Pub Date : 2025-12-15DOI: 10.1007/s40263-025-01252-x
Marianna Vinokur, Eric S Schwenk, Sawsan Alabad, Winston Hamilton, Hsiangkuo Yuan, Samuel Fallon, Michael J Marmura
<p><strong>Background: </strong>Ketamine infusions are used in the inpatient setting for refractory chronic migraine but are associated with neurotoxicity in rodents at high doses and memory deficits in illicit users. The relationship between ketamine infusions and cognitive function in refractory chronic migraine patients after infusions is unknown. We aimed to determine if patients receiving ketamine infusions for refractory chronic migraine experience changes in cognitive function as assessed by the telephone Montreal Cognitive Assessment.</p><p><strong>Methods: </strong>Adults 18 years or older who were diagnosed with refractory chronic migraine and met criteria for hospitalization with an elective ketamine infusion were recruited for this prospective observational study. All patients had Migraine Disability Assessment grade IV (severe disability) and had previously failed an inpatient treatment with at least one other intravenous infusion. Baseline assessments included current and average pain levels, monthly migraine days, depression history, medications, and initial assessments of the telephone Montreal Cognitive Assessment, Migraine Disability Assessment, and Headache Impact Test-6. Patients were admitted to a dedicated headache unit within 1 month of baseline assessments to undergo a 5-day continuous ketamine infusion to a maximum rate of 1 mg/kg/h. The aforementioned assessments were then repeated at 1, 6, and 12 months. The primary outcome was the change over time in the telephone Montreal Cognitive Assessment, which was analyzed using generalized estimating equations adjusted for age and sex.</p><p><strong>Results: </strong>A total of 23 patients were analyzed. The mean age was 44.8 <math><mo>±</mo></math> 11.5 years, and 87% were female. A history of depression was present in 82.6%. The estimated marginal mean telephone Montreal Cognitive Assessment score changed from 18.8 <math><mo>±</mo></math> 0.7 to 19.9 <math><mo>±</mo></math> 0.7 at 1 month (p < 0.001), 19.2 <math><mo>±</mo></math> 0.8 at 6 months (p = 0.390), and 18.3 <math><mo>±</mo></math> 0.9 at 12 months (p = 0.382). Only the change at 1 month reached the minimal clinically important difference of 1 point. Monthly migraine days decreased from a baseline of 27.1 <math><mo>±</mo></math> 1.6 to 24.4 <math><mo>±</mo></math> 2.1 at 1 month (p = 0.08), 22.4 <math><mo>±</mo></math> 2.3 at 6 months (p = 0.05), and 22.3 <math><mo>±</mo></math> 2.2 at 12 months (p = 0.026).</p><p><strong>Conclusion: </strong>The study suggests that measurable cognitive impairment did not occur over the course of the 1-year study period in most patients with refractory chronic migraine after receiving a ketamine infusion. However, a few patients experienced worsening telephone Montreal Cognitive Assessment scores, and furthermore, the small sample size and lack of a control group prevent any definitive conclusions. Larger follow-up studies would further establish the safety of ketamine treatment in h
{"title":"Assessing Cognitive Function over Time in Patients with Refractory Chronic Migraine Who Received Ketamine Infusions: A Prospective, Observational Study.","authors":"Marianna Vinokur, Eric S Schwenk, Sawsan Alabad, Winston Hamilton, Hsiangkuo Yuan, Samuel Fallon, Michael J Marmura","doi":"10.1007/s40263-025-01252-x","DOIUrl":"https://doi.org/10.1007/s40263-025-01252-x","url":null,"abstract":"<p><strong>Background: </strong>Ketamine infusions are used in the inpatient setting for refractory chronic migraine but are associated with neurotoxicity in rodents at high doses and memory deficits in illicit users. The relationship between ketamine infusions and cognitive function in refractory chronic migraine patients after infusions is unknown. We aimed to determine if patients receiving ketamine infusions for refractory chronic migraine experience changes in cognitive function as assessed by the telephone Montreal Cognitive Assessment.</p><p><strong>Methods: </strong>Adults 18 years or older who were diagnosed with refractory chronic migraine and met criteria for hospitalization with an elective ketamine infusion were recruited for this prospective observational study. All patients had Migraine Disability Assessment grade IV (severe disability) and had previously failed an inpatient treatment with at least one other intravenous infusion. Baseline assessments included current and average pain levels, monthly migraine days, depression history, medications, and initial assessments of the telephone Montreal Cognitive Assessment, Migraine Disability Assessment, and Headache Impact Test-6. Patients were admitted to a dedicated headache unit within 1 month of baseline assessments to undergo a 5-day continuous ketamine infusion to a maximum rate of 1 mg/kg/h. The aforementioned assessments were then repeated at 1, 6, and 12 months. The primary outcome was the change over time in the telephone Montreal Cognitive Assessment, which was analyzed using generalized estimating equations adjusted for age and sex.</p><p><strong>Results: </strong>A total of 23 patients were analyzed. The mean age was 44.8 <math><mo>±</mo></math> 11.5 years, and 87% were female. A history of depression was present in 82.6%. The estimated marginal mean telephone Montreal Cognitive Assessment score changed from 18.8 <math><mo>±</mo></math> 0.7 to 19.9 <math><mo>±</mo></math> 0.7 at 1 month (p < 0.001), 19.2 <math><mo>±</mo></math> 0.8 at 6 months (p = 0.390), and 18.3 <math><mo>±</mo></math> 0.9 at 12 months (p = 0.382). Only the change at 1 month reached the minimal clinically important difference of 1 point. Monthly migraine days decreased from a baseline of 27.1 <math><mo>±</mo></math> 1.6 to 24.4 <math><mo>±</mo></math> 2.1 at 1 month (p = 0.08), 22.4 <math><mo>±</mo></math> 2.3 at 6 months (p = 0.05), and 22.3 <math><mo>±</mo></math> 2.2 at 12 months (p = 0.026).</p><p><strong>Conclusion: </strong>The study suggests that measurable cognitive impairment did not occur over the course of the 1-year study period in most patients with refractory chronic migraine after receiving a ketamine infusion. However, a few patients experienced worsening telephone Montreal Cognitive Assessment scores, and furthermore, the small sample size and lack of a control group prevent any definitive conclusions. Larger follow-up studies would further establish the safety of ketamine treatment in h","PeriodicalId":10508,"journal":{"name":"CNS drugs","volume":" ","pages":""},"PeriodicalIF":7.4,"publicationDate":"2025-12-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145755513","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-12DOI: 10.1007/s40263-025-01249-6
Tommaso B Jannini, Ludovico Alisi, Francesca Giovannetti, Marta Armentano, Giorgio Di Lorenzo, Cinzia Niolu, Alberto Siracusano
Background and objectives: Glaucoma is one of the leading causes of irreversible blindness worldwide and is increasingly recognized as a potential adverse effect of various pharmacological agents. It has been suggested that psychotropic medications influence glaucoma risk, but findings across studies have remained inconsistent. We aimed to clarify the association between psychotropic drug use and glaucoma through a Bayesian meta-analysis.
