1
|
Tan W, Thiruppathi J, Hong SH, Puth S, Pheng S, Mun BR, Choi WS, Lee KH, Park HS, Nguyen DT, Lee MC, Jeong K, Zheng JH, Kim Y, Lee SE, Rhee JH. Development of an anti-tauopathy mucosal vaccine specifically targeting pathologic conformers. NPJ Vaccines 2024; 9:108. [PMID: 38879560 PMCID: PMC11180213 DOI: 10.1038/s41541-024-00904-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 05/28/2024] [Indexed: 06/19/2024] Open
Abstract
Alzheimer's disease (AD) and related tauopathies are associated with pathological tau protein aggregation, which plays an important role in neurofibrillary degeneration and dementia. Targeted immunotherapy to eliminate pathological tau aggregates is known to improve cognitive deficits in AD animal models. The tau repeat domain (TauRD) plays a pivotal role in tau-microtubule interactions and is critically involved in the aggregation of hyperphosphorylated tau proteins. Because TauRD forms the structural core of tau aggregates, the development of immunotherapies that selectively target TauRD-induced pathological aggregates holds great promise for the modulation of tauopathies. In this study, we generated recombinant TauRD polypeptide that form neurofibrillary tangle-like structures and evaluated TauRD-specific immune responses following intranasal immunization in combination with the mucosal adjuvant FlaB. In BALB/C mice, repeated immunizations at one-week intervals induced robust TauRD-specific antibody responses in a TLR5-dependent manner. Notably, the resulting antiserum recognized only the aggregated form of TauRD, while ignoring monomeric TauRD. The antiserum effectively inhibited TauRD filament formation and promoted the phagocytic degradation of TauRD aggregate fragments by microglia. The antiserum also specifically recognized pathological tau conformers in the human AD brain. Based on these results, we engineered a built-in flagellin-adjuvanted TauRD (FlaB-TauRD) vaccine and tested its efficacy in a P301S transgenic mouse model. Mucosal immunization with FlaB-TauRD improved quality of life, as indicated by the amelioration of memory deficits, and alleviated tauopathy progression. Notably, the survival of the vaccinated mice was dramatically extended. In conclusion, we developed a mucosal vaccine that exclusively targets pathological tau conformers and prevents disease progression.
Collapse
Affiliation(s)
- Wenzhi Tan
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Jayalakshmi Thiruppathi
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Seol Hee Hong
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sao Puth
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Sophea Pheng
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Bo-Ram Mun
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Won-Seok Choi
- School of Biological Sciences and Technology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Kyung-Hwa Lee
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Pathology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Hyun-Sun Park
- Department of Pharmacology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Duc Tien Nguyen
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Min-Cheol Lee
- Department of Pathology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Seegene Inc, Seoul, 05548, Republic of Korea
| | - Kwangjoon Jeong
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea
| | - Jin Hai Zheng
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410082, China
| | - Young Kim
- Department of Oral Pathology, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Shee Eun Lee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea.
- National Immunotherapy Innovation Center, Hwasun-gun, Jeonnam, 58128, Republic of Korea.
- Department of Pharmacology and Dental Therapeutics, School of Dentistry, Chonnam National University, Gwangju, 61186, Republic of Korea.
| | - Joon Haeng Rhee
- Clinical Vaccine R&D Center, Chonnam National University, Hwasun-gun, Jeonnam, 58128, Republic of Korea.
- Department of Microbiology, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea.
- Combinatorial Tumor Immunotherapy MRC, Chonnam National University Medical School, Hwasun-gun, Jeonnam, 58128, Republic of Korea.
| |
Collapse
|
2
|
Hsueh SCC, Nijland M, Aina A, Plotkin SS. Cyclization Scaffolding for Improved Vaccine Immunogen Stability: Application to Tau Protein in Alzheimer's Disease. J Chem Inf Model 2024; 64:2035-2044. [PMID: 38427576 DOI: 10.1021/acs.jcim.3c01556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2024]
Abstract
Effective scaffolding of immunogens is crucial for generating conformationally selective antibodies through active immunization, particularly in the treatment of protein misfolding diseases such as Alzheimer's and Parkinson's disease. Previous computational work has revealed that a disorder-prone region of the tau protein, when in a stacked form, is predicted to structurally resemble a small, soluble protofibril, having conformational properties similar to those of experimental in vitro tau oligomers. Such an oligomeric structural mimic has the potential to serve as a vaccine immunogen design for Alzheimer's disease. In this study, we developed a cyclization scaffolding method in Rosetta, in which multiple cyclic peptides are stacked into a protofibril. Cyclization results in significant stabilization of protofibril-like structures by constraining the conformational space. Applying this method to the disorder-prone region of the tau fibril, we evaluated the metastability of the cyclized tau immunogen using molecular dynamics simulations, and we identified sequences of two cyclic constructs having high metastability in the protofibril. We then assessed their thermodynamic stability by computing the free energy required to separate a distal chain from the rest of the stacked structure. Our computational results, based on molecular dynamics simulations and free energy calculations, demonstrate that two cyclized constructs, cyclo-(VKSEKLDFKDRVQSKIFyN) and cyclo-(VKSEKLDFKDRVQSKIYvG) (lowercase letters indicate d-form amino acids), possess significantly increased thermodynamic stability in the protofibril over an uncyclized linear construct VKSEKLDFKDRVQSKI. The cyclization scaffolding approach proposed here holds promise as a means to effectively design immunogens for protein misfolding diseases, particularly those involving liposome-conjugated peptide constructs.
Collapse
Affiliation(s)
- Shawn C C Hsueh
- Department of Physics and Astronomy, The University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Mark Nijland
- Laboratory of Physical Chemistry, Wageningen University, Wageningen 6708 WG, The Netherlands
| | - Adekunle Aina
- Department of Physics and Astronomy, The University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| | - Steven S Plotkin
- Department of Physics and Astronomy, The University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
- Genome Science and Technology Program, The University of British Columbia, Vancouver, British Columbia V6T 1Z1, Canada
| |
Collapse
|
3
|
Liu N, Liang X, Chen Y, Xie L. Recent trends in treatment strategies for Alzheimer 's disease and the challenges: A topical advancement. Ageing Res Rev 2024; 94:102199. [PMID: 38232903 DOI: 10.1016/j.arr.2024.102199] [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: 11/14/2023] [Accepted: 01/12/2024] [Indexed: 01/19/2024]
Abstract
Alzheimer's Disease (AD) is an irreversible and progressive neurological disease that has affected at least 50 million people around the globe. Considering the severity of the disease and the continuous increase in the number of patients, the development of new effective drugs or intervention strategies for AD has become urgent. AD is caused by a combination of genetic, environmental, and lifestyle factors, but its exact cause has not yet been clarified. Given the current challenges being faced in the clinical treatment of AD, such as complex AD pathological network and insufficient early diagnosis, herein, we have focused on the three core pathological features of AD, including amyloid-β (Aβ) aggregation, tau phosphorylation and tangles, and activation of inflammatory factors. In this review, we have briefly underscored the primary evidence supporting each pathology and discuss AD pathological network among Aβ, tau, and inflammation. We have also comprehensively summarized the most instructive drugs and their treatment strategies against Aβ, tau, or neuroinflammation used in basic research and clinical trials. Finally, we have discussed and outlined the pros and cons of each pathological approach and looked forward to potential personalized diagnosis and treatment strategies that are beneficial to AD patients.
Collapse
Affiliation(s)
- Ni Liu
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Xiaohan Liang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Yu Chen
- College of Public Health, Zhengzhou University, Zhengzhou 450000, China.
| | - Lihang Xie
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| |
Collapse
|
4
|
Gharat R, Dixit G, Khambete M, Prabhu A. Targets, trials and tribulations in Alzheimer therapeutics. Eur J Pharmacol 2024; 962:176230. [PMID: 38042464 DOI: 10.1016/j.ejphar.2023.176230] [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: 08/22/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/04/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by abnormal accumulation of extracellular amyloid beta senile plaques and intracellular neurofibrillary tangles in the parts of the brain responsible for cognition. The therapeutic burden for the management of AD relies solely on cholinesterase inhibitors that provide only symptomatic relief. The urgent need for disease-modifying drugs has resulted in intensive research in this domain, which has led to better understanding of the disease pathology and identification of a plethora of new pathological targets. Currently, there are over a hundred and seventy clinical trials exploring disease modification, cognitive enhancement, and reduction of neuro-psychiatric complications. However, the path to developing safe and efficacious AD therapeutics has not been without challenges. Several clinical trials have been terminated in advanced stages due to lack of therapeutic translation or increased incidence of adverse events. This review presents an in-depth look at the various therapeutic targets of AD and the lessons learnt during their clinical assessment. Comprehensive understanding of the implication of modulating various aspects of Alzheimer brain pathology is crucial for development of drugs with potential to halt disease progression in Alzheimer therapeutics.
Collapse
Affiliation(s)
- Ruchita Gharat
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, VM Road, Vile Parle (West), Mumbai, 400056, Maharashtra, India
| | - Gargi Dixit
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, VM Road, Vile Parle (West), Mumbai, 400056, Maharashtra, India
| | - Mihir Khambete
- Department of Chemistry, Yale University, New Haven, CT, USA
| | - Arati Prabhu
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, VM Road, Vile Parle (West), Mumbai, 400056, Maharashtra, India.
| |
Collapse
|
5
|
Stefanik O, Majerova P, Kovac A, Mikus P, Piestansky J. Capillary electrophoresis in the analysis of therapeutic peptides-A review. Electrophoresis 2024; 45:120-164. [PMID: 37705480 DOI: 10.1002/elps.202300141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/10/2023] [Accepted: 08/14/2023] [Indexed: 09/15/2023]
Abstract
Therapeutic peptides are a growing class of innovative drugs with high efficiency and a low risk of adverse effects. These biomolecules fall within the molecular mass range between that of small molecules and proteins. However, their inherent instability and potential for degradation underscore the importance of reliable and effective analytical methods for pharmaceutical quality control, therapeutic drug monitoring, and compliance testing. Liquid chromatography-mass spectrometry (LC-MS) has long time been the "gold standard" conventional method for peptide analysis, but capillary electrophoresis (CE) is increasingly being recognized as a complementary and, in some cases, superior, highly efficient, green, and cost-effective alternative technique. CE can separate peptides composed of different amino acids owing to differences in their net charge and size, determining their migration behavior in an electric field. This review provides a comprehensive overview of therapeutic peptides that have been used in the clinical environment for the last 25 years. It describes the properties, classification, current trends in development, and clinical use of therapeutic peptides. From the analytical point of view, it discusses the challenges associated with the analysis of therapeutic peptides in pharmaceutical and biological matrices, as well as the evaluation of CE as a whole and the comparison with LC methods. The article also highlights the use of microchip electrophoresis, nonaqueous CE, and nonconventional hydrodynamically closed CE systems and their applications. Overall, the article emphasizes the importance of developing new CE-based analytical methods to ensure the high quality, safety, and efficacy of therapeutic peptides in clinical practice.
Collapse
Affiliation(s)
- Ondrej Stefanik
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Petra Majerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Andrej Kovac
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
| | - Peter Mikus
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
| | - Juraj Piestansky
- Toxicological and Antidoping Center, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovak Republic
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University in Bratislava, Bratislava, Slovak Republic
| |
Collapse
|
6
|
Sun Y, Zhao J, Lu Y, Ngo FY, Shuai B, Zhang ZJ, Feng Y, Rong J. In Silico Prediction of Quercetin Analogs for Targeting Death-Associated Protein Kinase 1 (DAPK1) Against Alzheimer's Disease. Curr Neuropharmacol 2024; 22:2353-2367. [PMID: 38752632 PMCID: PMC11451310 DOI: 10.2174/1570159x22666240515090434] [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: 11/08/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 10/06/2024] Open
Abstract
Alzheimer's Disease (AD) is a progressive neurodegenerative disorder that greatly affects the health and life quality of the elderly population. Existing drugs mainly alleviate symptoms but fail to halt disease progression, underscoring the urgent need for the development of novel drugs. Based on the neuroprotective effects of flavonoid quercetin in AD, this study was designed to identify potential AD-related targets for quercetin and perform in silico prediction of promising analogs for the treatment of AD. Database mining suggested death-associated protein kinase 1 (DAPK1) as the most promising AD-related target for quercetin among seven protein candidates. To achieve better biological effects for the treatment of AD, we devised a series of quercetin analogs as ligands for DAPK1, and molecular docking analyses, absorption, distribution, metabolism, and excretion (ADME) predictions, as well as molecular dynamics (MD) simulations, were performed. The energy for drug-protein interaction was predicted and ranked. As a result, quercetin-A1a and quercetin-A1a1 out of 19 quercetin analogs exhibited the lowest interaction energy for binding to DAPK1 than quercetin, and they had similar dynamics performance with quercetin. In addition, quercetin-A1a and quercetin-A1a1 were predicted to have better water solubility. Thus, quercetin-A1a and quercetin-A1a1 could be promising agents for the treatment of AD. Our findings paved the way for further experimental studies and the development of novel drugs.
