immunoglobulins IgG, IgA, and IgM in serum were detected by immunoturbidimetry. T-cell subsets (CD3+, CD3+CD4+, and CD3+CD8+) and B-cell subsets [CD3-CD19+ and CD3-CD16+CD56+ natural killer (NK) cells] were detected by flow cytometry, and the ratio of CD3+CD4+/CD3+CD8+ was calculated. Serum levels of interleukin-1β (IL-1β), interleukin-2R (IL-2R), and interleukin-6 (IL-6) cytokines were detected by the enzyme-linked immunosorbent assay, and changes in serum cytokine and immunoglobulin levels in the two groups were compared before therapy, whereas in observation group one, these comparisons were made both before and after ACTH therapy. Results Compared to the control group, the observation group showed significantly increased serum levels of immunoglobulins IgG and IgM before therapy, while the level of IgA was significantly decreased (p < 0.05). Also, the percentage of CD3-CD19+ B cells was significantly increased, while the percentages of CD3+ T cells and CD3+CD4+ T cells were significantly decreased (p < 0.05). The percentages of CD3+CD8+ T cells, CD3-CD16+CD56+ NK cells, and CD3+CD4+/CD3+CD8+ cells did not change significantly (p > 0.05); the levels of cytokines IL-1 β, IL-2R, and IL-6 were significantly increased (p < 0.05). Compared to levels before treatment, the serum level of immunoglobulin IgG in the observation group after ACTH therapy was significantly reduced (p < 0.05), while the IgA and IgM levels did not change significantly (p > 0.05). The percentages of CD3+ T cells and CD3+CD4+ T cells were significantly increased, while the percentages of CD3-CD16+CD56+ NK cells and CD3-CD19+ B cells were significantly decreased (p < 0.05); however, the percentages of CD3+CD8+ T cells and the CD3+CD4+/CD3+CD8+ ratio did not change significantly (p > 0.05). Furthermore, the levels of cytokines IL-1 β, IL-2R, and IL-6 were significantly reduced (p < 0.05). Conclusion Children with IS exhibit immune dysfunction, and the changes in serological immune indices after ACTH treatment indicate that ACTH may control seizures in IS children by regulating and improving immune dysfunction. Therefore, the therapeutic effects of ACTH on IS can be evaluated by detecting the levels of cytokines and immunoglobulins.
Collapse
Affiliation(s)
| | | | | | | | | | - Bin Yang
- Department of Neurology, Anhui Provincial Children’s Hospital, Hefei, Anhui, China
| |
Collapse
|
8
|
Rajaraman RR, Smith RJ, Oana S, Daida A, Shrey DW, Nariai H, Lopour BA, Hussain SA. Computational EEG attributes predict response to therapy for epileptic spasms. Clin Neurophysiol 2024; 163:39-46. [PMID: 38703698 DOI: 10.1016/j.clinph.2024.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 03/10/2024] [Accepted: 03/28/2024] [Indexed: 05/06/2024]
Abstract
OBJECTIVE We set out to evaluate whether response to treatment for epileptic spasms is associated with specific candidate computational EEG biomarkers, independent of clinical attributes. METHODS We identified 50 children with epileptic spasms, with pre- and post-treatment overnight video-EEG. After EEG samples were preprocessed in an automated fashion to remove artifacts, we calculated amplitude, power spectrum, functional connectivity, entropy, and long-range temporal correlations (LRTCs). To evaluate the extent to which each feature is independently associated with response and relapse, we conducted logistic and proportional hazards regression, respectively. RESULTS After statistical adjustment for the duration of epileptic spasms prior to treatment, we observed an association between response and stronger baseline and post-treatment LRTCs (P = 0.042 and P = 0.004, respectively), and higher post-treatment entropy (P = 0.003). On an exploratory basis, freedom from relapse was associated with stronger post-treatment LRTCs (P = 0.006) and higher post-treatment entropy (P = 0.044). CONCLUSION This study suggests that multiple EEG features-especially LRTCs and entropy-may predict response and relapse. SIGNIFICANCE This study represents a step toward a more precise approach to measure and predict response to treatment for epileptic spasms.
Collapse
Affiliation(s)
- Rajsekar R Rajaraman
- Division of Pediatric Neurology, UCLA Mattel Children's Hospital and University of California, Los Angeles, Los Angeles, CA, USA
| | - Rachel J Smith
- Department of Electrical and Computer Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shingo Oana
- Division of Pediatric Neurology, UCLA Mattel Children's Hospital and University of California, Los Angeles, Los Angeles, CA, USA
| | - Atsuro Daida
- Division of Pediatric Neurology, UCLA Mattel Children's Hospital and University of California, Los Angeles, Los Angeles, CA, USA
| | - Daniel W Shrey
- Division of Pediatric Neurology, University of California, Irvine, Irvine, CA, USA; Department of Neurology, Children's Hospital of Orange County, Orange, CA, USA
| | - Hiroki Nariai
- Division of Pediatric Neurology, UCLA Mattel Children's Hospital and University of California, Los Angeles, Los Angeles, CA, USA
| | - Beth A Lopour
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, USA
| | - Shaun A Hussain
- Division of Pediatric Neurology, UCLA Mattel Children's Hospital and University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
9
|
Macdonald-Laurs E, Dzau W, Warren AEL, Coleman M, Mignone C, Stephenson SEM, Howell KB. Identification and treatment of surgically-remediable causes of infantile epileptic spasms syndrome. Expert Rev Neurother 2024; 24:661-680. [PMID: 38814860 DOI: 10.1080/14737175.2024.2360117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
INTRODUCTION Infantile epileptic spasms syndrome (IESS) is a common developmental and epileptic encephalopathy with poor long-term outcomes. A substantial proportion of patients with IESS have a potentially surgically remediable etiology. Despite this, epilepsy surgery is underutilized in this patient group. Some surgically remediable etiologies, such as focal cortical dysplasia and malformation of cortical development with oligodendroglial hyperplasia in epilepsy (MOGHE), are under-diagnosed in infants and young children. Even when a surgically remediable etiology is recognised, for example, tuberous sclerosis or focal encephalomalacia, epilepsy surgery may be delayed or not considered due to diffuse EEG changes, unclear surgical boundaries, or concerns about operating in this age group. AREAS COVERED In this review, the authors discuss the common surgically remediable etiologies of IESS, their clinical and EEG features, and the imaging techniques that can aid in their diagnosis. They then describe the surgical approaches used in this patient group, and the beneficial impact that early epilepsy surgery can have on developing brain networks. EXPERT OPINION Epilepsy surgery remains underutilized even when a potentially surgically remediable cause is recognized. Overcoming the barriers that result in under-recognition of surgical candidates and underutilization of epilepsy surgery in IESS will improve long-term seizure and developmental outcomes.
Collapse
Affiliation(s)
- Emma Macdonald-Laurs
- Department of Neurology, The Royal Children's Hospital, Parkville, VIC, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Winston Dzau
- Neurosciences Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Aaron E L Warren
- Department of Medicine (Austin Health), The University of Melbourne, Melbourne, VIC, Australia
- Brigham and Women's Hospital, Harvard Medical School, Massachusetts, USA
| | - Matthew Coleman
- Neurosciences Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Cristina Mignone
- Department of Medical Imaging, The Royal Children's Hospital, Parkville, VIC, Australia
| | - Sarah E M Stephenson
- Neurosciences Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
- Department of Paediatrics, The University of Melbourne, Melbourne, VIC, Australia
| | - Katherine B Howell
- Department of Neurology, The Royal Children's Hospital, Parkville, VIC, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Parkville, VIC, Australia
| |
Collapse
|
10
|
Deckard E, Sathe R, Tabibzadeh D, Terango A, Groves A, Rajaraman RR, Nariai H, Hussain SA. Epileptic spasms relapse is associated with response latency but not conventional attributes of post-treatment EEG. Epilepsia Open 2024; 9:1034-1041. [PMID: 38588009 PMCID: PMC11145600 DOI: 10.1002/epi4.12931] [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: 09/20/2023] [Revised: 02/06/2024] [Accepted: 03/09/2024] [Indexed: 04/10/2024] Open
Abstract
OBJECTIVE Relapse of epileptic spasms after initial treatment of infantile epileptic spasms syndrome (IESS) is common. However, past studies of small cohorts have inconsistently linked relapse risk to etiology, treatment modality, and EEG features upon response. Using a large single-center IESS cohort, we set out to quantify the risk of epileptic spasms relapse and identify specific risk factors. METHODS We identified all children with epileptic spasms at our center using a clinical EEG database. Using the electronic medical record, we confirmed IESS syndrome classification and ascertained treatment, response, time to relapse, etiology, EEG features, and other demographic factors. Relapse-free survival analysis was carried out using Cox proportional hazards regression. RESULTS Among 599 children with IESS, 197 specifically responded to hormonal therapy and/or vigabatrin (as opposed to surgery or other second-line treatments). In this study, 41 (21%) subjects exhibited relapse of epileptic spasms within 12 months of response. Longer duration of IESS prior to response (>3 months) was strongly associated with shorter latency to relapse (hazard ratio = 3.11; 95% CI 1.59-6.10; p = 0.001). Relapse was not associated with etiology, developmental status, or any post-treatment EEG feature. SIGNIFICANCE This study suggests that long duration of IESS before response is the single largest clinical predictor of relapse risk, and therefore underscores the importance of prompt and successful initial treatment. Further study is needed to evaluate candidate biomarkers of epileptic spasms relapse and identify treatments to mitigate this risk. PLAIN LANGUAGE SUMMARY Relapse of infantile spasms is common after initially successful treatment. With study of a large group of children with infantile spasms, we determined that relapse is linked to long duration of infantile spasms. In contrast, relapse was not associated with the cause of infantile spasms, developmental measures, or EEG features at the time of initial response. Further study is needed to identify tools to predict impending relapse of infantile spasms.
Collapse
Affiliation(s)
- Emmi Deckard
- Department of PediatricsDivision of NeurologyUniversity of California Los Angeles and UCLA Mattel Children's HospitalCaliforniaLos AngelesUSA
| | - Rujuta Sathe
- Department of PediatricsDivision of NeurologyUniversity of California Los Angeles and UCLA Mattel Children's HospitalCaliforniaLos AngelesUSA
| | - David Tabibzadeh
- Department of PediatricsDivision of NeurologyUniversity of California Los Angeles and UCLA Mattel Children's HospitalCaliforniaLos AngelesUSA
| | - Aria Terango
- Department of PediatricsDivision of NeurologyUniversity of California Los Angeles and UCLA Mattel Children's HospitalCaliforniaLos AngelesUSA
| | - Aran Groves
- Department of PediatricsDivision of NeurologyUniversity of California Los Angeles and UCLA Mattel Children's HospitalCaliforniaLos AngelesUSA
| | - Rajsekar R. Rajaraman
- Department of PediatricsDivision of NeurologyUniversity of California Los Angeles and UCLA Mattel Children's HospitalCaliforniaLos AngelesUSA
| | - Hiroki Nariai
- Department of PediatricsDivision of NeurologyUniversity of California Los Angeles and UCLA Mattel Children's HospitalCaliforniaLos AngelesUSA
| | - Shaun A. Hussain
- Department of PediatricsDivision of NeurologyUniversity of California Los Angeles and UCLA Mattel Children's HospitalCaliforniaLos AngelesUSA
| |
Collapse
|
11
|
Russo A, Mazzone S, Landolina L, Colucci R, Baccari F, Fetta A, Boni A, Cordelli DM. Efficacy and Safety of Pulse Intravenous Methylprednisolone in Pediatric Epileptic Encephalopathies: Timing and Networks Consideration. J Clin Med 2024; 13:2497. [PMID: 38731025 PMCID: PMC11084200 DOI: 10.3390/jcm13092497] [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: 03/05/2024] [Revised: 04/10/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
Background: Epileptic encephalopathies (EE) are characterized by severe drug-resistant seizures, early onset, and unfavorable developmental outcomes. This article discusses the use of intravenous methylprednisolone (IVMP) pulse therapy in pediatric patients with EE to evaluate its efficacy and tolerability. Methods: This is a retrospective study from 2020 to 2023. Inclusion criteria were ≤18 years at the time of IVMP pulse therapy and at least 6 months of follow-up. Efficacy and outcome, defined as seizure reduction > 50% (responder rate), were evaluated at 6 and 9 months of therapy, and 6 months after therapy suspension; quality of life (QoL) was also assessed. Variables predicting positive post-IVMP outcomes were identified using statistical analysis. Results: The study included 21 patients, with a responder rate of 85.7% at 6 and 9 months of therapy, and 80.9% at 6 months after therapy suspension. Variables significantly predicting favorable outcome were etiology (p = 0.0475) and epilepsy type (p = 0.0475), with the best outcome achieved in patients with genetic epilepsy and those with encephalopathy related to electrical status epilepticus during slow-wave sleep (ESES). All patients evidenced improvements in QoL at the last follow-up, with no relevant adverse events reported. Conclusions: Our study confirmed the efficacy and high tolerability of IVMP pulse therapy in pediatric patients with EE. Genetic epilepsy and ESES were positive predictors of a favorable clinical outcome. QOL, EEG tracing, and postural-motor development showed an improving trend as well. IVMP pulse therapy should be considered earlier in patients with EE.
Collapse
Affiliation(s)
- Angelo Russo
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria Dell’età Pediatrica, 40139 Bologna, Italy; (S.M.); (L.L.); (R.C.); (A.F.); (A.B.); (D.M.C.)
| | - Serena Mazzone
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria Dell’età Pediatrica, 40139 Bologna, Italy; (S.M.); (L.L.); (R.C.); (A.F.); (A.B.); (D.M.C.)
| | - Laura Landolina
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria Dell’età Pediatrica, 40139 Bologna, Italy; (S.M.); (L.L.); (R.C.); (A.F.); (A.B.); (D.M.C.)
- Dipartimento di Scienze Mediche e Chirurgiche (DIMEC), Università di Bologna, 40126 Bologna, Italy
| | - Roberta Colucci
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria Dell’età Pediatrica, 40139 Bologna, Italy; (S.M.); (L.L.); (R.C.); (A.F.); (A.B.); (D.M.C.)
- Dipartimento di Scienze Mediche e Chirurgiche (DIMEC), Università di Bologna, 40126 Bologna, Italy
| | - Flavia Baccari
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, UOS Epidemiologia e Biostatistica, 40139 Bologna, Italy;
| | - Anna Fetta
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria Dell’età Pediatrica, 40139 Bologna, Italy; (S.M.); (L.L.); (R.C.); (A.F.); (A.B.); (D.M.C.)
