Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 80
Filter
1.
J Alzheimers Dis ; 92(1): 341-348, 2023.
Article in English | MEDLINE | ID: mdl-36744336

ABSTRACT

BACKGROUND: Delay of progression from prodromal Alzheimer's disease (AD) to dementia is an important outcome in AD trials. Centralized adjudication is intended to improve the consistency of dementia diagnosis but has not been scrutinized. OBJECTIVE: To evaluate centralized adjudication for determining progression to dementia compared with Site Investigator opinion or change in Clinical Dementia Rating (CDR). METHODS: We used data from the 2-year APECS trial of verubecestat versus placebo in 1,451 prodromal AD participants. Cases were triggered for central adjudication if: 1) the Site Investigator judged the participant had progressed to dementia, or 2) the participant's CDR sum-of-boxes score increased ≥2 points from baseline. Post-hoc analyses were performed on pooled treatment-group data to compare methods of assessing progression. RESULTS: 581/1,451 (40%) participants had changes triggering adjudication and most (83%) were confirmed as progression to dementia. Only 66% of those who met CDR criteria (regardless of whether they also met Site Investigator criteria) were adjudicated to have progressed to dementia and just 15% of those who met only CDR criteria were adjudicated to have progressed, representing 5% of progressors. In contrast, 99% of those who met Site Investigator criteria (regardless of whether they also met CDR criteria) were adjudicated to have progressed, and the same was true for those who met only Site Investigator criteria. CONCLUSION: A positive Site Investigator opinion is an excellent predictor for a positive adjudication decision regarding onset of dementia. Conversely, sole use of CDR sum-of-boxes change ≥2 is inadequate. The benefit of centralized adjudication appears doubtful.


Subject(s)
Alzheimer Disease , Dementia , Thiadiazines , Humans , Alzheimer Disease/diagnosis , Dementia/drug therapy , Cyclic S-Oxides/therapeutic use , Thiadiazines/therapeutic use , Disease Progression
2.
Neuropathol Appl Neurobiol ; 48(3): e12781, 2022 04.
Article in English | MEDLINE | ID: mdl-34825396

ABSTRACT

We report the neuropathological examination of a patient with Alzheimer's disease (AD) treated for 38 months with low doses of the BACE-1 inhibitor verubecestat. Brain examination showed small plaque size, reduced dystrophic neurites around plaques and reduced synaptic-associated Aß compared with a group of age-matched untreated sporadic AD (SAD) cases. Our findings suggest that BACE-1 inhibition has an impact on synaptic soluble Aß accumulation and neuritic derangement in AD.


Subject(s)
Alzheimer Disease , Thiadiazines , Alzheimer Disease/drug therapy , Alzheimer Disease/pathology , Amyloid beta-Peptides/metabolism , Brain/pathology , Cyclic S-Oxides/therapeutic use , Humans , Plaque, Amyloid/drug therapy , Plaque, Amyloid/pathology , Thiadiazines/therapeutic use
3.
Acta Biochim Biophys Sin (Shanghai) ; 53(12): 1670-1680, 2021 Dec 08.
Article in English | MEDLINE | ID: mdl-34693451

ABSTRACT

Osteosarcoma (OS), the most common malignant bone tumor with high metastatic potential, frequently affects children and adolescents. Epidermal growth factor receptor (EGFR)-targeted tyrosine kinase inhibitors exhibit encouraging anti-tumor activity for patients with solid tumors, whereas their effects on OS remain controversial. In the present study, we aimed to elucidate the anti-tumor activity of gefitinib for OS, as well as to explore the underlying mechanisms. Gefitinib inhibits cell viability, tumor growth, cell migration, and invasion and promotes cell apoptosis and G1 cycle arrest in OS at a relatively high concentration via suppressing the PI3K/Akt and ERK pathways. However, gefitinib treatment results in the feedback activation of signal transducer and activator of transcription 3 (STAT3) induced by interleukin 6 (IL-6) secretion. Combined treatment with gefitinib and stattic, an inhibitor for STAT3 phosphorylation, engenders more evident inhibitory effects on cell proliferation, migration, and invasion and promotive effects on cell apoptosis and G1 phase arrest in OS, compared with the single exposure to gefitinib or stattic. Western blot analysis demonstrates that stattic treatment in gefitinib-treated OS abrogates the IL-6-induced STAT3 activation and subsequently further restrains the activities of EGFR, Akt, and ERK pathways in tumor cells. This study confirms that the EGFR inhibitor of gefitinib has moderate anti-tumor effects on OS through IL-6 secretion-mediated STAT3 activation. Additional administration of stattic in EGFR-targeted therapies may contribute to improve the efficacy for OS.


