1
|
Manns MP, Bergquist A, Karlsen TH, Levy C, Muir AJ, Ponsioen C, Trauner M, Wong G, Younossi ZM. Primary sclerosing cholangitis. Nat Rev Dis Primers 2025; 11:17. [PMID: 40082445 DOI: 10.1038/s41572-025-00600-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/07/2025] [Indexed: 03/16/2025]
Abstract
Primary sclerosing cholangitis (PSC) is a chronic biliary inflammation associated with periductular fibrosis of the intrahepatic and extrahepatic bile ducts leading to strictures, bacterial cholangitis, decompensated liver disease and need for liver transplantation. This rare focal liver disease affects all races and ages, with a predominance of young males. There is an up to 88% association with inflammatory bowel disease. Although the aetiology is unknown and the pathophysiology is poorly understood, PSC is regarded as an autoimmune liver disease based on a strong immunogenetic background. Further, the associated risk for various malignancies, particularly cholangiocellular carcinoma, is also poorly understood. No medical therapy has been approved so far nor has been shown to improve transplant-free survival. However, ursodeoxycholic acid is widely used since it improves the biochemical parameters of cholestasis and is safe at low doses. MRI of the biliary tract is the primary imaging technology for diagnosis. Endoscopic interventions of the bile ducts should be limited to clinically relevant strictures for balloon dilatation, biopsy and brush cytology. End-stage liver disease with decompensation is an indication for liver transplantation with recurrent PSC in up to 38% of patients. Several novel therapeutic strategies are in various stages of development, including apical sodium-dependent bile acid transporter and ileal bile acid transporter inhibitors, integrin inhibitors, peroxisome proliferator-activated receptor agonists, CCL24 blockers, recombinant FGF19, CCR2/CCR5 inhibitors, farnesoid X receptor bile acid receptor agonists, and nor-ursodeoxycholic acid. Manipulation of the gut microbiome includes faecal microbiota transplantation. This article summarizes present knowledge and defines unmet medical needs to improve quality of life and survival.
Collapse
Affiliation(s)
- Michael P Manns
- Hannover Medical School (MHH) and Centre for Individualised Infection Medicine (CiiM), Hannover, Germany.
| | - Annika Bergquist
- Division of Hepatology, Department of Upper Gastrointestinal Disease, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Tom H Karlsen
- Norwegian PSC Research Center, Department of Transplantation Medicine, Clinic of Surgery and Specialized medicine, Oslo University Hospital, Oslo, Norway
- Research Institute of Internal Medicine, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Cynthia Levy
- Division of Digestive Health and Liver Diseases, University of Miami School of Medicine, Miami, FL, USA
| | - Andrew J Muir
- Division of Gastroenterology, Duke University School of Medicine, Durham, NC, USA
| | - Cyriel Ponsioen
- Department of Gastroenterology & Hepatology, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Grace Wong
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Union Hospital, Hong Kong SAR, China
| | | |
Collapse
|
2
|
Kanda T, Sasaki-Tanaka R, Kimura N, Abe H, Yoshida T, Hayashi K, Sakamaki A, Yokoo T, Kamimura H, Tsuchiya A, Kamimura K, Terai S. Pruritus in Chronic Cholestatic Liver Diseases, Especially in Primary Biliary Cholangitis: A Narrative Review. Int J Mol Sci 2025; 26:1883. [PMID: 40076514 PMCID: PMC11900276 DOI: 10.3390/ijms26051883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Patients with chronic cholestatic liver diseases often experience itch and struggle with this symptom. We discuss the mechanism of itch in patients with chronic cholestatic liver diseases, such as primary biliary cholangitis (PBC) and others, and their therapies, including ileal bile acid transporter (IBAT) inhibitors. In patients with PBC, there are high serum/plasma concentrations of multiple factors, including bile salts, bilirubin, endogenous opioids, lysophosphatidic acid (LPA), autotaxin, and histamine. Bile salts, bilirubin, LPA, and autotaxin affect itch mediators in the skin and sensory nerves, while the endogenous opioid balance affects mediators in the spinal cord. Itch is sensitized by both the peripheral and central nervous systems. Both mechanisms are involved in itch in patients with chronic cholestatic liver disease. Although IBAT inhibitors have been approved for use in pediatric cholestatic conditions, such as progressive familial intrahepatic cholestasis and Alagille syndrome, IBAT inhibition seems to be a promising treatment for chronic refractory itch in patients with PBC. A traditional non-systematic review results in this narrative review. Multidisciplinary cooperation, involving hepatologists, dermatologists, and pharmacists, could provide better treatment for PBC patients suffering from refractory itch. In conclusion, we summarized the existing knowledge on itch caused by chronic cholestatic liver diseases, especially in PBC with a focus on the mechanisms and therapies. This narrative review provides the mechanisms and therapeutic options for itch in patients with chronic cholestatic liver diseases.
Collapse
Affiliation(s)
- Tatsuo Kanda
- Division of Gastroenterology and Hepatology, Uonuma Institute of Community Medicine, Niigata University Medical and Dental Hospital, Uonuma Kikan Hospital, Minamiuonuma 949-7302, Japan
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-9510, Japan (K.H.); (A.S.)
| | - Reina Sasaki-Tanaka
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-9510, Japan (K.H.); (A.S.)
| | - Naruhiro Kimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-9510, Japan (K.H.); (A.S.)
| | - Hiroyuki Abe
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-9510, Japan (K.H.); (A.S.)
| | - Tomoaki Yoshida
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-9510, Japan (K.H.); (A.S.)
| | - Kazunao Hayashi
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-9510, Japan (K.H.); (A.S.)
| | - Akira Sakamaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-9510, Japan (K.H.); (A.S.)
| | - Takeshi Yokoo
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-9510, Japan (K.H.); (A.S.)
| | - Hiroteru Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-9510, Japan (K.H.); (A.S.)
| | - Atsunori Tsuchiya
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Chuo 409-3898, Japan;
| | - Kenya Kamimura
- Department of General Medicine, Niigata University School of Medicine, Niigata 951-9510, Japan;
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata 951-9510, Japan (K.H.); (A.S.)
| |
Collapse
|
3
|
Vitale G, Sciveres M, Mandato C, d'Adamo AP, Di Giorgio A. Genotypes and different clinical variants between children and adults in progressive familial intrahepatic cholestasis: a state-of-the-art review. Orphanet J Rare Dis 2025; 20:80. [PMID: 39984942 PMCID: PMC11846186 DOI: 10.1186/s13023-025-03599-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/06/2025] [Indexed: 02/23/2025] Open
Abstract
INTRODUCTION Progressive Familial intrahepatic cholestasis (PFIC) are rare disorders of bile acid (BAs) secretion and transport with a genetic background. PFIC are paediatric manifestations, but the same variants causing PFIC can also cause cholestasis with a later paediatric onset or adult-onset cholestatic disease (AOCD). Pruritus is a symptom of cholestasis that can be so devastating that it requires a liver transplant (LT) in children; some PFIC types have been described as at risk of liver cancer development. Commonly prescribed medications for PFIC symptoms can partially relieve pruritus without changing the natural history of the disease. Recently, a therapy reducing the intestinal resorption of BAs has been approved; it is effective on both pruritus and cholestasis in PFIC, potentially being a disease-modifying intervention. AREAS COVERED The clinical and genetic characteristics of different PFIC and AOCD are summarized to provide a common background for geneticists and paediatric and adult hepatologists in diagnosis and management. EXPERT OPINION Collaboration between paediatric and adult hepatologists and geneticists will become crucial for cholestatic disease research and patient treatment. Therefore, adult hepatologists will need to learn more about FIC. This might enable the implementation of individualized surveillance in FIC patients and the evaluation of patient family histories.
Collapse
Affiliation(s)
- Giovanni Vitale
- Internal Medicine Unit for the Treatment of Severe Organ Failure, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Marco Sciveres
- Epatologia e Clinica dei Trapianti, Ospedale Pediatrico IRCCS Bambino Gesù, Rome, Italy
| | - Claudia Mandato
- Dipartimento di Medicina, Chirurgia e Odontoiatria "Scuola Medica Salernitana", Section of Pediatrics, Baronissi (Salerno), Italy
| | - Adamo Pio d'Adamo
- Department of Medicine, Surgery and Health Sciences, University of Trieste, Trieste, Italy.
