1
|
Yang H, Byun MS, Ha NY, Yang J, Park SY, Park JE, Yi D, Chang YT, Jung WS, Kim JY, Kim J, Lee DY, Bae H. A preclinical and phase I clinical study of ex vivo-expanded amyloid beta-specific human regulatory T cells in Alzheimer's disease. Biomed Pharmacother 2024; 181:117721. [PMID: 39626378 DOI: 10.1016/j.biopha.2024.117721] [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: 08/09/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 12/21/2024] Open
Abstract
INTRODUCTION Despite advancements in adoptive regulatory T cell (Treg) therapy, its application in Alzheimer's disease (AD) remains constrained by challenges in ex vivo Treg selection and expansion with antigen specificity. Our previous findings demonstrated the bystander suppressive immunomodulatory mechanism of ex vivo expanded amyloid β-specific mouse Tregs in AD models, prompting inquiry into the efficacy of ex vivo expanded human Tregs in AD. METHODS We developed an effective ex vivo expansion method for manufacturing amyloid β-specific human Tregs (Aβ-hTreg) and evaluated their safety and efficacy in 3xTg mouse models of AD and a phase 1 clinical trial with six AD patients. The phenotype of Aβ-hTreg was analyzed using single-cell transcriptomics. The clinical trial involved intravenous administration of Aβ-hTreg, with three patients receiving a low dose and three receiving a high dose. Exploratory assessments of effectiveness, including cognitive tasks and functional evaluations, were conducted ninety days post-treatment. RESULTS Behavioral spatial learning and memory impairment, neuroinflammatory and amyloid pathology were dramatically ameliorated by single intrathecal administration of ex vivo expanded Aβ-hTreg to 3xTg AD mice. Single cell transcriptomics analysis revealed alterations in five key genes within a cluster of Tregs under antigen-specific manufacturing conditions. In the clinical trial with six AD patients, dose-limiting toxicity was experienced by none of the participants within five days of receiving GMP-grade Aβ-hTreg (VT301), indicating its good tolerability. Although exploratory assessments of effectiveness did not reach statistically significant values among the groups, these findings offer valuable insights for AD treatment and management, guiding the planning of the next phase of clinical trials. DISCUSSION This study suggests that hTregs may modulate Alzheimer's disease pathology by suppressing neuroinflammation, while VT301 shows promise as a safe treatment option. However, further research is necessary to confirm its clinical efficacy and optimize treatment strategies. TRIAL REGISTRATION Title: A Study of Possibility of Using Regulatory T Cells (VT301) for Treatment of Alzheimer's Disease, ClinicalTrials.gov NCT05016427, Study approval date: Ministry of Food and Drug Safety of the Republic of Korea (MFDS) - August 31st, 2020, Institutional Review Board (IRB) of Seoul National University Hospital, Republic of Korea - September 29th, 2020, The date of first patient enrollment: December 7th, 2020. https://clinicaltrials.gov/study/NCT05016427.
Collapse
Affiliation(s)
- Hyejin Yang
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Min Soo Byun
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Psychiatry, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Na-Yeon Ha
- Department of Digestive Diseases, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Division of Digestive Diseases, Department of Korean Internal Medicine, Kyung Hee University Korean Medicine Hospital, Seoul 02447, Republic of Korea
| | - Juwon Yang
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seon-Young Park
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jee Eun Park
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Psychiatry, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul 03080, Republic of Korea
| | - Dahyun Yi
- Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul 03080, Republic of Korea
| | - Young-Tae Chang
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Republic of Korea
| | - Woo Sang Jung
- Department of Cardiovascular and Neurologic Diseases, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Jae Yoon Kim
- Institute of Life Science & Biotechnology, VT Bio.Co., Ltd., Seoul 06185, Republic of Korea
| | - Jinsung Kim
- Department of Digestive Diseases, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea; Division of Digestive Diseases, Department of Korean Internal Medicine, Kyung Hee University Korean Medicine Hospital, Seoul 02447, Republic of Korea
| | - Dong Young Lee
- Department of Neuropsychiatry, Seoul National University Hospital, Seoul 03080, Republic of Korea; Department of Psychiatry, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; Institute of Human Behavioral Medicine, Medical Research Center Seoul National University, Seoul 03080, Republic of Korea.
| | - Hyunsu Bae
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
2
|
Moyer H, Mellett R, Vigneault K, McKeown M, Karlawish J, Augustine E, Schneider L, Kimmelman J. Prevalence and Impact of Bypassing or Overriding Phase 2 Trials in Neurologic Drug Development. Neurology 2024; 103:e209533. [PMID: 38833654 PMCID: PMC11226307 DOI: 10.1212/wnl.0000000000209533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 04/09/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Pivotal trials for neurologic drugs in clinical development are often initiated without a phase 2 trial ("bypass") or despite a negative phase 2 efficacy result ("override"). Such practices may degrade the risk/benefit ratio of phase 3 trials. The aim of this study is to estimate the proportion of phase 3 trials for 10 neurologic diseases started without a positive phase 2 trial, to identify factors associated with this practice, and to investigate any association with unfavorable phase 3 trial outcomes. METHODS We searched ClinicalTrials.gov for phase 3 trials completed during 2011-2021, with at least 1 research site in the United States, Canada, the European Union, the United Kingdom, or Australia, and investigating drugs or biologics for treatment of 10 neurologic conditions. Our primary objective was to assess the prevalence of phase 2 bypass/override by searching for preceding phase 2 trials. We used Fisher exact tests to determine whether phase 3 trial characteristics and trial results were associated with phase 2 bypass/override. RESULTS Of the 1,188 phase 3 trials captured in our search, 113 met eligibility for inclusion. Of these, 46% were not preceded by a phase 2 trial that was positive on an efficacy endpoint (31% bypassed and 15% overrode phase 2 trial). Phase 2 bypass/override was not associated with industry funding (77% vs 89%, 95% CI 0.75-7.55, p = 0.13) or testing already approved interventions (23% vs 15%, 95% CI 0.60-5.14, p = 0.33). Overall, phase 3 trials based on phase 2 bypassed/override were statistically significantly less likely to be positive on their primary outcome (31% vs 57%, respectively, 95% CI 1.21-6.92, p = 0.01). This effect disappeared when indications characterized by nearly universal positive or negative results were excluded. Trials that bypassed/overrode phase 2 trials were not statistically significantly more likely to be terminated early because of safety or futility (29% vs 15%, respectively, 95% CI 0.15-1.18, p = 0.11) and did not show increased risk of adverse events in experimental arms (RR = 1.46, 95% CI 1.19-1.79, vs RR = 1.36, 95% CI 1.10-1.69, respectively, p = 0.65). DISCUSSION Almost half of the neurologic disease phase 3 trials were initiated without the support of a positive phase 2 trial. Although our analysis does not establish harm with bypass/override, its prevalence and the scientific rationale for phase 2 trial testing favor development of criteria defining when phase 2 bypass/override is justified.
