1
|
Karimi-Sani I, Sharifi M, Abolpour N, Lotfi M, Atapour A, Takhshid MA, Sahebkar A. Drug repositioning for Parkinson's disease: An emphasis on artificial intelligence approaches. Ageing Res Rev 2025; 104:102651. [PMID: 39755176 DOI: 10.1016/j.arr.2024.102651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/09/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025]
Abstract
Parkinson's disease (PD) is one of the most incapacitating neurodegenerative diseases (NDDs). PD is the second most common NDD worldwide which affects approximately 1-2 percent of people over 65 years. It is an attractive pursuit for artificial intelligence (AI) to contribute to and evolve PD treatments through drug repositioning by repurposing existing drugs, shelved drugs, or even candidates that do not meet the criteria for clinical trials. A search was conducted in three databases Web of Science, Scopus, and PubMed. We reviewed the data related to the last years (1975-present) to identify those drugs currently being proposed for repositioning in PD. Moreover, we reviewed the present status of the computational approach, including AI/Machine Learning (AI/ML)-powered pharmaceutical discovery efforts and their implementation in PD treatment. It was found that the number of drug repositioning studies for PD has increased recently. Repositioning of drugs in PD is taking off, and scientific communities are increasingly interested in communicating its results and finding effective treatment alternatives for PD. A better chance of success in PD drug discovery has been made possible due to AI/ML algorithm advancements. In addition to the experimentation stage of drug discovery, it is also important to leverage AI in the planning stage of clinical trials to make them more effective. New AI-based models or solutions that increase the success rate of drug development are greatly needed.
Collapse
Affiliation(s)
- Iman Karimi-Sani
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrdad Sharifi
- Emergency Medicine Department, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran; Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Nahid Abolpour
- Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mehrzad Lotfi
- Artificial Intelligence Department, Shiraz University of Medical Sciences, Shiraz, Iran; Medical Imaging Research Center, Department of Radiology, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amir Atapour
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Mohammad-Ali Takhshid
- Division of Medical Biotechnology, Department of Laboratory Sciences, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran; Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College & Hospitals, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Bhatia T, Sharma S. Drug Repurposing: Insights into Current Advances and Future Applications. Curr Med Chem 2025; 32:468-510. [PMID: 37946344 DOI: 10.2174/0109298673266470231023110841] [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: 08/06/2023] [Revised: 09/04/2023] [Accepted: 09/11/2023] [Indexed: 11/12/2023]
Abstract
Drug development is a complex and expensive process that involves extensive research and testing before a new drug can be approved for use. This has led to a limited availability of potential therapeutics for many diseases. Despite significant advances in biomedical science, the process of drug development remains a bottleneck, as all hypotheses must be tested through experiments and observations, which can be timeconsuming and costly. To address this challenge, drug repurposing has emerged as an innovative strategy for finding new uses for existing medications that go beyond their original intended use. This approach has the potential to speed up the drug development process and reduce costs, making it an attractive option for pharmaceutical companies and researchers alike. It involves the identification of existing drugs or compounds that have the potential to be used for the treatment of a different disease or condition. This can be done through a variety of approaches, including screening existing drugs against new disease targets, investigating the biological mechanisms of existing drugs, and analyzing data from clinical trials and electronic health records. Additionally, repurposing drugs can lead to the identification of new therapeutic targets and mechanisms of action, which can enhance our understanding of disease biology and lead to the development of more effective treatments. Overall, drug repurposing is an exciting and promising area of research that has the potential to revolutionize the drug development process and improve the lives of millions of people around the world. The present review provides insights on types of interaction, approaches, availability of databases, applications and limitations of drug repurposing.
Collapse
Affiliation(s)
- Trisha Bhatia
- School of Pharmacy, National Forensic Sciences University, Gandhinagar, Gujarat, 382007, India
| | - Shweta Sharma
- School of Pharmacy, National Forensic Sciences University, Gandhinagar, Gujarat, 382007, India
| |
Collapse
|
3
|
Bellini G, D'Antongiovanni V, Palermo G, Antonioli L, Fornai M, Ceravolo R, Bernardini N, Derkinderen P, Pellegrini C. α-Synuclein in Parkinson's Disease: From Bench to Bedside. Med Res Rev 2024. [PMID: 39704040 DOI: 10.1002/med.22091] [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: 05/12/2024] [Revised: 09/24/2024] [Accepted: 11/07/2024] [Indexed: 12/21/2024]
Abstract
α-Synuclein (α-syn), a pathological hallmark of PD, is emerging as a bridging element at the crossroads between neuro/immune-inflammatory responses and neurodegeneration in PD. Several evidence show that pathological α-syn accumulates in neuronal and non-neuronal cells (i.e., neurons, microglia, macrophages, skin cells, and intestinal cells) in central and peripheral tissues since the prodromal phase of the disease, contributing to brain pathology. Indeed, pathological α-syn deposition can promote neurogenic/immune-inflammatory responses that contribute to systemic and central neuroinflammation associated with PD. After providing an overview of the structure and functions of physiological α-syn as well as its pathological forms, we review current studies about the role of neuronal and non-neuronal α-syn at the crossroads between neuroinflammation and neurodegeneration in PD. In addition, we provide an overview of the correlation between the accumulation of α-syn in central and peripheral tissues and PD, related symptoms, and neuroinflammation. Special attention was paid to discussing whether targeting α-syn can represent a suitable therapeutical approach for PD.
Collapse
Affiliation(s)
- Gabriele Bellini
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Department of Neurology, The Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, NYU Langone Health, New York City, New York, USA
| | - Vanessa D'Antongiovanni
- Unit of Histology and Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giovanni Palermo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Luca Antonioli
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Roberto Ceravolo
- Center for Neurodegenerative Diseases, Unit of Neurology, Parkinson's Disease and Movement Disorders, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Nunzia Bernardini
- Unit of Histology and Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Pascal Derkinderen
- Department of Neurology, Nantes Université, CHU Nantes, INSERM, Nantes, France
| | - Carolina Pellegrini
- Unit of Histology and Embryology, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
4
|
Huang C, Zheng X, Yan S, Zhang Z. Advances in Clinical Therapies for Huntington's Disease and the Promise of Multi-Targeted/Functional Drugs Based on Clinicaltrials.gov. Clin Pharmacol Ther 2024; 116:1452-1471. [PMID: 38863261 DOI: 10.1002/cpt.3341] [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] [Received: 03/08/2024] [Accepted: 05/27/2024] [Indexed: 06/13/2024]
Abstract
Huntington's disease (HD) is a dominantly inherited neurodegenerative disorder characterized by a triad of motor, cognitive, and psychiatric problems. Caused by CAG repeat expansion in the huntingtin gene (HTT), the disease involves a complex network of pathogenic mechanisms, including synaptic dysfunction, impaired autophagy, neuroinflammation, oxidative damage, mitochondrial dysfunction, and extrasynaptic excitotoxicity. Although current therapies targeting the pathogenesis of HD primarily aim to reduce mHTT levels by targeting HTT DNA, RNA, or proteins, these treatments only ameliorate downstream pathogenic effects. While gene therapies, such as antisense oligonucleotides, small interfering RNAs and gene editing, have emerged in the field of HD treatment, their safety and efficacy are still under debate. Therefore, pharmacological therapy remains the most promising breakthrough, especially multi-target/functional drugs, which have diverse pharmacological effects. This review summarizes the latest progress in HD drug development based on clinicaltrials.gov search results (Search strategy: key word "Huntington's disease" in HD clinical investigational drugs registered as of December 31, 2023), and highlights the key role of multi-target/functional drugs in HD treatment strategies.
Collapse
Affiliation(s)
- Chunhui Huang
- School of Medicine, Jinan University, Guangzhou, China
- Guangdong Key Laboratory of Non-Human Primate Models, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic, Constituents of TCM and New Drugs Research and Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Xiao Zheng
- Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Sen Yan
- School of Medicine, Jinan University, Guangzhou, China
- Guangdong Key Laboratory of Non-Human Primate Models, Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou, China
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic, Constituents of TCM and New Drugs Research and Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| | - Zaijun Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Province Key Laboratory of Pharmacodynamic, Constituents of TCM and New Drugs Research and Institute of New Drug Research, College of Pharmacy, Jinan University, Guangzhou, China
| |
Collapse
|
5
|
Ahanger IA, Dar TA. Small molecule modulators of alpha-synuclein aggregation and toxicity: Pioneering an emerging arsenal against Parkinson's disease. Ageing Res Rev 2024; 101:102538. [PMID: 39389237 DOI: 10.1016/j.arr.2024.102538] [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: 06/21/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/12/2024]
Abstract
Parkinson's disease (PD) is primarily characterized by loss of dopaminergic neurons in the substantia nigra pars compacta region of the brain and accumulation of aggregated forms of alpha-synuclein (α-Syn), an intrinsically disordered protein, in the form of Lewy Bodies and Lewy Neurites. Substantial evidences point to the aggregated/fibrillar forms of α-Syn as a central event in PD pathogenesis, underscoring the modulation of α-Syn aggregation as a promising strategy for PD treatment. Consequently, numerous anti-aggregation agents, spanning from small molecules to polymers, have been scrutinized for their potential to mitigate α-Syn aggregation and its associated toxicity. Among these, small molecule modulators like osmoprotectants, polyphenols, cellular metabolites, metals, and peptides have emerged as promising candidates with significant potential in PD management. This article offers a comprehensive overview of the effects of these small molecule modulators on the aggregation propensity and associated toxicity of α-Syn and its PD-associated mutants. It serves as a valuable resource for identifying and developing potent, non-invasive, non-toxic, and highly specific small molecule-based therapeutic arsenal for combating PD. Additionally, it raises pertinent questions aimed at guiding future research endeavours in the field of α-Syn aggregation remodelling.
Collapse
Affiliation(s)
- Ishfaq Ahmad Ahanger
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India.
| | - Tanveer Ali Dar
- Department of Clinical Biochemistry, University of Kashmir, Srinagar, Jammu and Kashmir 190006, India.
| |
Collapse
|
6
|
Faouzi A, Arnaud A, Hallé F, Roussel J, Aymard M, Denavit V, Do CV, Mularoni A, Salah M, ElHady A, Pham TN, Bancet A, Le Borgne M, Terreux R, Barret R, Engel M, Lomberget T. Design, synthesis, and structure-activity relationship studies of 6 H-benzo[ b]indeno[1,2- d]thiophen-6-one derivatives as DYRK1A/CLK1/CLK4/haspin inhibitors. RSC Med Chem 2024; 16:d4md00537f. [PMID: 39430953 PMCID: PMC11487425 DOI: 10.1039/d4md00537f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 09/06/2024] [Indexed: 10/22/2024] Open
Abstract
A series of sulfur-containing tetracycles was designed and evaluated for their ability to inhibit protein kinase DYRK1A, a target known to have several potential therapeutic applications including cancers, Down syndrome or Alzheimer's disease. Our medicinal chemistry strategy relied on the design of new compounds using ring contraction/isosteric replacement and constrained analogy of known DYRK1A inhibitors, thus resulting in their DYRK1A inhibitory activity enhancement. Whereas a good inhibitory effect of targeted DYRK1A protein was observed for 5-hydroxy compounds 4i-k (IC50 = 35-116 nM) and the 5-methoxy derivative 4e (IC50 = 52 nM), a fairly good selectivity towards its known DYRK1B off-target was observed for 4k. In addition, the most active compound 4k, having an ATP-competitive mechanism of action, proved to be also a potent inhibitor of CLK1/CLK4 (IC50 = 20 and 26 nM) and, to a lesser extent, of haspin (IC50 = 76 nM) kinases. In silico docking studies within the DYRK1A, CLK1/CLK4 and haspin ATP binding sites were carried out to understand the interactions of our tetracyclic derivatives 4 with these targets. Antiproliferative activities on U87/U373 glioblastoma cell lines of the most potent compound 4k showed a moderate effect (IC50 values between 33 and 46 μM). Microsomal stabilities of the designed compounds 4a-m were also investigated, showing great disparities, depending on benzo[b]thiophene ring 5-substitution.
