Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Sci Transl Med ; 16(739): eadk9109, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38507469

ABSTRACT

Myasthenia gravis (MG) is a neuromuscular disease that results in compromised transmission of electrical signals at the neuromuscular junction (NMJ) from motor neurons to skeletal muscle fibers. As a result, patients with MG have reduced skeletal muscle function and present with symptoms of severe muscle weakness and fatigue. ClC-1 is a skeletal muscle specific chloride (Cl-) ion channel that plays important roles in regulating neuromuscular transmission and muscle fiber excitability during intense exercise. Here, we show that partial inhibition of ClC-1 with an orally bioavailable small molecule (NMD670) can restore muscle function in rat models of MG and in patients with MG. In severely affected MG rats, ClC-1 inhibition enhanced neuromuscular transmission, restored muscle function, and improved mobility after both single and prolonged administrations of NMD670. On this basis, NMD670 was progressed through nonclinical safety pharmacology and toxicology studies, leading to approval for testing in clinical studies. After successfully completing phase 1 single ascending dose in healthy volunteers, NMD670 was tested in patients with MG in a randomized, placebo-controlled, single-dose, three-way crossover clinical trial. The clinical trial evaluated safety, pharmacokinetics, and pharmacodynamics of NMD670 in 12 patients with mild MG. NMD670 had a favorable safety profile and led to clinically relevant improvements in the quantitative myasthenia gravis (QMG) total score. This translational study spanning from single muscle fiber recordings to patients provides proof of mechanism for ClC-1 inhibition as a potential therapeutic approach in MG and supports further development of NMD670.


Subject(s)
Chlorides , Myasthenia Gravis , Humans , Rats , Animals , Chlorides/therapeutic use , Myasthenia Gravis/drug therapy , Muscle, Skeletal/physiology , Neuromuscular Junction , Chloride Channels
2.
Eur J Heart Fail ; 25(4): 478-487, 2023 04.
Article in English | MEDLINE | ID: mdl-36924142

ABSTRACT

There are many consequences of heart failure (HF), including symptoms, impaired health-related quality of life (HRQoL), and physical and social limitations (functional status). These have a substantial impact on patients' lives, yet are not routinely captured in clinical trials. Patient-reported outcomes (PROs) can quantify patients' experiences of their disease and its treatment. Steps can be taken to improve the use of PROs in HF trials, in regulatory and payer decisions, and in patient care. Importantly, PRO measures (PROMs) must be developed with involvement of patients, family members, and caregivers from diverse demographic groups and communities. PRO data collection should become more routine not only in clinical trials but also in clinical practice. This may be facilitated by the use of digital tools and interdisciplinary patient advocacy efforts. There is a need for standardization, not only of the PROM instruments, but also in procedures for analysis, interpretation and reporting PRO data. More work needs to be done to determine the degree of change that is important to patients and that is associated with increased risks of clinical events. This 'minimal clinically important difference' requires further research to determine thresholds for different PROMs, to assess consistency across trial populations, and to define standards for improvement that warrant regulatory and reimbursement approvals. PROs are a vital part of patient care and drug development, and more work should be done to ensure that these measures are both reflective of the patient experience and that they are more widely employed.


Subject(s)
Heart Failure , Quality of Life , Humans , Heart Failure/therapy , Patient Participation , Patient Reported Outcome Measures , Caregivers
3.
Mol Ther Nucleic Acids ; 29: 176-188, 2022 Sep 13.
Article in English | MEDLINE | ID: mdl-35860384

ABSTRACT

The introduction of sulfur into the phosphate linkage of chemically synthesized oligonucleotides creates the stereocenters on phosphorus atoms. Researchers have valued the nature of backbone stereochemistry and early on investigated drug properties for the individual stereocenters in dimers or short oligomers. Only very recently, it has become possible to synthesize fully stereodefined antisense oligonucleotides in good yield and purity. Non-bridging phosphorodithioate (PS2) introduces second sulfur into the phosphorothioate linkage to remove the chirality of phosphorus atom. Here, we describe the application of symmetrical non-bridging PS2 linkages in the context of stereodefined locked nucleic acids (LNAs) antisense oligonucleotides with the goal of reducing chiral complexity and, ultimately, resulting in single molecules. In addition, we propose a rather simple strategy to rapidly identify stereodefined gapmers, combining PS2 and a preferred stereochemistry motif (RSSR), which supports RNase-H-mediated target knockdown. Pharmacological efficacy and metabolic stability are investigated systematically using ApoB as a target sequence, where in vivo data correlate well to what is observed in vitro.

