Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 338
Filter
1.
BMJ Open ; 14(7): e081623, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38991669

ABSTRACT

INTRODUCTION: Patients with clinically significant portal hypertension (CSPH) are recommended to be treated with non-selective beta-blockers (ie, carvedilol) to prevent the first hepatic decompensation event by the renewing Baveno VII consensus. CSPH is defined by hepatic venous pressure gradient (HVPG)≥10 mm Hg; however, the HVPG measurement is not widely adopted due to its invasiveness. Liver stiffness (LS)≥25 kPa can be used as a surrogate of HVPG≥10 mm Hg to rule in CSPH with 90% of the positive predicting value in majority aetiologies of patients. A compelling argument is existing for using LS≥25 kPa to diagnose CSPH and then to initiate carvedilol in patients with compensated cirrhosis, and about 5%-6% of patients under this diagnosis criteria may not be benefited from carvedilol and are at risk of lower heart rate and mean arterial pressure. Randomised controlled trial on the use of carvedilol to prevent liver decompensation in CSPH diagnosed by LS remains to elucidate. Therefore, we aimed to investigate if compensated cirrhosis patients with LS≥25 kPa may benefit from carvedilol therapy. METHODS AND ANALYSIS: This study is a randomised, double-blind, placebo-controlled, multicentre trial. We will randomly assign 446 adult compensated cirrhosis patients with LS≥25 kPa and without any previous decompensated event and without high-risk gastro-oesophageal varices. Patients are randomly divided into two groups, with 223 subjects in group A and 223 subjects in group B. Group A is a carvedilol intervention group, while group B is a placebo group. All patients in both groups will receive aetiology therapies and are followed up at an interval of 6 months. The 3-year incidences of decompensated events of cirrhosis-related and liver-related death are the primary outcome. The secondary outcomes include development of each complication of portal hypertension individually (ascites, variceal bleeding or overt hepatic encephalopathy), development of spontaneous bacterial peritonitis and other bacterial infections, development of new varices, growth of small varices to large varices, delta changes in LS and spleen stiffness, change in hepatic dysfunction assessed by Child-Pugh and model for end-stage liver disease score, change in platelet count, development of hepatocellular carcinoma, development of portal vein thrombosis and adverse events with a 3-year follow-up. A predefined interim analysis will be performed to ensure that the calculation is reasonable. ETHICS AND DISSEMINATION: The study protocol has been approved by the ethics committees of the Sixth People's Hospital of Shenyang (2023-05-003-01) and independent ethics committee for clinical research of Zhongda Hospital, affiliated to Southeast University (2023ZDSYLL433-P01). The results from this trial will be submitted for publication in peer-reviewed journals and will be presented at international conferences. TRIAL REGISTRATION NUMBER: ChiCTR2300073864.


Subject(s)
Carvedilol , Hypertension, Portal , Liver Cirrhosis , Carvedilol/therapeutic use , Carvedilol/pharmacology , Humans , Hypertension, Portal/drug therapy , Hypertension, Portal/etiology , Liver Cirrhosis/complications , Double-Blind Method , China/epidemiology , Multicenter Studies as Topic , Randomized Controlled Trials as Topic , Adrenergic beta-Antagonists/therapeutic use , Female , Liver/drug effects , Liver/physiopathology , Portal Pressure/drug effects , Esophageal and Gastric Varices/etiology , Esophageal and Gastric Varices/prevention & control , Elasticity Imaging Techniques , Adult , Male
2.
Transplantation ; 105(2): 346-353, 2021 02 01.
Article in English | MEDLINE | ID: mdl-32235258

ABSTRACT

BACKGROUND: Portopulmonary hypertension is present in an estimated 5.3% to 8.5% of liver transplant candidates. Untreated, 5-year survival is estimated between 14% and 28%. Moderate-severe disease is a contraindication to liver transplant due to the high perioperative mortality, but patients optimized with pulmonary vasodilator therapy can become eligible for transplant. There is minimal data regarding posttransplant outcomes and ability to discontinue pulmonary vasodilator therapy posttransplant. METHODS: We performed a single-center retrospective analysis to evaluate long-term outcomes of patients with moderate-severe portopulmonary hypertension who were optimized with pulmonary vasodilator therapy, became eligible for liver transplant, and subsequently underwent transplant. We identified 24 patients optimized with pulmonary vasodilator therapy who underwent subsequent liver transplantation and 25 patients who were treated with pulmonary vasodilator therapy alone. RESULTS: In the transplanted cohort, 1-year survival from portopulmonary hypertension diagnosis date: 95.8%, 3-year survival: 90.9%, and 5-year survival: 90.9%. Posttransplant; 1-, 3-, and 5-year survival was 86.9%. Among transplanted patients, 41.6% (10/24) were optimized with nonparenteral therapy. Following transplantation, 100% (14/14) of the surviving patients were able to discontinue parenteral therapy; median time: 7.2 months (interquartile range: 5.1-8.9 mo), while 61.9% (13/21) were able to discontinue pulmonary vasodilator therapy altogether; median time: 13.9 months (interquartile range: 5.1-17.6 mo). CONCLUSIONS: Patients who are optimized with pulmonary vasodilator therapy before liver transplant can have excellent long-term outcomes posttransplant. Oral pulmonary vasodilator therapy can be effective treatment to qualify a patient for transplant, and the majority are able to wean from pulmonary vasodilator therapy entirely posttransplant.


Subject(s)
Antihypertensive Agents/administration & dosage , Arterial Pressure/drug effects , End Stage Liver Disease/surgery , Hypertension, Portal/drug therapy , Liver Transplantation , Portal Pressure/drug effects , Pulmonary Arterial Hypertension/drug therapy , Pulmonary Artery/drug effects , Vasodilator Agents/administration & dosage , Administration, Oral , End Stage Liver Disease/diagnosis , End Stage Liver Disease/mortality , End Stage Liver Disease/physiopathology , Female , Humans , Hypertension, Portal/diagnosis , Hypertension, Portal/mortality , Hypertension, Portal/physiopathology , Liver Transplantation/adverse effects , Liver Transplantation/mortality , Male , Middle Aged , Pulmonary Arterial Hypertension/diagnosis , Pulmonary Arterial Hypertension/mortality , Pulmonary Arterial Hypertension/physiopathology , Pulmonary Artery/physiopathology , Retrospective Studies , Time Factors , Treatment Outcome
3.
J Hepatol ; 74(5): 1188-1199, 2021 05.
Article in English | MEDLINE | ID: mdl-33278455