Methods: We conducted a systematic literature search up to December 2024. Studies that examined the relationship between psychotropic medications and glaucoma or intraocular pressure (IOP), and reported odds ratios (ORs), relative risks (RRs), or mean differences, were eligible. Bayesian random-effects models were applied using informative priors based on existing evidence for specific compounds. Estimates were reported as pooled ORs and Hedges' g with corresponding 95% credible intervals (CrIs).
Results: A total of 22 observational studies, including 293,228 users of psychotropic medications, met the inclusion criteria. Selective serotonin reuptake inhibitors (SSRIs) were associated with a modestly reduced risk of open-angle glaucoma (OR = 0.832, 95% CrI: 0.753-0.921) and a small but consistent reduction in IOP (Hedges' g = -0.332, 95% CrI: -0.487 to -0.179). Although tricyclic antidepressants were expected to have a direct causative effect, results did not show a significant association with glaucoma risk (OR = 1.466, 95% CrI: 0.700-3.338). Benzodiazepines were associated with a significantly increased risk of glaucoma (OR = 1.550, 95% CrI: 1.436-1.674), with consistent effects across both short- and long-acting compounds. Topiramate demonstrated a strong association with acute angle-closure glaucoma (OR = 3.930, 95% CrI: 1.784-11.465), in accordance with its known mechanism of inducing anterior displacement of the lens-iris diaphragm. Studies on methylphenidate, limited to pediatric populations, suggested a modest but non-significant reduction in IOP compared with untreated individuals. Evidence on antipsychotics was inconsistent, precluding any quantitative synthesis.
Conclusions: While some drug classes (e.g., benzodiazepines, topiramate) show a strong association with glaucoma, results for other compounds must be taken judiciously. The high level of heterogeneity, and the presence of special populations suggest caution when moving to real-life scenarios. Nonetheless, our results highlight the importance of ophthalmologic monitoring in patients prescribed with psychiatric drugs (e.g., benzodiazepines or topiramate), at risk for angle closure.
{"title":"The Effect of Psychotropic Medications on Glaucoma Risk and Intraocular Pressure: A Bayesian Meta-Analysis.","authors":"Tommaso B Jannini, Ludovico Alisi, Francesca Giovannetti, Marta Armentano, Giorgio Di Lorenzo, Cinzia Niolu, Alberto Siracusano","doi":"10.1007/s40263-025-01249-6","DOIUrl":"https://doi.org/10.1007/s40263-025-01249-6","url":null,"abstract":"<p><strong>Background and objectives: </strong>Glaucoma is one of the leading causes of irreversible blindness worldwide and is increasingly recognized as a potential adverse effect of various pharmacological agents. It has been suggested that psychotropic medications influence glaucoma risk, but findings across studies have remained inconsistent. We aimed to clarify the association between psychotropic drug use and glaucoma through a Bayesian meta-analysis.</p><p><strong>Methods: </strong>We conducted a systematic literature search up to December 2024. Studies that examined the relationship between psychotropic medications and glaucoma or intraocular pressure (IOP), and reported odds ratios (ORs), relative risks (RRs), or mean differences, were eligible. Bayesian random-effects models were applied using informative priors based on existing evidence for specific compounds. Estimates were reported as pooled ORs and Hedges' g with corresponding 95% credible intervals (CrIs).</p><p><strong>Results: </strong>A total of 22 observational studies, including 293,228 users of psychotropic medications, met the inclusion criteria. Selective serotonin reuptake inhibitors (SSRIs) were associated with a modestly reduced risk of open-angle glaucoma (OR = 0.832, 95% CrI: 0.753-0.921) and a small but consistent reduction in IOP (Hedges' g = -0.332, 95% CrI: -0.487 to -0.179). Although tricyclic antidepressants were expected to have a direct causative effect, results did not show a significant association with glaucoma risk (OR = 1.466, 95% CrI: 0.700-3.338). Benzodiazepines were associated with a significantly increased risk of glaucoma (OR = 1.550, 95% CrI: 1.436-1.674), with consistent effects across both short- and long-acting compounds. Topiramate demonstrated a strong association with acute angle-closure glaucoma (OR = 3.930, 95% CrI: 1.784-11.465), in accordance with its known mechanism of inducing anterior displacement of the lens-iris diaphragm. Studies on methylphenidate, limited to pediatric populations, suggested a modest but non-significant reduction in IOP compared with untreated individuals. Evidence on antipsychotics was inconsistent, precluding any quantitative synthesis.</p><p><strong>Conclusions: </strong>While some drug classes (e.g., benzodiazepines, topiramate) show a strong association with glaucoma, results for other compounds must be taken judiciously. The high level of heterogeneity, and the presence of special populations suggest caution when moving to real-life scenarios. Nonetheless, our results highlight the importance of ophthalmologic monitoring in patients prescribed with psychiatric drugs (e.g., benzodiazepines or topiramate), at risk for angle closure.</p>","PeriodicalId":10508,"journal":{"name":"CNS drugs","volume":" ","pages":""},"PeriodicalIF":7.4,"publicationDate":"2025-12-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145741583","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
<p><strong>Background and objectives: </strong>The efficacy and safety of lemborexant, a dual orexin receptor antagonist, for treating insomnia associated with depressive episodes remain unclear. This pilot study aimed to evaluate the changes in clinical symptoms following the initiation of lemborexant treatment and its safety in patients with insomnia comorbid with depressive episodes.</p><p><strong>Methods: </strong>The inclusion criteria for this multicenter, prospective, interventional, open-label pilot study conducted in Japan were adults (≥ 18 years) diagnosed with insomnia and either major depressive disorder (MDD) or bipolar disorder (BD) according to DSM-5, receiving treatment for a depressive episode for ≥ 4 weeks, and having a baseline Hamilton Depression Rating Scale (HAM-D-17) score of ≥ 8. Patients were classified into four cohorts on the basis of their MDD or BD diagnosis and concomitant insomnia medication use (lemborexant add-on or monotherapy). After a 2-week lead-in period with unchanged prior medications for insomnia MDD or BD, 12-week lemborexant treatment was administered. The primary endpoint was the change from baseline in the Insomnia Severity Index (ISI) total score at week 4, which was analyzed using paired t-tests with a two-sided significance level of 5%. For other endpoints, summary statistics and 95% confidence intervals (CIs) for observed values and changes from baseline were calculated by cohort. Safety outcomes included the evaluation of the safety and tolerability of lemborexant during the treatment period.