151
|
Bradshaw EL, Spilker ME, Zang R, Bansal L, He H, Jones RD, Le K, Penney M, Schuck E, Topp B, Tsai A, Xu C, Nijsen MJ, Chan JR. Applications of Quantitative Systems Pharmacology in Model-Informed Drug Discovery: Perspective on Impact and Opportunities. CPT Pharmacometrics Syst Pharmacol 2019; 8:777-791. [PMID: 31535440 PMCID: PMC6875708 DOI: 10.1002/psp4.12463] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 07/19/2019] [Indexed: 12/15/2022] Open
Abstract
Quantitative systems pharmacology (QSP) approaches have been increasingly applied in the pharmaceutical since the landmark white paper published in 2011 by a National Institutes of Health working group brought attention to the discipline. In this perspective, we discuss QSP in the context of other modeling approaches and highlight the impact of QSP across various stages of drug development and therapeutic areas. We discuss challenges to the field as well as future opportunities.
Collapse
Affiliation(s)
| | - Mary E. Spilker
- Pfizer Worldwide Research and DevelopmentSan DiegoCaliforniaUSA
| | | | | | - Handan He
- Novartis Institutes for Biomedical ResearchEast HanoverNew JerseyUSA
| | | | - Kha Le
- AgiosCambridgeMassachusettsUSA
| | | | | | | | - Alice Tsai
- Vertex Pharmaceuticals IncorporatedBostonMassachusettsUSA
| | | | | | | |
Collapse
|
152
|
Johnsen KB, Burkhart A, Thomsen LB, Andresen TL, Moos T. Targeting the transferrin receptor for brain drug delivery. Prog Neurobiol 2019; 181:101665. [DOI: 10.1016/j.pneurobio.2019.101665] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 07/10/2019] [Accepted: 07/18/2019] [Indexed: 02/07/2023]
|
153
|
Reynolds DS. A short perspective on the long road to effective treatments for Alzheimer's disease. Br J Pharmacol 2019; 176:3636-3648. [PMID: 30657599 PMCID: PMC6715596 DOI: 10.1111/bph.14581] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 11/21/2018] [Accepted: 12/03/2018] [Indexed: 12/13/2022] Open
Abstract
Globally, there are approximately 47 million people living with dementia, and about two thirds of those have Alzheimer's disease (AD). Age is the single biggest risk factor for the vast majority of sporadic AD cases, and because the world's population is aging, the number of people living with AD is set to rise dramatically over the coming decades. There are currently no disease-modifying treatments for AD, and the few symptomatic agents available have limited impact on the disease. Perhaps surprisingly, there is relatively little activity in the AD research and development field compared with other diseases with a high mortality burden, such as cancer. There is enormous economic incentive to discover and develop the first disease-modifying treatment, but previous failure has significantly reduced further industrial investment in this field. The short review looks at the historical path trodden to develop treatments and reflects on the journey down the road to truly effective treatments for people living with AD. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
|
154
|
Yang T, Dang Y, Ostaszewski B, Mengel D, Steffen V, Rabe C, Bittner T, Walsh DM, Selkoe DJ. Target engagement in an alzheimer trial: Crenezumab lowers amyloid β oligomers in cerebrospinal fluid. Ann Neurol 2019; 86:215-224. [PMID: 31168802 PMCID: PMC6771589 DOI: 10.1002/ana.25513] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Oligomeric forms of amyloid β protein (oAβ) are believed to be principally responsible for neurotoxicity in Alzheimer disease (AD), but it is not known whether anti-Aβ antibodies are capable of lowering oAβ levels in humans. METHODS We developed an ultrasensitive immunoassay and used it to measure oAβ in cerebrospinal fluid (CSF) from 104 AD subjects participating in the ABBY and BLAZE phase 2 trials of the anti-Aβ antibody crenezumab. Patients received subcutaneous (SC) crenezumab (300mg) or placebo every 2 weeks, or intravenous (IV) crenezumab (15mg/kg) or placebo every 4 weeks for 68 weeks. Ninety-eight of the 104 patients had measurable baseline oAβ levels, and these were compared to levels at week 69 in placebo (n = 28), SC (n = 35), and IV (n = 35) treated patients. RESULTS Among those receiving crenezumab, 89% of SC and 86% of IV patients had lower levels of oAβ at week 69 versus baseline. The difference in the proportion of patients with decreasing levels was significant for both treatment arms: p = 0.0035 for SC and p = 0.01 for IV crenezumab versus placebo. The median percentage change was -48% in the SC arm and -43% in the IV arm. No systematic change was observed in the placebo group, with a median change of -13% and equivalent portions with negative and positive change. INTERPRETATION Crenezumab lowered CSF oAβ levels in the large majority of treated patients tested. These results support engagement of the principal pathobiological target in AD and identify CSF oAβ as a novel pharmacodynamic biomarker for use in trials of anti-Aβ agents. ANN NEUROL 2019;86:215-224.