- Institute for Maternal and Child Health-IRCCS "Burlo Garofolo"-Trieste, 34137, Trieste, Italy.
| | - Angelo Di Giorgio
- Pediatric Hepatology Gastroenterology and Transplantation, Hospital Papa Giovanni XIII, Bergamo, Italy
- Department of Medicine, Hospital Santa Maria della Misericordia, University of Udine, Udine, Italy
| |
Collapse
|
4
|
Bufler P, Howard R, Quadrado L, Lacey G, Terner-Rosenthal J, Goldstein A, Vig P, Kelly D. The burden of Alagille syndrome: uncovering the potential of emerging therapeutics - a comprehensive systematic literature review. J Comp Eff Res 2025; 14:e240188. [PMID: 39807752 PMCID: PMC11773862 DOI: 10.57264/cer-2024-0188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/17/2024] [Indexed: 01/16/2025] Open
Abstract
Aim: Alagille syndrome (ALGS) is a rare, cholestatic multiorgan disease associated with bile duct paucity, leading to cholestasis. Clinical symptoms of cholestasis include debilitating pruritus, xanthomas, fat-soluble vitamin deficiencies, growth failure, renal disease and impaired health-related quality of life (HRQoL). The main objective was to review the current literature on the epidemiological, clinical, psychosocial and economic burden of ALGS in view of the development of ileal bile acid transporter (IBAT) inhibitors. Methods: Electronic literature databases were searched in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses checklist. Results: 330 publications were screened, 119 were relevant: 11 randomized controlled trials (RCTs), 21 non-RCTs, 10 HRQoL studies, two studies assessing cost/resource use and 77 epidemiological studies across several databases through 31 July 2024. Studies confirm that patients with ALGS experience cardiac anomalies, impaired growth, renal disease, poor HRQoL, fat-soluble vitamin deficiencies and debilitating pruritus; until the approval of IBAT inhibitors for the treatment of cholestatic pruritus in patients with ALGS, supportive management was the standard of care. Conclusion: This review confirms the substantial clinical, economic and HRQoL burden associated with ALGS and consolidates current treatment evidence. Data from recent trials in ALGS demonstrate the potential impact of IBAT inhibitors to transform lives by improving cholestatic pruritus symptoms, HRQoL and native liver survival.
Collapse
Affiliation(s)
- Philip Bufler
- Department of Pediatric Gastroenterology, Nephrology & Metabolic Diseases, Charité – Universitätsmedizin Berlin, Berlin, Germany
- German Center for Child & Adolescent Health (DZKJ), partner site Berlin, Berlin, Germany
| | - Robin Howard
- Mirum Pharmaceuticals, Inc., Foster City, CA, USA
| | | | - Guy Lacey
- Mirum Pharmaceuticals, Inc., Foster City, CA, USA
| | | | | | - Pamela Vig
- Mirum Pharmaceuticals, Inc., Foster City, CA, USA
| | - Deirdre Kelly
- Guy's & St Thomas' NHS Foundation Trust, London, UK
- Evelina London Women's & Children's Clinical Group, Evelina London Children's Hospital, London, UK
| |
Collapse
|
5
|
Ganschow R, Maucksch C, Rauschkolb P, Schneider MBE. Odevixibat treatment in a child with hypoplastic left heart syndrome and severe cholestatic pruritus: a case report. Front Pediatr 2025; 12:1443338. [PMID: 39917088 PMCID: PMC11799544 DOI: 10.3389/fped.2024.1443338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 12/03/2024] [Indexed: 02/09/2025] Open
Abstract
Liver-related abnormalities are commonly observed in patients with congenital heart disease, and these may lead to secondary manifestations such as pruritus. Odevixibat is an ileal bile acid transporter inhibitor under investigation for the treatment of cholestatic liver diseases. Here, we describe the effects of odevixibat treatment in a pediatric patient with congenital heart disease and severe cholestatic pruritus. A 2-year-old male with Kleefstra syndrome, hypoplastic left heart syndrome, and a history of Giessen procedure and biventricular correction surgery presented to the pediatric cardiology and hepatology outpatient clinics at University Children's Hospital Bonn. Portal hypertension was evident on imaging, and the patient was experiencing severe itching attacks that did not respond to treatment with naltrexone, ursodeoxycholic acid, dimetindene, or rifampicin. Sleep and quality of life were poor. Treatment with odevixibat was initiated off label due to refractory pruritus and elevated serum bile acids. Improvements in pruritus and sleep occurred rapidly with odevixibat and were sustained for the duration of treatment. The patient's serum bile acids decreased from 111 μmol/L before treatment with odevixibat to 24 μmol/L within 1 month of initiating therapy. Relief from pruritus had positive effects on psychomotor development and quality of life. Mild diarrhea lasting 2 days was reported by the patient's mother. In this case report, odevixibat was effective and well tolerated. Together with those of previous studies in patients with progressive familial intrahepatic cholestasis and Alagille syndrome, these results suggest that odevixibat warrants further study as a potential treatment option for patients with cholestatic pruritus of diverse etiologies.
Collapse
Affiliation(s)
- Rainer Ganschow
- Department of Pediatrics, University Children’s Hospital, Bonn, Germany
| | | | | | - Martin B. E. Schneider
- Department of Cardiology, Pediatric Heart Center, University Hospital of Bonn, Bonn, Germany
| |
Collapse
|
6
|
Marx M, Hartleif S, Hilberath J, Berg CP, Tsiflikas I, Singer S, Sturm E. Practical Considerations for Odevixibat Treatment in Patients with Progressive Familial Intrahepatic Cholestasis: A Single-Center Case Series. J Clin Med 2024; 13:7508. [PMID: 39768432 PMCID: PMC11676709 DOI: 10.3390/jcm13247508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/26/2024] [Accepted: 11/28/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Patients with progressive familial intrahepatic cholestasis (PFIC) experience cholestasis-associated symptoms, including severe pruritus. Odevixibat is an ileal bile acid transporter inhibitor indicated for treatment of PFIC in the European Union and for the treatment of pruritus in PFIC in the United States. The aim of the current study was to characterize the real-world effectiveness and safety of odevixibat in patients with PFIC. Methods: This retrospective study included 9 patients with PFIC treated with odevixibat in a single center in Tübingen, Germany. Data were recorded using case report forms. Results: Of the 9 patients (PFIC1, n = 2; PFIC2, n = 7), 5 had improved serum bile acid levels, pruritus, liver function tests, and sleep with odevixibat treatment. Two siblings with periodic relapses of PFIC symptoms also had improved pruritus and sleep within 4 months of treatment. Two siblings with complete loss of bile salt export pump (BSEP) protein did not respond to treatment; both underwent liver transplantation (indications: hepatocellular carcinoma [HCC] manifestation [n = 1] and severe failure to thrive and refractory pruritus [n = 1]). Four patients reported abdominal complaints that were transient or responded to dose reduction; no other safety issues were reported. Conclusions: In this case series, clinical benefits were observed in most patients with PFIC1 and PFIC2 treated with odevixibat. In patients with periodic relapse of PFIC symptoms, ≥3 months of treatment with odevixibat may be required for symptom control. Patients with complete loss of BSEP did not have consistent symptom relief and require careful monitoring. Effectiveness and feasibility results from our cohort demonstrate potential for long-term benefits with odevixibat in real-world treatment of patients with PFIC.
