1
|
Di Paolo ML, Salerno S, Nordio G, Piazzola F, Sarno S, Sarno G, Natale B, Poggetti V, Borreca A, Baglini E, Barresi E, Da Settimo F, Cosconati S, Castellano S, Taliani S, Dalla Via L. 2-(Phenylamino)-7,8-dihydroquinazolin-5(6H)-one, a promising scaffold for MAO-B inhibitors with potential GSK3β targeting. Eur J Med Chem 2025; 291:117580. [PMID: 40186896 DOI: 10.1016/j.ejmech.2025.117580] [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: 01/31/2025] [Revised: 03/19/2025] [Accepted: 03/28/2025] [Indexed: 04/07/2025]
Abstract
Neurodegenerative disorders, such as Parkinson's disease and Alzheimer's disease, constitute pathological conditions of great relevance on health span and quality of life. The identification of novel therapeutic options, able to modulate the processes involved in the insurgence and progression of neurodegenerative disorders, represents an intriguing challenge of current research. Herein, a library of 36-membered 2-(phenylamino)-7,8-dihydroquinazolinone derivatives was synthesized and biologically evaluated as human MAO inhibitors. Some compounds able to inhibit MAO-B potently and selectively (Ki in the nanomolar range) were identified, and robust structure-activity relationships were drawn, supported by computational studies. Further biological assays revealed a safe profile for all derivatives and, for compounds selected as the best MAO-B inhibitors (4, 5, 13, 14) the following properties also emerged: (i) the ability to inhibit MAO-B activity in whole cells, with an effectiveness comparable or slight lower with respect to the reference safinamide; (ii) physicochemical parameters suggesting drug-likeness properties; (iii) the ability to inhibit, albeit weakly, GSK3β kinase (for compound 4). Within the whole series, compound 4 stood out as a promising lead for future optimization campaigns aimed to obtain useful drugs for the treatment of Alzheimer's and Parkinson's diseases.
Collapse
Affiliation(s)
| | - Silvia Salerno
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy.
| | - Giulia Nordio
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy.
| | - Francesco Piazzola
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy.
| | - Stefania Sarno
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy.
| | - Giuliana Sarno
- Department of Pharmacy, University of Salerno, Fisciano, SA, 84084, Italy.
| | - Benito Natale
- DiSTABiF, University of Campania Luigi Vanvitelli, 81100, Caserta, Italy.
| | | | - Antonella Borreca
- Institute of Neuroscience (IN-CNR), Consiglio Nazionale delle Ricerche, Milan, Italy; IRCCS Humanitas Research Hospital, Via Manzoni 56, Rozzano, Milan, 20089, Italy.
| | - Emma Baglini
- Institute of Clinical Physiology, National Research Council of Italy, CNR Research Area, 56124, Pisa, Italy.
| | | | | | - Sandro Cosconati
- DiSTABiF, University of Campania Luigi Vanvitelli, 81100, Caserta, Italy.
| | - Sabrina Castellano
- Department of Pharmacy, University of Salerno, Fisciano, SA, 84084, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, 56126, Pisa, Italy.
| | - Lisa Dalla Via
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131, Padova, Italy.
| |
Collapse
|
2
|
Lemke J, Mengers N, Schmidt L, Schulig L, König S, Rosendahl P, Bartz FM, Garscha U, Bednarski PJ, Link A. Lead Optimization of Positive Allosteric K V7.2/3 Channel Modulators toward Improved Balance of Lipophilicity and Aqueous Solubility. J Med Chem 2025; 68:8377-8399. [PMID: 40198203 DOI: 10.1021/acs.jmedchem.4c03112] [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: 04/10/2025]
Abstract
The voltage-gated potassium channel KV7.2/3 is gaining attention for its association with several medical indications. While recently reported, potent compounds aimed to fill the therapeutic gap left by market-withdrawn activators, key physicochemical parameters did not meet the requirements of potential drug candidates. Targeting the membrane-located channel requires subtly balancing lipophilicity, activity, and aqueous solubility. This publication describes the lead optimization of a highly active compound toward optimized physicochemical parameters. Out of 42 newly synthesized compounds, 30 showed activity on KV7.2/3 channels, and 15 had also an increased solubility compared the to hit compound. The integration of a three-dimensional bulky structure and the probable onset of chameleonic behavior, led to a 20-fold solubility increase (S = 21.7 vs 1.1 μM) and only slightly reduced potency (pEC50 = 7.42 vs 7.96) for the lead. Additionally, the target engagement of the compound was theoretically enhanced by a reduction of membrane retention.
Collapse
Affiliation(s)
- Jana Lemke
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Nadine Mengers
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Louis Schmidt
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Lukas Schulig
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Stefanie König
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Pascal Rosendahl
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Frieda-Marie Bartz
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Ulrike Garscha
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Patrick J Bednarski
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| | - Andreas Link
- Institute of Pharmacy, University of Greifswald, Friedrich-Ludwig-Jahn-Str. 17, Greifswald 17489, Germany
| |
Collapse
|
3
|
Poggialini F, Governa P, Vagaggini C, Maramai S, Lamponi S, Mugnaini C, Brizzi A, Purgatorio R, de Candia M, Catto M, Dreassi E, Manetti F, Corelli F, Altomare CD, Cappelli A, Paolino M. Light-mediated activation/deactivation control and in vitro ADME-Tox profiling of a donepezil-like Dual AChE/MAO-B Inhibitor. Eur J Pharm Sci 2025; 209:107066. [PMID: 40064401 DOI: 10.1016/j.ejps.2025.107066] [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: 01/07/2025] [Revised: 02/25/2025] [Accepted: 03/07/2025] [Indexed: 03/21/2025]
Abstract
The possibility to control the effects of drugs in time and space represents an ideal condition for developing safer and more personalized therapies against different disorders. In this context, photopharmacology has paved the way for the use of light in the modulation of drugs activity. Our interest is directed to photo-switchable molecules, capable of interconverting between two different isoforms upon light irradiation. We recently reported 1, a donepezil-like compound based on 2-benzylidenindan-1-one structure, as a dual AChE and MAO-B inhibitor, which can be converted into the E- (active form) and Z- (about tenfold less active form) diastereoisomers by irradiating with UV-vis light. Aiming at identifying compounds with remarkable activity in physiological conditions, we herein report a fine characterization of 1 in PBS solutions. First, we evaluated its ability to act as a photoswitch comparing PBS solution with organic solvents (e.g. methanol), demonstrating that a wavelength in the UV range (330 nm) can convert the E- into the Z-diastereoisomer, while the use of a visible light (400 nm) allows the interconversion from Z to E in both media. Along with its photoinducible behavior, we investigated the passive diffusion across cellular membrane with PAMPA experiments, plasma and microsomal stability, and binding to plasma proteins. Interestingly, the results of such studies suggested that 1 could persist in the blood circulation for a long time, which is desirable for application in photopharmacological therapies. Cytotoxicity studies highlighted the potential of our prototypic compound as a lead photodrug against neurodegenerative disorders, deserving to advance in molecular optimization studies and further in vitro and in vivo characterization.
Collapse
Affiliation(s)
- Federica Poggialini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Paolo Governa
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Chiara Vagaggini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Samuele Maramai
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Stefania Lamponi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Claudia Mugnaini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Antonella Brizzi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Rosa Purgatorio
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Modesto de Candia
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Marco Catto
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Fabrizio Manetti
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Federico Corelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Cosimo Damiano Altomare
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari Aldo Moro, Via E. Orabona 4, 70125, Bari, Italy
| | - Andrea Cappelli
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy
| | - Marco Paolino
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via A. Moro 2, I-53100, Siena, Italy.
| |
Collapse
|
4
|
Rodrigues B, Ventura E, Moreira P, Resende R, Bicker J, Santos AE, Pereira CF, Cruz MT, Robalo MP, Silva A, Silva S. New low-dose curcumin derivative with therapeutic potential in Alzheimer's disease: Results from an in vitro and in vivo study in mice. Neurobiol Aging 2025; 147:105-123. [PMID: 39733760 DOI: 10.1016/j.neurobiolaging.2024.12.005] [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: 04/16/2024] [Revised: 12/08/2024] [Accepted: 12/17/2024] [Indexed: 12/31/2024]
Abstract
Curcumin has been proposed as a potential treatment for Alzheimer's disease (AD) due to its ability to inhibit amyloid-β (Aβ) peptide aggregates and to destabilise pre-formed ones. Derivative 27 was synthesized to improve low-dose efficacy in the context of AD. Its anti-inflammatory, antioxidant and anti-amyloidogenic activities were evaluated in chemico, in vitro using AD and neuroinflammation cell models, and in vivo using the double-transgenic APP/PS1 mice. In vitro, this curcumin derivative significantly reduced nitric oxide (NO) production and levels of pro-inflammatory proteins, inducible NO synthase, pro-interleukin-1β (Pro-IL-1β) and cyclooxygenase-2. Furthermore, Derivative 27 activated nuclear factor erythroid 2-related factor 2 transcription factor (Nrf2) and significantly increased Nrf2 and heme-oxygenase-1 protein levels in the nucleus and in the cytoplasm, respectively. In one-year-old APP/PS1 mice, orally administered-Derivative 27 (50 mg/Kg/day) for 28 days improved spatial short-term memory and significantly decreased hippocampal Pro-IL-1β and amyloid precursor protein levels, as well as Aβ levels in the hippocampus and plasma. This study supports developing new chemical approaches to alter curcumin molecule, enabling lower doses, while increasing the effectiveness in AD treatment.
Collapse
Affiliation(s)
- Beatriz Rodrigues
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Centro Clínico e Académico de Coimbra, Coimbra, Portugal.
| | - Eduarda Ventura
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Centro Clínico e Académico de Coimbra, Coimbra, Portugal
| | - Patrícia Moreira
- Centro Clínico e Académico de Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Rosa Resende
- Centro Clínico e Académico de Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Joana Bicker
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Centro Clínico e Académico de Coimbra, Coimbra, Portugal; CIBIT/ICNAS, Coimbra Institute for Biomedical Imaging and Translation Research, University of Coimbra, Coimbra, Portugal
| | - Armanda E Santos
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Centro Clínico e Académico de Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Cláudia Fragão Pereira
- Centro Clínico e Académico de Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria Teresa Cruz
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Centro Clínico e Académico de Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Maria Paula Robalo
- CQE, Institute of Molecular Sciences, Instituto Superior Técnico, University of Lisbon, Lisbon, Portugal; Instituto Superior de Engenharia de Lisboa (ISEL), Instituto Politécnico de Lisboa, Lisbon, Portugal
| | - Ana Silva
- Centro Clínico e Académico de Coimbra, Coimbra, Portugal; Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Sónia Silva
- Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal; Centro Clínico e Académico de Coimbra, Coimbra, Portugal; Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal; Coimbra Institute of Clinical and Biomedical Research, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
5
|
Poggetti V, Angeloni E, Germelli L, Natale B, Waqas M, Sarno G, Angeli A, Daniele S, Salerno S, Barresi E, Cosconati S, Castellano S, Da Pozzo E, Costa B, Supuran CT, Da Settimo F, Taliani S. Discovery of the First-in-Class Dual TSPO/Carbonic Anhydrase Modulators with Promising Neurotrophic Activity. ACS Chem Neurosci 2025; 16:1-15. [PMID: 39545683 DOI: 10.1021/acschemneuro.4c00477] [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] [Indexed: 11/17/2024] Open
Abstract
In searching for putative new therapeutic strategies to treat neurodegenerative diseases, the mitochondrial 18 kDa translocator protein (TSPO) and cerebral isoforms of carbonic anhydrase (CA) were exploited as potential targets. Based on the structures of a class of highly affine and selective TSPO ligands and a class of CA activators, both developed by us in recent years, a small library of 2-phenylindole-based dual TSPO/CA modulators was developed, able to bind TSPO and activate CA VII in the low micromolar/submicromolar range. The interaction with the two targets was corroborated by computational studies. Biological investigation on human microglia C20 cells identified derivative 3 as a promising lead compound worthy of future optimization due to its (i) lack of cytotoxicity, (ii) ability to stimulate TSPO steroidogenic function and activate CA VII, and (iii) ability to effectively upregulate gene expression of the brain-derived neurotrophic factor.