Collapse
Affiliation(s)
- Ting Yang
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMA
| | - Yifan Dang
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMA
| | - Beth Ostaszewski
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMA
| | - David Mengel
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMA
| | - Verena Steffen
- Genentech (a member of the Roche Group)South San FranciscoCA
| | - Christina Rabe
- Genentech (a member of the Roche Group)South San FranciscoCA
| | - Tobias Bittner
- Genentech (a member of the Roche Group)South San FranciscoCA
| | - Dominic M. Walsh
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMA
| | - Dennis J. Selkoe
- Ann Romney Center for Neurologic Diseases, Department of NeurologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMA
| |
Collapse
|
155
|
Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer's disease drug development pipeline: 2019. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2019; 5:272-293. [PMID: 31334330 PMCID: PMC6617248 DOI: 10.1016/j.trci.2019.05.008] [Citation(s) in RCA: 506] [Impact Index Per Article: 84.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Alzheimer's disease (AD) has few available treatments, and there is a high rate of failure in AD drug development programs. Study of the AD drug development pipeline can provide insight into the evolution of drug development and how best to optimize development practices. METHODS We reviewed clinicaltrials.gov and identified all pharmacologic AD trials of all agents currently being developed for treatment of AD. RESULTS There are 132 agents in clinical trials for the treatment of AD. Twenty-eight agents are in 42 phase 3 trials; 74 agents are in 83 phase 2 trials; and 30 agents are in 31 phase 1 trials. There is an increase in the number of agents in each phase compared with that in the 2018 pipeline. Nineteen agents in trials target cognitive enhancement, and 14 are intended to treat neuropsychiatric and behavioral symptoms. There are 96 agents in disease modification trials; of these, 38 (40%) have amyloid as the primary target or as one of several effects. Eighteen of the antiamyloid agents are small molecules, and 20 are monoclonal antibodies or biological therapies. Seven small molecules and ten biologics have tau as a primary or combination target (18%). Amyloid is the most common specific target in phase 3 and phase 2 disease modification trials. Novel biomarkers (e.g., neurofilament light), new outcomes (e.g., AD Composite Score [ADCOMS]), enrollment of earlier populations, and innovative trial designs (e.g., Bayesian adaptive designs) are new features in recent clinical trials. DISCUSSION Drug development continues robustly at all phases despite setbacks in several programs in the recent past. Continuing unmet needs require a commitment to growing and accelerating the pipeline.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Department of Brain Health, University of Nevada, Las Vegas (UNLV), School of Integrated Health Sciences, Las Vegas, NV, USA
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Garam Lee
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Aaron Ritter
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Marwan Sabbagh
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | | |
Collapse
|
156
|
Gibbs E, Silverman JM, Zhao B, Peng X, Wang J, Wellington CL, Mackenzie IR, Plotkin SS, Kaplan JM, Cashman NR. A Rationally Designed Humanized Antibody Selective for Amyloid Beta Oligomers in Alzheimer's Disease. Sci Rep 2019; 9:9870. [PMID: 31285517 PMCID: PMC6614461 DOI: 10.1038/s41598-019-46306-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 06/25/2019] [Indexed: 11/29/2022] Open
Abstract
Advances in the understanding of Alzheimer’s disease (AD) suggest that pathogenesis is not directly related to plaque burden, but rather to soluble toxic amyloid-beta oligomers (AßO). Therapeutic antibodies targeting Aß monomers and/or plaque have shown limited efficacy and dose-limiting adverse events in clinical trials. These findings suggest that antibodies capable of selectively neutralizing toxic AßO may achieve improved efficacy and safety. To this end, we generated monoclonal antibodies against a conformational Aß epitope predicted by computational modeling to be presented on toxic AßO but not monomers or fibrils. The resulting lead antibody, PMN310, showed the desired AßO-selective binding profile. In vitro, PMN310 inhibited AßO propagation and toxicity. In vivo, PMN310 prevented AßO-induced loss of memory formation and reduced synaptic loss and inflammation. A humanized version (huPMN310) compared favorably to other Aß-directed antibodies showing a lack of adverse event-associated binding to Aß deposits in AD brains, and greater selective binding to AßO-enriched AD brain fractions that contain synaptotoxic Aß species. Systemic administration of huPMN310 in mice resulted in brain exposure and kinetics comparable to those of other therapeutic human monoclonal antibodies. Greater selectivity for AßO and the potential to safely administer high doses of huPMN310 are expected to result in enhanced safety and therapeutic potency.
Collapse
Affiliation(s)
- Ebrima Gibbs
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, V6T 2B5, Canada
| | - Judith M Silverman
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, V6T 2B5, Canada
| | - Beibei Zhao
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, V6T 2B5, Canada
| | - Xubiao Peng
- University of British Columbia, Department of Physics and Astronomy, Vancouver, BC, V6T 1Z1, Canada
| | - Jing Wang
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, V6T 2B5, Canada
| | - Cheryl L Wellington
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, V6T 2B5, Canada
| | - Ian R Mackenzie
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, V6T 2B5, Canada
| | - Steven S Plotkin
- University of British Columbia, Department of Physics and Astronomy and Genome Sciences and Technology Program, Vancouver, BC, V6T 1Z1, Canada.,ProMIS Neurosciences, Cambridge, MA, 02142, USA
| | | | - Neil R Cashman
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC, V6T 2B5, Canada. .,ProMIS Neurosciences, Cambridge, MA, 02142, USA.
