Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters











Publication year range
1.
Int J Mol Sci ; 24(13)2023 Jun 28.
Article in English | MEDLINE | ID: mdl-37445987

ABSTRACT

Tau protein aggregations are important contributors to the etiology of Alzheimer's disease (AD). Hydromethylthionine (HMT) is a potent inhibitor of tau aggregation in vitro and in vivo and is being developed as a possible anti-dementia medication. HMT was also shown to affect the cholinergic system and to interact with mitochondria. Here, we used tau-transgenic (L1 and L66) and wild-type NMRI mice that were treated with HMT, rivastigmine and memantine and with combinations thereof, for 2-4 weeks. We measured HMT concentrations in both brain homogenates and isolated mitochondria and concentrations of glucose, lactate and pyruvate in brain by microdialysis. In isolated brain mitochondria, we recorded oxygen consumption of mitochondrial complexes by respirometry. While rivastigmine and memantine lowered mitochondrial respiration, HMT did not affect respiration in wild-type animals and increased respiration in tau-transgenic L1 mice. Glucose and lactate levels were not affected by HMT administration. The presence of HMT in isolated mitochondria was established. In summary, traditional anti-dementia drugs impair mitochondrial function while HMT has no adverse effects on mitochondrial respiration in tau-transgenic mice. These results support the further development of HMT as an anti-dementia drug.


Subject(s)
Alzheimer Disease , Memantine , Mice , Animals , Rivastigmine/pharmacology , Memantine/pharmacology , Memantine/therapeutic use , tau Proteins/genetics , tau Proteins/metabolism , Mice, Transgenic , Cholinesterase Inhibitors/pharmacology , Alzheimer Disease/drug therapy , Alzheimer Disease/genetics , Alzheimer Disease/chemically induced , Mitochondria/metabolism
2.
Int J Anal Chem ; 2022: 9171836, 2022.
Article in English | MEDLINE | ID: mdl-36110254

ABSTRACT

Formaldehyde is commonly used as an alkylating agent in the pharmaceutical industry. Consequently, its residual level in drug substances and/or their intermediates needs to be accurately quantified. Formaldehyde is a small, volatile molecule with a weak chromophore (the carbonyl group), and its direct analysis by GC-FID and HPLC-UV is difficult. For these reasons, the majority of papers found in the literature are based upon a derivatisation process (most commonly using the desensitised explosive 2,4-dinitrophenylhydrazine) prior to the analysis of formaldehyde. A novel high-performance liquid chromatography (HPLC) method with UV detection for its quantification in a pharmaceutical is described in this paper. The method proposed herein is based upon a derivatisation reaction between formaldehyde and 4-methylbenzenesulfonohydrazide (MBSH) before analysis by HPLC-UV. Selectivity, linearity, limit of quantification, accuracy, repeatability, intermediate precision, and solution stability were successfully assessed as per ICH guideline Q2(R1), and the method has also been validated in a good manufacturing practice (GMP) laboratory in the UK.

3.
Biomedicines ; 10(4)2022 Apr 07.
Article in English | MEDLINE | ID: mdl-35453617

ABSTRACT

The tau protein aggregation inhibitor hydromethylthionine mesylate (HMTM) was shown recently to have concentration-dependent pharmacological activity in delaying cognitive decline and brain atrophy in phase 3 Alzheimer's disease (AD) clinical trials; the activity was reduced in patients receiving symptomatic therapies. The methylthionine (MT) moiety has been reported to increase the clearance of pathological tau and to enhance mitochondrial activity, which is impaired in AD patients. In line 1 (L1) mice (a model of AD), HMTM (5/15 mg/kg) was administered either as a monotherapy or as an add-on to a chronic administration with the cholinesterase inhibitor rivastigmine (0.1/0.5 mg/kg) to explore mitochondrial function and energy substrate utilization as potential targets of drug interference. Compared with wild-type NMRI mice, the L1 mice accumulated greater levels of l-lactate and of the LDH-A subunit responsible for the conversion of pyruvate into l-lactate. In contrast, the levels of LDH-B and mitochondrial ETC subunits and the activity of complexes I and IV was not altered in the L1 mice. The activity of complex I and complex IV tended to increase with the HMTM dosing, in turn decreasing l-lactate accumulation in the brains of the L1 mice, despite increasing the levels of LDH-A. The chronic pre-dosing of the L1 mice with rivastigmine partially prevented the enhancement of the activity of complexes I and IV by HMTM and the increase in the levels of LDH-A while further reducing the levels of l-lactate. Thus, HMTM in combination with rivastigmine leads to a depletion in the energy substrate l-lactate, despite bioenergetic production not being favoured. In this study, the changes in l-lactate appear to be regulated by LDH-A, since neither of the experimental conditions affected the levels of LDH-B. The data show that HMTM monotherapy facilitates the use of substrates for energy production, particularly l-lactate, which is provided by astrocytes, additionally demonstrating that a chronic pre-treatment with rivastigmine prevented most of the HMTM-associated effects.