Subject(s)
Antineoplastic Agents/pharmacology , Cyclic S-Oxides/pharmacology , ErbB Receptors/antagonists & inhibitors , Interleukin-6/metabolism , Osteosarcoma/drug therapy , Protein Kinase Inhibitors/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cyclic S-Oxides/therapeutic use , Female , Gefitinib/pharmacology , Gefitinib/therapeutic use , Humans , Mice, Inbred BALB C , Mice, Nude , Osteosarcoma/pathology , Protein Kinase Inhibitors/therapeutic use , STAT3 Transcription Factor/metabolism , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
4.
Eur J Med Chem ; 219: 113441, 2021 Jul 05.
Article in English | MEDLINE | ID: mdl-33862517

ABSTRACT

Cumulative evidence suggests that ß-amyloid and oxidative stress are closely related with each other and play key roles in the process of Alzheimer's disease (AD). Multitarget regulation of both pathways might represent a promising therapeutic strategy. Here, a series of selenium-containing compounds based on ebselen and verubecestat were designed and synthesized. Biological evaluation showed that 13f exhibited good BACE-1 inhibitory activity (IC50 = 1.06 µΜ) and potent GPx-like activity (ν0 = 183.0 µM min-1). Aß production experiment indicated that 13f could reduce the secretion of Aß1-40 in HEK APPswe 293T cells. Moreover, 13f exerted a cytoprotective effect against the H2O2 or 6-OHDA caused cell damage via alleviation of intracellular ROS, mitochondrial dysfunction, Ca2+ overload and cell apoptosis. The mechanism studies indicated that 13f exhibited cytoprotective effect by activating the Keap1-Nrf2-ARE pathway and stimulating downstream anti-oxidant protein including HO-1, NQO1, TrxR1, GCLC, and GCLM. In addition, 13f significantly reduced the production of NO and IL-6 induced by LPS in BV2 cells, which confirmed its anti-inflammatory activity as a Nrf2 activator. The BBB permeation assay predicted that 13f was able to cross the BBB. In summary, 13f might be a promising multi-target-directed ligand for the treatment of AD.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Ligands , NF-E2-Related Factor 2/antagonists & inhibitors , Neuroprotective Agents/chemical synthesis , Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Antioxidants/metabolism , Aspartic Acid Endopeptidases/metabolism , Azoles/chemistry , Azoles/metabolism , Azoles/pharmacology , Azoles/therapeutic use , Binding Sites , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Cyclic S-Oxides/chemistry , Cyclic S-Oxides/metabolism , Cyclic S-Oxides/pharmacology , Cyclic S-Oxides/therapeutic use , Drug Design , Humans , Interleukin-6/metabolism , Isoindoles , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Docking Simulation , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Organoselenium Compounds/chemistry , Organoselenium Compounds/metabolism , Organoselenium Compounds/pharmacology , Organoselenium Compounds/therapeutic use , Oxidative Stress/drug effects , Peptide Fragments/metabolism , Reactive Oxygen Species/metabolism , Selenium/chemistry , Signal Transduction/drug effects , Thiadiazines/chemistry , Thiadiazines/metabolism , Thiadiazines/pharmacology , Thiadiazines/therapeutic use
5.
Innate Immun ; 27(2): 201-209, 2021 02.
Article in English | MEDLINE | ID: mdl-33576722

ABSTRACT

Increasing evidence indicates that signal transducer and activator of transcription 3 (STAT3), a vital transcription factor, plays crucial roles in the regulation of inflammation. STAT3 has become a novel therapeutic target for intervention in inflammation-related disorders. However, it remains unclear whether STAT3 plays a part in acute hepatic damage. To investigate the effects of STAT3 here, LPS/d-GalN-induced hepatic damage was induced in mice, the STAT3 inhibitor Stattic was administered, and the degree of liver injury, inflammation, and hepatocyte apoptosis were investigated. The results showed that Stattic mitigated the hepatic morphologic abnormalities and decreased the level of aminotransferase in LPS/D-GalN-insulted mice. The results also indicated that Stattic decreased the levels of TNF-α and IL-6, prevented the activation of the caspase cascade, suppressed cleavage of PARP, and decreased the quantity of TUNEL-positive cells. These results suggest that Stattic provided protective benefits in LPS/d-GalN-induced hepatic damage, and the protective effects might be associated with its anti-inflammatory and anti-apoptotic effects. Therefore, STAT3 might become a novel target for intervening in inflammation-based and apoptosis-based hepatic disorders.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cyclic S-Oxides/therapeutic use , Liver Failure, Acute/drug therapy , Liver/pathology , STAT3 Transcription Factor/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacology , Apoptosis , Caspases/metabolism , Cyclic S-Oxides/pharmacology , Disease Models, Animal , Galactosamine/metabolism , Humans , Interleukin-6/metabolism , Lipopolysaccharides/metabolism , Liver/immunology , Liver Failure, Acute/immunology , Male , Mice , Mice, Inbred BALB C , Tumor Necrosis Factor-alpha/metabolism
6.
J Alzheimers Dis ; 79(1): 275-287, 2021.
Article in English | MEDLINE | ID: mdl-33252075