4.
Bioorg Med Chem Lett ; 72: 128879, 2022 09 15.
Article in English | MEDLINE | ID: mdl-35809818

ABSTRACT

The N-demethylation of zicronapine (7) and three of its deuterated analogs 8 - 10 has been studied in human in vitro metabolism systems. While the N-deuterio-methyl analog 8 did not behave differently from the parent in human liver microsomes, a significantly reduced rate of N-demethylation was observed as a consequence of benzene ring deuteration (compound 7vs.9). Additional deuteration of the N-methyl group, which as mentioned had shown no effect in isolation, further decreased the rate of the N-demethylation reaction (compound 10vs.9). This paper presents and discusses this unprecedented 'distal kinetic isotope effect' that was observed when incubating the test compounds with human liver microsomes or recombinant human CYP450 liver enzymes.


Subject(s)
Cytochrome P-450 Enzyme System , Microsomes, Liver , Cytochrome P-450 Enzyme System/metabolism , Demethylation , Deuterium/metabolism , Humans , Kinetics , Microsomes, Liver/metabolism
5.
Eur Heart J ; 43(41): 4362-4373, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35900838

ABSTRACT

AIMS: To investigate the impact of patiromer on the serum potassium level and its ability to enable specified target doses of renin-angiotensin-aldosterone system inhibitor (RAASi) use in patients with heart failure and reduced ejection fraction (HFrEF). METHODS AND RESULTS: A total of 1642 patients with HFrEF and current or a history of RAASi-related hyperkalemia were screened and 1195 were enrolled in the run-in phase with patiromer and optimization of the RAASi therapy [≥50% recommended dose of angiotensin-converting enzyme inhibitor/angiotensin receptor blocker/angiotensin receptor-neprilysin inhibitor, and 50 mg of mineralocorticoid receptor antagonist (MRA) spironolactone or eplerenone]. Specified target doses of the RAASi therapy were achieved in 878 (84.6%) patients; 439 were randomized to patiromer and 439 to placebo. All patients, physicians, and outcome assessors were blinded to treatment assignment. The primary endpoint was between-group difference in the adjusted mean change in serum potassium. Five hierarchical secondary endpoints were assessed. At the end of treatment, the median (interquartile range) duration of follow-up was 27 (13-43) weeks, the adjusted mean change in potassium was +0.03 mmol/l in the patiromer group and +0.13 mmol/l in the placebo group [difference in the adjusted mean change between patiromer and placebo: -0.10 mmol/l (95% confidence interval, CI -0.13, 0.07); P < 0.001]. Risk of hyperkalemia >5.5 mmol/l [hazard ratio (HR) 0.63; 95% CI 0.45, 0.87; P = 0.006), reduction of MRA dose (HR 0.62; 95% CI 0.45, 0.87; P = 0.006), and total adjusted hyperkalemia events/100 person-years (77.7 vs. 118.2; HR 0.66; 95% CI 0.53, 0.81; P < 0.001) were lower with patiromer. Hyperkalemia-related morbidity-adjusted events (win ratio 1.53, P < 0.001) and total RAASi use score (win ratio 1.25, P = 0.048) favored the patiromer arm. Adverse events were similar between groups. CONCLUSION: Concurrent use of patiromer and high-dose MRAs reduces the risk of recurrent hyperkalemia (ClinicalTrials.gov: NCT03888066).


Subject(s)
Heart Failure , Hyperkalemia , Humans , Hyperkalemia/drug therapy , Hyperkalemia/complications , Heart Failure/complications , Heart Failure/drug therapy , Stroke Volume , Mineralocorticoid Receptor Antagonists/adverse effects , Renin-Angiotensin System , Potassium
6.
PLoS One ; 17(5): e0268159, 2022.
Article in English | MEDLINE | ID: mdl-35576218

ABSTRACT

BACKGROUND: Despite the discovery of familial cases with mutations in Cu/Zn-superoxide dismutase (SOD1), Guanine nucleotide exchange C9orf72, TAR DNA-binding protein 43 (TARDBP) and RNA-binding protein FUS as well as a number of other genes linked to Amyotrophic Lateral Sclerosis (ALS), the etiology and molecular pathogenesis of this devastating disease is still not understood. As proteins do not act alone, conducting an analysis of ALS at the system level may provide new insights into the molecular biology of ALS and put it into relationship to other neurological diseases. METHODS: A set of ALS-associated genes/proteins were collected from publicly available databases and text mining of scientific literature. We used these as seed proteins to build protein-protein interaction (PPI) networks serving as a scaffold for further analyses. From the collection of networks, a set of core modules enriched in seed proteins were identified. The molecular biology of the core modules was investigated, as were their associations to other diseases. To assess the core modules' ability to describe unknown or less well-studied ALS biology, they were queried for proteins more recently associated to ALS and not involved in the primary analysis. RESULTS: We describe a set of 26 ALS core modules enriched in ALS-associated proteins. We show that these ALS core modules not only capture most of the current knowledge about ALS, but they also allow us to suggest biological interdependencies. In addition, new associations of ALS networks with other neurodegenerative diseases, e.g. Alzheimer's, Huntington's and Parkinson's disease were found. A follow-up analysis of 140 ALS-associated proteins identified since 2014 reveals a significant overrepresentation of new ALS proteins in these 26 disease modules. CONCLUSIONS: Using protein-protein interaction networks offers a relevant approach for broadening the understanding of the biological context of known ALS-associated genes. Using a bottom-up approach for the analysis of protein-protein interaction networks is a useful method to avoid bias caused by over-connected proteins. Our ALS-enriched modules cover most known biological functions associated with ALS. The presence of recently identified ALS-associated proteins in the core modules highlights the potential for using these as a scaffold for identification of novel ALS disease mechanisms.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Protein Interaction Maps , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Computational Biology/methods , Humans , Mutation , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/pathology , RNA-Binding Protein FUS/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/genetics , Superoxide Dismutase-1/metabolism
7.
Nat Commun ; 13(1): 494, 2022 01 25.
Article in English | MEDLINE | ID: mdl-35078977