Collapse
Affiliation(s)
- Yilu Sun
- Department of Chinese Medicine, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
- School of Chinese Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, China
| | - Jia Zhao
- Department of Chinese Medicine, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
- School of Chinese Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, China
- Zhu Nansun’s Workstation and Yu Jin’s Workstation, School of Chinese Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, China
| | - Yizhu Lu
- School of Chinese Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, China
| | - Fung Yin Ngo
- School of Chinese Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, China
| | - Bo Shuai
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang-Jin Zhang
- Department of Chinese Medicine, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
- School of Chinese Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, China
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, China
| | - Jianhui Rong
- School of Chinese Medicine, The University of Hong Kong, 3 Sassoon Road, Pokfulam, Hong Kong, China
| |
Collapse
|
7
|
Davidson R, Krider RI, Borsellino P, Noorda K, Alhwayek G, Vida TA. Untangling Tau: Molecular Insights into Neuroinflammation, Pathophysiology, and Emerging Immunotherapies. Curr Issues Mol Biol 2023; 45:8816-8839. [PMID: 37998730 PMCID: PMC10670294 DOI: 10.3390/cimb45110553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 10/26/2023] [Accepted: 10/30/2023] [Indexed: 11/25/2023] Open
Abstract
Neuroinflammation, a core pathological feature observed in several neurodegenerative diseases, including Alzheimer's disease (AD), is rapidly gaining attention as a target in understanding the molecular underpinnings of these disorders. Glial cells, endothelial cells, peripheral immune cells, and astrocytes produce a variety of pro-inflammatory mediators that exacerbate the disease progression. Additionally, microglial cells play a complex role in AD, facilitating the clearance of pathological amyloid-beta peptide (Aβ) plaques and aggregates of the tau protein. Tau proteins, traditionally associated with microtubule stabilization, have come under intense scrutiny for their perturbed roles in neurodegenerative conditions. In this narrative review, we focus on recent advances from molecular insights that have revealed aberrant tau post-translational modifications, such as phosphorylation and acetylation, serving as pathological hallmarks. These modifications also trigger the activation of CNS-resident immune cells, such as microglia and astrocytes substantially contributing to neuroinflammation. This intricate relationship between tau pathologies and neuroinflammation fosters a cascading impact on neural pathophysiology. Furthermore, understanding the molecular mechanisms underpinning tau's influence on neuroinflammation presents a frontier for the development of innovative immunotherapies. Neurodegenerative diseases have been relatively intractable to conventional pharmacology using small molecules. We further comprehensively document the many alternative approaches using immunotherapy targeting tau pathological epitopes and structures with a wide array of antibodies. Clinical trials are discussed using these therapeutic approaches, which have both promising and disappointing outcomes. Future directions for tau immunotherapies may include combining treatments with Aβ immunotherapy, which may result in more significant clinical outcomes for neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas A. Vida
- Kirk Kerkorian School of Medicine at UNLV, 625 Shadow Lane, Las Vegas, NV 89106, USA; (R.D.); (R.I.K.); (P.B.); (K.N.); (G.A.)
| |
Collapse
|
8
|
Meskova K, Martonova K, Hrasnova P, Sinska K, Skrabanova M, Fialova L, Njemoga S, Cehlar O, Parmar O, Kolenko P, Pevala V, Skrabana R. Cost-Effective Protein Production in CHO Cells Following Polyethylenimine-Mediated Gene Delivery Showcased by the Production and Crystallization of Antibody Fabs. Antibodies (Basel) 2023; 12:51. [PMID: 37606435 PMCID: PMC10443350 DOI: 10.3390/antib12030051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/02/2023] [Accepted: 08/03/2023] [Indexed: 08/23/2023] Open
Abstract
Laboratory production of recombinant mammalian proteins, particularly antibodies, requires an expression pipeline assuring sufficient yield and correct folding with appropriate posttranslational modifications. Transient gene expression (TGE) in the suspension-adapted Chinese Hamster Ovary (CHO) cell lines has become the method of choice for this task. The antibodies can be secreted into the media, which facilitates subsequent purification, and can be glycosylated. However, in general, protein production in CHO cells is expensive and may provide variable outcomes, namely in laboratories without previous experience. While achievable yields may be influenced by the nucleotide sequence, there are other aspects of the process which offer space for optimization, like gene delivery method, cultivation process or expression plasmid design. Polyethylenimine (PEI)-mediated gene delivery is frequently employed as a low-cost alternative to liposome-based methods. In this work, we are proposing a TGE platform for universal medium-scale production of antibodies and other proteins in CHO cells, with a novel expression vector allowing fast and flexible cloning of new genes and secretion of translated proteins. The production cost has been further reduced using recyclable labware. Nine days after transfection, we routinely obtain milligrams of antibody Fabs or human lactoferrin in a 25 mL culture volume. Potential of the platform is established based on the production and crystallization of antibody Fabs and their complexes.
Collapse
Affiliation(s)
- Klaudia Meskova
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
- Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Katarina Martonova
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
- Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Patricia Hrasnova
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Kristina Sinska
- AXON Neuroscience R&D Services SE, 811 02 Bratislava, Slovakia
| | - Michaela Skrabanova
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Lubica Fialova
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
- AXON Neuroscience R&D Services SE, 811 02 Bratislava, Slovakia
| | - Stefana Njemoga
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
- Faculty of Natural Sciences, Comenius University, 842 15 Bratislava, Slovakia
| | - Ondrej Cehlar
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Olga Parmar
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Petr Kolenko
- Faculty of Nuclear Sciences and Physical Engineering, Czech Technical University in Prague, 115 19 Prague, Czech Republic
| | - Vladimir Pevala
- Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| | - Rostislav Skrabana
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| |
Collapse
|
9
|
Imran Sajid M, Sultan Sheikh F, Anis F, Nasim N, Sumbria RK, Nauli SM, Kumar Tiwari R. siRNA drug delivery across the blood-brain barrier in Alzheimer's disease. Adv Drug Deliv Rev 2023; 199:114968. [PMID: 37353152 PMCID: PMC10528676 DOI: 10.1016/j.addr.2023.114968] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/29/2023] [Accepted: 06/19/2023] [Indexed: 06/25/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease with a few FDA-approved drugs that provide modest symptomatic benefits and only two FDA-approved disease-modifying treatments for AD. The advancements in understanding the causative genes and non-coding sequences at the molecular level of the pathophysiology of AD have resulted in several exciting research papers that employed small interfering RNA (siRNA)-based therapy. Although siRNA is being sought by academia and biopharma industries, several challenges still need to be addressed. We comprehensively report the latest advances in AD pathophysiology, druggable targets, ongoing clinical trials, and the siRNA-based approaches across the blood-brain barrier for addressing AD. This review describes the latest delivery systems employed to address this barrier. Critical insights and future perspectives on siRNA therapy for AD are also provided.
Collapse
Affiliation(s)
- Muhammad Imran Sajid
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University School of Pharmacy, Irvine, CA 92618, USA; Faculty of Pharmacy, University of Central Punjab, Lahore 54000, Pakistan
| | - Fahad Sultan Sheikh
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millat University, Islamabad 44000, Pakistan
| | - Faiza Anis
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Federal Urdu University of Arts, Science and Technology, Karachi, Pakistan
| | - Nourina Nasim
- Department of Chemistry and Chemical Engineering, Syed Baber Ali School of Science and Engineering, Lahore University of Management Sciences, 54792 Lahore, Pakistan
| | - Rachita K Sumbria
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University School of Pharmacy, Irvine, CA 92618, USA; Department of Neurology, University of California, Irvine, CA, 92868, USA
| | - Surya M Nauli
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University School of Pharmacy, Irvine, CA 92618, USA
| | - Rakesh Kumar Tiwari
- Department of Biomedical and Pharmaceutical Sciences, Harry and Diane Rinker Health Science Campus, Chapman University School of Pharmacy, Irvine, CA 92618, USA.
| |
Collapse
|
10
|
Parrocha CMT, Nowick JS. Current Peptide Vaccine and Immunotherapy Approaches Against Alzheimer's Disease. Pept Sci (Hoboken) 2023; 115:e24289. [PMID: 36778914 PMCID: PMC9916509 DOI: 10.1002/pep2.24289] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/11/2022] [Accepted: 06/11/2022] [Indexed: 11/06/2022]
Abstract
Peptide vaccines and immunotherapies against aggregating proteins involved in the pathogenesis and progression of Alzheimer's disease (AD) - the β-amyloid peptide (Aβ) and tau - are promising therapeutic avenues against AD. Two decades of effort has led to the controversial FDA approval of the monoclonal antibody Aducanumab (Aduhelm), which has subsequentially sparked the revival and expedited review of promising monoclonal antibody immunotherapies that target Aβ. In this review, we explore the development of Aβ and tau peptide vaccines and immunotherapies with monoclonal antibodies in clinical trials against AD.
Collapse
Affiliation(s)
| | - James S. Nowick
- Department of Pharmaceutical SciencesUniversity of California IrvineIrvineCaliforniaUSA
- Department of ChemistryUniversity of California IrvineIrvineCaliforniaUSA
| |
Collapse
|
11
|
Congdon EE, Pan R, Jiang Y, Sandusky-Beltran LA, Dodge A, Lin Y, Liu M, Kuo MH, Kong XP, Sigurdsson EM. Single domain antibodies targeting pathological tau protein: Influence of four IgG subclasses on efficacy and toxicity. EBioMedicine 2022; 84:104249. [PMID: 36099813 PMCID: PMC9475275 DOI: 10.1016/j.ebiom.2022.104249] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/15/2022] [Accepted: 08/19/2022] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Eleven tau immunoglobulin G (IgG) antibodies have entered clinical trials to treat tauopathies, including Alzheimer's disease, but it is unclear which IgG subclass/subtype has the ideal efficacy and safety profile. Only two subtypes, with or without effector function, have been examined in the clinic and not for the same tau antibody. The few preclinical studies on this topic have only compared two subtypes of one antibody each and have yielded conflicting results. METHODS We selected two single domain antibodies (sdAbs) derived from a llama immunized with tau proteins and utilized them to generate an array of Fc-(sdAb)2 subclasses containing identical tau binding domains but differing Fc region. Unmodified sdAbs and their IgG subclasses were tested for efficacy in primary cultures and in vivo microdialysis using JNPL3 tauopathy mice. FINDINGS Unmodified sdAbs were non-toxic, blocked tau toxicity and promoted tau clearance. However, the efficacy/safety profile of their Fc-(sdAb)2 subclasses varied greatly within and between sdAbs. For one of them, all its subtypes were non-toxic, only those with effector function cleared tau, and were more effective in vivo than unmodified sdAb. For the other sdAb, all its subtypes were toxic in tauopathy cultures but not in wild-type cells, suggesting that bivalent binding of its tau epitope stabilizes a toxic conformation of tau, with major implications for tau pathogenesis. Likewise, its subclasses were less effective than the unmodified sdAb in clearing tau in vivo. INTERPRETATION These findings indicate that tau antibodies with effector function are safe and better at clearing pathological tau than effectorless antibodies, Furthermore, tau antibodies can provide a valuable insight into tau pathogenesis, and some may aggravate it. FUNDING Funding for these studies was provided by the National Institute of Health (R01 AG032611, R01 NS077239, RF1 NS120488, R21 AG 069475, R21 AG 058282, T32AG052909), and the NYU Alzheimer's Disease Center Pilot Grant Program (via P30 AG008051).
Collapse
Affiliation(s)
- Erin E Congdon
- Department of Neuroscience and Physiology, and the Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Ruimin Pan
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Yixiang Jiang
- Department of Neuroscience and Physiology, and the Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Leslie A Sandusky-Beltran
- Department of Neuroscience and Physiology, and the Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Andie Dodge
- Department of Neuroscience and Physiology, and the Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Yan Lin
- Department of Neuroscience and Physiology, and the Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA
| | - Mengyu Liu
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, East Lansing, MI, 48824, USA
| | - Min-Hao Kuo
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, East Lansing, MI, 48824, USA
| | - Xiang-Peng Kong
- Department of Biochemistry and Molecular Pharmacology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, and the Neuroscience Institute, New York University Grossman School of Medicine, 435 East 30th Street, New York, NY 10016, USA; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
12
|
Li L, Miao J, Chu D, Jin N, Tung YC, Dai C, Hu W, Gong C, Iqbal K, Liu F. Tau antibody 77G7 targeting microtubule binding domain suppresses proteopathic tau to seed tau aggregation. CNS Neurosci Ther 2022; 28:2245-2259. [PMID: 36114722 PMCID: PMC9627375 DOI: 10.1111/cns.13970] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Neurofibrillary tangle (NFT) of hyperphosphorylated tau is a hallmark of Alzheimer's disease (AD) and related tauopathies. Tau lesion starts in the trans-entorhinal cortex, from where it spreads to limbic regions, followed by neocortical areas. The regional distribution of NFTs associates with the progression of AD. Accumulating evidence suggests that proteopathic tau can seed tau aggregation in a prion-like fashion in vitro and in vivo. Inhibition of tau seeding activity could provide a potential therapeutic opportunity to block the propagation of tau pathology in AD and related tauopathies. AIMS In the present study, we investigated the role of 77G7, a monoclonal tau antibody to the microtubule-binding repeats, in repressing the seeding activity of proteopathic tau. RESULTS We found that 77G7 had a higher affinity toward aggregated pathological tau fractions than un-aggregated tau derived from AD brain. 77G7 inhibited the internalization of tau aggregates by cells, blocked AD O-tau to capture normal tau, and to seed tau aggregation in vitro and in cultured cells. Tau pathology induced by hippocampal injection of AD O-tau in 3xTg-AD mice was suppressed by mixing 77G7 with AD O-tau. Intravenous administration of 77G7 ameliorated site-specific hyperphosphorylation of tau induced by AD O-tau in the hippocampi of Tg/hTau mice. CONCLUSION These findings indicate that 77G7 can effectively suppress the seeding activity of AD O-tau and thus could be developed as a potential immunotherapeutic drug to inhibit the propagation of tau pathology in AD and related tauopathies.