ABSTRACT

BACKGROUND: We performed exploratory analyses of retinal thickness data from a clinical trial of the AßPP cleaving enzyme (BACE) inhibitor verubecestat in patients with Alzheimer's disease (AD). OBJECTIVE: To evaluate: 1) possible retinal thickness changes following BACE inhibition; and 2) possible association between retinal thickness and brain atrophy. METHODS: Retinal thickness was measured using spectral-domain optical coherence tomography in a 78-week randomized placebo-controlled trial of verubecestat in 1,785 patients with mild-to-moderate AD. Changes from baseline in retinal pigment epithelium, macular grid retinal nerve fiber layer, central subfield retinal thickness, and macular grid volume were evaluated for verubecestat versus placebo. Correlation analyses were performed to investigate the potential association between macular grid retinal nerve fiber layer and central subfield retinal thickness with brain volumetric magnetic resonance imaging (vMRI) data at baseline, as well as correlations for changes from baseline at Week 78 in patients receiving placebo. RESULTS: Verubecestat did not significantly alter retinal thickness during the trial compared with placebo. At baseline, mean macular grid retinal nerve fiber layer and central subfield retinal thickness were weakly but significantly correlated (Pearson's r values≤0.23, p-values < 0.01) with vMRI of several brain regions including whole brain, hippocampus, and thalamus. At Week 78, correlations between retinal thickness and brain vMRI changes from baseline in the placebo group were small and mostly not statistically significant. CONCLUSION: BACE inhibition by verubecestat was not associated with adverse effects on retinal thickness in patients with mild-to-moderate AD. Correlations between retinal thickness and brain volume were observed at baseline. TRIAL REGISTRATION: Clinicaltrials.gov NCT01739348 (registered December 3, 2012; https://clinicaltrials.gov/ct2/show/NCT01739348).


Subject(s)
Alzheimer Disease/drug therapy , Brain/diagnostic imaging , Cyclic S-Oxides/therapeutic use , Retina/diagnostic imaging , Thiadiazines/therapeutic use , Aged , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/physiopathology , Atrophy , Brain/pathology , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Organ Size , Retina/pathology , Tomography, Optical Coherence
7.
Invest New Drugs ; 39(3): 764-774, 2021 06.
Article in English | MEDLINE | ID: mdl-33245464

ABSTRACT

Anaplastic thyroid cancer (ATC) is a rare type of thyroid cancer (TC) with no effective therapeutic strategy. Although surgery, chemotherapy and radiation are all available for ATC treatment, the median survival for ATC patients is less than 6 months. In this study, we aimed to study on resistant mechanisms to B-Raf proto-oncogene serine/threonine kinase (BRAF) inhibitor and identify effective combinational therapy for ATC patients. TC cells were treated with Vemurafenib and cell apoptosis and viability were analyzed by flow cytometry and MTT assay. Monolayer and sphere cells were isolated from ATC cells to detect the mRNA level of stem cell markers and differentiation markers by RT-PCR. Phosphor-STAT3 level in sphere and monolayer cells was tested by Western blotting. The xenotransplantation animal model has established to analyze the anti-tumor effect of Vemurafenib and Stattic combinational therapy. Undifferentiated TC cells were resistant to Vemurafenib treatment. Sphere cells isolated from ATC showed no significant change in cell viability and apoptosis upon Vemurafenib treatment, and expressed a high level of stem cell marker and phosphor-STAT3. STAT3 inhibition enhanced the tumorigenic capacity and increased Vemurafenib sensitivity in ATC cell lines. Stattic significantly enhanced anti-tumor effect of Vemurafenib in mouse model. Our findings demonstrate that the combinational therapy of Vemurafenib and Stattic is an effective therapeutic treatment for ATC patients.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cyclic S-Oxides/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Thyroid Carcinoma, Anaplastic/drug therapy , Thyroid Neoplasms/drug therapy , Vemurafenib/therapeutic use , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cyclic S-Oxides/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Synergism , Female , Humans , Mice, Nude , STAT3 Transcription Factor/metabolism , Thyroid Carcinoma, Anaplastic/metabolism , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Tumor Burden/drug effects , Vemurafenib/pharmacology
8.
ACS Appl Mater Interfaces ; 12(51): 57281-57289, 2020 Dec 23.
Article in English | MEDLINE | ID: mdl-33296171

ABSTRACT

As a critical component for photodynamic therapy toward cancer treatment, photosensitizers require high photoinduced reactive oxygen species generation efficiency, good biocompatibility, and high phototoxicity. Herein, a series of donor-acceptor conjugated polymers containing dibenzothiophene-S,S-dioxide derivatives are designed and synthesized, which can be used as effective photosensitizers. The resulting copolymer PTA5 shows strong green light emission with high photoluminescence quantum yields owing to the intercrossed excited state of local existed and charge transfer states. The PTA5 nanoparticles can be fabricated by encapsulation with a biocompatible polymer matrix. Upon excitation at 800 nm, these nanoparticles present a relatively large two-photon absorption cross section of 3.29 × 106 GM. These nanoparticles also exhibit good photostability in water and thus can be utilized for bioimaging. The tissue-penetrating depths of up to 170 µm for hepatic vessels and 380 µm for blood vessels of mouse ear were achieved using PTA5 nanoparticles. Furthermore, PTA5 nanoparticles show impressive reactive oxygen species generation capability under the irradiation of a white light source. This can be attributed to the effective intersystem crossing between high-level excited state. Upon irradiation with white light (400-700 nm) at 50 mW cm-2 for 5 min every other day, the tumor growth can be effectively suppressed in the presence of PTA5 nanoparticles. These findings demonstrate that PTA5 nanoparticles can be used as a photosensitizer for photodynamic therapy.