- Dipartimento di Scienze Mediche e Chirurgiche (DIMEC), Università di Bologna, 40126 Bologna, Italy
| | - Antonella Boni
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria Dell’età Pediatrica, 40139 Bologna, Italy; (S.M.); (L.L.); (R.C.); (A.F.); (A.B.); (D.M.C.)
| | - Duccio Maria Cordelli
- IRCCS, Istituto delle Scienze Neurologiche di Bologna, UOC Neuropsichiatria Dell’età Pediatrica, 40139 Bologna, Italy; (S.M.); (L.L.); (R.C.); (A.F.); (A.B.); (D.M.C.)
- Dipartimento di Scienze Mediche e Chirurgiche (DIMEC), Università di Bologna, 40126 Bologna, Italy
| |
Collapse
|
12
|
Ramani PK, Briscoe Abath C, Donatelli S, Hadjinicolaou A, Vega Toro S, Acevedo K, Astorga KR, Parbhoo K, Singh A, Catenaccio E, Jain P, Sahu JK, Samanta D, Harini C. Initial combination versus early sequential standard therapies for Infantile Epileptic Spasms Syndrome-Feedback from stakeholders. Epilepsia Open 2024; 9:819-822. [PMID: 38217384 PMCID: PMC10984285 DOI: 10.1002/epi4.12895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 12/26/2023] [Indexed: 01/15/2024] Open
Affiliation(s)
- Praveen Kumar Ramani
- Department of Pediatrics, Division of Pediatric NeurologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Christina Briscoe Abath
- Division of Pediatric NeurologyChildren's Hospital of Philadelphia, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Stephanie Donatelli
- Department of Neurology, Boston Children's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| | - Aristides Hadjinicolaou
- Division of Neurology, Department of Pediatrics, Comprehensive Epilepsy ProgramCHU Sainte‐JustineMontrealQuebecCanada
| | - Sebastian Vega Toro
- Pediatric Neurology Unit, Department of PediatricsHospital Carlos Van BurenValparaísoChile
| | - Keryma Acevedo
- Pediatric Neurology Unit, Division of PediatricsPontificia Universidad Católica de ChileSantiagoChile
| | - Karina Rosso Astorga
- Pediatric Neurology Unit, Department of PediatricsHospital Carlos Van BurenValparaísoChile
| | - Kaajal Parbhoo
- Division of Paediatric NeurologyNelson Mandela Children's HospitalJohannesburgSouth Africa
| | - Avantika Singh
- Department of Neurology, Division of Child NeurologyMedical College of WisconsinMilwaukeeWisconsinUSA
| | - Eva Catenaccio
- Division of Pediatric NeurologyChildren's Hospital of Philadelphia, Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Puneet Jain
- Division of Neurology, Department of Pediatrics, Epilepsy ProgramHospital for Sick ChildrenTorontoOntarioCanada
| | - Jitendra Kumar Sahu
- Pediatric Neurology UnitPostgraduate Institute of Medical Education & ResearchChandigarhIndia
| | - Debopam Samanta
- Department of Pediatrics, Division of Pediatric NeurologyUniversity of Arkansas for Medical SciencesLittle RockArkansasUSA
| | - Chellamani Harini
- Department of Neurology, Boston Children's HospitalHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
13
|
Bashiri FA, Hundallah K, Al-Baradie R, Al-Otaibi A, Ismayl O, AlMalik ME, Muthaffar OY, Futaisi AA, Kurdi D, Tawari AA, AlSowat D, Shafi SA, Ali A, AlHajjar LM, Aldakhil A. Diagnosis and management of infantile epileptic spasms syndrome (IESS) in Gulf Cooperation Council (GCC) countries: Expert consensus statement. Seizure 2024; 117:174-182. [PMID: 38432081 DOI: 10.1016/j.seizure.2024.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/20/2024] [Accepted: 02/22/2024] [Indexed: 03/05/2024] Open
Abstract
Despite the availability of international recommendations for the management of Infantile Epileptic Spasms Syndrome (IESS), there is a lack of recommendations adapted to the local context of clinical practice of pediatric neurology in the Gulf Cooperation Council (GCC) countries. By an initiative from the Saudi Pediatric Neurology Society (SPNS), a literature review was performed and an expert panel comprised of 13 pediatric neurologists from all GCC countries (Saudi Arabia, Kuwait, Bahrain, Oman, Qatar, and the United Arab Emirates) was subsequently convened to discuss all issues related to the management and diagnosis practices of IESS in the GCC. The overall aim of this consensus document was to develop practical recommendations to support the care of patients with IESS in the GCC and to reflect on how clinical management approaches compare with those adopted internationally.
Collapse
Affiliation(s)
- Fahad A Bashiri
- Pediatric Neurology Division, Department of Pediatrics, College of Medicine, King Saud University Medical City, King Saud University, Riyadh, Saudi Arabia.
| | - Khalid Hundallah
- Pediatric Neurology Division, Department of Pediatrics, Prince Sultan Military Medical City, Riyadh 13317, Saudi Arabia.
| | - Raidah Al-Baradie
- Neuroscience Center, King Fahd Specialist Hospital, PO Box 15215, Dammam 314444, Kingdom of Saudi Arabia.
| | - Ali Al-Otaibi
- Pediatric Neurology Division, Department of Neurophysiology, National Neuroscience Institute, King Fahad Medical City, As Sulimaniyah, Riyadh 12231, Saudi Arabia.
| | - Omar Ismayl
- Department of Child Neurology, Sheikh Khalifah Medical City, Al Karamah Street, Abu Dhabi, United Arab Emirates.
| | - Mohamed Elhadi AlMalik
- Department of Pediatric Neurology, Mediclinic Al Jowhara Hospital, Al Ain City, United Arab Emirates.
| | - Osama Y Muthaffar
- Pediatric Neurology Division, Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Amna Al Futaisi
- Pediatric Neurology Division, Child Health Department, College of Medicine and Health Sciences, Sultan Qaboos University, P.O.Box 35, P.C 123 Al Khoud, Sultanate of Oman.
| | - Daniah Kurdi
- Pediatric Neurology Division, Department of Pediatrics, College of Medicine, King Saud University, Riyadh 12372, Saudi Arabia.
| | - Asmaa Al Tawari
- Pediatric Neurology Division, Department of Pediatrics, Al Sabah Hospital, Ministry of Health, Sabah Health Region, Shuwaikh Industrial, State of Kuwait.
| | - Daad AlSowat
- Pediatric Neurology Division, Department of Pediatrics, King Faisal Specialist Hospital & Research Centre, Riyadh 11211, Saudi Arabia.
| | - Shatha Al Shafi
- Neurology Division, Epilepsy and EEG Fellowship Program, Department of Pediatrics, Prince Sultan Military Medical City, Riyadh 13317, Saudi Arabia.
| | - Ayman Ali
- Pediatric Neurology Division, Department of Pediatrics, AlSalam Specialist Hospital, Riffa 80278, Bahrain.
| | - Lynn M AlHajjar
- Itkan Health Consulting Group, Al Olaya, Riyadh 12221, Saudi Arabia.
| | - Abdullah Aldakhil
- Itkan Health Consulting Group, Al Olaya, Riyadh 12221, Saudi Arabia.
| |
Collapse
|
14
|
Hollenshead PP, Jackson CN, Cross JV, Witten TE, Anwar AI, Ahmadzadeh S, Shekoohi S, Kaye AD. Treatment modalities for infantile spasms: current considerations and evolving strategies in clinical practice. Neurol Sci 2024; 45:507-514. [PMID: 37736852 DOI: 10.1007/s10072-023-07078-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 09/13/2023] [Indexed: 09/23/2023]
Abstract
Infantile spasms, newly classified as infantile epileptic spasm syndrome (IESS), occur in children under 2 years of age and present as an occur as brief, symmetrical, contractions of the musculature of the neck, trunk, and extremities. When infantile spasms occur with a concomitant hypsarrhythmia on electroencephalogram (EEG) and developmental regression, it is known as West Syndrome. There is no universally accepted mainstay of treatment for this condition, but some options include synthetic adrenocorticotropic hormone (ACTH), repository corticotropin injection (RCI/Acthar Gel), corticosteroids, valproic acid, vigabatrin, and surgery. Without effective treatment, infantile spasms can cause an impairment of psychomotor development and/or cognitive and behavioral functions. The first-line treatment in the USA is ACTH related to high efficacy for cessation of infantile spasms long-term and low-cost profile. Acthar Gel is a repository corticotropin intramuscular injection that became FDA-approved for the treatment of IESS in 2010. Though it is believed that ACTH, Acthar Gel, and corticosteroids all work via a negative feedback pathway to decrease corticotropin-releasing hormone (CRH) release, their safety and efficacy profiles all vary. Vigabatrin and valproic acid are both anti-seizure medications that work by increasing GABA concentrations in the CNS and decreasing excitatory activity. Acthar Gel has been shown to have superior efficacy and a diminished side effect profile when compared with other treatment modalities.
Collapse
Affiliation(s)
- Payton P Hollenshead
- School of Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, 71103, USA
| | - Corrie N Jackson
- School of Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, 71103, USA
| | - Jordan V Cross
- School of Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, 71103, USA
| | - Taylor E Witten
- School of Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, 71103, USA
| | - Ahmed I Anwar
- Department of Behavioral Neuroscience, Quinnipiac University, 275 Mount Carmel Ave, Hamden, CT, 06518, USA
| | - Shahab Ahmadzadeh
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, 71103, USA
| | - Sahar Shekoohi
- Department of Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, 71103, USA.
| | - Alan D Kaye
- Department of Anesthesiology, Department of Pharmacology, Toxicology, and Neurosciences, Louisiana State University Health Sciences Center at Shreveport, Shreveport, LA, 71103, USA
| |
Collapse
|
15
|
Zheng Q, He S, Xu SL, Ma MD, Fan M, Ge JF. Pharmacokinetics and tissue distribution of vigabatrin enantiomers in rats. Saudi Pharm J 2024; 32:101934. [PMID: 38223203 PMCID: PMC10787297 DOI: 10.1016/j.jsps.2023.101934] [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: 09/04/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024] Open
Abstract
Purpose To investigate the pharmacokinetics and tissue distribution of VGB racemate and its single enantiomers, and explore the potential of clinic development for single enantiomer S-VGB. Methods In the pharmacokinetics study, male Sprague-Dawley rats were gavaged with VGB racemate or its single enantiomers dosing 50, 100 or 200 mg/kg, and the blood samples were collected during 12 h at regular intervals. In the experiment of tissue distribution, VGB and its single enantiomers were administered intravenously dosing 200 mg/kg, and the tissues including heart, liver, spleen, lung and kidney, eyes, hippocampus, and prefrontal cortex were separated at different times. The concentrations of R-VGB and S-VGB in the plasma and tissues were measured using HPLC. Results Both S-VGB and R-VGB could be detected in the plasma of rats administered with VGB racemate, reaching Cmax at approximately 0.5 h with t1/2 2-3 h. There was no significant pharmacokinetic difference between the two enantiomers when VGB racemate was given 200 mg/kg and 100 mg/kg. However, when given at the dose of 50 mg/kg, S-VGB presented a shorter t1/2 and a higher Cl/F than R-VGB, indicating a faster metabolism of S-VGB. Furthermore, when single enantiomer was administered respectively, S-VGB presented a slower metabolism than R-VGB, as indicated by a longer t1/2 and MRT but a lower Cmax. Moreover, compared with the VGB racemate, the single enantiomers S-VGB and R-VGB had shorter t1/2 and MRT, higher Cmax and AUC/D, and lower Vz/F and Cl/F, indicating the stronger oral absorption and faster metabolism of single enantiomer. In addition, regardless of VGB racemate administration or single enantiomer administration, S-VGB and R-VGB had similar characteristics in tissue distribution, and the content of S-VGB in hippocampus, prefrontal cortex and liver was much higher than that of R-VGB. Conclusions Although there is no transformation between S-VGB and R-VGB in vivo, those two enantiomers display certain disparities in the pharmacokinetics and tissue distribution, and interact with each other. These findings might be a possible interpretation for the pharmacological and toxic effects of VGB and a potential direction for the development and optimization of the single enantiomer S-VGB.
Collapse
Affiliation(s)
- Qiang Zheng
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, PR China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, Anhui 230032, PR China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Shuai He
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, PR China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, Anhui 230032, PR China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Song-Lin Xu
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, PR China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, Anhui 230032, PR China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Meng-Die Ma
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, PR China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, Anhui 230032, PR China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Min Fan
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, PR China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, Anhui 230032, PR China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, PR China
| | - Jin-Fang Ge
- School of Pharmacy, Anhui Medical University, Hefei, Anhui 230032, PR China
- Anhui Provincial Laboratory of Inflammatory and Immune Disease, Anhui Institute of Innovative Drugs, Hefei, Anhui 230032, PR China
- The Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, Anhui 230032, PR China
| |
Collapse
|
16
|
Kuchenbuch M, Lo Barco T, Chemaly N, Chiron C, Nabbout R. Fifteen years of real-world data on the use of vigabatrin in individuals with infantile epileptic spasms syndrome. Epilepsia 2024; 65:430-444. [PMID: 37872396 DOI: 10.1111/epi.17808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 10/14/2023] [Accepted: 10/19/2023] [Indexed: 10/25/2023]
Abstract
OBJECTIVE This study was undertaken to evaluate our treatment algorithm for infantile epileptic spasms syndrome (IESS) used between 2000 and 2018. We initiated vigabatrin (VGB), and steroids were added if the electroclinical response (spasms and electroencephalogram [EEG]) to VGB was not obtained or incomplete. METHODS Individuals with IESS treated with VGB were recruited from our hospital clinical data warehouse based on electronic health records (EHRs) generated since 2009 and containing relevant keywords. We confirmed the diagnosis of IESS. Clinical, EEG, imaging, and biological data were extracted from the EHRs. We analyzed factors associated with short-term response, time to response, relapse, time to relapse of spasms, and the presence of spasms at last follow-up. RESULTS We collected data from 198 individuals (female: 46.5%, IESS onset: 6 [4.5-10.3] months, follow-up: 4.6 [2.5-7.6] years, median [Q1-Q3]) including 129 (65.2%) with identifiable etiology. VGB was started 17 (5-57.5) days after IESS diagnosis. A total of 113 individuals were responders (57.1% of the cohort), 64 with VGB alone and 38 with VGB further combined with steroids (56.6% and 33.6% of responders, respectively). Among responders, 33 (29%) experienced relapses of spasms, mostly those with later onset of spasms (p = .002) and those who received VGB for <24 months after spasms cessation compared to a longer duration on VGB (45% vs. 12.8%, p = .003). At follow-up, 92 individuals were seizure-free (46.5% of the whole cohort), including 26 free of therapy (13.1%). One hundred twelve individuals (56.6%) were still receiving VGB, with a duration of 3.2 (1.75-5.7) years. SIGNIFICANCE Our sequential protocol introducing VGB then adding steroids is an effective alternative to a combined VGB-steroids approach in IESS. It avoids steroid-related adverse events, as well as those from VGB-steroid combination. According to our data, a period of 7 days seems sufficient to assess VGB response and enables the addition of steroids rapidly if needed. Continuing VGB for 2 years may balance the risk of relapse and treatment-induced adverse events.