Collapse
Affiliation(s)
- Hannah Moyer
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Robyn Mellett
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Karine Vigneault
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Maya McKeown
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Jason Karlawish
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Erika Augustine
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Lon Schneider
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| | - Jonathan Kimmelman
- From the Department of Equity, Ethics and Policy (H.M., R.M., K.V., M.M., J. Kimmelman), McGill University, Montreal, Quebec, Canada; Penn Memory Center (J. Karlawish), University of Pennsylvania, Philadelphia; Clinical Trials Unit (E.A.), Kennedy Krieger Institute, Baltimore, MD; Departments of Psychiatry and Behavioral Sciences and Neurology, Keck School of Medicine (L.S.), and L. Davis School of Gerontology (L.S.), University of Southern California, Los Angeles
| |
Collapse
|
3
|
Rhodus EK, Barber J, Abner EL, Bardach SH, Gibson A, Jicha GA. Comparison of behaviors characteristic of autism spectrum disorder behaviors and behavioral and psychiatric symptoms of dementia. Aging Ment Health 2022; 26:586-594. [PMID: 33222510 PMCID: PMC8212388 DOI: 10.1080/13607863.2020.1849025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Similarities exist in behavioral expression of autism spectrum disorder (ASD) and Alzheimer's disease and related dementias (ADRD). The purpose of this study was to assess presence of behavioral and psychiatric symptoms of dementia (BPSD) and ASD-like behaviors in adults with ADRD. METHODS Using a cross-sectional design, data from University of Kentucky Alzheimer's Disease Center participant cohort were used. Hierarchical linear regression was used to assess (1) the relationship between ASD-like behaviors (measured by the Gilliam Autism Rating Scale-Second Edition, GARS-2) and BPSD measured by the Neuropsychiatric Inventory (NPI), and (2) the relationship between ASD-like behaviors and dementia severity (measured by the Clinical Dementia Rating [CDR] sum of boxes), when controlling for BPSD. RESULTS Complete data were available for 142 participants. Using α of 0.05, analyses identified ASD behaviors were significantly associated with BPSD severity ratings (r = 0.47; p < 0.001) and dementia severity (r = 0.46; p < 0.001). GARS-2 explained 6.1% (p < 0.001) of variance in CDR sum of boxes when controlling for NPI and other covariates. DISCUSSION There is significant overlap in behaviors characteristic of ASD and BPSD as assessed by the NPI and GARS-2, despite the use of these instruments in disparate developmental vs. aging settings. ASD behaviors appear to not be solely present in early childhood as a manifestation of ASD but are also present in older adults with neurodegenerative cognitive impairment. Such associations warrant additional research into causation, assessment, and behavioral interventions to further enable new therapeutic approaches targeting ASD behaviors across the lifespan.
Collapse
Affiliation(s)
| | - Justin Barber
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY
| | - Erin L. Abner
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY,Department of Epidemiology, University of Kentucky, Lexington, KY
| | - Shoshana H. Bardach
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY,Graduate Center for Gerontology, University of Kentucky, Lexington, KY
| | - Allison Gibson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY,College of Social Work, University of Kentucky, Lexington, KY
| | - Gregory A. Jicha
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY,Department of Behavioral Science, University of Kentucky, Lexington, KY,Department of Neurology, University of Kentucky, Lexington, KY
| |
Collapse
|
4
|
Independent replication of advanced brain age in mild cognitive impairment and dementia: detection of future cognitive dysfunction. Mol Psychiatry 2022; 27:5235-5243. [PMID: 35974140 PMCID: PMC9763106 DOI: 10.1038/s41380-022-01728-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 07/18/2022] [Accepted: 07/26/2022] [Indexed: 01/14/2023]
Abstract
We previously developed a novel machine-learning-based brain age model that was sensitive to amyloid. We aimed to independently validate it and to demonstrate its utility using independent clinical data. We recruited 650 participants from South Korean memory clinics to undergo magnetic resonance imaging and clinical assessments. We employed a pretrained brain age model that used data from an independent set of largely Caucasian individuals (n = 757) who had no or relatively low levels of amyloid as confirmed by positron emission tomography (PET). We investigated the association between brain age residual and cognitive decline. We found that our pretrained brain age model was able to reliably estimate brain age (mean absolute error = 5.68 years, r(650) = 0.47, age range = 49-89 year) in the sample with 71 participants with subjective cognitive decline (SCD), 375 with mild cognitive impairment (MCI), and 204 with dementia. Greater brain age was associated with greater amyloid and worse cognitive function [Odds Ratio, (95% Confidence Interval {CI}): 1.28 (1.06-1.55), p = 0.030 for amyloid PET positivity; 2.52 (1.76-3.61), p < 0.001 for dementia]. Baseline brain age residual was predictive of future cognitive worsening even after adjusting for apolipoprotein E e4 and amyloid status [Hazard Ratio, (95% CI): 1.94 (1.33-2.81), p = 0.001 for total 336 follow-up sample; 2.31 (1.44-3.71), p = 0.001 for 284 subsample with baseline Clinical Dementia Rating ≤ 0.5; 2.40 (1.43-4.03), p = 0.001 for 240 subsample with baseline SCD or MCI]. In independent data set, these results replicate our previous findings using this model, which was able to delineate significant differences in brain age according to the diagnostic stages of dementia as well as amyloid deposition status. Brain age models may offer benefits in discriminating and tracking cognitive impairment in older adults.
Collapse
|
5
|
Nuño MM, Grill JD, Gillen DL. On the design of early-phase Alzheimer's disease clinical trials with cerebrospinal fluid tau outcomes. Clin Trials 2021; 18:714-723. [PMID: 34325548 PMCID: PMC8595611 DOI: 10.1177/17407745211034497] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND/AIMS The focus of Alzheimer's disease studies has shifted to earlier disease stages, including mild cognitive impairment. Biomarker inclusion criteria are often incorporated into mild cognitive impairment clinical trials to identify individuals with "prodromal Alzheimer's disease" to ensure appropriate drug targets and enrich for participants likely to develop Alzheimer's disease dementia. The use of these eligibility criteria may affect study power. METHODS We investigated outcome variability and study power in the setting of proof-of-concept prodromal Alzheimer's disease trials that incorporate cerebrospinal fluid levels of total tau (t-tau) and phosphorylated (p-tau) as primary outcomes and how differing biomarker inclusion criteria affect power. We used data from the Alzheimer's Disease Neuroimaging Initiative to model trial scenarios and to estimate the variance and within-subject correlation of total and phosphorylated tau. These estimates were then used to investigate the differences in study power for trials considering these two surrogate outcomes. RESULTS Patient characteristics were similar for all eligibility criteria. The lowest outcome variance and highest within-subject correlation were obtained when phosphorylated tau was used as an eligibility criterion, compared to amyloid beta or total tau, regardless of whether total tau or phosphorylated tau were used as primary outcomes. Power increased when eligibility criteria were broadened to allow for enrollment of subjects with either low amyloid beta or high phosphorylated tau. CONCLUSION Specific biomarker inclusion criteria may impact statistical power in trials using total tau or phosphorylated tau as the primary outcome. In concert with other important considerations such as treatment target and population of clinical interest, these results may have implications to the integrity and efficiency of prodromal Alzheimer's disease trial designs.