Collapse
Affiliation(s)
- Valeria Poggetti
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Elisa Angeloni
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Lorenzo Germelli
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Benito Natale
- DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi, 43, 81100 Caserta, Italy
| | - Muhammad Waqas
- DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi, 43, 81100 Caserta, Italy
| | - Giuliana Sarno
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| | - Andrea Angeli
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, Polo Scientifico, University of Florence, Via U. Schiff, 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Simona Daniele
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Silvia Salerno
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Elisabetta Barresi
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Sandro Cosconati
- DiSTABiF, University of Campania Luigi Vanvitelli, Via Vivaldi, 43, 81100 Caserta, Italy
| | - Sabrina Castellano
- Department of Pharmacy, University of Salerno, Via Giovanni Paolo II, 132, 84084 Fisciano, SA, Italy
| | - Eleonora Da Pozzo
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Barbara Costa
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, Polo Scientifico, University of Florence, Via U. Schiff, 6, Sesto Fiorentino, 50019 Firenze, Italy
| | - Federico Da Settimo
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| | - Sabrina Taliani
- Department of Pharmacy, University of Pisa, Via Bonanno, 6, 56126 Pisa, Italy
| |
Collapse
|
6
|
Alvarez-Figueroa MJ, Nuñez-Navarro F, Recabarren-Gajardo G, González-Aramundiz JV. Design of an innovative nanovehicle to enhance brain permeability of a novel 5-HT6 receptor antagonist. Int J Pharm X 2024; 8:100296. [PMID: 39525530 PMCID: PMC11550157 DOI: 10.1016/j.ijpx.2024.100296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/02/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
An innovative nanovehicle based on lipid nanocapsules (LNC) was designed to facilitate the passage of a new 5-HT6 receptor antagonist, namely PUC-10, through the blood-brain barrier. PUC-10 is a new synthetic N-arylsulfonylindole that has demonstrated potent 5-HT6 receptor antagonist activity, but it exhibits poor solubility in water, which indicates limited absorption. The lipid nanocapsules designed had a nanometric size (53 nm), a monomodal distribution (PI<0.2), a negative Z potential (-17 ± 7 mV) and allowed efficient PUC-10 encapsulation (74 %). Furthermore, the LNC demonstrated to be stable for at least 4 weeks at 4 °C (storage conditions), for at least 4 h in DMEM at pH 7.4, and for 18 h in water with 5 % DMSO, with both latter conditions maintained at 37 °C. They also demonstrated that cell viability was not affected at the different concentrations studied. Finally, in vitro studies that simulate the blood brain barrier (PAMPA-BBB) demonstrated that the nanoencapsulation of PUC-10 promoted their penetration through the blood-brain barrier, with a calculated permeability of 1.3 × 10-8 cm/s, compared to the null permeability exhibited by non-nanoencapsulated PUC-10.
Collapse
Affiliation(s)
| | - Francisco Nuñez-Navarro
- Departamento de Farmacia, Facultad de Química, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Gonzalo Recabarren-Gajardo
- Departamento de Farmacia, Facultad de Química, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro Interdisciplinario de Neurociencias, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José Vicente González-Aramundiz
- Departamento de Farmacia, Facultad de Química, Pontificia Universidad Católica de Chile, Santiago, Chile
- Centro de Investigación en Nanotecnología y Materiales Avanzados "CIEN-UC ", Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
7
|
Moreira P, Macedo J, Matos P, Bicker J, Fortuna A, Figueirinha A, Salgueiro L, Batista MT, Silva A, Silva S, Resende R, Branco PC, Cruz MT, Pereira CF. Effect of bioactive extracts from Eucalyptus globulus leaves in experimental models of Alzheimer's disease. Biomed Pharmacother 2024; 181:117652. [PMID: 39486370 DOI: 10.1016/j.biopha.2024.117652] [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: 08/01/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Current therapies for Alzheimer's disease (AD) do not delay its progression, therefore, novel disease-modifying strategies are urgently needed. Recently, an increasing number of compounds from natural origin with protective properties against AD have been identified. Mixtures or extracts obtained from natural products containing several bioactive compounds have multifunctional properties and have drawn the attention because multiple AD pathways can be simultaneously modulated. This study evaluated the in vitro and in vivo effect of the essential oil (EO) obtained from the hydrodistillation of Eucalyptus globulus leaves, and an extract obtained from the hydrodistillation residual water (HRW). It was observed that EO and HRW have anti-inflammatory effect in brain immune cells modeling AD, namely lipopolysaccharide (LPS)- and amyloid-beta (Aβ)-stimulated microglia. In cell models that mimic AD-related neuronal dysfunction, HRW attenuated Aβ secretion and Aβ-induced mitochondrial dysfunction. Since the HRW's major components did not cross the blood-brain barrier, both EO and HRW were administered to the APP/PS1 transgenic AD mouse model by an intranasal route, which reduced cortical and hippocampal Aβ levels, and to rescue memory deficits and anxiety-like behaviors. Finally, HRW and EO were found to regulate cholesterol levels in aged mice after intranasal administration, suggesting that these extracts can reduce hypercholesterolemia and avoid risk for AD development. Overall, findings support a protective role of E. globulus extracts against AD‑like pathology and cognitive impairment highlighting the underlying mechanisms. These extracts obtained from underused forest biomass could be useful to develop nutraceutical supplements helpful to avoid AD risk and to prevent its progression.
Collapse
Affiliation(s)
- Patrícia Moreira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal.
| | - Jéssica Macedo
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Patrícia Matos
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; LAQV, REQUIMTE, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Joana Bicker
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Ana Fortuna
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Artur Figueirinha
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; LAQV, REQUIMTE, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Lígia Salgueiro
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Maria Teresa Batista
- CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Ana Silva
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Sónia Silva
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; iCBR-Coimbra Institute for Clinical and Biomedical Research, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Rosa Resende
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Pedro Costa Branco
- RAIZ-Forest and Paper Research Institute, Eixo, Aveiro 3800-783, Portugal
| | - Maria Teresa Cruz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Cláudia Fragão Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal; Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| |
Collapse
|
8
|
Maramai S, Saletti M, Paolino M, Giuliani G, Cazzola J, Spaiardi P, Talpo F, Frosini M, Pifferi A, Ballarotto M, Carotti A, Poggialini F, Vagaggini C, Dreassi E, Giorgi G, Dondio G, Cappelli A, Rosario Biella G, Anzini M. Novel multitarget directed ligands inspired by riluzole: A serendipitous synthesis of substituted benzo[b][1,4]thiazepines potentially useful as neuroprotective agents. Bioorg Med Chem 2024; 112:117872. [PMID: 39153378 DOI: 10.1016/j.bmc.2024.117872] [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: 07/22/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024]
Abstract
Riluzole, the first clinically approved treatment for amyotrophic lateral sclerosis (ALS), represents a successful example of a drug endowed with a multimodal mechanism of action. In recent years, different series of riluzole-based compounds have been reported, including several agents acting as Multi-Target-Directed Ligands (MTLDs) endowed with neuroprotective effects. Aiming at identical twin structures inspired by riluzole (2a-c), a synthetic procedure was planned, but the reactivity of the system took a different path, leading to the serendipitous isolation of benzo[b][1,4]thiazepines 3a-c and expanded intermediates N-cyano-benzo[b][1,4]thiazepines 4a-c, which were fully characterized. The newly obtained structures 3a-c, bearing riluzole key elements, were initially tested in an in vitro ischemia/reperfusion injury protocol, simulating the cerebral stroke. Results identified compound 3b as the most effective in reverting the injury caused by an ischemia-like condition, and its activity was comparable, or even higher than that of riluzole, exhibiting a concentration-dependent neuroprotective effect. Moreover, derivative 3b completely reverted the release of Lactate Dehydrogenase (LDH), lowering the values to those of the control slices. Based on its very promising pharmacological properties, compound 3b was then selected to assess its effects on voltage-dependent Na+ and K+ currents. The results indicated that derivative 3b induced a multifaceted inhibitory effect on voltage-gated currents in SH-SY5Y differentiated neurons, suggesting its possible applications in epilepsy and stroke management, other than ALS. Accordingly, brain penetration was also measured for 3b, as it represents an elegant example of a MTDL and opens the way to further ex-vivo and/or in-vivo characterization.
Collapse
Affiliation(s)
- Samuele Maramai
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Mario Saletti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Marco Paolino
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Germano Giuliani
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Jessica Cazzola
- Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", Università di Pavia, Via Adolfo Ferrata 9, 27100 Pavia, Italy
| | - Paolo Spaiardi
- Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", Università di Pavia, Via Adolfo Ferrata 9, 27100 Pavia, Italy; INFN - Sezione di Pavia, Dipartimento di Fisica, Via Agostino Bassi 6, 27100 Pavia, Italy
| | - Francesca Talpo
- Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", Università di Pavia, Via Adolfo Ferrata 9, 27100 Pavia, Italy
| | - Maria Frosini
- Dipartimento di Scienze della Vita, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Alice Pifferi
- Dipartimento di Scienze della Vita, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Marco Ballarotto
- Dipartimento di Scienze Farmaceutiche, Università di Perugia, Via Fabretti 48, 06123 Perugia, Italy
| | - Andrea Carotti
- Dipartimento di Scienze Farmaceutiche, Università di Perugia, Via Fabretti 48, 06123 Perugia, Italy
| | - Federica Poggialini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Chiara Vagaggini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Elena Dreassi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Gianluca Giorgi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Giulio Dondio
- Aphad SrL, Via della Resistenza 65, 20090 Buccinasco, Italy
| | - Andrea Cappelli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Gerardo Rosario Biella
- Dipartimento di Biologia e Biotecnologie "Lazzaro Spallanzani", Università di Pavia, Via Adolfo Ferrata 9, 27100 Pavia, Italy; INFN - Sezione di Pavia, Dipartimento di Fisica, Via Agostino Bassi 6, 27100 Pavia, Italy
| | - Maurizio Anzini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy.
| |
Collapse
|
9
|
Manhas D, Dhiman S, Kour H, Kour D, Sharma K, Wazir P, Vij B, Kumar A, Sawant SD, Ahmed Z, Nandi U. ADME/PK Insights of Crocetin: A Molecule Having an Unusual Chemical Structure with Druglike Features. ACS OMEGA 2024; 9:21494-21509. [PMID: 38764638 PMCID: PMC11097163 DOI: 10.1021/acsomega.4c02116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 03/20/2024] [Indexed: 05/21/2024]
Abstract
Crocetin is a promising phyto-based molecule to treat Alzheimer's disease (AD). The chemical structure of crocetin is incongruent with various standard structural features of CNS drugs. As poor pharmacokinetic behavior is the major hurdle for any candidate to become a drug, we elucidated its druggable characteristics by implementing in silico, in vitro, and in vivo approaches, as limited ADME/PK information is available. Results demonstrate several attributes of crocetin based on rules of drug-likeness, lipophilicity, pKa, P-gp inhibitory activity, plasma stability, RBC partitioning, metabolic stability, CYP inhibitory action, blood-brain barrier (BBB) permeability, oral bioavailability, and pharmacokinetic interaction with marketed anti-Alzheimer's drugs (memantine, donepezil, galantamine, and rivastigmine). However, aqueous solubility, chemical stability, plasma protein binding, and P-gp induction are some concerns associated with this molecule that should be taken into consideration during its further development. Overall results indicate favorable ADME/PK behavior and potential druggable candidature of crocetin.
Collapse
Affiliation(s)
- Diksha Manhas
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sumit Dhiman
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Harpreet Kour
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Natural
Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Dilpreet Kour
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Kuhu Sharma
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Priya Wazir
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
| | - Bhavna Vij
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
| | - Ajay Kumar
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sanghapal D. Sawant
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Natural
Products & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
| | - Zabeer Ahmed
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Utpal Nandi
- Pharmacology
Division, CSIR-Indian Institute of Integrative
Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
10
|
Wasielewska JM, Szostak K, McInnes LE, Quek H, Chaves JCS, Liddell JR, Koistinaho J, Oikari LE, Donnelly PS, White AR. Patient-Derived Blood-Brain Barrier Model for Screening Copper Bis(thiosemicarbazone) Complexes as Potential Therapeutics in Alzheimer's Disease. ACS Chem Neurosci 2024; 15:1432-1455. [PMID: 38477556 DOI: 10.1021/acschemneuro.3c00743] [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] [Indexed: 03/14/2024] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia characterized by a progressive cognitive decline. Addressing neuroinflammation represents a promising therapeutic avenue to treat AD; however, the development of effective antineuroinflammatory compounds is often hindered by their limited blood-brain barrier (BBB) permeability. Consequently, there is an urgent need for accurate, preclinical AD patient-specific BBB models to facilitate the early identification of immunomodulatory drugs capable of efficiently crossing the human AD BBB. This study presents a unique approach to BBB drug permeability screening as it utilizes the familial AD patient-derived induced brain endothelial-like cell (iBEC)-based model, which exhibits increased disease relevance and serves as an improved BBB drug permeability assessment tool when compared to traditionally employed in vitro models. To demonstrate its utility as a small molecule drug candidate screening platform, we investigated the effects of diacetylbis(N(4)-methylthiosemicarbazonato)copper(II) (CuII(atsm)) and a library of metal bis(thiosemicarbazone) complexes─a class of compounds exhibiting antineuroinflammatory therapeutic potential in neurodegenerative disorders. By evaluating the toxicity, cellular accumulation, and permeability of those compounds in the AD patient-derived iBEC, we have identified 3,4-hexanedione bis(N(4)-methylthiosemicarbazonato)copper(II) (CuII(dtsm)) as a candidate with good transport across the AD BBB. Furthermore, we have developed a multiplex approach where AD patient-derived iBEC were combined with immune modulators TNFα and IFNγ to establish an in vitro model representing the characteristic neuroinflammatory phenotype at the patient's BBB. Here, we observed that treatment with CuII(dtsm) not only reduced the expression of proinflammatory cytokine genes but also reversed the detrimental effects of TNFα and IFNγ on the integrity and function of the AD iBEC monolayer. This suggests a novel pathway through which copper bis(thiosemicarbazone) complexes may exert neurotherapeutic effects on AD by mitigating BBB neuroinflammation and related BBB integrity impairment. Together, the presented model provides an effective and easily scalable in vitro BBB platform for screening AD drug candidates. Its improved translational potential makes it a valuable tool for advancing the development of metal-based compounds aimed at modulating neuroinflammation in AD.