</p><p><strong>Results: </strong>A total of 83 patients with comorbid insomnia and depressive episodes were enrolled (MDD add-on cohort, n = 29; MDD monotherapy, n = 23; BD add-on, n = 16; and BD monotherapy, n = 15). The average age was 47.2 years and 64.6% were female. Mean baseline ISI total score (SD) was 14.5 (4.8), 13.2 (5.5), 13.5 (6.0), and 11.8 (4.6), respectively. At week 4, mean change from baseline in the ISI total score [95% CI, p-value] was - 2.3 [- 3.9, - 0.7, p = 0.0065], - 3.2 [- 5.0, - 1.3, p = 0.0018], - 5.2 [- 7.9, - 2.5, p = 0.0012], - 4.5 [- 6.3, - 2.6, p = 0.0001], respectively. Improvements were sustained through week 12. Sleep diary measures such as sleep onset latency and total sleep time improved in some cohorts, but the results were inconsistent. Mean baseline HAM-D-17 scores were 13.6 (5.1), 12.6 (5.1), 13.9 (4.3), and 11.4 (3.4), respectively, with mean changes at week 12 of - 6.4 [- 7.8, - 4.9], - 6.2 [- 7.7, - 4.6], - 5.2 [- 8.4, - 2.1], and - 5.4 [- 7.2, - 3.5], respectively. No serious treatment-emergent adverse events (TEAEs) were reported, with TEAE incidence ranging from 30% to 40%, all mild to moderate in severity. Somnolence was the most common adverse reaction, reported in 10.3-25.0% of cohorts except BD monotherapy. Nightmare followed, occurring in 4.3-12.5% across cohorts.</p><p><strong>Conclusions: </strong>Lemborexant was associated with improvements in insomnia seve
{"title":"A 12-Week, Open-Label, Multicenter Pilot Study to Evaluate the Efficacy and Safety of Lemborexant in Patients with Insomnia Comorbid with Depressive Episodes (SELENADE).","authors":"Yoshikazu Takaesu, Hiroyuki Muraoka, Masahiro Takeshima, Masaki Kato, Hirofumi Hirakawa, Hikaru Hori, Ken Inada, Hitoshi Sakurai, Motohiro Ozone, Yosuke Koshikawa, Tomohiko Murao, Takeshi Terao, Koichiro Watanabe, Haruka Yokoyama, Michinori Koebisu, Yuka Kawatsu, Yoshiteru Takekita","doi":"10.1007/s40263-025-01245-w","DOIUrl":"https://doi.org/10.1007/s40263-025-01245-w","url":null,"abstract":"<p><strong>Background and objectives: </strong>The efficacy and safety of lemborexant, a dual orexin receptor antagonist, for treating insomnia associated with depressive episodes remain unclear. This pilot study aimed to evaluate the changes in clinical symptoms following the initiation of lemborexant treatment and its safety in patients with insomnia comorbid with depressive episodes.</p><p><strong>Methods: </strong>The inclusion criteria for this multicenter, prospective, interventional, open-label pilot study conducted in Japan were adults (≥ 18 years) diagnosed with insomnia and either major depressive disorder (MDD) or bipolar disorder (BD) according to DSM-5, receiving treatment for a depressive episode for ≥ 4 weeks, and having a baseline Hamilton Depression Rating Scale (HAM-D-17) score of ≥ 8. Patients were classified into four cohorts on the basis of their MDD or BD diagnosis and concomitant insomnia medication use (lemborexant add-on or monotherapy). After a 2-week lead-in period with unchanged prior medications for insomnia MDD or BD, 12-week lemborexant treatment was administered. The primary endpoint was the change from baseline in the Insomnia Severity Index (ISI) total score at week 4, which was analyzed using paired t-tests with a two-sided significance level of 5%. For other endpoints, summary statistics and 95% confidence intervals (CIs) for observed values and changes from baseline were calculated by cohort. Safety outcomes included the evaluation of the safety and tolerability of lemborexant during the treatment period.</p><p><strong>Results: </strong>A total of 83 patients with comorbid insomnia and depressive episodes were enrolled (MDD add-on cohort, n = 29; MDD monotherapy, n = 23; BD add-on, n = 16; and BD monotherapy, n = 15). The average age was 47.2 years and 64.6% were female. Mean baseline ISI total score (SD) was 14.5 (4.8), 13.2 (5.5), 13.5 (6.0), and 11.8 (4.6), respectively. At week 4, mean change from baseline in the ISI total score [95% CI, p-value] was - 2.3 [- 3.9, - 0.7, p = 0.0065], - 3.2 [- 5.0, - 1.3, p = 0.0018], - 5.2 [- 7.9, - 2.5, p = 0.0012], - 4.5 [- 6.3, - 2.6, p = 0.0001], respectively. Improvements were sustained through week 12. Sleep diary measures such as sleep onset latency and total sleep time improved in some cohorts, but the results were inconsistent. Mean baseline HAM-D-17 scores were 13.6 (5.1), 12.6 (5.1), 13.9 (4.3), and 11.4 (3.4), respectively, with mean changes at week 12 of - 6.4 [- 7.8, - 4.9], - 6.2 [- 7.7, - 4.6], - 5.2 [- 8.4, - 2.1], and - 5.4 [- 7.2, - 3.5], respectively. No serious treatment-emergent adverse events (TEAEs) were reported, with TEAE incidence ranging from 30% to 40%, all mild to moderate in severity. Somnolence was the most common adverse reaction, reported in 10.3-25.0% of cohorts except BD monotherapy. Nightmare followed, occurring in 4.3-12.5% across cohorts.</p><p><strong>Conclusions: </strong>Lemborexant was associated with improvements in insomnia seve","PeriodicalId":10508,"journal":{"name":"CNS drugs","volume":" ","pages":""},"PeriodicalIF":7.4,"publicationDate":"2025-12-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145667383","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Background: Prolactin elevation associated with antipsychotic use significantly affects medication adherence and long-term treatment outcomes in patients with schizophrenia. The currently available data are insufficient to guide the monitoring and management of elevated prolactin levels in patients on antipsychotic medications. This study aimed to explore the patterns of prolactin level elevation associated with nine second-generation antipsychotics (SGAs) in a real-world setting and compare the associated risks.
Methods: This retrospective cohort study utilized data from the inpatient electronic medical records of a large mental health center in China from 2007 to 2019. The study included patients diagnosed with schizophrenia (ICD-10 criteria) who received SGA therapy and whose serum prolactin levels were measured. Exposures were the use of nine specific SGAs (amisulpride, risperidone, paliperidone, ziprasidone, olanzapine, perospirone, quetiapine, clozapine, or aripiprazole), including polytherapy and monotherapy. The primary outcome was incident prolactin elevation in patients during hospitalization. An adjusted stratified Cox proportional hazards regression analysis was used to compare the hazard ratios (HRs) of prolactin level elevation across the nine SGAs. In addition, a dose-response analysis of these SGAs was conducted using the defined daily dose (DDD) method. Dose categories were as follows: < 0.6 DDDs/day (low dose), 0.6 to < 1.1 DDDs/day (medium dose), and ≥ 1.1 DDDs/day (high dose).