Subject(s)
Cyclic S-Oxides/therapeutic use , Nanoparticles/therapeutic use , Neoplasms/drug therapy , Photosensitizing Agents/therapeutic use , Polymers/therapeutic use , Thiophenes/therapeutic use , Aniline Compounds/chemical synthesis , Aniline Compounds/radiation effects , Aniline Compounds/therapeutic use , Animals , Cyclic S-Oxides/chemical synthesis , Cyclic S-Oxides/radiation effects , Fluorescent Dyes/chemical synthesis , Fluorescent Dyes/radiation effects , Fluorescent Dyes/therapeutic use , HeLa Cells , Humans , Mice , Nanoparticles/radiation effects , Optical Imaging , Photochemotherapy , Photons , Photosensitizing Agents/chemical synthesis , Photosensitizing Agents/radiation effects , Polymers/chemical synthesis , Polymers/radiation effects , Reactive Oxygen Species/metabolism , Thiophenes/chemical synthesis , Thiophenes/radiation effects
9.
Brain ; 143(12): 3816-3826, 2020 12 01.
Article in English | MEDLINE | ID: mdl-33253354

ABSTRACT

In the phase 3 EPOCH trial (Clinicaltrials.gov; NCT01739348), treatment with the BACE inhibitor verubecestat failed to improve cognition in patients with mild-to-moderate Alzheimer's disease, but was associated with reduced hippocampal volume after 78 weeks as assessed by MRI. The aims of the present exploratory analyses were to: (i) characterize the effect of verubecestat on brain volume by evaluating the time course of volumetric MRI changes for a variety of brain regions; and (ii) understand the mechanism through which verubecestat might cause hippocampal (and other brain region) volume loss by assessing its relationship to measures of amyloid, neurodegeneration, and cognition. Participants were aged 55-85 years with probable Alzheimer's disease dementia and a Mini Mental State Examination score ≥15 and ≤26. MRIs were obtained at baseline and at Weeks 13, 26, 52 and 78 of treatment. MRIs were segmented using Freesurfer and analysed using a tensor-based morphometry method. PET amyloid data were obtained with 18F-flutemetamol (Vizamyl®) at baseline and Week 78. Standardized uptake value ratios were generated with subcortical white matter as a reference region. Neurofilament light chain in the CSF was assessed as a biomarker of neurodegeneration. Compared with placebo, verubecestat showed increased MRI brain volume loss at Week 13 with no evidence of additional loss through Week 78. The verubecestat-related volumetric MRI loss occurred predominantly in amyloid-rich brain regions. Correlations between amyloid burden at baseline and verubecestat-related volumetric MRI reductions were not significant (r = 0.05 to 0.26, P-values > 0.27). There were no significant differences between verubecestat and placebo in changes from baseline in CSF levels of neurofilament light chain at Week 78 (increases of 7.2 and 14.6 pg/ml for verubecestat versus 19.7 pg/ml for placebo, P-values ≥ 0.1). There was a moderate correlation between volumetric MRI changes and cognitive decline in all groups including placebo at Week 78 (e.g. r = -0.45 to -0.55, P < 0.001 for whole brain), but the correlations were smaller at Week 13 and significant only for the verubecestat groups (e.g. r = -0.15 and -0.11, P < 0.04 for whole brain). Our results suggest that the verubecestat-associated MRI brain volume loss is not due to generalized, progressive neurodegeneration, but may be mediated by specific effects on BACE-related amyloid processes.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Brain/diagnostic imaging , Cyclic S-Oxides/therapeutic use , Enzyme Inhibitors/therapeutic use , Thiadiazines/therapeutic use , Aged , Aged, 80 and over , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Brain/drug effects , Diffusion Tensor Imaging , Double-Blind Method , Female , Hippocampus/diagnostic imaging , Hippocampus/drug effects , Humans , Magnetic Resonance Imaging , Male , Mental Status and Dementia Tests , Middle Aged , Neurofilament Proteins/cerebrospinal fluid , Positron-Emission Tomography , Treatment Outcome , White Matter/diagnostic imaging , White Matter/metabolism
10.
J Neurol Neurosurg Psychiatry ; 91(12): 1316-1324, 2020 12.
Article in English | MEDLINE | ID: mdl-33046560