Collapse
Affiliation(s)
- Michelle M. Nuño
- Children’s Oncology Group, Monrovia, CA, USA
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA, USA
| | - Joshua D. Grill
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA
| | - Daniel L. Gillen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
- Department of Statistics, University of California, Irvine, Irvine, CA, USA
| | | |
Collapse
|
6
|
Yao L, Li M, Sun S, Xu M, Yu S, Zhang Z, Zhang L, Zheng H, Zhong Z, Ma S, Huang H, Wang H. Multimodal brain imaging effect of "Adjust Zang-fu and Arouse Spirit" electroacupuncture on diabetic cognitive impairment: study protocol of a randomized, sham-controlled pilot trial. Trials 2021; 22:847. [PMID: 34823569 PMCID: PMC8620192 DOI: 10.1186/s13063-021-05842-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 11/18/2021] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Diabetic cognitive impairment (DCI) is a serious chronic complication caused by diabetes. The pathogenesis of DCI is complex, but brain nerve injury and brain nerve cell apoptosis are important pathological changes. Multimodal brain imaging is one of the most important techniques to study the neural mechanism of the brain. For the clinical treatment of DCI, there is no effective targeted Western medicine and a lack of clear drug intervention methods. Therefore, there is an urgent need to find effective complementary and alternative methods and clarify their mechanism. This research seeks to explore the multimodal brain imaging effect of "Adjust Zang-fu and Arouse Spirit" electroacupuncture for DCI. METHODS This clinical research will be a randomized, sham-controlled pilot trial. Eligible participants will be randomly assigned to the intervention group (n = 60) and the control group (n = 30). The intervention group will be divided into the "Adjust Zang-fu and Arouse Spirit" electroacupuncture group (n = 30) and sham electroacupuncture group (n = 30). All participants will continue to receive routine hypoglycemic therapy. The treatment period is the same in both groups. The primary outcomes include functional magnetic resonance imaging (fMRI), magnetic resonance spectroscopy (MRS), Montreal Cognitive Assessment Scale (MoCA), and Clinical Dementia Rating (CDR). The secondary outcomes include blood glucose and blood lipid tests, Instrumental Activities of Daily Living Scale (IADL), Hachinski Ischemic Scale (HIS), Self-Rating Anxiety Scale (SAS), and Self-Rating Depression Scale (SDS). Outcomes will be assessed at baseline and before and after treatment, and adverse events will be examined. Inter- and intragroup analyses will be performed. DISCUSSION This randomized controlled study, combined with multimodal brain imaging techniques and a clinical evaluation scale, was designed to explore the mechanism of "Adjust Zang-fu and Arouse Spirit" electroacupuncture in improving the central nervous system in DCI. TRIAL REGISTRATION Chinese Clinical Trial Registration ChiCTR2000040268 . Registered on 26 November 2020.
Collapse
Affiliation(s)
- Lin Yao
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China
| | - Mengyuan Li
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China
| | - Shunan Sun
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China
| | - Ming Xu
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China
| | - Shuo Yu
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China
| | - Ziyang Zhang
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China
| | - Liying Zhang
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China
| | - Haizhu Zheng
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China
| | - Zhen Zhong
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China
| | - Shiqi Ma
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China
| | - Haipeng Huang
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China
| | - Hongfeng Wang
- Changchun University of Chinese Medicine, No.1035 Boshuo Road, Nanguan District, Changchun, Jilin, 130117, China.
| |
Collapse
|
7
|
Li QS, Vasanthakumar A, Davis JW, Idler KB, Nho K, Waring JF, Saykin AJ. Association of peripheral blood DNA methylation level with Alzheimer's disease progression. Clin Epigenetics 2021; 13:191. [PMID: 34654479 PMCID: PMC8518178 DOI: 10.1186/s13148-021-01179-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 09/29/2021] [Indexed: 12/11/2022] Open
Abstract
Background Identifying biomarkers associated with Alzheimer’s disease (AD) progression may enable patient enrichment and improve clinical trial designs. Epigenome-wide association studies have revealed correlations between DNA methylation at cytosine-phosphate-guanine (CpG) sites and AD pathology and diagnosis. Here, we report relationships between peripheral blood DNA methylation profiles measured using Infinium® MethylationEPIC BeadChip and AD progression in participants from the Alzheimer’s Disease Neuroimaging Initiative (ADNI) cohort. Results The rate of cognitive decline from initial DNA sampling visit to subsequent visits was estimated by the slopes of the modified Preclinical Alzheimer Cognitive Composite (mPACC; mPACCdigit and mPACCtrailsB) and Clinical Dementia Rating Scale Sum of Boxes (CDR-SB) plots using robust linear regression in cognitively normal (CN) participants and patients with mild cognitive impairment (MCI), respectively. In addition, diagnosis conversion status was assessed using a dichotomized endpoint. Two CpG sites were significantly associated with the slope of mPACC in CN participants (P < 5.79 × 10−8 [Bonferroni correction threshold]); cg00386386 was associated with the slope of mPACCdigit, and cg09422696 annotated to RP11-661A12.5 was associated with the slope of CDR-SB. No significant CpG sites associated with diagnosis conversion status were identified. Genes involved in cognition and learning were enriched. A total of 19, 13, and 5 differentially methylated regions (DMRs) associated with the slopes of mPACCtrailsB, mPACCdigit, and CDR-SB, respectively, were identified by both comb-p and DMRcate algorithms; these included DMRs annotated to HOXA4. Furthermore, 5 and 19 DMRs were associated with conversion status in CN and MCI participants, respectively. The most significant DMR was annotated to the AD-associated gene PM20D1 (chr1: 205,818,956 to 205,820,014 [13 probes], Sidak-corrected P = 7.74 × 10−24), which was associated with both the slope of CDR-SB and the MCI conversion status. Conclusion Candidate CpG sites and regions in peripheral blood were identified as associated with the rate of cognitive decline in participants in the ADNI cohort. While we did not identify a single CpG site with sufficient clinical utility to be used by itself due to the observed effect size, a biosignature composed of DNA methylation changes may have utility as a prognostic biomarker for AD progression. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01179-2.