Results: This study included 6489 patients with schizophrenia (mean [SD] age, 35.1 [14.2] years; 3396 males [52.3%]). Compared with the nonexposure, amisulpride (HR 2.76, 95% confidence interval [CI] 2.12-3.59), risperidone (HR 2.70, 95% CI 2.30-3.16), paliperidone (HR 1.84, 95% CI 1.37-2.46), and ziprasidone (HR 1.36, 95% CI 1.06-1.76) were associated with the highest risk of prolactin elevation. In contrast, quetiapine (HR 0.73, 95% CI 0.61-0.87), clozapine (HR 0.59, 95% CI 0.46-0.76), and aripiprazole (HR 0.30, 95% CI 0.23-0.37) were associated with the lowest risk. Amisulpride posed the highest risk among male patients, whereas risperidone posed the highest risk among female patients. Different types of dose-response associations were detected in seven SGAs.
Conclusion: This cohort study, conducted in an inpatient setting, identified different risks of prolactin elevation associated with SGAs, along with their dose-response curves. Sex and age must be considered when prolactin elevation is analyzed in patients with schizophrenia who are treated with SGAs.
背景:催乳素升高与抗精神病药物的使用显著影响精神分裂症患者的药物依从性和长期治疗结果。目前可用的数据不足以指导使用抗精神病药物的患者催乳素水平升高的监测和管理。本研究旨在探讨现实环境中9种第二代抗精神病药物(SGAs)的催乳素水平升高模式,并比较相关风险。方法:本回顾性队列研究利用中国一家大型精神卫生中心2007 - 2019年住院患者电子病历数据。该研究包括接受SGA治疗的精神分裂症患者(ICD-10标准),并测量其血清催乳素水平。暴露是使用九种特定的SGAs(氨硫pride,利培酮,帕利哌酮,齐拉西酮,奥氮平,perospirone,喹硫平,氯氮平或阿立哌唑),包括综合治疗和单一治疗。主要结局是住院期间患者的泌乳素升高。采用调整后的分层Cox比例风险回归分析比较9个SGAs催乳素水平升高的风险比(hr)。此外,采用限定日剂量(DDD)法对这些SGAs进行了剂量-反应分析。剂量分类为:< 0.6 DDDs/天(低剂量)、0.6 ~ < 1.1 DDDs/天(中剂量)、≥1.1 DDDs/天(高剂量)。结果:本研究纳入6489例精神分裂症患者(平均[SD]年龄35.1[14.2]岁,男性3396例[52.3%])。与未暴露组相比,氨硫傲(HR 2.76, 95%可信区间[CI] 2.12-3.59)、利培酮(HR 2.70, 95% CI 2.30-3.16)、帕利培酮(HR 1.84, 95% CI 1.37-2.46)和齐拉西酮(HR 1.36, 95% CI 1.06-1.76)与泌乳素升高的最高风险相关。相反,喹硫平(HR 0.73, 95% CI 0.61-0.87)、氯氮平(HR 0.59, 95% CI 0.46-0.76)和阿立哌唑(HR 0.30, 95% CI 0.23-0.37)与最低风险相关。氨硫pride在男性患者中风险最高,而利培酮在女性患者中风险最高。在7个SGAs中检测到不同类型的剂量-反应关联。结论:这项在住院患者环境中进行的队列研究,确定了与SGAs相关的催乳素升高的不同风险,以及它们的剂量-反应曲线。在分析接受SGAs治疗的精神分裂症患者的催乳素升高时,必须考虑性别和年龄。试验注册:ClinicalTrials.gov标识符:NCT04002258。
{"title":"Patterns of Serum Prolactin Elevation Associated with Nine Second-Generation Antipsychotics in a Large Cohort of Patients with Schizophrenia.","authors":"Lei Zhang, Yuzhen Zheng, Jingjing Huang, Wenjuan Yu, Lihong Zhou, Luyao He, Yange Li, Hao Hu, Guanjun Li, Yifeng Shen, Jianping Zhang, Huafang Li","doi":"10.1007/s40263-025-01216-1","DOIUrl":"10.1007/s40263-025-01216-1","url":null,"abstract":"<p><strong>Background: </strong>Prolactin elevation associated with antipsychotic use significantly affects medication adherence and long-term treatment outcomes in patients with schizophrenia. The currently available data are insufficient to guide the monitoring and management of elevated prolactin levels in patients on antipsychotic medications. This study aimed to explore the patterns of prolactin level elevation associated with nine second-generation antipsychotics (SGAs) in a real-world setting and compare the associated risks.</p><p><strong>Methods: </strong>This retrospective cohort study utilized data from the inpatient electronic medical records of a large mental health center in China from 2007 to 2019. The study included patients diagnosed with schizophrenia (ICD-10 criteria) who received SGA therapy and whose serum prolactin levels were measured. Exposures were the use of nine specific SGAs (amisulpride, risperidone, paliperidone, ziprasidone, olanzapine, perospirone, quetiapine, clozapine, or aripiprazole), including polytherapy and monotherapy. The primary outcome was incident prolactin elevation in patients during hospitalization. An adjusted stratified Cox proportional hazards regression analysis was used to compare the hazard ratios (HRs) of prolactin level elevation across the nine SGAs. In addition, a dose-response analysis of these SGAs was conducted using the defined daily dose (DDD) method. Dose categories were as follows: < 0.6 DDDs/day (low dose), 0.6 to < 1.1 DDDs/day (medium dose), and ≥ 1.1 DDDs/day (high dose).</p><p><strong>Results: </strong>This study included 6489 patients with schizophrenia (mean [SD] age, 35.1 [14.2] years; 3396 males [52.3%]). Compared with the nonexposure, amisulpride (HR 2.76, 95% confidence interval [CI] 2.12-3.59), risperidone (HR 2.70, 95% CI 2.30-3.16), paliperidone (HR 1.84, 95% CI 1.37-2.46), and ziprasidone (HR 1.36, 95% CI 1.06-1.76) were associated with the highest risk of prolactin elevation. In contrast, quetiapine (HR 0.73, 95% CI 0.61-0.87), clozapine (HR 0.59, 95% CI 0.46-0.76), and aripiprazole (HR 0.30, 95% CI 0.23-0.37) were associated with the lowest risk. Amisulpride posed the highest risk among male patients, whereas risperidone posed the highest risk among female patients. Different types of dose-response associations were detected in seven SGAs.</p><p><strong>Conclusion: </strong>This cohort study, conducted in an inpatient setting, identified different risks of prolactin elevation associated with SGAs, along with their dose-response curves. Sex and age must be considered when prolactin elevation is analyzed in patients with schizophrenia who are treated with SGAs.</p><p><strong>Trial registration: </strong>ClinicalTrials.gov identifier: NCT04002258.</p>","PeriodicalId":10508,"journal":{"name":"CNS drugs","volume":" ","pages":"1317-1330"},"PeriodicalIF":7.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144882352","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-09-12DOI: 10.1007/s40263-025-01226-z
Christian Hölscher
Alzheimer's disease (AD) and Parkinson's disease (PD) are chronic neurodegenerative disorders with few effective drug treatments available. An underrated element of these diseases is that glucose uptake and energy utilization is much reduced in neurons. In the brains of patients, signaling of insulin, insulin-like growth factor 1, and other growth factors is downregulated early on. This leads to reduced glucose utilization and impaired mitochondrial function. In an attempt to compensate for the loss, other pathways are upregulated, e.g., the increased use of ketones produced from fatty acids by astrocytes that are shuttled to neurons. In addition, amino acids are increasingly used to generate energy. Despite this, neurons generate less and less energy over time, leading to impaired synaptic activity, reduced cell repair, mitogenesis, autophagy, the accumulation of misfolded proteins, and finally, to cell death. At the same time, the chronic inflammation response in the brain that is part of these diseases continues to damage neurons. Glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are peptide hormones and growth factors that have shown neuroprotective effects in animal studies and in clinical trials. GLP-1 and GIP receptor agonists were able to reduce inflammation while normalizing growth factor signaling and energy utilization in the brain. Insulin signaling was improved and energy utilization, glucose uptake, mitogenesis, and mitochondrial functionality was brought back to physiological levels. In addition, the chronic inflammation response and the levels of proinflammatory cytokines in the brain were much reduced. Clinical trials testing GLP-1 receptor agonists in patients with AD or PD have been conducted and have shown first successes, serving as proof of concept that activating GLP-1 receptor is a sensible strategy to treat AD/PD. A phase II study testing liraglutide in patients with AD showed first improvements, and two phase II trials testing exendin-4 (exenatide, Bydureon®) or lixisenatide showed improvements in patients with PD. A recent phase III trial testing exendin-4 did not show an improvement, which may be linked to the lack of insulin desensitization in the study participants. Semaglutide (Rybelsus®; Wegovy®; Ozempic®) is currently in two phase III trials for AD. Current drugs that are on the market have a long half-life in the blood and do not readily cross the blood-brain barrier (BBB). Newer dual GLP-1/GIP receptor agonists have been developed that can more easily cross the BBB and that show improved protection in animal models of AD and PD. Therefore, GLP-1 and GIP receptor agonists that can cross the BBB show promise as treatments for chronic neurodegenerative disorders.