Collapse
Affiliation(s)
- Mathieu Kuchenbuch
- Department of Pediatric Neurology, Reference Center for Rare Epilepsies, Hôpital Necker-Enfants Malades, member of ERN EpiCARE, Paris, France
- Laboratory of Translational Research for Neurological Disorders, INSERM MR1163, Imagine Institute, Paris, France
- Service de Pédiatrie, Reference Center for Rare Epilepsies, member of ERN EpiCARE, Université de Lorraine, CHRU-Nancy, Nancy, France
| | - Tommaso Lo Barco
- Department of Pediatric Neurology, Reference Center for Rare Epilepsies, Hôpital Necker-Enfants Malades, member of ERN EpiCARE, Paris, France
| | - Nicole Chemaly
- Department of Pediatric Neurology, Reference Center for Rare Epilepsies, Hôpital Necker-Enfants Malades, member of ERN EpiCARE, Paris, France
- Laboratory of Translational Research for Neurological Disorders, INSERM MR1163, Imagine Institute, Paris, France
| | - Catherine Chiron
- Department of Pediatric Neurology, Reference Center for Rare Epilepsies, Hôpital Necker-Enfants Malades, member of ERN EpiCARE, Paris, France
| | - Rima Nabbout
- Department of Pediatric Neurology, Reference Center for Rare Epilepsies, Hôpital Necker-Enfants Malades, member of ERN EpiCARE, Paris, France
- Laboratory of Translational Research for Neurological Disorders, INSERM MR1163, Imagine Institute, Paris, France
- Université de Paris Cité, Paris, France
| |
Collapse
|
17
|
Abath CB, Gupta N, Hadjinicolaou A, Donatelli S, Singh A, Merchant S, Ryan ME, Soby M, Ryan C, Nelson AK, Maldonado Pacheco JE, Zhang B, Williams DN, Yuskaitis CJ, Harini C. Delays to care in infantile epileptic spasms syndrome: Racial and ethnic inequities. Epilepsia 2024; 65:107-114. [PMID: 37953072 DOI: 10.1111/epi.17827] [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: 05/30/2023] [Revised: 11/06/2023] [Accepted: 11/09/2023] [Indexed: 11/14/2023]
Abstract
OBJECTIVE Non-Hispanic (NH) Black children are less likely to receive a standard treatment course for infantile epileptic spasms syndrome (IESS) than White/NH children at pediatric tertiary care epilepsy centers in the United States. However, if inequities exist in time to diagnosis is unknown. Diagnostic delays as little as 1 week can be associated with worse developmental outcomes. METHODS Diagnostic delays were evaluated in a retrospective cohort of 100 children with new onset IESS between January 2019 and May 2022. RESULTS Children with Black, Indigenous, and People of Color (BIPOC) caregivers were more likely to experience clinically significant delays in referral from first provider to neurologist, when compared to White/NH children, even after controlling for other demographic and clinical variables (odds ratio = 4.98, confidence interval = 1.24-19.94, p = .023). SIGNIFICANCE Disproportionate diagnostic delays place BIPOC children at risk of adverse developmental and epilepsy outcomes. Further interventional prospective and qualitative studies are needed to address inequities in care.
Collapse
Affiliation(s)
- Christina Briscoe Abath
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Nishtha Gupta
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| | | | - Stephanie Donatelli
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Avantika Singh
- Department of Neurology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Sabrina Merchant
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Morgan E Ryan
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Meghann Soby
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Christopher Ryan
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Adrianne Katrina Nelson
- Tulane University School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - John E Maldonado Pacheco
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Bo Zhang
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| | - David N Williams
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Christopher J Yuskaitis
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Chellamani Harini
- Department of Neurology, Comprehensive Epilepsy Program, Division of Epilepsy, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
18
|
Sun Y, Chen J, Shi X, Li Z, Wan L, Yan H, Chen Y, Wang J, Wang J, Zou L, Reiter R, Zhang B, Yang G. Safety and efficacy of melatonin supplementation as an add-on treatment for infantile epileptic spasms syndrome: A randomized, placebo-controlled, double-blind trial. J Pineal Res 2024; 76:e12922. [PMID: 37909654 DOI: 10.1111/jpi.12922] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 08/19/2023] [Accepted: 10/20/2023] [Indexed: 11/03/2023]
Abstract
This was a prospective, randomized, double-blind, single-center placebo-controlled trial to assess the efficacy and safety of melatonin as an add-on treatment for infantile epileptic spasms syndrome (IESS). Participants aged 3 months to 2 years with a primary diagnosis of IESS were recruited and assigned to two groups in a 1:1 ratio. Both treatment groups received a combination of adrenocorticotrophic hormone (ACTH) and magnesium sulfate (MgSO4 ) for 2 weeks, and the treatment group also received melatonin (3 mg) between 20:00 and 21:00 daily, 0.5-1 h before bedtime. The study's primary endpoint was the average reduction rate in spasm frequency assessed by seizure diaries. Secondary endpoints included assessment of the response rate, EEG hypsarrhythmia (Kramer score), and psychomotor development (Denver Developmental Screening Test, DDST). Sleep quality was assessed by using the Brief Infant Sleep Questionnaire (BISQ), the Infant Sleep Assessment Scale (ISAS), and actigraphy. Safety parameters were also evaluated. Statistical analyses were conducted on intention-to-treat and per-protocol populations. The trial is registered at Clinicaltrials.gov (ChiCTR2000036208). Out of 119 screened patients, 70 were randomized and 66 completed treatments. In the intention-to-treat population, there were no significant differences in the average percentage reduction of spasm frequency (median [interquartile range, IQR: Q3-Q1], 100% [46.7%] vs. 66.7% [55.3%], p = .288), the 3-day response rate (51.4% vs. 37.1%, p = .229), the 28-day response rate (42.9% vs. 28.6%, p = .212), EEG Kramer scores (2 [3.5] vs. 2 [3], p = .853), or DDST comprehensive months (5 [2.5] vs. 6 [6], p = .239) between the melatonin (n = 35) and placebo (n = 35) groups. However, caregivers reported improved sleep quality after melatonin treatment, with 85.7% reporting regular sleep compared to 42.9% with placebo (42.9%, p < .001). The melatonin group had lower ISAS scores in 4-11-month-old patients compared to the placebo (mean ± SD, 29.3 ± 4.4 vs. 35.2 ± 5.9, p < .001). Moreover, the median (IQR) value of sleep-onset latency was shortened by 6.0 (24.5) min after melatonin treatment, while that in the placebo group was extended by 3.0 (22.0) min (p = .030). The serum melatonin (6:00 h) level (pg/mL) of the children in the melatonin group after treatment was significantly higher than in the placebo group (median [IQR], 84.8 [142] vs. 17.5 [37.6], p < .001). No adverse effects related to melatonin were observed in the study, and there were no significant differences in adverse effects between the melatonin and placebo groups. Although not statistically significant, the results of this randomized clinical trial proved that melatonin supplementation, as an add-on treatment, can improve spasm control rate in the treatment of IESS. For IESS children treated with ACTH, the addition of melatonin was found to improve sleep quality, shorten sleep onset latency, and increase blood melatonin levels. Moreover, it was observed to be a safe treatment option.
Collapse
Affiliation(s)
- Yulin Sun
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- Tongji University School of Medicine, Shanghai, China
| | - Jian Chen
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiuyu Shi
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zhichao Li
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lin Wan
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Huimin Yan
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yuehao Chen
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jiaxin Wang
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jing Wang
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Liping Zou
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Russel Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, Texas, USA
| | - Bo Zhang
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Biostatistics and Research Design Center, Institutional Centers for Clinical & Translational Research, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Guang Yang
- Senior Department of Pediatrics, The Seventh Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Pediatrics, The First Medical Center of Chinese PLA General Hospital, Beijing, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
19
|
Pavone P, Falsaperla R. Real-life data comparing the efficacy of vigabatrin and oral steroids given sequentially or combined for infantile epileptic spasms syndrome. Eur J Paediatr Neurol 2024; 48:A1. [PMID: 38429149 DOI: 10.1016/j.ejpn.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Affiliation(s)
- Piero Pavone
- Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, 95124, Catania, Italy.
| | - Raffaele Falsaperla
- Section of Pediatrics and Child Neuropsychiatry, Department of Clinical and Experimental Medicine, University of Catania, 95124, Catania, Italy
| |
Collapse
|
20
|
Dozieres-Puyravel B, Nasser H, Mauvais FX, De Saint Martin A, Perriard C, Di Meglio C, Cances C, Hachon-LE Camus C, Milh M, Auvin S. Real-life data comparing the efficacy of vigabatrin and oral steroids given sequentially or combined for infantile epileptic spasms syndrome. Eur J Paediatr Neurol 2024; 48:61-66. [PMID: 38041897 DOI: 10.1016/j.ejpn.2023.11.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Revised: 11/19/2023] [Accepted: 11/20/2023] [Indexed: 12/04/2023]
Abstract
AIMS The prognosis of Infantile epileptic spasm syndrome (IESS), relates to the underlying etiology and delay in controlling epileptic spasms. Based on the spasm-free rate, a randomized controlled trial has demonstrated the superiority of combining oral steroids and vigabatrin over oral steroids alone but confirmation in real-life conditions is mandatory. METHODS We compared two real-life IESS cohorts: a multicenter, retrospective cohort of 40 infants treated with vigabatrin followed by a sequential (ST) addition of steroids, and a prospective, single-center cohort of 58 infants treated with an immediate combination of vigabatrin and steroids (CT). RESULTS The two cohorts were similar. When the rate of spasm-free infants in the two cohorts was compared on day 14, a significant difference was observed between the ST (27,5 %) and CT cohorts (64 %) (p < 0.0004). This difference remained significant on day 30, with 55 % spasm-free patients in the ST cohort compared to 76 % in the CT cohort (p = 0.03). After the infants had received both vigabatrin and steroids, without taking into account the time point after treatment initiation, no significant difference was observed in the spasm-free rate between the two cohorts (p = 0.38). INTERPRETATION Real-life data confirm the interest of combination therapy as a first-line treatment for IESS.
Collapse
Affiliation(s)
| | - Hala Nasser
- Service de Physiologie - Explorations Fonctionnelles Pédiatriques - Centre Pédiatrique des Pathologies du Sommeil, AP-HP, Hôpital Universitaire Robert Debré, F-75019, Paris, France.
| | - François-Xavier Mauvais
- Service de Physiologie - Explorations Fonctionnelles Pédiatriques - Centre Pédiatrique des Pathologies du Sommeil, AP-HP, Hôpital Universitaire Robert Debré, F-75019, Paris, France; Université Paris Cité, CNRS, Inserm, Institut Necker-Enfants Malades, F- 75015, Paris, France.
| | | | | | - Chloé Di Meglio
- Aix-Marseille Univ, APHM, Service de Neurologie Pédiatrique, Hopital de la Timone-Enfants. Marseille, France.
| | - Claude Cances
- Service de neurologie pédiatrique, CHU de Toulouse, France.
| | | | - Mathieu Milh
- Aix-Marseille Univ, APHM, Service de Neurologie Pédiatrique, Hopital de la Timone-Enfants. Marseille, France.
| | - Stéphane Auvin
- APHP. Service de Neurologie Pédiatrique, EpiCARE ERN Membre, Hôpital Robert Debré, Paris, France; Université Paris-Cité, INSERM NeuroDiderot, Paris, France; Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
21
|
Sands TT, Gelinas JN. Epilepsy and Encephalopathy. Pediatr Neurol 2024; 150:24-31. [PMID: 37948790 DOI: 10.1016/j.pediatrneurol.2023.09.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 09/14/2023] [Accepted: 09/24/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND Epilepsy encompasses more than the predisposition to unprovoked seizures. In children, epileptic activity during (ictal) and between (interictal) seizures has the potential to disrupt normal brain development. The term "epileptic encephalopathy (EE)" refers to the concept that such abnormal activity may contribute to cognitive and behavioral impairments beyond that expected from the underlying cause of the epileptic activity. METHODS In this review, we survey the concept of EE across a diverse selection of syndromes to illustrate its broad applicability in pediatric epilepsy. We review experimental evidence that provides mechanistic insights into how epileptic activity has the potential to impact normal brain processes and the development of neural networks. We then discuss opportunities to improve developmental outcomes in epilepsy now and in the future. RESULTS Epileptic activity in the brain poses a threat to normal physiology and brain development. CONCLUSION Until we have treatments that reliably target and effectively treat the underlying causes of epilepsy, a major goal of management is to prevent epileptic activity from worsening developmental outcomes.
Collapse
Affiliation(s)
- Tristan T Sands
- Center for Translational Research in Neurodevelopmental Disease, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Departments of Neurology and Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York.
| | - Jennifer N Gelinas
- Center for Translational Research in Neurodevelopmental Disease, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York; Departments of Neurology and Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York
| |
Collapse
|
22
|
Jones KEA, Howells R, Mallick AA, Paul SP, Dey I. NICE guideline review: Epilepsies in children, young people and adults NG217. Arch Dis Child Educ Pract Ed 2023; 108:416-421. [PMID: 37339862 DOI: 10.1136/archdischild-2022-324427] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 05/15/2023] [Indexed: 06/22/2023]
Affiliation(s)
| | - Rachel Howells
- Department of Paediatrics, Royal Devon University Healthcare NHS Foundation Trust, Exeter, UK
| | - Andrew A Mallick
- Department of Paediatric Neurology, Bristol Royal Hospital for Children, Bristol, UK
| | | | - Indranil Dey
- Department of Paediatrics, Torbay and South Devon NHS Foundation Trust, Torquay, UK
| |
Collapse
|
23
|
Tierradentro-García LO, Zandifar A, Stern J, Nel JH, Ub Kim JD, Andronikou S. Magnetic Resonance Imaging-Based Distribution and Reversibility of Lesions in Pediatric Vigabatrin-Related Brain Toxicity. Pediatr Neurol 2023; 148:86-93. [PMID: 37690269 DOI: 10.1016/j.pediatrneurol.2023.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND We aimed to systematically characterize the magnetic resonance imaging (MRI) findings in vigabatrin-related neurotoxicity in children and determine the reversibility of lesions based on follow-up images. METHODS We evaluated children with a history of refractory seizures who had a brain MRI while on vigabatrin therapy. We included available brain MRI studies before vigabatrin therapy initiation, during vigabatrin treatment, and after vigabatrin was discontinued. A pediatric neuroradiologist systematically assessed images on T2/fluid-attenuated inversion recovery (FLAIR) and diffusion-weighted imaging /apparent diffusion coefficient sequences to identify hyperintense lesions and/or restricted diffusion. The frequency of abnormal signal at each location was determined, as well as the reversibility of these after vigabatrin discontinuation. RESULTS MRIs of 43 patients were reviewed: 13 before vigabatrin initiation, 18 during treatment, and 12 after vigabatrin discontinuation. In the MRIs acquired during vigabatrin treatment, most lesions on T2/FLAIR occurred in the globus pallidi, thalami, and midbrain. Correspondingly, the most common locations for restricted diffusion were the globus pallidi, thalami, and subthalamic nuclei. On MRI after vigabatrin discontinuation, complete resolution of lesions on T2/FLAIR in all patients was seen in the midbrain, dentate nuclei, subthalamic nuclei, and hypothalami. Complete resolution of restricted diffusion was observed in the globus pallidi, midbrain, dentate nuclei, hippocampi, anterior commissure, and hypothalami. CONCLUSION Globus pallidi and thalami are the most commonly affected structures in vigabatrin-related toxicity, and most vigabatrin-related neuroimaging findings are reversible.
