1
|
El-Atawneh S, Goldblum A. A Machine Learning Algorithm Suggests Repurposing Opportunities for Targeting Selected GPCRs. Int J Mol Sci 2024; 25:10230. [PMID: 39337714 PMCID: PMC11432050 DOI: 10.3390/ijms251810230] [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: 07/17/2024] [Revised: 09/18/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Repurposing utilizes existing drugs with known safety profiles and discovers new uses by combining experimental and computational approaches. The integration of computational methods has greatly advanced drug repurposing, offering a rational approach and reducing the risk of failure in these efforts. Recognizing the potential for drug repurposing, we employed our Iterative Stochastic Elimination (ISE) algorithm to screen known drugs from the DrugBank database. Repurposing in our hands is based on computer models of the actions of ligands: the ISE algorithm is a machine learning tool that creates ligand-based models by distinguishing between the physicochemical properties of known drugs and those of decoys. The models are large sets of "filters" made out, each, of molecular properties. We screen and score external sets of molecules (in our case- the DrugBank molecules) by our agonism and antagonism models based on published data (i.e., IC50, Ki, or EC50) and pick the top-scoring molecules as candidates for experiments. Such agonist and antagonist models for six G-protein coupled receptors (GPCRs) families facilitated the identification of repurposing opportunities. Our screening revealed 5982 new potential molecular actions (agonists, antagonists), which suggest repurposing candidates for the cannabinoid 2 (CB2), histamine (H1, H3, and H4), and dopamine 3 (D3) receptors, which may be useful to treat conditions such as neuroinflammation, obesity, allergic dermatitis, and drug abuse. These sets of best candidates should now be examined by experimentalists: based on previous such experiments, there is a very high chance of discovering novel highly bioactive molecules.
Collapse
Affiliation(s)
- Shayma El-Atawneh
- Molecular Modelling and Drug Design Lab, Institute for Drug Research and Fraunhofer Project Center for Drug Discovery and Delivery, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Amiram Goldblum
- Molecular Modelling and Drug Design Lab, Institute for Drug Research and Fraunhofer Project Center for Drug Discovery and Delivery, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| |
Collapse
|
2
|
Singh R, Rani N, Kaur R, Chahal G, Kumar P, Kaur G. Exploring the Therapeutic Potential of Alkaloids in Alzheimer's Disease Management. Cent Nerv Syst Agents Med Chem 2024; 24:206-218. [PMID: 38213138 DOI: 10.2174/0118715249269092231109181638] [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: 07/20/2023] [Revised: 09/15/2023] [Accepted: 10/05/2023] [Indexed: 01/13/2024]
Abstract
BACKGROUND Alkaloids are important phytoconstituents obtained from various plant sources. The study's primary goal is to assess the anti-Alzheimer potential of alkaloids using a molecular docking study. Alzheimer's disease (AD) is considered a gradual decline in memory, reasoning, decision-making, orientation to one's physical surroundings, and language. MATERIALS AND METHODS The main target i.e. acetylcholinesterase proteins was selected for the molecular docking study. RESULTS The structures of various alkaloids were drawn using Chem Draw Software, PDB was retrieved from the RCSB PDB database, and molecular docking study was performed on Molergo Virtual Docker. The potential alkaloids were identified with anti-Alzheimer potency. CONCLUSION Reserpine, vinblastine, ergotamine, and tubocurarine were found to exhibit potential anti-Alzheimer potency.
Collapse
Affiliation(s)
- Randhir Singh
- Department of Pharmaceutical Sciences, Central University of Punjab, Bathinda, India
| | - Nidhi Rani
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Geeta Chahal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Praveen Kumar
- SunPharma, Hill Top Area, Vill. Bhatolikalan, Himachal Pradesh, India- 174103
| | - Gagandeep Kaur
- Chitkara College of Pharmacy, Chitkara University, Himachal Pradesh, India
| |
Collapse
|
3
|
Pak ME, Yang HJ, Li W, Kim JK, Go Y. Yuk-Gunja-Tang attenuates neuronal death and memory impairment via ERK/CREB/BDNF signaling in the hippocampi of experimental Alzheimer’s disease model. Front Pharmacol 2022; 13:1014840. [PMID: 36386241 PMCID: PMC9643579 DOI: 10.3389/fphar.2022.1014840] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/12/2022] [Indexed: 11/23/2022] Open
Abstract
Yuk-Gunja-Tang (YG) is the Korean traditional medicine in East Asia for gastrointestinal disorders. In the present study, we determined the protective effects of YG on glutamate-induced cytotoxicity in HT22 hippocampal neuronal cells and mice with scopolamine-induced memory impairment. In vitro assessments were performed using a cell viability assay, flow cytometry, and Western blotting, while in vivo assessments were performed in C57BL/6 mice administered with YG for 7 days and injected with scopolamine (1 mg/kg) for 7 days. We assessed the memory function using the Y-maze, novel object recognition, and passive avoidance tests. Protein expression analyses and histological analyses were performed using hippocampal tissues. YG treatment significantly restored cell viability against glutamate-induced apoptosis. It significantly suppressed glutamate-induced reactive oxygen species accumulation and mitochondrial dysfunction. It also increased Bcl-2 protein expression and decreased HO-1 protein expression. It activated the extracellular signal-regulated kinase/cAMP response element binding protein (ERK/CREB) signaling pathway and increased the expression of brain-derived neurotrophic factor (BDNF) under excitotoxic conditions. In the scopolamine-injected mice, YG ameliorated memory impairment in the Y-maze, novel object recognition, and passive avoidance tests; restored dysfunction in the acetylcholine, acetylcholinesterase expression levels; reduced neuronal damage in Nissl staining; and increased BDNF and phosphorylated ERK and CREB levels in Western blotting and immunofluorescence staining. Thus, YG exerted neuroprotective effects by activating ERK/CREB/BDNF signaling in the hippocampus, indicating its potential cognition-enhancing effects, especially in Alzheimer’s disease.
Collapse
|
4
|
Firdaus Z, Singh TD. An Insight in Pathophysiological Mechanism of Alzheimer's Disease and its Management Using Plant Natural Products. Mini Rev Med Chem 2021; 21:35-57. [PMID: 32744972 DOI: 10.2174/1389557520666200730155928] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 05/20/2020] [Accepted: 06/05/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is an age-associated nervous system disorder and a leading cause of dementia worldwide. Clinically, it is described by cognitive impairment and pathophysiologically by deposition of amyloid plaques and neurofibrillary tangles in the brain and neurodegeneration. This article reviews the pathophysiology, course of neuronal degeneration, and the various possible hypothesis of AD progression. These hypotheses include amyloid cascade, tau hyperphosphorylation, cholinergic disruption, metal dysregulation, vascular dysfunction, oxidative stress, and neuroinflammation. There is an exponential increase in the occurrence of AD in the recent few years that indicate an urgent need to develop some effective treatment. Currently, only 2 classes of drugs are available for AD treatment, namely acetylcholinesterase inhibitor and NMDA receptor antagonist. Since AD is a complex neurological disorder and these drugs use a single target approach, alternatives are needed due to limited effectiveness and unpleasant side-effects of these drugs. Currently, plants have been used for drug development research especially because of their multiple sites of action and fewer side effects. Uses of some herbs and phytoconstituents for the management of neuronal disorders like AD have been documented in this article. Phytochemical screening of these plants shows the presence of many beneficial constituents like flavonoids, triterpenes, alkaloids, sterols, polyphenols, and tannins. These compounds show a wide array of pharmacological activities, such as anti-amyloidogenic, anticholinesterase, and antioxidants. This article summarizes the present understanding of AD progression and gathers biochemical evidence from various works on natural products that can be useful in the management of this disease.