Collapse
Affiliation(s)
- Abdelfattah Faouzi
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Alexandre Arnaud
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - François Hallé
- Universite Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie, ISPB 8, avenue Rockefeller F-69373 Lyon Cedex 08 France
| | - Jean Roussel
- Universite Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie, ISPB 8, avenue Rockefeller F-69373 Lyon Cedex 08 France
| | - Mandy Aymard
- Universite Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie, ISPB 8, avenue Rockefeller F-69373 Lyon Cedex 08 France
| | - Vincent Denavit
- Universite Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie, ISPB 8, avenue Rockefeller F-69373 Lyon Cedex 08 France
| | - Cong Viet Do
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
- University of Science and Technology of HanoÏ USTH 18 Hoang Quoc Viet Hanoi 100000 Vietnam
| | - Angélique Mularoni
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Mohamed Salah
- Pharmaceutical and Medicinal Chemistry, Saarland University Campus C2.3 D-66123 Saarbrücken Germany
- Department of Pharmaceutical Chemistry, School of Pharmacy, Newgiza University (NGU) Newgiza, km 22 Cairo-Alexandria Desert Road 12577 Cairo Egypt
| | - Ahmed ElHady
- Pharmaceutical and Medicinal Chemistry, Saarland University Campus C2.3 D-66123 Saarbrücken Germany
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation New Administrative Capital Cairo 11865 Egypt
| | - Thanh-Nhat Pham
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Alexandre Bancet
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Marc Le Borgne
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Raphaël Terreux
- Universite Claude Bernard Lyon 1, CNRS UMR 5305, LBTI, ECMO Team, Institut de Biologie et Chimie des Protéines 7 Passage du Vercors 69367 Lyon Cedex 07 France
| | - Roland Barret
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| | - Matthias Engel
- Pharmaceutical and Medicinal Chemistry, Saarland University Campus C2.3 D-66123 Saarbrücken Germany
| | - Thierry Lomberget
- Universite Claude Bernard Lyon 1, CNRS UMR 5246, Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), COSSBA Team, Faculté de Pharmacie, ISPB 8, avenue Rockefeller F-69373 Lyon Cedex 08 France
- Universite Claude Bernard Lyon 1, EA 4446 Bioactive Molecules and Medicinal Chemistry, Faculté de Pharmacie, ISPB, Univ Lyon F-69373 Lyon Cedex 08 France
| |
Collapse
|
7
|
Dai L, Liu M, Ke W, Chen L, Fang X, Zhang Z. Lysosomal dysfunction in α-synuclein pathology: molecular mechanisms and therapeutic strategies. Cell Mol Life Sci 2024; 81:382. [PMID: 39223418 PMCID: PMC11368888 DOI: 10.1007/s00018-024-05419-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
In orchestrating cell signaling, facilitating plasma membrane repair, supervising protein secretion, managing waste elimination, and regulating energy consumption, lysosomes are indispensable guardians that play a crucial role in preserving intracellular homeostasis. Neurons are terminally differentiated post-mitotic cells. Neuronal function and waste elimination depend on normal lysosomal function. Converging data suggest that lysosomal dysfunction is a critical event in the etiology of Parkinson's disease (PD). Mutations in Glucosylceramidase Beta 1 (GBA1) and leucine-rich repeat kinase 2 (LRRK2) confer an increased risk for the development of parkinsonism. Furthermore, lysosomal dysfunction has been observed in the affected neurons of sporadic PD (sPD) patients. Given that lysosomal hydrolases actively contribute to the breakdown of impaired organelles and misfolded proteins, any compromise in lysosomal integrity could incite abnormal accumulation of proteins, including α-synuclein, the major component of Lewy bodies in PD. Clinical observations have shown that lysosomal protein levels in cerebrospinal fluid may serve as potential biomarkers for PD diagnosis and as signs of lysosomal dysfunction. In this review, we summarize the current evidence regarding lysosomal dysfunction in PD and discuss the intimate relationship between lysosomal dysfunction and pathological α-synuclein. In addition, we discuss therapeutic strategies that target lysosomes to treat PD.
Collapse
Affiliation(s)
- Lijun Dai
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Miao Liu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wei Ke
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Liam Chen
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, USA
| | - Xin Fang
- Department of Neurology, the First Affiliated Hospital of Nanchang University, Nanchang, 330000, China.
| | - Zhentao Zhang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
- TaiKang Center for Life and Medical Science, Wuhan University, Wuhan, 430000, China.
| |
Collapse
|
8
|
Das V, Miller JH, Alladi CG, Annadurai N, De Sanctis JB, Hrubá L, Hajdúch M. Antineoplastics for treating Alzheimer's disease and dementia: Evidence from preclinical and observational studies. Med Res Rev 2024; 44:2078-2111. [PMID: 38530106 DOI: 10.1002/med.22033] [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: 03/02/2023] [Revised: 02/15/2024] [Accepted: 03/04/2024] [Indexed: 03/27/2024]
Abstract
As the world population ages, there will be an increasing need for effective therapies for aging-associated neurodegenerative disorders, which remain untreatable. Dementia due to Alzheimer's disease (AD) is one of the leading neurological diseases in the aging population. Current therapeutic approaches to treat this disorder are solely symptomatic, making the need for new molecular entities acting on the causes of the disease extremely urgent. One of the potential solutions is to use compounds that are already in the market. The structures have known pharmacokinetics, pharmacodynamics, toxicity profiles, and patient data available in several countries. Several drugs have been used successfully to treat diseases different from their original purposes, such as autoimmunity and peripheral inflammation. Herein, we divulge the repurposing of drugs in the area of neurodegenerative diseases, focusing on the therapeutic potential of antineoplastics to treat dementia due to AD and dementia. We briefly touch upon the shared pathological mechanism between AD and cancer and drug repurposing strategies, with a focus on artificial intelligence. Next, we bring out the current status of research on the development of drugs, provide supporting evidence from retrospective, clinical, and preclinical studies on antineoplastic use, and bring in new areas, such as repurposing drugs for the prion-like spreading of pathologies in treating AD.
Collapse
Affiliation(s)
- Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - John H Miller
- School of Biological Sciences and Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Charanraj Goud Alladi
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Narendran Annadurai
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
| | - Juan Bautista De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - Lenka Hrubá
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University and University Hospital Olomouc, Olomouc, Czech Republic
- Czech Advanced Technologies and Research Institute (CATRIN), Institute of Molecular and Translational Medicine, Palacký University Olomouc, Olomouc, Czech Republic
| |
Collapse
|
9
|
Gosch Berton G, Cyntia Lima Fonseca Rodrigues A, Dos Santos Borges R, Rodrigues Cardoso N, de Oliveira TA, Oliveira Marques MV. Abelson Tyrosine Kinase Inhibitors in Parkinson's Disease and Lewy Body Dementia: A Systematic Review, Meta-analysis, and Meta-regression. Clin Neuropharmacol 2024; 47:113-119. [PMID: 39008541 DOI: 10.1097/wnf.0000000000000597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
BACKGROUND Alpha-synucleinopathies are incurable neurodegenerative diseases. Abelson tyrosine kinase inhibitors (Abl TKIs) may be disease-modifying therapies. This systematic review, meta-analysis, and meta-regression evaluated the use of Abl TKIs in their treatment. METHODS We searched PubMed, Embase, and Cochrane databases for trials using Abl TKIs in patients with Parkinson's disease and Lewy body dementia published until July 2023. The outcome was the change in the MDS-UPDRS-III (Movement Disorder Society-Sponsored Revision of the Unified Parkinson's Disease Rating Scale III). DerSimonian-Laird random-effects model was used to calculate the pooled effect estimates. Leave-one-out forest plots were used for the sensitivity analysis, and meta-regression (restricted maximum likelihood) was performed. RESULTS Five studies (197 patients) were included. Nilotinib 300 mg had an effect size of -1.154 (95% confidence interval [CI], -3.000 to 0.692). Nilotinib 150 mg and bosutinib 100 mg versus placebo yielded 0.82 (95% CI, -3.76 to 5.41). Sensitivity analysis showed that 1 trial changed the significance of the nilotinib 300 mg single-arm analysis (MD = -1.723; 95% CI, -2.178 to -1.268). Meta-regression revealed that lower age (EC = -0.9103, SE = 0.2286, P < 0.0001) and higher baseline MDS-UPDRS-III scores (EC = 0.1210, SE = 0.0168, P < 0.0001) could explain the inefficacy of nilotinib 300 mg. CONCLUSIONS Nilotinib (300 mg) proved effective postsensitivity analysis, unlike lower doses and bosutinib in Parkinson's disease/Lewy body dementia. Abl TKIs showed reduced efficacy in younger, more impaired patients, indicating the need for further testing with higher-potency drugs in patients who have diseases that are in the early stage but with a later onset.
Collapse
|
10
|
Lin YM, Banoth D, Wali MH, Bekova K, Abdulla N, Gurugubelli S, Khan S. The Role of Alpha-Synuclein and Tubulin-Associated Unit (Tau) Proteins in the Diagnosis, Prognosis, and Treatment of Parkinson's Disease: A Systematic Review. Cureus 2024; 16:e64766. [PMID: 39156411 PMCID: PMC11330161 DOI: 10.7759/cureus.64766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
Parkinson's disease (PD) is a degenerative neurological disorder resulting from the death of dopaminergic neurons, which, in turn, results in impaired motor and cognitive functions. Early diagnosis is important in achieving a good prognosis for PD. Currently, the only approved way to diagnose PD is through medical history, current symptoms, and neurological examination. This, however, can only happen after PD progresses far enough in patients. Biomarkers in cerebrospinal fluid (CSF) and blood plasma, however, may provide insight into the early progress of PD and potentially concurrent dementia, which can also aid in the development of novel treatments. Specifically, this systematic review explores alpha-synuclein (α-syn) and tubulin-associated unit (Tau) proteins and analyzes their potential roles as biomarkers while also touching on nilotinib and immunotherapy as potential treatment options. PubMed, PubMed Central (PMC), Medline, and Cochrane Library serve as the databases for relevant literature, upon which eligibility criteria and quality checks - Assessment of Multiple Systematic Review (AMSTAR) tool, Newcastle-Ottawa Quality Assessment Scale, Cochrane risk-of-bias assessment 2 (RoB2), and Scale for the Assessment of Narrative Review (SANRA) - were applied. The remaining literature examines the various aspects of PD and Parkinson's disease dementia (PDD) and associated biomarkers. From 10 studies, 2,361 participants, both PD patients and healthy controls (HCs), were assessed and compared. Various assessment scales, such as the Unified Parkinson's Disease Rating Scale part III (UPDRS III), were used to ascertain the severity or progression of PD in patients while also seeking a noticeable correlation with α-syn and total Tau (t-Tau). The lack of standardized clinical testing has led to conflicting reports. Thus, while the articles generally agree on the potential efficacy of α-syn and Tau protein analysis in the diagnosis, prognosis, and treatment of PD and PDD, they also argue for further testing and trials.
Collapse
Affiliation(s)
- Yi Mon Lin
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Devendar Banoth
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Muhammad Hassaan Wali
- Surgery, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Khava Bekova
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Noor Abdulla
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Simhachalam Gurugubelli
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Safeera Khan
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| |
Collapse
|
11
|
Lee HJ, Choi HJ, Jeong YJ, Na YH, Hong JT, Han JM, Hoe HS, Lim KH. Developing theragnostics for Alzheimer's disease: Insights from cancer treatment. Int J Biol Macromol 2024; 269:131925. [PMID: 38685540 DOI: 10.1016/j.ijbiomac.2024.131925] [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: 01/01/2024] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2024]
Abstract
The prevalence of Alzheimer's disease (AD) and its associated economic and societal burdens are on the rise, but there are no curative treatments for AD. Interestingly, this neurodegenerative disease shares several biological and pathophysiological features with cancer, including cell-cycle dysregulation, angiogenesis, mitochondrial dysfunction, protein misfolding, and DNA damage. However, the genetic factors contributing to the overlap in biological processes between cancer and AD have not been actively studied. In this review, we discuss the shared biological features of cancer and AD, the molecular targets of anticancer drugs, and therapeutic approaches. First, we outline the common biological features of cancer and AD. Second, we describe several anticancer drugs, their molecular targets, and their effects on AD pathology. Finally, we discuss how protein-protein interactions (PPIs), receptor inhibition, immunotherapy, and gene therapy can be exploited for the cure and management of both cancer and AD. Collectively, this review provides insights for the development of AD theragnostics based on cancer drugs and molecular targets.
