Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
Gut ; 73(8): 1343-1349, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38418210

ABSTRACT

BACKGROUND: Dynamic changes in non-invasive tests, such as changes in alanine aminotransferase (ALT) and MRI proton-density-fat-fraction (MRI-PDFF), may help to detect metabolic dysfunction-associated steatohepatitis (MASH) resolution, but a combination of non-invasive tests may be more accurate than either alone. We developed a novel non-invasive score, the MASH Resolution Index, to detect the histological resolution of MASH. METHODS: This study included a derivation cohort of 95 well-characterised adult participants (67% female) with biopsy-confirmed MASH who underwent contemporaneous laboratory testing, MRI-PDFF and liver biopsy at two time points. The primary objective was to develop a non-invasive score to detect MASH resolution with no worsening of fibrosis. The most predictive logistic regression model was selected based on the highest area under the receiver operating curve (AUC), and the lowest Akaike information criterion and Bayesian information criterion. The model was then externally validated in a distinct cohort of 163 participants with MASH from a clinical trial. RESULTS: The median (IQR) age and body mass index were 55 (45-62) years and 32.0 (30-37) kg/m2, respectively, in the derivation cohort. The most accurate model (MASH Resolution Index) included MRI-PDFF, ALT and aspartate aminotransferase. The index had an AUC of 0.81 (95% CI 0.69 to 0.93) for detecting MASH resolution in the derivation cohort. The score calibrated well and performed robustly in a distinct external validation cohort (AUC 0.83, 95% CI 0.76 to 0.91), and outperformed changes in ALT and MRI-PDFF. CONCLUSION: The MASH Resolution Index may be a useful score to non-invasively identify MASH resolution.


Subject(s)
Liver , Magnetic Resonance Imaging , Humans , Female , Middle Aged , Male , Magnetic Resonance Imaging/methods , Liver/pathology , Liver/diagnostic imaging , Biopsy , Alanine Transaminase/blood , Non-alcoholic Fatty Liver Disease/pathology , Non-alcoholic Fatty Liver Disease/diagnostic imaging , Non-alcoholic Fatty Liver Disease/diagnosis , ROC Curve , Fatty Liver/diagnostic imaging , Fatty Liver/pathology , Fatty Liver/diagnosis
2.
N Engl J Med ; 389(11): 998-1008, 2023 Sep 14.
Article in English | MEDLINE | ID: mdl-37356033

ABSTRACT

BACKGROUND: Pegozafermin is a long-acting glycopegylated (pegylated with the use of site-specific glycosyltransferases) fibroblast growth factor 21 (FGF21) analogue in development for the treatment of nonalcoholic steatohepatitis (NASH) and severe hypertriglyceridemia. The efficacy and safety of pegozafermin in patients with biopsy-proven noncirrhotic NASH are not well established. METHODS: In this phase 2b, multicenter, double-blind, 24-week, randomized, placebo-controlled trial, we randomly assigned patients with biopsy-confirmed NASH and stage F2 or F3 (moderate or severe) fibrosis to receive subcutaneous pegozafermin at a dose of 15 mg or 30 mg weekly or 44 mg once every 2 weeks or placebo weekly or every 2 weeks. The two primary end points were an improvement in fibrosis (defined as reduction by ≥1 stage, on a scale from 0 to 4, with higher stages indicating greater severity), with no worsening of NASH, at 24 weeks and NASH resolution without worsening of fibrosis at 24 weeks. Safety was also assessed. RESULTS: Among the 222 patients who underwent randomization, 219 received pegozafermin or placebo. The percentage of patients who met the criteria for fibrosis improvement was 7% in the pooled placebo group, 22% in the 15-mg pegozafermin group (difference vs. placebo, 14 percentage points; 95% confidence interval [CI], -9 to 38), 26% in the 30-mg pegozafermin group (difference, 19 percentage points; 95% CI, 5 to 32; P = 0.009), and 27% in the 44-mg pegozafermin group (difference, 20 percentage points; 95% CI, 5 to 35; P = 0.008). The percentage of patients who met the criteria for NASH resolution was 2% in the placebo group, 37% in the 15-mg pegozafermin group (difference vs. placebo, 35 percentage points; 95% CI, 10 to 59), 23% in the 30-mg pegozafermin group (difference, 21 percentage points; 95% CI, 9 to 33), and 26% in the 44-mg pegozafermin group (difference, 24 percentage points; 95% CI, 10 to 37). The most common adverse events associated with pegozafermin therapy were nausea and diarrhea. CONCLUSIONS: In this phase 2b trial, treatment with pegozafermin led to improvements in fibrosis. These results support the advancement of pegozafermin into phase 3 development. (Funded by 89bio; ENLIVEN ClinicalTrials.gov number, NCT04929483.).