Collapse
Affiliation(s)
- Zeba Firdaus
- Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi - 221005, India
| | - Tryambak Deo Singh
- Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi - 221005, India
| |
Collapse
|
5
|
Law CSW, Yeong KY. Repurposing Antihypertensive Drugs for the Management of Alzheimer's Disease. Curr Med Chem 2021; 28:1716-1730. [PMID: 32164502 DOI: 10.2174/0929867327666200312114223] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/04/2020] [Accepted: 02/18/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that has affected millions of people worldwide. However, currently, there is no treatment to cure the disease. The AD drugs available in the market only manage the disease symptomatically and the effects are usually short-term. Thus, there is a need to look at alternatives AD therapies. This literature review aims to shed some light on the potential of repurposing antihypertensives to treat AD. Mid-life hypertension has not only been recognised as a risk factor for AD, but its relation with AD has also been well established. Hence, antihypertensives were postulated to be beneficial in managing AD. Four classes of antihypertensives, as well as their potential limitations and prospects in being utilised as AD therapeutics, were discussed in this review.
Collapse
Affiliation(s)
- Christine Shing Wei Law
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Keng Yoon Yeong
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| |
Collapse
|
6
|
Ayoub BM, Michel HE, Mowaka S, Hendy MS, Tadros MM. Repurposing of Omarigliptin as a Neuroprotective Agent Based on Docking with A 2A Adenosine and AChE Receptors, Brain GLP-1 Response and Its Brain/Plasma Concentration Ratio after 28 Days Multiple Doses in Rats Using LC-MS/MS. Molecules 2021; 26:molecules26040889. [PMID: 33567615 PMCID: PMC7915074 DOI: 10.3390/molecules26040889] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 12/26/2022] Open
Abstract
The authors in the current work suggested the potential repurposing of omarigliptin (OMR) for neurodegenerative diseases based on three new findings that support the preliminary finding of crossing BBB after a single dose study in the literature. The first finding is the positive results of the docking study with the crystal structures of A2A adenosine (A2AAR) and acetylcholine esterase (AChE) receptors. A2AAR is a member of non-dopaminergic GPCR superfamily receptor proteins and has essential role in regulation of glutamate and dopamine release in Parkinson’s disease while AChE plays a major role in Alzheimer’s disease as the primary enzyme responsible for the hydrolytic metabolism of the neurotransmitter acetylcholine into choline and acetate. Docking showed that OMR perfectly fits into A2AAR binding pocket forming a distinctive hydrogen bond with Threonine 256. Besides other non-polar interactions inside the pocket suggesting the future of the marketed anti-diabetic drug (that cross BBB) as a potential antiparkinsonian agent while OMR showed perfect fit inside AChE receptor binding site smoothly because of its optimum length and the two fluorine atoms that enables quite lean fitting. Moreover, a computational comparative study of OMR docking, other 12 DPP-4 inhibitors and 11 SGLT-2 inhibitors was carried out. Secondly, glucagon-like peptide-1 (GLP-1) concentration in rats’ brain tissue was determined by the authors using sandwich GLP-1 ELISA kit bio-analysis to ensure the effect of OMR after the multiple doses’ study. Brain GLP-1 concentration was elevated by 1.9-fold following oral multiple doses of OMR (5 mg/kg/day, p.o. for 28 days) as compared to the control group. The third finding is the enhanced BBB crossing of OMR after 28 days of multiple doses that had been studied using LC-MS/MS method with enhanced liquid–liquid extraction. A modified LC-MS/MS method was established for bioassay of OMR in rats’ plasma (10–3100 ng/mL) and rats’ brain tissue (15–2900 ng/mL) using liquid–liquid extraction. Alogliptin (ALP) was chosen as an internal standard (IS) due to its LogP value of 1.1, which is very close to the LogP of OMR. Extraction of OMR from samples of both rats’ plasma and rats’ brain tissue was effectively achieved with ethyl acetate as the extracting solvent after adding 1N sodium carbonate to enhance the drug migration, while choosing acetonitrile to be the diluent solvent for the IS to effectively decrease any emulsion between the layers in the stated method of extraction. Validation results were all pleasing including good stability studies with bias of value below 20%. Concentration of OMR in rats’ plasma were determined after 2 h of the latest dose from 28 days multiple doses, p.o, 5 mg/kg/day. It was found to be 1295.66 ± 684.63 ng/mL estimated from the bio-analysis regression equation. OMR passed through the BBB following oral administration and exhibited concentration of 543.56 ± 344.15 ng/g in brain tissue, taking in consideration the dilution factor of 10. The brain/plasma concentration ratio of 0.42 (543.56/1295.66) was used to illustrate the penetration power through the BBB after the multiple doses for 28 days. Results showed that OMR passed through the BBB more effectively in the multiple dose study as compared to the previously published single dose study by the authors. Thus, the present study suggests potential repositioning of OMR as antiparkinsonian agent that will be of interest for researchers interested in neurodegenerative diseases.
Collapse
Affiliation(s)
- Bassam M. Ayoub
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt; (S.M.); (M.S.H.)
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt
- Correspondence: ; Tel.: +20-226-890-000; Fax: +20-226-300-010
| | - Haidy E. Michel
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Ain Shams University, Organization of African Unity Street, Abassia, Cairo 11566, Egypt;
| | - Shereen Mowaka
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt; (S.M.); (M.S.H.)
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt
- Analytical Chemistry Department, Faculty of Pharmacy, Helwan University, Ain Helwan, Cairo 11795, Egypt
| | - Moataz S. Hendy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt; (S.M.); (M.S.H.)
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt
| | - Mariam M. Tadros
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Organization of African Unity Street, Abassia, Cairo 11566, Egypt;
| |
Collapse
|
7
|
Yang D, Zhou Q, Labroska V, Qin S, Darbalaei S, Wu Y, Yuliantie E, Xie L, Tao H, Cheng J, Liu Q, Zhao S, Shui W, Jiang Y, Wang MW. G protein-coupled receptors: structure- and function-based drug discovery. Signal Transduct Target Ther 2021; 6:7. [PMID: 33414387 PMCID: PMC7790836 DOI: 10.1038/s41392-020-00435-w] [Citation(s) in RCA: 261] [Impact Index Per Article: 87.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 02/08/2023] Open
Abstract
As one of the most successful therapeutic target families, G protein-coupled receptors (GPCRs) have experienced a transformation from random ligand screening to knowledge-driven drug design. We are eye-witnessing tremendous progresses made recently in the understanding of their structure-function relationships that facilitated drug development at an unprecedented pace. This article intends to provide a comprehensive overview of this important field to a broader readership that shares some common interests in drug discovery.