Collapse
Affiliation(s)
- Joanna M Wasielewska
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- Faculty of Medicine, University of Queensland, Herston, QLD 4006, Australia
| | - Kathryn Szostak
- School of Chemistry, Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Lachlan E McInnes
- School of Chemistry, Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Hazel Quek
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- School of Biomedical Science, University of Queensland, St. Lucia, QLD 4067, Australia
| | - Juliana C S Chaves
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Jeffrey R Liddell
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Jari Koistinaho
- Drug Research Program, Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki 00014,Finland
- Neuroscience Centre, Helsinki Institute of Life Science, University of Helsinki, Helsinki 00014, Finland
| | - Lotta E Oikari
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
| | - Paul S Donnelly
- School of Chemistry, Bio21 Institute for Molecular Science and Biotechnology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Anthony R White
- Mental Health and Neuroscience Program, QIMR Berghofer Medical Research Institute, Herston, QLD 4006, Australia
- School of Biomedical Science, University of Queensland, St. Lucia, QLD 4067, Australia
| |
Collapse
|
11
|
Cham PS, Deepika, Bhat R, Raina D, Manhas D, Kotwal P, Mindala DP, Pandey N, Ghosh A, Saran S, Nandi U, Khan IA, Singh PP. Exploring the Antibacterial Potential of Semisynthetic Phytocannabinoid: Tetrahydrocannabidiol (THCBD) as a Potential Antibacterial Agent against Sensitive and Resistant Strains of Staphylococcus aureus. ACS Infect Dis 2024; 10:64-78. [PMID: 38051636 DOI: 10.1021/acsinfecdis.3c00154] [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] [Indexed: 12/07/2023]
Abstract
Antimicrobial resistance (AMR) is one of the most challenging problems and is responsible for millions of deaths every year. We therefore urgently require new chemical entities with novel mechanisms of action. Phytocannabinoids have been adequately reported for the antimicrobial effect but not seriously pursued because of either stringent regulatory issues or poor drug-like properties. In this regard, the current work demonstrated the antibacterial potential of tetrahydrocannabidiol (THCBD, 4), a semisynthetic phytocannabinoid, against Staphylococcus aureus, the second-most widespread bug recognized by the WHO. THCBD (4) was generated from cannabidiol and subjected to extensive antibacterial screening. In in vitro studies, THCBD (4) demonstrated a potent MIC of 0.25 μg/mL against Gram-positive bacteria, S. aureus ATCC-29213. It is interesting to note that THCBD (4) has demonstrated strong effectiveness against efflux pump-overexpressing (SA-1199B, SA-K2191, SA-K2192, and Mupr-1) and multidrug-resistant (MRSA-15187) S. aureus strains. THCBD (4) has also shown a good effect in kill kinetic assays against ATCC-29213 and MRSA-15187. In the checkerboard assay, THCBD (4) has shown additive/indifference effects with several well-known clinically used antibiotics, tetracycline, mupirocin, penicillin G, and ciprofloxacin. THCBD (4) also exhibited good permeability in the artificial skin model. Most importantly, THCBD (4) has significantly reduced CFU in mice's in vivo skin infection models and also demonstrated decent plasma exposure with 16-17% oral bioavailability. Acute dermal toxicity of THCBD (4) suggests no marked treatment-related impact on gross pathophysiology. This attractive in vitro and in vivo profile of plant-based compounds opens a new direction for new-generation antibiotics and warrants further detailed investigation.
Collapse
Affiliation(s)
- Pankaj Singh Cham
- Natural Product & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Deepika
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
| | - Rahul Bhat
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Diksha Raina
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Diksha Manhas
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Pankul Kotwal
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Durga Prasad Mindala
- Natural Product & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Noopur Pandey
- Solid State Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology Mesra Ranchi, Jharkhand 835215, India
| | - Animesh Ghosh
- Solid State Pharmaceutics Research Laboratory, Department of Pharmaceutical Sciences and Technology, Birla Institute of Technology Mesra Ranchi, Jharkhand 835215, India
| | - Saurabh Saran
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Fermentation and Microbial Biotechnology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Utpal Nandi
- Pharmacology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Inshad Ali Khan
- Clinical Microbiology Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan 305817, India
| | - Parvinder Pal Singh
- Natural Product & Medicinal Chemistry Division, CSIR-Indian Institute of Integrative Medicine (CSIR-IIIM), Canal Road, Jammu 180001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
12
|
Poonsiri T, Dell’Accantera D, Loconte V, Casnati A, Cervoni L, Arcovito A, Benini S, Ferrari A, Cipolloni M, Cacioni E, De Franco F, Giacchè N, Rinaldo S, Folli C, Sansone F, Berni R, Cianci M. 3-O-Methyltolcapone and Its Lipophilic Analogues Are Potent Inhibitors of Transthyretin Amyloidogenesis with High Permeability and Low Toxicity. Int J Mol Sci 2023; 25:479. [PMID: 38203650 PMCID: PMC10779086 DOI: 10.3390/ijms25010479] [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: 12/03/2023] [Revised: 12/22/2023] [Accepted: 12/24/2023] [Indexed: 01/12/2024] Open
Abstract
Transthyretin (TTR) is an amyloidogenic homotetramer involved in the transport of thyroxine in blood and cerebrospinal fluid. To date, more than 130 TTR point mutations are known to destabilise the TTR tetramer, leading to its extracellular pathological aggregation accumulating in several organs, such as heart, peripheral and autonomic nerves, and leptomeninges. Tolcapone is an FDA-approved drug for Parkinson's disease that has been repurposed as a TTR stabiliser. We characterised 3-O-methyltolcapone and two newly synthesized lipophilic analogues, which are expected to be protected from the metabolic glucuronidation that is responsible for the lability of tolcapone in the organism. Immunoblotting assays indicated the high degree of TTR stabilisation, coupled with binding selectivity towards TTR in diluted plasma of 3-O-methyltolcapone and its lipophilic analogues. Furthermore, in vitro toxicity data showed their several-fold improved neuronal and hepatic safety compared to tolcapone. Calorimetric and structural data showed that both T4 binding sites of TTR are occupied by 3-O-methyltolcapone and its lipophilic analogs, consistent with an effective TTR tetramer stabilisation. Moreover, in vitro permeability studies showed that the three compounds can effectively cross the blood-brain barrier, which is a prerequisite for the inhibition of TTR amyloidogenesis in the cerebrospinal fluid. Our data demonstrate the relevance of 3-O-methyltolcapone and its lipophilic analogs as potent inhibitors of TTR amyloidogenesis.
Collapse
Affiliation(s)
- Thanalai Poonsiri
- Bioorganic Chemistry and Bio-Crystallography Laboratory (B2Cl), Faculty of Agricultural, Environmental and Food Sciences, Free University of Bolzano, 39100 Bolzano, Italy; (T.P.); (S.B.)
| | - Davide Dell’Accantera
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy; (D.D.); (A.C.); (F.S.); (R.B.)
| | - Valentina Loconte
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA;
- Lawrence Berkeley National Laboratory, Molecular Biophysics and Integrated Bioimaging Division, Berkeley, CA 94720, USA
| | - Alessandro Casnati
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy; (D.D.); (A.C.); (F.S.); (R.B.)
| | - Laura Cervoni
- Department of Biochemical Sciences, University of Rome “La Sapienza”, P.le Aldo Moro 5, 00185 Rome, Italy; (L.C.); (S.R.)
| | - Alessandro Arcovito
- Department of Biotechnological Sciences and Intensive Care, Catholic University of Sacred Heart, Largo F. Vito 1, 00168 Rome, Italy;
- Fondazione Policlinico Universitario A. Gemelli—IRCCS, 00168 Rome, Italy
| | - Stefano Benini
- Bioorganic Chemistry and Bio-Crystallography Laboratory (B2Cl), Faculty of Agricultural, Environmental and Food Sciences, Free University of Bolzano, 39100 Bolzano, Italy; (T.P.); (S.B.)
| | - Alberto Ferrari
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (A.F.); (C.F.)
| | - Marco Cipolloni
- TES Pharma S.r.l., Via P. Togliatti 20, Corciano, 06073 Perugia, Italy; (M.C.); (E.C.); (F.D.F.); (N.G.)
| | - Elisa Cacioni
- TES Pharma S.r.l., Via P. Togliatti 20, Corciano, 06073 Perugia, Italy; (M.C.); (E.C.); (F.D.F.); (N.G.)
| | - Francesca De Franco
- TES Pharma S.r.l., Via P. Togliatti 20, Corciano, 06073 Perugia, Italy; (M.C.); (E.C.); (F.D.F.); (N.G.)
| | - Nicola Giacchè
- TES Pharma S.r.l., Via P. Togliatti 20, Corciano, 06073 Perugia, Italy; (M.C.); (E.C.); (F.D.F.); (N.G.)
| | - Serena Rinaldo
- Department of Biochemical Sciences, University of Rome “La Sapienza”, P.le Aldo Moro 5, 00185 Rome, Italy; (L.C.); (S.R.)
| | - Claudia Folli
- Department of Food and Drug, University of Parma, 43124 Parma, Italy; (A.F.); (C.F.)
| | - Francesco Sansone
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy; (D.D.); (A.C.); (F.S.); (R.B.)
| | - Rodolfo Berni
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 17/a, 43124 Parma, Italy; (D.D.); (A.C.); (F.S.); (R.B.)
| | - Michele Cianci
- Department of Agricultural, Food and Environmental Sciences, Università Politecnica delle Marche, Via Brecce Bianche, 60131 Ancona, Italy
| |
Collapse
|
13
|
Henriques P, Bicker J, Silva S, Doktorovová S, Fortuna A. Nasal-PAMPA: A novel non-cell-based high throughput screening assay for prediction of nasal drug permeability. Int J Pharm 2023; 643:123252. [PMID: 37479103 DOI: 10.1016/j.ijpharm.2023.123252] [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: 05/10/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 07/23/2023]
Abstract
In nasal drug product development, screening studies are vital to select promising compounds or formulations. The Parallel Artificial Membrane Permeability Assay (PAMPA), a high throughput screening tool, has been applied to evaluate drug permeability across several barriers such as the skin or blood-brain barrier. Herein, a new nasal-PAMPA model was optimized to predict nasal permeability, using a biorelevant donor medium containing mucin. The apparent permeability (Papp) of 15 reference compounds was assessed in six different experimental conditions, and the most discriminating and predictive model was applied to a test drug (piroxicam) and mucoadhesive powder formulations loading the same drug. The model with 0.5% (w/v) mucin in the donor compartment and 2% (w/v) phosphatidylcholine in the lipid membrane accurately distinguished high and low permeable compounds. Additionally, it exhibited the highest correlation with permeation across human nasal epithelial cells, RPMI 2650 (R2 = 0.93). When applied to powder formulations, this model was sensitive to the presence of mucoadhesive excipients and the drug solid state. Overall, the nasal-PAMPA model was more rapid than cell-based assays, without requiring specialized training or equipment, showing to be a promising in vitro tool that can be applied in drug and formulation screening for nasal delivery.