Collapse
Affiliation(s)
- Longfei Li
- Department of NeurochemistryInge Grundke‐Iqbal Research FloorNew York State Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNantong UniversityNantongChina
| | - Jin Miao
- Department of NeurochemistryInge Grundke‐Iqbal Research FloorNew York State Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA,Laboratory Animal CenterNantong UniversityNantongChina
| | - Dandan Chu
- Department of NeurochemistryInge Grundke‐Iqbal Research FloorNew York State Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNantong UniversityNantongChina
| | - Nana Jin
- Department of NeurochemistryInge Grundke‐Iqbal Research FloorNew York State Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of EducationNantong UniversityNantongChina
| | - Yunn Chyn Tung
- Department of NeurochemistryInge Grundke‐Iqbal Research FloorNew York State Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA
| | - Chun‐Ling Dai
- Department of NeurochemistryInge Grundke‐Iqbal Research FloorNew York State Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA
| | - Wen Hu
- Department of NeurochemistryInge Grundke‐Iqbal Research FloorNew York State Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA
| | - Cheng‐Xin Gong
- Department of NeurochemistryInge Grundke‐Iqbal Research FloorNew York State Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA
| | - Khalid Iqbal
- Department of NeurochemistryInge Grundke‐Iqbal Research FloorNew York State Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA
| | - Fei Liu
- Department of NeurochemistryInge Grundke‐Iqbal Research FloorNew York State Institute for Basic Research in Developmental DisabilitiesStaten IslandNew YorkUSA
| |
Collapse
|
13
|
Congdon EE, Jiang Y, Sigurdsson EM. Targeting tau only extracellularly is likely to be less efficacious than targeting it both intra- and extracellularly. Semin Cell Dev Biol 2022; 126:125-137. [PMID: 34896021 PMCID: PMC9680670 DOI: 10.1016/j.semcdb.2021.12.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 12/01/2021] [Accepted: 12/01/2021] [Indexed: 12/11/2022]
Abstract
Aggregation of the tau protein is thought to be responsible for the neurodegeneration and subsequent functional impairments in diseases that are collectively named tauopathies. Alzheimer's disease is the most common tauopathy, but the group consists of over 20 different diseases, many of which have tau pathology as their primary feature. The development of tau therapies has mainly focused on preventing the formation of and/or clearing these aggregates. Of these, immunotherapies that aim to either elicit endogenous tau antibodies or deliver exogenous ones are the most common approach in clinical trials. While their mechanism of action can involve several pathways, both extra- and intracellular, pharmaceutical companies have primarily focused on antibody-mediated clearance of extracellular tau. As we have pointed out over the years, this is rather surprising because it is well known that most of pathological tau protein is found intracellularly. It has been repeatedly shown by several groups over the past decades that antibodies can enter neurons and that their cellular uptake can be enhanced by various means, particularly by altering their charge. Here, we will briefly describe the potential extra- and intracellular mechanisms involved in antibody-mediated clearance of tau pathology, discuss these in the context of recent failures of some of the tau antibody trials, and finally provide a brief overview of how the intracellular efficacy of tau antibodies can potentially be further improved by certain modifications that aim to enhance tau clearance via specific intracellular degradation pathways.
Collapse
Affiliation(s)
- Erin E Congdon
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, United States.
| | - Yixiang Jiang
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, United States
| | - Einar M Sigurdsson
- Department of Neuroscience and Physiology, Neuroscience Institute, New York University Grossman School of Medicine, New York, NY 10016, United States; Department of Psychiatry, New York University Grossman School of Medicine, New York, NY 10016, United States.
| |
Collapse
|
14
|
Helboe L, Rosenqvist N, Volbracht C, Pedersen LØ, Pedersen JT, Christensen S, Egebjerg J, Christoffersen CT, Bang-Andersen B, Beach TG, Serrano GE, Falsig J. Highly Specific and Sensitive Target Binding by the Humanized pS396-Tau Antibody hC10.2 Across a Wide Spectrum of Alzheimer’s Disease and Primary Tauopathy Postmortem Brains. J Alzheimers Dis 2022; 88:207-228. [DOI: 10.3233/jad-220125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Deposits of hyperphosphorylated tau fibrils are hallmarks of a broad spectrum of tauopathies, including Alzheimer’s disease (AD). Objective: To investigate heterogeneity of tau pathology across brain extracts from a broad selection of different tauopathies and examine the binding properties of the humanized pS396-tau antibody hC10.2 and six other anti-tau antibodies. Methods: 76 individual tauopathy tissue samples were analyzed in a battery of assays: immunohistochemistry, ELISA, tau aggregation assay, western blot, [3H]PI-2620 and [3H]MK-6240 tau tracer binding, and aggregated seeding activity in RD_P301S HEK293T Biosensor cells. The efficiency of seven anti-tau antibodies to engage with pathological tau species was directly compared. Results: Our data indicate that a strong correlation existed between the tau tracer binding, amount of tau aggregates, pS396-tau phosphorylation, and seeding activity. The hC10.2 antibody, which has entered clinical development, effectively engaged with its epitope across all individual cases of mid-stage and late AD, and primary tauopathies. hC10.2 was superior compared to other phospho- and total tau antibodies to prevent seeded tau aggregation in the biosensor cells. hC10.2 effectively depleted hyperphosphorylated and aggregated tau species across all tauopathy samples proportionally to the amount of tau aggregates. In AD samples, hC10.2 bound to ghost tangles which represent extracellular pathological tau species. Conclusion: S396 hyperphosphorylation is a feature of the formation of seeding-competent tau across different tauopathies and it is present both in intra- and extracellular pathological tau. hC10.2 represents an excellent candidate for a hyperphosphorylation-selective therapeutic tau antibody for the treatment of AD and primary tauopathies.
Collapse
|
15
|
Hamid FA, Marker CL, Raleigh MD, Khaimraj A, Winston S, Pentel PR, Pravetoni M. Pre-clinical safety and toxicology profile of a candidate vaccine to treat oxycodone use disorder. Vaccine 2022; 40:3244-3252. [DOI: 10.1016/j.vaccine.2022.03.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/17/2022] [Accepted: 03/22/2022] [Indexed: 10/18/2022]
|
16
|
Kovacech B, Fialova L, Filipcik P, Skrabana R, Zilkova M, Paulenka-Ivanovova N, Kovac A, Palova D, Rolkova GP, Tomkova K, Csokova NT, Markova K, Skrabanova M, Sinska K, Basheer N, Majerova P, Hanes J, Parrak V, Prcina M, Cehlar O, Cente M, Piestansky J, Fresser M, Novak M, Slavikova M, Borsova K, Cabanova V, Brejova B, Vinař T, Nosek J, Klempa B, Eyer L, Hönig V, Palus M, Ruzek D, Vyhlidalova T, Strakova P, Mrazkova B, Zudova D, Koubkova G, Novosadova V, Prochazka J, Sedlacek R, Zilka N, Kontsekova E. Monoclonal antibodies targeting two immunodominant epitopes on the Spike protein neutralize emerging SARS-CoV-2 variants of concern. EBioMedicine 2022; 76:103818. [PMID: 35078012 PMCID: PMC8782626 DOI: 10.1016/j.ebiom.2022.103818] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/05/2022] [Accepted: 01/05/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The emergence of new SARS-CoV-2 variants of concern B.1.1.7 (Alpha), B.1.351 (Beta), P.1 (Gamma) and B.1.617.2 (Delta) that harbor mutations in the viral S protein raised concern about activity of current vaccines and therapeutic antibodies. Independent studies have shown that mutant variants are partially or completely resistant against some of the therapeutic antibodies authorized for emergency use. METHODS We employed hybridoma technology, ELISA-based and cell-based S-ACE2 interaction assays combined with authentic virus neutralization assays to develop second-generation antibodies, which were specifically selected for their ability to neutralize the new variants of SARS-CoV-2. FINDINGS AX290 and AX677, two monoclonal antibodies with non-overlapping epitopes, exhibit subnanomolar or nanomolar affinities to the receptor binding domain of the viral Spike protein carrying amino acid substitutions N501Y, N439K, E484K, K417N, and a combination N501Y/E484K/K417N found in the circulating virus variants. The antibodies showed excellent neutralization of an authentic SARS-CoV-2 virus representing strains circulating in Europe in spring 2020 and also the variants of concern B.1.1.7 (Alpha), B.1.351 (Beta) and B.1.617.2 (Delta). In addition, AX677 is able to bind Omicron Spike protein just like the wild type Spike. The combination of the two antibodies prevented the appearance of escape mutations of the authentic SARS-CoV-2 virus. Prophylactic administration of AX290 and AX677, either individually or in combination, effectively reduced viral burden and inflammation in the lungs, and prevented disease in a mouse model of SARS-CoV-2 infection. INTERPRETATION The virus-neutralizing properties were fully reproduced in chimeric mouse-human versions of the antibodies, which may represent a promising tool for COVID-19 therapy. FUNDING The study was funded by AXON Neuroscience SE and AXON COVIDAX a.s.
Collapse
Affiliation(s)
- Branislav Kovacech
- AXON COVIDAX a. s.; Bratislava, 811 02, Slovakia; AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia.
| | - Lubica Fialova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Peter Filipcik
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | | | - Monika Zilkova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | | | - Andrej Kovac
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Denisa Palova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | | | | | - Natalia Turic Csokova
- Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | - Karina Markova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Michaela Skrabanova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | - Kristina Sinska
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Neha Basheer
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Petra Majerova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Jozef Hanes
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | - Vojtech Parrak
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Michal Prcina
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | - Ondrej Cehlar
- Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | - Martin Cente
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| | | | - Michal Fresser
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia
| | | | - Monika Slavikova
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences; Bratislava, 845 05, Slovakia
| | - Kristina Borsova
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences; Bratislava, 845 05, Slovakia; Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava; Bratislava, 842 15, Slovakia
| | - Viktoria Cabanova
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences; Bratislava, 845 05, Slovakia
| | - Bronislava Brejova
- Department of Computer Science, Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava; Bratislava, 842 48, Slovakia
| | - Tomas Vinař
- Department of Applied Informatics, Faculty of Mathematics, Physics and Informatics, Comenius University in Bratislava; Bratislava, 842 48, Slovakia
| | - Jozef Nosek
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University in Bratislava; Bratislava, 842 15, Slovakia
| | - Boris Klempa
- Biomedical Research Center, Institute of Virology, Slovak Academy of Sciences; Bratislava, 845 05, Slovakia
| | - Ludek Eyer
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic
| | - Vaclav Hönig
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic
| | - Martin Palus
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic
| | - Daniel Ruzek
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic; Department of Experimental Biology, Faculty of Science, Masaryk University, Kamenice 753/5, CZ-62500 Brno, Czech Republic
| | - Tereza Vyhlidalova
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic
| | - Petra Strakova
- Institute of Parasitology, Biology Centre of the Czech Academy of Sciences, Branisovska 31, CZ-37005 Ceske Budejovice, Czech Republic; Veterinary Research Institute, Hudcova 70, CZ-62100 Brno, Czech Republic
| | - Blanka Mrazkova
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Dagmar Zudova
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Gizela Koubkova
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Vendula Novosadova
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Jan Prochazka
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Radislav Sedlacek
- Czech Centre of Phenogenomics, Institute of Molecular Genetics, ASCR v.v.i, Prumyslova 595, 252 50, Vestec, Czech Republic
| | - Norbert Zilka
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia.
| | - Eva Kontsekova
- AXON Neuroscience R&D Services SE; Bratislava, 811 02, Slovakia; Institute of Neuroimmunology, Slovak Academy of Sciences; Bratislava, 845 10, Slovakia
| |
Collapse
|
17
|
Zhao X, Hasan S, Liou B, Lin Y, Sun Y, Liu C. Analysis of the Biomarkers for Neurodegenerative Diseases in Aged Progranulin Deficient Mice. Int J Mol Sci 2022; 23:629. [PMID: 35054815 PMCID: PMC8775568 DOI: 10.3390/ijms23020629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/23/2021] [Accepted: 01/03/2022] [Indexed: 11/28/2022] Open
Abstract
Neurodegenerative diseases are debilitating impairments that affect millions of people worldwide and are characterized by progressive degeneration of structure and function of the central or peripheral nervous system. Effective biomarkers for neurodegenerative diseases can be used to improve the diagnostic workup in the clinic as well as facilitate the development of effective disease-modifying therapies. Progranulin (PGRN) has been reported to be involved in various neurodegenerative disorders. Hence, in the current study we systematically compared the inflammation and accumulation of typical neurodegenerative disease markers in the brain tissue between PGRN knockout (PGRN KO) and wildtype (WT) mice. We found that PGRN deficiency led to significant neuron loss as well as activation of microglia and astrocytes in aged mice. Several characteristic neurodegenerative markers, including α-synuclein, TAR DNA-binding protein 43 (TDP-43), Tau, and β-amyloid, were all accumulated in the brain of PGRN-deficient mice as compared to WT mice. Moreover, higher aggregation of lipofuscin was observed in the brain tissue of PGRN-deficient mice compared with WT mice. In addition, the autophagy was also defective in the brain of PGRN-deficient mice, indicated by the abnormal expression level of autophagy marker LC3-II. Collectively, comprehensive assays support the idea that PGRN plays an important role during the development of neurodegenerative disease, indicating that PGRN might be a useful biomarker for neurodegenerative diseases in clinical settings.