Collapse
Affiliation(s)
- Dehua Yang
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Qingtong Zhou
- School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China
| | - Viktorija Labroska
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Shanshan Qin
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Sanaz Darbalaei
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yiran Wu
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Elita Yuliantie
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Linshan Xie
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Houchao Tao
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Jianjun Cheng
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China
| | - Qing Liu
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.,The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China
| | - Suwen Zhao
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China.,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Wenqing Shui
- iHuman Institute, ShanghaiTech University, 201210, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China.
| | - Yi Jiang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China.
| | - Ming-Wei Wang
- The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China. .,The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, China. .,School of Basic Medical Sciences, Fudan University, 200032, Shanghai, China. .,University of Chinese Academy of Sciences, 100049, Beijing, China. .,School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China. .,School of Pharmacy, Fudan University, 201203, Shanghai, China.
| |
Collapse
|
8
|
Dighe SN, Tippana M, van Akker S, Collet TA. Structure-Based Scaffold Repurposing toward the Discovery of Novel Cholinesterase Inhibitors. ACS OMEGA 2020; 5:30971-30979. [PMID: 33324805 PMCID: PMC7726787 DOI: 10.1021/acsomega.0c03848] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/12/2020] [Indexed: 05/06/2023]
Abstract
Cholinesterases (ChE) are well-known drug targets for the treatment of Alzheimer's disease (AD). In continuation of work to develop novel cholinesterase inhibitors, we utilized a structure-based scaffold repurposing approach and discovered six novel ChE inhibitors from our recently developed DNA gyrase inhibitor library. Among the identified hits, two compounds (denoted 3 and 18) were found to be the most potent inhibitor of acetylcholinesterase (AChE, IC50 = 6.10 ± 1.01 μM) and butyrylcholinesterase (BuChE, IC50 = 5.50 ± 0.007 μM), respectively. Compound 3 was responsible for the formation of H-bond and π-π stacking interactions within the active site of AChE. In contrast, compound 18 was well fitted in the choline-binding pocket and catalytic site of BuChE. Results obtained from in vitro cytotoxicity assays and in silico derived physicochemical and absorption, distribution, metabolism, and excretion (ADME) properties indicate that repurposed scaffold 3 and 18 could be potential drug candidates for further development as novel ChE inhibitors.
Collapse
|
9
|
Bauzon J, Lee G, Cummings J. Repurposed agents in the Alzheimer's disease drug development pipeline. Alzheimers Res Ther 2020; 12:98. [PMID: 32807237 PMCID: PMC7433208 DOI: 10.1186/s13195-020-00662-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/29/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Treatments are needed to address the growing prevalence of Alzheimer's disease (AD). Clinical trials have failed to produce any AD drugs for Food and Drug Administration (FDA) approval since 2003, and the pharmaceutical development process is both time-consuming and costly. Drug repurposing provides an opportunity to accelerate this process by investigating the AD-related effects of agents approved for other indications. These drugs have known safety profiles, pharmacokinetic characterization, formulations, doses, and manufacturing processes. METHODS We assessed repurposed AD therapies represented in Phase I, Phase II, and Phase III of the current AD pipeline as registered on ClinicalTrials.gov as of February 27, 2020. RESULTS We identified 53 clinical trials involving 58 FDA-approved agents. Seventy-eight percent of the agents in trials had putative disease-modifying mechanisms of action. Of the repurposed drugs in the pipeline 20% are hematologic-oncologic agents, 18% are drugs derived from cardiovascular indications, 14% are agents with psychiatric uses, 12% are drug used to treat diabetes, 10% are neurologic agents, and the remaining 26% of drugs fall under other conditions. Intellectual property strategies utilized in these programs included using the same drug but altering doses, routes of administration, or formulations. Most repurposing trials were supported by Academic Medical Centers and were not funded through the biopharmaceutical industry. We compared our results to a European trial registry and found results similar to those derived from ClinicalTrials.gov. CONCLUSIONS Drug repurposing is a common approach to AD drug development and represents 39% of trials in the current AD pipeline. Therapies from many disease areas provide agents potentially useful in AD. Most of the repurposed agents are generic and a variety of intellectual property strategies have been adopted to enhance their economic value.
Collapse
Affiliation(s)
- Justin Bauzon
- School of Medicine, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, 89154, USA
| | - Garam Lee
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, 89106, USA
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, 89106, USA.
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas (UNLV), Las Vegas, NV, 89154, USA.
| |
Collapse
|
10
|
Zhu Y, Jung W, Wang F, Che C. Drug repurposing against Parkinson's disease by text mining the scientific literature. LIBRARY HI TECH 2020. [DOI: 10.1108/lht-08-2019-0170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
PurposeDrug repurposing involves the identification of new applications for existing drugs. Owing to the enormous rise in the costs of pharmaceutical R&D, several pharmaceutical companies are leveraging repurposing strategies. Parkinson's disease is the second most common neurodegenerative disorder worldwide, affecting approximately 1–2 percent of the human population older than 65 years. This study proposes a literature-based drug repurposing strategy in Parkinson's disease.Design/methodology/approachThe literature-based drug repurposing strategy proposed herein combined natural language processing, network science and machine learning methods for analyzing unstructured text data and producing actional knowledge for drug repurposing. The approach comprised multiple computational components, including the extraction of biomedical entities and their relationships, knowledge graph construction, knowledge representation learning and machine learning-based prediction.FindingsThe proposed strategy was used to mine information pertaining to the mechanisms of disease treatment from known treatment relationships and predict drugs for repurposing against Parkinson's disease. The F1 score of the best-performing method was 0.97, indicating the effectiveness of the proposed approach. The study also presents experimental results obtained by combining the different components of the strategy.Originality/valueThe drug repurposing strategy proposed herein for Parkinson's disease is distinct from those existing in the literature in that the drug repurposing pipeline includes components of natural language processing, knowledge representation and machine learning for analyzing the scientific literature. The results of the study provide important and valuable information to researchers studying different aspects of Parkinson's disease.
Collapse
|
11
|
Lebouvier T, Chen Y, Duriez P, Pasquier F, Bordet R. Antihypertensive agents in Alzheimer's disease: beyond vascular protection. Expert Rev Neurother 2019; 20:175-187. [PMID: 31869274 DOI: 10.1080/14737175.2020.1708195] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Introduction: Midlife hypertension has been consistently linked with increased risk of cognitive decline and Alzheimer's disease (AD). Observational studies and randomized trials show that the use of antihypertensive therapy is associated with a lesser incidence or prevalence of cognitive impairment and dementia. However, whether antihypertensive agents specifically target the pathological process of AD remains elusive.Areas covered: This review of literature provides an update on the clinical and preclinical arguments supporting anti-AD properties of antihypertensive drugs. The authors focused on validated all classes of antihypertensive treatments such as angiotensin-converting enzyme inhibitors (ACEi), angiotensin receptor blockers (ARB), calcium channel blockers (CCB), β-blockers, diuretics, neprilysin inhibitors, and other agents. Three main mechanisms can be advocated: action on the concurrent vascular pathology, action on the vascular component of Alzheimer's pathophysiology, and action on nonvascular targets.Expert opinion: In 2019, while there is no doubt that hypertension should be treated in primary prevention of vascular disease and in secondary prevention of stroke and mixed dementia, the place of antihypertensive agents in the secondary prevention of 'pure' AD remains an outstanding question.