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is a leading cause of death worldwide, however our understanding of cell specific mechanisms underlying COPD pathobiology remains incomplete. Here, we analyze single-cell RNA sequencing profiles of explanted lung tissue from subjects with advanced COPD or control lungs, and we validate findings using single-cell RNA sequencing of lungs from mice exposed to 10 months of cigarette smoke, RNA sequencing of isolated human alveolar epithelial cells, functional in vitro models, and in situ hybridization and immunostaining of human lung tissue samples. We identify a subpopulation of alveolar epithelial type II cells with transcriptional evidence for aberrant cellular metabolism and reduced cellular stress tolerance in COPD. Using transcriptomic network analyses, we predict capillary endothelial cells are inflamed in COPD, particularly through increased CXCL-motif chemokine signaling. Finally, we detect a high-metallothionein expressing macrophage subpopulation enriched in advanced COPD. Collectively, these findings highlight cell-specific mechanisms involved in the pathobiology of advanced COPD.


Subject(s)
Alveolar Epithelial Cells/metabolism , Lung/metabolism , Pulmonary Disease, Chronic Obstructive/genetics , RNA-Seq/methods , Single-Cell Analysis/methods , A549 Cells , Alveolar Epithelial Cells/classification , Animals , Cells, Cultured , Cluster Analysis , Epithelial Cells/metabolism , Female , Gene Expression Profiling/methods , Gene Regulatory Networks , Humans , Lung/cytology , Male , Mice, Inbred C57BL , Mice, Transgenic , Pulmonary Disease, Chronic Obstructive/pathology , Signal Transduction/genetics
8.
Nutrients ; 13(12)2021 Nov 27.
Article in English | MEDLINE | ID: mdl-34959831

ABSTRACT

This study investigated the postprandial plasma metabolome following consumption of four dairy matrices different in texture and structure: cheddar cheese (Cheese), homogenized cheddar cheese (Hom. Cheese), and micellar casein isolate (MCI) with cream (MCI Drink) or a MCI Gel. An acute, randomized, crossover trial in male participants (n = 25) with four test days was conducted. Blood samples were collected during an 8-h postprandial period after consumption of a meal similar in micro- and macronutrients containing one of the four dairy matrices, and the metabolome was analyzed using nuclear magnetic resonance (NMR) spectroscopy. A liquid dairy matrix (MCI Drink) resulted in a faster absorption of amino acids compared to products, representing either a semi-solid (MCI Gel and Hom. Cheese) or solid (Cheese) dairy matrix. For the MCI Gel, plasma concentration of acetic acid and formic acid increased approximately 2 h following consumption, while 3-hydroxybyturate and acetoacetic acid increased approximately 6 h after consumption. The structure and texture of the dairy matrix affected the postprandial absorption of amino acids, as revealed by the plasma metabolome. Our study furthermore pointed at endogenous effects associated with consumption of dairy products containing glucono-δ-lactone.


Subject(s)
Amino Acids/blood , Dairy Products/analysis , Gastrointestinal Absorption/physiology , Metabolome/drug effects , Postprandial Period/physiology , Adult , Caseins/pharmacology , Cheese/analysis , Gluconates/pharmacology , Humans , Lactones/pharmacology , Magnetic Resonance Spectroscopy , Male , Meals , Young Adult
9.
Am J Clin Nutr ; 114(5): 1729-1742, 2021 11 08.
Article in English | MEDLINE | ID: mdl-34477812