Collapse
Affiliation(s)
- Xiangli Zhao
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA; (X.Z.); (S.H.)
| | - Sadaf Hasan
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA; (X.Z.); (S.H.)
| | - Benjamin Liou
- The Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (B.L.); (Y.L.); (Y.S.)
| | - Yi Lin
- The Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (B.L.); (Y.L.); (Y.S.)
| | - Ying Sun
- The Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (B.L.); (Y.L.); (Y.S.)
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Chuanju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA; (X.Z.); (S.H.)
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
18
|
Peptide-Based Vaccines for Neurodegenerative Diseases: Recent Endeavors and Future Perspectives. Vaccines (Basel) 2021; 9:vaccines9111278. [PMID: 34835209 PMCID: PMC8622585 DOI: 10.3390/vaccines9111278] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/25/2021] [Accepted: 10/30/2021] [Indexed: 02/06/2023] Open
Abstract
The development of peptide-based vaccines for treating human neurodegenerative diseases has been the eventual aim of many research endeavors, although no active immunotherapies have been approved for clinical use till now. A typical example of such endeavors is the effort to develop vaccines for Alzheimer’s disease based on the beta-amyloid peptide, which continues to be intensively investigated despite previous setbacks. In this paper, recent developments in peptide-based vaccines which target beta-amyloid as well as tau protein and α-synuclein are presented. Particular focus has been directed toward peptide epitopes and formulation systems selected/developed and employed to enhance vaccine efficacy and safety. Results from both, human clinical trials and animal preclinical studies conducted mainly in transgenic mice have been included. Future perspectives on the topic are also briefly discussed.
Collapse
|
19
|
Carroll T, Guha S, Nehrke K, Johnson GVW. Tau Post-Translational Modifications: Potentiators of Selective Vulnerability in Sporadic Alzheimer's Disease. BIOLOGY 2021; 10:1047. [PMID: 34681146 PMCID: PMC8533264 DOI: 10.3390/biology10101047] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/14/2022]
Abstract
Sporadic Alzheimer's Disease (AD) is the most common form of dementia, and its severity is characterized by the progressive formation of tau neurofibrillary tangles along a well-described path through the brain. This spatial progression provides the basis for Braak staging of the pathological progression for AD. Tau protein is a necessary component of AD pathology, and recent studies have found that soluble tau species with selectively, but not extensively, modified epitopes accumulate along the path of disease progression before AD-associated insoluble aggregates form. As such, modified tau may represent a key cellular stressing agent that potentiates selective vulnerability in susceptible neurons during AD progression. Specifically, studies have found that tau phosphorylated at sites such as T181, T231, and S396 may initiate early pathological changes in tau by disrupting proper tau localization, initiating tau oligomerization, and facilitating tau accumulation and extracellular export. Thus, this review elucidates potential mechanisms through which tau post-translational modifications (PTMs) may simultaneously serve as key modulators of the spatial progression observed in AD development and as key instigators of early pathology related to neurodegeneration-relevant cellular dysfunctions.
Collapse
Affiliation(s)
- Trae Carroll
- Department of Pathology, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Sanjib Guha
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Keith Nehrke
- Department of Medicine, Nephrology Division, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| | - Gail V. W. Johnson
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center (URMC), Rochester, NY 14642, USA;
| |
Collapse
|
20
|
Quint WH, Matečko-Burmann I, Schilcher I, Löffler T, Schöll M, Burmann BM, Vogels T. Bispecific Tau Antibodies with Additional Binding to C1q or Alpha-Synuclein. J Alzheimers Dis 2021; 80:813-829. [PMID: 33579845 DOI: 10.3233/jad-201334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) and other tauopathies are neurodegenerative disorders characterized by cellular accumulation of aggregated tau protein. Tau pathology within these disorders is accompanied by chronic neuroinflammation, such as activation of the classical complement pathway by complement initiation factor C1q. Additionally, about half of the AD cases present with inclusions composed of aggregated alpha-synuclein called Lewy bodies. Lewy bodies in disorders such as Parkinson's disease and Lewy body dementia also frequently occur together with tau pathology. OBJECTIVE Immunotherapy is currently the most promising treatment strategy for tauopathies. However, the presence of multiple pathological processes within tauopathies makes it desirable to simultaneously target more than one disease pathway. METHODS Herein, we have developed three bispecific antibodies based on published antibody binding region sequences. One bispecific antibody binds to tau plus alpha-synuclein and two bispecific antibodies bind to tau plus C1q. RESULTS Affinity of the bispecific antibodies to their targets compared to their monospecific counterparts ranged from nearly identical to one order of magnitude lower. All bispecific antibodies retained binding to aggregated protein in patient-derived brain sections. The bispecific antibodies also retained their ability to inhibit aggregation of recombinant tau, regardless of whether the tau binding sites were in IgG or scFv format. Mono- and bispecific antibodies inhibited cellular seeding induced by AD-derived pathological tau with similar efficacy. Finally, both Tau-C1q bispecific antibodies completely inhibited the classical complement pathway. CONCLUSION Bispecific antibodies that bind to multiple pathological targets may therefore present a promising approach to treat tauopathies and other neurodegenerative disorders.
Collapse
Affiliation(s)
| | - Irena Matečko-Burmann
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
| | | | - Tina Löffler
- QPS Austria GmbH, Neuropharmacology, Grambach, Austria
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK
| | - Björn Marcus Burmann
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden.,Department of Chemistry and Molecular Biology, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Vogels
- Maptimmune BV, The Hague, The Netherlands.,Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden.,Department of Neurodegenerative Disease, UCL Queen Square, Institute of Neurology, University College London, London, UK
| |
Collapse
|
21
|
Abstract
This scientific commentary refers to ‘CSF tau microtubule binding region identifies tau tangle and clinical stages of Alzheimer’s disease’, by Horie et al. (doi:10.1093/brain/awaa373).
Collapse
Affiliation(s)
- Jamie Toombs
- Centre for Discovery Brain Sciences, UK Dementia Research Institute, The University of Edinburgh, UK
| | - Henrik Zetterberg
- UK Dementia Research Institute at UCL, London, UK.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
22
|
Li Q, Xiong C, Liu H, Ge H, Yao X, Liu H. Computational Insights Into the Inhibition Mechanism of Proanthocyanidin B2 on Tau Hexapeptide (PHF6) Oligomer. Front Chem 2021; 9:666043. [PMID: 34336783 PMCID: PMC8316602 DOI: 10.3389/fchem.2021.666043] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 04/14/2021] [Indexed: 11/13/2022] Open
Abstract
The formation of amyloid fibrils from Tau is a key pathogenic feature of Alzheimer's disease (AD). To disturb the formation of Tau aggregates is considered as a promising therapeutic strategy for AD. Recently, a natural product proanthocyanidin B2 (PB2) was confirmed to not only inhibit Tau aggregation, but also disaggregate Tau fibrils. Herein, to explore the inhibition mechanism of PB2 against Tau fibril and to provide the useful information for drug design and discovery, all-atom molecular dynamics simulations were carried out for the ordered Tau hexapeptide PHF6 oligomer in the presence and absence of PB2. The obtained result shows that PB2 can transform PHF6 oligomer from the ordered β-sheet structure into disordered one. Moreover, the clustering analysis and binding free energy calculations identify that S3 site is the most potential binding site. At S3 site, by hydrophobic and hydrogen bond interactions, the residues V309, Y310 and K311 are essential for binding with PB2, especially K311. In a word, our study reveals the molecular mechanism of PB2 inhibiting PHF6 aggregation and it will provide some valuable information for the development of Tau aggregation inhibitors.
Collapse
Affiliation(s)
- Qin Li
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Chunmei Xiong
- School of Pharmacy, Lanzhou University, Lanzhou, China
| | - Hongli Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Huizhen Ge
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou, China
| | - Xiaojun Yao
- State Key Laboratory of Applied Organic Chemistry and Department of Chemistry, Lanzhou University, Lanzhou, China
| | - Huanxiang Liu
- School of Pharmacy, Lanzhou University, Lanzhou, China
| |
Collapse
|
23
|
Ojha R, Prajapati VK. Cognizance of posttranslational modifications in vaccines: A way to enhanced immunogenicity. J Cell Physiol 2021; 236:8020-8034. [PMID: 34170014 PMCID: PMC8427110 DOI: 10.1002/jcp.30483] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/21/2021] [Accepted: 06/14/2021] [Indexed: 12/25/2022]
Abstract
Vaccination is a significant advancement or preventative strategy for controlling the spread of various severe infectious and noninfectious diseases. The purpose of vaccination is to stimulate or activate the immune system by injecting antigens, i.e., either whole microorganisms or using the pathogen's antigenic part or macromolecules. Over time, researchers have made tremendous efforts to reduce vaccine side effects or failure by developing different strategies combining with immunoinformatic and molecular biology. These newly designed vaccines are composed of single or several antigenic molecules derived from a pathogenic organism. Although, whole‐cell vaccines are still in use against various diseases but due to their ineffectiveness, other vaccines like DNA‐based, RNA‐based, and protein‐based vaccines, with the addition of immunostimulatory agents, are in the limelight. Despite this, many researchers escape the most common fundamental phenomenon of protein posttranslational modifications during the development of vaccines, which regulates protein functional behavior, evokes immunogenicity and stability, etc. The negligence about post translational modification (PTM) during vaccine development may affect the vaccine's efficacy and immune responses. Therefore, it becomes imperative to consider these modifications of macromolecules before finalizing the antigenic vaccine construct. Here, we have discussed different types of posttranslational/transcriptional modifications that are usually considered during vaccine construct designing: Glycosylation, Acetylation, Sulfation, Methylation, Amidation, SUMOylation, Ubiquitylation, Lipidation, Formylation, and Phosphorylation. Based on the available research information, we firmly believe that considering these modifications will generate a potential and highly immunogenic antigenic molecule against communicable and noncommunicable diseases compared to the unmodified macromolecules.
Collapse
Affiliation(s)
- Rupal Ojha
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| |
Collapse
|
24
|
Karelina T, Lerner S, Stepanov A, Meerson M, Demin O. Monoclonal antibody therapy efficacy can be boosted by combinations with other treatments: Predictions using an integrated Alzheimer's Disease Platform. CPT Pharmacometrics Syst Pharmacol 2021; 10:543-550. [PMID: 33818905 PMCID: PMC8213414 DOI: 10.1002/psp4.12628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/02/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
For many years, clinical research in Alzheimer's disease (AD) has focused on attempts to identify the most explicit biomarker, namely amyloid beta. Unfortunately, the numerous therapies that have been developed have failed in clinical practice. AD arises as a consequence of multiple factors, and as such it requires a more mechanistic analytical approach than statistical modeling. Quantitative systems pharmacology modeling is a valuable tool for drug development. It utilizes in vitro data for the calibration of parameters, embeds them into physiologically based structures, and explores translation between animals and humans. Such an approach allows for a quantitative study of the dynamics of the interactions between multiple factors or variables. Here, we present an overview of the quantitative translational model in AD, which embraces current preclinical and clinical data. The previously published description of amyloid physiology has been updated and joined with a model for tau pathology and multiple intraneuronal processes responsible for cellular transport, metabolism, or proteostasis. In addition, several hypotheses regarding the best correlates of cognitive deterioration have been validated using clinical data. Here, the amyloid hypothesis was unable to predict the aducanumab clinical trial data, whereas simulations of cognitive impairment coupled with tau seeding or neuronal breakdown (expressed as caspase activity) matched the data. A satisfactory validation of the data from multiple preclinical and clinical studies was followed by an attempt to predict the results of combinatorial treatment with targeted immunotherapy and activation of autophagy using rapamycin. The combination is predicted to yield better efficacy than immunotherapy alone.
Collapse
|
25
|
Novak P, Kovacech B, Katina S, Schmidt R, Scheltens P, Kontsekova E, Ropele S, Fialova L, Kramberger M, Paulenka-Ivanovova N, Smisek M, Hanes J, Stevens E, Kovac A, Sutovsky S, Parrak V, Koson P, Prcina M, Galba J, Cente M, Hromadka T, Filipcik P, Piestansky J, Samcova M, Prenn-Gologranc C, Sivak R, Froelich L, Fresser M, Rakusa M, Harrison J, Hort J, Otto M, Tosun D, Ondrus M, Winblad B, Novak M, Zilka N. ADAMANT: a placebo-controlled randomized phase 2 study of AADvac1, an active immunotherapy against pathological tau in Alzheimer's disease. NATURE AGING 2021; 1:521-534. [PMID: 37117834 DOI: 10.1038/s43587-021-00070-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 04/28/2021] [Indexed: 04/30/2023]
Abstract
Alzheimer's disease (AD) pathology is partly characterized by accumulation of aberrant forms of tau protein. Here we report the results of ADAMANT, a 24-month double-blinded, parallel-arm, randomized phase 2 multicenter placebo-controlled trial of AADvac1, an active peptide vaccine designed to target pathological tau in AD (EudraCT 2015-000630-30). Eleven doses of AADvac1 were administered to patients with mild AD dementia at 40 μg per dose over the course of the trial. The primary objective was to evaluate the safety and tolerability of long-term AADvac1 treatment. The secondary objectives were to evaluate immunogenicity and efficacy of AADvac1 treatment in slowing cognitive and functional decline. A total of 196 patients were randomized 3:2 between AADvac1 and placebo. AADvac1 was safe and well tolerated (AADvac1 n = 117, placebo n = 79; serious adverse events observed in 17.1% of AADvac1-treated individuals and 24.1% of placebo-treated individuals; adverse events observed in 84.6% of AADvac1-treated individuals and 81.0% of placebo-treated individuals). The vaccine induced high levels of IgG antibodies. No significant effects were found in cognitive and functional tests on the whole study sample (Clinical Dementia Rating-Sum of the Boxes scale adjusted mean point difference -0.360 (95% CI -1.306, 0.589)), custom cognitive battery adjusted mean z-score difference of 0.0008 (95% CI -0.169, 0.172). We also present results from exploratory and post hoc analyses looking at relevant biomarkers and clinical outcomes in specific subgroups. Our results show that AADvac1 is safe and immunogenic, but larger stratified studies are needed to better evaluate its potential clinical efficacy and impact on disease biomarkers.