Collapse
Affiliation(s)
- Patrícia Henriques
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; R&D, Drug Product Development, Hovione FarmaCiencia SA, Lisbon, Portugal
| | - Joana Bicker
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | - Soraia Silva
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal
| | | | - Ana Fortuna
- University of Coimbra, Faculty of Pharmacy, Coimbra, Portugal; University of Coimbra, Coimbra Institute for Biomedical Imaging and Translational Research, Coimbra, Portugal.
| |
Collapse
|
14
|
Singh S, Agrawal M, Vashist R, Patel RK, Sangave SD, Alexander A. Recent advancements on in vitro blood-brain barrier model: A reliable and efficient screening approach for preclinical and clinical investigation. Expert Opin Drug Deliv 2023; 20:1839-1857. [PMID: 38100459 DOI: 10.1080/17425247.2023.2295940] [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: 09/06/2023] [Accepted: 12/13/2023] [Indexed: 12/17/2023]
Abstract
INTRODUCTION The efficiency of brain therapeutics is greatly hindered by the blood-brain barrier (BBB). BBB's protective function, selective permeability, and dynamic functionality maintain the harmony between the brain and peripheral region. Thus, the design of any novel drug carrier system requires the complete study and investigation of BBB permeability, efflux transport, and the effect of associated cellular and non-vascular unit trafficking on BBB penetrability. The in vitro BBB models offer a most promising, and reliable mode of initial investigation of BBB permeability and associated factors as strong evidence for further preclinical and clinical investigation. AREA COVERED This review work covers the structure and functions of BBB components and different types of in vitro BBB models along with factors affecting BBB model development and model selection criteria. EXPERT OPINION In vivo models assume to reciprocate the physiological environment to the maximum extent. However, the interspecies variability, NVUs trafficking, dynamic behavior of BBB, etc., lead to non-reproducible results. The in vitro models are comparatively less complex, and flexible, as per the study design, could generate substantial evidence and help identify suitable in vivo animal model selection.
Collapse
Affiliation(s)
- Snigdha Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Mukta Agrawal
- School of Pharmacy and Technology Management, Narsee Monjee Institute of Management Studies, Mahbubnagar, India
| | - Rajat Vashist
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Rohit K Patel
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | | | - Amit Alexander
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| |
Collapse
|
15
|
Vincze A, Dékány G, Bicsak R, Formanek A, Moreau Y, Koplányi G, Takács G, Katona G, Balogh-Weiser D, Arany Á, Balogh GT. Natural Lipid Extracts as an Artificial Membrane for Drug Permeability Assay: In Vitro and In Silico Characterization. Pharmaceutics 2023; 15:pharmaceutics15030899. [PMID: 36986760 PMCID: PMC10053807 DOI: 10.3390/pharmaceutics15030899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/04/2023] [Accepted: 03/06/2023] [Indexed: 03/12/2023] Open
Abstract
In vitro non-cellular permeability models such as the parallel artificial membrane permeability assay (PAMPA) are widely applied tools for early-phase drug candidate screening. In addition to the commonly used porcine brain polar lipid extract for modeling the blood–brain barrier’s permeability, the total and polar fractions of bovine heart and liver lipid extracts were investigated in the PAMPA model by measuring the permeability of 32 diverse drugs. The zeta potential of the lipid extracts and the net charge of their glycerophospholipid components were also determined. Physicochemical parameters of the 32 compounds were calculated using three independent forms of software (Marvin Sketch, RDKit, and ACD/Percepta). The relationship between the lipid-specific permeabilities and the physicochemical descriptors of the compounds was investigated using linear correlation, Spearman correlation, and PCA analysis. While the results showed only subtle differences between total and polar lipids, permeability through liver lipids highly differed from that of the heart or brain lipid-based models. Correlations between the in silico descriptors (e.g., number of amide bonds, heteroatoms, and aromatic heterocycles, accessible surface area, and H-bond acceptor–donor balance) of drug molecules and permeability values were also found, which provides support for understanding tissue-specific permeability.
Collapse
Affiliation(s)
- Anna Vincze
- Department of Chemical and Environmental Process Engineering, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Gergely Dékány
- Department of Chemical and Environmental Process Engineering, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Richárd Bicsak
- Department of Chemical and Environmental Process Engineering, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - András Formanek
- ESAT-STADIUS KU LEUVEN, 3001 Leuven, Belgium
- Department of Measurement and Information Systems, Faculty of Electrical Engineering and Informatics, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Yves Moreau
- ESAT-STADIUS KU LEUVEN, 3001 Leuven, Belgium
| | - Gábor Koplányi
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Gergely Takács
- Department of Chemical and Environmental Process Engineering, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
- Mcule.com Kft, Bartók Béla út 105-113, H-1115 Budapest, Hungary
| | - Gábor Katona
- Institute of Pharmaceutical Technology and Regulatory Affairs, Faculty of Pharmacy, University of Szeged, Eötvös Str. 6, H-6720 Szeged, Hungary
| | - Diána Balogh-Weiser
- Department of Organic Chemistry and Technology, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
- Department of Physical Chemistry and Materials Science, Faculty of Chemical Technology and Biotechnology, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
| | - Ádám Arany
- ESAT-STADIUS KU LEUVEN, 3001 Leuven, Belgium
| | - György T. Balogh
- Department of Chemical and Environmental Process Engineering, Budapest University of Technology and Economics, Műegyetem rkp. 3, H-1111 Budapest, Hungary
- Institute of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
- Correspondence:
| |
Collapse
|
16
|
Mathew AA, Mohapatra S, Panonnummal R. Formulation and evaluation of magnesium sulphate nanoparticles for improved CNS penetrability. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:567-576. [PMID: 36474021 DOI: 10.1007/s00210-022-02356-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Abstract
Magnesium (Mg2+) is the fourth most abundant cation in the human body and is involved in maintaining varieties of cellular and neurological functions. Magnesium deficiency has been associated with numerous diseases, particularly neurological disorders, and its supplementation has proven beneficial. However, magnesium therapy in neurological diseases is limited because of the inability of magnesium to cross the blood-brain barrier (BBB). The present study focuses on developing magnesium sulphate nanoparticles (MGSN) to improve blood-brain barrier permeability. MGSN was prepared by precipitation technique with probe sonication. The developed formulation was characterized by DLS, EDAX, FT-IR and quantitative and qualitative estimation of magnesium. According to the DLS report, the average size of the prepared MGSN is found to be 247 nm. The haemocompatibility assay studies revealed that the prepared MGSN are biocompatible at different concentrations. The in vitro BBB permeability assay conducted by Parallel Artificial Membrane Permeability Assay (PAMPA) using rat brain tissue revealed that the prepared MGSN exhibited enhanced BBB permeability as compared to the marketed i.v. MgSO4 injection. The reversal effect of MGSN to digoxin-induced Na+/K+ ATPase enzyme inhibition using brain microslices confirmed that MGSN could attenuate the altered levels of Na+ and K+ and is useful in treating neurological diseases with altered expression of Na+/K+ ATPase activity.
Collapse
Affiliation(s)
- Aparna Ann Mathew
- Amrita School of Pharmacy, Amrita Institute of Medical Science & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Sudeshna Mohapatra
- Amrita School of Pharmacy, Amrita Institute of Medical Science & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Rajitha Panonnummal
- Amrita School of Pharmacy, Amrita Institute of Medical Science & Research Centre, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| |
Collapse
|
17
|
Blood brain barrier-on-a-chip to model neurological diseases. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
18
|
Sekikawa A, Wharton W, Butts B, Veliky CV, Garfein J, Li J, Goon S, Fort A, Li M, Hughes TM. Potential Protective Mechanisms of S-equol, a Metabolite of Soy Isoflavone by the Gut Microbiome, on Cognitive Decline and Dementia. Int J Mol Sci 2022; 23:11921. [PMID: 36233223 PMCID: PMC9570153 DOI: 10.3390/ijms231911921] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 09/27/2022] [Accepted: 10/01/2022] [Indexed: 11/16/2022] Open
Abstract
S-equol, a metabolite of soy isoflavone daidzein transformed by the gut microbiome, is the most biologically potent among all soy isoflavones and their metabolites. Soy isoflavones are phytoestrogens and exert their actions through estrogen receptor-β. Epidemiological studies in East Asia, where soy isoflavones are regularly consumed, show that dietary isoflavone intake is inversely associated with cognitive decline and dementia; however, randomized controlled trials of soy isoflavones in Western countries did not generally show their cognitive benefit. The discrepant results may be attributed to S-equol production capability; after consuming soy isoflavones, 40-70% of East Asians produce S-equol, whereas 20-30% of Westerners do. Recent observational and clinical studies in Japan show that S-equol but not soy isoflavones is inversely associated with multiple vascular pathologies, contributing to cognitive impairment and dementia, including arterial stiffness and white matter lesion volume. S-equol has better permeability to the blood-brain barrier than soy isoflavones, although their affinity to estrogen receptor-β is similar. S-equol is also the most potent antioxidant among all known soy isoflavones. Although S-equol is available as a dietary supplement, no long-term trials in humans have examined the effect of S-equol supplementation on arterial stiffness, cerebrovascular disease, cognitive decline, or dementia.
Collapse
Affiliation(s)
- Akira Sekikawa
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Whitney Wharton
- School of Nursing and Medicine, Emory University, Atlanta, GA 30322, USA
| | - Brittany Butts
- School of Nursing and Medicine, Emory University, Atlanta, GA 30322, USA
| | - Cole V. Veliky
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Joshua Garfein
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jiatong Li
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Shatabdi Goon
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Annamaria Fort
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Mengyi Li
- Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Timothy M. Hughes
- Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| |
Collapse
|
19
|
Self-Emulsifying Drug Delivery Systems: An Alternative Approach to Improve Brain Bioavailability of Poorly Water-Soluble Drugs through Intranasal Administration. Pharmaceutics 2022; 14:pharmaceutics14071487. [PMID: 35890385 PMCID: PMC9319231 DOI: 10.3390/pharmaceutics14071487] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 11/30/2022] Open
Abstract
Efforts in discovering new and effective neurotherapeutics are made daily, although most fail to reach clinical trials. The main reason is their poor bioavailability, related to poor aqueous solubility, limited permeability through biological membranes, and the hepatic first-pass metabolism. Nevertheless, crossing the blood–brain barrier is the major drawback associated with brain drug delivery. To overcome it, intranasal administration has become more attractive, in some cases even surpassing the oral route. The unique anatomical features of the nasal cavity allow partial direct drug delivery to the brain, circumventing the blood–brain barrier. Systemic absorption through the nasal cavity also avoids the hepatic first-pass metabolism, increasing the systemic bioavailability of highly metabolized entities. Nevertheless, most neurotherapeutics present physicochemical characteristics that require them to be formulated in lipidic nanosystems as self-emulsifying drug delivery systems (SEDDS). These are isotropic mixtures of oils, surfactants, and co-surfactants that, after aqueous dilution, generate micro or nanoemulsions loading high concentrations of lipophilic drugs. SEDDS should overcome drug precipitation in absorption sites, increase their permeation through absorptive membranes, and enhance the stability of labile drugs against enzymatic activity. Thus, combining the advantages of SEDDS and those of the intranasal route for brain delivery, an increase in drugs’ brain targeting and bioavailability could be expected. This review deeply characterizes SEDDS as a lipidic nanosystem, gathering important information regarding the mechanisms associated with the intranasal delivery of drugs loaded in SEDDS. In the end, in vivo results after SEDDS intranasal or oral administration are discussed, globally revealing their efficacy in comparison with common solutions or suspensions.
Collapse
|
20
|
Kosinska GP, Ognichenko LM, Shyrykalova AO, Burdina YF, Artemenko AG, Kuz’min VE. Influence of Chemical Structure of Molecules on Blood–Brain Barrier Permeability on the Pampa Model. THEOR EXP CHEM+ 2022. [DOI: 10.1007/s11237-022-09718-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
21
|
Guo J, Xu A, Cheng M, Wan Y, Wang R, Fang Y, Jin Y, Xie SS, Liu J. Design, Synthesis and Biological Evaluation of New 3,4-Dihydro-2(1H)-Quinolinone-Dithiocarbamate Derivatives as Multifunctional Agents for the Treatment of Alzheimer’s Disease. Drug Des Devel Ther 2022; 16:1495-1514. [PMID: 35611357 PMCID: PMC9124477 DOI: 10.2147/dddt.s354879] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 05/10/2022] [Indexed: 12/01/2022] Open
Abstract
Background Alzheimer’s disease (AD) belongs to neurodegenerative disease, and the increasing number of AD patients has placed a heavy burden on society, which needs to be addressed urgently. ChEs/MAOs dual-target inhibitor has potential to treat AD according to reports. Purpose To obtain effective multi-targeted agents for the treatment of AD, a novel series of hybrid compounds were designed and synthesized by fusing the pharmacophoric features of 3,4-dihydro-2 (1H)-quinolinone and dithiocarbamate. Methods All compounds were evaluated for their inhibitory abilities of ChEs and MAOs. Then, further biological activities of the most promising candidate 3e were determined, including the ability to cross the blood-brain barrier (BBB), kinetics and molecular model analysis, cytotoxicity in vitro and acute toxicity studies in vivo. Results Most compounds showed potent and clear inhibition to AChE and MAOs. Among them, compound 3e was considered to be the most effective and balanced inhibitor to both AChE and MAOs (IC50=0.28 µM to eeAChE; IC50=0.34 µM to hAChE; IC50=2.81 µM to hMAO-B; IC50=0.91 µM to hMAO-A). In addition, 3e showed mixed inhibition of hAChE and competitive inhibition of hMAO-B in the enzyme kinetic studies. Further studies indicated that 3e could penetrate the BBB and showed no toxicity on PC12 cells and HT-22 cells when the concentration of 3e was lower than 12.5 µM. More importantly, 3e lacked acute toxicity in mice even at high dose (2500 mg/kg, P.O.). Conclusion This work indicated that compound 3e with a six-carbon atom linker and a piperidine moiety at terminal position was a promising candidate and was worthy of further study.