| |
Collapse
|
157
|
Chantran Y, Capron J, Alamowitch S, Aucouturier P. Anti-Aβ Antibodies and Cerebral Amyloid Angiopathy Complications. Front Immunol 2019; 10:1534. [PMID: 31333665 PMCID: PMC6620823 DOI: 10.3389/fimmu.2019.01534] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 06/19/2019] [Indexed: 11/13/2022] Open
Abstract
Cerebral amyloid angiopathy (CAA) corresponds to the deposition of amyloid material in the cerebral vasculature, leading to structural modifications of blood vessel walls. The most frequent form of sporadic CAA involves fibrillar β-amyloid peptide (Aβ) deposits, mainly the 40 amino acid form (Aβ1-40), which are commonly found in the elderly with or without Alzheimer's disease. Sporadic CAA usually remains clinically silent. However, in some cases, acute complications either hemorrhagic or inflammatory can occur. Similar complications occurred after active or passive immunization against Aβ in experimental animal models exhibiting CAA, and in subjects with Alzheimer's disease during clinical trials. The triggering of these adverse events by active immunization and monoclonal antibody administration in CAA-bearing individuals suggests that analogous mechanisms could be involved during spontaneous CAA complications, drawing particular attention to the role of anti-Aβ antibodies. However, antibodies that react with several monomeric and aggregated forms of Aβ spontaneously occur in virtually all human individuals, hence being part of the "natural antibody" repertoire. Natural antibodies are usually described as having low-affinity and high cross-reactivity toward microbial components and autoantigens. Although frequently of the IgM class, they also belong to IgG and IgA isotypes. They likely display homeostatic functions and protective roles in aging. Until recently, the peculiar properties of these natural antibodies have hindered proper analysis of the Aβ-reactive antibody repertoire and the study of their implication in CAA complications. Herein, we review and comment the evidences of an auto-immune nature of spontaneous CAA complications, and discuss implications for forthcoming research and clinical practice.
Collapse
Affiliation(s)
- Yannick Chantran
- Sorbonne Université, Inserm, UMRS 938, Hôpital St-Antoine, AP-HP, Paris, France.,Département d'Immunologie Biologique, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Jean Capron
- Sorbonne Université, Inserm, UMRS 938, Hôpital St-Antoine, AP-HP, Paris, France.,Département de Neurologie, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Sonia Alamowitch
- Sorbonne Université, Inserm, UMRS 938, Hôpital St-Antoine, AP-HP, Paris, France.,Département de Neurologie, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Pierre Aucouturier
- Sorbonne Université, Inserm, UMRS 938, Hôpital St-Antoine, AP-HP, Paris, France.,Département d'Immunologie Biologique, Hôpital Saint-Antoine, AP-HP, Paris, France
| |
Collapse
|
158
|
Duggal P, Mehan S. Neuroprotective Approach of Anti-Cancer Microtubule Stabilizers Against Tauopathy Associated Dementia: Current Status of Clinical and Preclinical Findings. J Alzheimers Dis Rep 2019; 3:179-218. [PMID: 31435618 PMCID: PMC6700530 DOI: 10.3233/adr-190125] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Neuronal microtubule (MT) tau protein provides cytoskeleton to neuronal cells and plays a vital role including maintenance of cell shape, intracellular transport, and cell division. Tau hyperphosphorylation mediates MT destabilization resulting in axonopathy and neurotransmitter deficit, and ultimately causing Alzheimer’s disease (AD), a dementing disorder affecting vast geriatric populations worldwide, characterized by the existence of extracellular amyloid plaques and intracellular neurofibrillary tangles in a hyperphosphorylated state. Pre-clinically, streptozotocin stereotaxically mimics the behavioral and biochemical alterations similar to AD associated with tau pathology resulting in MT assembly defects, which proceed neuropathological cascades. Accessible interventions like cholinesterase inhibitors and NMDA antagonist clinically provides only symptomatic relief. Involvement of microtubule stabilizers (MTS) prevents tauopathy particularly by targeting MT oriented cytoskeleton and promotes polymerization of tubulin protein. Multiple in vitro and in vivo research studies have shown that MTS can hold substantial potential for the treatment of AD-related tauopathy dementias through restoration of tau function and axonal transport. Moreover, anti-cancer taxane derivatives and epothiolones may have potential to ameliorate MT destabilization and prevent the neuronal structural and functional alterations associated with tauopathies. Therefore, this current review strictly focuses on exploration of various clinical and pre-clinical features available for AD to understand the neuropathological mechanisms as well as introduce pharmacological interventions associated with MT stabilization. MTS from diverse natural sources continue to be of value in the treatment of cancer, suggesting that these agents have potential to be of interest in the treatment of AD-related tauopathy dementia in the future.
Collapse
Affiliation(s)
- Pallavi Duggal
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
159
|
Pardridge WM. Alzheimer’s disease: future drug development and the blood-brain barrier. Expert Opin Investig Drugs 2019; 28:569-572. [DOI: 10.1080/13543784.2019.1627325] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
160
|
Govindpani K, McNamara LG, Smith NR, Vinnakota C, Waldvogel HJ, Faull RL, Kwakowsky A. Vascular Dysfunction in Alzheimer's Disease: A Prelude to the Pathological Process or a Consequence of It? J Clin Med 2019; 8:E651. [PMID: 31083442 PMCID: PMC6571853 DOI: 10.3390/jcm8050651] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Despite decades of research following several theoretical and clinical lines, all existing treatments for the disorder are purely symptomatic. AD research has traditionally been focused on neuronal and glial dysfunction. Although there is a wealth of evidence pointing to a significant vascular component in the disease, this angle has been relatively poorly explored. In this review, we consider the various aspects of vascular dysfunction in AD, which has a significant impact on brain metabolism and homeostasis and the clearance of β-amyloid and other toxic metabolites. This may potentially precede the onset of the hallmark pathophysiological and cognitive symptoms of the disease. Pathological changes in vessel haemodynamics, angiogenesis, vascular cell function, vascular coverage, blood-brain barrier permeability and immune cell migration may be related to amyloid toxicity, oxidative stress and apolipoprotein E (APOE) genotype. These vascular deficits may in turn contribute to parenchymal amyloid deposition, neurotoxicity, glial activation and metabolic dysfunction in multiple cell types. A vicious feedback cycle ensues, with progressively worsening neuronal and vascular pathology through the course of the disease. Thus, a better appreciation for the importance of vascular dysfunction in AD may open new avenues for research and therapy.