ABSTRACT

BACKGROUND & AIMS: In advanced chronic liver disease (ACLD), deregulated hepatic necroinflammatory processes play a key role in the development of liver microvascular dysfunction, fibrogenesis, and increased hepatic vascular tone, resulting in progression of ACLD and portal hypertension. Given the current lack of an effective treatment, we aimed to characterise the effects of the pan-peroxisome proliferator-activated receptor (pan-PPAR) agonist lanifibranor in 2 preclinical models of ACLD, as well as in liver cells from patients with ACLD. METHODS: Cirrhotic rats (thioacetamide or common bile duct ligation; TAA or cBDL) randomly received lanifibranor (100 mg/kg/day, po) or vehicle for 14 days (n = 12/group). PPAR expression, systemic and hepatic haemodynamics, presence of ascites, liver sinusoidal endothelial cell (LSEC) phenotype, hepatic stellate cell (HSC) activation, serum transaminases and albumin, hepatic macrophage infiltration, cytokine expression, and liver fibrosis were determined. Hepatic cells were isolated from the livers of patients with cirrhosis and their phenotype was evaluated after treatment with either lanifibranor or vehicle. RESULTS: TAA-cirrhotic rats receiving lanifibranor showed significantly lower portal pressure compared with vehicle-treated animals (-15%; p = 0.003) without decreasing portal blood flow, indicating improved hepatic vascular resistance. Moreover, lanifibranor-treated TAA-rats showed decreased ascites, improved LSEC and HSC phenotypes, ameliorated hepatic microvascular function, reduced hepatic inflammation, and significant fibrosis regression (-32%; p = 0.020). These findings were confirmed in the cBDL rat model as well as in human liver cells from patients with cirrhosis, which exhibited phenotypic improvement upon treatment with lanifibranor. CONCLUSIONS: Lanifibranor ameliorates fibrosis and portal hypertension in preclinical models of decompensated cirrhosis. Promising results in human hepatic cells further support its clinical evaluation for the treatment of ACLD. LAY SUMMARY: Advanced chronic liver disease (ACLD) constitutes a serious public health issue for which safe and effective treatments are lacking. This study shows that lanifibranor improves portal hypertension and liver fibrosis, 2 key elements of the pathophysiology of ACLD, in preclinical models of the disease. Evaluation of lanifibranor in liver cells from patients with ACLD further supports its beneficial effects.


Subject(s)
Benzothiazoles/pharmacology , Hypertension, Portal , Liver Cirrhosis , Peroxisome Proliferator-Activated Receptors/agonists , Sulfonamides/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Antifibrotic Agents/pharmacology , Antihypertensive Agents/pharmacology , Cells, Cultured , Disease Models, Animal , Drug Evaluation, Preclinical , Humans , Hypertension, Portal/drug therapy , Hypertension, Portal/etiology , Hypertension, Portal/metabolism , Liver/drug effects , Liver/pathology , Liver Cirrhosis/complications , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Portal Pressure/drug effects , Rats , Vascular Resistance/drug effects
4.
Chirurgia (Bucur) ; 115(6): 707-714, 2020.
Article in English | MEDLINE | ID: mdl-33378629

ABSTRACT

Background-Objectives: It has been reported, that high posthepatectomy portal vein pressure (PVP) has deleterious effect on the liver parenchyma and causes posthepatectomy liver failure (PHLF) and increased 90-day mortality. Terlipressin, is widely used to mitigate the effects of portal hyper-tension. Randomised clinical trials (RCTs) demonstrated encouraging results of use of terlipressin for modulation of increased posthepatectomy PVP. The aim of the present study was to evaluate the effectiveness of the pharmacological modulation of the increased posthepatectomy PVP after major hepatectomy. Methods: Systematic literature searches of electronic databases in accordance with PRISMA was conducted. Meta-analysis was conducted using both fixed- and random-effects models. Results: Three randomised controlled trials (RCTs) comparing terlipressin versus placebo including 284 patients of pooled 60 studies were selected. Placebo cohort patients were significantly younger by 5 years compared to terlipressin cohort. However, the terlipressin cohort demonstrated significantly shorter intensive care unit (ICU) stay compared to placebo cohort. Conclusions: The first meta-analysis demonstrated that terlipressin cohort patients although significantly older by 5 years had significantly shorter ICU stay compared to placebo cohort. Furthermore, though statistically nonsignificant only 6% of terlipressin patients needed inotropic support compared to 16.4% of placebo cohort.


Subject(s)
Cardiovascular Agents/pharmacology , Hepatectomy , Liver Diseases/surgery , Portal Pressure/drug effects , Portal Vein , Terlipressin/pharmacology , Hepatectomy/adverse effects , Humans , Liver Cirrhosis/surgery , Portal Vein/drug effects , Portal Vein/surgery , Randomized Controlled Trials as Topic , Treatment Outcome
5.
United European Gastroenterol J ; 8(10): 1174-1185, 2020 12.
Article in English | MEDLINE | ID: mdl-32878579

ABSTRACT

BACKGROUND: In cirrhosis, the nitric oxide-soluble guanylyl cyclase (sGC)-cyclic guanosine monophosphate (cGMP) pathway is impaired, which contributes to increased intrahepatic vascular resistance (IHVR) and fibrogenesis. We investigated if sGC stimulation (riociguat (RIO)), sGC activation (cinaciguat (CINA)) or phosphodiesterase (PDE)-5 inhibition (tadalafil (TADA)) improves portal hypertension (PHT) and liver fibrosis. METHODS: Fifty male Sprague-Dawley rats underwent bile-duct ligation (BDL) or sham operation. RIO (0.5 mg/kg), CINA (1 mg/kg), TADA (1.5 mg/kg) or vehicle (VEH) was administered from weeks 2 to 4 after BDL. At week 4, invasive haemodynamic measurements were performed, and liver fibrosis was assessed by histology (chromotrope-aniline blue (CAB), Picro-Sirius red (PSR)) and hepatic hydroxyproline content. RESULTS: Cirrhotic bile duct-ligated rats presented with PHT (13.1 ± 1.0 mmHg) and increased IHVR (4.9 ± 0.5 mmHg⋅min/mL). Both RIO (10.0 ± 0.7 mmHg, p = 0.021) and TADA (10.3 ± 0.9 mmHg, p = 0.050) decreased portal pressure by reducing IHVR (RIO: -41%, p = 0.005; TADA: -21%, p = 0.199) while not impacting heart rate, mean arterial pressure and portosystemic shunting. Hepatic cGMP levels increased upon RIO (+239%, p = 0.006) and TADA (+32%, p = 0.073) therapy. In contrast, CINA dosed at 1 mg/kg caused weight loss, arterial hypotension and hyperlactataemia in bile duct-ligated rats. Liver fibrosis area was significantly decreased by RIO (CAB: -48%, p = 0.011; PSR: -27%, p = 0.121) and TADA (CAB: -21%, p = 0.342; PSR: -52%, p = 0.013) compared to VEH-treated bile duct-ligated rats. Hepatic hydroxyproline content was reduced by RIO (from 503 ± 20 to 350 ± 30 µg/g, p = 0.003) and TADA (282 ± 50 µg/g, p = 0.003), in line with a reduction of the hepatic stellate cell activation markers smooth-muscle actin and phosphorylated moesin. Liver transaminases decreased under RIO (AST: -36%; ALT: -32%) and TADA (AST: -24%; ALT: -27%) treatment. Hepatic interleukin 6 gene expression was reduced in the RIO group (-56%, p = 0.053). CONCLUSION: In a rodent model of biliary cirrhosis, the sGC stimulator RIO and the PDE-5 inhibitor TADA improved PHT. The decrease of sinusoidal vascular resistance was paralleled by a reduction in liver fibrosis and hepatic inflammation, while systemic haemodynamics were not affected.