ABSTRACT

OBJECTIVE: To assess the efficacy and safety of Aß-targeting agents for mild to moderate Alzheimer's disease. METHODS: The MEDLINE, Embase, Cochrane Central Register of Controlled Trials, PsycINFO, ClinicalTrials.gov and the WHO's International Clinical Trials Registry Platform search portal were searched from their inception to April 2020. We generated pooled estimates using random effects meta-analyses. RESULTS: Nineteen randomised controlled trials, of which 17 had a low risk of bias, included 12 903 participants. The meta-analysis showed no difference in the cognitive subscale of Alzheimer's Disease Assessment Scale (ADAS-Cog) between anti-Aß drugs and placebo (mean difference (MD): 0.20, 95% CI -0.40 to 0.81; I 2=99.8%; minimal important difference 3.1-3.8 points, moderate-certainty evidence). For ADAS-Cog, results suggested that one drug that increases Aß clearance may differ in effect (MD: -0.96, 95% CI -0.99 to -0.92) from drugs that reduce Aß production (MD: 0.78, 95% CI 0.25 to 1.32) (interaction p<0.000001); this difference also existed in the outcome of MMSE and CDR-SOB. Compared with placebo, anti-Aß drug-related adverse events were as follows: anxiety, depression, diarrhoea, fatigue, rash, syncope and vomit. DISCUSSION: From current evidence, anti-Aß interventions are unlikely to have an important impact on slowing cognitive or functional decline. Although the subgroup analysis suggested possible benefits from Aß clearance drugs, the analysis has limited credibility, and a benefit from drugs that increase clearance, if real, is very small. TRIAL REGISTRATION NUMBER: PROSPERO registration number CRD42019126272.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Acitretin/therapeutic use , Alanine/analogs & derivatives , Alanine/therapeutic use , Alzheimer Disease/metabolism , Alzheimer Disease/physiopathology , Alzheimer Disease/psychology , Amyloid beta-Peptides/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Anxiety/chemically induced , Azepines/therapeutic use , Clioquinol/analogs & derivatives , Clioquinol/therapeutic use , Copper/therapeutic use , Cyclic S-Oxides/therapeutic use , Depression/chemically induced , Diarrhea/chemically induced , Exanthema/chemically induced , Fatigue/chemically induced , Flurbiprofen/therapeutic use , Humans , Immunoglobulins, Intravenous/therapeutic use , Inositol/therapeutic use , Mental Status and Dementia Tests , Minimal Clinically Important Difference , Orotic Acid/therapeutic use , Oxadiazoles/therapeutic use , Severity of Illness Index , Sulfonamides/therapeutic use , Syncope/chemically induced , Thiadiazines/therapeutic use , Treatment Outcome , Vomiting/chemically induced
11.
Alzheimers Dement ; 16(11): 1483-1492, 2020 11.
Article in English | MEDLINE | ID: mdl-33049114

ABSTRACT

INTRODUCTION: The APECS and AMARANTH trials showed that beta-secretase (BACE) inhibitors verubecestat and lanabecestat failed to slow cognitive and functional decline in individuals with prodromal or early Alzheimer's disease. Here, the performance on secondary and exploratory cognitive measures in both studies is reported. METHODS: APECS (verubecestat) and AMARANTH (lanabecestat) were randomized, double-blind, placebo-controlled, parallel-group, 104-week clinical trials conducted by different sponsors. Measures included the 3-Domain Composite Cognition Score (CCS-3D), Repeatable Battery for the Assessment of Neuropsychological Status (RBANS), Letter/Category Fluency, and Digit Symbol Coding. RESULTS: Verubecestat showed worsening on the CCS-3D Total Score, Episodic Memory, and Attention/Processing Speed domains. Lanabecestat showed worsening on the RBANS Total Score, Immediate Memory, and Visuospatial/Constructional Indexes. Both BACE inhibitors showed worsening on Digit Symbol Coding and improvements on Letter/Category Fluency. DISCUSSION: In both studies, many measures showed treatment-associated cognitive worsening, whereas verbal fluency tasks showed improvement.


Subject(s)
Alzheimer Disease/drug therapy , Cognition/drug effects , Cyclic S-Oxides/therapeutic use , Imidazoles/therapeutic use , Spiro Compounds/therapeutic use , Thiadiazines/therapeutic use , Treatment Outcome , Aged , Double-Blind Method , Female , Humans , Male
12.
Article in English | MEDLINE | ID: mdl-31383666

ABSTRACT

The carbapenem-hydrolyzing class D ß-lactamases (CHDLs) are the main mechanism of carbapenem resistance in Acinetobacter baumannii CHDLs are not effectively inactivated by clinically available ß-lactam-type inhibitors. We have previously described the in vitro efficacy of the inhibitor LN-1-255 in combination with carbapenems. The aim of this study was to compare the efficacy of LN-1-255 with that of imipenem in murine pneumonia using A. baumannii strains carrying their most extended carbapenemases, OXA-23 and OXA-24/40. The blaOXA-23 and blaOXA-24/40 genes were cloned into the carbapenem-susceptible A. baumannii ATCC 17978 strain. Clinical isolates Ab1 and JC12/04, producing the enzymes OXA-23 and OXA-24/40, respectively, were used in the study. Pharmacokinetic (PK) parameters were determined. An experimental pneumonia model was used to evaluate the efficacy of the combined imipenem-LN-1-255 therapy. MICs of imipenem decreased between 32- and 128-fold in the presence of LN-1-255. Intramuscular treatment with imipenem-LN-1-255 (30/50 mg/kg) decreased the bacterial burden by (i) 4 and 1.7 log10 CFU/g lung in the infection with the ATCC 17978-OXA-23 and Ab1 strains, respectively, and by (ii) 2.5 and 4.5 log10 CFU/g lung in the infection produced by the ATCC 17978-OXA-24/40 and the JC12/04 strains, respectively. In all assays, combined therapy offered higher protection against pneumonia than that provided by monotherapy. No toxicity was observed in treated mice. Imipenem treatment combined with LN-1-255 treatment significantly reduced the severity of infection by carbapenem-resistant A. baumannii strains carrying CHDLs. Preclinical assays demonstrated the potential of LN-1-255 and imipenem therapy as a new antibacterial treatment.