Collapse
Affiliation(s)
- Karan Govindpani
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Laura G McNamara
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Nicholas R Smith
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Chitra Vinnakota
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Richard Lm Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
161
|
Gandbhir O, Sundaram P. Pre-Clinical Safety and Efficacy Evaluation of Amytrap, a Novel Therapeutic to Treat Alzheimer's Disease. J Alzheimers Dis Rep 2019; 3:77-94. [PMID: 31259305 PMCID: PMC6597960 DOI: 10.3233/adr-190107] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common cause of dementia. Amyloid-β (Aβ42) is implicated in AD pathogenesis. We have designed a non-immune based proprietary therapeutic, called Amytrap, a conjugate containing a retro-inverso peptide, polyethylene glycol, and human serum albumin. Amytrap not only binds Aβ42 but also prevents and dissociates aggregated Aβ42. Amytrap binds to the region in Aβ42 known to trigger its self-aggregation, thus disrupting aggregation. We have obtained proof of concept on AmyTrap in a clinically relevant mouse model, namely, AD-APPSWE/Tg2576. We synthesized and characterized Amytrap and confirmed its authenticity. Efficacy evaluations were performed on young (5 months) and old (9 months) model mice. Amytrap was injected biweekly for a period of five months. Pharmacokinetics and safety toxicology were assessed in normal mice and rats, respectively. Post treatment, younger mice showed significant improvements in cognition and Aβ42 levels in plasma, brain, and cerebrospinal fluid, while older mice showed less significant benefits. Immunohistochemistry of brain sections showed similar differences between young and old mice. They all had diminished size and number of plaques in the brain of Amytrap-treated mice. Further, treated mice did not develop antibodies to Amytrap, suggesting Amytrap is non-immunogenic. Safety toxicological studies in rats showed that Amytrap was well tolerated and therefore safe (even at 50 X the efficacy dose). Stability tests showed Amytrap is stable at 4°C for up to one year. Efficacy and safety features make Amytrap a promising candidate for treating or modulating AD.
Collapse
|
162
|
Manolopoulos A, Andreadis P, Malandris K, Avgerinos I, Karagiannis T, Kapogiannis D, Tsolaki M, Tsapas A, Bekiari E. Intravenous Immunoglobulin for Patients With Alzheimer's Disease: A Systematic Review and Meta-Analysis. Am J Alzheimers Dis Other Demen 2019; 34:281-289. [PMID: 30987435 DOI: 10.1177/1533317519843720] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIM To assess the efficacy and safety of intravenous immunoglobulin (IVIg) for patients with Alzheimer's disease (AD). MATERIALS AND METHODS We searched electronic databases and other sources for randomized controlled trials comparing IVIg with placebo or other treatment for adults with AD. Primary outcome was change from baseline in Alzheimer's Disease Assessment Scale-Cognitive subscale (ADAS-Cog). RESULTS Five placebo-controlled trials were included in the meta-analysis. Compared to placebo, IVIg 0.2 and 0.4 g/kg once every two weeks did not change ADAS-Cog score (weighted mean difference: 0.37, 95% confidence interval: -1.46 to 2.20 and 0.77, -1.34 to 2.88, respectively). Furthermore, except for an increase in the incidence of rash, IVIg did not affect the incidence of other adverse events. CONCLUSION IVIg, albeit safe, is inefficacious for treatment of patients with AD. Future trials targeting earlier stages of disease or applying different dosing regimens may be warranted to clarify its therapeutic potential.