Collapse
Affiliation(s)
- Hyun-Ju Lee
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Hee-Jeong Choi
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea
| | - Yoo Joo Jeong
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333, Techno jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea
| | - Yoon-Hee Na
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea
| | - Jin Tae Hong
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea
| | - Ji Min Han
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea.
| | - Hyang-Sook Hoe
- Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu 41062, Republic of Korea; Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology (DGIST), 333, Techno jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu 42988, Republic of Korea.
| | - Key-Hwan Lim
- College of Pharmacy, Chungbuk National University, Cheongju-si 28160, Republic of Korea.
| |
Collapse
|
12
|
Mansour HM. The interference between SARS-COV-2 and Alzheimer's disease: Potential immunological and neurobiological crosstalk from a kinase perspective reveals a delayed pandemic. Ageing Res Rev 2024; 94:102195. [PMID: 38244862 DOI: 10.1016/j.arr.2024.102195] [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: 11/14/2022] [Revised: 01/06/2024] [Accepted: 01/08/2024] [Indexed: 01/22/2024]
Abstract
Coronavirus disease 2019 (COVID-19) has infected over 700 million people, with up to 30% developing neurological manifestations, including dementias. However, there is a lack of understanding of common molecular brain markers causing Alzheimer's disease (AD). COVID-19 has etiological cofactors with AD, making patients with AD a vulnerable population at high risk of experiencing more severe symptoms and worse consequences. Both AD and COVID-19 have upregulated several shared kinases, leading to the repositioning of kinase inhibitors (KIs) for the treatment of both diseases. This review provides an overview of the interactions between the immune system and the nervous system in relation to receptor tyrosine kinases, including epidermal growth factor receptors, vascular growth factor receptors, and non-receptor tyrosine kinases such as Bruton tyrosine kinase, spleen tyrosine kinase, c-ABL, and JAK/STAT. We will discuss the promising results of kinase inhibitors in pre-clinical and clinical studies for both COVID-19 and Alzheimer's disease (AD), as well as the challenges in repositioning KIs for these diseases. Understanding the shared kinases between AD and COVID-19 could help in developing therapeutic approaches for both.
Collapse
Affiliation(s)
- Heba M Mansour
- General Administration of Innovative Products, Central Administration of Biological, Innovative Products, and Clinical Studies (Bio-INN), Egyptian Drug Authority (EDA), Giza, Egypt.
| |
Collapse
|
13
|
Miao Y, Meng H. The involvement of α-synucleinopathy in the disruption of microglial homeostasis contributes to the pathogenesis of Parkinson's disease. Cell Commun Signal 2024; 22:31. [PMID: 38216911 PMCID: PMC10785555 DOI: 10.1186/s12964-023-01402-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/18/2023] [Indexed: 01/14/2024] Open
Abstract
The intracellular deposition and intercellular transmission of α-synuclein (α-syn) are shared pathological characteristics among neurodegenerative disorders collectively known as α-synucleinopathies, including Parkinson's disease (PD). Although the precise triggers of α-synucleinopathies remain unclear, recent findings indicate that disruption of microglial homeostasis contributes to the pathogenesis of PD. Microglia play a crucial role in maintaining optimal neuronal function by ensuring a homeostatic environment, but this function is disrupted during the progression of α-syn pathology. The involvement of microglia in the accumulation, uptake, and clearance of aggregated proteins is critical for managing disease spread and progression caused by α-syn pathology. This review summarizes current knowledge on the interrelationships between microglia and α-synucleinopathies, focusing on the remarkable ability of microglia to recognize and internalize extracellular α-syn through diverse pathways. Microglia process α-syn intracellularly and intercellularly to facilitate the α-syn neuronal aggregation and cell-to-cell propagation. The conformational state of α-synuclein distinctly influences microglial inflammation, which can affect peripheral immune cells such as macrophages and lymphocytes and may regulate the pathogenesis of α-synucleinopathies. We also discuss ongoing research efforts to identify potential therapeutic approaches targeting both α-syn accumulation and inflammation in PD. Video Abstract.
Collapse
Affiliation(s)
- Yongzhen Miao
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China
| | - Hongrui Meng
- Institute of Neuroscience, Soochow University, Suzhou, Jiangsu, China.
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
14
|
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
|
15
|
Wyse RK, Isaacs T, Barker RA, Cookson MR, Dawson TM, Devos D, Dexter DT, Duffen J, Federoff H, Fiske B, Foltynie T, Fox S, Greenamyre JT, Kieburtz K, Kordower JH, Krainc D, Matthews H, Moore DJ, Mursaleen L, Schwarzschild MA, Stott SR, Sulzer D, Svenningsson P, Tanner CM, Carroll C, Simon DK, Brundin P. Twelve Years of Drug Prioritization to Help Accelerate Disease Modification Trials in Parkinson's Disease: The International Linked Clinical Trials Initiative. JOURNAL OF PARKINSON'S DISEASE 2024; 14:657-666. [PMID: 38578902 PMCID: PMC11191436 DOI: 10.3233/jpd-230363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/10/2024] [Indexed: 04/07/2024]
Abstract
In 2011, the UK medical research charity Cure Parkinson's set up the international Linked Clinical Trials (iLCT) committee to help expedite the clinical testing of potentially disease modifying therapies for Parkinson's disease (PD). The first committee meeting was held at the Van Andel Institute in Grand Rapids, Michigan in 2012. This group of PD experts has subsequently met annually to assess and prioritize agents that may slow the progression of this neurodegenerative condition, using a systematic approach based on preclinical, epidemiological and, where possible, clinical data. Over the last 12 years, 171 unique agents have been evaluated by the iLCT committee, and there have been 21 completed clinical studies and 20 ongoing trials associated with the initiative. In this review, we briefly outline the iLCT process as well as the clinical development and outcomes of some of the top prioritized agents. We also discuss a few of the lessons that have been learnt, and we conclude with a perspective on what the next decade may bring, including the introduction of multi-arm, multi-stage clinical trial platforms and the possibility of combination therapies for PD.
Collapse
Affiliation(s)
| | | | - Roger A. Barker
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - David Devos
- Department of Medical Pharmacology and Neurology, University of Lille, CHU Lille, Lille Neurosciences and Cognition Inserm UMR-S-U1172, Lille, France
| | | | | | - Howard Federoff
- Henry and Susan Samueli College of Health Sciences, University of California, Irvine CA, USA
| | - Brian Fiske
- Research Programs, The Michael J. Fox Foundation for Parkinson’s Research, New York, NY, USA
| | - Thomas Foltynie
- Department of Clinical and Movement Neurosciences, UCL Institute of Neurology, National Hospital for Neurology and Neurosurgery, London, UK
| | - Susan Fox
- Edmond J. Safra Program in Parkinson’s Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, University Health Network, Division of Neurology, University of Toronto, Toronto, ON, Canada
| | - J. Timothy Greenamyre
- Department of Neurology, Pittsburgh Institute for Neurodegenerative Diseases, University of Pittsburgh, Pittsburgh, PA, USA
| | - Karl Kieburtz
- Department of Neurology Center for Health and Technology, University of Rochester, Rochester, NY, USA
| | - Jeffrey H. Kordower
- ASU-Banner Neurodegenerative Disease Research Center and School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Dimitri Krainc
- Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | | | | | | | | | - David Sulzer
- Department of Neurology, Columbia University, New York, NY, USA
| | | | - Caroline M. Tanner
- Department of Neurology, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - Camille Carroll
- Translational and Clinical Research Institute, Newcastle University, Newcastle, UK
| | - David K. Simon
- Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Patrik Brundin
- Neuroscience and Rare Diseases, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| |
Collapse
|
16
|
Khan T, Waseem R, Shahid M, Ansari J, Ahanger IA, Hassan I, Islam A. Recent advancement in therapeutic strategies for Alzheimer's disease: Insights from clinical trials. Ageing Res Rev 2023; 92:102113. [PMID: 37918760 DOI: 10.1016/j.arr.2023.102113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/04/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, characterized by the presence of plaques of amyloid beta and Tau proteins. There is currently no permanent cure for AD; the only medications approved by the FDA for mild to moderate AD are cholinesterase inhibitors, NMDA receptor antagonists, and immunotherapies against core pathophysiology, that provide temporary relief only. Researchers worldwide have made significant attempts to find new targets and develop innovative therapeutic molecules to treat AD. The FDA-approved drugs are palliative and couldn't restore the damaged neuron cells of AD. Stem cells have self-differentiation properties, making them prospective therapeutics to treat AD. The promising results in pre-clinical studies of stem cell therapy for AD seek attention worldwide. Various stem cells, mainly mesenchymal stem cells, are currently in different phases of clinical trials and need more advancements to take this therapy to the translational level. Here, we review research from the past decade that has identified several hypotheses related to AD pathology. Moreover, this article also focuses on the recent advancement in therapeutic strategies for AD treatment including immunotherapy and stem cell therapy detailing the clinical trials that are currently undergoing development.
Collapse
Affiliation(s)
- Tanzeel Khan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Rashid Waseem
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Mohammad Shahid
- Department of Basic Medical Sciences, College of Medicine, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Jaoud Ansari
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Ishfaq Ahmad Ahanger
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India; Department of Clinical Biochemistry, University of Kashmir,190006, India
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
17
|
Tolea MI, Ezzeddine R, Camacho S, Galvin JE. Emerging drugs for dementia with Lewy Bodies: a review of Phase II & III trials. Expert Opin Emerg Drugs 2023; 28:167-180. [PMID: 37531299 DOI: 10.1080/14728214.2023.2244425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/04/2023]
Abstract
INTRODUCTION Despite faster cognitive decline and greater negative impact on patients and family caregivers, drug development efforts in Dementia with Lewy Bodies (DLB) fall behind those for Alzheimer's Disease (AD). Current off-label drug DLB treatment options are limited to symptomatic agents developed to address cognitive deficits in AD, motor deficits in Parkinson's Disease, or behavioral symptoms in psychiatric disease. Aided by recent improvements in DLB diagnosis, a new focus on the development of disease-modifying agents (DMA) is emerging. AREAS COVERED Driven by evidence supporting different pathological mechanisms in DLB and PDD, this review assesses the evidence on symptomatic drug treatments and describes current efforts in DMA development in DLB. Specifically, our goals were to: (1) review evidence supporting the use of symptomatic drug treatments in DLB; (2) review the current DMA pipeline in DLB with a focus on Phase II and III clinical trials; and (3) identify potential issues with the development of DMA in DLB. Included in this review were completed and ongoing drug clinical trials in DLB registered on ClinicalTrials.gov (no time limits set for the search) or disseminated at the 2023 international conference on Clinical Trials in AD. Drug clinical trials registered in non-US clinical trial registries were not included. EXPERT OPINION Adoption of current symptomatic drug treatments used off-label in DLB relied on efficacy of benefits in other disorders rather than evidence from randomized controlled clinical trials. Symptoms remain difficult to manage. Several DMA drugs are currently being evaluated as either repurposing candidates or novel small molecules. Continued improvement in methodological aspects including development of DLB-specific outcome measures and biomarkers is needed to move the field of DMA drug development forward.
Collapse
Affiliation(s)
- Magdalena I Tolea
- Comprehensive Center for Brain Health, Lewy Body Dementia Research Center of Excellence, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Reem Ezzeddine
- Comprehensive Center for Brain Health, Lewy Body Dementia Research Center of Excellence, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Simone Camacho
- Comprehensive Center for Brain Health, Lewy Body Dementia Research Center of Excellence, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - James E Galvin
- Comprehensive Center for Brain Health, Lewy Body Dementia Research Center of Excellence, Department of Neurology, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
18
|
Makeeva VS, Dyrkheeva NS, Lavrik OI, Zakian SM, Malakhova AA. Mutant-Huntingtin Molecular Pathways Elucidate New Targets for Drug Repurposing. Int J Mol Sci 2023; 24:16798. [PMID: 38069121 PMCID: PMC10706709 DOI: 10.3390/ijms242316798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 11/18/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The spectrum of neurodegenerative diseases known today is quite extensive. The complexities of their research and treatment lie not only in their diversity. Even many years of struggle and narrowly focused research on common pathologies such as Alzheimer's, Parkinson's, and other brain diseases have not brought cures for these illnesses. What can be said about orphan diseases? In particular, Huntington's disease (HD), despite affecting a smaller part of the human population, still attracts many researchers. This disorder is known to result from a mutation in the HTT gene, but having this information still does not simplify the task of drug development and studying the mechanisms of disease progression. Nonetheless, the data accumulated over the years and their analysis provide a good basis for further research. Here, we review studies devoted to understanding the mechanisms of HD. We analyze genes and molecular pathways involved in HD pathogenesis to describe the action of repurposed drugs and try to find new therapeutic targets.