Collapse
Affiliation(s)
- Petr Novak
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia.
| | | | | | - Reinhold Schmidt
- Clinical Division of Neurogeriatrics, Department of Neurology, Medical University Graz, Graz, Austria
| | - Philip Scheltens
- Alzheimer Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | | | - Stefan Ropele
- Clinical Division of General Neurology, Department of Neurology, Medical University Graz, Graz, Austria
| | | | - Milica Kramberger
- Department of Neurology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | | | | | - Jozef Hanes
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Eva Stevens
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Andrej Kovac
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Stanislav Sutovsky
- 1st Department of Neurology, Faculty of Medicine, Comenius University and University Hospital, Bratislava, Slovakia
| | | | - Peter Koson
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | - Michal Prcina
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | | | - Martin Cente
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Tomas Hromadka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | | | | | - Maria Samcova
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | | | - Roman Sivak
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | - Lutz Froelich
- Department of Geriatric Psychiatry, Zentralinstitut für Seelische Gesundheit, Medical Faculty Mannheim University of Heidelberg, Heidelberg, Germany
| | | | - Martin Rakusa
- Department of Neurological Diseases, University Medical Centre Maribor, Maribor, Slovenia
| | - John Harrison
- Alzheimer Center, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Jakub Hort
- Memory Clinic, Department of Neurology, Charles University, 2nd Faculty of Medicine and Motol University Hospital, Prague, Czech Republic
| | - Markus Otto
- Department of Neurology, Ulm University Hospital, Ulm, Germany
| | - Duygu Tosun
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, CA, USA
| | - Matej Ondrus
- AXON Neuroscience CRM Services SE, Bratislava, Slovakia
| | - Bengt Winblad
- Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Solna, Sweden
- Theme Inflammation and Aging, Karolinska University Hospital, Huddinge, Sweden
| | | | - Norbert Zilka
- AXON Neuroscience R&D Services SE, Bratislava, Slovakia
| |
Collapse
|
26
|
Affiliation(s)
- Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, USA.
| |
Collapse
|
27
|
Giunta M, Solje E, Gardoni F, Borroni B, Benussi A. Experimental Disease-Modifying Agents for Frontotemporal Lobar Degeneration. J Exp Pharmacol 2021; 13:359-376. [PMID: 33790662 PMCID: PMC8005747 DOI: 10.2147/jep.s262352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia is a clinically, genetically and pathologically heterogeneous neurodegenerative disorder, enclosing a wide range of different pathological entities, associated with the accumulation of proteins such as tau and TPD-43. Characterized by a high hereditability, mutations in three main genes, MAPT, GRN and C9orf72, can drive the neurodegenerative process. The connection between different genes and proteinopathies through specific mechanisms has shed light on the pathophysiology of the disease, leading to the identification of potential pharmacological targets. New experimental strategies are emerging, in both preclinical and clinical settings, which focus on small molecules rather than gene therapy. In this review, we provide an insight into the aberrant mechanisms leading to FTLD-related proteinopathies and discuss recent therapies with the potential to ameliorate neurodegeneration and disease progression.
Collapse
Affiliation(s)
- Marcello Giunta
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Eino Solje
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
28
|
Toniolo S, Sen A, Husain M. Modulation of Brain Hyperexcitability: Potential New Therapeutic Approaches in Alzheimer's Disease. Int J Mol Sci 2020; 21:E9318. [PMID: 33297460 PMCID: PMC7730926 DOI: 10.3390/ijms21239318] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
People with Alzheimer's disease (AD) have significantly higher rates of subclinical and overt epileptiform activity. In animal models, oligomeric Aβ amyloid is able to induce neuronal hyperexcitability even in the early phases of the disease. Such aberrant activity subsequently leads to downstream accumulation of toxic proteins, and ultimately to further neurodegeneration and neuronal silencing mediated by concomitant tau accumulation. Several neurotransmitters participate in the initial hyperexcitable state, with increased synaptic glutamatergic tone and decreased GABAergic inhibition. These changes appear to activate excitotoxic pathways and, ultimately, cause reduced long-term potentiation, increased long-term depression, and increased GABAergic inhibitory remodelling at the network level. Brain hyperexcitability has therefore been identified as a potential target for therapeutic interventions aimed at enhancing cognition, and, possibly, disease modification in the longer term. Clinical trials are ongoing to evaluate the potential efficacy in targeting hyperexcitability in AD, with levetiracetam showing some encouraging effects. Newer compounds and techniques, such as gene editing via viral vectors or brain stimulation, also show promise. Diagnostic challenges include identifying best biomarkers for measuring sub-clinical epileptiform discharges. Determining the timing of any intervention is critical and future trials will need to carefully stratify participants with respect to the phase of disease pathology.
Collapse
Affiliation(s)
- Sofia Toniolo
- Cognitive Neurology Group, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
- Wellcome Trust Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford OX2 6AE, UK
| | - Arjune Sen
- Oxford Epilepsy Research Group, Nuffield Department Clinical Neurosciences, John Radcliffe Hospital, Oxford OX3 9DU, UK;
| | - Masud Husain
- Cognitive Neurology Group, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
- Wellcome Trust Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford OX2 6AE, UK
| |
Collapse
|
29
|
Oakley SS, Maina MB, Marshall KE, Al-Hilaly YK, Harrington CR, Wischik CM, Serpell LC. Tau Filament Self-Assembly and Structure: Tau as a Therapeutic Target. Front Neurol 2020; 11:590754. [PMID: 33281730 PMCID: PMC7688747 DOI: 10.3389/fneur.2020.590754] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 09/04/2020] [Indexed: 12/12/2022] Open
Abstract
Tau plays an important pathological role in a group of neurodegenerative diseases called tauopathies, including Alzheimer's disease, Pick's disease, chronic traumatic encephalopathy and corticobasal degeneration. In each disease, tau self-assembles abnormally to form filaments that deposit in the brain. Tau is a natively unfolded protein that can adopt distinct structures in different pathological disorders. Cryo-electron microscopy has recently provided a series of structures for the core of the filaments purified from brain tissue from patients with different tauopathies and revealed that they share a common core region, while differing in their specific conformation. This structurally resolvable part of the core is contained within a proteolytically stable core region from the repeat domain initially isolated from AD tau filaments. Tau has recently become an important target for therapy. Recent work has suggested that the prevention of tau self-assembly may be effective in slowing the progression of Alzheimer's disease and other tauopathies. Here we review the work that explores the importance of tau filament structures and tau self-assembly mechanisms, as well as examining model systems that permit the exploration of the mode of action of potential inhibitors.
Collapse
Affiliation(s)
- Sebastian S. Oakley
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Mahmoud B. Maina
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- College of Medical Sciences, Yobe State University, Damaturu, Nigeria
| | - Karen E. Marshall
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| | - Youssra K. Al-Hilaly
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
- Chemistry Department, College of Science, Mustansiriyah University, Baghdad, Iraq
| | - Charlie R. Harrington
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
- TauRx Therapeutics Ltd., Aberdeen, United Kingdom
| | - Claude M. Wischik
- School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, United Kingdom
- TauRx Therapeutics Ltd., Aberdeen, United Kingdom
| | - Louise C. Serpell
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, United Kingdom
| |
Collapse
|
30
|
Ng PY, Chang IS, Koh RY, Chye SM. Recent advances in tau-directed immunotherapy against Alzheimer's disease: an overview of pre-clinical and clinical development. Metab Brain Dis 2020; 35:1049-1066. [PMID: 32632666 DOI: 10.1007/s11011-020-00591-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 06/23/2020] [Indexed: 02/01/2023]
Abstract
Alzheimer's disease (AD) has been a worldwide concern for many years now. This is due to the fact that AD is an irreversible and progressive neurodegenerative disease that affects quality of life. Failure of some Phase II/III clinical trials in AD targeting accumulation of β-amyloid in the brain has led to an increase in interest in studying alternative treatments against tubulin-associated unit (Tau) pathology. These alternative treatments include active and passive immunisation. Based on numerous studies, Tau is reported as a potential immunotherapeutic target for tauopathy-related diseases including AD. Accumulation and aggregation of hyperphosphorylated Tau as neuropil threads and neurofibrillary tangles (NFT) are pathological hallmarks of AD. Both active and passive immunisation targeting Tau protein have shown the capabilities to decrease or prevent Tau pathology and improve either motor or cognitive impairment in various animal models. In this review, we summarise recent advances in active and passive immunisation targeting pathological Tau protein, and will discuss with data obtained from both animal and human trials. Together, we give a brief overview about problems being encountered in these immunotherapies.
Collapse
Affiliation(s)
- Pei Ying Ng
- School of Postgraduate, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - I Shuen Chang
- School of Health Science, Division of Biomedical Science and Biotechnology, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Rhun Yian Koh
- School of Health Science, Division of Biomedical Science and Biotechnology, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- School of Health Science, Division of Biomedical Science and Biotechnology, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
31
|
Plotkin SS, Cashman NR. Passive immunotherapies targeting Aβ and tau in Alzheimer's disease. Neurobiol Dis 2020; 144:105010. [PMID: 32682954 PMCID: PMC7365083 DOI: 10.1016/j.nbd.2020.105010] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 07/01/2020] [Accepted: 07/07/2020] [Indexed: 12/21/2022] Open
Abstract
Amyloid-β (Aβ) and tau proteins currently represent the two most promising targets to treat Alzheimer's disease. The most extensively developed method to treat the pathologic forms of these proteins is through the administration of exogenous antibodies, or passive immunotherapy. In this review, we discuss the molecular-level strategies that researchers are using to design an effective therapeutic antibody, given the challenges in treating this disease. These challenges include selectively targeting a protein that has misfolded or is pathological rather than the more abundant, healthy protein, designing strategic constructs for immunizing an animal to raise an antibody that has the appropriate conformational selectivity to achieve this end, and clearing the pathological protein species before prion-like cell-to-cell spread of misfolded protein has irreparably damaged neurons, without invoking damaging inflammatory responses in the brain that naturally arise when the innate immune system is clearing foreign agents. The various solutions to these problems in current clinical trials will be discussed.
Collapse
Affiliation(s)
- Steven S Plotkin
- University of British Columbia, Department of Physics and Astronomy and Genome Sciences and Technology Program, Vancouver, BC V6T 1Z1, Canada.
| | - Neil R Cashman
- University of British Columbia, Djavad Mowafaghian Centre for Brain Health, Vancouver, BC V6T 2B5, Canada.
| |
Collapse
|
32
|
VandeVrede L, Ljubenkov PA, Rojas JC, Welch AE, Boxer AL. Four-Repeat Tauopathies: Current Management and Future Treatments. Neurotherapeutics 2020; 17:1563-1581. [PMID: 32676851 PMCID: PMC7851277 DOI: 10.1007/s13311-020-00888-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Four-repeat tauopathies are a neurodegenerative disease characterized by brain parenchymal accumulation of a specific isoform of the protein tau, which gives rise to a wide breadth of clinical syndromes encompassing diverse symptomatology, with the most common syndromes being progressive supranuclear palsy-Richardson's and corticobasal syndrome. Despite the lack of effective disease-modifying therapies, targeted treatment of symptoms can improve quality of life for patients with 4-repeat tauopathies. However, managing these symptoms can be a daunting task, even for those familiar with the diseases, as they span motor, sensory, cognitive, affective, autonomic, and behavioral domains. This review describes current approaches to symptomatic management of common clinical symptoms in 4-repeat tauopathies with a focus on practical patient management, including pharmacologic and nonpharmacologic strategies, and concludes with a discussion of the history and future of disease-modifying therapeutics and clinical trials in this population.
Collapse
Affiliation(s)
- Lawren VandeVrede
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA.
| | - Peter A Ljubenkov
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Julio C Rojas
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Ariane E Welch
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
33
|
VandeVrede L, Boxer AL, Polydoro M. Targeting tau: Clinical trials and novel therapeutic approaches. Neurosci Lett 2020; 731:134919. [PMID: 32380145 PMCID: PMC9212860 DOI: 10.1016/j.neulet.2020.134919] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 02/25/2020] [Accepted: 03/17/2020] [Indexed: 12/12/2022]
Abstract
Tauopathies are a group of over 20 clinicopathological neurodegenerative diseases including Alzheimer's disease (AD), the most common type of dementia, progressive supranuclear palsy, Pick's disease, corticobasal degeneration, among others. Tauopathies are defined by neurodegeneration and the presence of tau aggregates in affected brains regions. Interestingly, regional tau aggregation burden correlates with clinical phenotype and predicts cognitive status. Autosomal dominant mutations in the MAPT gene lead to tau deposition and clinical FTD syndromes with cognitive, behavioral, and motor impairment. Polymorphisms in or around the MAPT gene have also been strongly linked to other proteinopathies including synucleinopathies. Taken together these findings suggests that tau plays a critical role in neurodegeneration and proteinopathies, supporting the idea that tau targeted approaches can be disease-modifying and lead to clinically meaningful benefits in slowing or reversing disease progression. Increasingly, human clinical trials are testing this hypothesis. This article reviews tau-targeted therapies tested in clinical trials as well as agents currently in active development based on publicly disclosed information. We describe the therapeutic approaches of these trials based on the potential pathogenic mechanism they target.