Collapse
Affiliation(s)
- Qingqin S Li
- Neuroscience, Janssen Research and Development, LLC, 1125 Trenton-Harbourton Road, Titusville, NJ, 08560, USA.
| | | | - Justin W Davis
- Genomics Research Center, AbbVie, North Chicago, IL, USA
| | | | - Kwangsik Nho
- Indiana Alzheimer's Disease Research Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Andrew J Saykin
- Indiana Alzheimer's Disease Research Center, Department of Radiology and Imaging Sciences, Indiana University School of Medicine, Indianapolis, IN, USA
| | | |
Collapse
|
8
|
Cummings J, Bauzon J, Lee G. Who funds Alzheimer's disease drug development? ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2021; 7:e12185. [PMID: 34095442 PMCID: PMC8145442 DOI: 10.1002/trc2.12185] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Despite the increase in Alzheimer's disease (AD) cases in the United States, no new treatments have been approved in the United States since 2003. The costs associated with drug development programs are high and serve as a significant deterrent to AD therapeutic investigations. In this study, we analyze the sponsorship data for AD clinical trials conducted since 2016 to assess the fiscal support for AD clinical trials. METHODS We analyzed the funding sources of all AD trials over the past 5 years as reported on ClinicalTrials.gov. RESULTS There were 136 trials being conducted for treatments in the US AD therapeutic pipeline on the index date of this study. Among non-prevention trials, disease-modifying therapies (DMT) in Phase 3 were almost entirely sponsored by the biopharmaceutical industry; Phase 2 DMT trials were split between the biopharmaceutical industry and funding from the National Institutes of Health (NIH) to academic medical centers (AMCs). The majority of prevention trials received sponsorship from public-private partnerships (PPP). Trials of symptomatic agents are equally likely to have biopharmaceutical or NIH/AMC sponsorship. Most trials with repurposed agents had NIH/AMC funding (89%). Since 2016, there has been consistent growth in the number of trials sponsored both in part and fully by NIH/AMC sources and in PPP, and there has been a reduction in biopharmaceutical company-sponsored trials. DISCUSSION The number of trials supported by the biopharmaceutical industry has decreased over the past 5 years; trials supported from federal sources and PPP have increased. Repurposed compounds are mostly in Phase 2 trials and provide critical mechanistic information.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Chambers‐Grundy Center for Transformative NeuroscienceDepartment of Brain HealthSchool of Integrated Health SciencesUniversity of NevadaLas Vegas (UNLV)Las VegasNevadaUSA
| | - Justin Bauzon
- University of NevadaLas Vegas (UNLV)School of MedicineLas VegasNevadaUSA
| | - Garam Lee
- Biogen, Medical AffairsWestonMassachusettsUSA
| |
Collapse
|
9
|
Cummings J. Drug Development for Psychotropic, Cognitive-Enhancing, and Disease-Modifying Treatments for Alzheimer's Disease. J Neuropsychiatry Clin Neurosci 2020; 33:3-13. [PMID: 33108950 PMCID: PMC7989572 DOI: 10.1176/appi.neuropsych.20060152] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with limited available therapies. There is progress in developing treatments for neuropsychiatric indications in AD, including agitation, psychosis, apathy, and sleep disorders. Candidate therapies progress from nonclinical and animal assessment to trials in normal volunteers (phase 1), small phase-2 trials, and larger confirmatory phase-3 trials. Biomarkers play an increasingly important role in selecting participants, stratifying populations, demonstrating target engagement, supporting disease modification, and monitoring safety. There are currently 121 agents in clinical trials, including treatments for neuropsychiatric symptoms, cognition enhancement, and disease progression. There are 27 agents in phase-1 trials, 65 in phase-2 trials, and 29 in phase-3 trials. Most of the agents in trials (80%) target disease modification. Treatments are being assessed in secondary prevention trials with cognitively normal individuals at high risk for the development of AD. There is progress in target diversification, trial designs, outcome measures, biomarkers, and trial population definitions that promise to accelerate developing new therapies for those with or at risk for AD.
Collapse
Affiliation(s)
- Jeffrey Cummings
- The Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, and the Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas
| |
Collapse
|
10
|
Hashweh NN, Bartochowski Z, Khoury R, Grossberg GT. An evaluation of hydromethylthionine as a treatment option for Alzheimer's disease. Expert Opin Pharmacother 2020; 21:619-627. [PMID: 32037892 DOI: 10.1080/14656566.2020.1719066] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a major cause of morbidity worldwide and its prevalence is expected to rise. Previous studies involving compounds that target the accumulation of amyloid β protein have been unsuccessful, renewing interest in therapies directed against intracellular deposits of tau proteins. Derived from methylene blue, hydromethylthionine is a tau aggregation inhibitor that recently emerged as a promising disease-modifying treatment for AD. AREAS COVERED Herein, the authors cover the chemistry, pharmacodynamics and pharmacokinetics of hydromethylthionine and its oxidized form methylthionine chloride (MTC) that was first studied, as well as clinical efficacy and safety of hydromethylthionine in the treatment of mild to moderate AD. EXPERT OPINION Randomized clinical trials with hydromethylthionine failed to show any impact of the doses used on the disease course. Data analysis from a non-randomized cohort showed that a smaller dose of the drug previously thought to be ineffective and used as placebo, prescribed as monotherapy rather than as add-on to AD approved symptomatic therapies may slow cognitive decline. This finding was further confirmed by a pharmacokinetic analysis study showing a dose/response relationship with doses around 16 mg daily. Future trials need to study the pharmacological properties of hydromethylthionine and ascertain the optimal safe and effective dose to be used.