Collapse
Affiliation(s)
- Jie Guo
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Airen Xu
- Clinical Pharmacology Research Center, The Second Hospital of Yinzhou, Ningbo, Zhejiang, People’s Republic of China
| | - Maojun Cheng
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Yang Wan
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Rikang Wang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Yuanying Fang
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Yi Jin
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
| | - Sai-Sai Xie
- National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
- Correspondence: Sai-Sai Xie, National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Chinese Medicine, No. 56, Yangming Road, Donghu District, Nanchang City, Jiangxi Province, 330006, People’s Republic of China, Email
| | - Jing Liu
- School of Pharmacy, Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, People’s Republic of China
- Jing Liu, School of Pharmacy, Jiangxi University of Chinese Medicine, No. 56, Yangming Road, Donghu District, Nanchang City, Jiangxi Province, 330006, People’s Republic of China, Email
| |
Collapse
|
22
|
Abstract
Levodopa treatment remains the gold standard for Parkinson's disease, but shortcomings related to the pharmacological profile, notably, oral administration and the consequent occurrence of motor complications, have led to the development of several add-on levodopa treatments or to research to improve the method of delivery. Motor fluctuations, and to a lesser extent non-motor fluctuations, concern half of the patients with Parkinson's disease after 5 years of disease and patients identified them as one of their most bothersome symptoms. Catechol-O-methyl transferase inhibitors (COMT-Is) are one of the recommended first-line levodopa add-on therapies for the amelioration of end-of dose motor fluctuations in patient with advanced Parkinson's disease. Currently, two peripheral COMT-Is are considered as first-line choices - entacapone (ENT), which was approved by the US Food and Drug Administration in 1999 and the European Committee in 1998; and opicapone (OPC), which was approved by the European Committee in 2016. A second-line COMT-I that requires regular hepatic monitoring, tolcapone (TOL), was approved by the Food and Drug Administration in 1998 and the European Committee in 1997. Of note, OPC also received Food and Drug Administration approval in 2021, but it is still only marketed in a few countries, including Germany, UK, Spain, Portugal, Italy, Japan, and USA, while ENT and TOL have a wider market. Our narrative review summarizes the pharmacokinetic/pharmacodynamic properties, clinical efficacy in terms of motor fluctuations, motor/non-motor symptoms, quality of life, and safety data of these three COMT-Is, as evidenced by randomized clinical trials, as well as by real-life observational studies. Overall, a phase III non-inferiority trial showed a similar effect between ENT and OPC on off-time (-60.8 min/day and -40.3 min/day, vs placebo, respectively), with a possible additional off-time reduction of 39 min/day, obtained when there is a switch from ENT to OPC. Concomitantly, TOL can reduce off-time by an average of 98 min/day. A significant though discrete concomitant reduction on the Unified Parkinson's Disease Rating Scale motor section (2-3 points) is obtained with all three drugs vs placebo. Data on quality of life are fewer and more heterogeneous, with positive results obtained especially in open-label studies. Effects on non-motor symptoms were investigated as secondary outcome only in a few studies, frequently by means of non-specific scales and a benefit was observed in open-label studies. Dopaminergic adverse effects were the most frequent, dyskinesia being the most common for the three drugs eventually requiring levodopa dose reductions. No urine discoloration and a very low incidence of diarrhea were found with OPC compared with ENT and TOL. Regular hepatic monitoring is needed only for TOL. A combination of COMT-Is with new formulations of levodopa, including the subcutaneous, intrajejunal, or new extended-release formulation, merits further exploration to improve the management of both mild and severe motor fluctuations.
Collapse
|
23
|
Cao Y, Zhang L, Geng Y, Li Y, Zhao Q, Huang J, Ning P, Tian S. Evaluation of the permeability and potential toxicity of polycyclic aromatic hydrocarbons to pulmonary surfactant membrane by the parallel artificial membrane permeability assay model. CHEMOSPHERE 2022; 290:132485. [PMID: 34627814 DOI: 10.1016/j.chemosphere.2021.132485] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 09/11/2021] [Accepted: 10/04/2021] [Indexed: 06/13/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) can penetrate and accumulate in the pulmonary surfactant (PS) membranes, leading to abnormalities of biological macromolecules and the destruction of membrane structure and properties. In the present study, the bioavailability, apparent permeability, effective permeability and residual coefficient of 10 PAHs on PS membrane was assessed by the parallel artificial membrane permeability assay (PAMPA). The influence of various forces on permeability is obtained by analyzing the correlation between parameters and physicochemical properties. Research shows that octanol-water partition coefficient (Kow) cannot directly predict permeability, and permeability has no significant relationship with polarity. Dispersion, induction, coupling/polarization promote permeation, while hydrogen bonded acid and n-n electron pair inhibit permeation. Further surface pressure-area (π-A) isotherms test and Brewster angle microscope observation manifested that there are huge differences in the transmembrane ability and effects on the membrane of PAHs with different structures. This work has considerable significance that will help to evaluate the bioavailability and human health risk of PAHs.
Collapse
Affiliation(s)
- Yan Cao
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Linfeng Zhang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Yingxue Geng
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Yingjie Li
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Qun Zhao
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China.
| | - Jianhong Huang
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Ping Ning
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
| | - Senlin Tian
- Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China.
| |
Collapse
|
24
|
Murata Y, Neuhoff S, Rostami-Hodjegan A, Takita H, Al-Majdoub ZM, Ogungbenro K. In Vitro to In Vivo Extrapolation Linked to Physiologically Based Pharmacokinetic Models for Assessing the Brain Drug Disposition. AAPS J 2022; 24:28. [PMID: 35028763 PMCID: PMC8817058 DOI: 10.1208/s12248-021-00675-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/09/2021] [Indexed: 11/30/2022] Open
Abstract
Drug development for the central nervous system (CNS) is a complex endeavour with low success rates, as the structural complexity of the brain and specifically the blood-brain barrier (BBB) poses tremendous challenges. Several in vitro brain systems have been evaluated, but the ultimate use of these data in terms of translation to human brain concentration profiles remains to be fully developed. Thus, linking up in vitro-to-in vivo extrapolation (IVIVE) strategies to physiologically based pharmacokinetic (PBPK) models of brain is a useful effort that allows better prediction of drug concentrations in CNS components. Such models may overcome some known aspects of inter-species differences in CNS drug disposition. Required physiological (i.e. systems) parameters in the model are derived from quantitative values in each organ. However, due to the inability to directly measure brain concentrations in humans, compound-specific (drug) parameters are often obtained from in silico or in vitro studies. Such data are translated through IVIVE which could be also applied to preclinical in vivo observations. In such exercises, the limitations of the assays and inter-species differences should be adequately understood in order to verify these predictions with the observed concentration data. This report summarizes the state of IVIVE-PBPK-linked models and discusses shortcomings and areas of further research for better prediction of CNS drug disposition. Graphical abstract ![]()
Collapse
Affiliation(s)
- Yukiko Murata
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, University of Manchester, Manchester, M13 9PT, UK.,Sohyaku.Innovative Research Division, Mitsubishi Tanabe Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, Kanagawa, 227-0033, Japan
| | - Sibylle Neuhoff
- Certara UK Ltd, Simcyp Division, 1 Concourse Way, Level 2-Acero, Sheffield, S1 2BJ, UK
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, University of Manchester, Manchester, M13 9PT, UK.,Certara UK Ltd, Simcyp Division, 1 Concourse Way, Level 2-Acero, Sheffield, S1 2BJ, UK
| | - Hiroyuki Takita
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, University of Manchester, Manchester, M13 9PT, UK.,Development Planning, Clinical Development Center, Asahi Kasei Pharma Corporation, Hibiya Mitsui Tower, 1-1-2 Yurakucho, Chiyoda-ku, Tokyo, 100-0006, Japan
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, University of Manchester, Manchester, M13 9PT, UK
| | - Kayode Ogungbenro
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
25
|
Sandbhor P, Goda J, Mohanty B, Chaudhari P, Dutt S, Banerjee R. Non-invasive transferrin targeted nanovesicles sensitize resistant glioblastoma multiforme tumors and improve survival in orthotopic mouse models. NANOSCALE 2021; 14:108-126. [PMID: 34897360 DOI: 10.1039/d1nr05460k] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The blood-brain barrier (BBB) and tumor heterogeneity have resulted in abysmally poor clinical outcomes in glioblastoma (GBM) with the standard therapeutic regimen. Despite several anti-glioma drug delivery strategies, the lack of adequate chemotherapeutic bioavailability in gliomas has led to a suboptimal therapeutic gain in terms of improvement in survival and increased systemic toxicities. This has paved the way for designing highly specific and non-invasive drug delivery approaches for treating GBM. The intranasal (IN) route is one such delivery strategy that has the potential to reach the brain parenchyma by circumventing the BBB. We recently showed that in situ hydrogel embedded with miltefosine (HePc, proapoptotic anti-tumor agent) and temozolomide (TMZ, DNA methylating agent) loaded targeted nanovesicles prevented tumor relapses in orthotopic GBM mouse models. In this study, we specifically investigated the potential of a non-invasive IN route of TMZ delivered from lipid nanovesicles (LNs) decorated with surface transferrin (Tf) and co-encapsulated with HePc to reach the brain by circumventing the BBB in glioma bearing mice. The targeted nanovesicles (228.3 ± 10 nm, -41.7 ± 4 mV) exhibited mucoadhesiveness with 2% w/v mucin suggesting their potential to increase brain drug bioavailability after IN administration. The optimized TLNs had controlled, tunable and significantly different release kinetics in simulated cerebrospinal fluid and simulated nasal fluid demonstrating efficient release of the payload upon reaching the brain. Drug synergy (combination index, 0.7) showed a 6.4-fold enhanced cytotoxicity against resistant U87MG cells compared to free drugs. In vivo gamma scintigraphy of 99mTc labeled LNs showed 500- and 280-fold increased brain concentration post 18 h of treatment. The efficacy of the TLNs increased by 1.8-fold in terms of survival of tumor-bearing mice compared to free drugs. These findings suggested that targeted drug synergy has the potential to intranasally deliver a high therapeutic dose of the chemotherapy agent (TMZ) and could serve as a platform for future clinical application.
Collapse
Affiliation(s)
- Puja Sandbhor
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai, Mumbai, India.
| | - Jayant Goda
- Department of Radiation Oncology ACTREC, Tata Memorial Center, Kharghar Navi-Mumbai, India.
| | - Bhabani Mohanty
- Department of Comparative Oncology and Small Animal Imaging Facility, ACTREC, Tata Memorial Center, Kharghar Navi-Mumbai, India
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai, Mumbai, India.
| | - Pradip Chaudhari
- Department of Radiation Oncology ACTREC, Tata Memorial Center, Kharghar Navi-Mumbai, India.
- Department of Comparative Oncology and Small Animal Imaging Facility, ACTREC, Tata Memorial Center, Kharghar Navi-Mumbai, India
| | - Shilpee Dutt
- Department Shilpee Lab/DNA Repair and Cellular Oncology Lab, ACTREC, Tata Memorial Center, Kharghar Navi-Mumbai, India
| | - Rinti Banerjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology, Bombay, Powai, Mumbai, India.
| |
Collapse
|
26
|
Hyun S, Park N, Nam SH, Cheon DH, Lee Y, Lim HS, Yu J. One-bead-one-compound screening approach to the identification of cyclic peptoid inhibitors of cyclophilin D as neuroprotective agents from mitochondrial dysfunction. Chem Commun (Camb) 2021; 57:2388-2391. [PMID: 33538721 DOI: 10.1039/d0cc08268f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In an effort designed to discover superior inhibitors of cyclophilin D (CypD), we identified and screened members of a one-bead-one-compound (OBOC) library of cyclic peptoid analogues of cyclosporin A (CsA). The results show that the one member of this cyclic peptoid family, I11, inhibits mitochondrial membrane potential changes mediated by CypD.