Collapse
Affiliation(s)
- Apostolos Manolopoulos
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Panagiotis Andreadis
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Konstantinos Malandris
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Ioannis Avgerinos
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Thomas Karagiannis
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Dimitrios Kapogiannis
- 2 Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, Bethesda, MD, USA
| | - Magda Tsolaki
- 3 First Department of Neurology, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Apostolos Tsapas
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece.,4 Harris Manchester College, University of Oxford, Oxford, United Kingdom
| | - Eleni Bekiari
- 1 Clinical Research and Evidence-Based Medicine Unit, Second Medical Department, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| |
Collapse
|
163
|
Zhao L, Cheng X, Zhong C. Implications of Successful Symptomatic Treatment in Parkinson's Disease for Therapeutic Strategies of Alzheimer's Disease. ACS Chem Neurosci 2019; 10:922-930. [PMID: 30474958 DOI: 10.1021/acschemneuro.8b00450] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Alzheimer's disease (AD) has been a devastating neurodegenerative disorder and lacks effective treatment to improve the prognosis for patients. Symptomatic treatment for AD mainly includes two categories: Acetylcholinesterase inhibitors and the N-methyl-d-aspartate (NMDA) receptor antagonist (memantine). They cannot significantly improve the quality of life and extend survival time for AD patients. Worse, almost all clinical trials for disease-modifying drugs have failed, and the reduction of brain β-amyloid (Aβ) deposition by multiple approaches, including inhibitors of β- or γ-secretase, vaccines, and antibodies against Aβ deposition, was found to have little effect on AD progression. A new therapeutic strategy for AD is urgently needed. Parkinson's disease also is a neurodegenerative disease having no effective treatment for modifying the disease. Nevertheless, successful symptomatic treatment using the combined therapies of l-DOPA supplement and modulators of l-DOPA metabolism greatly improves the prognosis for PD patients; the average survival time of the patient has been extended from 3-4 years to 10-15 years although dopaminergic neurons are still progressively decreasing. This provides useful implications for AD therapeutic strategies. AD patients manifest global cognitive decline, prominently represented by memory deficit, especially in the early stages of the disease. Further, the degree of decreased cognitive abilities correlates with cholinergic dysfunction and the hypometabolism of glucose, the dominant energy fuel for brain. Thus, the amelioration of brain cholinergic function and brain energy metabolism may be effective treatment to improve cognitive abilities of AD patients. Here, we highlighted the explorations of symptomatic therapeutics through modulating brain cholinergic function and energy metabolism in AD.
Collapse
Affiliation(s)
- Lei Zhao
- Department of Neurology, Shanghai Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200111, China
| | - Xiaoqin Cheng
- Department of Neurology, Zhongshan Hospital, The State Key Laboratory of Medical Neurobiology, The Institute of Brain Science, Fudan University, Shanghai 200032, China
| | - Chunjiu Zhong
- Department of Neurology, Zhongshan Hospital, The State Key Laboratory of Medical Neurobiology, The Institute of Brain Science, Fudan University, Shanghai 200032, China
| |
Collapse
|
164
|
Mullane K, Williams M. The de-Alzheimerization of age-related dementias: implications for drug targets and approaches to effective therapeutics. Curr Opin Pharmacol 2019; 44:62-75. [PMID: 30795894 DOI: 10.1016/j.coph.2019.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 01/10/2019] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD) was differentiated from senile dementia (SD) in 1910 due to its early onset and pathological severity. In 1976, this distinction was upended when SD was redesignated as AD to focus efforts and funding in dementia-related research. AD then became conflated with amyloid plaques and, to a lesser degree, neurofibrillary tangles complicating efforts in understanding dementia causality and its treatment. The resultant four-decade search for therapies-based almost exclusively on amyloid was an exercise in futility. While dementia is a complex, multifactorial syndrome, AD is viewed as a homogeneous, linear disease. An amyloid-agnostic approach is necessary to discover therapeutics for age-related dementias.
Collapse
Affiliation(s)
- Kevin Mullane
- Gladstone Institutes, San Francisco, CA, United States
| | - Michael Williams
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, OH, United States.
| |
Collapse
|
165
|
Nikseresht S, Bush AI, Ayton S. Treating Alzheimer's disease by targeting iron. Br J Pharmacol 2019; 176:3622-3635. [PMID: 30632143 DOI: 10.1111/bph.14567] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 10/14/2018] [Accepted: 11/27/2018] [Indexed: 12/30/2022] Open
Abstract
No disease modifying drugs have been approved for Alzheimer's disease despite recent major investments by industry and governments throughout the world. The burden of Alzheimer's disease is becoming increasingly unsustainable, and given the last decade of clinical trial failures, a renewed understanding of the disease mechanism is called for, and trialling of new therapeutic approaches to slow disease progression is warranted. Here, we review the evidence and rational for targeting brain iron in Alzheimer's disease. Although iron elevation in Alzheimer's disease was reported in the 1950s, renewed interest has been stimulated by the advancement of fluid and imaging biomarkers of brain iron that predict disease progression, and the recent discovery of the iron-dependent cell death pathway termed ferroptosis. We review these emerging clinical and biochemical findings and propose how this pathway may be targeted therapeutically to slow Alzheimer's disease progression. LINKED ARTICLES: This article is part of a themed section on Therapeutics for Dementia and Alzheimer's Disease: New Directions for Precision Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.18/issuetoc.
Collapse
Affiliation(s)
- Sara Nikseresht
- The Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Ashley I Bush
- The Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| | - Scott Ayton
- The Melbourne Dementia Research Centre, The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
166
|
Dong Y, Li X, Cheng J, Hou L. Drug Development for Alzheimer's Disease: Microglia Induced Neuroinflammation as a Target? Int J Mol Sci 2019; 20:E558. [PMID: 30696107 PMCID: PMC6386861 DOI: 10.3390/ijms20030558] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/09/2019] [Accepted: 01/13/2019] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most common causes of dementia. Its pathogenesis is characterized by the aggregation of the amyloid-β (Aβ) protein in senile plaques and the hyperphosphorylated tau protein in neurofibrillary tangles in the brain. Current medications for AD can provide temporary help with the memory symptoms and other cognitive changes of patients, however, they are not able to stop or reverse the progression of AD. New medication discovery and the development of a cure for AD is urgently in need. In this review, we summarized drugs for AD treatments and their recent updates, and discussed the potential of microglia induced neuroinflammation as a target for anti-AD drug development.