Collapse
Affiliation(s)
- Thibaud Lebouvier
- Inserm URM_S1172, University of Lille, Lille, France.,DISTALZ, University of Lille, Lille, France
| | - Yaohua Chen
- DISTALZ, University of Lille, Lille, France.,Inserm, CHU Lille, University of Lille, Lille, France
| | | | - Florence Pasquier
- DISTALZ, University of Lille, Lille, France.,Inserm, CHU Lille, University of Lille, Lille, France
| | - Régis Bordet
- Inserm, CHU Lille, University of Lille, Lille, France
| |
Collapse
|
12
|
Vyas S, Kothari S, Kachhwaha S. Nootropic medicinal plants: Therapeutic alternatives for Alzheimer’s disease. J Herb Med 2019. [DOI: 10.1016/j.hermed.2019.100291] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
13
|
Papanagnou P, Papadopoulos GE, Stivarou T, Pappas A. Toward fully exploiting the therapeutic potential of marketed pharmaceuticals: the use of octreotide and chloroquine in oncology. Onco Targets Ther 2018; 12:319-339. [PMID: 30643430 PMCID: PMC6317484 DOI: 10.2147/ott.s182685] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Pleiotropy in biological systems and their targeting allows many pharmaceuticals to be used for multiple therapeutic purposes. Fully exploiting the therapeutic properties of drugs that are already marketed would be highly advantageous. This is especially the case in the field of oncology, where the ineffectiveness of typical anticancer agents is a common issue, while the development of novel anticancer agents is a costly and particularly time-consuming process. Octreotide and chloroquine are two pharmaceuticals that exhibit profound antitumorigenic activities. However, the current therapeutic use of octreotide is restricted primarily to the management of acromegaly and neuroendocrine tumors, both of which are rare medical conditions. Similarly, chloroquine is used mainly for the treatment of malaria, which is designated as a rare disease in Western countries. This limited exploitation contradicts the experimental findings of numerous studies outlining the possible expansion of the use of octreotide to include the treatment of common human malignancies and the repositioning of chloroquine in oncology. Herein, we review the current knowledge on the antitumor function of these two agents stemming from preclinical or clinical experimentation. In addition, we present in silico evidence on octreotide potentially binding to multiple Wnt-pathway components. This will hopefully aid in the design of new efficacious anticancer therapeutic regimens with minimal toxicity, which represents an enormous unmet demand in oncology.
Collapse
Affiliation(s)
| | | | - Theodora Stivarou
- Immunology Laboratory, Immunology Department, Hellenic Pasteur Institute, Athens, Greece
| | - Anastasios Pappas
- Department of Urology, Agios Savvas Cancer Hospital, Athens 11522, Greece,
| |
Collapse
|
14
|
Ayoub BM, Mowaka S, Safar MM, Ashoush N, Arafa MG, Michel HE, Tadros MM, Elmazar MM, Mousa SA. Repositioning of Omarigliptin as a once-weekly intranasal Anti-parkinsonian Agent. Sci Rep 2018; 8:8959. [PMID: 29895906 PMCID: PMC5997767 DOI: 10.1038/s41598-018-27395-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/30/2018] [Indexed: 12/13/2022] Open
Abstract
Drug repositioning is a revolution breakthrough of drug discovery that presents outstanding privilege with already safer agents by scanning the existing candidates as therapeutic switching or repurposing for marketed drugs. Sitagliptin, vildagliptin, saxagliptin & linagliptin showed antioxidant and neurorestorative effects in previous studies linked to DPP-4 inhibition. Literature showed that gliptins did not cross the blood brain barrier (BBB) while omarigliptin was the first gliptin that crossed it successfully in the present work. LC-MS/MS determination of once-weekly anti-diabetic DPP-4 inhibitors; omarigliptin & trelagliptin in plasma and brain tissue was employed after 2 h of oral administration to rats. The brain/plasma concentration ratio was used to deduce the penetration power through the BBB. Results showed that only omarigliptin crossed the BBB due to its low molecular weight & lipophilic properties suggesting its repositioning as antiparkinsonian agent. The results of BBB crossing will be of interest for researchers interested in Parkinson's disease. A novel intranasal formulation was developed using sodium lauryl sulphate surfactant to solubilize the lipophilic omarigliptin with penetration enhancing & antimicrobial properties. Intranasal administration showed enhanced brain/plasma ratio by 3.3 folds compared to the oral group accompanied with 2.6 folds increase in brain glucagon-like peptide-1 concentration compared to the control group.
Collapse
Affiliation(s)
- Bassam M Ayoub
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt.
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt.
| | - Shereen Mowaka
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt
- Analytical Chemistry Department, Faculty of Pharmacy, Helwan University, Ein Helwan, Cairo, Egypt
| | - Marwa M Safar
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt
- Pharmacology & Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Cairo University, Kasr El-Aini st., Cairo, Egypt
| | - Nermeen Ashoush
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt
- Clinical Pharmacy and Pharmacy Practice Department, Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt
| | - Mona G Arafa
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt
- Pharmaceutics Department, Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt
- Chemotheraputic Unit, Mansoura University Hospitals, Mansoura, 35516, Egypt
| | - Haidy E Michel
- Pharmacology & Toxicology Department, Faculty of Pharmacy, Ain Shams University, El-Abaseya, Cairo, Egypt
| | - Mariam M Tadros
- Analytical Chemistry Department, Faculty of Pharmacy, Ain Shams University, El-Abaseya, Cairo, Egypt
| | - Mohamed M Elmazar
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt
- Pharmacology & Biochemistry Department, Faculty of Pharmacy, The British University in Egypt, El-Sherouk city, Cairo, Egypt
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY, United States
| |
Collapse
|
15
|
Gong CX, Liu F, Iqbal K. Multifactorial Hypothesis and Multi-Targets for Alzheimer’s Disease. J Alzheimers Dis 2018; 64:S107-S117. [DOI: 10.3233/jad-179921] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Cheng-Xin Gong
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA
| |
Collapse
|
16
|
Abstract
AbstractRapid advances in pharmacotherapy and bioinformatics has led to the discovery and growing popularity of drug repositioning which includes re-investigating or recycling of existing drugs for new indications. There are innumerable advantages as well as challenges of drug repositioning. Since de-novo drug discovery takes plenty of time, effort and money, it has proved to a preferred alternative strategy for accelerated drug discovery. Moreover it is relatively inexpensive and carries minimal risk due to availability of previous pharmacological, safety and toxicology data. The strategies used are Known drug – new target/Drug focus/Drug-centric, Known target- new indication/Target focus/Target-centric and Disease focus/Disease-centric. Drug repositioning is a new breakthrough strategy to benefit patients by offering safer and effective treatment using shelved drugs.