Collapse
Affiliation(s)
- Milena Marx
- Pediatric Gastroenterology and Hepatology, University Children’s Hospital Tübingen, Hoppe-Seyler Str. 1, 72076 Tübingen, Germany
| | - Steffen Hartleif
- Pediatric Gastroenterology and Hepatology, University Children’s Hospital Tübingen, Hoppe-Seyler Str. 1, 72076 Tübingen, Germany
| | - Johannes Hilberath
- Pediatric Gastroenterology and Hepatology, University Children’s Hospital Tübingen, Hoppe-Seyler Str. 1, 72076 Tübingen, Germany
| | - Christoph P. Berg
- Internal Medicine I, University Hospital of Tübingen, Otfried-Müller-Str. 10, 72076 Tübingen, Germany
| | - Ilias Tsiflikas
- Department of Diagnostic and Interventional Radiology, University Hospital of Tübingen, Hoppe-Seyler Str. 3, 72076 Tübingen, Germany
| | - Stephan Singer
- Institute of Pathology, Department for General and Molecular Pathology, University Hospital of Tübingen, Liebermeisterstrasse 8, 72076 Tübingen, Germany
| | - Ekkehard Sturm
- Pediatric Gastroenterology and Hepatology, University Children’s Hospital Tübingen, Hoppe-Seyler Str. 1, 72076 Tübingen, Germany
| |
Collapse
|
7
|
Song H, Xiao L, Meng X, Li Y, Song J. Two episodes of jaundice within 1 year in a teenager. J Hepatol 2024; 81:e218-e220. [PMID: 39419571 DOI: 10.1016/j.jhep.2024.06.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 10/19/2024]
Affiliation(s)
- Hanyi Song
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, PR China
| | - Lin Xiao
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, PR China
| | - Xiangzhen Meng
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, PR China
| | - Yang Li
- Department of Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, PR China.
| | - Junmin Song
- Department of Gastroenterology, Shengjing Hospital of China Medical University, Shenyang 110004, Liaoning Province, PR China.
| |
Collapse
|
8
|
Peng Z, Mpofo M, Jie Z, Ye L. Stereoselective Synthesis of Maralixibat via VO(acac) 2/Schiff Base-Catalyzed Asymmetric Oxidation of Its Sulfide Intermediate. J Org Chem 2024; 89:14510-14514. [PMID: 39292517 DOI: 10.1021/acs.joc.4c01443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
The stereoselective synthesis of maralixibat was achieved by harnessing the chiral transferring effect of the stereogenic R-sulfoxide functionality, which was obtained via the VO(acac)2/Schiff base-catalyzed asymmetric oxidation of a phenylthiophenol prochiral intermediate. The R-sulfoxide intermediate underwent a ring closure reaction to form the seven-membered ring core structure with the desired stereochemistry, ultimately ensuring the drug's exceptional isomeric purity and synthetic efficiency.
Collapse
Affiliation(s)
- Ziyu Peng
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, Hubei Province, P. R. China 430081
| | - Mercy Mpofo
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, Hubei Province, P. R. China 430081
| | - Zheng Jie
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, Hubei Province, P. R. China 430081
| | - Long Ye
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan, Hubei Province, P. R. China 430081
| |
Collapse
|
9
|
Pinon M, Kamath BM. What's new in pediatric genetic cholestatic liver disease: advances in etiology, diagnostics and therapeutic approaches. Curr Opin Pediatr 2024; 36:524-536. [PMID: 38957097 DOI: 10.1097/mop.0000000000001380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
PURPOSE OF REVIEW To highlight recent advances in pediatric cholestatic liver disease, including promising novel prognostic markers and new therapies. FINDINGS Additional genetic variants associated with the progressive familial intrahepatic cholestasis (PFIC) phenotype and new genetic cholangiopathies, with an emerging role of ciliopathy genes, are increasingly being identified. Genotype severity predicts outcomes in bile salt export pump (BSEP) deficiency, and post-biliary diversion serum bile acid levels significantly affect native liver survival in BSEP and progressive familial intrahepatic cholestasis type 1 (FIC1 deficiency) patients. Heterozygous variants in the MDR3 gene have been associated with various cholestatic liver disease phenotypes in adults. Ileal bile acid transporter (IBAT) inhibitors, approved for pruritus in PFIC and Alagille Syndrome (ALGS), have been associated with improved long-term quality of life and event-free survival. SUMMARY Next-generation sequencing (NGS) technologies have revolutionized diagnostic approaches, while discovery of new intracellular signaling pathways show promise in identifying therapeutic targets and personalized strategies. Bile acids may play a significant role in hepatic damage progression, suggesting their monitoring could guide cholestatic liver disease management. IBAT inhibitors should be incorporated early into routine management algorithms for pruritus. Data are emerging as to whether IBAT inhibitors are impacting disease biology and modifying the natural history of the cholestasis.
Collapse
Affiliation(s)
- Michele Pinon
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children, University of Toronto, Toronto, Canada
| | | |
Collapse
|
10
|
Hof WFJ, de Boer JF, Verkade HJ. Emerging drugs for the treatment of progressive familial intrahepatic cholestasis: a focus on phase II and III trials. Expert Opin Emerg Drugs 2024; 29:305-320. [PMID: 38571480 DOI: 10.1080/14728214.2024.2336986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 03/27/2024] [Indexed: 04/05/2024]
Abstract
INTRODUCTION Progressive familial intrahepatic cholestasis (PFIC) is a group of disorders characterized by inappropriate bile formation, causing hepatic accumulation of bile acids and, subsequently, liver injury. Until recently, no approved treatments were available for these patients. AREAS COVERED Recent clinical trials for PFIC treatment have focused on intestine-restricted ileal bile acid transporter (IBAT) inhibitors. These compounds aim to reduce the pool size of bile acids by interrupting their enterohepatic circulation. Other emerging treatments in the pipeline include systemic IBAT inhibitors, synthetic bile acid derivatives, compounds targeting bile acid synthesis via the FXR/FGF axis, and chaperones/potentiators that aim to enhance the residual activity of the mutated transporters. EXPERT OPINION Substantial progress has been made in drug development for PFIC patients during the last couple of years. Although data concerning long-term efficacy are as yet only scarcely available, new therapies have demonstrated robust efficacy in a considerable fraction of patients at least on the shorter term. However, a substantial fraction of PFIC patients do not respond to these novel therapies and thus still requires surgical treatment, including liver transplantation before adulthood. Hence, there is still an unmet medical need for long-term effective medical, preferably non-surgical, treatment for all PFIC patients.
Collapse
Affiliation(s)
- Willemien F J Hof
- Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Freark de Boer
- Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Henkjan J Verkade
- Department of Pediatrics, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
11
|
Yan J, Huang Y, Cao L, Dong Y, Xu Z, Wang F, Gao Y, Feng D, Zhang M. Clinical, pathological and genetic characteristics of 17 unrelated children with Alagille Syndrome. BMC Pediatr 2024; 24:532. [PMID: 39164659 PMCID: PMC11334458 DOI: 10.1186/s12887-024-04973-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Accepted: 07/25/2024] [Indexed: 08/22/2024] Open
Abstract
BACKGROUND Alagille syndrome (ALGS) is a multisystem genetic disorder frequently characterized by hepatic manifestations. This study analyzed the clinical, pathological, and molecular genetic features of ALGS to improve the efficiency of clinical diagnosis. METHODS We retrospectively analyzed the clinical manifestations, pathological examination findings, and genetic testing results of 17 children diagnosed with ALGS based on the revised criteria and hospitalized at our center from January 2012 to January 2022. RESULTS The clinical manifestations are as follows: Cholestasis (16/17, 94%), characteristic facies (15/17, 88%), heart disease (12/16, 75%), butterfly vertebrae (12/17, 71%) and posterior embryotoxon (7/12, 58%). Among the 15 patients who underwent liver pathology examination, 13 (87%) were found to have varying degrees of bile duct paucity. Genetic testing was performed on 15 children, and pathogenic variants of the jagged canonical Notch ligand 1 (JAG1) gene were identified in 13 individuals, including 4 novel variants. No pathogenic variant in the notch homolog 2 (NOTCH2) gene were identified, and 2 children exhibited none of the aforementioned gene pathogenic variants. The median follow-up duration was 7 years. Of the remaining 15 patients (excluding 2 lost to follow-up), 11 remained stable, 4 deteriorated, and no patient died during the follow-up period. CONCLUSIONS Among children diagnosed with ALGS, cholestasis stands as the most common feature. To minimize the risk of misdiagnosis, genetic testing should be performed on children exhibiting cholestasis, followed by the application of the revised diagnostic criteria for ALGS. While pharmacological therapy has shown effectiveness for ALGS patients, liver transplantation may be considered in instances of severe pruritus.