Subject(s)
Fibroblast Growth Factors , Fibrosis , Gastrointestinal Agents , Non-alcoholic Fatty Liver Disease , Humans , Biopsy , Double-Blind Method , Fibroblast Growth Factors/analogs & derivatives , Fibrosis/drug therapy , Fibrosis/etiology , Fibrosis/pathology , Gastrointestinal Agents/administration & dosage , Gastrointestinal Agents/therapeutic use , Injections, Subcutaneous , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Treatment Outcome
3.
Lancet Gastroenterol Hepatol ; 7(7): 603-616, 2022 07.
Article in English | MEDLINE | ID: mdl-35325622

ABSTRACT

BACKGROUND: Non-alcoholic steatohepatitis (NASH) is characterised by hepatic steatosis, inflammation, and injury, and is associated with an increased risk of liver transplantation and death. NASH affects more than 16 million people in the USA, and there is no approved therapy. The aim of this study was to evaluate the safety and efficacy of aldafermin, an engineered analogue of the gut hormone fibroblast growth factor 19 (FGF19). METHODS: In this randomised, double-blind, placebo-controlled, phase 2b study (ALPINE 2/3) in patients with biopsy-confirmed NASH and stage 2 or 3 fibrosis, we randomly assigned patients stratified by fibrosis stage in a 1:1:1:1 ratio to receive placebo, aldafermin 0·3 mg, 1·0 mg, or 3·0 mg once daily for 24 weeks at 30 study sites in the USA. Patients, investigators, the funder, and all other staff, were masked to treatment assignment throughout the study. The primary endpoint was an improvement in liver fibrosis of at least one stage with no worsening of NASH at week 24. Analyses were done by intention-to-treat. This trial is registered with ClinicalTrials.gov, number NCT03912532, and has been completed. FINDINGS: Between May 16, 2019, and Sept 4, 2020, 786 patients were screened, of whom 171 were randomly assigned to a treatment group and included in the intention-to-treat population: 43 in the 0·3 mg aldafermin group, 42 in the 1·0 mg group, 43 in the 3·0 mg group, and 43 in the placebo group. In total, 145 (85%) of patients completed treatment. At week 24, among patients with biopsies at both baseline and week 24, was seven (19%) of 36 patients in the placebo group, 11 (31%) of 36 in the 0·3 mg aldafermin group (difference 90% CI 12% [-9 to 33]; p=0·11), five (15%) of 34 patients in the 1·0 mg group (difference -5% [-24 to 13]; p=0·80), and 11 (30%) of 37 patients in the 3·0 mg group (difference 10% [-9 to 30]; p=0·12) had an improvement in liver fibrosis of at least one stage with no worsening of NASH, without meeting the prespecified significance for dose response (p=0·55). Adverse events were mostly mild or moderate in severity. Diarrhoea occurred in six (14%) of 43 patients in the placebo group, three (7%) of 43 patients in the 0·3 mg aldafermin group, five (12%) of 41 patients in the 1·0 mg group, and ten (23%) of 43 patients in the 3·0 mg group. Incidences of serious adverse events and discontinuations owing to adverse events were similar between groups. INTERPRETATION: Aldafermin was generally well tolerated but did not produce a significant dose response on fibrosis improvement of at least one stage with no worsening of NASH, despite positive effects on a number of secondary endpoints. The findings of this trial may have implications for the design of future NASH trials. FUNDING: NGM Biopharmaceuticals.