Collapse
Affiliation(s)
- Lawren VandeVrede
- Memory and Aging Center, Department of Neurology, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Adam L Boxer
- Memory and Aging Center, Department of Neurology, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | | |
Collapse
|
34
|
Kwon S, Iba M, Kim C, Masliah E. Immunotherapies for Aging-Related Neurodegenerative Diseases-Emerging Perspectives and New Targets. Neurotherapeutics 2020; 17:935-954. [PMID: 32347461 PMCID: PMC7222955 DOI: 10.1007/s13311-020-00853-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neurological disorders such as Alzheimer's disease (AD), Lewy body dementia (LBD), frontotemporal dementia (FTD), and vascular dementia (VCID) have no disease-modifying treatments to date and now constitute a dementia crisis that affects 5 million in the USA and over 50 million worldwide. The most common pathological hallmark of these age-related neurodegenerative diseases is the accumulation of specific proteins, including amyloid beta (Aβ), tau, α-synuclein (α-syn), TAR DNA-binding protein 43 (TDP43), and repeat-associated non-ATG (RAN) peptides, in the intra- and extracellular spaces of selected brain regions. Whereas it remains controversial whether these accumulations are pathogenic or merely a byproduct of disease, the majority of therapeutic research has focused on clearing protein aggregates. Immunotherapies have garnered particular attention for their ability to target specific protein strains and conformations as well as promote clearance. Immunotherapies can also be neuroprotective: by neutralizing extracellular protein aggregates, they reduce spread, synaptic damage, and neuroinflammation. This review will briefly examine the current state of research in immunotherapies against the 3 most commonly targeted proteins for age-related neurodegenerative disease: Aβ, tau, and α-syn. The discussion will then turn to combinatorial strategies that enhance the effects of immunotherapy against aggregating protein, followed by new potential targets of immunotherapy such as aging-related processes.
Collapse
Affiliation(s)
- Somin Kwon
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michiyo Iba
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Changyoun Kim
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Eliezer Masliah
- Laboratory of Neurogenetics, Molecular Neuropathology Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, 20892, USA.
- Division of Neuroscience, National Institute on Aging/National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
35
|
Zilkova M, Nolle A, Kovacech B, Kontsekova E, Weisova P, Filipcik P, Skrabana R, Prcina M, Hromadka T, Cehlar O, Rolkova GP, Maderova D, Novak M, Zilka N, Hoozemans JJM. Humanized tau antibodies promote tau uptake by human microglia without any increase of inflammation. Acta Neuropathol Commun 2020; 8:74. [PMID: 32471486 PMCID: PMC7257136 DOI: 10.1186/s40478-020-00948-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 05/13/2020] [Indexed: 01/10/2023] Open
Abstract
Immunotherapies targeting pathological tau have recently emerged as a promising approach for treatment of neurodegenerative disorders. We have previously showed that the mouse antibody DC8E8 discriminates between healthy and pathological tau, reduces tau pathology in murine tauopathy models and inhibits neuronal internalization of AD tau species in vitro. Here we show, that DC8E8 and antibodies elicited against the first-in-man tau vaccine, AADvac1, which is based on the DC8E8 epitope peptide, both promote uptake of pathological tau by mouse primary microglia. IgG1 and IgG4 isotypes of AX004, the humanized versions of DC8E8, accelerate tau uptake by human primary microglia isolated from post-mortem aged and diseased brains. This promoting activity requires the presence of the Fc-domain of the antibodies. The IgG1 isotype of AX004 showed greater ability to promote tau uptake compared to the IgG4 isotype, while none of the antibody-tau complexes provoked increased pro-inflammatory activity of microglia. Our data suggest that IgG1 has better suitability for therapeutic development.
Collapse
|
36
|
Corsetti V, Borreca A, Latina V, Giacovazzo G, Pignataro A, Krashia P, Natale F, Cocco S, Rinaudo M, Malerba F, Florio R, Ciarapica R, Coccurello R, D’Amelio M, Ammassari-Teule M, Grassi C, Calissano P, Amadoro G. Passive immunotherapy for N-truncated tau ameliorates the cognitive deficits in two mouse Alzheimer's disease models. Brain Commun 2020; 2:fcaa039. [PMID: 32954296 PMCID: PMC7425324 DOI: 10.1093/braincomms/fcaa039] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/05/2020] [Accepted: 02/12/2020] [Indexed: 12/12/2022] Open
Abstract
Clinical and neuropathological studies have shown that tau pathology better correlates with the severity of dementia than amyloid plaque burden, making tau an attractive target for the cure of Alzheimer's disease. We have explored whether passive immunization with the 12A12 monoclonal antibody (26-36aa of tau protein) could improve the Alzheimer's disease phenotype of two well-established mouse models, Tg2576 and 3xTg mice. 12A12 is a cleavage-specific monoclonal antibody which selectively binds the pathologically relevant neurotoxic NH226-230 fragment (i.e. NH2htau) of tau protein without cross-reacting with its full-length physiological form(s). We found out that intravenous administration of 12A12 monoclonal antibody into symptomatic (6 months old) animals: (i) reaches the hippocampus in its biologically active (antigen-binding competent) form and successfully neutralizes its target; (ii) reduces both pathological tau and amyloid precursor protein/amyloidβ metabolisms involved in early disease-associated synaptic deterioration; (iii) improves episodic-like type of learning/memory skills in hippocampal-based novel object recognition and object place recognition behavioural tasks; (iv) restores the specific up-regulation of the activity-regulated cytoskeleton-associated protein involved in consolidation of experience-dependent synaptic plasticity; (v) relieves the loss of dendritic spine connectivity in pyramidal hippocampal CA1 neurons; (vi) rescues the Alzheimer's disease-related electrophysiological deficits in hippocampal long-term potentiation at the CA3-CA1 synapses; and (vii) mitigates the neuroinflammatory response (reactive gliosis). These findings indicate that the 20-22 kDa NH2-terminal tau fragment is crucial target for Alzheimer's disease therapy and prospect immunotherapy with 12A12 monoclonal antibody as safe (normal tau-preserving), beneficial approach in contrasting the early Amyloidβ-dependent and independent neuropathological and cognitive alterations in affected subjects.
Collapse
Affiliation(s)
| | - Antonella Borreca
- Humanitas University Laboratory of Pharmacology and Brain Pathology, Neuro Center, 20089 Milan, Italy
- Institute of Neuroscience, 20129 Milan, Italy
| | | | | | | | - Paraskevi Krashia
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Department of Medicine, University Campus Bio-Medico, 00128 Rome, Italy
- Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128 Rome, Italy
| | - Francesca Natale
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Sara Cocco
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | - Marco Rinaudo
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
| | | | - Rita Florio
- European Brain Research Institute (EBRI), 00161 Rome, Italy
| | | | - Roberto Coccurello
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Institute for Complex Systems (ISC), CNR, 00185 Rome, Italy
| | - Marcello D’Amelio
- IRCSS Santa Lucia Foundation, 00143 Rome, Italy
- Department of Medicine, University Campus Bio-Medico, 00128 Rome, Italy
- Department of Science and Technology for Humans and Environment, University Campus Bio-medico, 00128 Rome, Italy
| | | | - Claudio Grassi
- Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Rome, Italy
- Institute of Human Physiology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | | | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), 00161 Rome, Italy
- Institute of Translational Pharmacology (IFT)–National Research Council (CNR), 00133 Rome, Italy
| |
Collapse
|
37
|
Abstract
Vaccines have had a profound impact on the management and prevention of infectious disease. In addition, the development of vaccines against chronic diseases has attracted considerable interest as an approach to prevent, rather than treat, conditions such as cancer, Alzheimer's disease, and others. Subunit vaccines consist of nongenetic components of the infectious agent or disease-related epitope. In this Review, we discuss peptide-based vaccines and their potential in three therapeutic areas: infectious disease, Alzheimer's disease, and cancer. We discuss factors that contribute to vaccine efficacy and how these parameters may potentially be modulated by design. We examine both clinically tested vaccines as well as nascent approaches and explore current challenges and potential remedies. While peptide vaccines hold substantial promise in the prevention of human disease, many obstacles remain that have hampered their clinical use; thus, continued research efforts to address these challenges are warranted.
Collapse
Affiliation(s)
- Ryan J. Malonis
- Department of Biochemistry, Albert Einstein College of Medicine, Michael F. Price Center for Translational Research, 1301 Morris Park Avenue, Bronx, NY 10461
| | - Jonathan R. Lai
- Department of Biochemistry, Albert Einstein College of Medicine, Michael F. Price Center for Translational Research, 1301 Morris Park Avenue, Bronx, NY 10461
| | - Olivia Vergnolle
- Department of Biochemistry, Albert Einstein College of Medicine, Michael F. Price Center for Translational Research, 1301 Morris Park Avenue, Bronx, NY 10461
| |
Collapse
|
38
|
Xie J, Liang R, Wang Y, Huang J, Cao X, Niu B. Progress in Target Drug Molecules for Alzheimer's Disease. Curr Top Med Chem 2020; 20:4-36. [DOI: 10.2174/1568026619666191203113745] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/20/2019] [Accepted: 10/31/2019] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease that 4 widespread in the elderly.
The etiology of AD is complicated, and its pathogenesis is still unclear. Although there are many
researches on anti-AD drugs, they are limited to reverse relief symptoms and cannot treat diseases.
Therefore, the development of high-efficiency anti-AD drugs with no side effects has become an urgent
need. Based on the published literature, this paper summarizes the main targets of AD and their drugs,
and focuses on the research and development progress of these drugs in recent years.
Collapse
Affiliation(s)
- Jiayang Xie
- School of Life Science, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Ruirui Liang
- School of Life Science, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Yajiang Wang
- School of Life Science, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Junyi Huang
- School of Life Science, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| | - Xin Cao
- Zhongshan Hospital Institute of Clinical Science, Fudan University Shanghai Medical College, Shanghai, China
| | - Bing Niu
- School of Life Science, Shanghai University, 99 Shangda Road, Shanghai 200444, China
| |
Collapse
|
39
|
Ziu I, Laryea ET, Alashkar F, Wu CG, Martic S. A dip-and-read optical aptasensor for detection of tau protein. Anal Bioanal Chem 2020; 412:1193-1201. [PMID: 31900535 DOI: 10.1007/s00216-019-02350-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/03/2019] [Accepted: 12/11/2019] [Indexed: 12/20/2022]
Abstract
Neurodegeneration currently remains without a differential diagnosis or cure. Tau protein is one of the biomarkers of neurodegenerative diseases commonly known as tauopathies. Tau protein plays an integral role in stabilizing microtubules and cell structure; however, due to post-translational modifications, tau protein undergoes self-assembly into cytotoxic structures and is co-localized intra- and extracellularly. Hence, tau protein is a viable biomarker associated with protein pathogenesis and neurodegeneration. The novel optical biosensor for tau441 protein is based on the aptamer recognition probe and the biolayer interferometry (BLI) method for detection. The current biotin-aptasensor in combination with the streptavidin surface provides real-time monitoring of tau441 protein in the nanomolar range, with the limit of detection at 6.7 nM in vitro. The tau441 detection is achieved with high selectivity over other neurodegeneration biomarkers which include amyloid-β and α-synuclein. The aptasensor also allows for tau441 protein detection in a complex matrix such as fetal bovine serum, indicating its utility in other biological fluids for diagnostic applications. The optical method is simple, rapid and highly selective for point-of-care application which is critical for achieving the early and differential diagnosis of neurodegenerative diseases and identifying their treatments. Graphical abstract.