Collapse
Affiliation(s)
- Jianguo Yan
- Senior Department of Liver Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yuanzhi Huang
- Peking University 302 Clinical Medical School, 38 Xueyuan Road, 100191, Beijing, China
- Senior Department of Liver Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Lili Cao
- Senior Department of Liver Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yi Dong
- Senior Department of Liver Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhiqiang Xu
- Senior Department of Liver Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Fuchuan Wang
- Senior Department of Liver Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yinjie Gao
- Senior Department of Liver Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Danni Feng
- Senior Department of Liver Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Min Zhang
- Peking University 302 Clinical Medical School, 38 Xueyuan Road, 100191, Beijing, China.
- Senior Department of Liver Diseases, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|
12
|
Ziegler A, Sæves I, Almaas R. Differences in bile acid profiles between cholestatic diseases - Development of a high throughput assay for dried bloodspots. Clin Chim Acta 2024; 562:119864. [PMID: 38992821 DOI: 10.1016/j.cca.2024.119864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 07/01/2024] [Accepted: 07/08/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Cholestasis causes accumulation of bile acids (BAs) and changes the circulating bile acid profile. Quantification of circulating BAs in dried bloodspots (DBS) may demonstrate obstruction of bile flow and altered bile acid metabolism in the liver. High sample throughput enables rapid screening of cholestatic diseases. MATERIALS AND METHODS Ultra high performance liquid chromatography coupled to tandem mass spectrometry (UHPLC-MS/MS) was used for optimizing separation and detection of the primary unconjugated BAs cholic acid (CA) and chenodeoxycholic acid (CDCA); the secondary unconjugated BAs ursodeoxycholic acid (UDCA), hyodeoxycholic acid (HDCA) and deoxycholic acid (DCA), as well as the glycine- and taurine-conjugated variants of CA, CDCA, DCA and UDCA. Donor blood was obtained to prepare DBS calibrators and quality controls for method development and validation. RESULTS We developed a quantitative bile acid assay with a run-time of two minutes, and one-step sample preparation of 3.2 mm DBS discs. Validation results demonstrated overall good performance and was considered fit for purpose. Children with Alagille syndrome, Aagenaes syndrome and alpha-1 antitrypsin deficiency had increased BAs in DBS from newborn screening samples compared with age matched controls, and had different bile acids profiles. CONCLUSION We propose that our high throughput assay allows bile acid profiling in DBS that can be a valuable assessment tool for early screening of cholestasis in children. Assaying BAs in dried bloodspots is key for early detection of cholestasis, and provides transferability to a newborn screening setting.
Collapse
Affiliation(s)
- Anders Ziegler
- Department of Pediatric Research, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway; Department of Newborn Screening, Oslo University Hospital, Forskningsveien 2B, 0373 Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Ingjerd Sæves
- Department of Newborn Screening, Oslo University Hospital, Forskningsveien 2B, 0373 Oslo, Norway
| | - Runar Almaas
- Department of Pediatric Research, Oslo University Hospital, Sognsvannsveien 20, 0372 Oslo, Norway; Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
13
|
Miethke AG, Moukarzel A, Porta G, Covarrubias Esquer J, Czubkowski P, Ordonez F, Mosca A, Aqul AA, Squires RH, Sokal E, D'Agostino D, Baumann U, D'Antiga L, Kasi N, Laborde N, Arikan C, Lin CH, Gilmour S, Mittal N, Chiou FK, Horslen SP, Huber WD, Jaecklin T, Nunes T, Lascau A, Longpre L, Mogul DB, Garner W, Vig P, Hupertz VF, Gonzalez-Peralta RP, Ekong U, Hartley J, Laverdure N, Ovchinsky N, Thompson RJ. Maralixibat in progressive familial intrahepatic cholestasis (MARCH-PFIC): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Gastroenterol Hepatol 2024; 9:620-631. [PMID: 38723644 DOI: 10.1016/s2468-1253(24)00080-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/08/2024] [Accepted: 03/12/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND Progressive familial intrahepatic cholestasis (PFIC) is a group of autosomal recessive disorders, the most prevalent being BSEP deficiency, resulting in disrupted bile formation, cholestasis, and pruritus. Building on a previous phase 2 study, we aimed to evaluate the efficacy and safety of maralixibat-an ileal bile acid transporter inhibitor-in participants with all types of PFIC. METHODS MARCH-PFIC was a multicentre, randomised, double-blind, placebo-controlled, phase 3 study conducted in 29 community and hospital centres across 16 countries in Europe, the Americas, and Asia. We recruited participants aged 1-17 years with PFIC with persistent pruritus (>6 months; average of ≥1·5 on morning Itch-Reported Outcome [Observer; ItchRO(Obs)] during the last 4 weeks of screening) and biochemical abnormalities or pathological evidence of progressive liver disease, or both. We defined three analysis cohorts. The BSEP (or primary) cohort included only those with biallelic, non-truncated BSEP deficiency without low or fluctuating serum bile acids or previous biliary surgery. The all-PFIC cohort combined the BSEP cohort with participants with biallelic FIC1, MDR3, TJP2, or MYO5B deficiencies without previous surgery but regardless of bile acids. The full cohort had no exclusions. Participants were randomly assigned (1:1) to receive oral maralixibat (starting dose 142·5 μg/kg, then escalated to 570 μg/kg) or placebo twice daily for 26 weeks. The primary endpoint was the mean change in average morning ItchRO(Obs) severity score between baseline and weeks 15-26 in the BSEP cohort. The key secondary efficacy endpoint was the mean change in total serum bile acids between baseline and the average of weeks 18, 22, and 26 in the BSEP cohort. Efficacy analyses were done in the intention-to-treat population (all those randomly assigned) and safety analyses were done in all participants who received at least one dose of study drug. This completed trial is registered with ClinicalTrials.gov, NCT03905330, and EudraCT, 2019-001211-22. FINDINGS Between July 9, 2019, and March 4, 2022, 125 patients were screened, of whom 93 were randomly assigned to maralixibat (n=47; 14 in the BSEP cohort and 33 in the all-PFIC cohort) or placebo (n=46; 17 in the BSEP cohort and 31 in the all-PFIC cohort), received at least one dose of study drug, and were included in the intention-to-treat and safety populations. The median age was 3·0 years (IQR 2·0-7·0) and 51 (55%) of 93 participants were female and 42 (45%) were male. In the BSEP cohort, least-squares mean change from baseline in morning ItchRO(Obs) was -1·7 (95% CI -2·3 to -1·2) with maralixibat versus -0·6 (-1·1 to -0·1) with placebo, with a significant between-group difference of -1·1 (95% CI -1·8 to -0·3; p=0·0063). Least-squares mean change from baseline in total serum bile acids was -176 μmol/L (95% CI -257 to -94) for maralixibat versus 11 μmol/L (-58 to 80) for placebo, also representing a significant difference of -187 μmol/L (95% CI -293 to -80; p=0·0013). The most common adverse event was diarrhoea (27 [57%] of 47 patients on maralixibat vs nine [20%] of 46 patients on placebo; all mild or moderate and mostly transient). There were five (11%) participants with serious treatment-emergent adverse events in the maralixibat group versus three (7%) in the placebo group. No treatment-related deaths occurred. INTERPRETATION Maralixibat improved pruritus and predictors of native liver survival in PFIC (eg, serum bile acids). Maralixibat represents a non-surgical, pharmacological option to interrupt the enterohepatic circulation and improve the standard of care in patients with PFIC. FUNDING Mirum Pharmaceuticals.