Subject(s)
Fibroblast Growth Factors , Non-alcoholic Fatty Liver Disease , Double-Blind Method , Fibroblast Growth Factors/analogs & derivatives , Fibroblast Growth Factors/therapeutic use , Humans , Liver Cirrhosis/complications , Liver Cirrhosis/drug therapy , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Treatment Outcome
4.
Mov Disord ; 36(10): 2408-2412, 2021 10.
Article in English | MEDLINE | ID: mdl-34191352

ABSTRACT

BACKGROUND: Solriamfetol is approved (US and EU) for excessive daytime sleepiness (EDS) in narcolepsy and obstructive sleep apnea. OBJECTIVES: Evaluate solriamfetol safety/efficacy for EDS in Parkinson's disease (PD). METHODS: Phase 2, double-blind, 4-week, crossover trial: adults with PD and EDS were randomized to sequence A (placebo, solriamfetol 75, 150, 300 mg/d), B (solriamfetol 75, 150, 300 mg/d, placebo), or C (placebo). Outcomes (safety/tolerability [primary]; Epworth Sleepiness Scale [ESS]; Maintenance of Wakefulness Test [MWT]) were assessed weekly. P values are nominal. RESULTS: Common adverse events (n = 66): nausea (10.7%), dizziness (7.1%), dry mouth (7.1%), headache (7.1%), anxiety (5.4%), constipation (5.4%), dyspepsia (5.4%). ESS decreased both placebo (-4.78) and solriamfetol (-4.82 to -5.72; P > 0.05). MWT improved dose-dependently with solriamfetol, increasing by 5.05 minutes with 300 mg relative to placebo (P = 0.0098). CONCLUSIONS: Safety/tolerability was consistent with solriamfetol's known profile. There were no significant improvements on ESS; MWT results suggest possible benefit with solriamfetol in PD. © 2021 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Carbamates/therapeutic use , Disorders of Excessive Somnolence , Parkinson Disease , Phenylalanine/therapeutic use , Adult , Disorders of Excessive Somnolence/drug therapy , Disorders of Excessive Somnolence/etiology , Double-Blind Method , Humans , Parkinson Disease/complications , Parkinson Disease/drug therapy , Phenylalanine/analogs & derivatives
5.
Gastroenterology ; 160(1): 219-231.e1, 2021 01.
Article in English | MEDLINE | ID: mdl-32781086

ABSTRACT

BACKGROUND & AIMS: Aldafermin, an engineered analog of fibroblast growth factor 19, inhibits bile acid synthesis and regulates metabolic homeostasis. We report results from a 24-week, phase 2 study, with serial liver biopsies, of patients with nonalcoholic steatohepatitis (NASH). METHODS: We performed a double-blind study of 78 patients with NASH at 9 centers in the United States. Key inclusion criteria were biopsy-proven NASH with Nonalcoholic Fatty Liver Disease Activity Score ≥4, stage 2 or 3 fibrosis by NASH Clinical Research Network classification, and absolute liver fat content ≥8%, measured by magnetic resonance imaging-proton density fat fraction. Patients were randomly assigned (1:2) to groups given subcutaneous placebo (n = 25) or aldafermin 1 mg (n = 53) daily for 24 weeks. The primary outcome was change in absolute liver fat content from baseline at week 24. Secondary outcomes included serum markers and histologic measures of fibrosis improvement and NASH resolution. RESULTS: At week 24, the aldafermin group had a significant reduction in absolute liver fat content (reduction of 7.7%) compared with placebo (reduction of 2.7%; difference, reduction of 5.0%; 95% confidence interval, reduction of 8.0%-1.9%; P = .002). Aldafermin produced significantly greater decreases in levels of 7α-hydroxy-4-cholesten-3-one, bile acids, alanine and aspartate aminotransferases, and neoepitope-specific N-terminal pro-peptide of type III collagen (Pro-C3) than placebo. Fibrosis improvement (≥1 stage) with no worsening of NASH was achieved in 38% of patients receiving aldafermin vs 18% of patients receiving placebo (P = .10). NASH resolution with no worsening of fibrosis was observed in 24% of patients given aldafermin vs 9% of patients given placebo (P = .20). Discontinuations due to adverse events occurred in no patients in the aldafermin group and 4% of patients in the placebo group. CONCLUSIONS: In a phase 2 trial of patients with NASH, aldafermin reduced liver fat and produced a trend toward fibrosis improvement. ClinicalTrials.gov, Number: NCT02443116.