Subject(s)
Hypertension, Portal/drug therapy , Liver Cirrhosis/drug therapy , Phosphodiesterase 5 Inhibitors/therapeutic use , Soluble Guanylyl Cyclase/antagonists & inhibitors , Animals , Benzoates/pharmacology , Benzoates/therapeutic use , Bile Ducts/surgery , Disease Models, Animal , Humans , Hypertension, Portal/diagnosis , Hypertension, Portal/etiology , Hypertension, Portal/physiopathology , Ligation/adverse effects , Liver Cirrhosis/etiology , Male , Phosphodiesterase 5 Inhibitors/pharmacology , Portal Pressure/drug effects , Portal Pressure/physiology , Portal System/drug effects , Portal System/physiopathology , Pyrazoles/pharmacology , Pyrazoles/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Soluble Guanylyl Cyclase/metabolism , Tadalafil/pharmacology , Tadalafil/therapeutic use , Vascular Resistance/drug effects , Vascular Resistance/physiology
6.
Dig Liver Dis ; 52(9): 958-965, 2020 09.
Article in English | MEDLINE | ID: mdl-32736898

ABSTRACT

BACKGROUND: The effects of poorly/non-absorbable antibiotics on hepatic venous pressure gradient (HVPG) are debated. AIM: To analyze the effects of rifaximin or norfloxacin on HVPG and on markers of bacterial translocation and proinflammatory cytokines. METHODS: We performed a systematic search of randomized clinical trials (RCTs) involving patients with cirrhosis and portal hypertension, assessing the effect of rifaximin or norfloxacin vs control on HVPG. Pooled analyses were based on random-effects models, heterogeneity was assessed by Cochran's Q, I2 statistic and subgroup analyses. RESULTS: Five studies (215 patients) were included. Risk of bias was high in three. We found no significant differences using antibiotics versus control. The summary mean difference in HVPG was of -0.55 mmHg (95%CI:-1.52, 0.42; P = 0.27), with moderate heterogeneity (P = 0.15; I2 = 40%). RCTs with longer therapy (60-90 days) used non-selective-beta-blockers (NSBB) in both antibiotics and control arms. Subgroup analysis showed a significantly greater reduction in HVPG in the combination arm over controls (mean difference -1.46 mmHg [95%CI: -2.63, -0.28; P = 0.01]) with no heterogeneity (P = 0.46; I2 = 0%). Serum lipopolysaccharide-binding protein (LBP) significantly decreased with antibiotics, but with high heterogeneity (P < 0.001; I2 = 92%). CONCLUSIONS: Rifaximin or norfloxacin did not significantly reduce HVPG in patients with cirrhosis and portal hypertension. Studies using antibiotic for longer periods on top of NSBB showed a significant decrease in HVPG.


Subject(s)
Adrenergic beta-Antagonists/pharmacology , Anti-Bacterial Agents/pharmacology , Hypertension, Portal/drug therapy , Portal Pressure/drug effects , Bacterial Translocation/drug effects , Drug Therapy, Combination , Humans , Hypertension, Portal/etiology , Liver Cirrhosis/complications , Norfloxacin , Randomized Controlled Trials as Topic , Rifaximin
7.
PLoS One ; 15(5): e0233778, 2020.
Article in English | MEDLINE | ID: mdl-32469999

ABSTRACT

The selection of the anesthetic regime is a crucial component in many experimental animal studies. In rodent models of liver disease, the combination of ketamine and diazepam (KD), generally by the intramuscular (i.m.) route, has traditionally been the anesthesia of choice for the evaluation of systemic and hepatic hemodynamics but it presents several problems. Here, we compared the performance of inhalational sevoflurane (Sevo) against the KD combination as the anesthesia used for hemodynamic studies involving the measurement of portal pressure in normal rats (Ctrl) and rats with non-cirrhotic portal hypertension induced by partial portal vein ligation (PPVL). Compared with Ctrl rats, rats with PPVL presented characteristic alterations that were not influenced by the anesthetic regime, which included liver atrophy, splenomegaly, increased plasma fibrinogen, decreased alkaline phosphatase and glycemia, and frequent ascites. The use of the KD combination presented several disadvantages compared with the inhalational anesthesia with sevoflurane, including considerable mortality, a higher need of dose adjustments to maintain an optimal depth of anesthesia, increases of heart rate, and alteration of blood biochemical parameters such as the concentration of aspartate aminotransferase, lactate, and lactic dehydrogenase. Rats anesthetized with sevoflurane, on the other hand, presented lower respiratory rates. Importantly, the anesthetic regime did not influence the measurement of portal pressure either in Ctrl or PPVL rats, with the increase of portal pressure being similar in Sevo- and KD- anesthetized groups of PPVL rats compared with their respective control groups. Overall, our results suggest that anesthesia with sevoflurane is preferable to the combination of KD for performing systemic and hepatic hemodynamic studies in rats with non-cirrhotic portal hypertension.


Subject(s)
Diazepam/pharmacology , Hypertension, Portal/drug therapy , Ketamine/pharmacology , Portal Pressure/drug effects , Sevoflurane/pharmacology , Anesthesia, Inhalation , Anesthesiology , Anesthetics, Dissociative/pharmacology , Anesthetics, Inhalation/pharmacology , Animals , Disease Models, Animal , Liver/blood supply , Male , Portal Vein/drug effects , Rats , Rats, Sprague-Dawley
8.
Trials ; 21(1): 260, 2020 Mar 12.
Article in English | MEDLINE | ID: mdl-32164767

ABSTRACT

BACKGROUND: In preclinical models, recombinant human relaxin-2 (serelaxin) had anti-fibrotic effects and ameliorated portal hypertension (PH). A small exploratory study in patients with cirrhosis also suggested that serelaxin could reduce portal pressure. METHODS: In a phase 2, double-blind, randomised controlled study conducted in a single centre (Royal Infirmary of Edinburgh, UK), male and female adult participants with cirrhosis and clinically significant PH (CSPH; hepatic venous pressure gradient (HVPG) > 10 mmHg) were enrolled. Participants were allocated to serelaxin or placebo in a 3:1 ratio. The placebo was matched to serelaxin on appearance and administration protocol to create and maintain blinding. The primary endpoint was the change from baseline in fasting HVPG after 2 h of peripheral i.v. serelaxin infusion (80 µg/kg/day for 60 min followed by 30 µg/kg/day for at least 60 min). Secondary endpoints included the change from baseline in hepatic blood flow and systemic haemodynamics (cardiac index, systemic vascular resistance index and aortic pulse wave velocity). Short-term safety and tolerability of serelaxin were assessed. RESULTS: A total of 17 participants were screened, 15 were randomised and 11 completed the study (n = 9 serelaxin, n = 2 placebo). Reasons for withdrawal were baseline HVPG < 10 mmHg (n = 2) and technical failure (n = 2). The trial ended early due to manufacturer discontinuation of the study drug. The median age was 56 (range 43-69) years and 73% of participants were male. Alcohol was the commonest cirrhosis aetiology (n = 10). Participants had a median Model for End-Stage Liver Disease score of 10 (range 6-14). The mean baseline HVPG was 16.3 (range 10.3-21.7) mmHg. Individual responses were variable, but overall there was no statistically significant change in HVPG after 2 h of i.v. serelaxin (arithmetic mean of difference ± SD was 0.4 ± 3.5 mmHg (95% CI -2.3, 3.1; p = 0.76)). There were also no substantial changes from baseline in hepatic or systemic haemodynamics. We recorded 12 adverse events in 7 participants treated with serelaxin; none were significant, and most were unrelated to the investigational medicinal product. There were no serious adverse events. CONCLUSION: In a small randomised, phase 2, proof-of-concept study in patients with cirrhosis and CSPH, serelaxin infusion was safe and well-tolerated but had a neutral effect on HVPG. TRIAL REGISTRATION: ClinicalTrials.gov, NCT02669875. Registered on 1 February 2016.