ABSTRACT

BACKGROUND: The dairy matrix may influence digestion and absorption of lipids and thereby risk of cardiovascular diseases (CVDs). However, few postprandial studies have compared dairy products that differed only in terms of their matrix. OBJECTIVES: We aimed to investigate acute 8-h postprandial lipid, glycemic, and appetite responses after intake of isoenergetic dairy meals with different matrixes, but similar nutritional composition. METHODS: Twenty-five normal-weight men (18-40 y old) were enrolled in a randomized controlled crossover trial. On 4 test days, a meal with 1 of 4 dairy products was served: cheddar cheese (Cheese), homogenized Cheese (Hom. Cheese), micellar casein isolate (MCI) with cream (MCI Drink), and a gel produced from the MCI Drink by addition of Glucono Delta-Lactone (MCI Gel). The fat- and protein-matched dairy products differed in terms of their casein network, fat droplet size, and/or texture. Blood biochemistry and appetite responses were collected. RESULTS: Eighteen participants completed the trial. Postprandial triglycerides (TGs) (primary outcome) increased by (mean ± SEM) 0.24 ± 0.07 and 0.19 ± 0.07 mmol/L after MCI Gel compared with Cheese and Hom. Cheese, respectively (both P ≤ 0.05). Likewise, MCI Gel increased TG incremental AUC compared with Cheese and Hom. Cheese (both P < 0.05), and peak compared with Cheese (P < 0.05). ApoB-48 (primary outcome) was unaffected by dairy matrix. For free fatty acids (FFAs), glucose, and insulin, time × meal interactions were observed (all P < 0.001). During the first 2 h, FFAs were lower for Cheese than for MCI products, whereas the opposite was observed for glucose and insulin. CONCLUSIONS: Postprandial TG but not apoB-48 response was higher after MCI Gel, indicating that the type of casein network influences lipid responses. This suggests that the dairy matrix may also affect risk factors for CVDs. Reducing fat droplet size (i.e., Hom. Cheese) did not affect blood biochemistry.This trial was registered at clinicaltrials.gov as NCT03656367.


Subject(s)
Dairy Products , Triglycerides/blood , Adult , Apolipoprotein B-48/blood , Caseins , Cheese , Cross-Over Studies , Energy Intake , Fatty Acids, Nonesterified/blood , Glucagon-Like Peptide 1/blood , Humans , Insulin/blood , Male , Postprandial Period , Young Adult
10.
Metabolites ; 11(7)2021 Jul 14.
Article in English | MEDLINE | ID: mdl-34357348

ABSTRACT

Studies have indicated that the dairy matrix can affect postprandial responses of dairy products, but little is known about the effect on postprandial plasma phospholipid levels. This study investigated postprandial plasma phospholipid levels following consumption of four different dairy products that are similar in micro and macro nutrients, but different in texture and structure: cheddar cheese (Cheese), homogenized cheddar cheese (Hom. Cheese), micellar casein isolate with cream (MCI Drink) or a gel made from the MCI Drink (MCI Gel). The study was an acute randomized, crossover trial in human volunteers with four test days. Blood samples were collected during an 8 h postprandial period and the content of 53 plasma phospholipids was analysed using liquid chromatography-mass spectrometry (LC-MS). No meal-time interactions were revealed; however, for nine of the 53 phospholipids, a meal effect was found. Thus, the results indicated a lower plasma level of specific lyso-phosphatidylethanolamines (LPEs) and lyso-phosphatidylcholines (LPCs) following consumption of the MCI Gel compared to the MCI Drink and Hom. Cheese, which might be attributed to an effect of viscosity. However, further studies are needed in order to reveal more details on the effect of the dairy matrix on postprandial phospholipids.