Subject(s)
Fibroblast Growth Factors/analysis , Fibroblast Growth Factors/therapeutic use , Liver Cirrhosis/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Adult , Aged , Double-Blind Method , Female , Humans , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Treatment Outcome
6.
Contemp Clin Trials ; 47: 356-65, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26944023

ABSTRACT

BACKGROUND: Non-alcoholic steatohepatitis (NASH) is often accompanied by liver fibrosis, which can progress to cirrhosis; C-C chemokine receptors type 2 and 5 (CCR2/CCR5), which mediate interactions driving inflammation and fibrosis, are promising treatment targets. Cenicriviroc (CVC), a dual-CCR2/CCR5 antagonist, has potent anti-inflammatory and antifibrotic activity in animal models; in HIV-positive subjects it reduced soluble CD14 levels, aspartate aminotransferase-to-platelet count ratio index, and non-invasive hepatic fibrosis risk scores; favorable tolerability was demonstrated in ~600 subjects. Efficacy and safety of CVC 150 mg for treating NASH with liver fibrosis are being evaluated over 2 years (primary endpoint at Year 1 [Y1]). DESIGN: Phase 2b, randomized, double-blind, placebo-controlled, multinational study (CENTAUR; NCT02217475). Adults with histological evidence of NASH, non-alcoholic fatty liver disease activity score (NAS) ≥ 4, and liver fibrosis (stages 1-3 NASH clinical research network system) enrolled. Subjects have increased risk of progression to cirrhosis due to ≥1 characteristic: type 2 diabetes; body mass index > 25 kg/m(2) with ≥1 feature of metabolic syndrome; bridging fibrosis and/or NAS ≥ 5. Liver biopsy evaluation at Screening, Y1, and Year 2 (Y2). OBJECTIVES: Assess histologic improvement (≥2-point in NAS with ≥1-point improvement in >1 category) without worsening of fibrosis at Y1 (primary); evaluate complete NASH resolution without worsening of fibrosis at Y2 (key secondary). DISCUSSION: CENTAUR is the first prospective study evaluating an oral agent exclusively enrolling subjects with NASH and liver fibrosis, with increased risk of developing cirrhosis. It will compare shorter versus longer CVC treatment and assess correlations between decreased inflammation and fibrosis.


Subject(s)
CCR5 Receptor Antagonists/therapeutic use , Imidazoles/therapeutic use , Liver Cirrhosis/complications , Non-alcoholic Fatty Liver Disease/drug therapy , Administration, Oral , Adolescent , Adult , Aged , Clinical Protocols , Double-Blind Method , Drug Administration Schedule , Female , Follow-Up Studies , Humans , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/complications , Prospective Studies , Research Design , Sulfoxides , Treatment Outcome , Young Adult
7.
Clin Pharmacol Drug Dev ; 4(2): 121-9, 2015 03.
Article in English | MEDLINE | ID: mdl-27128216