Subject(s)
Acinetobacter Infections/drug therapy , Acinetobacter Infections/microbiology , Acinetobacter baumannii/drug effects , Acinetobacter baumannii/pathogenicity , Anti-Infective Agents/therapeutic use , Cyclic S-Oxides/therapeutic use , Imipenem/therapeutic use , Penicillins/therapeutic use , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Drug Resistance, Multiple, Bacterial , Male , Mice , Mice, Inbred BALB C , Microbial Sensitivity Tests , beta-Lactamase Inhibitors/therapeutic use , beta-Lactamases/genetics , beta-Lactamases/metabolism
13.
Alzheimers Res Ther ; 11(1): 68, 2019 08 07.
Article in English | MEDLINE | ID: mdl-31387606

ABSTRACT

BACKGROUND: Verubecestat, a BACE1 inhibitor that reduces Aß levels in the cerebrospinal fluid of humans, was not effective in a phase 3 trial (EPOCH) of mild-to-moderate AD and was associated with adverse events. To assist in the development of BACE1 inhibitors, we report detailed safety findings from EPOCH. METHODS: EPOCH was a randomized, double-blind, placebo-controlled 78-week trial evaluating verubecestat 12 mg and 40 mg in participants with mild-to-moderate AD diagnosed clinically. The trial was terminated due to futility close to its scheduled completion. Of 1957 participants who were randomized and took treatment, 652 were assigned to verubecestat 12 mg, 652 to verubecestat 40 mg, and 653 to placebo. Adverse events and relevant laboratory, vital sign, and ECG findings were assessed. RESULTS: Verubecestat 12 mg and 40 mg were associated with an increase in the percentage of participants reporting adverse events versus placebo (89 and 92% vs. 82%), although relatively few participants discontinued treatment due to adverse events (8 and 9% vs. 6%). Adverse events that were increased versus placebo included falls and injuries, suicidal ideation, weight loss, sleep disturbance, rash, and hair color change. Most were mild to moderate in severity. Treatment differences in suicidal ideation emerged within the first 3 months but did not appear to increase after 6 months. In contrast, treatment differences in falls and injuries continued to increase over time. CONCLUSIONS: Verubecestat was associated with increased risk for several types of adverse events. Falls and injuries were notable for progressive increases over time. While the mechanisms underlying the increased adverse events are unclear, they may be due to BACE inhibition and should be considered in future clinical development programs of BACE1 inhibitors. TRIAL REGISTRATION: ClinicalTrials.gov NCT01739348 , registered on 29 November 2012.


Subject(s)
Alzheimer Disease/drug therapy , Cyclic S-Oxides/therapeutic use , Thiadiazines/therapeutic use , Aged , Aged, 80 and over , Alzheimer Disease/psychology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Aspartic Acid Endopeptidases/antagonists & inhibitors , Cyclic S-Oxides/adverse effects , Double-Blind Method , Female , Humans , Male , Middle Aged , Neuropsychological Tests , Suicidal Ideation , Thiadiazines/adverse effects , Treatment Outcome
14.
Expert Opin Pharmacother ; 20(17): 2095-2099, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31423903

ABSTRACT

Introduction: The amyloid-beta (Aß) cascade hypothesis is that reducing Aß levels in the brain will be beneficial in treating Alzheimer's disease. Aß is formed by the cleavage of amyloid precursor protein by ß-site amyloid precure protein cleaving enzyme (BACE1) and the BACE1 inhibitor verubecestat was developed to lower the brain levels of Aß. However, in the EPOCH trial of verubecestat in mild-to-moderate Alzheimer's disease, it was not beneficial and increased adverse effects.Areas covered: Prior to completing EPOCH, APECS, which trialled verubecestat in prodromal Alzheimer's disease, was commenced. Like EPOCH, APECS was terminated early. In APECS, verubecestat 40 mg worsened cognition and increased adverse effects.Expert opinion: In recruiting subjects to clinical trials in Alzheimer's disease, a clinical diagnosis involving the measurement of Aß should be undertaken for all subjects, as this may help to clarify the findings. In my opinion, the failure of verubecestat in EPOCH and APECS probably could have been avoided if a safety and potential efficacy trial (phase 2) had been completed prior to starting phase 3. It seems to me that, as we have a poor understanding of the underlying mechanisms/cause of Alzheimer's disease, this is where the research emphasis should be, not phase 3 clinical trials.


Subject(s)
Alzheimer Disease/drug therapy , Cyclic S-Oxides/therapeutic use , Thiadiazines/therapeutic use , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/cerebrospinal fluid , Amyloid beta-Peptides/metabolism , Brain/metabolism , Cyclic S-Oxides/adverse effects , Depression/etiology , Dermatitis/etiology , Humans , Severity of Illness Index , Thiadiazines/adverse effects , Treatment Failure
16.
Cell Commun Signal ; 17(1): 45, 2019 05 17.
Article in English | MEDLINE | ID: mdl-31101053