4.
J Alzheimers Dis ; 75(2): 501-519, 2020.
Article in English | MEDLINE | ID: mdl-32280089

ABSTRACT

BACKGROUND: Hydromethylthionine is a potent inhibitor of pathological aggregation of tau and TDP-43 proteins. OBJECTIVE: To compare hydromethylthionine treatment effects at two doses and to determine how drug exposure is related to treatment response in bvFTD. METHODS: We undertook a 52-week Phase III study in 220 bvFTD patients randomized to compare hydromethylthionine at 200 mg/day and 8 mg/day (intended as a control). The principal outcomes were change on the Addenbrookes Cognitive Examination - Revised (ACE-R), the Functional Activities Questionnaire (FAQ), and whole brain volume. Secondary outcomes included Modified Clinical Global Impression of Change (Modified-CGIC). A population pharmacokinetic exposure-response analysis was undertaken in 175 of the patients with available blood samples and outcome data using a discriminatory plasma assay for the parent drug. RESULTS: There were no significant differences between the two doses as randomized. There were steep concentration-response relationships for plasma levels in the range 0.3-0.6 ng/ml at the 8 mg/day dose on clinical and MRI outcomes. There were significant exposure-dependent differences at 8 mg/day for FAQ, Modified-CGIC, and whole brain atrophy comparing patients with plasma levels greater than 0.346 ng/ml with having minimal drug exposure. The exposure-response is biphasic with worse outcomes at the high concentrations produced by 200 mg/day. CONCLUSIONS: Hydromethylthionine has a similar concentration-response profile for effects on clinical decline and brain atrophy at the 8 mg/day dose in bvFTD as recently reported in AD. Treatment responses in bvFTD are predicted to be maximal at doses in the range 20-60 mg/day. A confirmatory placebo-controlled trial is now planned.


Subject(s)
Atrophy/drug therapy , Brain/drug effects , Frontotemporal Dementia/drug therapy , Methylene Blue/analogs & derivatives , Adult , Aged , Atrophy/diagnostic imaging , Atrophy/pathology , Brain/diagnostic imaging , Brain/pathology , Disease Progression , Dose-Response Relationship, Drug , Double-Blind Method , Female , Frontotemporal Dementia/diagnostic imaging , Frontotemporal Dementia/pathology , Humans , Magnetic Resonance Imaging , Male , Methylene Blue/pharmacology , Methylene Blue/therapeutic use , Middle Aged , Treatment Outcome
5.
J Alzheimers Dis ; 72(3): 931-946, 2019.
Article in English | MEDLINE | ID: mdl-31658058

ABSTRACT

BACKGROUND: Although hydromethylthionine is a potent tau aggregation inhibitor, no difference was found in either of two Phase III trials in mild to moderate Alzheimer's disease (AD) comparing doses in the range 150-250 mg/day with 8 mg/day intended as a control. OBJECTIVE: To determine how drug exposure is related to treatment response. METHODS: A sensitive plasma assay for the drug was used in a population pharmacokinetic analysis of samples from 1,162 of the 1,686 patients who participated in either of the Phase III trials with available samples and efficacy outcome data. RESULTS: There are steep concentration-response relationships for steady state plasma levels in the range 0.3-0.8 ng/ml at the 8 mg/day dose. Using a threshold based on the lower limit of quantitation of the assay on Day 1, there are highly significant differences in cognitive decline and brain atrophy in patients with above threshold plasma levels, both for monotherapy and add-on therapy, but with effect sizes reduced by half as add-on. Plasma concentrations in the range 4-21 ng/ml produced by the high doses are not associated with any additional benefit. CONCLUSIONS: Hydromethylthionine has pharmacological activity on brain structure and function at the 8 mg/day dose as monotherapy or as add-on to symptomatic treatments. This combined with a plateau at higher doses is consistent with the lack of dose-response seen in the Phase III trials. Treatment benefit is predicted to be maximal at 16 mg/day as monotherapy. A placebo-controlled trial in mild/moderate AD is now ongoing to confirm efficacy at this dose.