Collapse
Affiliation(s)
- Nader Nael Hashweh
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine , Saint Louis, MO, USA
| | - Zachary Bartochowski
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine , Saint Louis, MO, USA
| | - Rita Khoury
- Department of Psychiatry & Clinical Psychology, St. George Hospital University Medical Center, Balamand University, Faculty of Medicine , Beirut, Lebanon.,Institute for Development, Research, Advocacy & Applied Care (IDRAAC) , Beirut, Lebanon
| | - George T Grossberg
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine , Saint Louis, MO, USA
| |
Collapse
|
11
|
Cummings J, Feldman HH, Scheltens P. The "rights" of precision drug development for Alzheimer's disease. Alzheimers Res Ther 2019; 11:76. [PMID: 31470905 PMCID: PMC6717388 DOI: 10.1186/s13195-019-0529-5] [Citation(s) in RCA: 145] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/13/2019] [Indexed: 01/12/2023]
Abstract
There is a high rate of failure in Alzheimer's disease (AD) drug development with 99% of trials showing no drug-placebo difference. This low rate of success delays new treatments for patients and discourages investment in AD drug development. Studies across drug development programs in multiple disorders have identified important strategies for decreasing the risk and increasing the likelihood of success in drug development programs. These experiences provide guidance for the optimization of AD drug development. The "rights" of AD drug development include the right target, right drug, right biomarker, right participant, and right trial. The right target identifies the appropriate biologic process for an AD therapeutic intervention. The right drug must have well-understood pharmacokinetic and pharmacodynamic features, ability to penetrate the blood-brain barrier, efficacy demonstrated in animals, maximum tolerated dose established in phase I, and acceptable toxicity. The right biomarkers include participant selection biomarkers, target engagement biomarkers, biomarkers supportive of disease modification, and biomarkers for side effect monitoring. The right participant hinges on the identification of the phase of AD (preclinical, prodromal, dementia). Severity of disease and drug mechanism both have a role in defining the right participant. The right trial is a well-conducted trial with appropriate clinical and biomarker outcomes collected over an appropriate period of time, powered to detect a clinically meaningful drug-placebo difference, and anticipating variability introduced by globalization. We lack understanding of some critical aspects of disease biology and drug action that may affect the success of development programs even when the "rights" are adhered to. Attention to disciplined drug development will increase the likelihood of success, decrease the risks associated with AD drug development, enhance the ability to attract investment, and make it more likely that new therapies will become available to those with or vulnerable to the emergence of AD.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Department of Brain Health, School of Integrated Health Sciences, UNLV and Cleveland Clinic Lou Ruvo Center for Brain Health, 888 West Bonneville Ave, Las Vegas, NV, 89106, USA.
| | - Howard H Feldman
- Department of Neurosciences, Alzheimer's Disease Cooperative Study, University of California San Diego, San Diego, CA, USA
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Moscoso A, Silva-Rodríguez J, Aldrey JM, Cortés J, Fernández-Ferreiro A, Gómez-Lado N, Ruibal Á, Aguiar P. Prediction of Alzheimer's disease dementia with MRI beyond the short-term: Implications for the design of predictive models. Neuroimage Clin 2019; 23:101837. [PMID: 31078938 PMCID: PMC6515129 DOI: 10.1016/j.nicl.2019.101837] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Revised: 02/12/2019] [Accepted: 04/24/2019] [Indexed: 11/22/2022]
Abstract
Magnetic resonance imaging (MRI) volumetric measures have become a standard tool for the detection of incipient Alzheimer's Disease (AD) dementia in mild cognitive impairment (MCI). Focused on providing an earlier and more accurate diagnosis, sophisticated MRI machine learning algorithms have been developed over the recent years, most of them learning their non-disease patterns from MCI that remained stable over 2-3 years. In this work, we analyzed whether these stable MCI over short-term periods are actually appropriate training examples of non-disease patterns. To this aim, we compared the diagnosis of MCI patients at 2 and 5 years of follow-up and investigated its impact on the predictive performance of baseline volumetric MRI measures primarily involved in AD, i.e., hippocampal and entorhinal cortex volumes. Predictive power was evaluated in terms of the area under the ROC curve (AUC), sensitivity, and specificity in a trial sample of 248 MCI patients followed-up over 5 years. We further compared the sensitivity in those MCI that converted before 2 years and those that converted after 2 years. Our results indicate that 23% of the stable MCI at 2 years progressed in the next three years and that MRI volumetric measures are good predictors of conversion to AD dementia even at the mid-term, showing a better specificity and AUC as follow-up time increases. The combination of hippocampus and entorhinal cortex yielded an AUC that was significantly higher for the 5-year follow-up (AUC = 73% at 2 years vs. AUC = 84% at 5 years), as well as for specificity (56% vs. 71%). Sensitivity showed a non-significant slight decrease (81% vs. 78%). Remarkably, the performance of this model was comparable to machine learning models at the same follow-up times. MRI correctly identified most of the patients that converted after 2 years (with sensitivity >60%), and these patients showed a similar degree of abnormalities to those that converted before 2 years. This implies that most of the MCI patients that remained stable over short periods and subsequently progressed to AD dementia had evident atrophies at baseline. Therefore, machine learning models that use these patients to learn non-disease patterns are including an important fraction of patients with evident pathological changes related to the disease, something that might result in reduced performance and lack of biological interpretability.
Collapse
Affiliation(s)
- Alexis Moscoso
- Nuclear Medicine Department and Molecular Imaging Group, University Hospital CHUS-IDIS, Travesía da Choupana s/n, Santiago de Compostela 15706, Spain
| | - Jesús Silva-Rodríguez
- Nuclear Medicine Department and Molecular Imaging Group, University Hospital CHUS-IDIS, Travesía da Choupana s/n, Santiago de Compostela 15706, Spain
| | - Jose Manuel Aldrey
- Neurology Department, University Hospital CHUS-IDIS, Travesía da Choupana s/n, Santiago de Compostela 15706, Spain
| | - Julia Cortés
- Nuclear Medicine Department and Molecular Imaging Group, University Hospital CHUS-IDIS, Travesía da Choupana s/n, Santiago de Compostela 15706, Spain
| | - Anxo Fernández-Ferreiro
- Pharmacy Department and Pharmacology Group, University Hospital CHUS-IDIS, Travesía da Choupana s/n, Santiago de Compostela 15706, Spain
| | - Noemí Gómez-Lado
- Nuclear Medicine Department and Molecular Imaging Group, University Hospital CHUS-IDIS, Travesía da Choupana s/n, Santiago de Compostela 15706, Spain
| | - Álvaro Ruibal
- Nuclear Medicine Department and Molecular Imaging Group, University Hospital CHUS-IDIS, Travesía da Choupana s/n, Santiago de Compostela 15706, Spain; Molecular Imaging Group, Department of Radiology, Faculty of Medicine, University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela 15782, Spain; Fundación Tejerina, Madrid 28003, Spain
| | - Pablo Aguiar
- Nuclear Medicine Department and Molecular Imaging Group, University Hospital CHUS-IDIS, Travesía da Choupana s/n, Santiago de Compostela 15706, Spain; Molecular Imaging Group, Department of Radiology, Faculty of Medicine, University of Santiago de Compostela (USC), Campus Vida, Santiago de Compostela 15782, Spain.