Collapse
Affiliation(s)
- Soonsil Hyun
- The Institute of Molecular Biology & Genetics, Seoul National University, Seoul 08826, Korea
| | - Namjoon Park
- Department of Chemistry & Education, Seoul National University, Seoul 08826, Korea.
| | - So Hee Nam
- Department of Chemistry, Seoul National University, Seoul 08826, Korea.
| | - Dae Hee Cheon
- Department of Chemistry, Seoul National University, Seoul 08826, Korea.
| | - Yan Lee
- Department of Chemistry, Seoul National University, Seoul 08826, Korea.
| | - Hyun-Suk Lim
- Department of Chemistry, Pohang University of Science and Technology, Pohang 37673, Korea.
| | - Jaehoon Yu
- Department of Chemistry & Education, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
27
|
Wu Z, Xian Z, Ma W, Liu Q, Huang X, Xiong B, He S, Zhang W. Artificial neural network approach for predicting blood brain barrier permeability based on a group contribution method. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2021; 200:105943. [PMID: 33515846 DOI: 10.1016/j.cmpb.2021.105943] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/11/2021] [Indexed: 06/12/2023]
Abstract
BACKGROUND AND OBJECTIVE The purpose of this study was to develop a quantitative structure-activity relationship (QSAR) model for the prediction of blood brain barrier (BBB) permeability by using artificial neural networks (ANN) in combination with molecular structure and property descriptors. METHODS Using a database composed of 300 compounds, 52 structure descriptors obtained based on the universal quasichemical functional group activity coefficients (UNIFAC) group contribution method and the selected 8 molecular property descriptors were used as the network inputs, whereas logBB values of compounds constituted its output. RESULTS The correlation coefficient R of the constructed prediction model, the relative error (RE) and the root mean square error (RMSE) was 0.956, 0.857, and 0.171, respectively. These indicators reflected the feasibility, robustness and accuracy of the prediction model. Compared with the previously published results, a significant improvement in the predictions of the proposed ANN model was observed. CONCLUSIONS ANN model based on the group contribution method could achieve a satisfactory performance for logBB prediction.
Collapse
Affiliation(s)
- Zeyu Wu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China; Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230601, China.
| | - Zhaojun Xian
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China; Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Wanru Ma
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China; Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Qingsong Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China; Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Xusheng Huang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China; Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Baoyi Xiong
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China; Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Shudong He
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China; Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230601, China
| | - Wencheng Zhang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei 230601, China; Engineering Research Center of Bio-Process, Ministry of Education, Hefei University of Technology, Hefei 230601, China.
| |
Collapse
|
28
|
Greenwood J, Pham H, Rey J. Opicapone: A third generation COMT inhibitor. Clin Park Relat Disord 2020; 4:100083. [PMID: 34316661 PMCID: PMC8299985 DOI: 10.1016/j.prdoa.2020.100083] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/22/2020] [Accepted: 11/24/2020] [Indexed: 11/30/2022] Open
Abstract
Opicapone is a 3rd generation catechol-O-methyl transferase (COMT) inhibitor approved in April 2020 by the FDA. Opicapone is used once daily as adjunctive to levodopa/carbidopa to reduce “off” periods in patients with Parkinson’s disease. In clinical trials BIPARK I and II, 50 mg of opicapone once daily was shown to be noninferior to entacapone and reduced the mean off time by about 50 min when compared to placebo. Most common treatment-emergent adverse events for opicapone were dyskinesia, falls, insomnia, and elevated blood creatine phosphokinase levels; it has not been associated with severe hepatic impairment.
Objective: To provide a drug review of the newly FDA approved catechol-O-methyl transferase (COMT) inhibitor, opicapone, for the use of end-of-motor motor fluctuation in adults with Parkinson’s disease. Data sources: A literature search of Pubmed was performed till May 2020 using the following key terms: opicapone, Ongentys, and BIA 9-1067. Review articles, clinical trials, and drug monographs were reviewed. Study selection and data extraction: Relevant English-language monographs and studies conducted in humans were considered. Data synthesis: Opicapone was FDA approved for the treatment of end-of-motor motor fluctuation in adults with Parkinson’s disease in April 2020 based on two published randomized clinical trials that were 14 to 15 weeks in duration called BIPARK I and BIPARK II. Based on the clinical trials, 50 mg of opicapone once daily was shown to be noninferior to entacapone and reduced the mean off time by about 50 min when compared to placebo. Most common treatment-emergent adverse events were dyskinesia, falls, insomnia, and elevated blood creatine phosphokinase levels. Relevance to patient care and clinical practice: Opicapone overcomes the limitations associated with other COMT inhibitors since it is dosed once daily, well tolerated, and has not been associated with the risk of hepatic failure. When switching from entacapone to opicapone a reduction in “off” time of −39.3 min was also seen. Conclusions: Opicapone is a once daily 3rd generation COMT inhibitor that has the potential to benefit patients with Parkinson’s disease who are experiencing end-of-motor fluctuations.
Collapse
|
29
|
Dey SK, Saini M, Prabhakar P, Kundu S. Dopamine β hydroxylase as a potential drug target to combat hypertension. Expert Opin Investig Drugs 2020; 29:1043-1057. [DOI: 10.1080/13543784.2020.1795830] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Sanjay Kumar Dey
- Department of Biochemistry, University of Delhi South Camp us , New Delhi, India
| | - Manisha Saini
- Department of Biochemistry, University of Delhi South Camp us , New Delhi, India
| | - Pankaj Prabhakar
- Department of Biochemistry, University of Delhi South Camp us , New Delhi, India
| | - Suman Kundu
- Department of Biochemistry, University of Delhi South Camp us , New Delhi, India
| |
Collapse
|
30
|
Bailey-Hytholt CM, Shen TL, Nie B, Tripathi A, Shukla A. Placental Trophoblast-Inspired Lipid Bilayers for Cell-Free Investigation of Molecular Interactions. ACS APPLIED MATERIALS & INTERFACES 2020; 12:31099-31111. [PMID: 32558532 DOI: 10.1021/acsami.0c06197] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
The placenta plays a key role in regulating the maternal-fetal transport but it is a difficult organ to study due to a lack of existing in vitro models. Lipid bilayers inspired by the placenta can provide a facile new in vitro tool with promise for screening molecular transport across this important organ. Here we developed lipid bilayers that mimic the composition of human placental trophoblast cells at different times during the course of pregnancy. Mass spectrometry identified five major lipid classes (phosphatidylcholine, phosphatidylethanolamine, phosphatidylinositol, phosphatidylserine, and sphingomyelin) present at varying concentrations in trophoblasts representative of the first and third trimesters and full-term placenta. We successfully developed supported and suspended lipid bilayers mimicking these trophoblast lipid compositions and then demonstrated the utility of these synthetic placenta models for investigating molecular interactions. Specifically, we investigated the interactions with di(2-ethylhexyl) phthalate (DEHP), a common plasticizer and environmental toxicant, and amphotericin B, a common yet toxic, antifungal therapeutic. Overall, we observed that DEHP adsorbs and potentially embeds itself within all placental lipid bilayers, with varying levels of interaction. For both amphotericin B and a liposomal formulation of amphotericin B, AmBisome, we noted lower levels of permeation in transport studies with bilayers and trophoblast cells compared with DEHP, likely driven by differences in size. AmBisome interacted less with both the supported and suspended placental lipid bilayers in comparison to amphotericin B, suggesting that drug delivery carriers can vary the impact of a pharmaceutical agent on these lipid structures. We found that the apparent permeability observed in suspended bilayers was approximately an order of magnitude less than those observed for trophoblast monolayers, which is typical of lipid bilayers. Ultimately, these placenta mimetic lipid bilayers can serve as a platform for the rapid initial screening of molecular interactions with the maternal-fetal interface to better inform future testing.
Collapse
Affiliation(s)
- Christina M Bailey-Hytholt
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Tun-Li Shen
- Department of Chemistry, Brown University, Providence, Rhode Island 02912, United States
| | - Bonnee Nie
- Department of Biochemistry and Molecular Biology, Brown University, Providence, Rhode Island 02912, United States
| | - Anubhav Tripathi
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| | - Anita Shukla
- School of Engineering, Center for Biomedical Engineering, Brown University, Providence, Rhode Island 02912, United States
| |
Collapse
|
31
|
Bailey-Hytholt CM, Puranik T, Tripathi A, Shukla A. Investigating interactions of phthalate environmental toxicants with lipid structures. Colloids Surf B Biointerfaces 2020; 190:110923. [DOI: 10.1016/j.colsurfb.2020.110923] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 02/24/2020] [Accepted: 03/01/2020] [Indexed: 11/29/2022]
|
32
|
Morais TR, Conserva GAA, Varela MT, Costa-Silva TA, Thevenard F, Ponci V, Fortuna A, Falcão AC, Tempone AG, Fernandes JPS, Lago JHG. Improving the drug-likeness of inspiring natural products - evaluation of the antiparasitic activity against Trypanosoma cruzi through semi-synthetic and simplified analogues of licarin A. Sci Rep 2020; 10:5467. [PMID: 32214193 PMCID: PMC7096397 DOI: 10.1038/s41598-020-62352-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 02/20/2020] [Indexed: 01/05/2023] Open
Abstract
Neolignan licarin A (1) was isolated from leaves of Nectandra oppositifolia (Lauraceae) and displayed activity against trypomastigote forms of the etiologic agent of American trypanosomiasis, Trypanosoma cruzi. Aiming for the establishment of SAR, five different compounds (1a - 1e) were prepared and tested against T. cruzi. The 2-allyl derivative of licarin A (1d) exhibited higher activity against trypomastigotes of T. cruzi (IC50 = 5.0 μM and SI = 9.0), while its heterocyclic derivative 1e displayed IC50 of 10.5 μM and reduced toxicity against NCTC cells (SI > 19.0). However, these compounds presented limited oral bioavailability estimation (<85%, Papp <1.0 × 10-6 cm/s) in parallel artificial membrane permeability assays (PAMPA) due to excessive lipophilicity. Based on these results, different simplified structures of licarin A were designed: vanillin (2), vanillyl alcohol (3), isoeugenol (4), and eugenol (5), as well as its corresponding methyl (a), acetyl (b), O-allyl (c), and C-allyl (d) analogues. Vanillin (2) and its acetyl derivative (2b) displayed expressive activity against intracellular amastigotes of T. cruzi with IC50 values of 5.5 and 5.6 μM, respectively, and reduced toxicity against NCTC cells (CC50 > 200 μM). In addition, these simplified analogues showed a better permeability profile (Papp > 1.0 × 10-6 cm/s) on PAMPA models, resulting in improved drug-likeness. Vanillyl alcohol acetyl derivative (3b) and isoeugenol methyl derivative (4a) displayed activity against the extracellular forms of T. cruzi (trypomastigotes) with IC50 values of 5.1 and 8.8 μM respectively. Based on these results, compounds with higher selectivity index against extracellular forms of the parasite (1d, 1e, 3d, and 4a) were selected for a mechanism of action study. After a short incubation period (1 h) all compounds increased the reactive oxygen species (ROS) levels of trypomastigotes, suggesting cellular oxidative stress. The ATP levels were increased after two hours of incubation, possibly involving a high energy expenditure of the parasite to control the homeostasis. Except for compound 4a, all compounds induced hyperpolarization of mitochondrial membrane potential, demonstrating a mitochondrial imbalance. Considering the unique mitochondria apparatus of T. cruzi and the lethal alterations induced by structurally based on licarin A, these compounds are interesting hits for future drug discovery studies in Chagas disease.
Collapse
Affiliation(s)
- Thiago R Morais
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, São Paulo, 09972-270, Brazil
| | - Geanne A Alves Conserva
- Center of Natural Sciences and Humanities, Universidade Federal do ABC, São Paulo, 09210-580, Brazil
| | - Marina T Varela
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, São Paulo, 09972-270, Brazil
| | - Thais A Costa-Silva
- Center of Natural Sciences and Humanities, Universidade Federal do ABC, São Paulo, 09210-580, Brazil
| | - Fernanda Thevenard
- Center of Natural Sciences and Humanities, Universidade Federal do ABC, São Paulo, 09210-580, Brazil
| | - Vitor Ponci
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, São Paulo, 09972-270, Brazil
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy of University of Coimbra, 3000-370, Coimbra, Portugal
- CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, 3000-370, Coimbra, Portugal
| | - Amílcar C Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy of University of Coimbra, 3000-370, Coimbra, Portugal
- CIBIT/ICNAS - Coimbra Institute for Biomedical Imaging and Translational Research, University of Coimbra, 3000-370, Coimbra, Portugal
| | - Andre G Tempone
- Centre for Parasitology and Mycology, Instituto Adolfo Lutz, São Paulo, 01246-000, Brazil
| | - João Paulo S Fernandes
- Institute of Environmental, Chemical and Pharmaceutical Sciences, Universidade Federal de São Paulo, São Paulo, 09972-270, Brazil.
| | - João Henrique G Lago
- Center of Natural Sciences and Humanities, Universidade Federal do ABC, São Paulo, 09210-580, Brazil.
| |
Collapse
|
33
|
Abstract
Knowledge about the transport of active compounds across the blood-brain barrier is of essential importance for drug development. Systemically applied drugs for the central nervous system (CNS) must be able to cross the blood-brain barrier in order to reach their target sites, whereas drugs that are supposed to act in the periphery should not permeate the blood-brain barrier so that they do not trigger any adverse central adverse effects. A number of approaches have been pursued, and manifold in silico, in vitro, and in vivo animal models were developed in order to be able to make a better prediction for humans about the possible penetration of active substances into the CNS. In this particular case, however, in vitro models play a special role, since the data basis for in silico models is usually in need of improvement, and the predictive power of in vivo animal models has to be checked for possible species differences. The blood-brain barrier is a dynamic, highly selective barrier formed by brain capillary endothelial cells. One of its main tasks is the maintenance of homeostasis in the CNS. The function of the barrier is regulated by cells of the microenvironment and the shear stress mediated by the blood flow, which makes the model development most complex. In general, one could follow the credo "as easy as possible, as complex as necessary" for the usage of in vitro BBB models for drug development. In addition to the description of the classical cell culture models (transwell, hollow fiber) and guidance how to apply them, the latest developments (spheroids, microfluidic models) will be introduced in this chapter, as it is attempted to get more in vivo-like and to be applicable for high-throughput usage with these models. Moreover, details about the development of models based on stem cells derived from different sources with a special focus on human induced pluripotent stem cells are presented.