Subject(s)
Alzheimer Disease/drug therapy , Brain/drug effects , Cognitive Dysfunction/drug therapy , Methylene Blue/analogs & derivatives , Aged , Aged, 80 and over , Alzheimer Disease/blood , Alzheimer Disease/diagnostic imaging , Atrophy , Brain/diagnostic imaging , Brain/metabolism , Cognitive Dysfunction/blood , Cognitive Dysfunction/diagnostic imaging , Dose-Response Relationship, Drug , Female , Humans , Male , Methylene Blue/administration & dosage , Methylene Blue/metabolism
6.
Acta Crystallogr E Crystallogr Commun ; 75(Pt 4): 516-523, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-31161067

ABSTRACT

The asymmetric unit of the title compound, C13H11N3O2S2, comprises two independent mol-ecules (A and B); the crystal structure was determined by employing synchrotron radiation. The mol-ecules exhibit essentially the same features with an almost planar benzo-thia-zole ring (r.m.s. deviation = 0.026 and 0.009 Šfor A and B, respectively), which forms an inclined dihedral angle with the phenyl ring [28.3 (3) and 29.1 (3)°, respectively]. A difference between the mol-ecules is noted in a twist about the N-S bonds [the C-S-N-N torsion angles = -56.2 (5) and -68.8 (5)°, respectively], which leads to a minor difference in orientation of the phenyl rings. In the mol-ecular packing, A and B are linked into a supra-molecular dimer via pairwise hydrazinyl-N-H⋯N(thiazol-yl) hydrogen bonds. Hydrazinyl-N-H⋯O(sulfon-yl) hydrogen bonds between A mol-ecules assemble the dimers into chains along the a-axis direction, while links between centrosymmetrically related B mol-ecules, leading to eight-membered {⋯HNSO}2 synthons, link the mol-ecules along [001]. The result is an undulating supra-molecular layer. Layers stack along the b-axis direction with benzo-thia-zole-C-H⋯O(sulfon-yl) points of contact being evident. The analyses of the calculated Hirshfeld surfaces confirm the relevance of the above inter-molecular inter-actions, but also serve to further differentiate the weaker inter-molecular inter-actions formed by the independent mol-ecules, such as π-π inter-actions. This is also highlighted in distinctive energy frameworks calculated for the individual mol-ecules.

7.
Steroids ; 140: 92-103, 2018 12.
Article in English | MEDLINE | ID: mdl-30273694

ABSTRACT

The crystal structures, Hirshfeld surface analyses and electrostatic potential surfaces of the di- and tri-hydrates of (5α,17E)-17-hydrazonoandrostan-3-ol, 3, namely [3·(H2O)2] and [3·(H2O)3], are reported. The trihydrate, isolated from a solution of 3 in moist methanol, recrystallizes in the orthorhombic space group, P212121, while that of the dihydrate, isolated from a 1:1 aqueous methanol solution, recrystallizes in the monoclinic space group, P21. The asymmetric unit of the trihydrate involves one steroid and three water molecules, while that of the dihydrate has two similar but independent steroid molecules and four hydrate molecules. Very similar conformations are found for the steroid molecules in both hydrates. As expected, the different mole ratios of water: steroid have major influences on the structures. In both cases, complex crystal structures are constructed from various classical hydrogen bonds, involving the hydrate molecules and the hydroxy and hydrazonyl moieties of the steroid. In the trihydrate, there are no direct connections between the steroid molecules, instead the water molecules link the steroid molecules, with only weak van der Waals forces between the steroid molecules. There are some direct links between the steroid molecules in the dihydrate, involving OH(steroid hydroxyl)⋯O(steroid oxo) hydrogen bonds, in a head to head fashion, and OH⋯N(hydrazonyl) hydrogen bonds, in a head to tail fashion. However, the major occurrence throughout the structure is of steroid molecules linked by water molecules.


Subject(s)
Androstanols/chemistry , Crystallography, X-Ray , Hydrogen Bonding , Methanol/chemistry , Models, Molecular , Molecular Conformation , Static Electricity , Surface Properties , Water/chemistry
8.
J Mol Biol ; 430(21): 4119-4131, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30121297

ABSTRACT

Alzheimer's disease is a tauopathy characterized by pathological fibrillization of tau protein to form the paired helical filaments (PHFs), which constitute neurofibrillary tangles. The methylthioninium (MT) moiety reverses the proteolytic stability of the PHF core and is in clinical development for treatment of Alzheimer's disease in a stable reduced form as leuco-MT. It has been hypothesized that MT acts via oxidation of cysteine residues, which is incompatible with activity in the predominantly reducing environment of living cells. We have shown recently that the PHF-core tau unit assembles spontaneously in vitro to form PHF-like filaments. Here we describe studies using circular dichroism, SDS-PAGE, transmission electron microscopy and site-directed mutagenesis to elucidate the mechanism of action of the MT moiety. We show that MT inhibitory activity is optimal in reducing conditions, that the active moiety is the reduced leuco-MT form of the molecule and that its mechanism of action is cysteine independent.