Subject(s)
Hypertension, Portal/drug therapy , Liver Cirrhosis/drug therapy , Portal Pressure/drug effects , Relaxin/therapeutic use , Vasodilator Agents/therapeutic use , Adult , Aged , Double-Blind Method , Female , Hemodynamics , Humans , Liver/physiopathology , Male , Middle Aged , Proof of Concept Study , Recombinant Proteins/administration & dosage , Recombinant Proteins/therapeutic use , Relaxin/administration & dosage , Severity of Illness Index , United Kingdom , Vasodilator Agents/administration & dosage
9.
Gut ; 69(5): 953-962, 2020 05.
Article in English | MEDLINE | ID: mdl-32139553

ABSTRACT

In the recent years, there have been an increasing number of reports on favourable effects of statins in patients with advanced chronic liver disease. These include reduction in portal pressure, improved liver sinusoidal endothelial and hepatic microvascular dysfunction, decreased fibrogenesis, protection against ischaemia/reperfusion injury, safe prolongation of ex vivo liver graft preservation, reduced sensitivity to endotoxin-mediated liver damage, protection from acute-on-chronic liver failure, prevention of liver injury following hypovolaemic shock and preventing/delaying progression of cirrhosis of any aetiology. Moreover, statins have been shown to have potential beneficial effects in the progression of other liver diseases, such as chronic sclerosing cholangitis and in preventing hepatocellular carcinoma. Because of these many theoretically favourable effects, statins have evolved from being considered a risk to kind of wonder drugs for patients with chronic liver diseases. The present article reviews the current knowledge on the potential applications of statins in chronic liver diseases, from its mechanistic background to objective evidence from clinical studies.


Subject(s)
Carcinoma, Hepatocellular/prevention & control , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Liver Cirrhosis/drug therapy , Liver Failure/prevention & control , Liver Neoplasms/surgery , Biopsy, Needle , Carcinoma, Hepatocellular/pathology , Disease Progression , Female , Graft Rejection/drug therapy , Humans , Immunohistochemistry , Liver Cirrhosis/pathology , Liver Neoplasms/prevention & control , Liver Transplantation/methods , Male , Portal Pressure/drug effects , Severity of Illness Index
10.
Gastroenterology ; 158(5): 1334-1345.e5, 2020 04.
Article in English | MEDLINE | ID: mdl-31812510

ABSTRACT

BACKGROUND & AIMS: Increased levels of galectin 3 have been associated with nonalcoholic steatohepatitis (NASH) and contribute to toxin-induced liver fibrosis in mice. GR-MD-02 (belapectin) is an inhibitor of galectin 3 that reduces liver fibrosis and portal hypertension in rats and was safe and well tolerated in phase 1 studies. We performed a phase 2b, randomized trial of the safety and efficacy of GR-MD-02 in patients with NASH, cirrhosis, and portal hypertension. METHODS: Patients with NASH, cirrhosis, and portal hypertension (hepatic venous pressure gradient [HVPG] ≥ 6 mm Hg) from 36 centers were randomly assigned, in a double-blind manner, to groups that received biweekly infusions of belapectin 2 mg/kg (n = 54), 8 mg/kg (n = 54), or placebo (n = 54) for 52 weeks. The primary endpoint was change in HVPG (Δ HVPG) at the end of the 52-week period compared with baseline. Secondary endpoints included changes in liver histology and development of liver-related outcomes. RESULTS: We found no significant difference in ΔHVPG between the 2 mg/kg belapectin group and placebo group (-0.28 mm HG vs 0.10 mm HG, P = 1.0) or between the 8 mg/kg belapectin and placebo group (-0.25 mm HG vs 0.10 mm HG, P = 1.0). Belapectin had no significant effect on fibrosis or nonalcoholic fatty liver disease activity score, and liver-related outcomes did not differ significantly among groups. In an analysis of a subgroup of patients without esophageal varices at baseline (n = 81), 2 mg/kg belapectin was associated with a reduction in HVPG at 52 weeks compared with baseline (P = .02) and reduced development of new varices (P = .03). Belapectin (2 mg/kg) was well tolerated and produced no safety signals. CONCLUSIONS: In a phase 2b study of 162 patients with NASH, cirrhosis, and portal hypertension, 1 year of biweekly infusion of belapectin was safe but not associated with significant reduction in HVPG or fibrosis compared with placebo. However, in a subgroup analysis of patients without esophageal varices, 2 mg/kg belapectin did reduce HVPG and development of varices. ClinicalTrials.gov number: NCT02462967.


Subject(s)
Galectin 3/antagonists & inhibitors , Hypertension, Portal/drug therapy , Liver Cirrhosis/drug therapy , Non-alcoholic Fatty Liver Disease/drug therapy , Pectins/administration & dosage , Aged , Biopsy , Blood Proteins , Double-Blind Method , Drug Administration Schedule , Female , Galectin 3/metabolism , Galectins , Humans , Hypertension, Portal/diagnosis , Hypertension, Portal/etiology , Hypertension, Portal/pathology , Infusions, Intravenous , Liver/drug effects , Liver/pathology , Liver Cirrhosis/diagnosis , Liver Cirrhosis/pathology , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/complications , Non-alcoholic Fatty Liver Disease/diagnosis , Non-alcoholic Fatty Liver Disease/pathology , Pectins/adverse effects , Placebos/administration & dosage , Placebos/adverse effects , Portal Pressure/drug effects , Severity of Illness Index , Treatment Outcome
11.
Lancet Gastroenterol Hepatol ; 5(1): 31-41, 2020 01.
Article in English | MEDLINE | ID: mdl-31607677