| |
Collapse
|
13
|
Kilic B, Gulcan HO, Aksakal F, Ercetin T, Oruklu N, Umit Bagriacik E, Dogruer DS. Design and synthesis of some new carboxamide and propanamide derivatives bearing phenylpyridazine as a core ring and the investigation of their inhibitory potential on in-vitro acetylcholinesterase and butyrylcholinesterase. Bioorg Chem 2018; 79:235-249. [PMID: 29775949 DOI: 10.1016/j.bioorg.2018.05.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 05/04/2018] [Accepted: 05/07/2018] [Indexed: 02/07/2023]
Abstract
A series of new carboxamide and propanamide derivatives bearing phenylpyridazine as a core ring were designed, synthesized and evaluated for their ability to inhibit both cholinesterase enzymes. In addition, a series of carboxamide and propanamide derivatives bearing biphenyl instead of phenylpyridazine were also synthesized to examine the inhibitory effect of pyridazine moiety on both cholinesterase enzymes. The inhibitory activity results revealed that compounds 5b, 5f, 5h, 5j, 5l pyridazine-3-carboxamide derivative, exhibited selective acetylcholinesterase (AChE) inhibition with IC50 values ranging from 0.11 to 2.69 µM. Among them, compound 5h was the most active one (IC50 = 0.11 µM) without cytotoxic effect at its effective concentration against AChE. Additionally, pyridazine-3-carboxamide derivative 5d (IC50 for AChE = 0.16 µM and IC50 for BChE = 9.80 µM) and biphenyl-4-carboxamide derivative 6d (IC50 for AChE = 0.59 µM and IC50 for BChE = 1.48 µM) displayed dual cholinesterase inhibitory activity. Besides, active compounds were also tested for their ability to inhibit Aβ aggregation. Theoretical physicochemical properties of the compounds were calculated by using Molinspiration Program as well. The Lineweaver-Burk plot and docking study showed that compound 5 h targeted both the catalytic active site (CAS) and the peripheral anionic site (PAS) of AChE.
Collapse
Affiliation(s)
- Burcu Kilic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey
| | - Hayrettin O Gulcan
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Eastern Mediterranean University, Gazimagosa, Cyprus
| | - Fatma Aksakal
- Department of Chemistry, Hacettepe University, Ankara, Turkey
| | - Tugba Ercetin
- Department of Pharmacognosy, Faculty of Pharmacy, Eastern Mediterranean University, Gazimagosa, Cyprus
| | - Nihan Oruklu
- Department of Immunology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - E Umit Bagriacik
- Department of Immunology, Faculty of Medicine, Gazi University, Ankara, Turkey
| | - Deniz S Dogruer
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Gazi University, Ankara, Turkey.
| |
Collapse
|
14
|
The contribution of transgenic and nontransgenic animal models in Alzheimer's disease drug research and development. Behav Pharmacol 2018; 28:95-111. [PMID: 28177983 DOI: 10.1097/fbp.0000000000000296] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Over the last few years, several papers have become available in the literature on both the main hallmarks of Alzheimer's disease (AD) and the several intracellular pathways whose alteration is responsible for its onset and progression. The use of transgenic and nontransgenic animal models has played a key role in achieving such a remarkable amount of preclinical data, allowing researchers to dissect the cellular changes occurring in the AD brain. In addition, the huge amount of preclinical evidence arising from these animal models was necessary for the further clinical development of pharmacological agents capable of interfering with most of the impaired neural pathways in AD patients. In this respect, a significant role is played by the dysfunction of excitatory and inhibitory neurotransmission responsible for the cognitive and behavioral symptoms described in AD patients. The aim of this review is to summarize the main animal models that contributed toward unraveling the pathological changes in neurotransmitter synthesis, release, and receptor binding in AD preclinical studies. The review also provides an updated description of the current pharmacological agents - still under clinical development - acting on the neurotransmitter systems.
Collapse
|
15
|
Effects of the Insulin Sensitizer Metformin in Alzheimer Disease: Pilot Data From a Randomized Placebo-controlled Crossover Study. Alzheimer Dis Assoc Disord 2017; 31:107-113. [PMID: 28538088 DOI: 10.1097/wad.0000000000000202] [Citation(s) in RCA: 237] [Impact Index Per Article: 29.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Epidemiological studies have identified a robust association between type II diabetes mellitus and Alzheimer disease (AD), and neurobiological studies have suggested the presence of central nervous system insulin resistance in individuals with AD. Given this association, we hypothesized that the central nervous system-penetrant insulin-sensitizing medication metformin would be beneficial as a disease-modifying and/or symptomatic therapy for AD, and conducted a placebo-controlled crossover study of its effects on cerebrospinal fluid (CSF), neuroimaging, and cognitive biomarkers. Twenty nondiabetic subjects with mild cognitive impairment or mild dementia due to AD were randomized to receive metformin then placebo for 8 weeks each or vice versa. CSF and neuroimaging (Arterial Spin Label MRI) data were collected for biomarker analyses, and cognitive testing was performed. Metformin was found to be safe, well-tolerated, and measureable in CSF at an average steady-state concentration of 95.6 ng/mL. Metformin was associated with improved executive functioning, and trends suggested improvement in learning/memory and attention. No significant changes in cerebral blood flow were observed, though post hoc completer analyses suggested an increase in orbitofrontal cerebral blood flow with metformin exposure. Further study of these findings is warranted.
Collapse
|
16
|
Gold M. Phase II clinical trials of anti-amyloid β antibodies: When is enough, enough? ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2017; 3:402-409. [PMID: 29067346 PMCID: PMC5651424 DOI: 10.1016/j.trci.2017.04.005] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Efforts to develop new therapies to combat Alzheimer's disease suffer from extraordinarily high failure rates that make it difficult to justify continued investment in the field. Although there are a number of plausible explanations for this extremely high attrition rate, one of the explanations that has received little attention is the lack of compelling data from Phase II studies for compounds that have been pushed into Phase III trials and then have failed. An analysis of publicly available data from the Phase II studies for bapineuzumab and solanezumab indicates that neither compound produced compelling evidence of drug-like behavior that would justify their progression into pivotal trials. The published data suggest that sponsors took decisions to move these compounds into Phase III on the basis of vastly limited data that were rife with type I error and probably driven by commercial concerns. The continued push to move compounds that are not likely to succeed in later stage clinical trials threatens to erode trust in the clinical research enterprise making it much harder to properly test truly promising compounds.