ABSTRACT

BACKGROUND: Endometriosis, characterized by the presence of functional endometrial tissues outside the uterus, is one of the most common gynecological disorders. Endometrial mesenchymal stem cells (MSCs) are crucial for the occurrence and development of endometriosis. Ectopic endometrial MSCs exist in the peritoneal cavity. Thus, the bioactive factors in endometriotic peritoneal fluid may regulate the biological behaviors of endometrial MSCs. METHODS: In this study, after assessing the concentration of Activin A in peritoneal fluid using ELISA, we isolated and cultured endometrial MSCs and investigated whether Activin A stimulated endometrial MSCs to differentiate into myofibroblasts and clarified the underlying mechanisms by quantitative real-time PCR, Western blot analysis, immunofluorescent staining, RNA interference and Chromatin immunoprecipitation. We also employed the inhibitors of Activin A to explore the possibility of suppressing the development of fibrosis in endometriosis using primary endometrial MSCs cultures and a mouse model of endometriosis. RESULTS: Here, we revealed that Activin A significantly elevated in endometriotic peritoneal fluid and activin receptor-like kinase (ALK4), the specific receptor for Activin A, obviously enhanced in ectopic endometrial MSCs compared with eutopic endometrial MSCs from women with or without endometriosis. Next, we found that Activin A drived myofibroblast differentiation of endometrial MSCs, with extremely enhanced expression of connective tissue growth factor (CTGF). CTGF was shown to be required for Activin A-induced expression of ACTA2, COL1A1 and FN1 in endometrial MSCs. CTGF induction by Activin A in endometrial MSCs involved the activation of Smad2/3, as evidenced by the phosphorylation and nuclear translocation of Smad2/3 as well as the binding of Smad2/3 to CTGF promoter. Furthermore, Smad/CTGF pathway in endometrial MSCs required activation of STAT3 while independent of PI3K, JNK and p-38 pathways. In addition, we also demonstrated that inhibition of Activin A pathway impeded myofibroblast differentiation of endometrial MSCs and ameliorated fibrosis in endometriosis mice. CONCLUSIONS: Activin A promotes myofibroblast differentiation of endometrial mesenchymal stem cells via STAT3-dependent Smad/CTGF pathway. The results provided the first evidence that STAT3 acted as a crucial Activin A downstream mediator to regulate CTGF production. Our data may supplement the stem cell theory of endometriosis and provide the experimental basis to treat endometriosis-associated fibrosis by manipulating Activin A signaling.


Subject(s)
Activins/metabolism , Cell Differentiation , Connective Tissue Growth Factor/metabolism , Endometriosis/metabolism , Myofibroblasts/metabolism , STAT3 Transcription Factor/metabolism , Smad Proteins/metabolism , Actins/genetics , Actins/metabolism , Activin Receptors, Type I/genetics , Activin Receptors, Type I/metabolism , Activins/genetics , Adult , Animals , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Cyclic S-Oxides/therapeutic use , Endometriosis/drug therapy , Endometrium/metabolism , Female , Fibronectins/genetics , Fibronectins/metabolism , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice , Mice, Inbred BALB C , Middle Aged , Myofibroblasts/cytology
17.
N Engl J Med ; 380(15): 1408-1420, 2019 Apr 11.
Article in English | MEDLINE | ID: mdl-30970186

ABSTRACT

BACKGROUND: Prodromal Alzheimer's disease offers an opportunity to test the effect of drugs that modify the deposition of amyloid in the brain before the onset of dementia. Verubecestat is an orally administered ß-site amyloid precursor protein-cleaving enzyme 1 (BACE-1) inhibitor that blocks production of amyloid-beta (Aß). The drug did not prevent clinical progression in a trial involving patients with mild-to-moderate dementia due to Alzheimer's disease. METHODS: We conducted a randomized, double-blind, placebo-controlled, 104-week trial to evaluate verubecestat at doses of 12 mg and 40 mg per day, as compared with placebo, in patients who had memory impairment and elevated brain amyloid levels but whose condition did not meet the case definition of dementia. The primary outcome was the change from baseline to week 104 in the score on the Clinical Dementia Rating Scale-Sum of Boxes (CDR-SB; scores range from 0 to 18, with higher scores indicating worse cognition and daily function). Secondary outcomes included other assessments of cognition and daily function. RESULTS: The trial was terminated for futility after 1454 patients had been enrolled; 485 had been assigned to receive verubecestat at a dose of 12 mg per day (the 12-mg group), 484 to receive verubecestat at a dose of 40 mg per day (the 40-mg group), and 485 to receive placebo. A total of 234 patients, 231 patients, and 239 patients per group, respectively, completed 104 weeks of the trial regimen. The estimated mean change from baseline to week 104 in the CDR-SB score was 1.65 in the 12-mg group, 2.02 in the 40-mg group, and 1.58 in the placebo group (P = 0.67 for the comparison between the 12-mg group and the placebo group and P = 0.01 for the comparison between the 40-mg group and the placebo group), suggesting a worse outcome in the higher-dose group than in the placebo group. The estimated rate of progression to dementia due to Alzheimer's disease was 24.5, 25.5, and 19.3 events per 100 patient-years in the 12-mg group, the 40-mg group, and the placebo group, respectively (hazard ratio for 40 mg vs. placebo, 1.38; 97.51% confidence interval, 1.07 to 1.79, not adjusted for multiple comparisons), favoring placebo. Adverse events were more common in the verubecestat groups than in the placebo group. CONCLUSIONS: Verubecestat did not improve clinical ratings of dementia among patients with prodromal Alzheimer's disease, and some measures suggested that cognition and daily function were worse among patients who received verubecestat than among those who received placebo. (Funded by Merck Sharp & Dohme; ClinicalTrials.gov number, NCT01953601.).