Collapse
Affiliation(s)
- Alexander G Miethke
- UC Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Adib Moukarzel
- Pediatric Gastroenterology, Hepatology and Nutrition, Hotel Dieu De France Saint Joseph University Hospital, Beirut, Lebanon
| | - Gilda Porta
- Gastroenterology and Pediatrics, Hospital Sirio Libanes, Sao Paulo, Brazil
| | | | - Piotr Czubkowski
- Department of Gastroenterology, Hepatology, and Immunology, The Children's Memorial Health Institute, Warsaw, Poland
| | - Felipe Ordonez
- Pediatric Gastroenterology, Cardioinfantil Foundation-Lacardio, Bogota, Colombia
| | - Antonella Mosca
- Transplant Department, Ospedale Pediatrico Bambino Gesu Irccs, Lazio, Italy
| | - Amal A Aqul
- Pediatric Gastroenterology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Robert H Squires
- Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Etienne Sokal
- Gastroenterology and Hepatology, UClouvain, Cliniques Universitaires St Luc, Brussels, Belgium
| | - Daniel D'Agostino
- Department of Pediatric Gastro-hepatology, Hospital Italiano De Buenos Aires, Buenos Aires, Argentina
| | - Ulrich Baumann
- Pediatric Gastroenterology and Hepatology, Hannover Medical School, Hannover, Germany
| | - Lorenzo D'Antiga
- Paediatric Hepatology, Gastroenterology and Transplantation, Hospital Papa Giovanni XXIII, Bergamo, Italy; Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Nagraj Kasi
- Pediatric Gastroenterology, Medical University of South Carolina, Charleston, SC, USA
| | - Nolwenn Laborde
- Pediatric Hereditary Metabolic Diseases, Hôpital Des Enfants-CHU Toulouse, Toulouse, France
| | - Cigdem Arikan
- Pediatrics Department, Koc University School of Medicine, Istanbul, Turkey
| | - Chuan-Hao Lin
- Gastroenterology, Hepatology and Nutrition, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Susan Gilmour
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Naveen Mittal
- Department of Pediatrics, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Fang Kuan Chiou
- Paediatric Gastroenterology, Hepatology and Nutrition, KK Women's and Children's Hospital, Singapore
| | - Simon P Horslen
- Pediatrics, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Wolf-Dietrich Huber
- Department of Pediatric Nephrology and Gastroentereology, Medical University of Vienna, Vienna, Austria
| | | | - Tiago Nunes
- Clinical Development, Scientific Affairs and Engagement, Biometrics, and Research Departments, Mirum Pharmaceuticals, Foster City, CA, USA
| | - Anamaria Lascau
- Clinical Development, Scientific Affairs and Engagement, Biometrics, and Research Departments, Mirum Pharmaceuticals, Foster City, CA, USA
| | - Lara Longpre
- Clinical Development, Scientific Affairs and Engagement, Biometrics, and Research Departments, Mirum Pharmaceuticals, Foster City, CA, USA
| | - Douglas B Mogul
- Clinical Development, Scientific Affairs and Engagement, Biometrics, and Research Departments, Mirum Pharmaceuticals, Foster City, CA, USA
| | - Will Garner
- Clinical Development, Scientific Affairs and Engagement, Biometrics, and Research Departments, Mirum Pharmaceuticals, Foster City, CA, USA
| | - Pamela Vig
- Clinical Development, Scientific Affairs and Engagement, Biometrics, and Research Departments, Mirum Pharmaceuticals, Foster City, CA, USA
| | - Vera F Hupertz
- Pediatric Gastroenterology, Hepatology, and Nutrition, Cleveland Clinic Children's, Cleveland, OH, USA
| | - Regino P Gonzalez-Peralta
- Pediatric Gastroenterology, AdventHealth for Children and AdventHealth Transplant Institute, Orlando, FL, USA
| | - Udeme Ekong
- Transplant Hepatology, Pediatric Hepatology, Medstar Georgetown Transplant Institute, Medstar Georgetown University Hospital, Washington, DC, USA
| | - Jane Hartley
- Paediatric Hepatology, Birmingham Women's and Children's Hospital, Birmingham, UK
| | - Noemie Laverdure
- Pediatric Hepatology, Gastroenterology, and Nutrition Unit, Hopital Femme Mere Enfant, Hospices Civils De Lyon, Lyon, France
| | - Nadia Ovchinsky
- Pediatric Gastroenterology and Hepatology, New York University Grossman School of Medicine, New York, NY, USA
| | - Richard J Thompson
- Department of Inflammation Biology, Institute of Liver Studies, King's College London, London, UK
| |
Collapse
|
14
|
Ovchinsky N, Aumar M, Baker A, Baumann U, Bufler P, Cananzi M, Czubkowski P, Durmaz Ö, Fischer R, Indolfi G, Karnsakul WW, Lacaille F, Lee WS, Maggiore G, Rosenthal P, Ruiz M, Sokal E, Sturm E, van der Woerd W, Verkade HJ, Wehrman A, Clemson C, Yu Q, Ni Q, Ruvido J, Manganaro S, Mattsson JP. Efficacy and safety of odevixibat in patients with Alagille syndrome (ASSERT): a phase 3, double-blind, randomised, placebo-controlled trial. Lancet Gastroenterol Hepatol 2024; 9:632-645. [PMID: 38670135 DOI: 10.1016/s2468-1253(24)00074-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 02/23/2024] [Accepted: 02/27/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND In patients with Alagille syndrome, cholestasis-associated clinical features can include high serum bile acids and severe pruritus that can necessitate liver transplantation. We aimed to evaluate the efficacy and safety of the ileal bile acid transporter inhibitor odevixibat versus placebo in patients with Alagille syndrome. METHODS The ASSERT study was a phase 3, double-blind, randomised, placebo-controlled trial that enrolled patients at 21 medical centres or hospitals in ten countries (Belgium, France, Germany, Italy, Malaysia, the Netherlands, Poland, Türkiye, the UK, and the USA). Eligible patients had a genetically confirmed diagnosis of Alagille syndrome, a history of significant pruritus, and elevated serum bile acids. Patients were randomly assigned (2:1) to receive oral odevixibat 120 μg/kg per day or placebo for 24 weeks (in a block size of six and stratified by age: <10 years and ≥10 years to <18 years) via a web-based system. Patients, clinicians, study staff, and people analysing the data were masked to treatment allocation. The primary efficacy endpoint was change in caregiver-reported scratching score (on the PRUCISION instrument; range 0-4) from baseline to weeks 21-24. The prespecified key secondary efficacy endpoint was change in serum bile acid concentration from baseline to the average of weeks 20 and 24. Outcomes were analysed in patients who received at least one dose of study drug (the full analysis set for efficacy outcomes and the safety analysis set for safety outcomes). This trial is registered on ClinicalTrials.gov (NCT04674761) and EudraCT (2020-004011-28), and is completed. FINDINGS Between Feb 26, 2021, and Sept 9, 2022, 52 patients were randomly assigned to receive odevixibat (n=35) or placebo (n=17), all of whom were included in the analysis sets. The median age was 5·5 years (IQR 3·2 to 8·9). 27 (52%) of 52 patients were male and 25 (48%) were female. The mean scratching score was elevated at baseline in both groups (2·8 [SD 0·5] for odevixibat vs 3·0 [0·6] for placebo). Mean scratching scores at weeks 21-24 were 1·1 (0·9) for odevixibat and 2·2 (1·0) for placebo, representing a least-squares (LS) mean change of -1·7 (95% CI -2·0 to -1·3) for odevixibat and -0·8 (-1·3 to -0·3) for placebo, which was significantly greater for odevixibat than for placebo (difference in LS mean change from baseline -0·9 [95% CI -1·4 to -0·3]; p=0·0024). Odevixibat also resulted in significantly greater reductions in mean serum bile acids from baseline versus placebo (237 μmol/L [SD 115] with odevixibat vs 246 μmol/L [121] with placebo) to the average of weeks 20 and 24 (149 μmol/L [102] vs 271 μmol/L [167]; LS mean change -90 μmol/L [95% CI -133 to -48] with odevixibat vs 22 μmol/L [-35 to 80] with placebo; difference in LS mean change -113 μmol/L [95% CI -179 to -47]; p=0·0012). The most common treatment-emergent adverse events were diarrhoea (ten [29%] of 35 patients in the odevixibat group vs one [6%] of 17 in the placebo group) and pyrexia (eight [23%] vs four [24%]). Seven patients had serious treatment-emergent adverse events during the treatment period: five (14%) in the odevixibat group and two (12%) in the placebo group. No patients discontinued treatment and there were no deaths. INTERPRETATION Odevixibat could be an efficacious non-surgical intervention to improve pruritus, reduce serum bile acids, and enhance the standard of care in patients with Alagille syndrome. Longer-term safety and efficacy data of odevixibat in this population are awaited from the ongoing, open-label ASSERT-EXT study. FUNDING Albireo Pharma, an Ipsen company.