Collapse
Affiliation(s)
- Man Mohan Mehndiratta
- Department of Neurology, Janakpuri Super Specialty Hospital, Janakpuri, New Delhi 110058, India
| | - Swati Wadhai
- Department of Pathology, Janakpuri Super Specialty Hospital, Janakpuri, New Delhi 110058, India
| | - Brij Tyagi
- Janakpuri Super Specialty Hospital, Janakpuri, New Delhi 110058, India
| | - Natasha Gulati
- Department of Pathology, Janakpuri Super Specialty Hospital, Janakpuri, New Delhi 110058, India
| | - Madhu Sinha
- Department of Pathology, Janakpuri Super Specialty Hospital, Janakpuri, New Delhi 110058, India
| |
Collapse
|
17
|
Wu H, Huang J, Zhong Y, Huang Q. DrugSig: A resource for computational drug repositioning utilizing gene expression signatures. PLoS One 2017; 12:e0177743. [PMID: 28562632 PMCID: PMC5451001 DOI: 10.1371/journal.pone.0177743] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 05/02/2017] [Indexed: 12/12/2022] Open
Abstract
Computational drug repositioning has been proved as an effective approach to develop new drug uses. However, currently existing strategies strongly rely on drug response gene signatures which scattered in separated or individual experimental data, and resulted in low efficient outputs. So, a fully drug response gene signatures database will be very helpful to these methods. We collected drug response microarray data and annotated related drug and targets information from public databases and scientific literature. By selecting top 500 up-regulated and down-regulated genes as drug signatures, we manually established the DrugSig database. Currently DrugSig contains more than 1300 drugs, 7000 microarray and 800 targets. Moreover, we developed the signature based and target based functions to aid drug repositioning. The constructed database can serve as a resource to quicken computational drug repositioning. Database URL: http://biotechlab.fudan.edu.cn/database/drugsig/.
Collapse
Affiliation(s)
- Hongyu Wu
- School of Life Sciences, Fudan University, Shanghai, China
- Shanghai High-Tech United Bio-Technological R&D Co., Ltd., Shanghai, China
| | - Jinjiang Huang
- School of Life Sciences, Fudan University, Shanghai, China
| | - Yang Zhong
- School of Life Sciences, Fudan University, Shanghai, China
| | - Qingshan Huang
- School of Life Sciences, Fudan University, Shanghai, China
- Shanghai High-Tech United Bio-Technological R&D Co., Ltd., Shanghai, China
- * E-mail:
| |
Collapse
|
18
|
Khan A, Corbett A, Ballard C. Emerging amyloid and tau targeting treatments for Alzheimer’s disease. Expert Rev Neurother 2017; 17:697-711. [DOI: 10.1080/14737175.2017.1326819] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Ayesha Khan
- Institute for NanoBiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Anne Corbett
- King’s College London, Wolfson Centre for Age-Related Diseases, London, UK
| | - Clive Ballard
- King’s College London, Wolfson Centre for Age-Related Diseases, London, UK
| |
Collapse
|
19
|
Khan A, Corbett A, Ballard C. Emerging treatments for Alzheimer's disease for non-amyloid and non-tau targets. Expert Rev Neurother 2017; 17:683-695. [PMID: 28490260 DOI: 10.1080/14737175.2017.1326818] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION The number of people with dementia, including Alzheimer's disease, is growing as a result of an ageing global population. Treatments available for AD only alleviate the symptoms of the disease, and are effective in some people with AD for a limited time. There is no disease-modifying treatment available, and despite research efforts, the underlying mechanisms of AD and optimal treatment targets have not been fully elucidated. Amyloid and tau are key pathological markers of AD with ongoing trials targeting both. However, there are also many trials at various stages of development that primarily target other markers and processes implicated in the disease, which are now being investigated. Areas covered: This review summarizes current treatment approaches for AD and explores both repositioned and novel therapies that target non amyloid and non tau mechanisms that are in the clinical trials pipeline. This includes treatments for cognitive and neuropsychiatric symptoms and potentially disease modifying therapies. The studies included in this review have been obtained from searches of PubMed and clinical trials databases. Expert commentary: There is a renewed energy in identifying better treatments for behavioural symptoms of AD using both novel drugs and repositioning existing drugs. Lack of success in clinical trials of drugs targeting amyloid and tau have led to a surge in targeting alternative mechanisms. Progress in the development of biomarkers will provide further tools for clinical trials of potential therapeutics for both symptomatic treatment and disease modification in AD.
Collapse
Affiliation(s)
- Ayesha Khan
- a Institute for NanoBiotechnology , Johns Hopkins University , Baltimore , Maryland , USA
| | - Anne Corbett
- b King's College London , Wolfson Centre for Age-Related Diseases , London , UK
| | - Clive Ballard
- b King's College London , Wolfson Centre for Age-Related Diseases , London , UK
| |
Collapse
|
20
|
Cerajewska TL, Davies M, West NX. Periodontitis: a potential risk factor for Alzheimer's disease. Br Dent J 2016; 218:29-34. [PMID: 25571822 DOI: 10.1038/sj.bdj.2014.1137] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2014] [Indexed: 01/12/2023]
Abstract
The role of periodontitis as a risk factor for multiple systemic diseases is widely accepted and there is growing evidence of an association between periodontitis and sporadic late onset Alzheimer's disease (SLOAD). Recent epidemiologic, microbiologic and inflammatory findings strengthen this association, indicating that periodontal pathogens are possible contributors to neural inflammation and SLOAD. The aim of this article is to present contemporary evidence of this association.
Collapse
Affiliation(s)
- T L Cerajewska
- Clinical Trials Group, School of Oral and Dental Science, Lower Maudlin Street, University of Bristol, Bristol, BS1 2LY
| | - M Davies
- Clinical Trials Group, School of Oral and Dental Science, Lower Maudlin Street, University of Bristol, Bristol, BS1 2LY
| | - N X West
- Clinical Trials Group, School of Oral and Dental Science, Lower Maudlin Street, University of Bristol, Bristol, BS1 2LY
| |
Collapse
|
21
|
Hughes RE, Nikolic K, Ramsay RR. One for All? Hitting Multiple Alzheimer's Disease Targets with One Drug. Front Neurosci 2016; 10:177. [PMID: 27199640 PMCID: PMC4842778 DOI: 10.3389/fnins.2016.00177] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 04/06/2016] [Indexed: 12/12/2022] Open
Abstract
HIGHLIGHTS Many AD target combinations are being explored for multi-target drug design.New databases and models increase the potential of computational drug designLiraglutide and other antidiabetics are strong candidates for repurposing to AD.Donecopride a dual 5-HT/AChE inhibitor shows promise in pre-clinical studies Alzheimer's Disease is a complex and multifactorial disease for which the mechanism is still not fully understood. As new insights into disease progression are discovered, new drugs must be designed to target those aspects of the disease that cause neuronal damage rather than just the symptoms currently addressed by single target drugs. It is becoming possible to target several aspects of the disease pathology at once using multi-target drugs (MTDs). Intended as an introduction for non-experts, this review describes the key MTD design approaches, namely structure-based, in silico, and data-mining, to evaluate what is preventing compounds progressing through the clinic to the market. Repurposing current drugs using their off-target effects reduces the cost of development, time to launch, and the uncertainty associated with safety and pharmacokinetics. The most promising drugs currently being investigated for repurposing to Alzheimer's Disease are rasagiline, originally developed for the treatment of Parkinson's Disease, and liraglutide, an antidiabetic. Rational drug design can combine pharmacophores of multiple drugs, systematically change functional groups, and rank them by virtual screening. Hits confirmed experimentally are rationally modified to generate an effective multi-potent lead compound. Examples from this approach are ASS234 with properties similar to rasagiline, and donecopride, a hybrid of an acetylcholinesterase inhibitor and a 5-HT4 receptor agonist with pro-cognitive effects. Exploiting these interdisciplinary approaches, public-private collaborative lead factories promise faster delivery of new drugs to the clinic.