{"title":"Incretin Hormones GLP-1 and GIP Normalize Energy Utilization and Reduce Inflammation in the Brain in Alzheimer's Disease and Parkinson's Disease: From Repurposed GLP-1 Receptor Agonists to Novel Dual GLP-1/GIP Receptor Agonists as Potential Disease-Modifying Therapies.","authors":"Christian Hölscher","doi":"10.1007/s40263-025-01226-z","DOIUrl":"10.1007/s40263-025-01226-z","url":null,"abstract":"<p><p>Alzheimer's disease (AD) and Parkinson's disease (PD) are chronic neurodegenerative disorders with few effective drug treatments available. An underrated element of these diseases is that glucose uptake and energy utilization is much reduced in neurons. In the brains of patients, signaling of insulin, insulin-like growth factor 1, and other growth factors is downregulated early on. This leads to reduced glucose utilization and impaired mitochondrial function. In an attempt to compensate for the loss, other pathways are upregulated, e.g., the increased use of ketones produced from fatty acids by astrocytes that are shuttled to neurons. In addition, amino acids are increasingly used to generate energy. Despite this, neurons generate less and less energy over time, leading to impaired synaptic activity, reduced cell repair, mitogenesis, autophagy, the accumulation of misfolded proteins, and finally, to cell death. At the same time, the chronic inflammation response in the brain that is part of these diseases continues to damage neurons. Glucagon-like peptide 1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) are peptide hormones and growth factors that have shown neuroprotective effects in animal studies and in clinical trials. GLP-1 and GIP receptor agonists were able to reduce inflammation while normalizing growth factor signaling and energy utilization in the brain. Insulin signaling was improved and energy utilization, glucose uptake, mitogenesis, and mitochondrial functionality was brought back to physiological levels. In addition, the chronic inflammation response and the levels of proinflammatory cytokines in the brain were much reduced. Clinical trials testing GLP-1 receptor agonists in patients with AD or PD have been conducted and have shown first successes, serving as proof of concept that activating GLP-1 receptor is a sensible strategy to treat AD/PD. A phase II study testing liraglutide in patients with AD showed first improvements, and two phase II trials testing exendin-4 (exenatide, Bydureon<sup>®</sup>) or lixisenatide showed improvements in patients with PD. A recent phase III trial testing exendin-4 did not show an improvement, which may be linked to the lack of insulin desensitization in the study participants. Semaglutide (Rybelsus<sup>®</sup>; Wegovy<sup>®</sup>; Ozempic<sup>®</sup>) is currently in two phase III trials for AD. Current drugs that are on the market have a long half-life in the blood and do not readily cross the blood-brain barrier (BBB). Newer dual GLP-1/GIP receptor agonists have been developed that can more easily cross the BBB and that show improved protection in animal models of AD and PD. Therefore, GLP-1 and GIP receptor agonists that can cross the BBB show promise as treatments for chronic neurodegenerative disorders.</p>","PeriodicalId":10508,"journal":{"name":"CNS drugs","volume":" ","pages":"1201-1220"},"PeriodicalIF":7.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12602575/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145039313","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-08-30DOI: 10.1007/s40263-025-01221-4
Bénédicte Nobile, Erika Nogue, M C Picot, Philippe Courtet, Chouki Chenaf, Nicolas Authier, Emilie Olié
Background and objectives: The rising prescription rates and opioid-related harms in France highlight the need for local data. Evaluating this association may help identify vulnerable subgroups and guide safer prescribing practices. This study aimed to assess the association between opioid analgesic dispensation and the risk of suicide attempt in the French population. Secondary objectives included evaluation of a dose-response relationship and examination of the potential additive effects of co-prescriptions with benzodiazepines or gabapentinoids.
Methods: We conducted a nationwide, population-based case-crossover study using data from the French National Health Insurance Database (Système National des Données de Santé, SNDS), covering 98.8% of the French population. Adults aged 18 years or older who were hospitalized for a first suicide attempt between 1 January 2013 and 31 December 2020, and who had received at least one opioid analgesic dispensation in the preceding year (excluding buprenorphine and methadone) were included. Opioid analgesic exposure during the 84 days before the attempt was compared with three earlier 84-day control periods.
Results: Among 158,400 patients (mean age 47.0 years; 64.0% women), opioid analgesic dispensation was associated with a higher risk of suicide attempt (odds ratio [OR] = 1.26; 95% confidence interval 1.25-1.28). The risk was greater for strong opioid analgesics (OR = 1.73) and higher morphine-equivalent doses. Co-prescription with benzodiazepines or gabapentinoids further increased risk.
Conclusions: Opioid analgesic use, especially at higher doses or in combination with benzodiazepines or gabapentinoids, was associated with an increased risk of suicide attempt. Clinical vigilance is warranted when prescribing these medications.
Trial registration: NCT04211077, registered 3 January 2020 (retrospectively registered).