Collapse
Affiliation(s)
- Vladlena S. Makeeva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia; (V.S.M.); (S.M.Z.); (A.A.M.)
| | - Nadezhda S. Dyrkheeva
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 8 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia;
| | - Olga I. Lavrik
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of Russian Academy of Sciences, 8 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia;
| | - Suren M. Zakian
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia; (V.S.M.); (S.M.Z.); (A.A.M.)
| | - Anastasia A. Malakhova
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, 10 Akad. Lavrentiev Ave., 630090 Novosibirsk, Russia; (V.S.M.); (S.M.Z.); (A.A.M.)
| |
Collapse
|
19
|
Bigi A, Cascella R, Cecchi C. α-Synuclein oligomers and fibrils: partners in crime in synucleinopathies. Neural Regen Res 2023; 18:2332-2342. [PMID: 37282450 PMCID: PMC10360081 DOI: 10.4103/1673-5374.371345] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023] Open
Abstract
The misfolding and aggregation of α-synuclein is the general hallmark of a group of devastating neurodegenerative pathologies referred to as synucleinopathies, such as Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. In such conditions, a range of different misfolded aggregates, including oligomers, protofibrils, and fibrils, are present both in neurons and glial cells. Growing experimental evidence supports the proposition that soluble oligomeric assemblies, formed during the early phases of the aggregation process, are the major culprits of neuronal toxicity; at the same time, fibrillar conformers appear to be the most efficient at propagating among interconnected neurons, thus contributing to the spreading of α-synuclein pathology. Moreover, α-synuclein fibrils have been recently reported to release soluble and highly toxic oligomeric species, responsible for an immediate dysfunction in the recipient neurons. In this review, we discuss the current knowledge about the plethora of mechanisms of cellular dysfunction caused by α-synuclein oligomers and fibrils, both contributing to neurodegeneration in synucleinopathies.
Collapse
Affiliation(s)
- Alessandra Bigi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Roberta Cascella
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| | - Cristina Cecchi
- Department of Experimental and Clinical Biomedical Sciences, Section of Biochemistry, University of Florence, Florence, Italy
| |
Collapse
|
20
|
Berg SZ, Berg J. Melanin: a unifying theory of disease as exemplified by Parkinson's, Alzheimer's, and Lewy body dementia. Front Immunol 2023; 14:1228530. [PMID: 37841274 PMCID: PMC10570809 DOI: 10.3389/fimmu.2023.1228530] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Melanin, a ubiquitous dark pigment, plays important roles in the immune system, including scavenging reactive oxygen species formed in response to ultraviolet radiation absorption, absorbing metals, thermal regulation, drug uptake, innate immune system functions, redox, and energy transduction. Many tissue types, including brain, heart, arteries, ovaries, and others, contain melanin. Almost all cells contain precursors to melanin. A growing number of diseases in which there is a loss of melanin and/or neuromelanin are increasingly thought to have infectious etiologies, for example, Alzheimer's disease (AD), Parkinson's disease (PD), Lewy Body Dementia (LBD), and vitiligo. AD, PD, LBD, and vitiligo have been linked with herpesvirus, which enters melanosomes and causes apoptosis, and with gut dysbiosis and inflammation. Herpesvirus is also linked with gut dysbiosis and inflammation. We theorize that under normal healthy states, melanin retains some of the energy it absorbs from electromagnetic radiation, which is then used to fuel cells, and energy from ATP is used to compliment that energy supply. We further theorize that loss of melanin reduces the energy supply of cells, which in the case of AD, PD, and LBD results in an inability to sustain immune system defenses and remove the plaques associated with the disease, which appear to be part of the immune system's attempt to eradicate the pathogens seen in these neurodegenerative diseases. In addition, in an attempt to explain why removing these plaques does not result in improvements in cognition and mood and why cognitions and moods in these individuals have ebbs and flows, we postulate that it is not the plaques that cause the cognitive symptoms but, rather, inflammation in the brain resulting from the immune system's response to pathogens. Our theory that energy retained in melanin fuels cells in an inverse relationship with ATP is supported by studies showing alterations in ATP production in relationship to melanin levels in melanomas, vitiligo, and healthy cells. Therefore, alteration of melanin levels may be at the core of many diseases. We propose regulating melanin levels may offer new avenues for treatment development.
Collapse
Affiliation(s)
- Stacie Z. Berg
- Department of Translational Biology, William Edwards LLC, Baltimore, MD, United States
| | - Jonathan Berg
- Department of Translational Biology, William Edwards LLC, Baltimore, MD, United States
| |
Collapse
|
21
|
Sesti F, Bortolami A, Kathera-Ibarra EF. Non-conducting functions of potassium channels in cancer and neurological disease. CURRENT TOPICS IN MEMBRANES 2023; 92:199-231. [PMID: 38007268 DOI: 10.1016/bs.ctm.2023.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2023]
Abstract
Cancer and neurodegenerative disease, albeit fundamental differences, share some common pathogenic mechanisms. Accordingly, both conditions are associated with aberrant cell proliferation and migration. Here, we review the causative role played by potassium (K+) channels, a fundamental class of proteins, in cancer and neurodegenerative disease. The concept that emerges from the review of the literature is that K+ channels can promote the development and progression of cancerous and neurodegenerative pathologies by dysregulating cell proliferation and migration. K+ channels appear to control these cellular functions in ways that not necessarily depend on their conducting properties and that involve the ability to directly or indirectly engage growth and survival signaling pathways. As cancer and neurodegenerative disease represent global health concerns, identifying commonalities may help understand the molecular basis for those devastating conditions and may facilitate the design of new drugs or the repurposing of existing drugs.
Collapse
Affiliation(s)
- Federico Sesti
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Hoes Ln. West, Piscataway, NJ, United States.
| | - Alessandro Bortolami
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Hoes Ln. West, Piscataway, NJ, United States
| | - Elena Forzisi Kathera-Ibarra
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, Rutgers University, Hoes Ln. West, Piscataway, NJ, United States
| |
Collapse
|
22
|
Lona-Durazo F, Reynolds RH, Scholz SW, Ryten M, Gagliano Taliun SA. Regional genetic correlations highlight relationships between neurodegenerative disease loci and the immune system. Commun Biol 2023; 6:729. [PMID: 37454237 PMCID: PMC10349864 DOI: 10.1038/s42003-023-05113-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 07/07/2023] [Indexed: 07/18/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer's and Parkinson's disease, are devastating complex diseases resulting in physical and psychological burdens on patients and their families. There have been important efforts to understand their genetic basis leading to the identification of disease risk-associated loci involved in several molecular mechanisms, including immune-related pathways. Regional, in contrast to genome-wide, genetic correlations between pairs of immune and neurodegenerative traits have not been comprehensively explored, but could uncover additional immune-mediated risk-associated loci. Here, we systematically assess the role of the immune system in five neurodegenerative diseases by estimating regional genetic correlations between these diseases and immune-cell-derived single-cell expression quantitative trait loci (sc-eQTLs). We also investigate correlations between diseases and protein levels. We observe significant (FDR < 0.01) correlations between sc-eQTLs and neurodegenerative diseases across 151 unique genes, spanning both the innate and adaptive immune systems, across most diseases tested. With Parkinson's, for instance, RAB7L1 in CD4+ naïve T cells is positively correlated and KANSL1-AS1 is negatively correlated across all adaptive immune cell types. Follow-up colocalization highlight candidate causal risk genes. The outcomes of this study will improve our understanding of the immune component of neurodegeneration, which can warrant repurposing of existing immunotherapies to slow disease progression.
Collapse
Affiliation(s)
- Frida Lona-Durazo
- Montréal Heart Institute, Montréal, QC, Canada
- Université de Montréal, Montréal, QC, Canada
| | - Regina H Reynolds
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Unit, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Mina Ryten
- Genetics and Genomic Medicine, Great Ormond Street Institute of Child Health, University College London, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- NIHR Great Ormond Street Hospital Biomedical Research Centre, University College London, London, UK
| | - Sarah A Gagliano Taliun
- Montréal Heart Institute, Montréal, QC, Canada.
- Department of Medicine & Department of Neurosciences, Université de Montréal, Montréal, QC, Canada.
| |
Collapse
|
23
|
Brembati V, Faustini G, Longhena F, Bellucci A. Alpha synuclein post translational modifications: potential targets for Parkinson's disease therapy? Front Mol Neurosci 2023; 16:1197853. [PMID: 37305556 PMCID: PMC10248004 DOI: 10.3389/fnmol.2023.1197853] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 04/27/2023] [Indexed: 06/13/2023] Open
Abstract
Parkinson's disease (PD) is the most common neurodegenerative disorder with motor symptoms. The neuropathological alterations characterizing the brain of patients with PD include the loss of dopaminergic neurons of the nigrostriatal system and the presence of Lewy bodies (LB), intraneuronal inclusions that are mainly composed of alpha-synuclein (α-Syn) fibrils. The accumulation of α-Syn in insoluble aggregates is a main neuropathological feature in PD and in other neurodegenerative diseases, including LB dementia (LBD) and multiple system atrophy (MSA), which are therefore defined as synucleinopathies. Compelling evidence supports that α-Syn post translational modifications (PTMs) such as phosphorylation, nitration, acetylation, O-GlcNAcylation, glycation, SUMOylation, ubiquitination and C-terminal cleavage, play important roles in the modulation α-Syn aggregation, solubility, turnover and membrane binding. In particular, PTMs can impact on α-Syn conformational state, thus supporting that their modulation can in turn affect α-Syn aggregation and its ability to seed further soluble α-Syn fibrillation. This review focuses on the importance of α-Syn PTMs in PD pathophysiology but also aims at highlighting their general relevance as possible biomarkers and, more importantly, as innovative therapeutic targets for synucleinopathies. In addition, we call attention to the multiple challenges that we still need to face to enable the development of novel therapeutic approaches modulating α-Syn PTMs.
Collapse
Affiliation(s)
| | | | | | - Arianna Bellucci
- Division of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| |
Collapse
|
24
|
Stevenson M, Varghese R, Hebron ML, Liu X, Ratliff N, Smith A, Turner RS, Moussa C. Inhibition of discoidin domain receptor (DDR)-1 with nilotinib alters CSF miRNAs and is associated with reduced inflammation and vascular fibrosis in Alzheimer's disease. J Neuroinflammation 2023; 20:116. [PMID: 37194065 PMCID: PMC10186647 DOI: 10.1186/s12974-023-02802-0] [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: 03/17/2023] [Accepted: 05/10/2023] [Indexed: 05/18/2023] Open
Abstract
Discoidin Domain Receptor (DDR)-1 is activated by collagen. Nilotinib is a tyrosine kinase inhibitor that is FDA-approved for leukemia and potently inhibits DDR-1. Individuals diagnosed with mild-moderate Alzheimer's disease (AD) treated with nilotinib (versus placebo) for 12 months showed reduction of amyloid plaque and cerebrospinal fluid (CSF) amyloid, and attenuation of hippocampal volume loss. However, the mechanisms are unclear. Here, we explored unbiased next generation whole genome miRNA sequencing from AD patients CSF and miRNAs were matched with their corresponding mRNAs using gene ontology. Changes in CSF miRNAs were confirmed via measurement of CSF DDR1 activity and plasma levels of AD biomarkers. Approximately 1050 miRNAs are detected in the CSF but only 17 miRNAs are specifically altered between baseline and 12-month treatment with nilotinib versus placebo. Treatment with nilotinib significantly reduces collagen and DDR1 gene expression (upregulated in AD brain), in association with inhibition of CSF DDR1. Pro-inflammatory cytokines, including interleukins and chemokines are reduced along with caspase-3 gene expression. Specific genes that indicate vascular fibrosis, e.g., collagen, Transforming Growth Factors (TGFs) and Tissue Inhibitors of Metalloproteases (TIMPs) are altered by DDR1 inhibition with nilotinib. Specific changes in vesicular transport, including the neurotransmitters dopamine and acetylcholine, and autophagy genes, including ATGs, indicate facilitation of autophagic flux and cellular trafficking. Inhibition of DDR1 with nilotinib may be a safe and effective adjunct treatment strategy involving an oral drug that enters the CNS and adequately engages its target. DDR1 inhibition with nilotinib exhibits multi-modal effects not only on amyloid and tau clearance but also on anti-inflammatory markers that may reduce cerebrovascular fibrosis.