11.
Lancet ; 396(10266): 1895-1904, 2020 12 12.
Article in English | MEDLINE | ID: mdl-33197395

ABSTRACT

BACKGROUND: Intravenous ferric carboxymaltose has been shown to improve symptoms and quality of life in patients with chronic heart failure and iron deficiency. We aimed to evaluate the effect of ferric carboxymaltose, compared with placebo, on outcomes in patients who were stabilised after an episode of acute heart failure. METHODS: AFFIRM-AHF was a multicentre, double-blind, randomised trial done at 121 sites in Europe, South America, and Singapore. Eligible patients were aged 18 years or older, were hospitalised for acute heart failure with concomitant iron deficiency (defined as ferritin <100 µg/L, or 100-299 µg/L with transferrin saturation <20%), and had a left ventricular ejection fraction of less than 50%. Before hospital discharge, participants were randomly assigned (1:1) to receive intravenous ferric carboxymaltose or placebo for up to 24 weeks, dosed according to the extent of iron deficiency. To maintain masking of patients and study personnel, treatments were administered in black syringes by personnel not involved in any study assessments. The primary outcome was a composite of total hospitalisations for heart failure and cardiovascular death up to 52 weeks after randomisation, analysed in all patients who received at least one dose of study treatment and had at least one post-randomisation data point. Secondary outcomes were the composite of total cardiovascular hospitalisations and cardiovascular death; cardiovascular death; total heart failure hospitalisations; time to first heart failure hospitalisation or cardiovascular death; and days lost due to heart failure hospitalisations or cardiovascular death, all evaluated up to 52 weeks after randomisation. Safety was assessed in all patients for whom study treatment was started. A pre-COVID-19 sensitivity analysis on the primary and secondary outcomes was prespecified. This study is registered with ClinicalTrials.gov, NCT02937454, and has now been completed. FINDINGS: Between March 21, 2017, and July 30, 2019, 1525 patients were screened, of whom 1132 patients were randomly assigned to study groups. Study treatment was started in 1110 patients, and 1108 (558 in the carboxymaltose group and 550 in the placebo group) had at least one post-randomisation value. 293 primary events (57·2 per 100 patient-years) occurred in the ferric carboxymaltose group and 372 (72·5 per 100 patient-years) occurred in the placebo group (rate ratio [RR] 0·79, 95% CI 0·62-1·01, p=0·059). 370 total cardiovascular hospitalisations and cardiovascular deaths occurred in the ferric carboxymaltose group and 451 occurred in the placebo group (RR 0·80, 95% CI 0·64-1·00, p=0·050). There was no difference in cardiovascular death between the two groups (77 [14%] of 558 in the ferric carboxymaltose group vs 78 [14%] in the placebo group; hazard ratio [HR] 0·96, 95% CI 0·70-1·32, p=0·81). 217 total heart failure hospitalisations occurred in the ferric carboxymaltose group and 294 occurred in the placebo group (RR 0·74; 95% CI 0·58-0·94, p=0·013). The composite of first heart failure hospitalisation or cardiovascular death occurred in 181 (32%) patients in the ferric carboxymaltose group and 209 (38%) in the placebo group (HR 0·80, 95% CI 0·66-0·98, p=0·030). Fewer days were lost due to heart failure hospitalisations and cardiovascular death for patients assigned to ferric carboxymaltose compared with placebo (369 days per 100 patient-years vs 548 days per 100 patient-years; RR 0·67, 95% CI 0·47-0·97, p=0·035). Serious adverse events occurred in 250 (45%) of 559 patients in the ferric carboxymaltose group and 282 (51%) of 551 patients in the placebo group. INTERPRETATION: In patients with iron deficiency, a left ventricular ejection fraction of less than 50%, and who were stabilised after an episode of acute heart failure, treatment with ferric carboxymaltose was safe and reduced the risk of heart failure hospitalisations, with no apparent effect on the risk of cardiovascular death. FUNDING: Vifor Pharma.


Subject(s)
Anemia, Iron-Deficiency/drug therapy , Ferric Compounds/therapeutic use , Heart Failure/drug therapy , Maltose/analogs & derivatives , Administration, Intravenous , Aged , Aged, 80 and over , Double-Blind Method , Female , Ferric Compounds/administration & dosage , Heart Failure/complications , Heart Failure/mortality , Hospitalization/statistics & numerical data , Humans , Male , Maltose/administration & dosage , Maltose/therapeutic use , Middle Aged , Patient Discharge , Treatment Outcome , Ventricular Function, Left
12.
J Med Microbiol ; 69(3): 387-395, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31958049

ABSTRACT

Introduction. Cefuroxime is an important antibiotic to treat several serious infections. Rapid elimination through the kidneys and the variation in MICs of various susceptible pathogens such as Escherichia coli, Klebsiella pneumoniae, Staphylococcus aureus and Streptococcus pneumoniae give rise to dosing issues, especially in otherwise healthy patients.Aim. To investigate the probability of target attainment (PTA) for obtaining the optimal dosage regimens for cefuroxime in healthy young people.Methodology. Two weeks apart 750 and 1500 mg cefuroxime were administered as an intravenous bolus to 20 healthy volunteers (mean age: 27 years). Population modelling and simulation studies were done based on the obtained data for cefuroxime plasma concentration.Results. With a target value of time above MIC (T >MIC) greater than 50 % the simulations revealed that a PTA of >99 % is obtained for S. pneumoniae with a dosage regimen of 750 mg q12h. For E. coli and K. pneumoniae the PTA was <90 % even with the highest, simulated dosage of 1500 mg q6h. For S. aureus a dosage of 1500 mg q8h gave a PTA above 97 %.Conclusions. S. pneumoniae is most likely treatable with a two-daily dose of 750 mg cefuroxime. Not treatable are K. pneumoniae and E. coli. For S. aureus 1500 mg q8h constitutes an optimal dosing schedule.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Cefuroxime/pharmacokinetics , Escherichia coli Infections/drug therapy , Klebsiella Infections/drug therapy , Staphylococcal Infections/drug therapy , Adult , Anti-Bacterial Agents/administration & dosage , Cefuroxime/administration & dosage , Escherichia coli/drug effects , Escherichia coli Infections/microbiology , Female , Healthy Volunteers , Humans , Klebsiella Infections/microbiology , Klebsiella pneumoniae/drug effects , Male , Microbial Sensitivity Tests , Middle Aged , Monte Carlo Method , Staphylococcal Infections/microbiology , Staphylococcus/drug effects , Streptococcus pneumoniae/drug effects , Young Adult
13.
Nat Rev Drug Discov ; 19(2): 131-148, 2020 02.
Article in English | MEDLINE | ID: mdl-31748707

ABSTRACT

Drug-induced liver injury (DILI) is a patient-specific, temporal, multifactorial pathophysiological process that cannot yet be recapitulated in a single in vitro model. Current preclinical testing regimes for the detection of human DILI thus remain inadequate. A systematic and concerted research effort is required to address the deficiencies in current models and to present a defined approach towards the development of new or adapted model systems for DILI prediction. This Perspective defines the current status of available models and the mechanistic understanding of DILI, and proposes our vision of a roadmap for the development of predictive preclinical models of human DILI.