ABSTRACT

BACKGROUND: Statins have beneficial effects on intrahepatic circulation and decrease portal hypertension and rifaximin modulates the gut microbiome and might prevent bacterial translocation in patients with cirrhosis. Therefore, this drug combination might be of therapeutic benefit in patients with decompensated cirrhosis. However, there is concern regarding the safety of statins in patients with decompensated cirrhosis. We assessed the safety of two different doses of simvastatin, in combination with rifaximin, in patients with decompensated cirrhosis. METHODS: We did a double-blind, randomised, placebo-controlled, phase 2 trial in patients with decompensated cirrhosis and moderate-to-severe liver failure from nine university hospitals in six European countries (Italy, France, Holland, Germany, the UK, and Spain). Patients older than 18 years with Child-Pugh class B or C disease were eligible. We randomly assigned patients (1:1:1) to receive either simvastatin 40 mg/day plus rifaximin 1200 mg/day, simvastatin 20 mg/day plus rifaximin 1200 mg/day, or placebo of both medications for 12 weeks. Randomisation was stratified according to Child-Pugh class (B vs C) and restricted using blocks of multiples of three. The primary endpoint was development of liver or muscle toxicity, as defined by changes in liver aminotransferases (aspartate aminotransferase [AST] and alanine aminotransferase [ALT]), alkaline phosphastase, and creatine kinase. The study is registered with the European Union Clinical Trials Register, 2016-004499-23, and with ClinicalTrials.gov, NCT03150459. FINDINGS: The study recruitment period was between July 28, 2017, and Jan 2, 2018. Follow-up finished on March 12, 2018. 50 patients were randomly assigned to simvastatin 40 mg/day plus rifaximin 1200 mg/day (n=18), simvastatin 20 mg/day plus rifaximin 1200 mg/day (n=16), or placebo of both medications (n=16). Six patients (two from each group) were excluded. Therefore, the full analysis set included 44 patients (16 in the simvastatin 40 mg/day plus rifaximin 1200 mg/day group, 14 in the simvastatin 20 mg/day plus rifaximin mg/day group, and 14 in the placebo group). After a safety analyses when the first ten patients completed treatment, treatment was stopped prematurely in the simvastatin 40 mg/day plus rifaximin group due to recommendations by the data safety monitoring board. Patients in the simvastatin 40 mg/day plus rifaximin group showed a significant increase in AST and ALT compared with the placebo group (mean differences between the groups at the end of treatment for AST 130 IU/L [95% CI 54 to 205; p=0·0009] and for ALT 61 IU/L [22 to 100; p=0·0025]. We observed no significant differences at 12 weeks in AST and ALT between the simvastatin 20 mg/day plus rifaximin and placebo group (for AST -14 IU/L [-91 to 64; p=0·728] and for ALT -8 IU/L [-49 to 33; p=0·698]). We observed no significant differences in alkaline phosphatase between the the simvastatin 40 mg/day plus rifaximin or the simvastatin 20 mg/day plus rifaximin groups compared with placebo. Patients in the simvastatin 40 mg/day plus rifaximin group showed an increase in creatine kinase at the end of treatment compared with patients in the placebo group (1009 IU/L [208 to 1809]; p=0·014). We observed no significant changes in creatine kinase in the simvastatin 20 mg/day plus rifaximin group (4·2 IU/L [-804 to 813]; p=0·992). Three (19%) patients in the simvastatin 40 mg/day group developed liver and muscle toxicity consistent with rhabdomyolysis. The number of patients who stopped treatment because of adverse events was significantly higher in the simvastatin 40 mg/day plus rifaximin group (nine [56%] of 16 patients) compared with the other two groups (two [14%] of 14 for both groups; p=0·017). There were no serious unexpected adverse reactions reported during the study. INTERPRETATION: Treatment with simvastatin 40 mg/day plus rifaximin in patients with decompensated cirrhosis was associated with a significant increase in adverse events requiring treatment withdrawal, particularly rhabdomyolysis, compared with simvastatin 20 mg/day plus rifaximin. We recommend simvastatin 20 mg/day as the dose to be used in studies investigating the role of statins in patients with decompensated cirrhosis. FUNDING: Horizon 20/20 European programme.


Subject(s)
Hypertension, Portal/prevention & control , Liver Cirrhosis/drug therapy , Rifaximin/administration & dosage , Simvastatin/administration & dosage , Dose-Response Relationship, Drug , Double-Blind Method , Drug Therapy, Combination , Follow-Up Studies , Gastrointestinal Agents/administration & dosage , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Hypertension, Portal/etiology , Hypertension, Portal/physiopathology , Liver Cirrhosis/complications , Liver Cirrhosis/diagnosis , Portal Pressure/drug effects , Treatment Outcome
12.
J Hepatol ; 72(5): 885-895, 2020 05.
Article in English | MEDLINE | ID: mdl-31870950

ABSTRACT

BACKGROUND & AIMS: Emricasan, an oral pan-caspase inhibitor, decreased portal pressure in experimental cirrhosis and in an open-label study in patients with cirrhosis and severe portal hypertension, defined as a hepatic venous pressure gradient (HVPG) ≥12 mmHg. We aimed to confirm these results in a placebo-controlled study in patients with non-alcoholic steatohepatitis (NASH)-related cirrhosis. METHODS: We performed a multicenter double-blinded study, randomizing 263 patients with NASH-related cirrhosis and baseline HVPG ≥12 mmHg to twice daily oral emricasan 5 mg, 25 mg, 50 mg or placebo in a 1:1:1:1 ratio for up to 48 weeks. The primary endpoint was change in HVPG (ΔHVPG) at week 24. Secondary endpoints were changes in biomarkers (aminotransferases, caspases, cytokeratins) and development of liver-related outcomes. RESULTS: There were no significant differences in ΔHVPG for any emricasan dose vs. placebo (-0.21, -0.45, -0.58 mmHg, respectively) adjusted for baseline HVPG, compensation status, and non-selective beta-blocker use. Compensated patients (n = 201 [76%]) tended to have a greater decrease in HVPG (emricasan all vs. placebo, p = 0.06), the decrease being greater in those with higher baseline HVPG (p = 0.018), with a significant interaction between baseline HVPG (continuous, p = 0.024; dichotomous at 16 mmHg [median], p = 0.013) and treatment. Biomarkers decreased significantly with emricasan at week 24 but returned to baseline levels by week 48. New or worsening decompensating events (∼10% over median exposure of 337 days), progression in model for end-stage liver disease and Child-Pugh scores, and treatment-emergent adverse events were similar among treatment groups. CONCLUSIONS: Despite a reduction in biomarkers indicating target engagement, emricasan was not associated with improvement in HVPG or clinical outcomes in patients with NASH-related cirrhosis and severe portal hypertension. Compensated patients with higher baseline HVPG had evidence of a small treatment effect. Emricasan treatment appeared safe and well-tolerated. LAY SUMMARY: Cirrhosis (scarring of the liver) is the main consequence of non-alcoholic steatohepatitis (NASH). Cirrhosis leads to high pressure in the portal vein which accounts for most of the complications of cirrhosis. Reducing portal pressure is beneficial in patients with cirrhosis. We studied the possibility that emricasan, a drug that improves inflammation and scarring in the liver, would reduce portal pressure in patients with NASH-related cirrhosis and severe portal hypertension. Our results in a large, prospective, double-blind study could not demonstrate a beneficial effect of emricasan in these patients. CLINICAL TRIAL NUMBER: Clinical Trials.gov #NCT02960204.


Subject(s)
Caspase Inhibitors/administration & dosage , Hypertension, Portal/complications , Hypertension, Portal/drug therapy , Liver Cirrhosis/complications , Non-alcoholic Fatty Liver Disease/complications , Pentanoic Acids/administration & dosage , Severity of Illness Index , Administration, Oral , Aged , Biomarkers/blood , Caspase Inhibitors/adverse effects , Double-Blind Method , Female , Follow-Up Studies , Humans , Hypertension, Portal/blood , Liver Cirrhosis/blood , Male , Middle Aged , Non-alcoholic Fatty Liver Disease/blood , Pentanoic Acids/adverse effects , Portal Pressure/drug effects , Prospective Studies , Treatment Outcome
13.
Gastroenterol. hepatol. (Ed. impr.) ; 42(9): 534-541, nov. 2019. ilus, graf, tab
Article in English | IBECS | ID: ibc-187914