Collapse
Affiliation(s)
- Max Stevenson
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Rency Varghese
- Genomics and Epigenomics Shared Resource, Department of Oncology, Georgetown University Medical Center, Building D, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Michaeline L Hebron
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Xiaoguang Liu
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Nick Ratliff
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Amelia Smith
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - R Scott Turner
- Memory Disorders Program, Department of Neurology, Georgetown University Medical Center, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Charbel Moussa
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Building D, Room 265, 4000 Reservoir Rd, NW, Washington, DC, 20057, USA.
| |
Collapse
|
25
|
Nakmode DD, Day CM, Song Y, Garg S. The Management of Parkinson's Disease: An Overview of the Current Advancements in Drug Delivery Systems. Pharmaceutics 2023; 15:pharmaceutics15051503. [PMID: 37242745 DOI: 10.3390/pharmaceutics15051503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/31/2023] [Accepted: 05/10/2023] [Indexed: 05/28/2023] Open
Abstract
Parkinson's disease (PD) has significantly affected a large proportion of the elderly population worldwide. According to the World Health Organization, approximately 8.5 million people worldwide are living with PD. In the United States, an estimated one million people are living with PD, with approximately 60,000 new cases diagnosed every year. Conventional therapies available for Parkinson's disease are associated with limitations such as the wearing-off effect, on-off period, episodes of motor freezing, and dyskinesia. In this review, a comprehensive overview of the latest advances in DDSs used to reduce the limitations of current therapies will be presented, and both their promising features and drawbacks will be discussed. We are also particularly interested in the technical properties, mechanism, and release patterns of incorporated drugs, as well as nanoscale delivery strategies to overcome the blood-brain barrier.
Collapse
Affiliation(s)
- Deepa D Nakmode
- Centre for Pharmaceutical Innovation, University of South Australia, North Terrace, Adelaide, SA 5000, Australia
| | - Candace M Day
- Centre for Pharmaceutical Innovation, University of South Australia, North Terrace, Adelaide, SA 5000, Australia
| | - Yunmei Song
- Centre for Pharmaceutical Innovation, University of South Australia, North Terrace, Adelaide, SA 5000, Australia
| | - Sanjay Garg
- Centre for Pharmaceutical Innovation, University of South Australia, North Terrace, Adelaide, SA 5000, Australia
| |
Collapse
|
26
|
Walsh RR, Damle NK, Mandhane S, Piccoli SP, Talluri RS, Love D, Yao SL, Ramanathan V, Hurko O. Plasma and cerebrospinal fluid pharmacokinetics of vodobatinib, a neuroprotective c-Abl tyrosine kinase inhibitor for the treatment of Parkinson's disease. Parkinsonism Relat Disord 2023; 108:105281. [PMID: 36717298 DOI: 10.1016/j.parkreldis.2023.105281] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/22/2022] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
BACKGROUND Preclinical evidence suggests that c-Abl is critical in the pathogenesis of Parkinson's Disease (PD). Vodobatinib (K0706) is a potent, specific Abl kinase inhibitor currently being developed for the treatment of PD. In previously reported studies, nilotinib, a multikinase c-Abl inhibitor, did not show clinical activity as evidenced by no improvement of symptoms or the rate of decline after one to six months of treatment at the maximum permissible dose, presumably because of insufficient CNS penetration. Here we report clinical PK and safety data for vodobatinib. OBJECTIVES To determine safety, plasma PK, and CSF penetration of vodobatinib in healthy volunteers and PD subjects following oral administration, and compare CSF levels to in vitro concentrations required for c-Abl inhibition relative to data reported for nilotinib. METHODS Inhibition of c-Abl kinase activity and c-Abl binding affinity were first assessed in vitro. Healthy human volunteers and PD patients received various oral doses of vodobatinib once-daily for seven and fourteen days respectively, to assess safety, and plasma and CSF PK. RESULTS In in vitro assays, vodobatinib was more potent (kinase IC50 = 0.9 nM) than nilotinib (kinase IC50 = 15-45 nM). Administration of vodobatinib 48, 192 and 384 mg to healthy subjects for 7 days yielded mean Cmax, CSF values of 1.8, 11.6, and 12.2 nM respectively, with the two highest doses exceeding the IC50 over the entire dosing interval. Cavg, CSF values were 6-8 times greater than the IC50. Comparable CSF levels were observed in PD patients. All doses were well tolerated in both cohorts. CONCLUSION Based on achieved CSF concentrations, the potential for c-Abl inhibition in the brain is substantially higher with vodobatinib than with nilotinib. The CSF PK profile of vodobatinib is suitable for determining if c-Abl inhibition will be neuroprotective in PD patients.
Collapse
Affiliation(s)
- Ryan R Walsh
- Sun Pharma Advanced Research Company Ltd, Cranbury, NJ, USA.
| | - Nitin K Damle
- Sun Pharma Advanced Research Company Ltd, Mumbai, India
| | | | | | | | - Damon Love
- Sun Pharma Advanced Research Company Ltd, Cranbury, NJ, USA
| | - Siu-Long Yao
- Sun Pharma Advanced Research Company Ltd, Cranbury, NJ, USA
| | | | - Orest Hurko
- Sun Pharma Advanced Research Company Ltd, Cranbury, NJ, USA
| |
Collapse
|
27
|
Zagórska A, Czopek A, Fryc M, Jaromin A, Boyd BJ. Drug Discovery and Development Targeting Dementia. Pharmaceuticals (Basel) 2023; 16:151. [PMID: 37259302 PMCID: PMC9965722 DOI: 10.3390/ph16020151] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/16/2023] [Accepted: 01/16/2023] [Indexed: 08/04/2023] Open
Abstract
Dementia, most often associated with neurodegenerative diseases, affects millions of people worldwide, predominantly the elderly. Unfortunately, no treatment is still available. Therefore, there is an urgent need to address this situation. This review presents the state of the art of drug discovery and developments in targeting dementia. Several approaches are discussed, such as drug repurposing, the use of small molecules, and phosphodiesterase inhibitors. Furthermore, the review also provides insights into clinical trials of these molecules. Emphasis has been placed on small molecules and multi-target-directed ligands, as well as disease-modifying therapies. Finally, attention is drawn to the possibilities of applications of nanotechnology in managing dementia.
Collapse
Affiliation(s)
- Agnieszka Zagórska
- Department of Medicinal Chemistry, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Anna Czopek
- Department of Medicinal Chemistry, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Monika Fryc
- Department of Medicinal Chemistry, Jagiellonian University Medical College, Medyczna 9, 30-688 Kraków, Poland
| | - Anna Jaromin
- Department of Lipids and Liposomes, Faculty of Biotechnology, University of Wrocław, Joliot-Curie 14a, 50-383 Wrocław, Poland
| | - Ben J. Boyd
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
- Drug Delivery Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University (Parkville Campus), 381 Royal Parade, Parkville, VIC 3052, Australia
| |
Collapse
|
28
|
Therapeutic Monitoring of Orally Administered, Small-Molecule Anticancer Medications with Tumor-Specific Cellular Protein Targets in Peripheral Fluid Spaces-A Review. Pharmaceutics 2023; 15:pharmaceutics15010239. [PMID: 36678867 PMCID: PMC9864625 DOI: 10.3390/pharmaceutics15010239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
Orally administered, small-molecule anticancer drugs with tumor-specific cellular protein targets (OACD) have revolutionized oncological pharmacotherapy. Nevertheless, the differences in exposure to these drugs in the systemic circulation and extravascular fluid compartments have led to several cases of therapeutic failure, in addition to posing unknown risks of toxicity. The therapeutic drug monitoring (TDM) of OACDs in therapeutically relevant peripheral fluid compartments is therefore essential. In this work, the available knowledge regarding exposure to OACD concentrations in these fluid spaces is summarized. A review of the literature was conducted by searching Embase, PubMed, and Web of Science for clinical research articles and case reports published between 10 May 2001 and 31 August 2022. Results show that, to date, penetration into cerebrospinal fluid has been studied especially intensively, in addition to breast milk, leukocytes, peripheral blood mononuclear cells, peritoneal fluid, pleural fluid, saliva and semen. The typical clinical indications of peripheral fluid TDM of OACDs were (1) primary malignancy, (2) secondary malignancy, (3) mental disorder, and (4) the assessment of toxicity. Liquid chromatography-tandem mass spectrometry was most commonly applied for analysis. The TDM of OACDs in therapeutically relevant peripheral fluid spaces is often indispensable for efficient and safe treatments.
Collapse
|
29
|
Abstract
Parkinson's disease (PD) is a neurodegenerative disease manifesting with motor and non-motor symptoms. Current treatment mainly relies on medication as a symptomatic therapy modulating neurotransmitters. Dopamine replacement therapy has been established, and levodopa is the gold standard for treatment of PD. However, the emergence of motor complications, such as a wearing-off phenomenon, is a clinical problem. Both primary symptoms and motor complications have been targets for the development of treatments for PD. Recent progression in the management of motor complications is supported by newly developed agents and advances in device and formulation technology to deliver drugs continuously. Elucidation of the pathophysiology of PD and the development of disease-modifying therapy that affects the underlying fundamental pathophysiology of the disease are also progressing. In this review, we introduce current knowledge on developments concerning medications for patients with PD.
Collapse
Affiliation(s)
- Hidetomo Murakami
- Department of Neurology, the Jikei University School of Medicine, Japan
| | | | - Tadashi Umehara
- Department of Neurology, the Jikei University School of Medicine, Japan
| | - Shusaku Omoto
- Department of Neurology, the Jikei University Katsushika Medical Center, Japan
| | - Yasuyuki Iguchi
- Department of Neurology, the Jikei University School of Medicine, Japan
| |
Collapse
|
30
|
Ozoran H, Srinivasan R. Astrocytes and Alpha-Synuclein: Friend or Foe? JOURNAL OF PARKINSON'S DISEASE 2023; 13:1289-1301. [PMID: 38007674 PMCID: PMC10741342 DOI: 10.3233/jpd-230284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/21/2023] [Indexed: 11/27/2023]
Abstract
Despite its devastating disease burden and alarming prevalence, the etiology of Parkinson's disease (PD) remains to be completely elucidated. PD is characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta and this correlates with the accumulation of misfolded α-synuclein. While the aggregation of α-synuclein in the form of Lewy bodies or Lewy neurites is a well-established intraneuronal hallmark of the disease process, our understanding of the glial contribution to aberrant α-synuclein proteostasis is lacking. In this regard, restoring astrocyte function during early PD could offer a promising therapeutic avenue and understanding the involvement of astrocytes in handling/mishandling of α-synuclein is of particular interest. Here, we explore the growing body of scientific literature implicating aberrant astrocytic α-synuclein proteostasis with the seemingly inexorable pathological sequelae typifying PD. We also provide a perspective on how heterogeneity in the morphological relationship between astrocytes and neurons will need to be considered in the context of PD pathogenesis.