Collapse
Affiliation(s)
- Rebecca E Hughes
- School of Biology, BMS Building, University of St Andrews St Andrews, UK
| | - Katarina Nikolic
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Belgrade Belgrade, Serbia
| | - Rona R Ramsay
- School of Biology, BMS Building, University of St Andrews St Andrews, UK
| |
Collapse
|
22
|
Lee HM, Kim Y. Drug Repurposing Is a New Opportunity for Developing Drugs against Neuropsychiatric Disorders. SCHIZOPHRENIA RESEARCH AND TREATMENT 2016; 2016:6378137. [PMID: 27073698 PMCID: PMC4814692 DOI: 10.1155/2016/6378137] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/24/2016] [Indexed: 01/03/2023]
Abstract
Better the drugs you know than the drugs you do not know. Drug repurposing is a promising, fast, and cost effective method that can overcome traditional de novo drug discovery and development challenges of targeting neuropsychiatric and other disorders. Drug discovery and development targeting neuropsychiatric disorders are complicated because of the limitations in understanding pathophysiological phenomena. In addition, traditional de novo drug discovery and development are risky, expensive, and time-consuming processes. One alternative approach, drug repurposing, has emerged taking advantage of off-target effects of the existing drugs. In order to identify new opportunities for the existing drugs, it is essential for us to understand the mechanisms of action of drugs, both biologically and pharmacologically. By doing this, drug repurposing would be a more effective method to develop drugs against neuropsychiatric and other disorders. Here, we review the difficulties in drug discovery and development in neuropsychiatric disorders and the extent and perspectives of drug repurposing.
Collapse
Affiliation(s)
- Hyeong-Min Lee
- Department of Cell Biology & Physiology, School of Medicine, University of North Carolina, 115 Mason Farm Road, Chapel Hill, NC 27599, USA
| | - Yuna Kim
- Department of Pediatrics, School of Medicine, Duke University, 905 S. LaSalle Street, Durham, NC 27710, USA
| |
Collapse
|
23
|
Joshi P, Chia S, Habchi J, Knowles TPJ, Dobson CM, Vendruscolo M. A Fragment-Based Method of Creating Small-Molecule Libraries to Target the Aggregation of Intrinsically Disordered Proteins. ACS COMBINATORIAL SCIENCE 2016; 18:144-53. [PMID: 26923286 DOI: 10.1021/acscombsci.5b00129] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The aggregation process of intrinsically disordered proteins (IDPs) has been associated with a wide range of neurodegenerative disorders, including Alzheimer's and Parkinson's diseases. Currently, however, no drug in clinical use targets IDP aggregation. To facilitate drug discovery programs in this important and challenging area, we describe a fragment-based approach of generating small-molecule libraries that target specific IDPs. The method is based on the use of molecular fragments extracted from compounds reported in the literature to inhibit of the aggregation of IDPs. These fragments are used to screen existing large generic libraries of small molecules to form smaller libraries specific for given IDPs. We illustrate this approach by describing three distinct small-molecule libraries to target, Aβ, tau, and α-synuclein, which are three IDPs implicated in Alzheimer's and Parkinson's diseases. The strategy described here offers novel opportunities for the identification of effective molecular scaffolds for drug discovery for neurodegenerative disorders and to provide insights into the mechanism of small-molecule binding to IDPs.
Collapse
Affiliation(s)
- Priyanka Joshi
- Department of Chemistry, University of Cambridge , Cambridge CB2 1EW, United Kingdom
| | - Sean Chia
- Department of Chemistry, University of Cambridge , Cambridge CB2 1EW, United Kingdom
| | - Johnny Habchi
- Department of Chemistry, University of Cambridge , Cambridge CB2 1EW, United Kingdom
| | - Tuomas P J Knowles
- Department of Chemistry, University of Cambridge , Cambridge CB2 1EW, United Kingdom
| | - Christopher M Dobson
- Department of Chemistry, University of Cambridge , Cambridge CB2 1EW, United Kingdom
| | - Michele Vendruscolo
- Department of Chemistry, University of Cambridge , Cambridge CB2 1EW, United Kingdom
| |
Collapse
|
24
|
Habchi J, Arosio P, Perni M, Costa AR, Yagi-Utsumi M, Joshi P, Chia S, Cohen SIA, Müller MBD, Linse S, Nollen EAA, Dobson CM, Knowles TPJ, Vendruscolo M. An anticancer drug suppresses the primary nucleation reaction that initiates the production of the toxic Aβ42 aggregates linked with Alzheimer's disease. SCIENCE ADVANCES 2016; 2:e1501244. [PMID: 26933687 PMCID: PMC4758743 DOI: 10.1126/sciadv.1501244] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 12/02/2015] [Indexed: 05/02/2023]
Abstract
The conversion of the β-amyloid (Aβ) peptide into pathogenic aggregates is linked to the onset and progression of Alzheimer's disease. Although this observation has prompted an extensive search for therapeutic agents to modulate the concentration of Aβ or inhibit its aggregation, all clinical trials with these objectives have so far failed, at least in part because of a lack of understanding of the molecular mechanisms underlying the process of aggregation and its inhibition. To address this problem, we describe a chemical kinetics approach for rational drug discovery, in which the effects of small molecules on the rates of specific microscopic steps in the self-assembly of Aβ42, the most aggregation-prone variant of Aβ, are analyzed quantitatively. By applying this approach, we report that bexarotene, an anticancer drug approved by the U.S. Food and Drug Administration, selectively targets the primary nucleation step in Aβ42 aggregation, delays the formation of toxic species in neuroblastoma cells, and completely suppresses Aβ42 deposition and its consequences in a Caenorhabditis elegans model of Aβ42-mediated toxicity. These results suggest that the prevention of the primary nucleation of Aβ42 by compounds such as bexarotene could potentially reduce the risk of onset of Alzheimer's disease and, more generally, that our strategy provides a general framework for the rational identification of a range of candidate drugs directed against neurodegenerative disorders.