Subject(s)
Chemical and Drug Induced Liver Injury/diagnosis , Disease Models, Animal , Drug-Related Side Effects and Adverse Reactions/prevention & control , Animals , Chemical and Drug Induced Liver Injury/etiology , Humans , Predictive Value of Tests
14.
Nucleic Acids Res ; 48(1): 63-74, 2020 01 10.
Article in English | MEDLINE | ID: mdl-31754711

ABSTRACT

The introduction of non-bridging phosphorothioate (PS) linkages in oligonucleotides has been instrumental for the development of RNA therapeutics and antisense oligonucleotides. This modification offers significantly increased metabolic stability as well as improved pharmacokinetic properties. However, due to the chiral nature of the phosphorothioate, every PS group doubles the amount of possible stereoisomers. Thus PS oligonucleotides are generally obtained as an inseparable mixture of a multitude of diastereoisomeric compounds. Herein, we describe the introduction of non-chiral 3' thiophosphate linkages into antisense oligonucleotides and report their in vitro as well as in vivo activity. The obtained results are carefully investigated for the individual parameters contributing to antisense activity of 3' and 5' thiophosphate modified oligonucleotides (target binding, RNase H recruitment, nuclease stability). We conclude that nuclease stability is the major challenge for this approach. These results highlight the importance of selecting meaningful in vitro experiments particularly when examining hitherto unexplored chemical modifications.


Subject(s)
Apolipoprotein B-100/genetics , Oligonucleotides/genetics , Phosphates/chemistry , Phosphorothioate Oligonucleotides/genetics , RNA, Long Noncoding/genetics , Animals , Apolipoprotein B-100/antagonists & inhibitors , Apolipoprotein B-100/metabolism , Cell Line, Tumor , Female , Humans , Kidney/cytology , Kidney/metabolism , Liver/cytology , Liver/metabolism , Lung/metabolism , Lung/pathology , Mice , Mice, Inbred C57BL , Oligonucleotides/chemical synthesis , Oligonucleotides/metabolism , Phosphates/metabolism , Phosphorothioate Oligonucleotides/chemical synthesis , Phosphorothioate Oligonucleotides/metabolism , RNA Stability , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/metabolism , Ribonuclease H/chemistry , Ribonuclease H/metabolism , Stereoisomerism
15.
Oncotarget ; 9(10): 9043-9060, 2018 Feb 06.
Article in English | MEDLINE | ID: mdl-29507673

ABSTRACT

Colorectal cancer (CRC) is a leading cause of death worldwide. Surgical intervention is a successful treatment for stage I patients, whereas other more advanced cases may require adjuvant chemotherapy. The selection of effective adjuvant treatments remains, however, challenging. Accurate patient stratification is necessary for the identification of the subset of patients likely responding to treatment, while sparing others from pernicious treatment. Targeted sequencing approaches may help in this regard, enabling rapid genetic investigation, and at the same time easily applicable in routine diagnosis. We propose a set of guidelines for the identification, including variant calling and filtering, of somatic mutations driving tumorigenesis in the absence of matched healthy tissue. We also discuss the inclusion criteria for the generation of our gene panel. Furthermore, we evaluate the prognostic impact of individual genes, using Cox regression models in the context of overall survival and disease-free survival. These analyses confirmed the role of commonly used biomarkers, and shed light on controversial genes such as CYP2C8. Applying those guidelines, we created a novel gene panel to investigate the onset and progression of CRC in 273 patients. Our comprehensive biomarker set includes 266 genes that may play a role in the progression through the different stages of the disease. Tracing the developmental state of the tumour, and its resistances, is instrumental in patient stratification and reliable decision making in precision clinical practice.