Collapse
Affiliation(s)
- Hakan Ozoran
- Clinical Medical School, University of Oxford, Oxford, UK
- Green Templeton College, University of Oxford, Oxford, UK
| | - Rahul Srinivasan
- Department of Neuroscience & Experimental Therapeutics, Texas A&M University School of Medicine, Bryan, TX, USA
- Texas A&M Institute for Neuroscience (TAMIN), College Station, TX, USA
| |
Collapse
|
31
|
Jayaswamy PK, Vijaykrishnaraj M, Patil P, Alexander LM, Kellarai A, Shetty P. Implicative role of epidermal growth factor receptor and its associated signaling partners in the pathogenesis of Alzheimer's disease. Ageing Res Rev 2023; 83:101791. [PMID: 36403890 DOI: 10.1016/j.arr.2022.101791] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 11/12/2022] [Accepted: 11/13/2022] [Indexed: 11/18/2022]
Abstract
Epidermal growth factor receptor (EGFR) plays a pivotal role in early brain development, although its expression pattern declines in accordance with the maturation of the active nervous system. However, recurrence of EGFR expression in brain cells takes place during neural functioning decline and brain atrophy in order to maintain the homeostatic neuronal pool. As a consequence, neurotoxic lesions such as amyloid beta fragment (Aβ1-42) formed during the alternative splicing of amyloid precursor protein in Alzheimer's disease (AD) elevate the expression of EGFR. This inappropriate peptide deposition on EGFR results in the sustained phosphorylation of the downstream signaling axis, leading to extensive Aβ1-42 production and tau phosphorylation as subsequent pathogenesis. Recent reports convey that the pathophysiology of AD is correlated with EGFR and its associated membrane receptor complex molecules. One such family of molecules is the annexin superfamily, which has synergistic relationships with EGFR and is known for membrane-bound signaling that contributes to a variety of inflammatory responses. Besides, Galectin-3, tissue-type activated plasminogen activator, and many more, which lineate the secretion of pro-inflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-18) result in severe neuronal loss. Altogether, we emphasized the perspectives of cellular senescence up-regulated by EGFR and its associated membrane receptor molecules in the pathogenesis of AD as a target for a therapeutical alternative to intervene in AD.
Collapse
Affiliation(s)
- Pavan K Jayaswamy
- Central Research Laboratory, KS. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore 575018, Karnataka, India
| | - M Vijaykrishnaraj
- Central Research Laboratory, KS. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore 575018, Karnataka, India
| | - Prakash Patil
- Central Research Laboratory, KS. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore 575018, Karnataka, India
| | - Lobo Manuel Alexander
- Department of Neurology, KS. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore 575018, Karnataka, India
| | - Adithi Kellarai
- Department of General Medicine, KS. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore 575018, Karnataka, India
| | - Praveenkumar Shetty
- Central Research Laboratory, KS. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore 575018, Karnataka, India; Department of Biochemistry, K.S. Hegde Medical Academy, Nitte (Deemed to be University), Deralakatte, Mangalore 575018, Karnataka, India.
| |
Collapse
|
32
|
From protein biomarkers to proteomics in dementia with Lewy Bodies. Ageing Res Rev 2023; 83:101771. [PMID: 36328346 DOI: 10.1016/j.arr.2022.101771] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 09/15/2022] [Accepted: 10/27/2022] [Indexed: 11/06/2022]
Abstract
Dementia with Lewy Bodies (DLB) is the second most common neurodegenerative dementia. Despite considerable research progress, there remain gaps in our understanding of the pathophysiology and there is no disease-modifying treatment. Proteomics is a powerful tool to elucidate complex biological pathways across heterogenous conditions. This review summarizes the widely used proteomic methods and presents evidence for protein dysregulation in the brain and peripheral tissues in DLB. Proteomics of post-mortem brain tissue shows that DLB shares common features with other dementias, such as synaptic dysfunction, but retains a unique protein signature. Promising diagnostic biomarkers are being identified in cerebrospinal fluid (CSF), blood, and peripheral tissues, such as serum Heart-type fatty acid binding protein. Research is needed to track these changes from the prodromal stage to established dementia, with standardized workflows to ensure replicability. Identifying novel protein targets in causative biological pathways could lead to the development of new targeted therapeutics or the stratification of participants for clinical trials.
Collapse
|
33
|
Mansour HM, El-Khatib AS. Repositioning of receptor tyrosine kinase inhibitors. RECEPTOR TYROSINE KINASES IN NEURODEGENERATIVE AND PSYCHIATRIC DISORDERS 2023:353-401. [DOI: 10.1016/b978-0-443-18677-6.00010-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
34
|
Alteration of Autophagy and Glial Activity in Nilotinib-Treated Huntington's Disease Patients. Metabolites 2022; 12:metabo12121225. [PMID: 36557263 PMCID: PMC9781133 DOI: 10.3390/metabo12121225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/03/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Nilotinib is a tyrosine kinase inhibitor that is safe and tolerated in neurodegeneration, it achieves CSF concentration that is adequate to inhibit discoidin domain receptor (DDR)-1. Nilotinib significantly affects dopamine metabolites, including Homovanillic acid (HVA), resulting in an increase in brain dopamine. HD is a hereditary disease caused by mutations in the Huntingtin's (HTT) gene and characterized by neurodegeneration and motor and behavioral symptoms that are associated with activation of dopamine receptors. We explored the effects of a low dose of nilotinib (150 mg) on behavioral changes and motor symptoms in manifest HD patients and examined the effects of nilotinib on several brain mechanisms, including dopamine transmission and gene expression via cerebrospinal fluid (CSF) miRNA sequencing. Nilotinib, 150 mg, did not result in any behavioral changes, although it significantly attenuated HVA levels, suggesting reduction of dopamine catabolism. There was no significant change in HTT, phosphorylated neuro-filament and inflammatory markers in the CSF and plasma via immunoassays. Whole miRNA genome sequencing of the CSF revealed significant longitudinal changes in miRNAs that control specific genes associated with autophagy, inflammation, microglial activity and basal ganglia neurotransmitters, including dopamine and serotonin.
Collapse
|
35
|
O'Brien JT, Chouliaras L, Sultana J, Taylor JP, Ballard C. RENEWAL: REpurposing study to find NEW compounds with Activity for Lewy body dementia-an international Delphi consensus. Alzheimers Res Ther 2022; 14:169. [PMID: 36369100 PMCID: PMC9650797 DOI: 10.1186/s13195-022-01103-7] [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] [Received: 07/05/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022]
Abstract
Drug repositioning and repurposing has proved useful in identifying new treatments for many diseases, which can then rapidly be brought into clinical practice. Currently, there are few effective pharmacological treatments for Lewy body dementia (which includes both dementia with Lewy bodies and Parkinson's disease dementia) apart from cholinesterase inhibitors. We reviewed several promising compounds that might potentially be disease-modifying agents for Lewy body dementia and then undertook an International Delphi consensus study to prioritise compounds. We identified ambroxol as the top ranked agent for repurposing and identified a further six agents from the classes of tyrosine kinase inhibitors, GLP-1 receptor agonists, and angiotensin receptor blockers that were rated by the majority of our expert panel as justifying a clinical trial. It would now be timely to take forward all these compounds to Phase II or III clinical trials in Lewy body dementia.
Collapse
Affiliation(s)
- John T O'Brien
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK.
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK.
| | - Leonidas Chouliaras
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Janet Sultana
- College of Medicine and Health, University of Exeter, Exeter, UK
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle, UK
| | - Clive Ballard
- College of Medicine and Health, University of Exeter, Exeter, UK
| |
Collapse
|
36
|
Erekat NS. Autophagy and Its Association with Genetic Mutations in Parkinson Disease. Med Sci Monit 2022; 28:e938519. [PMID: 36366737 PMCID: PMC9664771 DOI: 10.12659/msm.938519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 10/19/2022] [Indexed: 08/07/2023] Open
Abstract
Parkinson disease is the second most common neurodegenerative disorder, affecting 0.1-0.2% of the general population. It is a progressive debilitating disorder caused by degeneration of dopaminergic neurons in the substantia nigra pars compacta. It is characterized by motor and non-motor symptoms. Parkinson disease can be caused by mutations in genes that encode proteins involved in the autophagic process, resulting in impaired autophagy. Indeed, autophagy has been implicated in the pathogenesis of Parkinson disease, particularly because its impairment causes the buildup of proteins. Thus, this review aims to provide an overview of Parkinson disease-related genetic mutations and their association with autophagy impairment in Parkinson disease, which can be helpful in improving the understanding of the pathogenesis of Parkinson disease, illustrating the potential therapeutic implications of agents that can enhance autophagy in Parkinson disease. Additionally, we will highlight the essential need for the development of highly sensitive and specific assays for gene-based diagnostic biomarkers. Finally, we will provide an overview on the potential gene-based therapeutic approaches for Parkinson disease, which have been most advanced and are associated with the most common targets being alpha-synuclein (SNCA), leucine-rich repeat kinase-2 (LRRK2), and glucocerebrosidase (GBA).
Collapse
|
37
|
Molecular and Cellular Interactions in Pathogenesis of Sporadic Parkinson Disease. Int J Mol Sci 2022; 23:ijms232113043. [PMID: 36361826 PMCID: PMC9657547 DOI: 10.3390/ijms232113043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 10/16/2022] [Accepted: 10/25/2022] [Indexed: 11/23/2022] Open
Abstract
An increasing number of the population all around the world suffer from age-associated neurodegenerative diseases including Parkinson’s disease (PD). This disorder presents different signs of genetic, epigenetic and environmental origin, and molecular, cellular and intracellular dysfunction. At the molecular level, α-synuclein (αSyn) was identified as the principal molecule constituting the Lewy bodies (LB). The gut microbiota participates in the pathogenesis of PD and may contribute to the loss of dopaminergic neurons through mitochondrial dysfunction. The most important pathogenetic link is an imbalance of Ca2+ ions, which is associated with redox imbalance in the cells and increased generation of reactive oxygen species (ROS). In this review, genetic, epigenetic and environmental factors that cause these disorders and their cause-and-effect relationships are considered. As a constituent of environmental factors, the example of organophosphates (OPs) is also reviewed. The role of endothelial damage in the pathogenesis of PD is discussed, and a ‘triple hit hypothesis’ is proposed as a modification of Braak’s dual hit one. In the absence of effective therapies for neurodegenerative diseases, more and more evidence is emerging about the positive impact of nutritional structure and healthy lifestyle on the state of blood vessels and the risk of developing these diseases.
Collapse
|
38
|
Latif K, Ullah A, Shkodina AD, Boiko DI, Rafique Z, Alghamdi BS, Alfaleh MA, Ashraf GM. Drug reprofiling history and potential therapies against Parkinson's disease. Front Pharmacol 2022; 13:1028356. [PMID: 36386233 PMCID: PMC9643740 DOI: 10.3389/fphar.2022.1028356] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/03/2022] [Indexed: 12/02/2022] Open
Abstract
Given the high whittling down rates, high costs, and moderate pace of new medication, revelation, and improvement, repurposing "old" drugs to treat typical and uncommon illnesses is progressively becoming an appealing proposition. Drug repurposing is the way toward utilizing existing medications in treating diseases other than the purposes they were initially designed for. Faced with scientific and economic challenges, the prospect of discovering new medication indications is enticing to the pharmaceutical sector. Medication repurposing can be used at various stages of drug development, although it has shown to be most promising when the drug has previously been tested for safety. We describe strategies of drug repurposing for Parkinson's disease, which is a neurodegenerative condition that primarily affects dopaminergic neurons in the substantia nigra. We also discuss the obstacles faced by the repurposing community and suggest new approaches to solve these challenges so that medicine repurposing can reach its full potential.