Collapse
Affiliation(s)
- Michael Gold
- Global Product Development, Neuroscience, PPD, Morrisville, NC, USA
| |
Collapse
|
17
|
Satlin A, Wang J, Logovinsky V, Berry S, Swanson C, Dhadda S, Berry DA. Design of a Bayesian adaptive phase 2 proof-of-concept trial for BAN2401, a putative disease-modifying monoclonal antibody for the treatment of Alzheimer's disease. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2016; 2:1-12. [PMID: 29067290 PMCID: PMC5644271 DOI: 10.1016/j.trci.2016.01.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Introduction Recent failures in phase 3 clinical trials in Alzheimer's disease (AD) suggest that novel approaches to drug development are urgently needed. Phase 3 risk can be mitigated by ensuring that clinical efficacy is established before initiating confirmatory trials, but traditional phase 2 trials in AD can be lengthy and costly. Methods We designed a Bayesian adaptive phase 2, proof-of-concept trial with a clinical endpoint to evaluate BAN2401, a monoclonal antibody targeting amyloid protofibrils. The study design used dose response and longitudinal modeling. Simulations were used to refine study design features to achieve optimal operating characteristics. Results The study design includes five active treatment arms plus placebo, a clinical outcome, 12-month primary endpoint, and a maximum sample size of 800. The average overall probability of success is ≥80% when at least one dose shows a treatment effect that would be considered clinically meaningful. Using frequent interim analyses, the randomization ratios are adapted based on the clinical endpoint, and the trial can be stopped for success or futility before full enrollment. Discussion Bayesian statistics can enhance the efficiency of analyzing the study data. The adaptive randomization generates more data on doses that appear to be more efficacious, which can improve dose selection for phase 3. The interim analyses permit stopping as soon as a predefined signal is detected, which can accelerate decision making. Both features can reduce the size and duration of the trial. This study design can mitigate some of the risks associated with advancing to phase 3 in the absence of data demonstrating clinical efficacy. Limitations to the approach are discussed.
Collapse
Affiliation(s)
- Andrew Satlin
- Neuroscience & General Medicine, Eisai Inc., Woodcliff Lake, NJ, USA
| | - Jinping Wang
- Neuroscience & General Medicine, Eisai Inc., Woodcliff Lake, NJ, USA
| | | | | | - Chad Swanson
- Neuroscience & General Medicine, Eisai Inc., Woodcliff Lake, NJ, USA
| | - Shobha Dhadda
- Neuroscience & General Medicine, Eisai Inc., Woodcliff Lake, NJ, USA
| | | |
Collapse
|
18
|
Abstract
Neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and frontotemporal dementia have several important features in common. They are progressive, they affect a relatively inaccessible organ, and we have no disease-modifying therapies for them. For these brain-based diseases, current diagnosis and evaluation of disease severity rely almost entirely on clinical examination, which may be only a rough approximation of disease state. Thus, the development of biomarkers-objective, relatively easily measured, and precise indicators of pathogenic processes-could improve patient care and accelerate therapeutic discovery. Yet existing, rigorously tested neurodegenerative disease biomarkers are few, and even fewer biomarkers have translated into clinical use. To find new biomarkers for these diseases, an unbiased, high-throughput screening approach may be needed. In this review, I will describe the potential utility of such an approach to biomarker discovery, using Parkinson's disease as a case example.
Collapse
Affiliation(s)
- Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, 3 West Gates, 3400 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
19
|
Ghimire Gautam S, Komatsu M, Nishigaki K. Strong Inhibition of Beta-Amyloid Peptide Aggregation Realized by Two-Steps Evolved Peptides. Chem Biol Drug Des 2014; 85:356-68. [DOI: 10.1111/cbdd.12400] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 07/12/2014] [Accepted: 07/21/2014] [Indexed: 12/24/2022]
Affiliation(s)
- Sunita Ghimire Gautam
- Department of Functional Materials Science; Graduate School of Science and Engineering; Saitama University; 255 Shimo-okubo Sakura-Ku Saitama-Shi 338-8570 Japan
| | - Masayuki Komatsu
- Department of Functional Materials Science; Graduate School of Science and Engineering; Saitama University; 255 Shimo-okubo Sakura-Ku Saitama-Shi 338-8570 Japan
| | - Koichi Nishigaki
- Department of Functional Materials Science; Graduate School of Science and Engineering; Saitama University; 255 Shimo-okubo Sakura-Ku Saitama-Shi 338-8570 Japan
| |
Collapse
|
20
|
Lundkvist J, Halldin MM, Sandin J, Nordvall G, Forsell P, Svensson S, Jansson L, Johansson G, Winblad B, Ekstrand J. The battle of Alzheimer's Disease - the beginning of the future Unleashing the potential of academic discoveries. Front Pharmacol 2014; 5:102. [PMID: 24847271 PMCID: PMC4023025 DOI: 10.3389/fphar.2014.00102] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 04/18/2014] [Indexed: 01/21/2023] Open
Abstract
Alzheimer's Disease (AD) is the most common form of dementia, affecting approximately 36 million people worldwide. To date there is no preventive or curative treatment available for AD, and in absence of major progress in therapeutic development, AD manifests a concrete socioeconomic threat. The awareness of the growing problem of AD is increasing, exemplified by the recent G8 Dementia Summit, a meeting held in order to set the stage and steer the compass for the future. Simultaneously, and paradoxically, we have seen key players in the pharmaceutical industry that have recently closed or significantly decreased their R&D spending on AD and other CNS disorders. Given the pressing need for new treatments in this area, other actors need to step-in and enter this drug discovery arena complementing the industrial efforts, in order to turn biological and technological progress into novel therapeutics. In this article, we present an example of a novel drug discovery initiative that in a non-profit setting, aims to integrate with both preclinical and clinical academic groups and pharmaceutical industry to explore the therapeutic potential of new concepts in patients, using novel biology, state of the art technologies and rapid concept testing.