Subject(s)
Cysteine/metabolism , Methylene Blue/analogs & derivatives , Neurofibrillary Tangles/chemistry , Neurofibrillary Tangles/metabolism , tau Proteins/chemistry , tau Proteins/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Heparin/chemistry , Humans , Methylene Blue/chemistry , Molecular Structure , Neurofibrillary Tangles/ultrastructure , Recombinant Proteins , Spectrum Analysis
9.
Front Immunol ; 8: 1361, 2017.
Article in English | MEDLINE | ID: mdl-29109729

ABSTRACT

Molecular engineering to increase the percentage identity to common human immunoglobulin sequences of non-human therapeutic antibodies and scaffolds has become standard practice. This strategy is often used to reduce undesirable immunogenic responses, accelerating the clinical development of candidate domains. The first humanized shark variable domain (VNAR) was reported by Kovalenko and colleagues and used the anti-human serum albumin (HSA) domain, clone E06, as a model to construct a number of humanized versions including huE06v1.10. This study extends this work by using huE06v1.10 as a template to isolate domains with improved biophysical properties and reduced antigenicity. Random mutagenesis was conducted on huE06v1.10 followed by refinement of clones through an off-rate ranking-based selection on target antigen. Many of these next-generation binders retained high affinity for target, together with good species cross-reactivity. Lead domains were assessed for any tendency to dimerize, tolerance to N- and C-terminal fusions, affinity, stability, and relative antigenicity in human dendritic cell assays. Functionality of candidate clones was verified in vivo through the extension of serum half-life in a typical drug format. From these analyses the domain, BA11, exhibited negligible antigenicity, high stability and high affinity for mouse, rat, and HSA. When these attributes were combined with demonstrable functionality in a rat model of PK, the BA11 clone was established as our clinical candidate.

10.
Front Mol Neurosci ; 10: 447, 2017.
Article in English | MEDLINE | ID: mdl-29375308

ABSTRACT

α-Synuclein (α-Syn) aggregation is a pathological feature of synucleinopathies, neurodegenerative disorders that include Parkinson's disease (PD). We have tested whether N,N,N',N'-tetramethyl-10H-phenothiazine-3,7-diaminium bis(hydromethanesulfonate) (leuco-methylthioninium bis(hydromethanesulfonate); LMTM), a tau aggregation inhibitor, affects α-Syn aggregation in vitro and in vivo. Both cellular and transgenic models in which the expression of full-length human α-Syn (h-α-Syn) fused with a signal sequence peptide to promote α-Syn aggregation were used. Aggregated α-Syn was observed following differentiation of N1E-115 neuroblastoma cells transfected with h-α-Syn. The appearance of aggregated α-Syn was inhibited by LMTM, with an EC50 of 1.1 µM, with minimal effect on h-α-Syn mRNA levels being observed. Two independent lines of mice (L58 and L62) transgenic for the same fusion protein accumulated neuronal h-α-Syn that, with aging, developed into fibrillary inclusions characterized by both resistance to proteinase K (PK)-cleavage and their ability to bind thiazin red. There was a significant decrease in α-Syn-positive neurons in multiple brain regions following oral treatment of male and female mice with LMTM administered daily for 6 weeks at 5 and 15 mg MT/kg. The early aggregates of α-Syn and the late-stage fibrillar inclusions were both susceptible to inhibition by LMTM, a treatment that also resulted in the rescue of movement and anxiety-related traits in these mice. The results suggest that LMTM may provide a potential disease modification therapy in PD and other synucleinopathies through the inhibition of α-Syn aggregation.

11.
J Pharmacol Exp Ther ; 352(1): 110-8, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25320049

ABSTRACT

Methylthioninium (MT) is a tau aggregation inhibitor with therapeutic potential in Alzheimer's disease (AD). MT exists in equilibrium between reduced [leucomethylthioninium (LMT)] and oxidized (MT(+)) forms; as a chloride salt [methylthioninium chloride (MTC), "methylene blue"], it is stabilized in its MT(+) form. Although the results of a phase 2 study of MTC in 321 mild/moderate AD subjects identified a 138-mg MT/day dose as the minimum effective dose on cognitive and imaging end points, further clinical development of MT was delayed pending resolution of the unexpected lack of efficacy of the 228-mg MT/day dose. We hypothesized that the failure of dose response may depend on differences known at the time in dissolution in simulated gastric and intestinal fluids of the 100-mg MTC capsules used to deliver the 228-mg dose and reflect previously unsuspected differences in redox processing of MT at different levels in the gut. The synthesis of a novel chemical entity, LMTX (providing LMT in a stable anhydrous crystalline form), has enabled a systematic comparison of the pharmacokinetic properties of MTC and LMTX in preclinical and clinical studies. The quantity of MT released in water or gastric fluid within 60 minutes proved in retrospect to be an important determinant of clinical efficacy. A further factor was a dose-dependent limitation in the ability to absorb MT in the presence of food when delivered in the MT(+) form as MTC. A model is presented to account for the complexity of MT absorption, which may have relevance for other similar redox molecules.