Collapse
Affiliation(s)
- Iva Ziu
- Department of Chemistry, Oakland University, Rochester, MI, 48309, USA
| | - Erving T Laryea
- Department of Chemistry, Oakland University, Rochester, MI, 48309, USA
| | - Fayza Alashkar
- Department of Chemistry, Oakland University, Rochester, MI, 48309, USA
| | - Colin G Wu
- Department of Chemistry, Oakland University, Rochester, MI, 48309, USA
| | - Sanela Martic
- Department of Forensic Science, Environmental and Life Sciences, Trent University, Peterborough, ON, K9L 0G2, Canada.
| |
Collapse
|
40
|
Ghaffari M, Sanadgol N, Abdollahi M. A Systematic Review of Current Progresses in the Nucleic Acid-Based Therapies for Neurodegeneration with Implications for Alzheimer's Disease. Mini Rev Med Chem 2020; 20:1499-1517. [PMID: 32400332 DOI: 10.2174/1389557520666200513122357] [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] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/31/2020] [Accepted: 04/20/2020] [Indexed: 12/30/2022]
Abstract
Recently, manipulation of gene expression and switching genes on or off highlight the potential of nucleic acid-based therapies (NA-BTs). Alzheimer's Disease (AD) is a common devastating neurodegenerative disease (NDs) responsible for 60-80% of all cases of dementia and predicted as a main public health concern among aged populations. The aim of this study was to outline the current research in the field of NA-BTs for the treatment of AD disabilities, including strategies to suppress the memory and learning defects, to promote recovery processes, and to reinforce social relationships in these patients. This review was performed via evaluating PubMed reported studies from January 2010 to November 2019. Also, reference lists were checked to find additional studies. All intermediation or complementarity of animal models, case-control and cohort studies, and controlled trials (CTs) on specific NA-BTs to AD were acceptable, although in vitro studies were excluded due to the considerable diversities and heterogeneities. After removing the duplicates according to preferred reporting items for systematic reviews and meta-analyses (PRISMA) instruction, we merged remaining titles across search databases. There are 48 ongoing studies related to the application of nucleic acids in the treatment and diagnosis of AD where more consideration is given to DNA targeting strategies (18 targets for vectors and aptamers), antisense oligonucleotides (10 targets), micro-RNAs mimics (7 targets), antagomiRs (6 targets), small interferences-RNAs (5 targets), as well as mRNAs (2 targets) respectively. All of these targets are grouped into 4 categories according to their role in molecular pathways where amyloid-β (18 targets), neural survival (11 targets), memory and cognition (8 targets), and tau (3 targets) are more targeted pathways, respectively. With recent successes in the systemic delivery of nucleic acids via intravenous injection; it is worth investing in the production of new-generation medicines. There are still several challenges for NA-BTs including, their delivery to the effective modulators, mass production at low cost, sustaining efficacy and minimizing off-target effects. Regarding miRNA-based therapies, given the obvious involvement of miRNAs in numerous facets of brain disease, and the many sophisticated techniques for delivery to the brain, miRNA-based therapies will make new hope for the treatment of neurological diseases such as AD.
Collapse
Affiliation(s)
- Maryam Ghaffari
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran
| | - Nima Sanadgol
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran
| | - Mohammad Abdollahi
- Toxicology and Diseases Group, Pharmaceutical Sciences Research Center (PSRC), The Institute of Pharmaceutical Sciences (TIPS), and Department of Toxicology and Pharmacology, School of Pharmacy, Tehran University of Medical Sciences, Iran
| |
Collapse
|
41
|
Vander Zanden CM, Chi EY. Passive Immunotherapies Targeting Amyloid Beta and Tau Oligomers in Alzheimer's Disease. J Pharm Sci 2019; 109:68-73. [PMID: 31647950 DOI: 10.1016/j.xphs.2019.10.024] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is historically difficult to treat, in part because of the inaccessible nature of brain pathology. Amyloid beta and tau proteins drive pathology by forming toxic oligomers that eventually deposit as insoluble amyloid plaques and neurofibrillary tangles. Recent clinical studies suggest that effective drugs must specifically target oligomers, not native monomers or insoluble fibrils. Passive immunotherapy is a promising pharmaceutical strategy used to specifically target these oligomers in situ. Using the specificity of antibodies coupled with the natural power of the body's immune response, this treatment provides an opportunity for safe clearance of pathogenic protein species from the brain. Passive immunotherapies against amyloid beta and tau oligomers have progressed to clinical trials, with many currently in progress. Biochemical studies of antibody-oligomer complexes have helped identify previously unknown toxic epitopes, thus providing knowledge to the AD field as a whole. This mini-review focuses on the efforts to develop passive immunotherapy treatments for AD and discusses the knowledge gained from recent failures and clinical trials in progress.
Collapse
Affiliation(s)
- Crystal M Vander Zanden
- Center for Biomedical Engineering, Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131; Department of Chemistry and Biochemistry, University of Colorado at Colorado Springs, Colorado Springs, Colorado 80918.
| | - Eva Y Chi
- Center for Biomedical Engineering, Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131; Department of Chemical and Biological Engineering, University of New Mexico, Albuquerque, New Mexico 87131
| |
Collapse
|
42
|
Joly-Amado A, Davtyan H, Serraneau K, Jules P, Zitnyar A, Pressman E, Zagorski K, Antonyan T, Hovakimyan A, Paek HJ, Gordon MN, Cribbs DH, Petrovsky N, Agadjanyan MG, Ghochikyan A, Morgan D. Active immunization with tau epitope in a mouse model of tauopathy induced strong antibody response together with improvement in short memory and pSer396-tau pathology. Neurobiol Dis 2019; 134:104636. [PMID: 31629891 DOI: 10.1016/j.nbd.2019.104636] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/30/2019] [Accepted: 10/07/2019] [Indexed: 12/16/2022] Open
Abstract
Abnormal tau hyperphosphorylation and its aggregation into neurofibrillary tangles are a hallmark of tauopathies, neurodegenerative disorders that include Alzheimer's disease (AD). Active and passive Tau-immunotherapy has been proposed as a therapeutic approach to AD with mixed results. One of the limitations of active immunotherapy may be associated with the mediocre immunogenicity of vaccines that are not inducing therapeutically potent titers of antibodies. The aim of this study was to test the efficacy of an anti-tau vaccine, AV-1980R/A composed of N terminal peptide of this molecule fused with an immunogenic MultiTEP platform and formulated in a strong adjuvant, AdvaxCpG in a Tg4510 mouse model of tauopathy. Experimental mice were immunized with AV-1980R/A and a control group of mice were injected with adjuvant only. Nontransgenic and tetracycline transactivator (tTA) transgenic littermates were included as baseline controls to contrast with the tau phenotype. Active immunization with AV-1980R/A induced very strong anti-tau humoral immune responses in both nontransgenic and transgenic mice with evidence of IgG in brains of AV-1980R/A vaccinated mice. These experimental animals displayed an improvement in short-term memory during a novel object recognition test. However, impairments in other behavioral tasks were not prevented by AV-1980R/A vaccinations. At the same time, high titers of anti-tau antibodies reduced hyperphosphorylated pSer396 tau but did not lower the level of other phosphorylated tau species in the brains of AV-1980R/A vaccinated mice. These data indicate that active immunotherapy with an N-terminal Tau epitope was only partially effective in improving cognition and reducing pathology in the stringent Tg4510 mouse model of tauopathy.
Collapse
Affiliation(s)
- A Joly-Amado
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA.
| | - H Davtyan
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA; Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - K Serraneau
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| | - P Jules
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| | - A Zitnyar
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| | - E Pressman
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| | - K Zagorski
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - T Antonyan
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - A Hovakimyan
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - H J Paek
- Molecular Pharmacology and Physiology, College of Medicine, University of South Florida, Tampa, FL, USA
| | - M N Gordon
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| | - D H Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA 92697, USA
| | - N Petrovsky
- Flinders Med. Ctr., Bedford Park, Adelaide 5042, Australia
| | - M G Agadjanyan
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - A Ghochikyan
- The Institute for Molecular Medicine, Huntington Beach, CA 92647, USA
| | - D Morgan
- USF Health Byrd Alzheimer's Institute, Tampa, FL 33613, USA
| |
Collapse
|
43
|
Weisová P, Cehlár O, Škrabana R, Žilková M, Filipčík P, Kováčech B, Prčina M, Wojčiaková Ľ, Fialová Ľ, Smolek T, Kontseková E, Žilka N, Novák M. Therapeutic antibody targeting microtubule-binding domain prevents neuronal internalization of extracellular tau via masking neuron surface proteoglycans. Acta Neuropathol Commun 2019; 7:129. [PMID: 31391090 PMCID: PMC6685285 DOI: 10.1186/s40478-019-0770-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 07/15/2019] [Indexed: 02/04/2023] Open
Abstract
Pathologically altered tau protein is a common denominator of neurodegenerative disorders including Alzheimer's disease (AD) and other tauopathies. Therefore, promising immunotherapeutic approaches target and eliminate extracellular pathogenic tau species, which are thought to be responsible for seeding and propagation of tau pathology. Tau isoforms in misfolded states can propagate disease pathology in a template-dependent manner, proposed to be mediated by the release and internalization of extracellular tau. Monoclonal antibody DC8E8, binding four highly homologous and independent epitopes in microtubule-binding domain (MTBD) of diseased tau, inhibits tau-tau interaction, discriminates between healthy and pathologically truncated tau and reduces tau pathology in animal model in vivo. Here, we show that DC8E8 antibody acts via extracellular mechanism and does not influence viability and physiological functions of neurons. Importantly, in vitro functional assays showed that DC8E8 recognises pathogenic tau proteins of different size and origin, and potently blocks their entry into neurons. Next, we examined the mechanisms by which mouse antibody DC8E8 and its humanized version AX004 effectively block the neuronal internalization of extracellular AD tau species. We determined a novel mode of action of a therapeutic candidate antibody, which potently inhibits neuronal internalization of AD tau species by masking of epitopes present in MTBD important for interaction with neuron surface Heparan Sulfate Proteoglycans (HSPGs). We show that interference of tau-heparane sulfate interaction with DC8E8 antibody via steric hindrance represents an efficient and important therapeutic approach halting tau propagation.
Collapse
Affiliation(s)
- Petronela Weisová
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic.
| | - Ondrej Cehlár
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Rostislav Škrabana
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Monika Žilková
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Peter Filipčík
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Branislav Kováčech
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Michal Prčina
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Ľubica Wojčiaková
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Ľubica Fialová
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Tomáš Smolek
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Eva Kontseková
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Norbert Žilka
- Department of Neuroimmunology, Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, Bratislava, Slovak Republic
| | - Michal Novák
- Axon Neuroscience SE, Arch. Makariou & Kalogreon 4, Larnaca, Cyprus
| |
Collapse
|
44
|
Chen D, Drombosky KW, Hou Z, Sari L, Kashmer OM, Ryder BD, Perez VA, Woodard DR, Lin MM, Diamond MI, Joachimiak LA. Tau local structure shields an amyloid-forming motif and controls aggregation propensity. Nat Commun 2019; 10:2493. [PMID: 31175300 PMCID: PMC6555816 DOI: 10.1038/s41467-019-10355-1] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 05/08/2019] [Indexed: 11/09/2022] Open
Abstract
Tauopathies are neurodegenerative diseases characterized by intracellular amyloid deposits of tau protein. Missense mutations in the tau gene (MAPT) correlate with aggregation propensity and cause dominantly inherited tauopathies, but their biophysical mechanism driving amyloid formation is poorly understood. Many disease-associated mutations localize within tau's repeat domain at inter-repeat interfaces proximal to amyloidogenic sequences, such as 306VQIVYK311. We use cross-linking mass spectrometry, recombinant protein and synthetic peptide systems, in silico modeling, and cell models to conclude that the aggregation-prone 306VQIVYK311 motif forms metastable compact structures with its upstream sequence that modulates aggregation propensity. We report that disease-associated mutations, isomerization of a critical proline, or alternative splicing are all sufficient to destabilize this local structure and trigger spontaneous aggregation. These findings provide a biophysical framework to explain the basis of early conformational changes that may underlie genetic and sporadic tau pathogenesis.
Collapse
Affiliation(s)
- Dailu Chen
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Molecular Biophysics Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Kenneth W Drombosky
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Zhiqiang Hou
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Levent Sari
- Green Center for Molecular, Computational and Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Omar M Kashmer
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Bryan D Ryder
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Molecular Biophysics Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Valerie A Perez
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Molecular Biophysics Graduate Program, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - DaNae R Woodard
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Milo M Lin
- Green Center for Molecular, Computational and Systems Biology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
- Department of Biophysics, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Marc I Diamond
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lukasz A Joachimiak
- Center for Alzheimer's and Neurodegenerative Diseases, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
45
|
Katsinelos T, Tuck BJ, Mukadam AS, McEwan WA. The Role of Antibodies and Their Receptors in Protection Against Ordered Protein Assembly in Neurodegeneration. Front Immunol 2019; 10:1139. [PMID: 31214163 PMCID: PMC6554282 DOI: 10.3389/fimmu.2019.01139] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Accepted: 05/07/2019] [Indexed: 02/01/2023] Open
Abstract
Ordered assemblies of proteins are found in the postmortem brains of sufferers of several neurodegenerative diseases. The cytoplasmic microtubule associated protein tau and alpha-synuclein (αS) are found in an assembled state in Alzheimer's disease and Parkinson's disease, respectively. An accumulating body of evidence suggests a "prion-like" mechanism of spread of these assemblies through the diseased brain. Under this hypothesis, assembled variants of these proteins promote the conversion of native proteins to the assembled state. This likely inflicts pathology on cells of the brain through a toxic gain-of-function mechanism. Experiments in animal models of tau and αS pathology have demonstrated that the passive transfer of anti-tau or anti-αS antibodies induces a reduction in the levels of assembled proteins. This is further accompanied by improvements in neurological function and preservation of brain volume. Immunotherapy is therefore considered one of the brightest hopes as a therapeutic avenue in an area currently without disease-modifying therapy. Following a series of disappointing clinical trials targeting beta-amyloid, a peptide that accumulates in the extracellular spaces of the AD brain, attention is turning to active and passive immunotherapies that target tau and αS. However, there are several remaining uncertainties concerning the mechanism by which antibodies afford protection against self-propagating protein conformations. This review will discuss current understanding of how antibodies and their receptors can be brought to bear on proteins involved in neurodegeneration. Parallels will be made to antibody-mediated protection against classical viral infections. Common mechanisms that may contribute to protection against self-propagating protein conformations include blocking the entry of protein "seeds" to cells, clearance of immune complexes by microglia, and the intracellular protein degradation pathway initiated by cytoplasmic antibodies via the Fc receptor TRIM21. As with anti-viral immunity, protective mechanisms may be accompanied by the activation of immune signaling pathways and we will discuss the suitability of such activation in the neurological setting.