Collapse
Affiliation(s)
- Luis Octavio Tierradentro-García
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.
| | - Alireza Zandifar
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Joseph Stern
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jean Henri Nel
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Jorge Du Ub Kim
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Savvas Andronikou
- Department of Radiology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania; Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
24
|
Gatson TN, Freemont OA, Coleman RL, Lockett SJ, Spillers NJ, Ahmadzadeh S, Viswanath O, Varrassi G, Shekoohi S, Kaye AD. Vigabatrin (Sabril) for the Treatment of Refractory Complex Focal Seizures in Adults: Pharmacologic and Clinical Considerations. Cureus 2023; 15:e46414. [PMID: 37927747 PMCID: PMC10621625 DOI: 10.7759/cureus.46414] [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: 08/08/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
To review the pharmacokinetics, efficacy, and adverse effects of vigabatrin (Sabril) and its role in managing refractory focal unaware seizures in adults. In the present investigation, a search of English-language literature from 1999 through 2023 was conducted using vigabatrin and Sabril as search terms to identify relevant studies and review articles. A 2000 double-blind, placebo-controlled multicenter study found that out of 90 adult patients, 48% of those treated with vigabatrin achieved a 50% or greater reduction in the frequency of complex partial seizures, compared to 26% of placebo-treated patients. This study also observed that vigabatrin was well tolerated by 72.4% of patients, with the most common side effects being drowsiness, dizziness, headache, and fatigue. Further studies with a significant risk of vigabatrin-associated visual field loss necessitate vigabatrin only being an option for refractory cases. Additional studies suggest that despite the potential risk of vision loss and adverse effects, adult patients continue to use vigabatrin long-term. Sabril is an antiepileptic medication prescribed as an additional treatment for refractory complex partial seizures in patients at least ten years old who have not responded well to other alternative therapies. Multiple clinical trials indicate that Sabril sufficiently reduces the frequency of seizures when used as an adjunct treatment of refractory complex focal seizures. However, it is important to carefully monitor patients for any adverse effects, particularly long-term use, and to discontinue the drug if serious side effects occur.
Collapse
Affiliation(s)
- Taylor N Gatson
- Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Orlandria A Freemont
- Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Rachel L Coleman
- Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Stewart J Lockett
- Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Noah J Spillers
- Medicine, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Shahab Ahmadzadeh
- Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Omar Viswanath
- Pain Management, Valley Pain Consultants - Envision Physician Services, Phoenix, USA
| | | | - Sahar Shekoohi
- Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| | - Alan D Kaye
- Anesthesiology, Louisiana State University Health Sciences Center at Shreveport, Shreveport, USA
| |
Collapse
|
25
|
Chen H, Chen Y, Wu H, Qiu X, Yu X, Wang R, Zhong J, Peng J. De novo variants in PHF21A cause intellectual developmental disorder with behavioral abnormalities and craniofacial dysmorphism with or without seizures: A case report and literature review. Seizure 2023; 111:138-146. [PMID: 37633153 DOI: 10.1016/j.seizure.2023.08.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 08/07/2023] [Accepted: 08/17/2023] [Indexed: 08/28/2023] Open
Abstract
PURPOSE PHF21A has been associated with intellectual developmental disorder with behavioral abnormalities and craniofacial dysmorphism with or without seizures (IDDBCS). Here, we report a new patient with IDDBCS and review previously reported patients. METHODS We reviewed the phenotypic and genetic spectrum of the newly diagnosed patient and previously reported patients with IDDBCS. RESULTS Among 12 patients (11 whose cases were previously reported and the patient whose case we report here), all patients (100%) had intellectual disability (ID) and motor development delay. Three of 8 patients (37.5%) for whom information on cognition was available had severe ID; ID was moderate in two patients (25%) and mild in three patients (37.5%). Seven of the 12 patients (58.33%) had an epileptic phenotype, and the majority (5/7, 71.42%) of affected individuals developed developmental and epileptic encephalopathy (DEE). Of the 5 patients with DEE, three developed infantile epileptic spasm syndrome (IESS). The seizures of 2 patients (2/5, 40%) were controlled by antiseizure medications. Overgrowth, ADHD, hypotonia, ASD, and sleep disorders were observed in 100%, 77.78%, 70%, 50%, and 33.33% of patients, respectively. All of the variants (100%) were de novo heterozygous variants. Three of the 12 patients (25%) had the same variant (p.Arg580*). The most common types of variants were frameshift variants (7/12, 58.33%), followed by nonsense variants (4/12, 33.33%) and missense variants (1/12, 8.33%). Genotype-phenotype relationships for IDDBCS were uncertain, as phenotypic variability was observed among patients with the same variant (p.Arg580*). The patient whose case we report here had a novel PHF21A gene variant (p.Gln97fs*20), which caused neurodevelopmental delay, macrocephaly, and IESS. CONCLUSION The core phenotypes of IDDBCS include neurodevelopmental delay (intellectual disability and impaired motor skills), craniofacial abnormalities, and overgrowth. ADHD, hypotonia, epilepsy, ASD, and sleep disorders are common symptoms of IDDBCS. Notably, DEE is the dominant phenotype of epilepsy, especially IESS. PHF21A may be a candidate gene for DEE. De novo variants are the main mode of inheritance. The most common types of variants are frameshift variants, and the variant p.Arg580* in PHF21A is located at a mutation hot spot.
Collapse
Affiliation(s)
- Hui Chen
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, China
| | - Yong Chen
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, China
| | - Huaping Wu
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, China
| | - Xiaolu Qiu
- Child healthcare department, Children's Hospital of Jiangxi Province, Nanchang, China
| | - Xiongying Yu
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, China
| | - Ruiyan Wang
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, China.
| | - Jianmin Zhong
- Department of Neurology, Children's Hospital of Jiangxi Province, Nanchang, China.
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
26
|
Prezioso G, Chiarelli F, Matricardi S. Efficacy and safety of vigabatrin in patients with tuberous sclerosis complex and infantile epileptic spasm syndrome: a systematic review. Expert Rev Neurother 2023; 23:661-671. [PMID: 37243682 DOI: 10.1080/14737175.2023.2216385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/17/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Tuberous sclerosis complex (TSC) is a common genetic cause of epilepsy. Infantile epileptic spasm syndrome (IESS) is often the presenting neurologic feature, progressively evolving into refractory epilepsy. Vigabatrin (VGB) is often used in clinical practice as a first-line therapy in TSC with IESS. This systematic review aims to collect and analyze the efficacy data about VGB in TSC cases with IESS, in order to evaluate the strength of evidence in the literature. METHODS A systematic search of trials, observational studies, and case series involving patients with TSC and IESS treated with VGB was performed using MEDLINE, CENTRAL, and the US NIH Clinical Trials Registry. Single case studies, animal and non-English language studies were excluded. Seventeen studies were selected, of which 3 were RCTs and 14 were observational studies. RESULTS An overall response rate of 67% (231/343 responders) resulted from the analysis, with a spasm-free rate restricted to RCTs of 88% (29/33 subjects). CONCLUSIONS Although all the studies analyzed reported beneficial effects of VGB in TSC patients with IESS, with higher response rates in comparison to non-TSC subjects with IESS, a low level of evidence and high heterogeneity do not guarantee sufficient strength for therapeutic recommendations.
Collapse
Affiliation(s)
- Giovanni Prezioso
- Pediatric Neurologist, Department of Pediatrics, G. D'Annunzio University, Chieti, Italy
| | - Francesco Chiarelli
- Department of Pediatrics, Department of Pediatrics, G. D'Annunzio University, Chieti, Italy
| | - Sara Matricardi
- Pediatric Neurologist, Department of Pediatrics, G. D'Annunzio University, Chieti, Italy
| |
Collapse
|
27
|
Pisani F, Spagnoli C. What are the considerations when initiating treatment for epilepsy in children? Expert Rev Neurother 2023; 23:1081-1096. [PMID: 38032395 DOI: 10.1080/14737175.2023.2288107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/22/2023] [Indexed: 12/01/2023]
Abstract
INTRODUCTION There is a very wide spectrum of epilepsies and developmental and epileptic encephalopathies that affect children, from self-limited forms, not necessarily requiring treatment, to severe drug-resistant ones. AREAS COVERED In this perspective, the authors discuss the main factors to consider before drug prescription in children, considering the most recent clinical research, including age, seizure type, epilepsy syndrome, etiology, efficacy and safety profile, comorbidities, gender, available formulations, costs and drug coverage, and regulatory issues. The literature search was conducted through a PubMed search on antiseizure medications for patients aged 0-18, with respect to each of the aforementioned factors, and by checking the reference lists of relevant papers. EXPERT OPINION The most expanding field of research and innovation for clinical practice is precision medicine, which addresses the holistic treatment of genetic epilepsies and developmental and epileptic encephalopathies. It achieves this by addressing their detrimental effects on synapses, neurotransmission, and cellular signaling pathways with the double aim to treat seizures and to rescue neurodevelopmental trajectories, but also the issue of adverse events and drug resistance through pharmacogenomics.
Collapse
Affiliation(s)
- Francesco Pisani
- Human Neurosciences Department, Sapienza University of Rome, Rome, Italy
| | - Carlotta Spagnoli
- Child Neurology and Psychiatry Unit, Department of Pediatrics, Presidio Ospedaliero Santa Maria Nuova, AUSL-IRCCS di Reggio Emilia, Reggio Emilia, Italy
| |
Collapse
|
28
|
Olson HE, Demarest S, Pestana-Knight E, Moosa AN, Zhang X, Pérez-Pérez JR, Weisenberg J, O'Connor Prange E, Marsh ED, Rajaraman RR, Suter B, Katyayan A, Haviland I, Daniels C, Zhang B, Greene C, DeLeo M, Swanson L, Love-Nichols J, Benke T, Harini C, Poduri A. Epileptic spasms in CDKL5 deficiency disorder: Delayed treatment and poor response to first-line therapies. Epilepsia 2023; 64:1821-1832. [PMID: 37114835 PMCID: PMC10524264 DOI: 10.1111/epi.17630] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 04/25/2023] [Accepted: 04/25/2023] [Indexed: 04/29/2023]
Abstract
OBJECTIVE We aimed to assess the treatment response of infantile-onset epileptic spasms (ES) in CDKL5 deficiency disorder (CDD) vs other etiologies. METHODS We evaluated patients with ES from the CDKL5 Centers of Excellence and the National Infantile Spasms Consortium (NISC), with onset from 2 months to 2 years, treated with adrenocorticotropic hormone (ACTH), oral corticosteroids, vigabatrin, and/or the ketogenic diet. We excluded children with tuberous sclerosis complex, trisomy 21, or unknown etiology with normal development because of known differential treatment responses. We compared the two cohorts for time to treatment and ES remission at 14 days and 3 months. RESULTS We evaluated 59 individuals with CDD (79% female, median ES onset 6 months) and 232 individuals from the NISC database (46% female, median onset 7 months). In the CDD cohort, seizures prior to ES were common (88%), and hypsarrhythmia and its variants were present at ES onset in 34%. Initial treatment with ACTH, oral corticosteroids, or vigabatrin started within 1 month of ES onset in 27 of 59 (46%) of the CDD cohort and 182 of 232 (78%) of the NISC cohort (p < .0001). Fourteen-day clinical remission of ES was lower for the CDD group (26%, 7/27) than for the NISC cohort (58%, 106/182, p = .0002). Sustained ES remission at 3 months occurred in 1 of 27 (4%) of CDD patients vs 96 of 182 (53%) of the NISC cohort (p < .0001). Comparable results were observed with longer lead time (≥1 month) or prior treatment. Ketogenic diet, used within 3 months of ES onset, resulted in ES remission at 1 month, sustained at 3 months, in at least 2 of 13 (15%) individuals with CDD. SIGNIFICANCE Compared to the broad group of infants with ES, children with ES in the setting of CDD often experience longer lead time to treatment and respond poorly to standard treatments. Development of alternative treatments for ES in CDD is needed.