Collapse
Affiliation(s)
- Winfried Neuhaus
- Competence Unit Molecular Diagnostics, Center Health and Bioresources, AIT - Austrian Institute of Technology GmbH, Vienna, Austria.
| |
Collapse
|
34
|
Puscas I, Bernard-Patrzynski F, Jutras M, Lécuyer MA, Bourbonnière L, Prat A, Leclair G, Roullin VG. IVIVC Assessment of Two Mouse Brain Endothelial Cell Models for Drug Screening. Pharmaceutics 2019; 11:pharmaceutics11110587. [PMID: 31717321 PMCID: PMC6920823 DOI: 10.3390/pharmaceutics11110587] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 10/28/2019] [Accepted: 10/30/2019] [Indexed: 01/10/2023] Open
Abstract
Since most preclinical drug permeability assays across the blood-brain barrier (BBB) are still evaluated in rodents, we compared an in vitro mouse primary endothelial cell model to the mouse b.End3 and the acellular parallel artificial membrane permeability assay (PAMPA) models for drug screening purposes. The mRNA expression of key feature membrane proteins of primary and bEnd.3 mouse brain endothelial cells were compared. Transwell® monolayer models were further characterized in terms of tightness and integrity. The in vitro in vivo correlation (IVIVC) was obtained by the correlation of the in vitro permeability data with log BB values obtained in mice for seven drugs. The mouse primary model showed higher monolayer integrity and levels of mRNA expression of BBB tight junction (TJ) proteins and membrane transporters (MBRT), especially for the efflux transporter Pgp. The IVIVC and drug ranking underlined the superiority of the primary model (r2 = 0.765) when compared to the PAMPA-BBB (r2 = 0.391) and bEnd.3 cell line (r2 = 0.019) models. The primary monolayer mouse model came out as a simple and reliable candidate for the prediction of drug permeability across the BBB. This model encompasses a rapid set-up, a fair reproduction of BBB tissue characteristics, and an accurate drug screening.
Collapse
Affiliation(s)
- Ina Puscas
- Faculty of Pharmacy, Université de Montréal, CP6128 Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; (I.P.); (F.B.-P.); (M.J.)
| | - Florian Bernard-Patrzynski
- Faculty of Pharmacy, Université de Montréal, CP6128 Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; (I.P.); (F.B.-P.); (M.J.)
| | - Martin Jutras
- Faculty of Pharmacy, Université de Montréal, CP6128 Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; (I.P.); (F.B.-P.); (M.J.)
| | - Marc-André Lécuyer
- Department of Neuroscience, Faculty of Medicine, Université de Montréal and Centre de Recherc and du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada; (M.-A.L.); (L.B.); (A.P.)
- Centre for Biostructural Imaging of Neurodegeneration, Institute for Multiple Sclerosis Research and Neuroimmunology, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Lyne Bourbonnière
- Department of Neuroscience, Faculty of Medicine, Université de Montréal and Centre de Recherc and du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada; (M.-A.L.); (L.B.); (A.P.)
| | - Alexandre Prat
- Department of Neuroscience, Faculty of Medicine, Université de Montréal and Centre de Recherc and du CHUM (CRCHUM), Montréal, QC H2X 0A9, Canada; (M.-A.L.); (L.B.); (A.P.)
| | - Grégoire Leclair
- Faculty of Pharmacy, Université de Montréal, CP6128 Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; (I.P.); (F.B.-P.); (M.J.)
- Correspondence: (G.L.); (V.G.R.)
| | - V. Gaëlle Roullin
- Faculty of Pharmacy, Université de Montréal, CP6128 Succursale Centre-ville, Montreal, QC H3C 3J7, Canada; (I.P.); (F.B.-P.); (M.J.)
- Correspondence: (G.L.); (V.G.R.)
| |
Collapse
|
35
|
Liquid chromatographic methods for the determination of direct oral anticoagulant drugs in biological samples: A critical review. Anal Chim Acta 2019; 1076:18-31. [DOI: 10.1016/j.aca.2019.03.061] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 03/28/2019] [Accepted: 03/29/2019] [Indexed: 12/20/2022]
|
36
|
Johnson SL, Kirk RD, DaSilva NA, Ma H, Seeram NP, Bertin MJ. Polyphenol Microbial Metabolites Exhibit Gut and Blood⁻Brain Barrier Permeability and Protect Murine Microglia against LPS-Induced Inflammation. Metabolites 2019; 9:metabo9040078. [PMID: 31010159 PMCID: PMC6523162 DOI: 10.3390/metabo9040078] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/12/2022] Open
Abstract
Increasing evidence supports the beneficial effects of polyphenol-rich diets, including the traditional Mediterranean diet, for the management of cardiovascular disease, obesity and neurodegenerative diseases. However, a common concern when discussing the protective effects of polyphenol-rich diets against diseases is whether these compounds are present in systemic circulation in their intact/parent forms in order to exert their beneficial effects in vivo. Here, we explore two common classes of dietary polyphenols, namely isoflavones and lignans, and their gut microbial-derived metabolites for gut and blood-brain barrier predicted permeability, as well as protection against neuroinflammatory stimuli in murine BV-2 microglia. Polyphenol microbial metabolites (PMMs) generally showed greater permeability through artificial gut and blood-brain barriers compared to their parent compounds. The parent polyphenols and their corresponding PMMs were evaluated for protective effects against lipopolysaccharide-induced inflammation in BV-2 microglia. The lignan-derived PMMs, equol and enterolactone, exhibited protective effects against nitric oxide production, as well as against pro-inflammatory cytokines (IL-6 and TNF-α) in BV-2 microglia. Therefore, PMMs may contribute, in large part, to the beneficial effects attributed to polyphenol-rich diets, further supporting the important role of gut microbiota in human health and disease prevention.
Collapse
Affiliation(s)
- Shelby L Johnson
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA.
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Riley D Kirk
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Nicholas A DaSilva
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Hang Ma
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA.
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Navindra P Seeram
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA.
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| | - Matthew J Bertin
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA.
| |
Collapse
|
37
|
Physiologically-Based Pharmacokinetic Modeling for Drug-Drug Interactions of Procainamide and N-Acetylprocainamide with Cimetidine, an Inhibitor of rOCT2 and rMATE1, in Rats. Pharmaceutics 2019; 11:pharmaceutics11030108. [PMID: 30845766 PMCID: PMC6470842 DOI: 10.3390/pharmaceutics11030108] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 03/01/2019] [Accepted: 03/03/2019] [Indexed: 01/11/2023] Open
Abstract
Previous observations demonstrated that cimetidine decreased the clearance of procainamide (PA) and/or N-acetylprocainamide (NAPA; the primary metabolite of PA) resulting in the increased systemic exposure and the decrease of urinary excretion. Despite an abundance of in vitro and in vivo data regarding pharmacokinetic interactions between PA/NAPA and cimetidine, however, a mechanistic approach to elucidate these interactions has not been reported yet. The primary objective of this study was to construct a physiological model that describes pharmacokinetic interactions between PA/NAPA and cimetidine, an inhibitor of rat organic cation transporter 2 (rOCT2) and rat multidrug and toxin extrusion proteins (rMATE1), by performing extensive in vivo and in vitro pharmacokinetic studies for PA and NAPA performed in the absence or presence of cimetidine in rats. When a single intravenous injection of PA HCl (10 mg/kg) was administered to rats, co-administration of cimetidine (100 mg/kg) significantly increased systemic exposure and decreased the systemic (CL) and renal (CLR) clearance of PA, and reduced its tissue distribution. Similarly, cimetidine significantly decreased the CLR of NAPA formed by the metabolism of PA and increased the AUC of NAPA. Considering that these drugs could share similar renal secretory pathways (e.g., via rOCT2 and rMATE1), a physiologically-based pharmacokinetic (PBPK) model incorporating semi-mechanistic kidney compartments was devised to predict drug-drug interactions (DDIs). Using our proposed PBPK model, DDIs between PA/NAPA and cimetidine were successfully predicted for the plasma concentrations and urinary excretion profiles of PA and NAPA observed in rats. Moreover, sensitivity analyses of the pharmacokinetics of PA and NAPA showed the inhibitory effects of cimetidine via rMATE1 were probably important for the renal elimination of PA and NAPA in rats. The proposed PBPK model may be useful for understanding the mechanisms of interactions between PA/NAPA and cimetidine in vivo.
Collapse
|
38
|
Fabbri M, Ferreira JJ, Lees A, Stocchi F, Poewe W, Tolosa E, Rascol O. Opicapone for the treatment of Parkinson's disease: A review of a new licensed medicine. Mov Disord 2018; 33:1528-1539. [PMID: 30264443 DOI: 10.1002/mds.27475] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/09/2018] [Accepted: 05/21/2018] [Indexed: 01/08/2023] Open
Abstract
Catechol-O-methyl transferase inhibitors are currently used as first-line add-on therapy to levodopa for the treatment of end-of-dose motor fluctuations in Parkinson's disease patients, as they increase levodopa bioavailability. Several factors hamper the use of current available catechol-O-methyl transferase inhibitors, that is, the moderate efficacy and multiple dosing for entacapone and the risk of liver toxicity with tolcapone. Opicapone, a new long-acting, peripherally selective, once-daily catechol-O-methyl transferase inhibitor, was recently licensed in Europe. Two phase 3 double-blind clinical trials demonstrated opicapone efficacy in reducing OFF time by an average of about 60 minutes daily compared with placebo, without increasing ON time with troublesome dyskinesias. These effects were also maintained during a subsequent open-label extension consisting of 1-year follow-up. Opicapone showed a good safety profile. From June 2016, Opicapone received the approval for marketing authorization from the European Commission as adjunctive therapy to levodopa/DOPA decarboxylase inhibitors in patients with PD and end-of-dose motor fluctuations. We aimed to review the clinical pharmacological data of opicapone, summarize its clinical efficacy and safety issues, and discuss its potential role in the management of Parkinson's disease. © 2018 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Joaquim J Ferreira
- Instituto de Medicina Molecular, Lisbon, Portugal.,Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Medicine, University of Lisbon, Lisbon, Portugal.,CNS - Campus Neurológico Sénior, Torres Vedras, Portugal
| | - Andrew Lees
- The National Hospital, Queen Square, London, UK
| | - Fabrizio Stocchi
- University and Institute for Research and Medical Care, IRCCS San Raffaele, Rome, Italy
| | - Werner Poewe
- Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Eduardo Tolosa
- Movement Disorder Unit, Neurology Service, Hospital Clínic of Barcelona, IDIBAPS, CIBERNED, Barcelona, Spain
| | - Olivier Rascol
- Departments of Clinical Pharmacology and Neurosciences, Clinical Investigation Center CIC 1436, NS-Park/FCRIN network and NeuroToul COEN Center, INSERM, CHU de Toulouse, Université de Toulouse 3, Toulouse, France
| |
Collapse
|
39
|
Li H, Zeng H, He D, Wang M, Liu L, Liang W, Shu Y, Zhao S, Sun G, Lv C, Xiao C, Liu Y. A new approach to examining the extraction process of Zhishi and Zhiqiao considering the synergistic effect of complex mixtures by PAMPA. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1099:10-17. [PMID: 30236780 DOI: 10.1016/j.jchromb.2018.09.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 09/13/2018] [Indexed: 12/11/2022]
Abstract
Zhishi (ZS) and Zhiqiao (ZQ) are two important traditional Chinese medicines (TCMs) that exert various pharmacological functions due to their active ingredients. However, the oral absorption of these ingredients requires further study. At the early drug discovery stage, the high-throughput parallel artificial membrane permeability assay (PAMPA) is one of the most frequently used to predict transcellular passive absorption in in-vitro models. This study aims to establish a new approach to examine an optimal extraction process that can take into account not only the concentration of active ingredients but also the overall absorption properties of the mixtures extracted from TCMs. A high-performance liquid chromatography triple-quadrupole mass spectrometry (HPLC-QqQ-MS/MS) method was validated for the determination of the effective permeability value (Pe) applied to the above experimental medium. The PAMPA experiment showed that certain active ingredients such as diosmin, rhoifolin, eriocitrin, narirutin, naringin, hesperidin and neohesperidin were not detected in the permeability assay of mono-constituents but were well detected and achieved a better absorption in the permeability assay of the mixture, indicating that certain unknown ingredients may act as cosolvents to improve the solubility or permeability of other ingredients. Furthermore, solid phase extraction (SPE) as an enrichment and purification process enhances absorption. In the present study, a novel in vitro approach was developed to decipher the potential role of TCMs in global absorption, and the extraction process for complex TCMs was described and systematically optimized.