Collapse
Affiliation(s)
- Nadia Ovchinsky
- Pediatric Gastroenterology and Hepatology, Hassenfeld Children's Hospital, NYU Langone, New York, NY, USA
| | - Madeleine Aumar
- Univ Lille, CHU Lille, Pediatric Gastroenterology, Hepatology, and Nutrition, Inserm U1286 Infinite, CHU Lille Pôle Enfant, Lille, France
| | - Alastair Baker
- Paediatric Liver Centre, King's College Hospital, London, UK.
| | - Ulrich Baumann
- Pediatric Gastroenterology and Hepatology, Hannover Medical School, Hannover, Germany
| | - Philip Bufler
- Department of Pediatric Gastroenterology, Nephrology, and Metabolic Diseases, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Mara Cananzi
- Pediatric Gastroenterology, Digestive Endoscopy, Hepatology, and Care of the Child With Liver Transplantation, Department of Children's and Women's Health, University Hospital of Padova, Padova, Italy
| | - Piotr Czubkowski
- Department of Gastroenterology, Hepatology, Nutritional Disorders, and Pediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Özlem Durmaz
- Istanbul University, Istanbul Faculty of Medicine, Istanbul, Türkiye
| | - Ryan Fischer
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Children's Mercy Hospital, Kansas City, MO, USA
| | | | - Wikrom W Karnsakul
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Florence Lacaille
- Pediatric Gastroenterology-Hepatology-Nutrition Unit, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Way S Lee
- Department of Paediatrics, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Giuseppe Maggiore
- Hepatology, Gastroenterology, Nutrition, Digestive Endoscopy, and Liver Transplantation Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Philip Rosenthal
- Department of Pediatrics, Division of Gastroenterology, Hepatology, and Nutrition, University of California San Francisco, San Francisco, CA, USA
| | - Mathias Ruiz
- Hospices Civils de Lyon, Hôpital Femme-Mère-Enfant, Service d'Hépato-gastroentérologie et Nutrition Pédiatrique, Bron, France
| | - Etienne Sokal
- Université Catholique de Louvain, Cliniques St Luc, Service de Gastroentérologie et Hépatologie Pédiatrique, Brussels, Belgium
| | - Ekkehard Sturm
- Pediatric Gastroenterology and Hepatology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Wendy van der Woerd
- Department of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, Netherlands
| | - Henkjan J Verkade
- Pediatric Gastroenterology and Hepatology, Department of Pediatrics, University of Groningen, Beatrix Children's Hospital and University Medical Center Groningen, Groningen, Netherlands
| | - Andrew Wehrman
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Komaniecka N, Maroszek S, Drozdzik M, Oswald S, Drozdzik M. Transporter Proteins as Therapeutic Drug Targets-With a Focus on SGLT2 Inhibitors. Int J Mol Sci 2024; 25:6926. [PMID: 39000033 PMCID: PMC11241231 DOI: 10.3390/ijms25136926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/03/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
Membrane transporters interact not only with endogenous substrates but are also engaged in the transport of xenobiotics, including drugs. While the coordinated function of uptake (solute carrier family-SLC and SLCO) and efflux (ATP-binding cassette family-ABC, multidrug and toxic compound extrusion family-MATE) transporter system allows vectorial drug transport, efflux carriers alone achieve barrier functions. The modulation of transport functions was proved to be effective in the treatment strategies of various pathological states. Sodium-glucose cotransporter-2 (SGLT2) inhibitors are the drugs most widely applied in clinical practice, especially in the treatment of diabetes mellitus and heart failure. Sodium taurocholate co-transporting polypeptide (NTCP) serves as virus particles (HBV/HDV) carrier, and inhibition of its function is applied in the treatment of hepatitis B and hepatitis D by myrcludex B. Inherited cholestatic diseases, such as Alagille syndrome (ALGS) and progressive familial intrahepatic cholestasis (PFIC) can be treated by odevixibat and maralixibat, which inhibit activity of apical sodium-dependent bile salt transporter (ASBT). Probenecid can be considered to increase uric acid excretion in the urine mainly via the inhibition of urate transporter 1 (URAT1), and due to pharmacokinetic interactions involving organic anion transporters 1 and 3 (OAT1 and OAT3), it modifies renal excretion of penicillins or ciprofloxacin as well as nephrotoxicity of cidofovir. This review discusses clinically approved drugs that affect membrane/drug transporter function.
Collapse
Affiliation(s)
- Nina Komaniecka
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland; (N.K.); (S.M.); (M.D.)
| | - Sonia Maroszek
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland; (N.K.); (S.M.); (M.D.)
| | - Maria Drozdzik
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland; (N.K.); (S.M.); (M.D.)
| | - Stefan Oswald
- Institute of Pharmacology and Toxicology, Rostock University Medical Center, 18057 Rostock, Germany;
| | - Marek Drozdzik
- Department of Experimental and Clinical Pharmacology, Pomeranian Medical University, 70-111 Szczecin, Poland; (N.K.); (S.M.); (M.D.)
| |
Collapse
|
16
|
Hansen BE, Vandriel SM, Vig P, Garner W, Mogul DB, Loomes KM, Piccoli DA, Rand EB, Jankowska I, Czubkowski P, Gliwicz-Miedzińska D, Gonzales EM, Jacquemin E, Bouligand J, D’Antiga L, Nicastro E, Arnell H, Fischler B, Sokal É, Demaret T, Siew S, Stormon M, Karpen SJ, Romero R, Ebel NH, Feinstein JA, Roberts AJ, Evans HM, Sundaram SS, Chaidez A, Hardikar W, Shankar S, Fischer RT, Lacaille F, Debray D, Lin HC, Jensen MK, Jaramillo C, Karthikeyan P, Indolfi G, Verkade HJ, Larson-Nath C, Quiros-Tejeira RE, Valentino PL, Rogalidou M, Dezsőfi A, Squires JE, Schwarz K, Calvo PL, Bernabeu JQ, Zizzo AN, Nebbia G, Bulut P, Santos-Silva E, Fawaz R, Nastasio S, Karnsakul W, Tamara ML, Busoms CM, Kelly DA, Sandahl TD, Jimenez-Rivera C, Banales JM, Mujawar Q, Li LT, She H, Wang JS, Kim KM, Oh SH, Sanchez MC, Cavalieri ML, Lee WS, Hajinicolaou C, Lertudomphonwanit C, Waisbourd-Zinman O, Arikan C, Alam S, Carvalho E, Melere M, Eshun J, Önal Z, Desai DM, Wiecek S, Pinto RB, Wolters VM, Garcia J, Beretta M, Kerkar N, Brecelj J, Rock N, Lurz E, Blondet N, Shah U, Thompson RJ, Kamath BM. Event-free survival of maralixibat-treated patients with Alagille syndrome compared to a real-world cohort from GALA. Hepatology 2024; 79:1279-1292. [PMID: 38146932 PMCID: PMC11095900 DOI: 10.1097/hep.0000000000000727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/18/2023] [Indexed: 12/27/2023]
Abstract
BACKGROUND AND AIMS Alagille syndrome (ALGS) is characterized by chronic cholestasis with associated pruritus and extrahepatic anomalies. Maralixibat, an ileal bile acid transporter inhibitor, is an approved pharmacologic therapy for cholestatic pruritus in ALGS. Since long-term placebo-controlled studies are not feasible or ethical in children with rare diseases, a novel approach was taken comparing 6-year outcomes from maralixibat trials with an aligned and harmonized natural history cohort from the G lobal AL agille A lliance (GALA) study. APPROACH AND RESULTS Maralixibat trials comprise 84 patients with ALGS with up to 6 years of treatment. GALA contains retrospective data from 1438 participants. GALA was filtered to align with key maralixibat eligibility criteria, yielding 469 participants. Serum bile acids could not be included in the GALA filtering criteria as these are not routinely performed in clinical practice. Index time was determined through maximum likelihood estimation in an effort to align the disease severity between the two cohorts with the initiation of maralixibat. Event-free survival, defined as the time to first event of manifestations of portal hypertension (variceal bleeding, ascites requiring therapy), surgical biliary diversion, liver transplant, or death, was analyzed by Cox proportional hazards methods. Sensitivity analyses and adjustments for covariates were applied. Age, total bilirubin, gamma-glutamyl transferase, and alanine aminotransferase were balanced between groups with no statistical differences. Event-free survival in the maralixibat cohort was significantly better than the GALA cohort (HR, 0.305; 95% CI, 0.189-0.491; p <0.0001). Multiple sensitivity and subgroup analyses (including serum bile acid availability) showed similar findings. CONCLUSIONS This study demonstrates a novel application of a robust statistical method to evaluate outcomes in long-term intervention studies where placebo comparisons are not feasible, providing wide application for rare diseases. This comparison with real-world natural history data suggests that maralixibat improves event-free survival in patients with ALGS.