Subject(s)
Alzheimer Disease/prevention & control , Amyloid beta-Protein Precursor/antagonists & inhibitors , Cognitive Dysfunction/drug therapy , Cyclic S-Oxides/therapeutic use , Thiadiazines/therapeutic use , Aged , Amyloid beta-Peptides/analysis , Brain Chemistry , Cognitive Dysfunction/pathology , Cyclic S-Oxides/adverse effects , Disease Progression , Double-Blind Method , Enzyme Inhibitors/adverse effects , Enzyme Inhibitors/therapeutic use , Female , Hippocampus/pathology , Humans , Intention to Treat Analysis , Magnetic Resonance Imaging , Male , Organ Size , Plaque, Amyloid/diagnostic imaging , Positron-Emission Tomography , Prodromal Symptoms , Thiadiazines/adverse effects , Treatment Failure
19.
Oncogene ; 38(4): 533-548, 2019 01.
Article in English | MEDLINE | ID: mdl-30158673

ABSTRACT

Immunosuppressive myeloid-derived suppressor cells (MDSC) subvert antitumor immunity and limit the efficacy of chimeric antigen receptor T cells (CAR-T). Previously, we reported that the GM-CSF/JAK2/STAT3 axis drives liver-associated MDSC (L-MDSC) proliferation and blockade of this axis rescued antitumor immunity. We extended these findings in our murine liver metastasis (LM) model, by treating tumor-bearing mice with STAT3 inhibitors (STATTIC or BBI608) to further our understanding of how STAT3 drives L-MDSC suppressive function. STAT3 inhibition caused significant reduction of tumor burden as well as L-MDSC frequencies due to decrease in pSTAT3 levels. L-MDSC isolated from STATTIC or BBI608-treated mice had significantly reduced suppressive function. STAT3 inhibition of L-MDSC was associated with enhanced antitumor activity of CAR-T. Further investigation demonstrated activation of apoptotic signaling pathways in L-MDSC following STAT3 inhibition as evidenced by an upregulation of the pro-apoptotic proteins Bax, cleaved caspase-3, and downregulation of the anti-apoptotic protein Bcl-2. Accordingly, there was also a decrease of pro-survival markers, pErk and pAkt, and an increase in pro-death marker, Fas, with activation of downstream JNK and p38 MAPK. These findings represent a previously unrecognized link between STAT3 inhibition and Fas-induced apoptosis of MDSCs. Our findings suggest that inhibiting STAT3 has potential clinical application for enhancing the efficacy of CAR-T cells in LM through modulation of L-MDSC.


Subject(s)
Adenocarcinoma/secondary , Antineoplastic Agents/therapeutic use , Apoptosis/physiology , Benzofurans/therapeutic use , Cyclic S-Oxides/therapeutic use , Liver Neoplasms, Experimental/secondary , Molecular Targeted Therapy , Myeloid-Derived Suppressor Cells/pathology , Naphthoquinones/therapeutic use , Neoplasm Proteins/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , bcl-2-Associated X Protein/physiology , fas Receptor/physiology , Adenocarcinoma/drug therapy , Adenocarcinoma/immunology , Animals , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Benzofurans/pharmacology , Cell Line, Tumor , Colorectal Neoplasms/pathology , Cyclic S-Oxides/pharmacology , Drug Screening Assays, Antitumor , Gene Expression Regulation, Neoplastic , Immunotherapy , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naphthoquinones/pharmacology , Neoplasm Proteins/physiology , STAT3 Transcription Factor/physiology , Signal Transduction , Specific Pathogen-Free Organisms , Tumor Burden , Tumor Escape/physiology , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/genetics
20.
J Prev Alzheimers Dis ; 5(3): 171-174, 2018.
Article in English | MEDLINE | ID: mdl-29972209

ABSTRACT

Although the results were disappointing from two recent clinical trials of amyloid-targeting drugs in mild-to-moderate AD, the trials provided information that will be important for future studies, according to the EU-US CTAD Task Force, which met in November 2017 to discuss the EXPEDITION3 and EPOCH trials. These trials tested two of the predominant drug development strategies for AD: amyloid immunotherapy and BACE inhibition in populations largely composed of mild AD dementia patients. The results of these trials support the emerging consensus that effective amyloid-targeted treatment will require intervention in early, even pre-symptomatic stages of the disease. Further, the Task Force suggested that a refinement of the amyloid hypothesis may be needed and that other hypotheses should be more fully explored. In addition, they called for improved biomarkers and other outcome assessments to detect the earliest changes in the development of AD.


Subject(s)
Alzheimer Disease/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Clinical Trials, Phase II as Topic/statistics & numerical data , Clinical Trials, Phase III as Topic/statistics & numerical data , Cyclic S-Oxides/therapeutic use , Early Diagnosis , Thiadiazines/therapeutic use , Advisory Committees , Humans
SELECTION OF CITATIONS
SEARCH DETAIL