Collapse
Affiliation(s)
- Komal Latif
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Aman Ullah
- Shifa College of Pharmaceutical Sciences, Shifa Tameer-e-Millet University, Islamabad, Pakistan
| | - Anastasiia D. Shkodina
- Department of Neurological Diseases, Poltava State Medical University, Poltava, Ukraine
- Municipal Enterprise “1 City Clinical Hospital of Poltava City Council”, Poltava, Ukraine
| | - Dmytro I. Boiko
- Department of Psychiatry, Narcology and Medical Psychology, Poltava State Medical University, Poltava, Ukraine
| | - Zakia Rafique
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Islamabad, Pakistan
| | - Badrah S. Alghamdi
- Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohamed A. Alfaleh
- Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Division of Vaccines and Immunotherapy, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ghulam Md. Ashraf
- Department of Medical Laboratory Sciences, College of Health Sciences, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
39
|
Paik J. Targeting toxic forms of α-synuclein with immunotherapy could alter the progression of Parkinson’s disease. DRUGS & THERAPY PERSPECTIVES 2022. [DOI: 10.1007/s40267-022-00950-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
40
|
Kakoti BB, Bezbaruah R, Ahmed N. Therapeutic drug repositioning with special emphasis on neurodegenerative diseases: Threats and issues. Front Pharmacol 2022; 13:1007315. [PMID: 36263141 PMCID: PMC9574100 DOI: 10.3389/fphar.2022.1007315] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Accepted: 09/12/2022] [Indexed: 11/21/2022] Open
Abstract
Drug repositioning or repurposing is the process of discovering leading-edge indications for authorized or declined/abandoned molecules for use in different diseases. This approach revitalizes the traditional drug discovery method by revealing new therapeutic applications for existing drugs. There are numerous studies available that highlight the triumph of several drugs as repurposed therapeutics. For example, sildenafil to aspirin, thalidomide to adalimumab, and so on. Millions of people worldwide are affected by neurodegenerative diseases. According to a 2021 report, the Alzheimer's disease Association estimates that 6.2 million Americans are detected with Alzheimer's disease. By 2030, approximately 1.2 million people in the United States possibly acquire Parkinson's disease. Drugs that act on a single molecular target benefit people suffering from neurodegenerative diseases. Current pharmacological approaches, on the other hand, are constrained in their capacity to unquestionably alter the course of the disease and provide patients with inadequate and momentary benefits. Drug repositioning-based approaches appear to be very pertinent, expense- and time-reducing strategies for the enhancement of medicinal opportunities for such diseases in the current era. Kinase inhibitors, for example, which were developed for various oncology indications, demonstrated significant neuroprotective effects in neurodegenerative diseases. This review expounds on the classical and recent examples of drug repositioning at various stages of drug development, with a special focus on neurodegenerative disorders and the aspects of threats and issues viz. the regulatory, scientific, and economic aspects.
Collapse
Affiliation(s)
- Bibhuti Bhusan Kakoti
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, India
| | | | | |
Collapse
|
41
|
Salamon A, Zádori D, Szpisjak L, Klivényi P, Vécsei L. The genetic background of Parkinson's disease and novel therapeutic targets. Expert Opin Ther Targets 2022; 26:827-836. [PMID: 36524726 DOI: 10.1080/14728222.2022.2153037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Parkinson's disease (PD) is the second most common neurodegenerative disease worldwide. The median age of disease onset is around 60 years. From a genetic point of view, PD is basically considered a sporadic, idiopathic disease, however, hereditary components can be detected in 5-10% of patients. Expanding data are available regarding the targeted molecular therapy of the disease. AREAS COVERED The aim of this current review article is to provide brief clinical and molecular insight into three important genetic forms (LRRK2, SNCA, GBA) of hereditary PD subtypes and to present the human clinical trials in relation to these forms of the disease. EXPERT OPINION These small hereditary subgroups are crucially important in drug development, because the general trend is that clinical trials that treat PD patients as a large group, without any separation, do not meet expectations. As a result, no long term conclusions can currently be drawn regarding the effectiveness of the molecules tested in these phase 1 and 2 studies. Further precise studies are needed in the near future.
Collapse
Affiliation(s)
- András Salamon
- Interdisciplinary Excellence Centre, Department of Neurology, Albert Szent-Györgyi Faculty of Medicine School, University of Szeged, Szeged, Hungary
| | - Dénes Zádori
- Interdisciplinary Excellence Centre, Department of Neurology, Albert Szent-Györgyi Faculty of Medicine School, University of Szeged, Szeged, Hungary
| | - László Szpisjak
- Interdisciplinary Excellence Centre, Department of Neurology, Albert Szent-Györgyi Faculty of Medicine School, University of Szeged, Szeged, Hungary
| | - Péter Klivényi
- Interdisciplinary Excellence Centre, Department of Neurology, Albert Szent-Györgyi Faculty of Medicine School, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Interdisciplinary Excellence Centre, Department of Neurology, Albert Szent-Györgyi Faculty of Medicine School, University of Szeged, Szeged, Hungary.,Department of Neurology, ELKH-SZTE Neuroscience Research Group, Eötvös Loránd Research Network, University of Szeged, Szeged, Hungary
| |
Collapse
|
42
|
Xie X, Yuan P, Kou L, Chen X, Li J, Li Y. Nilotinib in Parkinson's disease: A systematic review and meta-analysis. Front Aging Neurosci 2022; 14:996217. [PMID: 36248007 PMCID: PMC9558096 DOI: 10.3389/fnagi.2022.996217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Accepted: 09/01/2022] [Indexed: 11/18/2022] Open
Abstract
Background Nilotinib, which inhibits cellular Abelson tyrosine kinase, may be an effective treatment for patients with Parkinson's disease (PD). The purpose of this study is to evaluate the outcomes of different doses of nilotinib in patients with PD. Methods We searched PubMed, Embase, Web of Science, and Cochrane Central Register of Controlled Clinical Trials from inception to 7 March 2022 to identify all randomized controlled trials (RCTs) of nilotinib reporting outcomes of interest in patients with PD. Outcomes included tolerability, efficacy, safety, and CSF biomarker levels. Review manager 5.4 software was used to analyze all data. Results Three RCTs with a total of 163 patients were included. No significant difference was found between 150 mg nilotinib or 300 mg nilotinib and placebo in terms of tolerability, adverse events, or HVA levels. 300 mg nilotinib showed significantly higher Movement Disorder Society Unified Parkinson's Disease Rating Scale III (MDS-UPDRS III) scores [SMD = 0.52, 95%CI = (0.12, 0.92), P = 0.01] and 3,4-dihydroxyphenylacetic acid (DOPAC) levels [SMD = 0.52, 95%CI = (0.12, 0.92), P = 0.01], and lower α-synuclein levels [SMD = −2.16, 95%CI = (−3.38, −1.84), P < 0.00001] compared with placebo. And compared with 150 mg nilotinib, 300 mg nilotinib showed significantly lower α-synuclein levels [SMD = −1.16, 95%CI = (−1.70, −0.61), P < 0.0001]. Conclusions Although our study demonstrated favorable tolerability and safety of different doses of nilotinib, and improvement in part of CSF biomarker levels of 300 mg nilotinib, the poor efficacy on motor outcomes indicated that nilotinib had no advantages in the clinic.
Collapse
Affiliation(s)
- Xiaolu Xie
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Ping Yuan
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Liqiu Kou
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiu Chen
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jun Li
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- *Correspondence: Jun Li
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Yaling Li
| |
Collapse
|
43
|
Therapeutic Strategies in Huntington’s Disease: From Genetic Defect to Gene Therapy. Biomedicines 2022; 10:biomedicines10081895. [PMID: 36009443 PMCID: PMC9405755 DOI: 10.3390/biomedicines10081895] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/29/2022] [Accepted: 08/03/2022] [Indexed: 12/14/2022] Open
Abstract
Despite the identification of an expanded CAG repeat on exon 1 of the huntingtin gene located on chromosome 1 as the genetic defect causing Huntington’s disease almost 30 years ago, currently approved therapies provide only limited symptomatic relief and do not influence the age of onset or disease progression rate. Research has identified various intricate pathogenic cascades which lead to neuronal degeneration, but therapies interfering with these mechanisms have been marked by many failures and remain to be validated. Exciting new opportunities are opened by the emerging techniques which target the mutant protein DNA and RNA, allowing for “gene editing”. Although some issues relating to “off-target” effects or immune-mediated side effects need to be solved, these strategies, combined with stem cell therapies and more traditional approaches targeting specific pathogenic cascades, such as excitotoxicity and bioavailability of neurotrophic factors, could lead to significant improvement of the outcomes of treated Huntington’s disease patients.
Collapse
|
44
|
Opportunities and challenges of alpha-synuclein as a potential biomarker for Parkinson's disease and other synucleinopathies. NPJ Parkinsons Dis 2022; 8:93. [PMID: 35869066 PMCID: PMC9307631 DOI: 10.1038/s41531-022-00357-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/24/2022] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD), the second most common progressive neurodegenerative disease, develops and progresses for 10–15 years before the clinical diagnostic symptoms of the disease are manifested. Furthermore, several aspects of PD pathology overlap with other neurodegenerative diseases (NDDs) linked to alpha-synuclein (aSyn) aggregation, also called synucleinopathies. Therefore, there is an urgent need to discover and validate early diagnostic and prognostic markers that reflect disease pathophysiology, progression, severity, and potential differences in disease mechanisms between PD and other NDDs. The close association between aSyn and the development of pathology in synucleinopathies, along with the identification of aSyn species in biological fluids, has led to increasing interest in aSyn species as potential biomarkers for early diagnosis of PD and differentiate it from other synucleinopathies. In this review, we (1) provide an overview of the progress toward mapping the distribution of aSyn species in the brain, peripheral tissues, and biological fluids; (2) present comparative and critical analysis of previous studies that measured total aSyn as well as other species such as modified and aggregated forms of aSyn in different biological fluids; and (3) highlight conceptual and technical gaps and challenges that could hinder the development and validation of reliable aSyn biomarkers; and (4) outline a series of recommendations to address these challenges. Finally, we propose a combined biomarker approach based on integrating biochemical, aggregation and structure features of aSyn, in addition to other biomarkers of neurodegeneration. We believe that capturing the diversity of aSyn species is essential to develop robust assays and diagnostics for early detection, patient stratification, monitoring of disease progression, and differentiation between synucleinopathies. This could transform clinical trial design and implementation, accelerate the development of new therapies, and improve clinical decisions and treatment strategies.
Collapse
|
45
|
Kim J, Lee HJ, Park JH, Cha BY, Hoe HS. Nilotinib modulates LPS-induced cognitive impairment and neuroinflammatory responses by regulating P38/STAT3 signaling. J Neuroinflammation 2022; 19:187. [PMID: 35841100 PMCID: PMC9288088 DOI: 10.1186/s12974-022-02549-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 07/05/2022] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND In chronic myelogenous leukemia, reciprocal translocation between chromosome 9 and chromosome 22 generates a chimeric protein, Bcr-Abl, that leads to hyperactivity of tyrosine kinase-linked signaling transduction. The therapeutic agent nilotinib inhibits Bcr-Abl/DDR1 and can cross the blood-brain barrier, but its potential impact on neuroinflammatory responses and cognitive function has not been studied in detail. METHODS The effects of nilotinib in vitro and in vivo were assessed by a combination of RT-PCR, real-time PCR, western blotting, ELISA, immunostaining, and/or subcellular fractionation. In the in vitro experiments, the effects of 200 ng/mL LPS or PBS on BV2 microglial cells, primary microglia or primary astrocytes pre- or post-treated with 5 µM nilotinib or vehicle were evaluated. The in vivo experiments involved wild-type mice administered a 7-day course of daily injections with 20 mg/kg nilotinib (i.p.) or vehicle before injection with 10 mg/kg LPS (i.p.) or PBS. RESULTS In BV2 microglial cells, pre- and post-treatment with nilotinib altered LPS-induced proinflammatory/anti-inflammatory cytokine mRNA levels by suppressing AKT/P38/SOD2 signaling. Nilotinib treatment also significantly downregulated LPS-stimulated proinflammatory cytokine levels in primary microglia and primary astrocytes by altering P38/STAT3 signaling. Experiments in wild-type mice showed that nilotinib administration affected LPS-mediated microglial/astroglial activation in a brain region-specific manner in vivo. In addition, nilotinib significantly reduced proinflammatory cytokine IL-1β, IL-6 and COX-2 levels and P38/STAT3 signaling in the brain in LPS-treated wild-type mice. Importantly, nilotinib treatment rescued LPS-mediated spatial working memory impairment and cortical dendritic spine number in wild-type mice. CONCLUSIONS Our results indicate that nilotinib can modulate neuroinflammatory responses and cognitive function in LPS-stimulated wild-type mice.