Collapse
Affiliation(s)
- Bettina E. Hansen
- Department of Hepatology, Toronto General Hospital University Health Network, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Shannon M. Vandriel
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada
| | - Pamela Vig
- Department of Scientific and Medical Affairs, Mirum Pharmaceuticals, Inc., Foster City, California, USA
| | - Will Garner
- Department of Scientific and Medical Affairs, Mirum Pharmaceuticals, Inc., Foster City, California, USA
| | - Douglas B. Mogul
- Department of Scientific and Medical Affairs, Mirum Pharmaceuticals, Inc., Foster City, California, USA
| | - Kathleen M. Loomes
- Department of Pathology and Laboratory Medicine, Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - David A. Piccoli
- Department of Pathology and Laboratory Medicine, Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Elizabeth B. Rand
- Department of Pathology and Laboratory Medicine, Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia and the University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Irena Jankowska
- Department of Gastroenterology, Hepatology, Nutrition Disturbances and Pediatrics, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Piotr Czubkowski
- Department of Gastroenterology, Hepatology, Nutrition Disturbances and Pediatrics, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Dorota Gliwicz-Miedzińska
- Department of Gastroenterology, Hepatology, Nutrition Disturbances and Pediatrics, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Emmanuel M. Gonzales
- Department of Pediatric Hepatology and Liver Transplantation, Service d’Hépatologie et de Transplantation Hépatique Pédiatriques, Centre de Référence de l’Atrésie des Voies Biliaires et des Cholestases Génétiques (AVB-CG), FSMR FILFOIE, ERN RARE LIVER, Hôpital Bicêtre, AP-HP, Faculté de Médecine Paris-Saclay, Le Kremlin-Bicêtre, and Inserm U1193, Hépatinov, Université Paris-Saclay, Orsay, France
| | - Emmanuel Jacquemin
- Department of Pediatric Hepatology and Liver Transplantation, Service d’Hépatologie et de Transplantation Hépatique Pédiatriques, Centre de Référence de l’Atrésie des Voies Biliaires et des Cholestases Génétiques (AVB-CG), FSMR FILFOIE, ERN RARE LIVER, Hôpital Bicêtre, AP-HP, Faculté de Médecine Paris-Saclay, Le Kremlin-Bicêtre, and Inserm U1193, Hépatinov, Université Paris-Saclay, Orsay, France
| | - Jérôme Bouligand
- Department of Molecular Genetics, Pharmacogenetics and Hormonology, Service de Génétique Moléculaire, Pharmacogénétique et Hormonologie, Hôpitaux Universitaires Paris-Saclay, Assistance PubliqueHôpitaux de Paris, Centre Hospitalier Universitaire de Bicêtre, Le Kremlin-Bicêtre, France
| | - Lorenzo D’Antiga
- Department of Pediatric Hepatology, Gastroenterology, and Transplantation, Ospedale Papa Giovanni XXIII, Pediatric Hepatology, Gastroenterology and Transplantation, Bergamo, Italy
| | - Emanuele Nicastro
- Department of Pediatric Hepatology, Gastroenterology, and Transplantation, Ospedale Papa Giovanni XXIII, Pediatric Hepatology, Gastroenterology and Transplantation, Bergamo, Italy
| | - Henrik Arnell
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Astrid Lindgren Children’s Hospital, Karolinska University Hospital and Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Björn Fischler
- Department of Paediatric Gastroenterology, Hepatology and Nutrition, Astrid Lindgren Children’s Hospital, Karolinska University Hospital and CLINTEC, Karolinska Institutet, Stockholm, Sweden
| | - Étienne Sokal
- Department of Pediatric GI and Hepatology, Cliniques Universitaires Saint-Luc, Service De Gastroentérologie & Hépatologie Pédiatrique, Brussels, Belgium
| | - Tanguy Demaret
- Department of Pediatric GI and Hepatology, Cliniques Universitaires Saint-Luc, Service De Gastroentérologie & Hépatologie Pédiatrique, Brussels, Belgium
| | - Susan Siew
- Department of Gastroenterology, The Children’s Hospital at Westmead, Sydney, New South Wales, Australia
| | - Michael Stormon
- Department of Gastroenterology, The Children’s Hospital at Westmead, Sydney, New South Wales, Australia
| | - Saul J. Karpen
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology & Nutrition, Children’s Healthcare of Atlanta & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Rene Romero
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology & Nutrition, Children’s Healthcare of Atlanta & Emory University School of Medicine, Atlanta, Georgia, USA
| | - Noelle H. Ebel
- Department of Pediatrics, Division of Gastroenterology, Stanford University School of Medicine, Palo Alto, California, USA
| | - Jeffrey A. Feinstein
- Department of Pediatrics (Cardiology), Stanford University School of Medicine, Lucile Packard Children’s Hospital, Palo Alto, California, USA
| | - Amin J. Roberts
- Starship Child Health, Department of Paediatric Gastroenterology, Auckland, New Zealand
| | - Helen M. Evans
- Starship Child Health, Department of Paediatric Gastroenterology, Auckland, New Zealand
| | - Shikha S. Sundaram
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics and the Digestive Health Institute, Children’s Hospital of Colorado and University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Alexander Chaidez
- Section of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics and the Digestive Health Institute, Children’s Hospital of Colorado and University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Winita Hardikar
- Department of Gastroenterology and Clinical Nutrition, Royal Children’s Hospital, Melbourne, Victoria, Australia
| | - Sahana Shankar
- Department of Pediatrics, Mazumdar Shaw Medical Center, Narayana Health, Bangalore, Karnataka, India
| | - Ryan T. Fischer
- Department of Gastroenterology, Children’s Mercy Kansas City, Section of Hepatology, Kansas City, Missouri, USA
| | - Florence Lacaille
- Department of Pediatric Gastroenterology and Nutrition, Necker-Enfants Malades Hospital, University of Paris, Paris, France
| | - Dominique Debray
- Department of Pediatric Gastroenterology and Hepatology, Pediatric Liver Unit, National Reference Centre for Rare Pediatric Liver Diseases (Biliary Atresia and Genetic Cholestasis), FILFOIE, ERN RARE LIVER, Necker-Enfants Malades Hospital, University of Paris, Paris, France
| | - Henry C. Lin
- Department of Pediatrics, Division of Pediatric Gastroenterology, Oregon Health and Science University, Portland, Oregon, USA
| | - M. Kyle Jensen
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Primary Children’s Hospital, University of Utah, Salt Lake City, Utah, USA
| | - Catalina Jaramillo
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, Primary Children’s Hospital, University of Utah, Salt Lake City, Utah, USA
| | - Palaniswamy Karthikeyan
- Department of Pediatrics, Leeds Teaching Hospitals NHS Trust, Leeds Children’s Hospital, Leeds, UK
| | - Giuseppe Indolfi
- Department Neurofarba, University of Florence and Meyer Children’s University Hospital, Paediatric and Liver Unit, Florence, Italy
| | - Henkjan J. Verkade
- Department of Pediatrics, Center for Liver, Digestive, and Metabolic Diseases, University Medical Center Groningen, Groningen, The Netherlands
| | - Catherine Larson-Nath
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology, and Nutrition, University of Minnesota, Minneapolis, Minnesota, USA
| | - Ruben E. Quiros-Tejeira
- Department of Pediatrics, Children’s Hospital & Medical Center and University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Pamela L. Valentino
- Department of Pediatrics, Gastroenterology & Hepatology Division, University of Washington, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Maria Rogalidou
- First Department of Pediatrics, Division of Gastroenterology & Hepatology, “Agia Sofia” Children’s Hospital, University of Athens, Athens, Greece
| | - Antal Dezsőfi