Collapse
Affiliation(s)
- Johnny Habchi
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Paolo Arosio
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Michele Perni
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Ana Rita Costa
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Maho Yagi-Utsumi
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Priyanka Joshi
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | - Sean Chia
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
| | | | - Martin B. D. Müller
- University of Groningen, University Medical Centre Groningen, European Research Institute for the Biology of Aging, 9700 AD Groningen, Netherlands
| | - Sara Linse
- Department of Biochemistry and Structural Biology, Center for Molecular Protein Science, Lund University, PO Box 124, 221 00 Lund, Sweden
| | - Ellen A. A. Nollen
- University of Groningen, University Medical Centre Groningen, European Research Institute for the Biology of Aging, 9700 AD Groningen, Netherlands
| | - Christopher M. Dobson
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- Corresponding author. E-mail: (M.V.); (T.P.J.K.); (C.M.D.)
| | - Tuomas P. J. Knowles
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- Corresponding author. E-mail: (M.V.); (T.P.J.K.); (C.M.D.)
| | - Michele Vendruscolo
- Department of Chemistry, University of Cambridge, Cambridge CB2 1EW, UK
- Corresponding author. E-mail: (M.V.); (T.P.J.K.); (C.M.D.)
| |
Collapse
|
25
|
Wang WY, Tan MS, Yu JT, Tan L. Role of pro-inflammatory cytokines released from microglia in Alzheimer's disease. ANNALS OF TRANSLATIONAL MEDICINE 2015. [PMID: 26207229 DOI: 10.3978/j.issn.2305-5839.2015.03.49] [Citation(s) in RCA: 477] [Impact Index Per Article: 53.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder of the brain, which is characterized by the formation of extracellular amyloid plaques (or senile plaques) and intracellular neurofibrillary tangles. However, increasing evidences demonstrated that neuroinflammatory changes, including chronic microgliosis are key pathological components of AD. Microglia, the resident immune cells of the brain, is constantly survey the microenvironment under physiological conditions. In AD, deposition of β-amyliod (Aβ) peptide initiates a spectrum of cerebral neuroinflammation mediated by activating microglia. Activated microglia may play a potentially detrimental role by eliciting the expression of pro-inflammatory cytokines such as interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α) influencing the surrounding brain tissue. Emerging studies have demonstrated that up-regulation of pro-inflammatory cytokines play multiple roles in both neurodegeneration and neuroprotection. Understanding the pro-inflammatory cytokines signaling pathways involved in the regulation of AD is crucial to the development of strategies for therapy. This review will discuss the mechanisms and important role of pro-inflammatory cytokines in the pathogenesis of AD, and the ongoing drug targeting pro-inflammatory cytokine for therapeutic modulation.
Collapse
Affiliation(s)
- Wen-Ying Wang
- 1 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China ; 2 Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Meng-Shan Tan
- 1 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China ; 2 Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Jin-Tai Yu
- 1 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China ; 2 Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing 210006, China
| | - Lan Tan
- 1 Department of Neurology, Qingdao Municipal Hospital, School of Medicine, Qingdao University, Qingdao 266071, China ; 2 Department of Neurology, Qingdao Municipal Hospital, College of Medicine and Pharmaceutics, Ocean University of China, Qingdao 266071, China ; 3 Department of Neurology, Qingdao Municipal Hospital, Nanjing Medical University, Nanjing 210006, China
| |
Collapse
|
26
|
Guibinga GH. MicroRNAs: tools of mechanistic insights and biological therapeutics discovery for the rare neurogenetic syndrome Lesch-Nyhan disease (LND). ADVANCES IN GENETICS 2015; 90:103-131. [PMID: 26296934 DOI: 10.1016/bs.adgen.2015.06.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
MicroRNAs (miRNAs) are small regulatory RNAs that modulate the translation of mRNA. They have emerged over the past few years as indispensable entities in the transcriptional regulation of genes. Their discovery has added additional layers of complexity to regulatory networks that control cellular homeostasis. Also, their dysregulated pattern of expression is now well demonstrated in myriad diseases and pathogenic processes. In the current review, we highlight the role of miRNAs in Lesch-Nyhan disease (LND), a rare neurogenetic syndrome caused by mutations in the purine metabolic gene encoding the hypoxanthine-guanine phosphoribosyltransferase (HPRT) enzyme. We describe how experimental and biocomputational approaches have helped to unravel genetic and signaling pathways that provide mechanistic understanding of some of the molecular and cellular basis of this ill-defined neurogenetic disorder. Through miRNA-based target predictions, we have identified signaling pathways that may be of significance in guiding biological therapeutic discovery for this incurable neurological disorder. We also propose a model to explain how a gene such as HPRT, mostly known for its housekeeping metabolic functions, can have pleiotropic effects on disparate genes and signal transduction pathways. Our hypothetical model suggests that HPRT mRNA transcripts may be acting as competitive endogenous RNAs (ceRNAs) intertwined in multiregulatory cross talk between key neural transcripts and miRNAs. Overall, this approach of using miRNA-based genomic approaches to elucidate the molecular and cellular basis of LND and guide biological target identification might be applicable to other ill-defined rare inborn-error metabolic diseases.
Collapse
Affiliation(s)
- Ghiabe-Henri Guibinga
- Division of Genetics, Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
27
|
Baek MC, Jung B, Kang H, Lee HS, Bae JS. Novel insight into drug repositioning: Methylthiouracil as a case in point. Pharmacol Res 2015; 99:185-93. [PMID: 26117428 DOI: 10.1016/j.phrs.2015.06.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/12/2015] [Accepted: 06/12/2015] [Indexed: 12/20/2022]
Abstract
Drug repositioning refers to the development of existing drugs for new indications. These drugs may have (I) failed to show efficacy in late stage clinical trials without safety issues; (II) stalled in the development for commercial reasons; (III) passed the point of patent expiry; or (IV) are being explored in new geographic markets. Over the past decade, pressure on the pharmaceutical industry caused by the 'innovation gap' owing to rising development costs and stagnant product output have become major reasons for the growing interest in drug repositioning. Companies that offer a variety of broad platforms for identifying new indications have emerged; some have been successful in building their own pipelines of candidates with reduced risks and timelines associated with further clinical development. The business models and platforms offered by these companies will be validated if they are able to generate positive proof-of-concept clinical data for their repositioned compounds. This review describes the strategy of biomarker-guided repositioning of chemotherapeutic drugs for inflammation therapy, considering the repositioning of methylthiouracil (MTU), an antithyroid drug, as a potential anti-inflammatory reagent.