16.
Diabetes Ther ; 9(1): 373-382, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29143919

ABSTRACT

INTRODUCTION: The treatment of patients with type 2 diabetes uncontrolled on basal insulin and oral glucose-lowering drugs was investigated previously in the LixiLan-L trial. In the LixiLan-L trial, patients experienced a 6-week run-in with insulin glargine U100 (iGlar) as part of the screening phase, followed by treatment with a fixed-ratio combination of iGlar + lixisenatide (iGlarLixi) or iGlar alone over 30 weeks. In the study reported here, we investigated the achievement of glycemic control in those who completed the 30-week LixiLan-L trial, as assessed by change in glycated hemoglobin (HbA1c) levels from screening, both for the overall category and for screening HbA1c subcategories. METHODS: This post hoc analysis of the LixiLan-L trial included both the screening phase and the treatment period for 30-week completers and evaluated the change in HbA1c from screening to Week 30, patients reaching HbA1c < 7% at Week 30, and iGlar and lixisenatide (Lixi) doses at Week 30 overall and according to HbA1c subcategory at screening (HbA1c ≤ 8%, 8% < HbA1c ≤ 9%, and HbA1c > 9%). Documented symptomatic hypoglycemia during the treatment period was also assessed. RESULTS: HbA1c reductions (least squares mean) from screening to Week 30 were greater for iGlarLixi than iGlar, both overall (- 1.7 vs. - 1.1%) and in all subgroups (HbA1c ≤ 8%, 8% < HbA1c ≤ 9%, and HbA1c > 9%): - 1.1, - 1.4, - 2.4 (iGlarLixi) vs. - 0.5, - 1.0, - 1.8% (iGlar), respectively (all p < 0.0001). The end-of-treatment mean HbA1c level for iGlarLixi across all groups was < 7%. More patients achieved an HbA1c of < 7% with iGlarLixi than with iGlar, both overall (59.9 vs. 31.2%) and within each subgroup [74.2, 54.7, 52.2 (iGlarLixi) vs. 37.2, 31.6, 23.5% (iGlar), respectively]. A higher initial screening HbA1c corresponded with a greater mean reduction in HbA1c for both treatment strategies. In all HbA1c screening categories, the risk of hypoglycemia was not increased with iGlarLixi versus iGlar during the treatment phase. CONCLUSION: iGlarLixi controlled HbA1c levels more effectively than iGlar across all HbA1c screening subgroups and in the overall study population without increasing the risk of hypoglycemia. TRIAL REGISTRATION: Clinicaltrials.gov Identifier: NCT02058160. FUNDING: Sanofi.

17.
Expert Opin Drug Metab Toxicol ; 13(7): 767-782, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28604124

ABSTRACT

INTRODUCTION: The liver is an important target for drug-induced toxicities. Early detection of hepatotoxic drugs requires use of well-characterized test systems, yet current knowledge, gaps and limitations of tests employed remains an important issue for drug development. Areas Covered: The current state of the science, understanding and application of test systems in use for the detection of drug-induced cytotoxicity, mitochondrial toxicity, cholestasis and inflammation is summarized. The test systems highlighted herein cover mostly in vitro and some in vivo models and endpoint measurements used in the assessment of small molecule toxic liabilities. Opportunities for research efforts in areas necessitating the development of specific tests and improved mechanistic understanding are highlighted. Expert Opinion: Use of in vitro test systems for safety optimization will remain a core activity in drug discovery. Substantial inroads have been made with a number of assays established for human Drug-induced Liver Injury. There nevertheless remain significant gaps with a need for improved in vitro tools and novel tests to address specific mechanisms of human Drug-Induced Liver Injury. Progress in these areas will necessitate not only models fit for application, but also mechanistic understanding of how chemical insult on the liver occurs in order to identify translational and quantifiable readouts for decision-making.


Subject(s)
Chemical and Drug Induced Liver Injury/etiology , Drug Discovery/methods , Toxicity Tests/methods , Animals , Chemical and Drug Induced Liver Injury/physiopathology , Drug Design , Drug-Related Side Effects and Adverse Reactions/diagnosis , Humans , Models, Biological , Risk Assessment/methods
18.
Drug Metab Dispos ; 45(1): 68-75, 2017 01.
Article in English | MEDLINE | ID: mdl-27737930

ABSTRACT

Inclusion of a microdose of 14C-labeled drug in the first-in-man study of new investigational drugs and subsequent analysis by accelerator mass spectrometry has become an integrated part of drug development at Lundbeck. It has been found to be highly informative with regard to investigations of the routes and rates of excretion of the drug and the human metabolite profiles according to metabolites in safety testing guidance and also when additional metabolism-related issues needed to be addressed. In the first-in-man study with the NCE Lu AF09535, contrary to anticipated, surprisingly low exposure was observed when measuring the parent compound using conventional bioanalysis. Parallel accelerator mass spectrometry analysis revealed that the low exposure was almost exclusively attributable to extensive metabolism. The metabolism observed in humans was mediated via a human specific metabolic pathway, whereas an equivalent extent of metabolism was not observed in preclinical species. In vitro, incubation studies in human liver cytosol revealed involvement of aldehyde oxidase (AO) in the biotransformation of Lu AF09535. In vivo, substantially lower plasma exposure of Lu AF09535 was observed in chimeric mice with humanized livers compared with control animals. In addition, Lu AF09535 exhibited very low oral bioavailability in monkeys despite relatively low clearance after intravenous administration in contrast to the pharmacokinetics in rats and dogs, both showing low clearance and high bioavailability. The in vitro and in vivo methods applied were proved useful for identifying and evaluating AO-dependent metabolism. Different strategies to integrate these methods for prediction of in vivo human clearance of AO substrates were evaluated.