背景和目标:法国不断上升的处方率和阿片类药物相关危害突出了对当地数据的需求。评估这种关联可能有助于确定易受伤害的亚群体,并指导更安全的处方实践。本研究旨在评估阿片类镇痛药分配与法国人群自杀企图风险之间的关系。次要目的包括评估剂量-反应关系和检查与苯二氮卓类药物或加巴喷丁类药物合用处方的潜在附加效应。方法:我们使用法国国家健康保险数据库(system National des donnsam, SNDS)的数据进行了一项全国性的、基于人群的病例交叉研究,覆盖了98.8%的法国人口。在2013年1月1日至2020年12月31日期间因首次自杀未遂住院并在前一年接受过至少一种阿片类镇痛药分配(不包括丁丙诺啡和美沙酮)的18岁或以上成年人被纳入研究对象。在尝试前84天的阿片类镇痛药物暴露与早期的三个84天对照期进行比较。结果:在158,400例患者(平均年龄47.0岁,64.0%为女性)中,阿片类镇痛药配药与自杀企图风险较高相关(优势比[OR] = 1.26; 95%可信区间1.25-1.28)。强阿片类镇痛药(OR = 1.73)和高吗啡当量剂量的风险更大。与苯二氮卓类药物或加巴喷丁类药物合用会进一步增加风险。结论:阿片类镇痛药的使用,特别是高剂量或与苯二氮卓类或加巴喷丁类药物联合使用,与自杀企图的风险增加有关。在处方这些药物时,临床警惕是有必要的。试验注册:NCT04211077,于2020年1月3日注册(回顾性注册)。
{"title":"Association Between Opioid Analgesics and Suicide Attempts: A Nationwide French Case-Crossover Study.","authors":"Bénédicte Nobile, Erika Nogue, M C Picot, Philippe Courtet, Chouki Chenaf, Nicolas Authier, Emilie Olié","doi":"10.1007/s40263-025-01221-4","DOIUrl":"10.1007/s40263-025-01221-4","url":null,"abstract":"<p><strong>Background and objectives: </strong>The rising prescription rates and opioid-related harms in France highlight the need for local data. Evaluating this association may help identify vulnerable subgroups and guide safer prescribing practices. This study aimed to assess the association between opioid analgesic dispensation and the risk of suicide attempt in the French population. Secondary objectives included evaluation of a dose-response relationship and examination of the potential additive effects of co-prescriptions with benzodiazepines or gabapentinoids.</p><p><strong>Methods: </strong>We conducted a nationwide, population-based case-crossover study using data from the French National Health Insurance Database (Système National des Données de Santé, SNDS), covering 98.8% of the French population. Adults aged 18 years or older who were hospitalized for a first suicide attempt between 1 January 2013 and 31 December 2020, and who had received at least one opioid analgesic dispensation in the preceding year (excluding buprenorphine and methadone) were included. Opioid analgesic exposure during the 84 days before the attempt was compared with three earlier 84-day control periods.</p><p><strong>Results: </strong>Among 158,400 patients (mean age 47.0 years; 64.0% women), opioid analgesic dispensation was associated with a higher risk of suicide attempt (odds ratio [OR] = 1.26; 95% confidence interval 1.25-1.28). The risk was greater for strong opioid analgesics (OR = 1.73) and higher morphine-equivalent doses. Co-prescription with benzodiazepines or gabapentinoids further increased risk.</p><p><strong>Conclusions: </strong>Opioid analgesic use, especially at higher doses or in combination with benzodiazepines or gabapentinoids, was associated with an increased risk of suicide attempt. Clinical vigilance is warranted when prescribing these medications.</p><p><strong>Trial registration: </strong>NCT04211077, registered 3 January 2020 (retrospectively registered).</p>","PeriodicalId":10508,"journal":{"name":"CNS drugs","volume":" ","pages":"1331-1340"},"PeriodicalIF":7.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"144945487","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-10-10DOI: 10.1007/s40263-025-01235-y
Luigi Francesco Iannone, Alberto Boccalini, Flavia Lo Castro, Daria Brovia, Marina Romozzi, Fabrizio Vernieri, Claudia Altamura, Simona Guérzoni
<p><strong>Background: </strong>Migraine is often associated with impaired sleep quality, including insomnia, fragmented sleep, and circadian rhythm disturbances. These factors can exacerbate migraine severity and chronification. Calcitonin gene-related peptide (CGRP), a key player in migraine pathophysiology, also influences sleep regulation. While CGRP monoclonal antibodies have shown mixed effects on sleep, no study to date has evaluated the impact of gepants on sleep quality. This study assessed whether atogepant, recently approved for migraine prevention, affects sleep quality and sleep-related adverse events in real-world settings.</p><p><strong>Methods: </strong>We conducted a prospective, observational, open-label, single-center study. All received atogepant 60 mg/day up to 12 weeks. Adults (≥ 18 years) with migraine (with/without aura or chronic migraine) experiencing ≥ 4 monthly migraine days were enrolled. Inclusion required ≥ 1 month of headache diaries and stable preventive or sleep treatments for ≥ 3 months. Patients were accepted regardless of prior preventive failures. Exclusion criteria were unstable treatments, recent sleep-impacting disease, and pregnancy. Sleep quality was assessed using five validated questionnaires (Pittsburgh Sleep Quality Index [PSQI], Athens Insomnia Scale [AIS], Bergen, Epworth Sleepiness Scale [ESS], Insomnia Severity Index [ISI]) at baseline and at follow-up. Migraine frequency, disability (Migraine Disability Assessment [MIDAS], Headache Impact Test [HIT-6]), allodynia (Allodynia Symptom Checklist [ASC-12]), acute medication use, and adverse events (AEs) were also recorded. Pre-post differences were assessed with Wilcoxon and McNemar's tests, while linear mixed-effects models were applied to evaluate the impact of clinical factors (response status, psychiatric comorbidities, prior anti-CGRP failures) on PSQI outcomes, with model fit estimated via REML and pseudo-R<sup>2</sup>.</p><p><strong>Results: </strong>The study population included 43 participants (93.0% female, mean age of 51.6 [IQR 48.4-54.8] years, mean age at disease onset of 18.9 [16.0-21.7] years); 30 (69.8%) participants had chronic migraine, and among them, 23 (76.7%) had a concomitant diagnosis of medication overuse headache. Atogepant significantly improved sleep quality with PSQI scores decreased from 9.6 to 8.2 (p = 0.002) and improvements in AIS (p = 0.014) and Bergen scores (p = 0.046). Sleep duration was the only PSQI subdomain with a statistically significant change. No differences were found in ESS or ISI scores. Notably, no patients reported sleep-related AEs such as somnolence, nightmares, or vivid dreams. Psychiatric comorbidities were associated with poorer baseline sleep but did not influence the magnitude of improvement. Prior anti-CGRP failure predicted a lesser sleep benefit. Finally, migraine burden improved across all evaluated migraine-related variables. Only two patients discontinued treatment.</p><p><strong>Conclusi