Collapse
Affiliation(s)
- Yuan Dong
- Department of Biochemistry, Medical College, Qingdao University, Qingdao 266071, China.
| | - Xiaoheng Li
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Jinbo Cheng
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Lin Hou
- Department of Biochemistry, Medical College, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
167
|
Panza F, Lozupone M, Logroscino G, Imbimbo BP. A critical appraisal of amyloid-β-targeting therapies for Alzheimer disease. Nat Rev Neurol 2019; 15:73-88. [DOI: 10.1038/s41582-018-0116-6] [Citation(s) in RCA: 459] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
168
|
Abstract
The most common neurodegenerative diseases are Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, frontotemporal lobar degeneration, and the motor neuron diseases, with AD affecting approximately 6% of people aged 65 years and older, and PD affecting approximately 1% of people aged over 60 years. Specific proteins are associated with these neurodegenerative diseases, as determined by both immunohistochemical studies on post-mortem tissue and genetic screening, where protein misfolding and aggregation are key hallmarks. Many of these proteins are shown to misfold and aggregate into soluble non-native oligomers and large insoluble protein deposits (fibrils and plaques), both of which may exert a toxic gain of function. Proteotoxicity has been examined intensively in cell culture and in in vivo models, and clinical trials of methods to attenuate proteotoxicity are relatively new. Therapies to enhance cellular protein quality control mechanisms such as upregulation of chaperones and clearance/degradation pathways, as well as immunotherapies against toxic protein conformations, are being actively pursued. In this article, we summarize the common pathophysiology of neurodegenerative disease, and review therapies in early-phase clinical trials that target the proteotoxic component of several neurodegenerative diseases.
Collapse
Affiliation(s)
- Luke McAlary
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, V6T 1Z1, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 2B5, Canada.
| | - Steven S Plotkin
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, V6T 1Z1, Canada.
- Genome Sciences and Technology Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada.
| | - Neil R Cashman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 2B5, Canada.
| |
Collapse
|
169
|
Cummings J. The Role of Biomarkers in Alzheimer's Disease Drug Development. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1118:29-61. [PMID: 30747416 DOI: 10.1007/978-3-030-05542-4_2] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Biomarkers have a key role in Alzheimer's disease (AD) drug development. Biomarkers can assist in diagnosis, demonstrate target engagement, support disease modification, and monitor for safety. The amyloid (A), tau (T), neurodegeneration (N) Research Framework emphasizes brain imaging and CSF measures relevant to disease diagnosis and staging and can be applied to drug development and clinical trials. Demonstration of target engagement in Phase 2 is critical before advancing a treatment candidate to Phase 3. Trials with biomarker outcomes are shorter and smaller than those required to show clinical benefit and are important to understanding the biological impact of an agent and inform go/no-go decisions. Companion diagnostics are required for safe and effective use of treatments and may emerge in AD drug development programs. Complementary biomarkers inform the use of therapies but are not mandatory for use. Biomarkers promise to de-risk AD drug development, attract sponsors to AD research, and accelerate getting new drugs to those with or at risk for AD.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA.
| |
Collapse
|
170
|
Pradier L, Blanchard-Brégeon V, Bohme A, Debeir T, Menager J, Benoit P, Barneoud P, Taupin V, Bertrand P, Dugay P, Cameron B, Shi Y, Naimi S, Duchesne M, Gagnaire M, Weeden T, Travaline T, Reczek D, Khiroug L, Slaoui M, Brunel P, Fukuyama H, Ravetch J, Canton T, Cohen C. SAR228810: an antibody for protofibrillar amyloid β peptide designed to reduce the risk of amyloid-related imaging abnormalities (ARIA). ALZHEIMERS RESEARCH & THERAPY 2018; 10:117. [PMID: 30486882 PMCID: PMC6264593 DOI: 10.1186/s13195-018-0447-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 11/04/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND Anti-amyloid β (Aβ) immunotherapy represents a major area of drug development for Alzheimer's disease (AD). However, Aβ peptide adopts multiple conformations and the pathological forms to be specifically targeted have not been identified. Aβ immunotherapy-related vasogenic edema has also been severely dose limiting for antibodies with effector functions binding vascular amyloid such as bapineuzumab. These two factors might have contributed to the limited efficacy demonstrated so far in clinical studies. METHODS To address these limitations, we have engineered SAR228810, a humanized monoclonal antibody (mAb) with limited Fc effector functions that binds specifically to soluble protofibrillar and fibrillar forms of Aβ peptide and we tested it together with its murine precursor SAR255952 in vitro and in vivo. RESULTS Unlike gantenerumab and BAN2401, SAR228810 and SAR255952 do not bind to Aβ monomers, low molecular weight Aβ oligomers or, in human brain sections, to Aβ diffuse deposits which are not specific of AD pathology. Both antibodies prevent Aβ42 oligomer neurotoxicity in primary neuronal cultures. In vivo, SAR255952, a mouse aglycosylated IgG1, dose-dependently prevented brain amyloid plaque formation and plaque-related inflammation with a minimal active dose of 3 mg/kg/week by the intraperitoneal route. No increase in plasma Aβ levels was observed with SAR255952 treatment, in line with its lack of affinity for monomeric Aβ. The effects of SAR255952 translated into synaptic functional improvement in ex-vivo hippocampal slices. Brain penetration and decoration of cerebral amyloid plaques was documented in live animals and postmortem. SAR255952 (up to 50 mg/kg/week intravenously) did not increase brain microhemorrhages and/or microscopic changes in meningeal and cerebral arteries in old APPSL mice while 3D6, the murine version of bapineuzumab, did. In immunotolerized mice, the clinical candidate SAR228810 demonstrated the same level of efficacy as the murine SAR255952. CONCLUSION Based on the improved efficacy/safety profile in non-clinical models of SAR228810, a first-in-man single and multiple dose administration clinical study has been initiated in AD patients.