Collapse
Affiliation(s)
- Hui Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Honglian Zeng
- Center for Certification and Evaluation, Guangdong Food and Drug Administration, Guangdong 510080, China
| | - Dan He
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Menglei Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Linlin Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Liang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yisong Shu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Siyu Zhao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China; State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangyu Sun
- Chaozhou People's Hospital, Guangdong 521000, China
| | - Cheng Lv
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China.
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing 100029, China.
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
40
|
Bicker J, Alves G, Fortuna A, Soares-da-Silva P, Falcão A. In vitro assessment of the interactions of dopamine β-hydroxylase inhibitors with human P-glycoprotein and Breast Cancer Resistance Protein. Eur J Pharm Sci 2018; 117:35-40. [PMID: 29428540 DOI: 10.1016/j.ejps.2018.02.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 01/06/2018] [Accepted: 02/05/2018] [Indexed: 01/31/2023]
Abstract
Inhibition of the biosynthesis of noradrenaline is a currently explored strategy for the treatment of hypertension, congestive heart failure and pulmonary arterial hypertension. While some dopamine β-hydroxylase (DBH) inhibitors cross the blood-brain barrier (BBB) and cause central as well as peripheral effects (nepicastat), others have limited access to the brain (etamicastat, zamicastat). In this context, peripheral selectivity is clinically advantageous, in order to prevent alterations of noradrenaline levels in the CNS and the occurrence of adverse central effects. A limited brain exposure results from the combination of several factors, such as a reduced passive permeability or affinity for efflux transporters, but efflux liabilities may also lead to unwanted drug-drug interactions (DDIs) in the presence of co-administered substrates or inhibitors. Thus, the purpose of the study herein presented was to explore the interaction of P-glycoprotein (P-gp) and Breast Cancer Resistance Protein (BCRP), the two major efflux transporters of the BBB that hamper the entry of several drugs to the brain, with the DBH inhibitors, etamicastat, nepicastat and zamicastat. Madin-Darby canine kidney cells (MDCK II) and transfected lines with human MDR1 (MDCK-MDR1) and ABCG2 (MDCK-BCRP) genes were used as a BBB surrogate model. P-gp and BCRP substrates and/or inhibitors were identified through intracellular accumulation and bidirectional permeability assays. The obtained data revealed that zamicastat is a concentration-dependent dual P-gp and BCRP inhibitor with IC50 values of 73.8 ± 7.2 μM and 17.0 ± 2.7 μM, while etamicastat and nepicastat inhibited BCRP to greater extent than P-gp, with IC50 values of 47.7 ± 1.8 μM and 59.2 ± 9.4 μM, respectively. Additionally, etamicastat was identified as P-gp and BCRP dual substrate, as demonstrated by net flux ratios of 5.84 and 3.87 and decreased >50% by verapamil and Ko143. Conversely, nepicastat revealed to be a P-gp-only substrate, with a net flux ratio of 2.01, reduced to 0.92 in the presence of verapamil. Furthermore, nepicastat displayed a consistently higher apparent permeability (>8.49 × 10-6 cm s-1) than etamicastat (<0.58 × 10-6 cm s-1). The identification of etamicastat as a dual efflux substrate suggests that P-gp and BCRP may be partially responsible for the limited central exposure of this compound, in association with its low passive permeability. Moreover, the weak efflux inhibitory potencies of etamicastat and nepicastat revealed a low DDI risk, while the dual P-gp/BCRP inhibition of zamicastat could be studied in the future with synergically effluxed compounds, for which BBB penetration is severely impaired.
Collapse
Affiliation(s)
- Joana Bicker
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Gilberto Alves
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; CICS-UBI - Health Sciences Research Centre, University of Beira Interior, Av. Infante D. Henrique, 6200-506 Covilhã, Portugal.
| | - Ana Fortuna
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Patrício Soares-da-Silva
- Department of Research and Development, BIAL, Av. da Siderurgia Nacional, 4745-457 S. Mamede do Coronado, Portugal; Department of Pharmacology and Therapeutics, Faculty of Medicine, University of Porto, 4200-319 Porto, Portugal
| | - Amílcar Falcão
- Laboratory of Pharmacology, Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal; CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| |
Collapse
|
41
|
Delrivo A, Aloisio C, Longhi MR, Granero G. Artificial Lipid Membrane Permeability Method for Predicting Intestinal Drug Transport: Probing the Determining Step in the Oral Absorption of Sulfadiazine; Influence of the Formation of Binary and Ternary Complexes with Cyclodextrins. AAPS PharmSciTech 2018; 19:1437-1447. [PMID: 29450829 DOI: 10.1208/s12249-018-0965-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/24/2018] [Indexed: 11/30/2022] Open
Abstract
We propose an in vitro permeability assay by using a modified lipid membrane to predict the in vivo intestinal passive permeability of drugs. Two conditions were tested, one with a gradient pH (pH 5.5 donor/pH 7.4 receptor) and the other with an iso-pH 7.4. The predictability of the method was established by correlating the obtained apparent intestinal permeability coefficients (Papp) and the oral dose fraction absorbed in humans (fa) of 16 drugs with different absorption properties. The Papp values correlated well with the absorption rates under the two conditions, and the method showed high predictability and good reproducibility. On the other hand, with this method, we successfully predicted the transport characteristics of oral sulfadiazine (SDZ). Also, the tradeoff between the increase in the solubility of SDZ by its complex formation with cyclodextrins and/or aminoacids and its oral permeability was assessed. Results suggest that SDZ is transported through the gastrointestinal epithelium by passive diffusion in a pH-dependent manner. These results support the classification of SDZ as a high/low borderline permeability compound and are in agreement with the Biopharmaceutics Classification Systems (BCS). This conclusion is consistent with the in vivo pharmacokinetic properties of SDZ.
Collapse
|
42
|
Screening of pharmacokinetic properties of fifty dihydropyrimidin(thi)one derivatives using a combo of in vitro and in silico assays. Eur J Pharm Sci 2017; 109:334-346. [DOI: 10.1016/j.ejps.2017.08.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Revised: 08/16/2017] [Accepted: 08/18/2017] [Indexed: 11/19/2022]
|
43
|
A New Generation of Arachidonic Acid Analogues as Potential Neurological Agent Targeting Cytosolic Phospholipase A 2. Sci Rep 2017; 7:13683. [PMID: 29057981 PMCID: PMC5651845 DOI: 10.1038/s41598-017-13996-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 10/04/2017] [Indexed: 11/08/2022] Open
Abstract
Cytosolic phospholipase A2 (cPLA2) is an enzyme that releases arachidonic acid (AA) for the synthesis of eicosanoids and lysophospholipids which play critical roles in the initiation and modulation of oxidative stress and neuroinflammation. In the central nervous system, cPLA2 activation is implicated in the pathogenesis of various neurodegenerative diseases that involves neuroinflammation, thus making it an important pharmacological target. In this paper, a new class of arachidonic acid (AA) analogues was synthesized and evaluated for their ability to inhibit cPLA2. Several compounds were found to inhibit cPLA2 more strongly than arachidonyl trifluoromethyl ketone (AACOCF3), an inhibitor that is commonly used in the study of cPLA2-related neurodegenerative diseases. Subsequent experiments concluded that one of the inhibitors was found to be cPLA2-selective, non-cytotoxic, cell and brain penetrant and capable of reducing reactive oxygen species (ROS) and nitric oxide (NO) production in stimulated microglial cells. Computational studies were employed to understand how the compound interacts with cPLA2.
Collapse
|
44
|
Annus Á, Vécsei L. Spotlight on opicapone as an adjunct to levodopa in Parkinson's disease: design, development and potential place in therapy. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:143-151. [PMID: 28123288 PMCID: PMC5234693 DOI: 10.2147/dddt.s104227] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Parkinson's disease (PD) is a progressive, chronic, neurodegenerative disease characterized by rigidity, tremor, bradykinesia and postural instability secondary to dopaminergic deficit in the nigrostriatal system. Currently, disease-modifying therapies are not available, and levodopa (LD) treatment remains the gold standard for controlling motor and nonmotor symptoms of the disease. LD is extensively and rapidly metabolized by peripheral enzymes, namely, aromatic amino acid decarboxylase and catechol-O-methyltransferase (COMT). To increase the bioavailability of LD, COMT inhibitors are frequently used in clinical settings. Opicapone is a novel COMT inhibitor that has been recently approved by the European Medicines Agency as an adjunctive therapy to combinations of LD and aromatic amino acid decarboxylase inhibitor in adult PD patients with end-of-dose motor fluctuations. We aimed to review the biochemical properties of opicapone, summarize its preclinical and clinical trials and discuss its future potential role in the treatment of PD.
Collapse
Affiliation(s)
- Ádám Annus
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged
| | - László Vécsei
- Department of Neurology, Faculty of Medicine, Albert Szent-Györgyi Clinical Center, University of Szeged; MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| |
Collapse
|
45
|
Zong SY, Pu YQ, Xu BL, Zhang T, Wang B. Study on the physicochemical properties and anti-inflammatory effects of paeonol in rats with TNBS-induced ulcerative colitis. Int Immunopharmacol 2016; 42:32-38. [PMID: 27863299 DOI: 10.1016/j.intimp.2016.11.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/18/2016] [Accepted: 11/11/2016] [Indexed: 12/13/2022]
Abstract
Paeonol, an active component from Paeonia suffruticosa Andr., has a variety of biological activities, such as vascular endothelial cell protection, anti-oxidation, and anti-inflammation. The aim of this study was to investigate the basic physicochemical properties of paeonol, including solubility, oil-water partition coefficient, and permeability. Then evaluated the anti-inflammatory effects of paeonol were evaluated on 2,4,6-trinitrobenzenesulfonic acid-induced ulcerative colitis in rats. The rats were divided randomly into 6 groups, namely, normal, model, paeonol-treated (100, 200, and 400mg/kg), and positive. Each group had 10 rats. Inhibition effects were evaluated by the disease activity index (DAI), colon weight/length ratio, as well as macroscopical and histological evaluations. Serum interleukin (IL)-17, IL-6 and transforming growth factor beta 1 (TGF-β1) levels were determined by enzyme-linked immunosorbent assay. The solubility and oil-water partition coefficient of paeonol in different phosphate buffer solutions were 284.06-598.23 and 461.97-981.17μg/mL, respectively. The effective passive permeability value Pe was 23.49×10-6cm/s. In terms of anti-inflammatory results, compared with the model group, treatment with 200 and 400mg/kg doses of paeonol had significantly decreased DAI, colon weight/length ratio, and macroscopic and histopathological scores. Furthermore, the serum levels of IL-17 and IL-6 were significantly reduced, whereas the TGF-β1 level was increased in the two paeonol-treated groups (medium- and high-dose group). Therefore, paeonol had poor water solubility, but oral absorption was good. In addition, paeonol had therapeutic effects on ulcerative colitis, and the therapeutic efficacy was dose dependent. The results presented in this study provide evidence for the development of a novel therapeutic agent in the treatment of UC. However, whether this agent could have therapeutic benefit or adverse effects in human IBD remains to be fully explored.
Collapse
Affiliation(s)
- Shi-Yu Zong
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New District, Shanghai 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New District, Shanghai 201203, China.
| | - Yi-Qiong Pu
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New District, Shanghai 201203, China.
| | - Ben-Liang Xu
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New District, Shanghai 201203, China.
| | - Tong Zhang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New District, Shanghai 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New District, Shanghai 201203, China.
| | - Bing Wang
- Experiment Center for Teaching and Learning, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New District, Shanghai 201203, China; School of Pharmacy, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New District, Shanghai 201203, China.
| |
Collapse
|
46
|
Grumetto L, Russo G, Barbato F. Immobilized Artificial Membrane HPLC Derived Parameters vs PAMPA-BBB Data in Estimating in Situ Measured Blood–Brain Barrier Permeation of Drugs. Mol Pharm 2016; 13:2808-16. [DOI: 10.1021/acs.molpharmaceut.6b00397] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Lucia Grumetto
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Via D. Montesano, 49, I-80131 Naples, Italy
| | - Giacomo Russo
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Via D. Montesano, 49, I-80131 Naples, Italy
| | - Francesco Barbato
- Dipartimento di Farmacia, Università degli Studi di Napoli Federico II, Via D. Montesano, 49, I-80131 Naples, Italy
| |
Collapse
|