Collapse
Affiliation(s)
- Johan Lundkvist
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | - Magnus M. Halldin
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | - Johan Sandin
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | - Gunnar Nordvall
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | - Pontus Forsell
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | - Samuel Svensson
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| | | | - Gunilla Johansson
- Center for Alzheimer Research at Karolinska Institutet and Swedish Brain PowerNovum, Huddinge, Sweden
| | - Bengt Winblad
- Center for Alzheimer Research at Karolinska Institutet and Swedish Brain PowerNovum, Huddinge, Sweden
| | - Jonas Ekstrand
- AlzeCure Foundation, Karolinska Institutet Science Park NovumHuddinge, Sweden
| |
Collapse
|
21
|
Chuang YF, Breitner JCS, Chiu YL, Khachaturian A, Hayden K, Corcoran C, Tschanz J, Norton M, Munger R, Welsh-Bohmer K, Zandi PP. Use of diuretics is associated with reduced risk of Alzheimer's disease: the Cache County Study. Neurobiol Aging 2014; 35:2429-2435. [PMID: 24910391 DOI: 10.1016/j.neurobiolaging.2014.05.002] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 03/27/2014] [Accepted: 05/02/2014] [Indexed: 10/25/2022]
Abstract
Although the use of antihypertensive medications has been associated with reduced risk of Alzheimer's disease (AD), it remains unclear which class provides the most benefit. The Cache County Study of Memory Health and Aging is a prospective longitudinal cohort study of dementing illnesses among the elderly population of Cache County, Utah. Using waves I to IV data of the Cache County Study, 3417 participants had a mean of 7.1 years of follow-up. Time-varying use of antihypertensive medications including different class of diuretics, angiotensin converting enzyme inhibitors, β-blockers, and calcium channel blockers was used to predict the incidence of AD using Cox proportional hazards analyses. During follow-up, 325 AD cases were ascertained with a total of 23,590 person-years. Use of any antihypertensive medication was associated with lower incidence of AD (adjusted hazard ratio [aHR], 0.77; 95% confidence interval [CI], 0.61-0.97). Among different classes of antihypertensive medications, thiazide (aHR, 0.7; 95% CI, 0.53-0.93), and potassium-sparing diuretics (aHR, 0.69; 95% CI, 0.48-0.99) were associated with the greatest reduction of AD risk. Thiazide and potassium-sparing diuretics were associated with decreased risk of AD. The inverse association of potassium-sparing diuretics confirms an earlier finding in this cohort, now with longer follow-up, and merits further investigation.
Collapse
Affiliation(s)
- Yi-Fang Chuang
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - John C S Breitner
- Department of Psychiatry, McGill University Faculty of Medicine, Montreal, Quebec, Canada
| | - Yen-Ling Chiu
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA; Department of Medicine, Far-Eastern Memorial Hospital, New Taipei City, Taiwan
| | - Ara Khachaturian
- National Supercomputing Center (NSCEE) for Energy and the Environment, University of Nevada, Las Vegas, NV, USA
| | - Kathleen Hayden
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Chris Corcoran
- Center for Epidemiologic Studies, Utah State University, Logan, UT, USA; Department of Mathematics and Statistics, Utah State University, Logan, UT, USA
| | - JoAnn Tschanz
- Center for Epidemiologic Studies, Utah State University, Logan, UT, USA; Department of Psychology, Utah State University, Logan, UT, USA
| | - Maria Norton
- Center for Epidemiologic Studies, Utah State University, Logan, UT, USA; Department of Psychology, Utah State University, Logan, UT, USA; Department of Family, Consumer, and Human Development, Utah State University, Logan, UT, USA
| | - Ron Munger
- Center for Epidemiologic Studies, Utah State University, Logan, UT, USA
| | - Kathleen Welsh-Bohmer
- Department of Psychiatry and Behavioral Sciences, Duke University School of Medicine, Durham, NC, USA
| | - Peter P Zandi
- Department of Mental Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA.
| | | |
Collapse
|
22
|
Scott TJ, O'Connor AC, Link AN, Beaulieu TJ. Economic analysis of opportunities to accelerate Alzheimer's disease research and development. Ann N Y Acad Sci 2014; 1313:17-34. [PMID: 24673372 PMCID: PMC4285871 DOI: 10.1111/nyas.12417] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The development of disease-modifying treatments for Alzheimer's disease (AD) faces a number of barriers. Among these are the lack of surrogate biomarkers, the exceptional size and duration of clinical trials, difficulties in identifying appropriate populations for clinical trials, and the limitations of monotherapies in addressing such a complex multifactorial disease. This study sets out to first estimate the consequent impact on the expected cost of developing disease-modifying treatments for AD and then to estimate the potential benefits of bringing together industry, academic, and government stakeholders to co-invest in, for example, developing better biomarkers and cognitive assessment tools, building out advanced registries and clinical trial-readiness cohorts, and establishing clinical trial platforms to investigate combinations of candidate drugs and biomarkers from the portfolios of multiple companies. Estimates based on interviews with experts on AD research and development suggest that the cost of one new drug is now $5.7 billion (95% confidence interval (CI) $3.7-9.5 billion) and could be reduced to $2.0 billion (95% CI $1.5-2.9 billion). The associated acceleration in the arrival of disease-modifying treatments could reduce the number of case years of dementia by 7.0 million (95% CI 4.4-9.4 million) in the United States from 2025 through 2040.
Collapse
Affiliation(s)
- Troy J Scott
- RTI International, Research Triangle Park, North Carolina
| | | | | | | |
Collapse
|
23
|
|
24
|
Kang JH, Korecka M, Toledo JB, Trojanowski JQ, Shaw LM. Clinical utility and analytical challenges in measurement of cerebrospinal fluid amyloid-β(1-42) and τ proteins as Alzheimer disease biomarkers. Clin Chem 2013; 59:903-16. [PMID: 23519967 DOI: 10.1373/clinchem.2013.202937] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND Over the past 2 decades, clinical studies have provided evidence that cerebrospinal fluid (CSF) amyloid β(1-42) (Aβ(1-42)), total τ (t-τ), and τ phosphorylated at Thr181 (p-τ(181)) are reliable biochemical markers of Alzheimer disease (AD) neuropathology. CONTENT In this review, we summarize the clinical performance and describe the major challenges for the analytical performance of the most widely used immunoassay platforms [based on ELISA or microbead-based multianalyte profiling (xMAP) technology] for the measurement of CSF AD biomarkers (Aβ(1-42), t-τ, and p-τ(181)). With foundational immunoassay data providing the diagnostic and prognostic values of CSF AD biomarkers, the newly revised criteria for the diagnosis of AD include CSF AD biomarkers for use in research settings. In addition, it has been suggested that the selection of AD patients at the predementia stage by use of CSF AD biomarkers can improve the statistical power of clinical trial design. Owing to the lack of a replenishable and commutable human CSF-based standardized reference material (SRM) and significant differences across different immunoassay platforms, the diagnostic-prognostic cutpoints of CSF AD biomarker concentrations are not universal at this time. These challenges can be effectively met in the future, however, through collaborative ongoing standardization efforts to minimize the sources of analytical variability and to develop reference methods and SRMs. SUMMARY Measurements of CSF Aβ(1-42), t-τ, and p-τ(181) with analytically qualified immunoassays reliably reflect the neuropathologic hallmarks of AD in patients at the early predementia stage of the disease and even in presymptomatic patients. Thus these CSF biomarker tests are useful for early diagnosis of AD, prediction of disease progression, and efficient design of drug intervention clinical trials.
Collapse
Affiliation(s)
- Ju-Hee Kang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|