ABSTRACT

Introduction and aim: Thromboxane (TX) A2 was identified as an important vasoconstrictor during Zymosan induced portal perfusion pressure (PP) increase. We aimed at investigating whether hepatic steatosis influences the extent of TXA2-induced portal hypertension. Materials and methods: Sprague-Dawley rats were randomly divided into control and steatosis (induced by the special diet) groups. PP and TXB2 (stable degradation product of TXA2) in the perfusate were measured after in situ liver perfusion with Zymosan (150μg/ml, 40-46min) or U46619 (TXA2 analog, 0.1μM/ml, 40-46min). The number of Kupffer cell (KC) was measured by immunohistochemistry with CD163. Results: Zymosan induced more TXB2 production and a higher PP increase in control group than in steatosis group despite more CD163 positive KCs in fatty livers. PP and TXB2 efflux revealed a strong correlation in control group and a moderate correlation in steatosis group. Contrary to the effect of Zymosan, U46619 induced a much higher PP increase in steatosis group than in control group. Conclusion: Severe steatosis increased number of KCs, however, PP increase and TXB2 efflux caused by Zymosan infusion in fatty livers were lower than those in healthy livers. In contrast, TXA2 analog caused higher PP increase in fatty livers. Targeting the more sensitive response to TXA2 in fatty livers might be a potential therapy of severe steatosis


Introducción y objetivos: Se ha identificado al tromboxano (TX) A2 como importante vasoconstrictor durante el aumento de la presión de perfusión portal (PP) inducida por zymosan. El objetivo ha sido analizar si la esteatosis hepática influye en el grado de hipertensión portal inducida por TXA2. Materiales y métodos: Las ratas Sprague-Dawley(R) se han dividido aleatoriamente en grupos de control y esteatosis (inducida por una dieta especial). Se midieron la PP y el TXB2 (producto de degradación estable de TXA2) en la perfusión después de la perfusión hepática in situ de zymosan (150μg/ml, minuto 40-46) o U46619 (análogo de TXA2, 0,1μM/ml, minuto 40-46). El número de células de Kupffer (CK) se midió mediante inmunohistoquímica con CD163. Resultados: Zymosan provocó más producción de TXB2 y mayor aumento de la PP en el grupo de control que en el grupo de esteatosis a pesar de hallar más CK positivas para CD163 en hígados grasos. El flujo de salida de la PP y el TXB2 reveló una fuerte correlación en el grupo de control y una correlación moderada en el grupo de esteatosis. De manera diferente al efecto de zymosan, U46619 indujo un aumento de la PP mucho mayor en el grupo de esteatosis que en el grupo de control. Conclusión: La esteatosis grave aumentó el número de CK; sin embargo, el aumento de la PP y el flujo de TXB2 provocado por la perfusión de zymosan en hígados grasos fueron menores que en los hígados sanos. En cambio, el análogo de TXA2 provocó un aumento de la PP en hígados grasos. Centrarse en la respuesta más sensible al TXA2 en hígados grasos podría convertirse en un tratamiento potencial de la esteatosis grave


Subject(s)
Animals , Rats , Fatty Liver/complications , Hypertension, Portal/chemically induced , Portal Pressure/drug effects , Thromboxane B2/biosynthesis , Zymosan/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Fatty Liver/pathology , Fatty Liver/physiopathology , Kupffer Cells/chemistry , Kupffer Cells/cytology , Perfusion/methods , Portal Pressure/physiology , Thromboxane A2/analogs & derivatives , Rats, Sprague-Dawley
14.
PLoS One ; 14(10): e0222840, 2019.
Article in English | MEDLINE | ID: mdl-31644538

ABSTRACT

BACKGROUND: Prostanoids are important regulators of platelet aggregation and thrombotic arterial diseases. Their involvement in the development of portal vein thrombosis, frequent in decompensated liver cirrhosis, is still not investigated. METHODS: Therefore, we used pro-thrombotic venous milieu generation by bare metal stent transjugular intrahepatic portosystemic shunt insertion, to study the role of prostanoids in decompensated liver cirrhosis. Here, 89 patients receiving transjugular intrahepatic portosystemic shunt insertion were included in the study, and baseline levels of thromboxane B2, prostaglandin D2 and prostaglandin E2 were measured in the portal and the hepatic vein. RESULTS: While the hepatic vein contained higher levels of thromboxane B2 than the portal vein, levels of prostaglandin E2 and D2 were higher in the portal vein (all P<0.0001). Baseline concentrations of thromboxane B2 in the portal vein were independently associated with an increase of portal hepatic venous pressure gradient during short term follow-up, as an indirect sign of thrombogenic potential (multivariable P = 0.004). Moreover, severity of liver disease was inversely correlated with portal as well as hepatic vein levels of prostaglandin D2 and E2 (all P<0.0001). CONCLUSIONS: Elevated portal venous thromboxane B2 concentrations are possibly associated with the extent of thrombogenic potential in patients with decompensated liver cirrhosis. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT03584204.


Subject(s)
Blood Coagulation/drug effects , Liver Cirrhosis/blood , Liver Cirrhosis/physiopathology , Portal Vein/physiopathology , Prostaglandins/blood , Female , Humans , Male , Middle Aged , Portal Pressure/drug effects , Portal Vein/drug effects , Portasystemic Shunt, Transjugular Intrahepatic , Regression Analysis , Survival Analysis
15.
World J Gastroenterol ; 25(39): 5953-5960, 2019 Oct 21.
Article in English | MEDLINE | ID: mdl-31660032

ABSTRACT

BACKGROUND: Portal hypertension (PHT) is primarily caused by an increase in resistance to portal outflow and secondarily by an increase in splanchnic blood flow. Vascular hyporeactivity both in systemic circulation and in the mesenteric artery plays a role in the hyperdynamic circulatory syndrome. AIM: To explore gender differences and the role of endogenous sex hormones in PHT and vascular reactivity of mesenteric arterioles in rats. METHODS: Cirrhosis and PHT were established by subcutaneous injection of carbon tetrachloride (CCl4) in both male and female integral and castrated rats (ovariectomized [OVX] in female rats, orchiectomy [ORX] in male rats). The third-order branch of the mensenteric artery was divided and used to measure vascular reactivity to vasoconstrictors. RESULTS: No significant difference in portal pressure was observed between integral and castrated male PHT rats (15.2 ± 2.1 mmHg vs 16.7 ± 2.7 mmHg, P > 0.05). The portal pressure in integral female PHT rats was lower than that in OVX female PHT rats (12.7 ± 2.7 mmHg vs 16.5 ± 2.4 mmHg, P < 0.05). In PHT rats, the concentration response curves of the mesenteric arterioles to norepinephrine were shifted to the right, and the maximal responses (Emax) values were decreased and effective concentrations causing half maximum responses (EC50) values were increased, compared to those of non-PHT rats, both in male and female rats. Compared to non-PHT integral male rats, the sensitivity of the mesenteric arterioles of non-PHT ORX male rats to norepinephrine was decreased (P > 0.05). However, there was no difference between integral and ORX male rats with PHT. In integral female PHT rats, the concentration response curves were shifted to the left (P < 0.05), and the Emax values were increased and EC50 values were decreased compared to OVX female PHT rats. CONCLUSION: Clear gender differences were observed in mesenteric vascular reactivity in CCl4-induced cirrhotic and PHT rats. Conservation of estrogen can retain the sensitivity of the mesenteric arterioles to vasoconstrictors and has a protective effect on splanchnic vascular function in PHT.