Collapse
Affiliation(s)
- Heather E Olson
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Scott Demarest
- Department of Pediatrics, School of Medicine, Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Elia Pestana-Knight
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ahsan N Moosa
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Xiaoming Zhang
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - José R Pérez-Pérez
- Epilepsy Center, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Judy Weisenberg
- Department of Pediatric Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Erin O'Connor Prange
- Division of Child Neurology, Children's Hospital of Philadelphia, Departments of Neurology and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Eric D Marsh
- Division of Child Neurology, Children's Hospital of Philadelphia, Departments of Neurology and Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rajsekar R Rajaraman
- Division of Pediatric Neurology, David Geffen School of Medicine and UCLA Mattel Children's Hospital, Los Angeles, California, USA
| | - Bernhard Suter
- Department of Pediatrics and Neurology, Baylor College of Medicine, Texas Children's Hospital, Houston, Houston, Texas, USA
| | - Akshat Katyayan
- Department of Pediatrics and Neurology, Baylor College of Medicine, Texas Children's Hospital, Houston, Houston, Texas, USA
| | - Isabel Haviland
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Carolyn Daniels
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Bo Zhang
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Caitlin Greene
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Michelle DeLeo
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Lindsay Swanson
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Jamie Love-Nichols
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Timothy Benke
- Department of Pediatrics, School of Medicine, Children's Hospital Colorado, University of Colorado, Aurora, Colorado, USA
| | - Chellamani Harini
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Annapurna Poduri
- Division of Epilepsy and Clinical Neurophysiology and Epilepsy Genetics Program, Department of Neurology, Boston Children's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
29
|
Rao CK, Nordli DR, Cousin JJ, Takacs DS, Sheth RD. The Effect of Smartphone Video on Lead Time to Diagnosis of Infantile Spasms. J Pediatr 2023; 258:113387. [PMID: 36931494 DOI: 10.1016/j.jpeds.2023.02.035] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 02/18/2023] [Accepted: 02/23/2023] [Indexed: 03/19/2023]
Abstract
OBJECTIVE To assess whether access to smartphone video capture of infantile spasms at initial presentation is associated with improved time to diagnosis and treatment. METHODS We conducted a collaborative retrospective cohort study of 80 consecutive infants with confirmed infantile epileptic spasms syndrome initially presenting from 2015 to 2021 at 2 US pediatric centers. Statistical methods used included Mann-Whitney U test to assess the difference in lead times to electroencephalogram (EEG), diagnosis, and treatment between groups with and without video capture. A χ2 analysis was used to assess differences in demographics, clinical characteristics, and treatment outcomes between groups. Multivariate regression analysis was used to account for etiology types and infantile spasms capture on EEG. RESULTS Patients with smartphone video infantile spasms capture initially presented a median of 9 days earlier (P = .02), had their first EEG 16 days earlier (P = .007), and were diagnosed and started treatment 17 days earlier (P = .006 and P = .008, respectively) compared with the nonvideo group. The video group had a 25% greater response to initial standard treatment (P = .02) and a 21% greater freedom from infantile spasms at long-term follow-up (P = .03), although this long-term outcome lost statistical significance after adjustment for etiology type (P = .07) and EEG capture of infantile spasms (P = .059). CONCLUSION Our findings suggest a benefit of smartphone video capture of infantile spasms in reduced time to diagnosis and initial standard treatment, which are associated with improved treatment response rates. Substantial differences in lead times and treatment response highlight the clinical importance of pediatricians recommending caregivers to obtain smartphone video of events concerning for infantile spasms.
Collapse
Affiliation(s)
- Chethan K Rao
- Division of Child and Adolescent Neurology, Mayo Clinic College of Medical Science Florida, Jacksonville, FL; Division of Neurology, Nemours Children's Health, Jacksonville, FL; Division of Child Neurology, Stanford University School of Medicine, Palo Alto, CA
| | - Douglas R Nordli
- Division of Child and Adolescent Neurology, Mayo Clinic College of Medical Science Florida, Jacksonville, FL; Division of Neurology, Nemours Children's Health, Jacksonville, FL
| | - Joshua J Cousin
- Neurology and Developmental Neuroscience, Texas Children's Hospital, Houston, TX; Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX
| | - Danielle S Takacs
- Neurology and Developmental Neuroscience, Texas Children's Hospital, Houston, TX; Department of Pediatrics and Neurology, Baylor College of Medicine, Houston, TX
| | - Raj D Sheth
- Division of Child and Adolescent Neurology, Mayo Clinic College of Medical Science Florida, Jacksonville, FL; Division of Neurology, Nemours Children's Health, Jacksonville, FL.
| |
Collapse
|
30
|
Thalwitzer KM, Xian J, deCampo D, Parthasarathy S, Magielski J, Sullivan KR, Goss J, Rigby CS, Boland M, Prosser B, Ruggiero SM, Syrbe S, Helbig I. Early life seizures and epileptic spasms in STXBP1-related disorders. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.26.23291892. [PMID: 37425705 PMCID: PMC10327252 DOI: 10.1101/2023.06.26.23291892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Background and Objectives Individuals with disease-causing variants in STXBP1 frequently have epilepsy onset in the first year of life with a variety of seizure types, including epileptic spasms. However, the impact of early-onset seizures and anti-seizure medication (ASM) on the risk of developing epileptic spasms and impact on their trajectory is poorly understood, limiting informed and anticipatory treatment, as well as trial design. Methods We retrospectively reconstructed seizure and medication histories in weekly intervals for individuals with STXBP1-related disorders with epilepsy onset in the first year of life and quantitatively analyzed longitudinal seizure histories and medication response. Results We included 61 individuals with early onset seizures, 29 of whom had epileptic spasms. Individuals with neonatal seizures were likely to have continued seizures after the neonatal period (25/26). The risk of developing epileptic spasms was not increased in individuals with neonatal seizures or early infantile seizures (21/41 vs. 8/16; OR 1, 95% CI 0.3-3.9, p = 1). We did not find any ASM associated with the development of epileptic spasms following prior seizures. Individuals with prior seizures (n = 16/21, 76%) had a higher risk to develop refractory epileptic spasms (n = 5/8, 63%, OR =1.9, 95% CI 0.2-14.6, p = 0.6). Individuals with refractory epileptic spasms had a later onset of epileptic spasms (n = 20, median 20 weeks) compared to individuals with non-refractory epileptic spasms (n = 8, median 13 weeks; p = 0.08). When assessing treatment response, we found that clonazepam (n = 3, OR 12.6, 95% CI 2.2-509.4; p < 0.01), clobazam (n=7, OR 3, 95% CI 1.6-6.2; p < 0.01), topiramate (n=9, OR 2.3, 95% CI 1.4-3.9; p < 0.01), and levetiracetam (n=16, OR 1.7, 95% CI 1.2-2.4; p < 0.01) were more likely to reduce seizure frequency and/or to maintain seizure freedom with regards to epileptic spasms than other medications. Discussion We provide a comprehensive assessment of early-onset seizures in STXBP1-related disorders and show that the risk of epileptic spasms is not increased following a prior history of early-life seizures, nor by certain ASM. Our study provides baseline information for targeted treatment and prognostication in early-life seizures in STXBP1-related disorders.
Collapse
Affiliation(s)
- Kim M Thalwitzer
- Division of Neurology, Children’s Hospital of Philadelphia, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, USA
- Division of Pediatric Epileptology, Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Germany
| | - Julie Xian
- Division of Neurology, Children’s Hospital of Philadelphia, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, USA
- Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, USA
| | - Danielle deCampo
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, USA
- Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, USA
- Division of Neurology, Children’s Hospital of Philadelphia, Philadelphia, USa
| | - Shridhar Parthasarathy
- Division of Neurology, Children’s Hospital of Philadelphia, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, USA
| | - Jan Magielski
- Division of Neurology, Children’s Hospital of Philadelphia, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, USA
- Neuroscience Program, University of Pennsylvania, Philadelphia, USA
| | - Katie Rose Sullivan
- Division of Neurology, Children’s Hospital of Philadelphia, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, USA
- Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, USA
| | | | | | - Michael Boland
- Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, USA
- Institute for Genomic Medicine, Columbia University, USA
| | - Ben Prosser
- Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, USA
- Department of Physiology, University of Pennsylvania Perelman School of Medicine, USA
| | - Sarah M Ruggiero
- Division of Neurology, Children’s Hospital of Philadelphia, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, USA
- Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, USA
| | - Steffen Syrbe
- Division of Pediatric Epileptology, Center for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Germany
| | - Ingo Helbig
- Division of Neurology, Children’s Hospital of Philadelphia, USA
- The Epilepsy NeuroGenetics Initiative (ENGIN), Children’s Hospital of Philadelphia, USA
- Department of Biomedical and Health Informatics (DBHi), Children’s Hospital of Philadelphia, USA
- Epilepsy and Neurodevelopmental Disorders Center (ENDD), Children’s Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, USA
- Department of Neurology, University of Pennsylvania, Perelman School of Medicine, USA
| |
Collapse
|
31
|
Takacs DS, Katyayan A, Vanderslice K, Riviello JJ. Inaccuracies in Parental Reporting of Treated Epileptic Spasms: Both Under- and Over-Reporting. Pediatr Neurol 2023; 145:119-123. [PMID: 37331271 DOI: 10.1016/j.pediatrneurol.2023.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 04/11/2023] [Accepted: 05/13/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND The purpose of this study was to evaluate the accuracy of parental reporting of epileptic spasms (ES) after 14 days of appropriate medical therapy for new-onset ES by comparison with extended video electroencephalography (vEEG) monitoring results. METHODS Fifty-eight patients were identified from August 2019 to February 2021 with new-onset ES, confirmed on vEEG. Patients were initiated on appropriate treatment (high-dose steroids or vigabatrin). After two weeks of therapy, patients underwent overnight (18 to 24 hours) vEEG monitoring in the epilepsy monitoring unit. Parental reporting of presence or absence of ES on admission was compared with results of vEEG monitoring. RESULTS The 58 patients ranged in age from three to 20 months (average 7.8 months). An underlying etiology was identified in 78%, whereas 22% patients had unknown etiology. The overall accuracy of parental reporting was 74% (43 of 58) when compared with results of vEEG within 14 to 18 days of starting therapy. Of these, 65% (28 of 43) reported ES resolution and 35% (15 of 43) reported continued ES. Of the 26% (15 of 58) families who were incorrect at two-week follow-up, 67% (10 of 15) reported resolution of ES. However, a minority of families, 33% (five of 15), who continued to report spasms clinically, were inaccurate. CONCLUSIONS Although a majority of inaccurate parental reports at two weeks of treatment were due to unrecognized ES (a widely known phenomenon), a minority were conversely inaccurate due to persistent over-reporting of ES. This fact highlights the importance of correlating parental history with objective vEEG monitoring, to prevent inappropriate escalation of medication therapy.
Collapse
Affiliation(s)
- Danielle Schwartzenburg Takacs
- Department of Pediatric Neurology and Developmental Neuroscience, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas.
| | - Akshat Katyayan
- Department of Pediatric Neurology and Developmental Neuroscience, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - Kari Vanderslice
- Department of Pediatric Neurology and Developmental Neuroscience, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| | - James John Riviello
- Department of Pediatric Neurology and Developmental Neuroscience, Texas Children's Hospital, Baylor College of Medicine, Houston, Texas
| |
Collapse
|
32
|
Raga SV, Essajee F, Solomons R, Van Toorn R, Wilmshurst JM. Epileptic spasms: A South African overview of aetiologies, interventions, and outcomes. Dev Med Child Neurol 2023; 65:526-533. [PMID: 36229895 DOI: 10.1111/dmcn.15433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 09/14/2022] [Accepted: 09/17/2022] [Indexed: 11/28/2022]
Abstract
AIM To better understand the aetiologies of epileptic spasms in infants, as well as the safety and efficacy of high dose corticosteroids in tuberculosis and human immunodeficiency virus (HIV) endemic resource-limited settings. METHOD This was a retrospective analysis of infants with epileptic spasms managed at the tertiary referral centres in the Western Cape, South Africa. RESULTS Of 175 children with epileptic spasms, the median age at onset was 6 months (interquartile range 4-8 months). Structural aetiologies were most common (115 out of 175 [66%]), with two-thirds related to perinatal insults. A lead time to treatment (LTTT) of less than 1 month was more likely in the epileptic encephalopathy/developmental and epileptic encephalopathy (DEE) group: 58 out of 92 (63%), compared to 28 out of 76 (37%) of those with developmental encephalopathy (p = 0.001). Failure to recognize preceding developmental delay was common. Ninety-nine children (57%) received first line hormonal therapy such as adrenocorticotropic hormone. A total of 111 out of 172 children (65%) from the developmental encephalopathy and epileptic encephalopathy/DEE groups had clinical and/or electroencephalogram resolution of spasms within 14 days. In our population, children in whom an aetiology could not be identified were statistically more likely to have moderate to profound developmental delay at 1 year of age: 33 out of 44 (p = 0.001). Based on reported incidence of epileptic spasms, 23 to 58 cases per annum would be expected but a far smaller proportion presented to our centres. INTERPRETATION Whilst this is the largest cohort of infants with epileptic spasms from sub-Saharan Africa, the study size is less than expected; this may reflect misdiagnosis and failure of referral pathways. Despite a reported shorter LTTT, infants with DEE had worse developmental outcomes compared to international studies. Hormonal therapy was safe and effective in our setting, despite exposure to high levels of tuberculosis and HIV. WHAT THIS PAPER ADDS The number of unreferred cases of epileptic spasms in South Africa remains high. Caregivers and health care workers in primary care facilities often fail to recognize developmental delay. The burden of disease from hypoxic-ischaemic encephalopathy remains high in our resource-limited setting. Hormonal treatment (e.g. adrenocorticotropic hormone) was safe and effective despite the high prevalence of human immunodeficiency virus and tuberculosis.
Collapse
Affiliation(s)
- Sharika V Raga
- Paediatric Neurology Division, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| | - Farida Essajee
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Regan Solomons
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Ronald Van Toorn
- Department of Paediatrics and Child Health, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Jo M Wilmshurst
- Paediatric Neurology Division, Department of Paediatrics and Child Health, Red Cross War Memorial Children's Hospital, Neuroscience Institute, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
33
|
Becker LL, Kaindl AM. Corticosteroids in childhood epilepsies: A systematic review. Front Neurol 2023; 14:1142253. [PMID: 36970534 PMCID: PMC10036579 DOI: 10.3389/fneur.2023.1142253] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 02/15/2023] [Indexed: 03/12/2023] Open
Abstract
Corticosteroids have been used for the treatment of patients with epilepsy for more than 6 decades, based on the hypothesis of inflammation in the genesis and/or promotion of epilepsy. We, therefore, aimed to provide a systematic overview of the use of corticosteroid regimes in childhood epilepsies in line with the PRISMA guidelines. We performed a structured literature search via PubMed and identified 160 papers with only three randomized controlled trials excluding the substantial trials on epileptic spasms. Corticosteroid regimes, duration of treatment (days to several months), and dosage protocols were highly variable in these studies. Evidence supports the use of steroids in epileptic spasms; however, there is only limited evidence for a positive effect for other epilepsy syndromes, e.g., epileptic encephalopathy with spike-and-wave activity in sleep [(D)EE-SWAS] or drug-resistant epilepsies (DREs). In (D)EE-SWAS (nine studies, 126 patients), 64% of patients showed an improvement either in the EEG or in their language/cognition following various steroid treatment regimes. In DRE (15 studies, 436 patients), a positive effect with a seizure reduction in 50% of pediatric and adult patients and seizure freedom in 15% was identified; however, no recommendation can be drawn due to the heterozygous cohort. This review highlights the immense need for controlled studies using steroids, especially in DRE, to offer patients new treatment options.