Subject(s)
Alzheimer Disease/drug therapy , Methylene Blue/metabolism , Methylene Blue/pharmacology , Protein Aggregation, Pathological/drug therapy , tau Proteins/chemistry , Absorption, Physicochemical , Administration, Oral , Adolescent , Adult , Animals , Biological Transport , Brain/metabolism , Dose-Response Relationship, Drug , Eating , Erythrocytes/metabolism , Female , Humans , Male , Methylene Blue/administration & dosage , Methylene Blue/therapeutic use , Mice , Oxidation-Reduction , Young Adult
12.
Acta Crystallogr Sect E Struct Rep Online ; 68(Pt 4): o1016-7, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22589890

ABSTRACT

In the title pyrazolone derivative, C(15)H(12)N(2)O, the five-membered ring is approximately planar (r.m.s. deviation = 0.018 Å), and the N- and C-bound benzene rings are inclined to this plane [dihedral angles = 21.45 (10) and 6.96 (10)°, respectively] and form a dihedral angle of 20.42 (10)° with each other. Supra-molecular layers are formed in the crystal structure via C-H⋯O and C-H⋯N inter-actions, and these are assembled into double layers by C-H⋯π and π-π inter-actions between the pyrazole and C-bound benzene rings [ring centroid-centroid distance = 3.6476 (12) Å]. The double layers stack along the a axis being connected by π-π inter-actions between the N- and C-bound benzene rings [ring centroid-centroid distance = 3.7718 (12) Å].

13.
Acta Crystallogr C ; 61(Pt 12): o711-4, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16330855

ABSTRACT

The structure of the title acid as the monohydrate, C14H18O11.H2O, displays hydrogen bonding which connects the molecules in layers parallel to (101). In the anhydrous glucopyranose, C14H20O10, only chain connectivity is attained but, due to disorder of the OH group, only partially and in two modes, one less favoured than the other. This provides incomplete connectivity between molecules in corrugated layers parallel to (010).

14.
Acta Crystallogr B ; 60(Pt 4): 461-71, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15258405

ABSTRACT

The structures of five substituted alpha,alpha'-trehalose trehalose derivatives have been determined, and these are compared with those of four previously published analogues. In 2,2',3,3',4,4'-hexaacetato-6,6'-bis-O-methylsulfonyl-alpha,alpha'-trehalose, C26H38O21S2, where the molecules lie across twofold rotation axes in the space group C2, a single C-H...O=S hydrogen bond links the molecules into sheets. 2,2',3,3',4,4'-Hexaacetato-6,6'-bis-O-(4-toluenesulfonyl)-alpha,alpha'-trehalose, C38H46O21S2, crystallizes with Z' = 2 in the space group P2(1)2(1)2(1) and a combination of three C-H...O hydrogen bonds, each having a carbonyl O atom as an acceptor, and a C-H...pi(arene) hydrogen bond link the molecules into a three-dimensional framework. 2,2',3,3',4,4'-Hexaacetato-6,6'-diazido-alpha,alpha'-trehalose, C24H32N6O15, crystallizes as a partial ethanol solvate and three C-H...O hydrogen bonds link the substituted trehalose molecules into a three-dimensional framework. In 2,2',3,3'-tetraacetato-6,6'-bis(N-acetylamino)-alpha,alpha'-trehalose dihydrate, C24H36N2O15...2H2O, the substituted trehalose molecules lie across twofold rotation axes in the space group P2(1)2(1)2 and a three-dimensional framework is generated by the combination of O-H...O and N-H...O hydrogen bonds. The diaminotrehalose molecules in 6,6'-diamino-alpha,alpha'-trehalose dihydrate, C12H24N2O9...2(H2O), lie across twofold rotation axes in the space group P4(3)2(1)2: a single O-H...N hydrogen bond links the trehalose molecules into sheets, which are linked into a three-dimensional framework by O-H...O hydrogen bonds.

SELECTION OF CITATIONS
SEARCH DETAIL