ABSTRACT

Ranolazine and metformin may be frequently co-administered in subjects with chronic angina and co-morbid type 2 diabetes mellitus (T2DM). The potential for a drug-drug interaction was explored in two phase 1 clinical studies in subjects with T2DM to evaluate the pharmacokinetics and safety of metformin 1000 mg BID when administered with ranolazine 1000 mg BID (Study 1, N = 28) or ranolazine 500 mg BID (Study 2, N = 25) as compared to metformin alone. Co-administration of ranolazine 1000 mg BID with metformin 1000 mg BID resulted in 1.53- and 1.79-fold increases in steady-state metformin Cmax and AUCtau , respectively; co-administration of ranolazine 500 mg BID with metformin 1000 mg BID resulted in 1.22- and 1.37-fold increases in steady-state metformin Cmax and AUCtau , respectively. Co-administration of ranolazine and metformin was well tolerated in these T2DM subjects, with no serious adverse events or drug-related adverse events leading to discontinuation. The most common adverse events were nausea, diarrhea, and dizziness. These findings are consistent with a dose-related interaction between ranolazine and metformin, and suggest that a dose adjustment of metformin may not be required with ranolazine 500 mg BID; whereas, the metformin dose should not exceed 1700 mg of total daily dose when using ranolazine 1000 mg BID.


Subject(s)
Angina Pectoris/drug therapy , Cardiovascular Agents/pharmacokinetics , Diabetes Mellitus, Type 2/drug therapy , Hypoglycemic Agents/pharmacokinetics , Metformin/pharmacokinetics , Ranolazine/pharmacokinetics , Adult , Aged , Angina Pectoris/blood , Angina Pectoris/diagnosis , Angina Pectoris/epidemiology , Animals , Area Under Curve , CHO Cells , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/adverse effects , Chronic Disease , Comorbidity , Cricetulus , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/epidemiology , Double-Blind Method , Drug Interactions , Female , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/adverse effects , Male , Metabolic Clearance Rate , Metformin/administration & dosage , Metformin/adverse effects , Middle Aged , Organic Cation Transporter 2/drug effects , Organic Cation Transporter 2/genetics , Organic Cation Transporter 2/metabolism , Polypharmacy , Ranolazine/administration & dosage , Ranolazine/adverse effects , United States
8.
Ann Neurol ; 69(6): 919-27, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21681795

ABSTRACT

Existing and emerging strategies for managing L-dopa-induced dyskinesias (LIDs) in patients with Parkinson disease have involved either delaying the introduction of L-dopa therapy, treatment with an antidyskinetic agent, using a therapy or delivery system that can provide continuous dopaminergic stimulation, or using novel agents that target receptors implicated in the mechanisms underlying LIDs. Treatment with dopamine agonists such as pramipexole or ropinirole allows levodopa to be delayed, but once levodopa is added to the drug regimen the usual course of onset of dyskinesias is observed. Amantadine, an N-methyl-D-aspartate antagonist, is so far the only approved compound with evidence of providing a sustained antidyskinetic benefit in the absence of unacceptable side effects. These findings support the hypothesis of glutamate overactivity in the development of dyskinesias. More continuous delivery of dopaminergic medication, such as through intraintestinal or subcutaneous routes, is promising but invasive and associated with injection site reactions. As a result of molecular research and elucidation of the role of a variety of neurotransmitters in the mechanism of LIDs, new compounds have been identified, including those that modulate the direct and indirect striatal output pathways; some of these new agents are in the early stages of development or undergoing proof-of-concept evaluation as antidyskinetic agents.


Subject(s)
Antiparkinson Agents/adverse effects , Dopamine/metabolism , Dyskinesia, Drug-Induced/etiology , Dyskinesia, Drug-Induced/therapy , Parkinson Disease/drug therapy , Animals , Dopamine Agents/therapeutic use , Drug Delivery Systems , Humans , Transcranial Magnetic Stimulation
9.
Clin Cardiol ; 33(6): 330-6, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20556802