Collapse
Affiliation(s)
- Lena-Luise Becker
- Department of Pediatric Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Center for Chronically Sick Children, Charité – Universitätsmedizin Berlin, Berlin, Germany
- German Epilepsy Center for Children and Adolescents, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Institute of Cell- and Neurobiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| | - Angela M. Kaindl
- Department of Pediatric Neurology, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Center for Chronically Sick Children, Charité – Universitätsmedizin Berlin, Berlin, Germany
- German Epilepsy Center for Children and Adolescents, Charité – Universitätsmedizin Berlin, Berlin, Germany
- Institute of Cell- and Neurobiology, Charité – Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
34
|
Fan HC, Chiang KL, Chang KH, Chen CM, Tsai JD. Epilepsy and Attention Deficit Hyperactivity Disorder: Connection, Chance, and Challenges. Int J Mol Sci 2023; 24:ijms24065270. [PMID: 36982345 PMCID: PMC10049646 DOI: 10.3390/ijms24065270] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/20/2023] [Accepted: 03/01/2023] [Indexed: 03/12/2023] Open
Abstract
Comorbidities are common in children with epilepsy, with nearly half of the patients having at least one comorbidity. Attention deficit hyperactivity disorder (ADHD) is a psychiatric disorder characterized by hyperactivity and inattentiveness level disproportional to the child’s developmental stage. The burden of ADHD in children with epilepsy is high and can adversely affect the patients’ clinical outcomes, psychosocial aspects, and quality of life. Several hypotheses were proposed to explain the high burden of ADHD in childhood epilepsy; the well-established bidirectional connection and shared genetic/non-genetic factors between epilepsy and comorbid ADHD largely rule out the possibility of a chance in this association. Stimulants are effective in children with comorbid ADHD, and the current body of evidence supports their safety within the approved dose. Nonetheless, safety data should be further studied in randomized, double-blinded, placebo-controlled trials. Comorbid ADHD is still under-recognized in clinical practice. Early identification and management of comorbid ADHD are crucial to optimize the prognosis and reduce the risk of adverse long-term neurodevelopmental outcomes. The identification of the shared genetic background of epilepsy and ADHD can open the gate for tailoring treatment options for these patients through precision medicine.
Collapse
Affiliation(s)
- Hueng-Chuen Fan
- Department of Pediatrics, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435, Taiwan
- Department of Rehabilitation, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli 356, Taiwan
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
| | - Kuo-Liang Chiang
- Department of Pediatric Neurology, Kuang-Tien General Hospital, Taichung 433, Taiwan
- Department of Nutrition, Hungkuang University, Taichung 433, Taiwan
| | - Kuang-Hsi Chang
- Department of Medical Research, Tungs’ Taichung Metroharbor Hospital, Wuchi, Taichung 435, Taiwan
| | - Chuan-Mu Chen
- Department of Life Sciences, Agricultural Biotechnology Center, National Chung Hsing University, Taichung 402, Taiwan
- The iEGG and Animal Biotechnology Center, and Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 402, Taiwan
- Correspondence: (C.-M.C.); (J.-D.T.); Tel.: +886-4-22840319-701 (C.-M.C.); +886-4-24730022-21731 (J.-D.T.)
| | - Jeng-Dau Tsai
- School of Medicine, Chung Shan Medical University, Taichung 402, Taiwan
- Department of Pediatrics, Chung Shan Medical University Hospital, Taichung 402, Taiwan
- Correspondence: (C.-M.C.); (J.-D.T.); Tel.: +886-4-22840319-701 (C.-M.C.); +886-4-24730022-21731 (J.-D.T.)
| |
Collapse
|
35
|
Sánchez Fernández I, Amengual-Gual M, Barcia Aguilar C, Romeu A, Sheikh T, Torres A, Chao J, Jonas R, Gaínza-Lein M, Harini C, Douglass L. Temporal trends in the cost and use of first-line treatments for infantile epileptic spasms syndrome. Epilepsia 2023; 64:630-640. [PMID: 36600453 DOI: 10.1111/epi.17498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 12/19/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To describe the temporal trends in the cost and use of adrenocorticotropic hormone (ACTH), oral prednisolone, and vigabatrin, the first-line treatments for infantile epileptic spasms syndrome (IESS). METHODS Retrospective observational study using the MarketScan Commercial database from 2006 to 2020. We identified patients with IESS diagnosed between birth and 18 months of age who received at least one of the first-line treatments within 60 days of diagnosis. Costs were adjusted for inflation using the Gross Domestic Product Implicit Price Deflator. RESULTS A total of 1131 patients received at least one first-line treatment (median [p25 -p75 ] age: 6.3 [4.5-8.3] months, 55% male), of whom 592 patients received ACTH, 363 patients received oral prednisolone, and 355 patients received vigabatrin. After adjusting for inflation, the median average wholesale price of a 14-day course of treatment increased for ACTH from $3718 in 2006 to $100 457 in 2020, ~2700% (by a factor of 27), whereas it decreased for oral prednisolone from $169 in 2006 to $89 in 2020, ~50% (by a factor of 0.5), and increased for vigabatrin from $1206 in 2009 (first year with data on vigabatrin used for IESS) to $4102 in 2020, ~340% (by a factor of 3.4). During the first 60 days after diagnosis, inpatient admission days and costs where higher for ACTH than for oral prednisolone and vigabatrin-5.0 (3.0-8.3) days vs 2.0 (0.0-5.0) days vs 2.0 (0.0-6.0) days, p < .0001; and $32 828 ($14 711-$67 216) vs $16 227 ($0-$35 829) vs $17 844 ($0-$47 642), p < .0001. ACTH use decreased from representing 78% of first-line treatments in 2006 to 18% in 2020 (p < .0001). Sensitivity analyses confirmed the robustness of the results. SIGNIFICANCE The gap between the cost of ACTH and the cost of oral prednisolone or vigabatrin has widened markedly from 2006 to 2020, whereas the relative proportion of ACTH use has decreased.
Collapse
Affiliation(s)
- Iván Sánchez Fernández
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Marta Amengual-Gual
- Pediatric Neurology Unit, Department of Pediatrics, Hospital Universitari Son Llàtzer, Universitat de les Illes Balears, Palma, Spain
| | - Cristina Barcia Aguilar
- Pediatric Neurology Unit, Department of Pediatrics, Complejo Hospitalario Universitario de La Coruña, La Coruña, Spain
| | - Amanda Romeu
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Tahir Sheikh
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Alcy Torres
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jessica Chao
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Rinat Jonas
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Marina Gaínza-Lein
- Instituto de Pediatría, Facultad de Medicina, Universidad Austral de Chile, Valdivia, Chile
- Servicio de Neuropsiquiatría Infantil, Hospital Clínico San Borja Arriarán, Universidad de Chile, Santiago, Chile
| | - Chellamani Harini
- Division of Epilepsy and Clinical Neurophysiology, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Laurie Douglass
- Division of Pediatric Neurology, Department of Pediatrics, Boston Medical Center, Boston University School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
36
|
Meng Y, Geng G, Ren Y, Zhang H, Gao Z, Liu Y, Shi J. Long-Term Outcome of Adrenocorticotropic Hormone Therapy in Children With New-Onset Infantile Spasms. Pediatr Neurol 2023; 143:100-105. [PMID: 37060643 DOI: 10.1016/j.pediatrneurol.2023.02.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Revised: 08/30/2022] [Accepted: 02/18/2023] [Indexed: 04/17/2023]
Abstract
BACKGROUND To investigate for pretreatment clinical variables to predict the outcome of new-onset epileptic spasms after adrenocorticotropic hormone (ACTH) therapy and to identify risk factors for poor long-term outcome. METHODS We retrospectively studied 129 consecutive patients with infantile spasms syndrome (ISS). These patients received ACTH with antiseizure medication therapy for the first time and were regularly followed up for more than six months at our hospital. The response to treatment was assessed after two weeks of ACTH injection. Kaplan-Meier survival analysis and the multivariate Cox proportional hazard regression model were used. RESULTS Among the 129 patients, 61 (47.3%) had a good response after two weeks of ACTH treatment. At the time of the latest follow-up, 71 (55%) patients were seizure-free (International League Against Epilepsy class1). The univariate analysis revealed that normal neurodevelopment (P = 0.018), time lag of less than one month (P = 0.026), no hypsarrhythmia on EEG (P = 0.004), and serum calcium level ≥2.50 mmol/L (P = 0.035) were significantly associated with a good response. Only a good response to ACTH therapy was significantly associated with a positive long-term outcome. The Kaplan-Meier survival analysis showed that serum calcium level ≧2.50 mmol/L was significantly associated with a positive long-term outcome (P = 0.030). Multivariate analysis confirmed that no response to ACTH therapy was an independent variable that predicted long-term seizure recurrence (P < 0.001, hazard ratio = 4.602, confidence interval = 2.252 to 9.406). CONCLUSIONS A good response to ACTH therapy had a significant predictive value for long-term seizure outcomes. Calcium may play an important role in the treatment of ISS with ACTH.
Collapse
Affiliation(s)
- Yao Meng
- Department of Epilepsy Center, Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, ShanDong, China; Department of Functional Neurosurgery, Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, ShanDong, China
| | - Guifu Geng
- Department of Epilepsy Center, Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, ShanDong, China; Department of Functional Neurosurgery, Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, ShanDong, China
| | - Ying Ren
- Department of Epilepsy Center, Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, ShanDong, China
| | - Hongwei Zhang
- Department of Epilepsy Center, Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, ShanDong, China
| | - Zaifen Gao
- Department of Epilepsy Center, Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, ShanDong, China
| | - Yong Liu
- Department of Epilepsy Center, Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, ShanDong, China
| | - Jianguo Shi
- Department of Epilepsy Center, Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, ShanDong, China; Department of Functional Neurosurgery, Children's Hospital Affiliated to Shandong University, Jinan Children's Hospital, Jinan, ShanDong, China.
| |
Collapse
|
37
|
Gettings JV, Shafi S, Boyd J, Snead OC, Rutka J, Drake J, McCoy B, Jain P, Whitney R, Go C. The Epilepsy Surgery Experience in Children With Infantile Epileptic Spasms Syndrome at a Tertiary Care Center in Canada. J Child Neurol 2023; 38:113-120. [PMID: 36788207 DOI: 10.1177/08830738231151993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
BACKGROUND Infantile epileptic spasms syndrome is an epileptic encephalopathy, characterized by spasms, hypsarrhythmia, and developmental regression. Appropriately selected patients with infantile epileptic spasms syndrome may be candidates for epilepsy surgery. METHODS This is a single-center retrospective case series of children 0-18 years with a current or previous diagnosis of infantile epileptic spasms syndrome with a lesion on magnetic resonance imaging (MRI) and/or positron emission tomography scan who underwent epilepsy surgery at The Hospital for Sick Children (HSC) in Toronto, Canada. The records of 223 patients seen in the infantile epileptic spasms syndrome clinic were reviewed. RESULTS Nineteen patients met inclusion criteria. The etiology of infantile epileptic spasms syndrome was encephalomalacia in 6 patients (32%), malformations of cortical development in 12 patients (63%), and atypical hypoglycemic injury in 1 patient (5%). Nine patients (47%) underwent hemispherectomy, and 10 patients (53%) underwent lobectomy/lesionectomy. Three patients (16%) underwent a second epilepsy surgery. Fifteen patients (79%) were considered ILAE seizure outcome class 1 (completely seizure free; no auras) at their most recent follow-up visit. The percentage of patients who were ILAE class 1 at most recent follow-up decreased with increasing duration of epilepsy prior to surgery. Developmental outcome after surgery was improved in 14 of 19 (74%) and stable in 5 of 19 (26%) patients. CONCLUSIONS Our study found excellent seizure freedom rates and improved developmental outcomes following epilepsy surgery in patients with a history of infantile epileptic spasms syndrome with a structural lesion detected on MRI brain. Patients who undergo surgery earlier have improved seizure freedom rates and improved developmental outcomes.
Collapse
Affiliation(s)
- Jennifer V Gettings
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - Shatha Shafi
- Division of Neurology, Department of Pediatrics, 37853Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Jennifer Boyd
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - O Carter Snead
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - James Rutka
- Division of Neurosurgery, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - James Drake
- Division of Neurosurgery, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - Bláthnaid McCoy
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - Puneet Jain
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada
| | - Robyn Whitney
- Division of Neurology, Department of Paediatrics, McMaster Children's Hospital (McMaster University), Hamilton, ON, Canada
| | - Cristina Go
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children (University of Toronto), Toronto, Ontario, Canada.,Division of Neurology, Department of Pediatrics, British Columbia Children's Hospital (University of British Columbia), Toronto, Ontario, Canada
| |
Collapse
|
38
|
Liao J, Hu Z, Lin S, Lu X, Wen J, Duan J, Zou D, Zou H, Yu M, Liu L, Qiao X, Ye Y. Long-term outcomes of infantile spasms in children treated with ketogenic diet therapy in combination with anti-seizure medications in a resource-limited region. FRONTIERS IN EPIDEMIOLOGY 2023; 2:1080068. [PMID: 38455303 PMCID: PMC10910894 DOI: 10.3389/fepid.2022.1080068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 12/19/2022] [Indexed: 03/09/2024]
Abstract
Objective Despite numerous guidelines, the overall outcome of infantile spasms is poor, with only a small number of patients being able to attend school. The purpose of this study was to investigate long-term outcomes. Patients had poor access to the recommended first-line anti-seizure medications (ASMs), such as hormones (corticotropin or prednisolone/prednisone) and vigabatrin, and their alternative treatment was other ASMs and a ketogenic diet. Methods Patients suffering from infantile spasms who had at least 2 years of medical records in the electronic medical record system between January 2014 and August 2022 were included in this study. Patient information was retrospectively reviewed. All patients had received ketogenic diet therapy (mainly classical ketogenic diet therapy). The ketogenic diet therapy was combined with ASMs not used as first-line therapies. The primary endpoint outcome measure was the number of patients with seizure freedom. The secondary measures included the duration of ketogenic diet therapy, choice of ASMs, and patient development at the last visit. Results A total of 177 patients with infantile spasms were included, and 152 (86%) of them had seizure freedom. The median duration from the first to the last hospital visit was 53.27 months, and the number of visits was 47.00. The median age at the initial hospital visit was 8.00 months, and the median age at initiation of the ketogenic diet was 17.73 months. At the last visit, the proportions of patients with neurodevelopmental delay, developmental epileptic encephalopathy, drug-resistant epilepsy, and generalized seizures increased significantly. The frequently used ASMs were topiramate, valproic acid, levetiracetam, nitrazepam, and vitamin B6 injection, while the recommended first-line drugs corticotropin and vigabatrin were rarely selected. The study duration of 9.5 years was divided into three periods but the prescription of ASMs did not change significantly between these periods. Conclusions Although the seizure freedom rate was high with ketogenic diet therapy combined with non-standard ASMs, the patients had a significant neurodevelopmental delay at the last visit, which was, however, similar to that of standard treatment. To improve the outcomes of infantile spasms, multicenter clinical trials of the ketogenic diet as a first-line treatment in combination with non-standard ASMs are needed.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Yuanzhen Ye
- Shenzhen Children's Hospital, Shenzhen, China
| |
Collapse
|
39
|
黄 茜, 彭 镜, 潘 邹, 彭 盼, 何 芳, 张 慈, 陈 晨, 刘 方, 尹 飞, 毛 蕾. [Factors influencing the efficacy of initial adrenocorticotropic hormone therapy for infantile epileptic spasms syndrome]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:60-66. [PMID: 36655665 PMCID: PMC9893818 DOI: 10.7499/j.issn.1008-8830.2207096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 11/23/2022] [Indexed: 01/20/2023]
Abstract
OBJECTIVES To study the factors influencing the short-term (28 days) efficacy of initial adrenocorticotropic hormone (ACTH) therapy for infantile epileptic spasms syndrome (IESS), as well as the factors influencing recurrence and prognosis. METHODS The clinical data were collected from the children with IESS who received ACTH therapy for the first time in the Department of Pediatric Neurology, Xiangya Hospital of Central South University, from April 2008 to January 2018 and were followed up for ≥2 years. The multivariate logistic regression analysis was used to evaluate the factors influencing the short-term efficacy of ACTH therapy, recurrence, and long-term prognosis. RESULTS ACTH therapy achieved a control rate of seizures of 55.5% (111/200) on day 28 of treatment. Of the 111 children, 75 (67.6%) had no recurrence of seizures within 12 months of follow-up. The possibility of seizure control on day 28 of ACTH therapy in the children without focal seizures was 2.463 times that in those with focal seizures (P<0.05). The possibility of seizure control on day 28 of ACTH therapy in the children without hypsarrhythmia on electroencephalography on day 14 of ACTH therapy was 2.415 times that in those with hypsarrhythmia (P<0.05). The possibility of recurrence within 12 months after treatment was increased by 11.8% for every 1-month increase in the course of the disease (P<0.05). The possibility of moderate or severe developmental retardation or death in the children without seizure control after 28 days of ACTH therapy was 8.314 times that in those with seizure control (P<0.05). The possibility of moderate or severe developmental retardation or death in the children with structural etiology was 14.448 times that in those with unknown etiology (P<0.05). CONCLUSIONS Presence or absence of focal seizures and whether hypsarrhythmia disappears after 14 days of treatment can be used as predictors for the short-term efficacy of ACTH therapy, while the course of disease before treatment can be used as the predictor for recurrence after seizure control by ACTH therapy. The prognosis of IESS children is associated with etiology, and early control of seizures after ACTH therapy can improve long-term prognosis.