Subject(s)
Arterioles/physiology , Gonadal Steroid Hormones/metabolism , Hypertension, Portal/physiopathology , Liver Cirrhosis, Experimental/physiopathology , Vascular Resistance/physiology , Animals , Arterioles/drug effects , Carbon Tetrachloride/toxicity , Female , Humans , Hypertension, Portal/chemically induced , Hypertension, Portal/metabolism , Liver Cirrhosis, Experimental/chemically induced , Liver Cirrhosis, Experimental/metabolism , Male , Mesenteric Arteries/drug effects , Mesenteric Arteries/physiopathology , Portal Pressure/drug effects , Portal Pressure/physiology , Rats , Rats, Sprague-Dawley , Sex Factors , Splanchnic Circulation/drug effects , Splanchnic Circulation/physiology , Vascular Resistance/drug effects , Vasoconstrictor Agents/administration & dosage
16.
Gastroenterol Hepatol ; 42(9): 534-541, 2019 Nov.
Article in English, Spanish | MEDLINE | ID: mdl-31326104

ABSTRACT

INTRODUCTION AND AIM: Thromboxane (TX) A2 was identified as an important vasoconstrictor during Zymosan induced portal perfusion pressure (PP) increase. We aimed at investigating whether hepatic steatosis influences the extent of TXA2-induced portal hypertension. MATERIALS AND METHODS: Sprague-Dawley rats were randomly divided into control and steatosis (induced by the special diet) groups. PP and TXB2 (stable degradation product of TXA2) in the perfusate were measured after in situ liver perfusion with Zymosan (150µg/ml, 40-46min) or U46619 (TXA2 analog, 0.1µM/ml, 40-46min). The number of Kupffer cell (KC) was measured by immunohistochemistry with CD163. RESULTS: Zymosan induced more TXB2 production and a higher PP increase in control group than in steatosis group despite more CD163 positive KCs in fatty livers. PP and TXB2 efflux revealed a strong correlation in control group and a moderate correlation in steatosis group. Contrary to the effect of Zymosan, U46619 induced a much higher PP increase in steatosis group than in control group. CONCLUSION: Severe steatosis increased number of KCs, however, PP increase and TXB2 efflux caused by Zymosan infusion in fatty livers were lower than those in healthy livers. In contrast, TXA2 analog caused higher PP increase in fatty livers. Targeting the more sensitive response to TXA2 in fatty livers might be a potential therapy of severe steatosis.


Subject(s)
Fatty Liver/complications , Hypertension, Portal/chemically induced , Portal Pressure/drug effects , Thromboxane B2/biosynthesis , Zymosan/pharmacology , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Cell Count , Diet, High-Fat , Fatty Liver/pathology , Fatty Liver/physiopathology , Kupffer Cells/chemistry , Kupffer Cells/cytology , Perfusion/methods , Portal Pressure/physiology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Cell Surface/analysis , Thromboxane A2/analogs & derivatives , Thromboxane B2/analysis , Vasoconstrictor Agents
17.
Acta Med Port ; 32(6): 420-426, 2019 Jun 28.
Article in English | MEDLINE | ID: mdl-31292022

ABSTRACT

INTRODUCTION: Excessive portal venous pressure in the liver remnant is an independent factor in the occurrence of posthepatectomy liver failure and small-for-size syndrome. The baseline portal pressure prior to hepatectomy was not considered previously. The aim of this study is to assess the impact of portal pressure change during hepatectomy on the patient outcome. MATERIAL AND METHODS: Prospective observational study including 30 patients subjected to intraoperative measurement of portal pressure before and after hepatectomy. This variation was related to the patient outcome. Control group evaluation was assessed. Patient, disease and procedure features were considered. The optimal cut-off of portal pressure variation was determined. Linear regression or logistic regression was applied to identify predictors of the outcome. RESULTS: The univariate analysis showed that portal pressure increase after hepatectomy was associated with coagulation impairment in the first 30 postoperative days (p < 0.05), and with the occurrence of major complications (p = 0.01), namely hepatic failure (p = 0.041). The multivariate analysis showed that portal venous pressure increase ≥ 2 mmHg is an independent factor for worse outcomes. DISCUSSION: As in previous studies, this study concludes that, after hepatectomy, in addition to the functional liver remnant, other factors are responsible for deterioration of liver function and patient outcome, such as the portal pressure increase and the exposure to chemotherapy prior to hepatectomy. This work may influence the definition of future indications for portal influx modulation. CONCLUSION: Patient outcomes are influenced by the portal venous pressure increase: an increment ≥ 2 mmHg after hepatectomy seems to increase the risk of major complications.


Introdução: O aumento da pressão venosa portal para o remanescente hepático é um fator independente para falência hepática após hepatectomia e síndrome small-for-size. Estudos anteriores não consideram o valor de pressão portal prévio à hepatectomia. O objetivo deste estudo é analisar o impacto da variação da pressão portal durante a hepatectomia na evolução clínica pós-operatória. Material e Métodos: Estudo observacional prospetivo, incluindo 30 doentes submetidos a medição intraoperatória da pressão portal antes e após hepatectomia, relacionando esta variação com a evolução clínica pós-operatória. Avaliação similar foi efetuada num grupo de controlo. Fatores relacionados com o doente, doença e procedimento foram considerados. Determinou-se o valor ideal de variação da pressão portal. Regressão linear ou logística foram aplicadas para identificar fatores preditores de evolução clínica. Resultados: A análise univariada mostrou que um aumento de pressão portal após hepatectomia associa-se a deterioração da coagulação nos primeiros 30 dias após hepatectomia (p < 0,05), a complicações major (p = 0,01) como a falência hepática após hepatectomia (p = 0,041). A análise multivariada mostrou que um aumento de pressão portal ≥ 2 mmHg é um fator independente para a evolução clínica pós-operatória desfavorável. Discussão: Após hepatectomia, para além do remanescente hepático funcional, outros fatores são responsáveis pela deterioração da função hepática e pela morbimortalidade, como o aumento da pressão portal e a exposição prévia a quimioterapia. Este trabalho contribui para a definição futura das indicações para modulação do influxo portal. Conclusão: Um aumento de pressão portal ≥ 2 mmHg após hepatectomia parece agravar o risco de complicações major.


Subject(s)
Blood Coagulation Disorders/etiology , Hepatectomy/adverse effects , Liver Failure/etiology , Portal Pressure/physiology , Postoperative Complications/etiology , Aged , Analysis of Variance , Area Under Curve , Blood Pressure Determination/methods , Case-Control Studies , Female , Hepatectomy/mortality , Humans , Hypertension , International Normalized Ratio , Intraoperative Care , Linear Models , Liver/enzymology , Liver Failure/mortality , Male , Middle Aged , Portal Pressure/drug effects , Postoperative Care , Postoperative Complications/mortality , Prospective Studies , Prothrombin Time , Time Factors , Treatment Outcome
18.
Sci Rep ; 9(1): 2256, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30783172