ABSTRACT

BACKGROUND: In patients with heart failure, few data describe the neurohormonal response to nesiritide and furosemide either alone or in combination. This study systematically compared the effects of nesiritide, furosemide, and their combination on natriuresis/diuresis and plasma aldosterone in patients with chronic stable heart failure who were relatively diuretic resistant. HYPOTHESIS: Natriuretic, diuretic, and neurohormonal responses to furosemide and nesiritide will differ when these agents are administered alone vs. in combination. METHODS: Twenty-eight subjects completed a multicenter, open-label, three-arm crossover study. Each subject received the following treatments in random order on alternate days: (1) furosemide, 40 mg intravenous bolus; (2) nesiritide, 2 microg/kg intravenous bolus followed by a 0.01 microg/kg/min infusion for 6 hours; (3) both furosemide and nesiritide, with furosemide given at least 15 minutes after initiation of nesiritide. RESULTS: Plasma aldosterone increased by 2.2 +/- 1.6 ng/dL after furosemide alone, decreased by 3.9 +/- 1.6 ng/dL after nesiritide alone (P = 0.005 vs furosemide alone and P = 0.56 vs furosemide plus nesiritide), and decreased by 2.8 +/- 1.6 ng/dL after furosemide plus nesiritide (P = 0.02 vs furosemide alone). CONCLUSIONS: Furosemide alone produced natriuresis/diuresis and a prompt rise in plasma aldosterone values. Nesiritide alone produced no significant natriuresis/diuresis, but decreased plasma aldosterone values. When furosemide was administered on a background of nesiritide infusion, the observed natriuresis/diuresis was similar to that seen with furosemide alone, without the anticipated increase in plasma aldosterone observed with furosemide alone.


Subject(s)
Aldosterone/blood , Furosemide/therapeutic use , Heart Failure/drug therapy , Natriuresis/drug effects , Natriuretic Agents/therapeutic use , Natriuretic Peptide, Brain/therapeutic use , Sodium Potassium Chloride Symporter Inhibitors/therapeutic use , Ventricular Dysfunction/drug therapy , Ventricular Function , Adult , Aged , Biomarkers/blood , Cross-Over Studies , Drug Resistance , Drug Therapy, Combination , Female , Furosemide/administration & dosage , Heart Failure/complications , Heart Failure/physiopathology , Humans , Infusions, Intravenous , Injections, Intravenous , Male , Middle Aged , Natriuretic Agents/administration & dosage , Natriuretic Peptide, Brain/administration & dosage , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Systole , Time Factors , Treatment Outcome , United States , Ventricular Dysfunction/etiology , Ventricular Dysfunction/physiopathology
10.
Drugs Today (Barc) ; 44(7): 531-45, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18806903

ABSTRACT

Rasagiline, a selective COMT inhibitor, and rotigotine, a transdermal dopamine (D2) agonist, are two new agents that have been approved in the U.S. and Europe for the treatment of Parkinson's disease. Rasagiline is approved in the U.S. for both monotherapy and as an adjunct to levodopa. Its role in preventing disease progression has yet to be proven, but a large-scale study (ADAGIO) is under way. Rotigotine is approved for early-stage disease in Europe and the U.S. but is only approved in Europe for late-stage disease. It has recently been recalled due to the formation of insoluble crystals that interfere with absorption and may reduce its efficacy. Measures are being taken by the manufacturer to solve this problem. Istradefylline, and adenosine receptor antagonist, showed early promise but efficacy has not been demonstrated consistently, possibly due to higher than expected placebo effect. This has resulted in a nonapprovable letter from the FDA. With regard to perampanel, additional studies are needed to demonstrate safety and efficacy. Sanifamide and pardoprunox are agents that target multiple receptors that may modulate dyskinesia and other nonmotor symptoms in addition to motor symptoms, but phase III data are not yet available. Lusuride is an older dopamine agonist that has been reformulated as a transdermal patch and as a subcutaneous injection and may offer advantages in refractory patients with motor fluctuations. Sphermaine is a novel cell therapy designed to provide a localized source of levodopa directly to the brain. Gene therapies including AAV-GAD, AAV-AADC and AAV2-neurturin are in early stages of development in patients with advanced-stage disease but early safety data are promising.


Subject(s)
Antiparkinson Agents/therapeutic use , Parkinson Disease/drug therapy , Genetic Therapy , Humans , Parkinson Disease/therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...