Collapse
|
40
|
GRIN2A-related epilepsy and speech disorders: A comprehensive overview with a focus on the role of precision therapeutics. Epilepsy Res 2023; 189:107065. [PMID: 36516565 DOI: 10.1016/j.eplepsyres.2022.107065] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/27/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
Language dysfunction is a common and serious comorbidity of epilepsy, especially in individuals with epilepsy aphasia spectrum syndromes. Childhood epilepsy with centrotemporal spikes is on the mild end of the spectrum, while epileptic encephalopathy with continuous spike-and-wave during sleep syndrome is on the severe end. Traditional antiseizure medicines and immunotherapy are currently used to treat severely affected patients, but the results are usually disappointing. The discovery that GRIN2A is the primary monogenic etiology of these diseases has opened the door to precision treatments. The GRIN2A gene encodes GluN2A protein, which constitutes a subunit of the NMDA receptor (NMDAR). The GRIN2A pathogenic variants cause gain or loss of function of NMDAR; the former can be treated with uncompetitive NMDAR antagonists, such as memantine, while the latter with NMDAR co-agonist serine. Hyper-precision therapies with various other effective agents are likely to be developed shortly to target the diverse functional effects of different variants. Precision treatments for GRIN2A-related disorders will benefit those who suffer from the condition and pave the way for new therapeutic approaches to a variety of other NMDAR-linked neurodegenerative and psychiatric diseases (schizophrenia, Parkinson's disease, Alzheimer's disease, and so on). Furthermore, more research into GRIN2A-related disorders will help us better understand the neuroinflammatory and neuroimmunological basis of epilepsy, as well as the pathological and physiological network activation mechanisms that cause sleep activation of central-temporal spikes and language impairment.
Collapse
|
41
|
Sharawat IK, Panda PK, Ramachandran A, Bhadoria AS. Cost-effectiveness of adrenocorticotropic hormone injection and oral prednisolone in patients with West syndrome: A comparative analysis. J Neurosci Rural Pract 2023; 14:103-110. [PMID: 36891085 PMCID: PMC9943942 DOI: 10.25259/jnrp-2022-6-31] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 09/20/2022] [Indexed: 12/23/2022] Open
Abstract
Objectives This study aims to compare the cost-effectiveness of oral prednisolone and adrenocorticotropic hormone injection in West syndrome patients, the two most common hormonal therapies used for this condition. Materials and Methods In this prospective and observational study, we documented sociodemographic, epilepsy, and development-related variables at baseline and up to 6 months after starting hormonal therapy, in all consecutive eligible patients of WS between August 2019 and June 2021, apart from the direct medical and non-medical costs and indirect health-care costs. We selected cost per quality-adjusted life-year (QALY) gained, per one patient with spasm freedom, one positive responder (>50% reduction in spasms), one relapse-free patient, and one patient with development gain. We determined whether incremental cost-effectiveness ratio for these parameters crossed the threshold value in base-case analysis and alternate scenario analysis. Results Out of 52 patients screened, 38 and 13 patients enrolled in ACTH and prednisolone group. On D28, 76% and 71% achieved spasm cessation (P = 0.78) and the total cost of treatment was INR 19783 and 8956 (P = 0.01), in ACTH and prednisolone group respectively. For all pre-specified parameters, the cost/effectiveness ratios including cost/QALY gain were higher in ACTH group and the corresponding ICER values for all these parameters crossed the threshold cost value of INR 148,777 in base-case analysis and also in alternative scenario analysis. Conclusion Treatment with oral prednisolone is more cost-effective as compared to ACTH injection for children with WS.
Collapse
Affiliation(s)
- Indar Kumar Sharawat
- Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Prateek Kumar Panda
- Department of Pediatrics, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| | - Aparna Ramachandran
- Department of Neurology, IQRAA International Hospital and Research Centre, Kozhikode, Kerala, India
| | - Ajeet Singh Bhadoria
- Department of Community and Family Medicine, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, India
| |
Collapse
|
42
|
Osborne JP, Edwards SW, Alber FD, Hancock E, Johnson AL, Kennedy CR, Likeman M, Lux AL, Mackay M, Mallick A, Newton RW, Nolan M, Pressler R, Rating D, Schmitt B, Verity CM, O'Callaghan FK. Prednisolone or tetracosactide depot for infantile epileptic spasms syndrome? A prospective analysis of data embedded within two randomised controlled trials. Eur J Paediatr Neurol 2023; 42:110-116. [PMID: 36621063 DOI: 10.1016/j.ejpn.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 12/20/2022] [Accepted: 12/21/2022] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To report a prospectively planned analysis of two randomised controlled trials with embedded comparisons of prednisolone versus tetracosactide depot for the treatment of infantile epileptic spasms syndrome (IESS). METHODS Individual patient data from patients randomly allocated to prednisolone or tetracosactide depot were analysed from two trials (UKISS, ICISS). The comparison was embedded within trials in which some patients also received vigabatrin but only patients receiving monotherapy with randomly allocated hormonal treatments are included in this analysis. The main outcome was cessation of spasms (Days 13-14 after randomisation). Lead time to treatment and underlying aetiology were taken into account. Cessation of spasms on Days 14-42 inclusive, electroclinical response (EEG Day 14), plus developmental and epilepsy outcomes (at 14 months in UKISS and 18 months in ICISS) are also reported. Minimum treatment was prednisolone 40 mg per day for two weeks or tetracosactide depot 0·5 mg IM on alternate days for two weeks, all followed by a reducing dose of prednisolone over two weeks. RESULTS 126 infants were included in this study. On tetracosactide depot, 47 of 62 (76%) were free of spasms on Days 13-14 compared to 43 of 64 (67%) on prednisolone (difference 9%, 95% CI -7·2% to +25·2%, chi square 1·15, p = 0·28). For Day 14-42 cessation of spasms, on tetracosactide depot, 41 of 61 (67%) were free of spasms compared to 35 of 62 (56%) on prednisolone (difference 11%, 95% CI -6·4% to +28·4%, chi square 1·51, p = 0·22). There was no significant difference in mean VABS score between infants who received prednisolone compared with those who received tetracosactide depot (74·8 (SD 18·3) versus 78·0 (SD 20·2) t = -0·91 p = 0·36). The proportion with ongoing epilepsy at the time of developmental assessment was 20 of 61 (33%) in the tetracosactide group compared with 26 out of 63 (41%) in the prednisolone group (difference 8%, 95% CI -9·2% to +25·2%, Chi [2] 0·95, p = 0·33). SIGNIFICANCE With hormone monotherapy, either prednisolone or tetracosactide depot may be recommended for infantile epileptic spasms syndrome.
Collapse
Affiliation(s)
- John P Osborne
- Department for Health, University of Bath, Claverton Down, Bath, BA2 7AY, UK; Children's Department, Royal United Hospitals Bath NHS Foundation Trust, Combe Park, Bath, BA1 3NG, UK
| | - Stuart W Edwards
- Department for Health, University of Bath, Claverton Down, Bath, BA2 7AY, UK; Children's Department, Royal United Hospitals Bath NHS Foundation Trust, Combe Park, Bath, BA1 3NG, UK
| | - Fabienne Dietrich Alber
- Division of Neurology/Neuropsychology, University Children's Hospital, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland
| | - Eleanor Hancock
- Children's Department, Royal United Hospitals Bath NHS Foundation Trust, Combe Park, Bath, BA1 3NG, UK
| | - Anthony L Johnson
- Medical Research Council Clinical Trials Unit at UCL, Institute of Clinical Trials Methodology, 90, High Holborn, London, WC1V 6LJ, UK
| | - Colin R Kennedy
- Clinical Neurosciences, Faculty of Medicine, University of Southampton. Mailpoint 803 G, Southampton General Hospital, Southampton, SO16 6YB, UK
| | - Marcus Likeman
- Department of Paediatric Radiology, Bristol Royal Hospital for Children, Upper Maudlin Street, Bristol, BS3 8AE, UK
| | - Andrew L Lux
- Department of Paediatric Neurology, Bristol Royal Hospital for Children, Upper Maudlin Street, Bristol, BS3 8AE, UK
| | - Mark Mackay
- Neurology Department, The Royal Children's Hospital Melbourne, 50 Flemington Road, Parkville, Victoria, 3058, Australia
| | - Andrew Mallick
- Department of Paediatric Neurology, Bristol Royal Hospital for Children, Upper Maudlin Street, Bristol, BS3 8AE, UK
| | - Richard W Newton
- Department of Neurology, Royal Manchester Children's Hospital, Hathersage Road, Manchester, M13 9WL, UK
| | - Melinda Nolan
- Starship Children's Health, Private bag 92024, Auckland, 1142, New Zealand
| | - Ronit Pressler
- UCL Institute of Child Health, Clinical Neurosciences, London, WC1N 1EH, UK
| | | | - Bernhard Schmitt
- Division of Paediatric Neurology, University Children's Hospital, Steinwiesstrasse 75, CH-8032, Zurich, Switzerland
| | | | - FinbarJ K O'Callaghan
- Head of Clinical Neurosciences Section, Room 41, 4th Floor PUW South, Institute of Child Health, University College London, London, WC1N 1EH, UK.
| |
Collapse
|
43
|
Iliopoulos G, Daoussis D. FDA-APPROVED INDICATIONS OF ADRENOCORTICOTROPIC HORMONE (ACTH) AS A DRUG: DOES IT HAVE A PLACE IN DISEASE MANAGEMENT TODAY? CENTRAL ASIAN JOURNAL OF MEDICAL HYPOTHESES AND ETHICS 2022. [DOI: 10.47316/cajmhe.2022.3.4.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
ACTH is a pituitary hormone important for proper function of adrenal glands, cortisol production as well as human physiology in general. It is involved in the pathogenesis of several endocrine disorders like Cushing syndrome and can be a useful diagnostic tool for diseases like primary adrenal insufficiency. Although popular as a hormone in endocrine system physiology and testing, ACTH has been used as a drug since the 1950s. Except for steroid-releasing properties, its mechanism of action involves a steroid-independent anti-inflammatory and possible immune-modulatory effect. Pharmaceutic ACTH has a wide range of indications approved by FDA and usually comes in the form of subcutaneous injections. In this narrative review, we accumulated what we considered as important data from reviews, cases and trials involving the most basic FDA-approved ACTH indications. A special emphasis was given on rheumatologic indications of ACTH. More large data studies need to be performed to assess ACTH usefulness, efficacy, safety and cost-effectiveness as a drug.
Collapse
|
44
|
Sakpichaisakul K, Boonkrongsak R, Lertbutsayanukul P, Iemwimangsa N, Klumsathian S, Panthan B, Trachoo O. Epileptic spasms related to neuronal differentiation factor 2 (NEUROD2) mutation respond to combined vigabatrin and high dose prednisolone therapy. BMC Neurol 2022; 22:461. [PMID: 36494631 PMCID: PMC9733267 DOI: 10.1186/s12883-022-02992-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Epileptic spasms are a devastating form of early infantile epileptic encephalopathy (EIEE) with various etiologies. Early diagnosis and a shorter lead time to treatment are crucial to stop the seizures and optimize the neurodevelopmental outcome. Genetic testing has become an integral part of epilepsy care that directly guides management and family planning and discovers new targeted treatments. Neuronal differentiation Factor 2 (NEUROD2) variants have recently been a cause of neurodevelopmental disorders (NDDs) and EIEEs with distinctive features. However, there is limited information about the clinical and electroencephalographic response of epileptic spasm treatment in NEUROD2-related NDD syndrome. CASE PRESENTATION We report a female patient of Southeast Asian ethnicity with global developmental delay and epileptic spasms commencing in the first few months of life. A novel de novo heterozygous pathogenic NEUROD2 variant, p. E130Q, was subsequently identified by whole-exome sequencing. Electroencephalogram before treatment showed multifocal independent spikes predominantly in both posterior head regions and demonstrated marked improvement following combined vigabatrin and high-dose prednisolone treatment. However, multiple courses of relapse occurred after weaning off the antiseizure medication. CONCLUSIONS We propose that epileptic spasms related to de novo NEUROD2 pathogenic variant respond well to combined vigabatrin and high-dose prednisolone therapy. These findings may imply the benefit of using combination therapy to treat epileptic spasms in NEUROD2-related NDD syndrome.