Collapse
Affiliation(s)
| | | | | | - William A. McEwan
- Department of Clinical Neurosciences, UK Dementia Research Institute at the University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
46
|
Panza F, Imbimbo BP, Lozupone M, Greco A, Seripa D, Logroscino G, Daniele A, Colosimo C. Disease-modifying therapies for tauopathies: agents in the pipeline. Expert Rev Neurother 2019; 19:397-408. [PMID: 30973276 DOI: 10.1080/14737175.2019.1606715] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Tauopathies are heterogeneous clinicopathological entities characterized by abnormal neuronal and/or glial inclusions of the microtubule-binding protein tau. Primary tauopathies considered to be diseases correspond to a major class of frontotemporal lobar degeneration (FTLD) neuropathology (FTLD-Tau), including several forms of frontotemporal dementia (FTD) clinical syndromes. Little progress has been made in the past 20 years in developing effective disease-modifying drugs for primary tauopathies and available symptomatic treatments have limited efficacy. Areas covered: Potential disease-modifying drugs in clinical development to slow neuropathological progression of primary tauopathies. Expert opinion: Since the underlying pathology of primary tauopathies consists of abnormal tau protein aggregates, treatments are being developed to interfere with the aggregation process or to promote the clearance of this protein. Unfortunately, disease-modifying treatments remain years away as demonstrated by the recent negative Phase III findings of a tau aggregation inhibitor (LMTM) for treating the behavioral variant of FTD. Further evidence will come from ongoing Phase I/II trials on novel drugs and immunotherapeutics with various targets - prevention of deposition or removal of tau aggregates, inhibition of tau phosphorylation/acetylation, modulation of O-GlcNAcylation, activation of autophagy or ubiquitin-proteasome system pathways, and rescue of selected tau loss of function or suppression of tau gene expression.
Collapse
Affiliation(s)
- Francesco Panza
- a Neurodegenerative Disease Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs , University of Bari "Aldo Moro" , Bari , Italy.,b Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain , University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico" , Lecce , Italy.,c Geriatric Unit, Fondazione IRCCS , "Casa Sollievo della Sofferenza" , Foggia , Italy
| | - Bruno P Imbimbo
- d Department of Research and Development , Chiesi Farmaceutici , Parma , Italy
| | - Madia Lozupone
- a Neurodegenerative Disease Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs , University of Bari "Aldo Moro" , Bari , Italy
| | - Antonio Greco
- c Geriatric Unit, Fondazione IRCCS , "Casa Sollievo della Sofferenza" , Foggia , Italy
| | - Davide Seripa
- c Geriatric Unit, Fondazione IRCCS , "Casa Sollievo della Sofferenza" , Foggia , Italy
| | - Giancarlo Logroscino
- a Neurodegenerative Disease Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs , University of Bari "Aldo Moro" , Bari , Italy.,b Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain , University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico" , Lecce , Italy
| | - Antonio Daniele
- e Institute of Neurology , Catholic University of Sacred Heart , Rome , Italy.,f Institute of Neurology , Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
| | - Carlo Colosimo
- g Department of Neurological Sciences , Santa Maria University Hospital , Terni , Italy
| |
Collapse
|
47
|
Bashir MA, Khan AU, Badshah H, Rodrigues-Filho E, Din ZU, Khan A. Synthesis, characterization, molecular docking evaluation, antidepressant, and anti-Alzheimer effects of dibenzylidene ketone derivatives. Drug Dev Res 2019; 80:595-605. [PMID: 30964563 DOI: 10.1002/ddr.21537] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 03/09/2019] [Accepted: 03/24/2019] [Indexed: 01/21/2023]
Abstract
Novel bioactive compounds as synthetic analogs of the potent herbal medicines can be optimized as potential drug candidates for various neurologic disorders. This study was performed to investigate the newly synthesized dibenzylidene ketone derivatives: (2E,6E)-2,6-dibenzylidene cyclohexanone (A1K1) and (1E,4E)-5-(2,3-dichlorophenyl)-1-(4-methoxyphenyl)-2-methylpenta-1,4-diene-3-one (A2K2) and evaluate its potential anti-Alzheimer's and anti-depressant properties. Both the derivatives are chemically characterized by using HNMR and CNMR techniques. Auto Dock Vina program was used to investigate ligand-protein affinity. Forced swim test, tail suspension test, open field test, Y-maze test, and Morris water maze test (MWM) models were employed to evaluate anti-depressant and anti-Alzheimer's activity of dibenzylidene ketone derivatives in mice. Both A1K1 and A2K2 showed high binding affinities against various proteins involved in depression and Alzheimer's mechanisms like monoamine oxidase B, acetylcholinesterase, norepinephrine transporter 2, serotonin transporter, dopamine receptor, serotonin receptor modulator, and beta-amyloid targets. A1K1 and A2K2 dose-dependently (0.1-1 mg/kg) decreased immobility time, while increased swimming and climbing time of mice in forced swim test (FST). A1K1 and A2K2 decreased animal immobility time in TST. In the open field test, both A1K1 and A2K2 increased the number of ambulations and rearings. A1K1 and A2K2 dose-dependently (0.5-1.0 mg/kg) increased spontaneous alternation behavior (%) and the number of entries of mice in Y-maze test. In the MWM test, A1K1 and A2K2 decreased escape latency time. Overall, both in-silico and in-vivo investigations of A1K1 and A2K2, report their therapeutic potential for antidepressant and anti-Alzheimer properties. Hence, these compounds possess potent neuroprotective properties and may be further evaluated for their therapeutic potential in various neurological disorders.
Collapse
Affiliation(s)
- Muhammad Adnan Bashir
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Arif-Ullah Khan
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Haroon Badshah
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan.,Department of Pharmacy, Abdul Wali Khan University, Mardan, Pakistan
| | - Edson Rodrigues-Filho
- Departament of Chemistry, Federal University of Sao Carlos, São Carlos, São Paulo, Brazil
| | - Zia Ud Din
- Departament of Chemistry, Federal University of Sao Carlos, São Carlos, São Paulo, Brazil.,Department of Chemistry, Woman University Swabi, Swabi, Pakistan
| | - Aslam Khan
- Basic Sciences Department, College of Science and Health Professions, King Saud bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
48
|
Melková K, Zapletal V, Narasimhan S, Jansen S, Hritz J, Škrabana R, Zweckstetter M, Ringkjøbing Jensen M, Blackledge M, Žídek L. Structure and Functions of Microtubule Associated Proteins Tau and MAP2c: Similarities and Differences. Biomolecules 2019; 9:biom9030105. [PMID: 30884818 PMCID: PMC6468450 DOI: 10.3390/biom9030105] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/09/2019] [Accepted: 03/13/2019] [Indexed: 12/16/2022] Open
Abstract
The stability and dynamics of cytoskeleton in brain nerve cells are regulated by microtubule associated proteins (MAPs), tau and MAP2. Both proteins are intrinsically disordered and involved in multiple molecular interactions important for normal physiology and pathology of chronic neurodegenerative diseases. Nuclear magnetic resonance and cryo-electron microscopy recently revealed propensities of MAPs to form transient local structures and long-range contacts in the free state, and conformations adopted in complexes with microtubules and filamentous actin, as well as in pathological aggregates. In this paper, we compare the longest, 441-residue brain isoform of tau (tau40), and a 467-residue isoform of MAP2, known as MAP2c. For both molecules, we present transient structural motifs revealed by conformational analysis of experimental data obtained for free soluble forms of the proteins. We show that many of the short sequence motifs that exhibit transient structural features are linked to functional properties, manifested by specific interactions. The transient structural motifs can be therefore classified as molecular recognition elements of tau40 and MAP2c. Their interactions are further regulated by post-translational modifications, in particular phosphorylation. The structure-function analysis also explains differences between biological activities of tau40 and MAP2c.
Collapse
Affiliation(s)
- Kateřina Melková
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
- Faculty of Science, National Centre for Biomolecular Research, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Vojtěch Zapletal
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
- Faculty of Science, National Centre for Biomolecular Research, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Subhash Narasimhan
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Séverine Jansen
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Jozef Hritz
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| | - Rostislav Škrabana
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dúbravská cesta 9, 845 10 Bratislava, Slovakia.
- Axon Neuroscience R&D Services SE, Dvořákovo nábrežie 10, 811 02 Bratislava, Slovakia.
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases (DZNE), Von-Siebold-Str. 3a, 37075 Göttingen, Germany.
- Department of NMR-Based Structural Biology, Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077 Göttingen, Germany.
| | | | | | - Lukáš Žídek
- Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
- Faculty of Science, National Centre for Biomolecular Research, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic.
| |
Collapse
|
49
|
Regional vulnerability and spreading of hyperphosphorylated tau in seeded mouse brain. Neurobiol Dis 2019; 127:398-409. [PMID: 30878534 DOI: 10.1016/j.nbd.2019.03.010] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/01/2019] [Accepted: 03/12/2019] [Indexed: 11/20/2022] Open
Abstract
We have exploited whole brain microscopy to map the progressive deposition of hyperphosphorylated tau in intact, cleared mouse brain. We found that the three-dimensional spreading pattern of hyperphosphorylated tau in the brain of an aging Tau.P301L mouse model did not resemble that observed in AD patients. Injection of synthetic or patient-derived tau fibrils in the CA1 region resulted in a more faithful spreading pattern. Atlas-guided volumetric analysis showed a connectome-dependent spreading from the injection site and also revealed hyperphosphorylated tau deposits beyond the direct anatomical connections. In fibril-injected brains, we also detected a persistent subpopulation of rod-like and swollen microglia. Furthermore, we showed that the hyperphosphorylated tau load could be reduced by intracranial co-administration of, and to a lesser extent, by repeated systemic dosing with an antibody targeting the microtubule-binding domain of tau. Thus, the combination of targeted seeding and in toto staging of tau pathology allowed assessing regional vulnerability in a comprehensive manner, and holds potential as a preclinical drug validation tool.
Collapse
|
50
|
Jadhav S, Avila J, Schöll M, Kovacs GG, Kövari E, Skrabana R, Evans LD, Kontsekova E, Malawska B, de Silva R, Buee L, Zilka N. A walk through tau therapeutic strategies. Acta Neuropathol Commun 2019; 7:22. [PMID: 30767766 PMCID: PMC6376692 DOI: 10.1186/s40478-019-0664-z] [Citation(s) in RCA: 185] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 01/21/2019] [Indexed: 12/18/2022] Open
Abstract
Tau neuronal and glial pathologies drive the clinical presentation of Alzheimer's disease and related human tauopathies. There is a growing body of evidence indicating that pathological tau species can travel from cell to cell and spread the pathology through the brain. Throughout the last decade, physiological and pathological tau have become attractive targets for AD therapies. Several therapeutic approaches have been proposed, including the inhibition of protein kinases or protein-3-O-(N-acetyl-beta-D-glucosaminyl)-L-serine/threonine Nacetylglucosaminyl hydrolase, the inhibition of tau aggregation, active and passive immunotherapies, and tau silencing by antisense oligonucleotides. New tau therapeutics, across the board, have demonstrated the ability to prevent or reduce tau lesions and improve either cognitive or motor impairment in a variety of animal models developing neurofibrillary pathology. The most advanced strategy for the treatment of human tauopathies remains immunotherapy, which has already reached the clinical stage of drug development. Tau vaccines or humanised antibodies target a variety of tau species either in the intracellular or extracellular spaces. Some of them recognise the amino-terminus or carboxy-terminus, while others display binding abilities to the proline-rich area or microtubule binding domains. The main therapeutic foci in existing clinical trials are on Alzheimer's disease, progressive supranuclear palsy and non-fluent primary progressive aphasia. Tau therapy offers a new hope for the treatment of many fatal brain disorders. First efficacy data from clinical trials will be available by the end of this decade.
Collapse
Affiliation(s)
- Santosh Jadhav
- Institute of Neuroimmunology, Slovak Academy of Sciences, Dubravska 9, 845 10, Bratislava, Slovakia
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia
| | - Jesus Avila
- Centro de Biologia Molecular "Severo Ochoa", Consejo Superior de Investigaciones, Cientificas, Universidad Autonoma de Madrid, C/ Nicolas Cabrera, 1. Campus de Cantoblanco, 28049, Madrid, Spain
- Networking Research Center on Neurodegenerative, Diseases (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Michael Schöll
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of, Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
- Dementia Research Centre, University College London, London, UK
| | - Gabor G Kovacs
- Institute of Neurology, Medical University of Vienna, AKH 4J, Währinger Gürtel 18-20, 1097, Vienna, Austria
| | - Enikö Kövari
- Department of Mental Health and Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Rostislav Skrabana
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia
| | - Lewis D Evans
- Gurdon Institute and Department of Biochemistry, University of Cambridge, Cambridge, CB2 1QN, UK
| | - Eva Kontsekova
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia
| | - Barbara Malawska
- Department of Physicochemical Drug Analysis, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688, Cracow, Poland
| | - Rohan de Silva
- Reta Lila Weston Institute and Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, 1 Wakefield Street, London, WC1N 1PJ, UK
| | - Luc Buee
- Universite of Lille, Inserm, CHU-Lille, UMRS1172, Alzheimer & Tauopathies, Place de Verdun, 59045, Lille cedex, France.
| | - Norbert Zilka
- AXON Neuroscience R&D Services SE, Dvorakovo nabrezie 10, 811 02, Bratislava, Slovakia.
| |
Collapse
|