Collapse
Affiliation(s)
- Moon-Chang Baek
- Department of Molecular Medicine, CMRI, School of Medicine, Kyungpook National University, Daegu 700-422, Republic of Korea
| | - Byeongjin Jung
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Hyejin Kang
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Hyun-Shik Lee
- ABRC, CMRI, School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 702-701, Republic of Korea
| | - Jong-Sup Bae
- College of Pharmacy, CMRI, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 702-701, Republic of Korea.
| |
Collapse
|
28
|
Jamal S, Goyal S, Shanker A, Grover A. Checking the STEP-Associated Trafficking and Internalization of Glutamate Receptors for Reduced Cognitive Deficits: A Machine Learning Approach-Based Cheminformatics Study and Its Application for Drug Repurposing. PLoS One 2015; 10:e0129370. [PMID: 26066505 PMCID: PMC4466797 DOI: 10.1371/journal.pone.0129370] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 05/07/2015] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Alzheimer's disease, a lethal neurodegenerative disorder that leads to progressive memory loss, is the most common form of dementia. Owing to the complexity of the disease, its root cause still remains unclear. The existing anti-Alzheimer's drugs are unable to cure the disease while the current therapeutic options have provided only limited help in restoring moderate memory and remain ineffective at restricting the disease's progression. The striatal-enriched protein tyrosine phosphatase (STEP) has been shown to be involved in the internalization of the receptor, N-methyl D-aspartate (NMDR) and thus is associated with the disease. The present study was performed using machine learning algorithms, docking protocol and molecular dynamics (MD) simulations to develop STEP inhibitors, which could be novel anti-Alzheimer's molecules. METHODS The present study deals with the generation of computational predictive models based on chemical descriptors of compounds using machine learning approaches followed by substructure fragment analysis. To perform this analysis, the 2D molecular descriptors were generated and machine learning algorithms (Naïve Bayes, Random Forest and Sequential Minimization Optimization) were utilized. The binding mechanisms and the molecular interactions between the predicted active compounds and the target protein were modelled using docking methods. Further, the stability of the protein-ligand complex was evaluated using MD simulation studies. The substructure fragment analysis was performed using Substructure fingerprint (SubFp), which was further explored using a predefined dictionary. RESULTS The present study demonstrates that the computational methodology used can be employed to examine the biological activities of small molecules and prioritize them for experimental screening. Large unscreened chemical libraries can be screened to identify potential novel hits and accelerate the drug discovery process. Additionally, the chemical libraries can be searched for significant substructure patterns as reported in the present study, thus possibly contributing to the activity of these molecules.
Collapse
Affiliation(s)
- Salma Jamal
- Department of Bioscience and Biotechnology, Banasthali University, Tonk, Rajasthan, India
| | - Sukriti Goyal
- Department of Bioscience and Biotechnology, Banasthali University, Tonk, Rajasthan, India
| | - Asheesh Shanker
- Department of Bioscience and Biotechnology, Banasthali University, Tonk, Rajasthan, India
| | - Abhinav Grover
- School of Biotechnology, Jawaharlal Nehru University, New Delhi, India
| |
Collapse
|
29
|
Patel H, Lucas X, Bendik I, Günther S, Merfort I. Target Fishing by Cross-Docking to Explain Polypharmacological Effects. ChemMedChem 2015; 10:1209-17. [DOI: 10.1002/cmdc.201500123] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 04/29/2015] [Indexed: 01/18/2023]
|
30
|
Predicting New Target Conditions for Drug Retesting Using Temporal Patterns in Clinical Trials: A Proof of Concept. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2015; 2015:445-9. [PMID: 26306283 PMCID: PMC4525223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Drug discovery is costly and time-consuming. Efficient drug repurposing promises to accelerate drug discovery with reduced cost. However, most successful repurposing cases so far have been achieved by serendipity. There is a need for more efficient computational methods for predicting new indications for existing drugs. This paper conducts a retrospective analysis of the temporal patterns of drug intervention trials for every drug in a pair of different conditions in ClinicalTrials.gov, including 550 drugs used for 451 conditions between 2003 and 2013. We found that drugs are often targeted towards conditions that are related by similar or identical eligibility criteria. We demonstrated the preliminary feasibility of predicting new target conditions for drug retesting among conditions with similar aggregated clinical trial eligibility criteria and confirmed this hypothesis using evidence from the literature.
Collapse
|
31
|
Identification of Nitazoxanide as a Group I Metabotropic Glutamate Receptor Negative Modulator for the Treatment of Neuropathic Pain: An In Silico Drug Repositioning Study. Pharm Res 2015; 32:2798-807. [PMID: 25762088 DOI: 10.1007/s11095-015-1665-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 03/02/2015] [Indexed: 10/23/2022]
Abstract
PURPOSE Drug repositioning strategies were employed to explore new therapeutic indications for existing drugs that may exhibit dual negative mGluR1/5 modulating activities as potential treatments for neuropathic pain. METHOD A customized in silico-in vitro-in vivo drug repositioning scheme was assembled and implemented to search available drug libraries for compounds with dual mGluR1/5 antagonistic activities, that were then evaluated using in vitro functional assays and, for validated hits, in an established animal model for neuropathic pain. RESULTS Tizoxanide, the primary active metabolite of the FDA approved drug nitazoxanide, fit in silico pharmacophore models constructed for both mGluR1 and mGluR5. Subsequent calcium (Ca++) mobilization functional assays confirmed that tizoxanide exhibited appreciable antagonist activity for both mGluR1 and mGluR5 (IC50 = 1.8 μM and 1.2 μM, respectively). The in vivo efficacy of nitazoxanide administered by intraperitoneal injection was demonstrated in a rat model for neuropathic pain. CONCLUSION The major aim of the present study was to demonstrate the utility of an in silico-in vitro-in vivo drug repositioning protocol to facilitate the repurposing of approved drugs for new therapeutic indications. As an example, this particular investigation successfully identified nitazoxanide and its metabolite tizoxanide as dual mGluR1/5 negative modulators. A key finding is the vital importance for drug screening libraries to include the structures of drug active metabolites, such as those emanating from prodrugs which are estimated to represent 5-7% of marketed drugs.
Collapse
|
32
|
Anighoro A, Bajorath J, Rastelli G. Polypharmacology: challenges and opportunities in drug discovery. J Med Chem 2014; 57:7874-87. [PMID: 24946140 DOI: 10.1021/jm5006463] [Citation(s) in RCA: 710] [Impact Index Per Article: 71.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
At present, the legendary magic bullet, i.e., a drug with high potency and selectivity toward a specific biological target, shares the spotlight with an emerging and alternative polypharmacology approach. Polypharmacology suggests that more effective drugs can be developed by specifically modulating multiple targets. It is generally thought that complex diseases such as cancer and central nervous system diseases may require complex therapeutic approaches. In this respect, a drug that "hits" multiple sensitive nodes belonging to a network of interacting targets offers the potential for higher efficacy and may limit drawbacks generally arising from the use of a single-target drug or a combination of multiple drugs. In this review, we will compare advantages and disadvantages of multitarget versus combination therapies, discuss potential drug promiscuity arising from off-target effects, comment on drug repurposing, and introduce approaches to the computational design of multitarget drugs.
Collapse
Affiliation(s)
- Andrew Anighoro
- Life Sciences Department, University of Modena and Reggio Emilia , Via Campi 183, 41125 Modena, Italy
| | | | | |
Collapse
|