ABSTRACT

Rho-kinase (ROCK) activation in hepatic stellate cells (HSC) is a key mechanism promoting liver fibrosis and portal hypertension (PTH). Specific delivery of ROCK-inhibitor Y-27632 (Y27) to HSC targeting mannose-6-phosphate-receptors reduces portal pressure and fibrogenesis. In decompensated cirrhosis, presence of ascites is associated with reduced renal perfusion. Since in cirrhosis, platelet-derived growth factor receptor beta (PDGFRß) is upregulated in the liver as well as the kidney, this study coupled Y27 to human serum albumin (HSA) substituted with PDGFRß-recognizing peptides (pPB), and investigated its effect on PTH in cirrhotic rats. In vitro collagen contraction assays tested biological activity on LX2 cells. Hemodynamics were analyzed in BDL and CCl4 cirrhotic rats 3 h, 6 h and 24 h after i.v. administration of Y27pPBHSA (0.5/1 mg/kg b.w). Phosphorylation of moesin and myosin light chain (MLC) assessed ROCK activity in liver, femoral muscle, mesenteric artery, kidney and heart. Three Y27 molecules were coupled to pPBHSA as confirmed by HPLC/MS, which was sufficient to relax LX2 cells. In vivo, Y27pPBHSA-treated rats exhibited lower portal pressure, hepatic vascular resistance without effect on systemic vascular resistance, but a tendency towards lower cardiac output compared to non-treated cirrhotic rats. Y27pPBHSA reduced intrahepatic resistance by reduction of phosphorylation of moesin and MLC in Y27pPBHSA-treated cirrhotic rats. Y27pPBHSA was found in the liver of rats up to 6 hours after its injection, in the HSC demonstrated by double-immunostainings. Interestingly, Y27pPBHSA increased renal arterial flow over time combined with an antifibrotic effect as shown by decreased renal acta2 and col1a1 mRNA expression. Therefore, targeting the ROCK inhibitor Y27 to PDGFRß decreases portal pressure with potential beneficial effects in the kidney. This unique approach should be tested in human cirrhosis.


Subject(s)
Drug Carriers , Enzyme Inhibitors , Kidney/blood supply , Liver Cirrhosis , Portal Pressure/drug effects , Serum Albumin, Human , rho-Associated Kinases/antagonists & inhibitors , Animals , Drug Carriers/chemistry , Drug Carriers/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Humans , Kidney/metabolism , Kidney/pathology , Liver Cirrhosis/drug therapy , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Perfusion , Rats , Rats, Sprague-Dawley , Serum Albumin, Human/chemistry , Serum Albumin, Human/pharmacology , rho-Associated Kinases/metabolism
19.
Clin Sci (Lond) ; 133(1): 153-166, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30606815

ABSTRACT

Liver failure is the major cause of death following liver resection. Post-resection portal venous pressure (PVP) predicts liver failure, is implicated in its pathogenesis, and when PVP is reduced, rates of liver dysfunction decrease. The aim of the present study was to characterize the hemodynamic, biochemical, and histological changes induced by 80% hepatectomy in non-cirrhotic pigs and determine if terlipressin or direct portacaval shunting can modulate these effects. Pigs were randomized (n=8/group) to undergo 80% hepatectomy alone (control); terlipressin (2 mg bolus + 0.5-1 mg/h) + 80% hepatectomy; or portacaval shunt (PCS) + 80% hepatectomy, and were maintained under terminal anesthesia for 8 h. The primary outcome was changed in PVP. Secondary outcomes included portal venous flow (PVF), hepatic arterial flow (HAF), and biochemical and histological markers of liver injury. Hepatectomy increased PVP (9.3 ± 0.4 mmHg pre-hepatectomy compared with 13.0 ± 0.8 mmHg post-hepatectomy, P<0.0001) and PVF/g liver (1.2 ± 0.2 compared with 6.0 ± 0.6 ml/min/g, P<0.0001) and decreased HAF (70.8 ± 5.0 compared with 41.8 ± 5.7 ml/min, P=0.002). Terlipressin and PCS reduced PVP (terlipressin = 10.4 ± 0.8 mmHg, P=0.046 and PCS = 8.3 ± 1.2 mmHg, P=0.025) and PVF (control = 869.0 ± 36.1 ml/min compared with terlipressin = 565.6 ± 25.7 ml/min, P<0.0001 and PCS = 488.4 ± 106.4 ml/min, P=0.002) compared with control. Treatment with terlipressin increased HAF (73.2 ± 11.3 ml/min) compared with control (40.3 ± 6.3 ml/min, P=0.026). The results of the present study suggest that terlipressin and PCS may have a role in the prevention and treatment of post-resection liver failure.


Subject(s)
Hepatectomy , Hepatic Artery/drug effects , Liver Circulation/drug effects , Liver Failure/prevention & control , Liver/blood supply , Portacaval Shunt, Surgical , Portal Pressure/drug effects , Portal Vein/drug effects , Terlipressin/pharmacology , Animals , Blood Flow Velocity , Disease Models, Animal , Hepatic Artery/physiopathology , Liver/pathology , Liver Failure/etiology , Liver Failure/pathology , Liver Failure/physiopathology , Male , Portal Vein/physiopathology , Sus scrofa
20.
BMC Gastroenterol ; 19(1): 231, 2019 Dec 30.
Article in English | MEDLINE | ID: mdl-31888534

ABSTRACT

BACKGROUND: Statin has been more and more widely used in chronic liver disease, however, existed studies have attained contradictory results. According to the present study, we aimed to test the efficacy and safety of statin via a meta-analysis. METHODS: Different databases were searched for full-text publication based on inclusion and exclusion criteria. For data-pooling, fixed-effect model was applied if heterogeneity wasn't detected. Otherwise, random-effect model was adopted. Heterogeneity was detected by I squire (I2) test. All results of analysis were illustrated as forest plots. Publication bias was assessed using the Begg's adjusted rank correlation test. Standard mean difference (SMD) was calculated in continuous variables. Pooled hazard ratio or odds ratio was calculated in catergorical variables. RESULTS: Seventeen clinical studies were finally included. Hepatic portal hemodynamic parameters were improved in statin users for a short-term response. For a long-term follow-up, statin treatment surprisingly decreased mortality rate (HR = 0.782, 95% CI: 0.718-0.846, I2 > 50%) and lower the occurrence of hepatocellular carcinoma (HR = 0.75, 95% CI: 0.64-0.86, I2 > 50%) in liver cirrhosis. Statin seemed not to decrease the risk of esophageal variceal bleeding and spontaneous bacterial peritonitis. However, statin was proved to decrease the risk of hepatic encephalopathy and ascites. Incidence of drug related adverse events didn't increase in statin users. Dose-dependent effects of statin on hepatocellular carcinoma development, decompensated cirrhosis events occurrence, and liver cirrhosis progression. CONCLUSION: Statin influenced parameters of hepatic portal vessel pressure in short-term treatment. Prognosis of liver cirrhosis benefited from statin treatment in long term follow-up. The efficacy and safety of statin in liver cirrhosis treatment is confirmed. To date, similar study is hardly seen before.


Subject(s)
Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Liver Cirrhosis/drug therapy , Ascites/prevention & control , Carcinoma, Hepatocellular/prevention & control , Disease Progression , Dose-Response Relationship, Drug , Esophageal and Gastric Varices/complications , Gastrointestinal Hemorrhage/prevention & control , Hepatic Encephalopathy/prevention & control , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Liver Cirrhosis/mortality , Liver Neoplasms/prevention & control , Non-Randomized Controlled Trials as Topic , Peritonitis/microbiology , Peritonitis/prevention & control , Portal Pressure/drug effects , Publication Bias , Randomized Controlled Trials as Topic
SELECTION OF CITATIONS
SEARCH DETAIL
...