Collapse
Affiliation(s)
- Laurent Pradier
- Sanofi R&D Neuroscience Unit, Sanofi, 1Av P. Brossolette, 91385, Chilly-Mazarin, France.
| | | | - Andrees Bohme
- Sanofi R&D Neuroscience Unit, Sanofi, 1Av P. Brossolette, 91385, Chilly-Mazarin, France
| | - Thomas Debeir
- Sanofi R&D Neuroscience Unit, Sanofi, 1Av P. Brossolette, 91385, Chilly-Mazarin, France
| | - Jean Menager
- Sanofi R&D Neuroscience Unit, Sanofi, 1Av P. Brossolette, 91385, Chilly-Mazarin, France
| | - Patrick Benoit
- Sanofi R&D Neuroscience Unit, Sanofi, 1Av P. Brossolette, 91385, Chilly-Mazarin, France
| | - Pascal Barneoud
- Sanofi R&D Neuroscience Unit, Sanofi, 1Av P. Brossolette, 91385, Chilly-Mazarin, France
| | - Véronique Taupin
- Sanofi R&D Neuroscience Unit, Sanofi, 1Av P. Brossolette, 91385, Chilly-Mazarin, France
| | - Philippe Bertrand
- Sanofi R&D Neuroscience Unit, Sanofi, 1Av P. Brossolette, 91385, Chilly-Mazarin, France
| | - Philippe Dugay
- Sanofi R&D Neuroscience Unit, Sanofi, 1Av P. Brossolette, 91385, Chilly-Mazarin, France
| | | | - Yi Shi
- Sanofi R&D Biotherapeutics Research, Vitry s/Seine, France
| | - Souad Naimi
- Sanofi R&D Biotherapeutics Research, Vitry s/Seine, France
| | - Marc Duchesne
- Sanofi R&D Biotherapeutics Research, Vitry s/Seine, France
| | - Marie Gagnaire
- Sanofi R&D Biotherapeutics Research, Vitry s/Seine, France
| | - Tim Weeden
- Sanofi R&D Biotherapeutics Research, Framingham, USA.,Present address: Dyne Therapeutics, Inc., 400 Technology Square, Cambridge, USA
| | | | - David Reczek
- Sanofi R&D Biotherapeutics Research, Framingham, USA
| | | | | | | | - Hidehiro Fukuyama
- The Rockefeller University, New-York City, USA.,Present address: Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | | | - Thierry Canton
- Sanofi R&D Neuroscience Unit, Sanofi, 1Av P. Brossolette, 91385, Chilly-Mazarin, France
| | - Caroline Cohen
- Sanofi R&D Neuroscience Unit, Sanofi, 1Av P. Brossolette, 91385, Chilly-Mazarin, France
| |
Collapse
|
171
|
Doggrell SA. Grasping at straws: the failure of solanezumab to modify mild Alzheimer's disease. Expert Opin Biol Ther 2018; 18:1189-1192. [PMID: 30376649 DOI: 10.1080/14712598.2018.1543397] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
INTRODUCTION The amyloid-beta (Aβ) cascade hypothesis is that reducing Aβ levels in the brain will be beneficial in the treatment of Alzheimer's disease. Solanezumab is a humanized analog of a murine antibody that selectively targets the central domain of the soluble form of Aβ. In the EXPEDITION 1 and 2 Phase 3 clinical trials, solanezumab was shown to be ineffective in subjects with mild-to-moderate Alzheimer's disease, and to have no effect on brain Aβ burden. Areas covered: This evaluation considers the secondary analysis of (EXPEDITION 1 and 2), which led to the EXPEDITION 3 trial of solanezumab in subjects with mild Alzheimer's disease, and the results of EXPEDITION 3. Expert opinion: The secondary analysis of EXPEDITION 1 and 2 was limited to mild Alzheimer's disease, and showed improvements on some scales, but not others. This analysis did not report data on Aβ burden. In my opinion, this was a questionable basis to undertake a further phase trial with solanezumab. The EXPEDITION 3 trial of solanezumab in subjects with mild Alzheimer's disease was terminated early for ineffectiveness. With hindsight, solanezumab should have been discontinued after EXPEDITION 1 and 2, especially as it had not been shown to reduce Aβ burden.