Collapse
Affiliation(s)
- Jieun Kim
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Korea
| | - Hyun-Ju Lee
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Korea
| | - Jin-Hee Park
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Korea.,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Korea
| | - Byung-Yoon Cha
- PharmacoRex Co., Ltd., 20 Techno 1-ro, Yuseong-gu, Daejeon, 34016, Korea
| | - Hyang-Sook Hoe
- Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), 61, Cheomdan-ro, Dong-gu, Daegu, 41062, Korea. .,Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science & Technology, Daegu, 42988, Korea.
| |
Collapse
|
46
|
Sidoroff V, Bower P, Stefanova N, Fanciulli A, Stankovic I, Poewe W, Seppi K, Wenning GK, Krismer F. Disease-Modifying Therapies for Multiple System Atrophy: Where Are We in 2022? JOURNAL OF PARKINSON'S DISEASE 2022; 12:1369-1387. [PMID: 35491799 PMCID: PMC9398078 DOI: 10.3233/jpd-223183] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Multiple system atrophy is a rapidly progressive and fatal neurodegenerative disorder. While numerous preclinical studies suggested efficacy of potentially disease modifying agents, none of those were proven to be effective in large-scale clinical trials. Three major strategies are currently pursued in preclinical and clinical studies attempting to slow down disease progression. These target α-synuclein, neuroinflammation, and restoration of neurotrophic support. This review provides a comprehensive overview on ongoing preclinical and clinical developments of disease modifying therapies. Furthermore, we will focus on potential shortcomings of previous studies that can be avoided to improve data quality in future studies of this rare disease.
Collapse
Affiliation(s)
- Victoria Sidoroff
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Pam Bower
- The Multiple System Atrophy Coalition, Inc., McLean, VA, USA
| | - Nadia Stefanova
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | | | - Iva Stankovic
- Neurology Clinic, University Clinical Center of Serbia, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Werner Poewe
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Klaus Seppi
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gregor K Wenning
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Florian Krismer
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
47
|
Ferguson MW, Kennedy CJ, Palpagama TH, Waldvogel HJ, Faull RLM, Kwakowsky A. Current and Possible Future Therapeutic Options for Huntington's Disease. J Cent Nerv Syst Dis 2022; 14:11795735221092517. [PMID: 35615642 PMCID: PMC9125092 DOI: 10.1177/11795735221092517] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 03/21/2022] [Indexed: 11/16/2022] Open
Abstract
Huntington's disease (HD) is an autosomal neurodegenerative disease that is characterized by an excessive number of CAG trinucleotide repeats within the huntingtin gene (HTT). HD patients can present with a variety of symptoms including chorea, behavioural and psychiatric abnormalities and cognitive decline. Each patient has a unique combination of symptoms, and although these can be managed using a range of medications and non-drug treatments there is currently no cure for the disease. Current therapies prescribed for HD can be categorized by the symptom they treat. These categories include chorea medication, antipsychotic medication, antidepressants, mood stabilizing medication as well as non-drug therapies. Fortunately, there are also many new HD therapeutics currently undergoing clinical trials that target the disease at its origin; lowering the levels of mutant huntingtin protein (mHTT). Currently, much attention is being directed to antisense oligonucleotide (ASO) therapies, which bind to pre-RNA or mRNA and can alter protein expression via RNA degradation, blocking translation or splice modulation. Other potential therapies in clinical development include RNA interference (RNAi) therapies, RNA targeting small molecule therapies, stem cell therapies, antibody therapies, non-RNA targeting small molecule therapies and neuroinflammation targeted therapies. Potential therapies in pre-clinical development include Zinc-Finger Protein (ZFP) therapies, transcription activator-like effector nuclease (TALEN) therapies and clustered regularly interspaced short palindromic repeats (CRISPR)/CRISPR-associated system (Cas) therapies. This comprehensive review aims to discuss the efficacy of current HD treatments and explore the clinical trial progress of emerging potential HD therapeutics.
Collapse
Affiliation(s)
- Mackenzie W. Ferguson
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Connor J. Kennedy
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Thulani H. Palpagama
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Henry J. Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard L. M. Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Pharmacology and Therapeutics, School of Medicine, Galway Neuroscience Centre, National University of Ireland Galway, Galway, Ireland
| |
Collapse
|
48
|
Rodriguez-Porcel F, Wyman-Chick KA, Abdelnour Ruiz C, Toledo JB, Ferreira D, Urwyler P, Weil RS, Kane J, Pilotto A, Rongve A, Boeve B, Taylor JP, McKeith I, Aarsland D, Lewis SJG. Clinical outcome measures in dementia with Lewy bodies trials: critique and recommendations. Transl Neurodegener 2022; 11:24. [PMID: 35491418 PMCID: PMC9059356 DOI: 10.1186/s40035-022-00299-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 03/31/2022] [Indexed: 12/28/2022] Open
Abstract
The selection of appropriate outcome measures is fundamental to the design of any successful clinical trial. Although dementia with Lewy bodies (DLB) is one of the most common neurodegenerative conditions, assessment of therapeutic benefit in clinical trials often relies on tools developed for other conditions, such as Alzheimer's or Parkinson's disease. These may not be sufficiently valid or sensitive to treatment changes in DLB, decreasing their utility. In this review, we discuss the limitations and strengths of selected available tools used to measure DLB-associated outcomes in clinical trials and highlight the potential roles for more specific objective measures. We emphasize that the existing outcome measures require validation in the DLB population and that DLB-specific outcomes need to be developed. Finally, we highlight how the selection of outcome measures may vary between symptomatic and disease-modifying therapy trials.
Collapse
Affiliation(s)
- Federico Rodriguez-Porcel
- Department of Neurology, Medical University of South Carolina, 208b Rutledge Av., Charleston, SC, 29403, USA.
| | - Kathryn A Wyman-Chick
- Department of Neurology, Center for Memory and Aging, HealthPartners, Saint Paul, MN, USA
| | | | - Jon B Toledo
- Fixel Institute for Neurological Diseases, University of Florida, Gainesville, FL, USA
| | - Daniel Ferreira
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences, and Society, Center for Alzheimer's Research, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Prabitha Urwyler
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Rimona S Weil
- Dementia Research Centre, University College London, London, UK
| | - Joseph Kane
- Centre for Public Health, Queen's University, Belfast, UK
| | - Andrea Pilotto
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Arvid Rongve
- Department of Research and Innovation, Helse Fonna, Haugesund Hospital, Haugesund, Norway
- Institute of Clinical Medicine (K1), The University of Bergen, Bergen, Norway
| | - Bradley Boeve
- Department of Neurology, Center for Sleep Medicine, Mayo Clinic, Rochester, MN, USA
| | - John-Paul Taylor
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Ian McKeith
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Dag Aarsland
- Department of Old Age Psychiatry Institute of Psychiatry Psychology and Neuroscience, King's College London, London, UK
| | - Simon J G Lewis
- ForeFront Parkinson's Disease Research Clinic, Brain and Mind Centre, School of Medical Sciences, University of Sydney, 100 Mallett Street, Camperdown, NSW, 2050, Australia
| |
Collapse
|
49
|
Vijayakumar D, Jankovic J. Slowing Parkinson's Disease Progression with Vaccination and Other Immunotherapies. CNS Drugs 2022; 36:327-343. [PMID: 35212935 DOI: 10.1007/s40263-022-00903-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/23/2022] [Indexed: 12/29/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. There are several recognized pathways leading up to dopaminergic neuron loss in the substantia nigra pars compacta and other cells in the brain as a result of age-related, genetic, environmental, and other processes. Of these, the most prominent is the role played by the protein α-synuclein, which aggregates and is the primary component of Lewy bodies, the histopathological hallmark of PD. The latest disease-modifying treatment options being investigated in PD are active and passive immunization against α-synuclein. There are currently five different monoclonal antibodies investigated as passive immunization and three drugs being studied as active immunization modalities in PD. These work through different mechanisms but with a common goal-to minimize or prevent α-synuclein-driven neurotoxicity by reducing α-synuclein synthesis, increasing α-synuclein degradation, and preventing aggregation and propagation from cell to cell. These promising strategies, along with other potential therapies, may favorably alter disease progression in PD.
Collapse
Affiliation(s)
- Dhanya Vijayakumar
- Department of Medicine, Prisma Health Upstate, The University of South Carolina School of Medicine Greenville, Greenville, South Carolina, USA
| | - Joseph Jankovic
- Distinguished Chair in Movement Disorders, Parkinson's Disease Center and Movement Disorders Clinic, Department of Neurology, Baylor College of Medicine, Baylor St. Luke's Medical Center at the McNair Campus, 7200 Cambridge, 9th Floor, Suite 9A, Houston, TX, 77030-4202, USA.
| |
Collapse
|
50
|
Marín T, Dulcey AE, Campos F, de la Fuente C, Acuña M, Castro J, Pinto C, Yañez MJ, Cortez C, McGrath DW, Sáez PJ, Gorshkov K, Zheng W, Southall N, Carmo-Fonseca M, Marugán J, Alvarez AR, Zanlungo S. c-Abl Activation Linked to Autophagy-Lysosomal Dysfunction Contributes to Neurological Impairment in Niemann-Pick Type A Disease. Front Cell Dev Biol 2022; 10:844297. [PMID: 35399514 PMCID: PMC8985125 DOI: 10.3389/fcell.2022.844297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 02/25/2022] [Indexed: 12/05/2022] Open
Abstract
Niemann-Pick type A (NPA) disease is a fatal lysosomal neurodegenerative disorder caused by the deficiency in acid sphingomyelinase (ASM) activity. NPA patients present severe and progressive neurodegeneration starting at an early age. Currently, there is no effective treatment for this disease and NPA patients die between 2 and 3 years of age. NPA is characterized by an accumulation of sphingomyelin in lysosomes and dysfunction in the autophagy-lysosomal pathway. Recent studies show that c-Abl tyrosine kinase activity downregulates autophagy and the lysosomal pathway. Interestingly, this kinase is also activated in other lysosomal neurodegenerative disorders. Here, we describe that c-Abl activation contributes to the mechanisms of neuronal damage and death in NPA disease. Our data demonstrate that: 1) c-Abl is activated in-vitro as well as in-vivo NPA models; 2) imatinib, a clinical c-Abl inhibitor, reduces autophagy-lysosomal pathway alterations, restores autophagy flux, and lowers sphingomyelin accumulation in NPA patient fibroblasts and NPA neuronal models and 3) chronic treatment with nilotinib and neurotinib, two c-Abl inhibitors with differences in blood-brain barrier penetrance and target binding mode, show further benefits. While nilotinib treatment reduces neuronal death in the cerebellum and improves locomotor functions, neurotinib decreases glial activation, neuronal disorganization, and loss in hippocampus and cortex, as well as the cognitive decline of NPA mice. Our results support the participation of c-Abl signaling in NPA neurodegeneration and autophagy-lysosomal alterations, supporting the potential use of c-Abl inhibitors for the clinical treatment of NPA patients.
Collapse
Affiliation(s)
- Tamara Marín
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Andrés E. Dulcey
- Early Translation Branch, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, United States
| | - Fabián Campos
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina de la Fuente
- Laboratory of Cell Signaling, Center for Aging and Regeneration (CARE), Millennium Institute on Immunology and Immunotherapy (IMII), Department of Cellular and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Mariana Acuña
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Juan Castro
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudio Pinto
- Laboratory of Cell Signaling, Center for Aging and Regeneration (CARE), Millennium Institute on Immunology and Immunotherapy (IMII), Department of Cellular and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María José Yañez
- School of Medical Technology, Health Sciences Faculty, Universidad San Sebastián, Santiago, Chile
| | - Cristian Cortez
- Center for Genomics and Bioinformatics, Faculty of Science, Universidad Mayor, Santiago, Chile
| | - David W. McGrath
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Pablo J. Sáez
- Cell Communication and Migration Laboratory, Institute of Biochemistry and Molecular Cell Biology, Center for Experimental Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kirill Gorshkov
- Early Translation Branch, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, United States
| | - Wei Zheng
- Early Translation Branch, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, United States
| | - Noel Southall
- Early Translation Branch, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, United States
| | - Maria Carmo-Fonseca
- Instituto de Medicina Molecular Joȧo Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Juan Marugán
- Early Translation Branch, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, MD, United States
- *Correspondence: Juan Marugán, ; Alejandra R. Alvarez, ; Silvana Zanlungo,
| | - Alejandra R. Alvarez
- Laboratory of Cell Signaling, Center for Aging and Regeneration (CARE), Millennium Institute on Immunology and Immunotherapy (IMII), Department of Cellular and Molecular Biology, Biological Sciences Faculty, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Juan Marugán, ; Alejandra R. Alvarez, ; Silvana Zanlungo,
| | - Silvana Zanlungo
- Department of Gastroenterology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- *Correspondence: Juan Marugán, ; Alejandra R. Alvarez, ; Silvana Zanlungo,
| |
Collapse
|