Collapse
Affiliation(s)
- Kullasate Sakpichaisakul
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, College of Medicine, Rangsit University, Bangkok, 10400, Thailand
| | - Rachata Boonkrongsak
- Department of Pediatrics, Queen Sirikit National Institute of Child Health, College of Medicine, Rangsit University, Bangkok, 10400, Thailand
| | | | - Nareenart Iemwimangsa
- Centre for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | - Sommon Klumsathian
- Centre for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand
| | | | - Objoon Trachoo
- Centre for Medical Genomics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, 10400, Thailand.
- Department of Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, 270 Rama 6 Road, Ratchathewi, Bangkok, 10400, Thailand.
| |
Collapse
|
45
|
Dzau W, Cheng S, Snell P, Fahey M, Scheffer IE, Harvey AS, Howell KB. Response to sequential treatment with prednisolone and vigabatrin in infantile spasms. J Paediatr Child Health 2022; 58:2197-2202. [PMID: 36054157 PMCID: PMC10087127 DOI: 10.1111/jpc.16181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 05/30/2022] [Accepted: 07/17/2022] [Indexed: 10/15/2022]
Abstract
AIM To report response to first treatment in infants with infantile spasms (IS), including incremental benefit of prednisolone 60 mg/day and vigabatrin following prednisolone 40 mg/day failure in infants commenced on the United Kingdom Infantile Spasms Study (UKISS) treatment sequence. METHODS In this retrospective analysis, we compared effectiveness of prednisolone, vigabatrin and nonstandard treatments as first treatment for IS. In infants who commenced the UKISS treatment sequence, we evaluated response to each step. Primary outcome was spasm cessation after 42 days. Secondary outcomes were severe side effects and spasm relapse after 42 days. RESULTS Treatment response data were available for 151 infants. First treatment was prednisolone in 99 infants, vigabatrin in 18 and nonstandard treatment in 34. The rate of spasm cessation with first treatment was significantly higher with prednisolone (62/99, 63%) than vigabatrin (5/18, 28%, P = 0.01) or nonstandard treatment (2/34, 5.9%, P < 0.01). Of 112 infants who commenced the UKISS treatment sequence, 71/112 (63%) responded to prednisolone 40 mg/day. Among non-responders, 12/29 (41%) subsequently responded to prednisolone 60 mg/day, and 10/22 (45%) to vigabatrin. Severe side effects and spasm relapse were not significantly different between each treatment. CONCLUSION We confirm higher rates of spasm cessation with initial treatment with prednisolone than vigabatrin and nonstandard therapy. Non-use of prednisolone as first treatment in over one third of infants highlights a concerning treatment gap. The UKISS treatment sequence has high overall treatment response (total 93/112; 83%), with similar benefit of subsequent prednisolone 60 mg/day and vigabatrin in prednisolone 40 mg/day non-responders.
Collapse
Affiliation(s)
- Winston Dzau
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia
| | - Sally Cheng
- Department of Neurology, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Penny Snell
- Neuroscience Research Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Michael Fahey
- Department of Paediatrics, Monash University, Melbourne, Victoria, Australia
| | - Ingrid E Scheffer
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.,Department of Neurology, The Royal Children's Hospital, Melbourne, Victoria, Australia.,Neuroscience Research Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Department of Medicine, University of Melbourne, Austin Health, Melbourne, Victoria, Australia.,Florey Institute of Neurosciences and Mental Health, Melbourne, Victoria, Australia
| | - A Simon Harvey
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.,Department of Neurology, The Royal Children's Hospital, Melbourne, Victoria, Australia.,Neuroscience Research Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Katherine B Howell
- Department of Paediatrics, University of Melbourne, Melbourne, Victoria, Australia.,Department of Neurology, The Royal Children's Hospital, Melbourne, Victoria, Australia.,Neuroscience Research Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia.,Florey Institute of Neurosciences and Mental Health, Melbourne, Victoria, Australia
| |
Collapse
|
46
|
Jain P, Sahu JK, Horn PS, Chau V, Go C, Mahood Q, Arya R. Treatment of children with infantile spasms: A network meta-analysis. Dev Med Child Neurol 2022; 64:1330-1343. [PMID: 35765990 DOI: 10.1111/dmcn.15330] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 05/28/2022] [Accepted: 05/30/2022] [Indexed: 12/12/2022]
Abstract
AIM We performed a systematic review and network meta-analysis (NMA) to obtain comparative effectiveness estimates and rankings of non-surgical interventions used to treat infantile spasms. METHOD All randomized controlled trials (RCTs) including children 2 months to 3 years of age with infantile spasms (with hypsarrhythmia or hypsarrhythmia variants on electroencephalography) receiving appropriate first-line medical treatment were included. Electroclinical and clinical remissions within 1 month of starting treatment were analyzed. RESULTS Twenty-two RCTs comparing first-line treatments for infantile spasms were reviewed; of these, 17 were included in the NMA. Both frequentist and Bayesian network rankings for electroclinical remission showed that high dose adrenocorticotropic hormone (ACTH), methylprednisolone, low dose ACTH and magnesium sulfate (MgSO4 ) combination, low dose ACTH, and high dose prednisolone were most likely to be the 'best' interventions, although these were not significantly different from each other. For clinical remission, low dose ACTH/MgSO4 combination, high dose ACTH (with/without vitamin B6 ), high dose prednisolone, and low dose ACTH were 'best'. INTERPRETATION Treatments including ACTH and high dose prednisolone are more effective in achieving electroclinical and clinical remissions for infantile spasms. WHAT THIS PAPER ADDS Adrenocorticotropic hormone and high dose prednisolone are more effective than other medications for infantile spasms. Symptomatic etiology decreases the likelihood of remission even after adjusting for treatment lag.
Collapse
Affiliation(s)
- Puneet Jain
- Epilepsy Program, Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Jitendra K Sahu
- Pediatric Neurology Unit, Department of Pediatrics, Postgraduate Institute of Medical Education & Research, Chandigarh, India
| | - Paul S Horn
- Comprehensive Epilepsy Center, Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Vann Chau
- Neonatal Neurology Program, Division of Neurology, Department of Pediatrics, Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Cristina Go
- Epilepsy Program, Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Quenby Mahood
- Hospital Library and Archives, Learning Institute, Toronto, ON, Canada
| | - Ravindra Arya
- Comprehensive Epilepsy Center, Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
47
|
Souza LDP, Bermudez BB, Bufara DC, Crippa ACDS. A Retrospective Cohort Study of Combined Therapy in West Syndrome associated with Trisomy 21. Child Neurol Open 2022; 9:2329048X221132639. [PMID: 36263394 PMCID: PMC9575436 DOI: 10.1177/2329048x221132639] [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: 06/30/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 11/19/2022] Open
Abstract
Background: West syndrome (WS) is a frequent epileptic encephalopathy associated with Down syndrome (DS). This study evaluated an outpatient protocol for WS in patients with DS who received vigabatrin (VGB) or VGB plus adrenocorticotrophic hormone. Methods: We analyzed infants treated in two neuropediatric centers from 2001-2021. We reviewed perinatal and familial history of epilepsy, spasm onset, treatment lag, electroencephalogram, neuroimaging, progression to epilepsy, and other neurological conditions. The outcomes were electroclinical resolution (ECR), relapses, and epilepsy progression. Results: Nineteen infants were included; 57.8% were male. The average spasm onset, follow-up, and treatment lag were 6.4 months, 8.15 years, and 2.33 months, respectively. Almost 74% had ECR after protocol intervention and minor epilepsy progression. Relapses occurred during combined therapy. Conclusions: The treatment protocol, especially combined therapy, was effective for WS in DS, impacting epilepsy progression and indicating the effectiveness of combined therapy to treat WS in patients with trisomy 21.
Collapse
Affiliation(s)
- Luciana de Paula Souza
- Paraná Federal University, Curitiba, PR, Brazil,Luciana de Paula Souza, Paraná Federal
University, Rua Quintino Bocaiuva, No. 325, Cabral 80060-900, Curitiba, Paraná,
Brazil.
| | | | | | | |
Collapse
|
48
|
Liu S, Fan M, Ma MD, Ge JF, Chen FH. Long non-coding RNAs: Potential therapeutic targets for epilepsy. Front Neurosci 2022; 16:986874. [PMID: 36278003 PMCID: PMC9582525 DOI: 10.3389/fnins.2022.986874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
Epilepsy is a common and disastrous neurological disorder characterized by abnormal firing of neurons in the brain, affecting about 70 million people worldwide. Long non-coding RNAs (LncRNAs) are a class of RNAs longer than 200 nucleotides without the capacity of protein coding, but they participate in a wide variety of pathophysiological processes. Alternated abundance and diversity of LncRNAs have been found in epilepsy patients and animal or cell models, suggesting a potential role of LncRNAs in epileptogenesis. This review will introduce the structure and function of LncRNAs, summarize the role of LncRNAs in the pathogenesis of epilepsy, especially its linkage with neuroinflammation, apoptosis, and transmitter balance, which will throw light on the molecular mechanism of epileptogenesis, and accelerate the clinical implementation of LncRNAs as a potential therapeutic target for treatment of epilepsy.
Collapse
Affiliation(s)
- Sen Liu
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Hefei, China
| | - Min Fan
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Hefei, China
| | - Meng-Die Ma
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Hefei, China
| | - Jin-Fang Ge
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Hefei, China
- *Correspondence: Jin-Fang Ge,
| | - Fei-Hu Chen
- School of Pharmacy, Anhui Medical University, Hefei, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Hefei, China
- Anhui Provincial Laboratory of Inflammatory and Immunity Disease, Hefei, China
- Fei-Hu Chen,
| |
Collapse
|
49
|
Jiang Y, Zou N, Luo Y, Cheng M, Liao S, Hong S, Liang X, Zhong M, Li T, Jiang L. Cohort study of infantile epileptic spasms syndrome: etiological analysis and treatment of corticosteroids. Seizure 2022; 101:120-126. [DOI: 10.1016/j.seizure.2022.07.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/24/2022] [Accepted: 07/30/2022] [Indexed: 11/13/2022] Open
|
50
|
Yan J, Kothur K, Innes EA, Han VX, Jones HF, Patel S, Tsang E, Webster R, Gupta S, Troedson C, Menezes MP, Antony J, Ardern-Holmes S, Tantsis E, Mohammad S, Wienholt L, Pires AS, Heng B, Guillemin GJ, Guller A, Gill D, Bandodkar S, Dale RC. Decreased cerebrospinal fluid kynurenic acid in epileptic spasms: A biomarker of response to corticosteroids. EBioMedicine 2022; 84:104280. [PMID: 36174397 PMCID: PMC9515432 DOI: 10.1016/j.ebiom.2022.104280] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/15/2022] Open
Abstract
Background Epileptic (previously infantile) spasms is the most common epileptic encephalopathy occurring during infancy and is frequently associated with abnormal neurodevelopmental outcomes. Epileptic spasms have a diverse range of known (genetic, structural) and unknown aetiologies. High dose corticosteroid treatment for 4 weeks often induces remission of spasms, although the mechanism of action of corticosteroid is unclear. Animal models of epileptic spasms have shown decreased brain kynurenic acid, which is increased after treatment with the ketogenic diet. We quantified kynurenine pathway metabolites in the cerebrospinal fluid (CSF) of infants with epileptic spasms and explored clinical correlations. Methods A panel of nine metabolites in the kynurenine pathway (tryptophan, kynurenine, kynurenic acid, 3-hydroxykynurenine, xanthurenic acid, anthranilic acid, 3-hydroxyanthranilic acid, quinolinic acid, and picolinic acid) were measured using liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS). CSF collected from paediatric patients less than 3 years of age with epileptic spasms (n=34, 19 males, mean age 0.85, median 0.6, range 0.3–3 yrs) were compared with other epilepsy syndromes (n=26, 9 males, mean age 1.44, median 1.45, range 0.3–3 yrs), other non-inflammatory neurological diseases (OND) (n=29, 18 males, mean age 1.47, median 1.6, range 0.1–2.9 yrs) and inflammatory neurological controls (n=12, 4 males, mean age 1.80, median 1.80, range 0.8–2.5 yrs). Findings There was a statistically significant decrease of CSF kynurenic acid in patients with epileptic spasms compared to OND (p<0.0001). In addition, the kynurenic acid/kynurenine (KYNA/KYN) ratio was lower in the epileptic spasms subgroup compared to OND (p<0.0001). Epileptic spasms patients who were steroid responders or partial steroid responders had lower KYNA/KYN ratio compared to patients who were refractory to steroids (p<0.005, p<0.05 respectively). Interpretation This study demonstrates decreased CSF kynurenic acid and KYNA/KYN in epileptic spasms, which may also represent a biomarker for steroid responsiveness. Given the anti-inflammatory and neuroprotective properties of kynurenic acid, further therapeutics able to increase kynurenic acid should be explored. Funding Financial support for the study was granted by Dale NHMRC Investigator grant APP1193648, Petre Foundation, Cerebral Palsy Alliance and Department of Biochemistry at the Children's Hospital at Westmead. Prof Guillemin is funded by NHMRC Investigator grant APP1176660 and Macquarie University.
Collapse
Affiliation(s)
- Jingya Yan
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Department of Biochemistry, The Children's Hospital at Westmead, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Kavitha Kothur
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Emily A Innes
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Velda X Han
- Khoo Teck Puat-National University Children's Medical Institute, National University Health System, Singapore, Singapore
| | - Hannah F Jones
- Starship Hospital, Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - Shrujna Patel
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Erica Tsang
- Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Richard Webster
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Sachin Gupta
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Christopher Troedson
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Manoj P Menezes
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Jayne Antony
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Simone Ardern-Holmes
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Esther Tantsis
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Shekeeb Mohammad
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Louise Wienholt
- Department of Clinical Immunology and Allergy, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Ananda S Pires
- Neuroinflammation Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, Australia
| | - Benjamin Heng
- Neuroinflammation Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, Australia
| | - Gilles J Guillemin
- Neuroinflammation Group, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, NSW, Australia
| | - Anna Guller
- Computational NeuroSurgery Lab, Macquarie University, Sydney, NSW, Australia
| | - Deepak Gill
- TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, The University of Sydney, Westmead, New South Wales, Australia
| | - Sushil Bandodkar
- Department of Biochemistry, The Children's Hospital at Westmead, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia
| | - Russell C Dale
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia; Clinical School, The Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, NSW, Australia.
| |
Collapse
|