Subject(s)
Aldehyde Oxidase/metabolism , Drugs, Investigational/pharmacokinetics , Liver/metabolism , Animals , Biological Availability , Biotransformation , Carbon Radioisotopes , Cytosol/metabolism , Double-Blind Method , Drug Evaluation, Preclinical , Drugs, Investigational/administration & dosage , Female , Healthy Volunteers , Humans , In Vitro Techniques , Liver/enzymology , Macaca fascicularis , Male , Mice , Species Specificity
19.
PLoS One ; 10(8): e0134503, 2015.
Article in English | MEDLINE | ID: mdl-26263558

ABSTRACT

We established a selection strategy to identify new models for an altered airway inflammatory response from a large compendium of mutant mouse lines that were systemically phenotyped in the German Mouse Clinic (GMC). As selection criteria we included published gene functional data, as well as immunological and transcriptome data from GMC phenotyping screens under standard conditions. Applying these criteria we identified a few from several hundred mutant mouse lines and further characterized the Cox4i2tm1Hutt, Ifit2tm1.1Ebsb, and Prdm11tm1.1ahl lines following ovalbumin (OVA) sensitization and repeated OVA airway challenge. Challenged Prdm11tm1.1ahl mice exhibited changes in B cell counts, CD4+ T cell counts, and in the number of neutrophils in bronchoalveolar lavages, whereas challenged Ifit2tm1.1Ebsb mice displayed alterations in plasma IgE, IgG1, IgG3, and IgM levels compared to the challenged wild type littermates. In contrast, challenged Cox4i2tm1Hutt mutant mice did not show alterations in the humoral or cellular immune response compared to challenged wild type mice. Transcriptome analyses from lungs of the challenged mutant mouse lines showed extensive changes in gene expression in Prdm11tm1.1ahl mice. Functional annotations of regulated genes of all three mutant mouse lines were primarily related to inflammation and airway smooth muscle (ASM) remodeling. We were thus able to define an effective selection strategy to identify new candidate genes for the predisposition to an altered airway inflammatory response under OVA challenge conditions. Similar selection strategies may be used for the analysis of additional genotype-envirotype interactions for other diseases.


Subject(s)
Carrier Proteins/genetics , Mutation , Proteins/genetics , Animals , Apoptosis Regulatory Proteins , Bronchoalveolar Lavage Fluid/cytology , Bronchoalveolar Lavage Fluid/immunology , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Genetic Predisposition to Disease , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulins/blood , Immunoglobulins/immunology , Inflammation/genetics , Inflammation/immunology , Lymphocyte Activation/immunology , Mice , Molecular Sequence Annotation , Ovalbumin/immunology , RNA-Binding Proteins , Respiratory Tract Diseases/genetics , Respiratory Tract Diseases/immunology , Selection, Genetic , Transcription Factors , Transcriptome
20.
Blood ; 125(8): 1272-81, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25499759

ABSTRACT

The PR-domain (PRDM) family of genes encodes transcriptional regulators, several of which are deregulated in cancer. By using a functional screening approach, we sought to identify novel tumor suppressors among the PRDMs. Here we demonstrate oncogenic collaboration between depletion of the previously uncharacterized PR-domain family member Prdm11 and overexpression of MYC. Overexpression of PRDM11 inhibits proliferation and induces apoptosis. Prdm11 knockout mice are viable, and loss of Prdm11 accelerates MYC-driven lymphomagenesis in the Eµ-Myc mouse model. Moreover, we show that patients with PRDM11-deficient diffuse large B-cell lymphomas (DLBCLs) have poorer overall survival and belong to the nongerminal center B-cell-like subtype. Mechanistically, genome-wide mapping of PRDM11 binding sites coupled with transcriptome sequencing in human DLBCL cells evidenced that PRDM11 associates with transcriptional start sites of target genes and regulates important oncogenes such as FOS and JUN. Hence, we characterize PRDM11 as a putative novel tumor suppressor that controls the expression of key oncogenes, and we add new mechanistic insight into B-cell lymphomagenesis.


Subject(s)
Carrier Proteins/genetics , Cell Transformation, Neoplastic/genetics , Lymphoma/genetics , Proto-Oncogene Proteins c-myc/physiology , Animals , Cells, Cultured , Embryo, Mammalian , Gene Deletion , Gene Expression Regulation, Neoplastic/physiology , Gene Knockout Techniques , HEK293 Cells , HeLa Cells , Humans , Lymphoma/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Mice , Molecular Sequence Data , Transcription Factors , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...