{"title":"Effect of Atogepant on Sleep Quality and Sleep-Related Adverse Events in Adult Patients with Migraine: A Prospective Observational 12-Week Study.","authors":"Luigi Francesco Iannone, Alberto Boccalini, Flavia Lo Castro, Daria Brovia, Marina Romozzi, Fabrizio Vernieri, Claudia Altamura, Simona Guérzoni","doi":"10.1007/s40263-025-01235-y","DOIUrl":"10.1007/s40263-025-01235-y","url":null,"abstract":"<p><strong>Background: </strong>Migraine is often associated with impaired sleep quality, including insomnia, fragmented sleep, and circadian rhythm disturbances. These factors can exacerbate migraine severity and chronification. Calcitonin gene-related peptide (CGRP), a key player in migraine pathophysiology, also influences sleep regulation. While CGRP monoclonal antibodies have shown mixed effects on sleep, no study to date has evaluated the impact of gepants on sleep quality. This study assessed whether atogepant, recently approved for migraine prevention, affects sleep quality and sleep-related adverse events in real-world settings.</p><p><strong>Methods: </strong>We conducted a prospective, observational, open-label, single-center study. All received atogepant 60 mg/day up to 12 weeks. Adults (≥ 18 years) with migraine (with/without aura or chronic migraine) experiencing ≥ 4 monthly migraine days were enrolled. Inclusion required ≥ 1 month of headache diaries and stable preventive or sleep treatments for ≥ 3 months. Patients were accepted regardless of prior preventive failures. Exclusion criteria were unstable treatments, recent sleep-impacting disease, and pregnancy. Sleep quality was assessed using five validated questionnaires (Pittsburgh Sleep Quality Index [PSQI], Athens Insomnia Scale [AIS], Bergen, Epworth Sleepiness Scale [ESS], Insomnia Severity Index [ISI]) at baseline and at follow-up. Migraine frequency, disability (Migraine Disability Assessment [MIDAS], Headache Impact Test [HIT-6]), allodynia (Allodynia Symptom Checklist [ASC-12]), acute medication use, and adverse events (AEs) were also recorded. Pre-post differences were assessed with Wilcoxon and McNemar's tests, while linear mixed-effects models were applied to evaluate the impact of clinical factors (response status, psychiatric comorbidities, prior anti-CGRP failures) on PSQI outcomes, with model fit estimated via REML and pseudo-R<sup>2</sup>.</p><p><strong>Results: </strong>The study population included 43 participants (93.0% female, mean age of 51.6 [IQR 48.4-54.8] years, mean age at disease onset of 18.9 [16.0-21.7] years); 30 (69.8%) participants had chronic migraine, and among them, 23 (76.7%) had a concomitant diagnosis of medication overuse headache. Atogepant significantly improved sleep quality with PSQI scores decreased from 9.6 to 8.2 (p = 0.002) and improvements in AIS (p = 0.014) and Bergen scores (p = 0.046). Sleep duration was the only PSQI subdomain with a statistically significant change. No differences were found in ESS or ISI scores. Notably, no patients reported sleep-related AEs such as somnolence, nightmares, or vivid dreams. Psychiatric comorbidities were associated with poorer baseline sleep but did not influence the magnitude of improvement. Prior anti-CGRP failure predicted a lesser sleep benefit. Finally, migraine burden improved across all evaluated migraine-related variables. Only two patients discontinued treatment.</p><p><strong>Conclusi","PeriodicalId":10508,"journal":{"name":"CNS drugs","volume":" ","pages":"1341-1354"},"PeriodicalIF":7.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC12602620/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145274012","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01DOI: 10.1007/s40263-025-01251-y
Susan Abushakra, P Murali Doraiswamy, John A Hey, Duygu Tosun, Frederik Barkhof, Jerome Barakos, Jeffrey Petrella, J Patrick Kesslak, Aidan Power, Marwan Sabbagh, Anton Porsteinsson, Sharon Cohen, Serge Gauthier, Craig Ritchie, David Watson, Emer McSweeney, Merce Boada, Earvin Liang, Luc Bracoud, Rosalind McLaine, Susan Flint, Jean F Schaefer, Jeremy Yu, Margaret Bray, Suzanne Hendrix, Sam Dickson, Abe Durrant, Adem Albayrak, Martin Tolar
<p><p>Amyloid-plaque reduction is currently the only recognized surrogate outcome for Alzheimer's disease (AD) trials, allowing accelerated approval of plaque-clearing amyloid antibodies. However, plaque reduction does not facilitate the development of new non-plaque-clearing treatments. The hippocampus is among the first brain regions affected by AD pathology, exhibiting synaptic dysfunction and neurodegeneration that manifests as hippocampal atrophy and memory decline. We evaluated hippocampal volume (HV) as a potential surrogate outcome that can predict clinical benefit in disease-modification trials. Using published data from observational and interventional studies that examined both cognition and HV on volumetric magnetic resonance imaging (vMRI), we evaluated the cross-sectional correlations of HV to cognitive performance, the longitudinal correlations of HV atrophy to cognitive decline, HV sensitivity to drug effects, and the correlations between drug effects on HV atrophy and cognitive decline. We also examined the magnitude of HV protection that corresponds to meaningful clinical benefit. Analyses from 30 observational studies encompassing 13,187 individuals (2633 cognitively normal; 10,554 early AD) showed significant cross-sectional correlations between baseline HV and cognition, and longitudinal correlations between HV atrophy and cognitive decline over ≥ 1 year. The relationship of HV-cognitive drug effects was examined at the group level in nine placebo-controlled trials of five antiamyloid agents that evaluated HV in early AD trials of at least 18 months' duration. These trials included four amyloid antibodies (aducanumab, lecanemab, donanemab, and gantenerumab) and one oral anti-oligomer agent (valiltramiprosate). Individual-level HV-cognition relationships were examined in two valiltramiprosate studies, one of which included diffusion tensor imaging (DTI) providing microstructural correlates of HV drug effects and helping distinguish neuroprotection from brain edema. Across these anti-amyloid drug trials (total N ~10,000), there was a linear relationship between drug effects on slowing of cognitive decline and slowing of HV atrophy. Two anti-oligomer trials (valiltramiprosate) reported significant subject-level correlations between drug effects on HV and cognition over 18-24 months (r = -0.40 to -0.44, p < 0.005, N = 50/69), with significant correlations of drug effects on brain microstructure (decreased mean diffusivity) with both HV and cognitive benefits, supporting reduced neurodegeneration. The minimal HV preservation at the mild cognitive impairment (MCI) stage that is associated with clinical benefit is estimated to be ≥ 40 mm<sup>3</sup> or ≥ 10% of atrophy in the placebo arm over 18 months. Our findings demonstrate that hippocampal atrophy is an early indicator of cognitive decline in AD, linked to amyloid and tau-related neurodegeneration. HV on standardized vMRI is sensitive to anti-amyloid treatments, demonstrating s