Collapse
Affiliation(s)
- Sheila A Doggrell
- a Faculty of Health , Queensland University of Technology , Brisbane , Australia
| |
Collapse
|
172
|
Salloway S, Honigberg LA, Cho W, Ward M, Friesenhahn M, Brunstein F, Quartino A, Clayton D, Mortensen D, Bittner T, Ho C, Rabe C, Schauer SP, Wildsmith KR, Fuji RN, Suliman S, Reiman EM, Chen K, Paul R. Amyloid positron emission tomography and cerebrospinal fluid results from a crenezumab anti-amyloid-beta antibody double-blind, placebo-controlled, randomized phase II study in mild-to-moderate Alzheimer's disease (BLAZE). ALZHEIMERS RESEARCH & THERAPY 2018; 10:96. [PMID: 30231896 PMCID: PMC6146627 DOI: 10.1186/s13195-018-0424-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 08/29/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND We investigated the effect of crenezumab, a humanized anti-amyloid-beta (Aβ) immunoglobulin (Ig)G4 monoclonal antibody, on biomarkers of amyloid pathology, neurodegeneration, and disease progression in patients with mild-to-moderate Alzheimer's disease (AD). METHODS This double-blind, placebo-controlled, randomized phase II study enrolled patients with mild-to-moderate AD and a Mini-Mental State Examination (MMSE) score of 18-26. In part 1 of the study, patients were 2:1 randomized to receive low-dose subcutaneous (SC) 300 mg crenezumab every 2 weeks (q2w) or placebo for 68 weeks; in part 2, patients were 2:1 randomized to receive high-dose intravenous (IV) 15 mg/kg crenezumab every 4 weeks (q4w) or placebo for 68 weeks. The primary endpoint was change in amyloid burden from baseline to week 69 assessed by florbetapir positron emission tomography (PET) in the modified intent-to-treat population. Secondary endpoints were change from baseline to week 69 in cerebrospinal fluid (CSF) biomarkers and fluorodeoxyglucose PET, and change from baseline to week 73 in 12-point Alzheimer's Disease Assessment Scale cognitive subscale (ADAS-Cog12) and Clinical Dementia Rating Sum of Boxes (CDR-SB). Safety was assessed in patients who received at least one dose of study treatment. RESULTS From August 2011 to September 2012, 91 patients were enrolled and randomized (low-dose SC cohort: crenezumab (n = 26) or placebo (n = 13); high-dose IV cohort: crenezumab (n = 36) or placebo (n = 16)). The primary endpoint was not met using a prespecified cerebellar reference region to calculate standard uptake value ratios (SUVRs) from florbetapir PET. Exploratory analyses using subcortical white matter reference regions showed nonsignificant trends toward slower accumulation of plaque amyloid in the high-dose IV cohort. In both cohorts, a significant mean increase from baseline in CSF Aβ(1-42) levels versus placebo was observed. Nonsignificant trends toward ADAS-Cog12 and CDR-SB benefits were identified in a mild (MMSE 20-26) subset of the high-dose IV cohort. No amyloid-related imaging abnormalities due to edema/effusion were observed. CONCLUSION The primary endpoint was not met. Exploratory findings suggest potential Aβ target engagement with crenezumab and possible slower accumulation of plaque amyloid. Studies investigating the effects of higher doses of crenezumab on amyloid load and disease progression are ongoing. TRIAL REGISTRATION ClinicalTrials.gov, NCT01397578 . Registered on 18 July 2011.
Collapse
Affiliation(s)
- Stephen Salloway
- Department of Neurology and Psychiatry, The Warren Alpert Medical School of Brown University, 345 Blackstone Boulevard, Providence, RI, 2906, USA.
| | | | - William Cho
- Genentech Inc., South San Francisco, CA, USA
| | | | | | | | | | | | | | | | - Carole Ho
- Genentech Inc., South San Francisco, CA, USA
| | | | | | | | | | | | | | - Kewei Chen
- Banner Alzheimer's Institute, Phoenix, AZ, USA
| | - Robert Paul
- Genentech Inc., South San Francisco, CA, USA
| |
Collapse
|
173
|
Herline K, Drummond E, Wisniewski T. Recent advancements toward therapeutic vaccines against Alzheimer's disease. Expert Rev Vaccines 2018; 17:707-721. [PMID: 30005578 DOI: 10.1080/14760584.2018.1500905] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a devastating neurodegenerative disease characterized by protein aggregates of amyloid β (Aβ) and tau. These proteins have normal physiological functions, but in AD, they undergo a conformational change and aggregate as toxic oligomeric and fibrillar species with a high β-sheet content. AREAS COVERED Active and passive immunotherapeutic approaches are among the most attractive methods for targeting misfolded Aβ and tau. Promising preclinical testing of various immunotherapeutic approaches has yet to translate to cognitive benefits in human clinical trials. Knowledge gained from these past failures has led to the development of second-generation Aβ-active immunotherapies, anti-Aβ monoclonal antibodies targeting a wide array of Aβ conformations, and to a number of immunotherapies targeting pathological tau. This review covers the more recent advances in vaccine development for AD from 2016 to present. EXPERT COMMENTARY Due to the complex pathophysiology of AD, greatest clinical efficacy will most likely be achieved by concurrently targeting the most toxic forms of both Aβ and tau.
Collapse
Affiliation(s)
- Krystal Herline
- a Center for Cognitive Neurology , New York University School of Medicine , New York , NY , USA.,b Departments of Neurology , New York University School of Medicine , New York , NY , USA
| | - Eleanor Drummond
- a Center for Cognitive Neurology , New York University School of Medicine , New York , NY , USA.,b Departments of Neurology , New York University School of Medicine , New York , NY , USA
| | - Thomas Wisniewski
- a Center for Cognitive Neurology , New York University School of Medicine , New York , NY , USA.,b Departments of Neurology , New York University School of Medicine , New York , NY , USA.,c Pathology , New York University School of Medicine , New York , NY , USA.,d Psychiatry , New York University School of Medicine , New York , NY , USA
| |
Collapse
|