{"title":"Hippocampal Atrophy on Magnetic Resonance Imaging as a Surrogate Marker for Clinical Benefit and Neurodegeneration in Early Symptomatic Alzheimer's Disease: Synthesis of Evidence from Observational and Interventional Trials.","authors":"Susan Abushakra, P Murali Doraiswamy, John A Hey, Duygu Tosun, Frederik Barkhof, Jerome Barakos, Jeffrey Petrella, J Patrick Kesslak, Aidan Power, Marwan Sabbagh, Anton Porsteinsson, Sharon Cohen, Serge Gauthier, Craig Ritchie, David Watson, Emer McSweeney, Merce Boada, Earvin Liang, Luc Bracoud, Rosalind McLaine, Susan Flint, Jean F Schaefer, Jeremy Yu, Margaret Bray, Suzanne Hendrix, Sam Dickson, Abe Durrant, Adem Albayrak, Martin Tolar","doi":"10.1007/s40263-025-01251-y","DOIUrl":"https://doi.org/10.1007/s40263-025-01251-y","url":null,"abstract":"<p><p>Amyloid-plaque reduction is currently the only recognized surrogate outcome for Alzheimer's disease (AD) trials, allowing accelerated approval of plaque-clearing amyloid antibodies. However, plaque reduction does not facilitate the development of new non-plaque-clearing treatments. The hippocampus is among the first brain regions affected by AD pathology, exhibiting synaptic dysfunction and neurodegeneration that manifests as hippocampal atrophy and memory decline. We evaluated hippocampal volume (HV) as a potential surrogate outcome that can predict clinical benefit in disease-modification trials. Using published data from observational and interventional studies that examined both cognition and HV on volumetric magnetic resonance imaging (vMRI), we evaluated the cross-sectional correlations of HV to cognitive performance, the longitudinal correlations of HV atrophy to cognitive decline, HV sensitivity to drug effects, and the correlations between drug effects on HV atrophy and cognitive decline. We also examined the magnitude of HV protection that corresponds to meaningful clinical benefit. Analyses from 30 observational studies encompassing 13,187 individuals (2633 cognitively normal; 10,554 early AD) showed significant cross-sectional correlations between baseline HV and cognition, and longitudinal correlations between HV atrophy and cognitive decline over ≥ 1 year. The relationship of HV-cognitive drug effects was examined at the group level in nine placebo-controlled trials of five antiamyloid agents that evaluated HV in early AD trials of at least 18 months' duration. These trials included four amyloid antibodies (aducanumab, lecanemab, donanemab, and gantenerumab) and one oral anti-oligomer agent (valiltramiprosate). Individual-level HV-cognition relationships were examined in two valiltramiprosate studies, one of which included diffusion tensor imaging (DTI) providing microstructural correlates of HV drug effects and helping distinguish neuroprotection from brain edema. Across these anti-amyloid drug trials (total N ~10,000), there was a linear relationship between drug effects on slowing of cognitive decline and slowing of HV atrophy. Two anti-oligomer trials (valiltramiprosate) reported significant subject-level correlations between drug effects on HV and cognition over 18-24 months (r = -0.40 to -0.44, p < 0.005, N = 50/69), with significant correlations of drug effects on brain microstructure (decreased mean diffusivity) with both HV and cognitive benefits, supporting reduced neurodegeneration. The minimal HV preservation at the mild cognitive impairment (MCI) stage that is associated with clinical benefit is estimated to be ≥ 40 mm<sup>3</sup> or ≥ 10% of atrophy in the placebo arm over 18 months. Our findings demonstrate that hippocampal atrophy is an early indicator of cognitive decline in AD, linked to amyloid and tau-related neurodegeneration. HV on standardized vMRI is sensitive to anti-amyloid treatments, demonstrating s","PeriodicalId":10508,"journal":{"name":"CNS drugs","volume":" ","pages":""},"PeriodicalIF":7.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145647122","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
Pub Date : 2025-12-01Epub Date: 2025-09-25DOI: 10.1007/s40263-025-01230-3
Yiwei Huang, Xinyuan Yu, Changxin Li
Sodium-glucose cotransporter 2 inhibitors (SGLT2i), a cornerstone therapy for type 2 diabetes mellitus (T2DM) and heart failure, are emerging as a promising therapeutic class for the management of ischemic stroke (IS). Given that T2DM is a significant risk factor for IS, understanding the potential neuroprotective role of SGLT2i is of paramount clinical importance. This review provides a systematic and logically structured synthesis of the current evidence, beginning with foundational preclinical studies in animal models that consistently demonstrate a reduction in infarct volume and improved neurological outcomes. We then transition to the extensive clinical evidence, primarily from large-scale real-world observational studies, that has confirmed a significant reduction in stroke risk, particularly in high-risk T2DM populations. The strengths and limitations of this evidence base are critically appraised, highlighting the robustness of the findings while acknowledging the predominantly observational nature of the data and the lack of stroke-specific primary endpoints in major trials. The neuroprotective benefits of SGLT2i appear to be multifactorial; this review delves into the potential mechanisms, emphasizing a foundational, glucose-dependent pathway of ameliorating hyperglycemia-induced neurotoxicity, which is complemented by a suite of pleiotropic, glucose-independent effects, including the induction of mild ketosis, attenuation of neuroinflammation, and preservation of the neurovascular unit. Finally, we address the key clinical challenges to their application, such as the management of euglycemic ketoacidosis, and outline crucial directions for future research, underscoring the need for dedicated randomized trials.
{"title":"Sodium-Glucose Cotransporter 2 Inhibitors: An Emerging Therapeutic Approach for Ischemic Stroke Management.","authors":"Yiwei Huang, Xinyuan Yu, Changxin Li","doi":"10.1007/s40263-025-01230-3","DOIUrl":"10.1007/s40263-025-01230-3","url":null,"abstract":"<p><p>Sodium-glucose cotransporter 2 inhibitors (SGLT2i), a cornerstone therapy for type 2 diabetes mellitus (T2DM) and heart failure, are emerging as a promising therapeutic class for the management of ischemic stroke (IS). Given that T2DM is a significant risk factor for IS, understanding the potential neuroprotective role of SGLT2i is of paramount clinical importance. This review provides a systematic and logically structured synthesis of the current evidence, beginning with foundational preclinical studies in animal models that consistently demonstrate a reduction in infarct volume and improved neurological outcomes. We then transition to the extensive clinical evidence, primarily from large-scale real-world observational studies, that has confirmed a significant reduction in stroke risk, particularly in high-risk T2DM populations. The strengths and limitations of this evidence base are critically appraised, highlighting the robustness of the findings while acknowledging the predominantly observational nature of the data and the lack of stroke-specific primary endpoints in major trials. The neuroprotective benefits of SGLT2i appear to be multifactorial; this review delves into the potential mechanisms, emphasizing a foundational, glucose-dependent pathway of ameliorating hyperglycemia-induced neurotoxicity, which is complemented by a suite of pleiotropic, glucose-independent effects, including the induction of mild ketosis, attenuation of neuroinflammation, and preservation of the neurovascular unit. Finally, we address the key clinical challenges to their application, such as the management of euglycemic ketoacidosis, and outline crucial directions for future research, underscoring the need for dedicated randomized trials.</p>","PeriodicalId":10508,"journal":{"name":"CNS drugs","volume":" ","pages":"1273-1295"},"PeriodicalIF":7.4,"publicationDate":"2025-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"145136674","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}