- First Department of Paediatrics, Semmelweis University, Budapest, Hungary
| | - James E. Squires
- Department of Pediatrics, Division of Pediatric Gastroenterology and Hepatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kathleen Schwarz
- Department of Pediatrics, Division of Pediatric Gastroenterology, Rady Children’s Hospital San Diego, University of California San Diego, San Diego, California, USA
| | - Pier Luigi Calvo
- Department of Pediatrics, Pediatric Gastroenterology Unit, Regina Margherita Children’s Hospital, Azienda Ospedaliera-Universitaria Citta’ della Salute e della Scienza, Turin, Italy
| | - Jesus Quintero Bernabeu
- Pediatric Hepatology and Liver Transplant Department, Hospital Universitari Vall d’Hebron, Barcelona, Spain
- Department of Liver and Gastrointestinal Diseases, Biodonostia Health Research Institute—Donostia University Hospital, University of the Basque Country (UPV/EHU), San Sebastian, Spain
| | - Andréanne N. Zizzo
- Department of Paediatrics, Division of Paediatric Gastroenterology and Hepatology, London Health Sciences Centre, Children’s Hospital, Western University, London, Ontario, Canada
| | - Gabriella Nebbia
- Department of Pediatric Hepatology, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Servizio di Epatologia Pediatrica, Milan, Italy
| | - Pinar Bulut
- Department of Pediatrics, Division of Pediatric Gastroenterology and Hepatology, Phoenix Children’s Hospital, Phoenix, Arizona, USA
| | - Ermelinda Santos-Silva
- Department of Pediatrics, Centro Hospitalar Universitário Do Porto, Pediatric Gastroenterology Unit, Porto, Portugal
| | - Rima Fawaz
- Department of Pediatrics, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Silvia Nastasio
- Department of Pediatrics, Division of Gastroenterology, Hepatology, & Nutrition, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Wikrom Karnsakul
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - María Legarda Tamara
- Department of Pediatrics, Paediatric Gastroenterology Unit, Cruces University Hospital, Bilbao, Spain
| | - Cristina Molera Busoms
- Department of Gastroenterology, Hepatology and Nutrition, Pediatric Gastroenterology Hepatology and Nutrition Unit, Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Deirdre A. Kelly
- Department of Paediatric Hepatology, Liver Unit, Birmingham Women’s & Children’s Hospital NHS Trust and University of Birmingham, Birmingham, UK
| | | | - Carolina Jimenez-Rivera
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition, Children’s Hospital of Eastern Ontario, Ottawa, Ontario, Canada
| | - Jesus M. Banales
- Department of Hepatology and Gastroenterology, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Biodonostia Health Research Institute—Donostia University Hospital, Universidad del País Vasco (UPV/EHU), San Sebastián, Spain
| | - Quais Mujawar
- Section of Pediatric Gastroenterology, Department of Pediatrics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Li-Ting Li
- Department of Pediatric Gastroenterology, Children’s Hospital of Fudan University, The Center for Pediatric Liver Diseases, Shanghai, China
| | - Huiyu She
- Department of Pediatric Gastroenterology, Children’s Hospital of Fudan University, The Center for Pediatric Liver Diseases, Shanghai, China
| | - Jian-She Wang
- Department of Pediatric Gastroenterology, Children’s Hospital of Fudan University, The Center for Pediatric Liver Diseases, Shanghai, China
| | - Kyung Mo Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children’s Hospital, Seoul, South Korea
| | - Seak Hee Oh
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children’s Hospital, Seoul, South Korea
| | - Maria Camila Sanchez
- Department of Pediatric Gastroenterology, Pediatric Gastroenterology and Hepatology Division, Hospital Italiano Buenos Aires, Buenos Aires, Argentina
| | - Maria Lorena Cavalieri
- Department of Pediatric Gastroenterology, Pediatric Gastroenterology and Hepatology Division, Hospital Italiano Buenos Aires, Buenos Aires, Argentina
| | - Way Seah Lee
- Department of Paediatrics, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Christina Hajinicolaou
- Department of Paediatrics and Child Health, Division of Paediatric Gastroenterology, Chris Hani Baragwanath Academic Hospital, University of the Witwatersrand, Johannesburg, South Africa
| | - Chatmanee Lertudomphonwanit
- Department of Pediatrics, Division of Gastroenterology, Ramathibodi Hospital Mahidol University, Bangkok, Thailand
| | - Orith Waisbourd-Zinman
- Department of Pediatrics, Schneider Children’s Medical Center of Israel, Institute of Gastroenterology, Nutrition and Liver Diseases, Petah Tikva, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Cigdem Arikan
- Department of Pediatric Gastroenterology and Organ Transplant, Koç University School of Medicine, Istanbul, Turkey
| | - Seema Alam
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Elisa Carvalho
- Pediatric Gastroenterology Department, Hospital de Base do Distrito Federal, Hospital da Criança de Brasília, Centro Universitário de Brasília, Brasília, DF, Brazil
| | - Melina Melere
- Departamento de Gastroenterologia e Hepatologia Pediátrica, Pediatric Gastroenterology Service, Hospital da Criança Santo Antônio, Universidade Federal de Ciências da Saúde de Porto Alegre, Complexo Hospitalar Santa Casa, Porto Alegre, RS, Brazil
| | - John Eshun
- Department of Pediatric Gastroenterology, Le Bonheur Children’s Hospital and The University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Zerrin Önal
- Pediatric Gastroenterology, Hepatology and Nutrition Department, Istanbul University Istanbul Medical Faculty, Istanbul, Turkey
| | - Dev M. Desai
- Solid Organ Transplant Department, Children’s Health—Children’s Medical Center, Dallas, Texas, USA
| | - Sabina Wiecek
- Department of Pediatrics, Medical University of Silesia in Katowice, Katowice, Poland
| | - Raquel Borges Pinto
- Department of Pediatric Gastroenterology, Division of Pediatric Gastroenterology of Hospital da Criança Conceição do Grupo Hospitalar Conceição, Porto Alegre, RS, Brazil
| | - Victorien M. Wolters
- Department of Pediatric Gastroenterology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jennifer Garcia
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition/Miami Transplant Institute, University of Miami, Miami, Florida, USA
| | - Marisa Beretta
- Department of Pediatric Intensive Care, Wits Donald Gordon Medical Centre, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Nanda Kerkar
- Department of Pediatrics, Division of Pediatric Gastroenterology, Hepatology and Nutrition, University of Rochester Medical Center, Rochester, New York, USA
| | - Jernej Brecelj
- Pediatric Gastroenterology, Hepatology and Nutrition, and Department of Pediatrics, Faculty of Medicine, University Medical Center Ljubljana, Ljubljana, Slovenia
| | - Nathalie Rock
- Department of Pediatrics, Gynecology, and Obstetrics, Division of Pediatric Specialties, Swiss Pediatric Liver Center, University Hospitals Geneva and University of Geneva, Geneva, Switzerland
| | - Eberhard Lurz
- Department of Pediatrics, Dr. von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Niviann Blondet
- Department of Pediatrics, Gastroenterology & Hepatology Division, University of Washington, Seattle Children’s Hospital, Seattle, Washington, USA
| | - Uzma Shah
- Department of Pediatrics, Harvard Medical School, Massachusetts General Hospital for Children, Boston, Massachusetts, USA
| | - Richard J. Thompson
- Department of Inflammation Biology, Institute of Liver Studies, King’s College London, London, UK
| | - Binita M. Kamath
- Department of Paediatrics, Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Geladari EV, Vallianou NG, Margellou E, Kounatidis D, Sevastianos V, Alexopoulou A. Benign Recurrent Intrahepatic Cholestasis: Where Are We Now? GASTROENTEROLOGY INSIGHTS 2024; 15:156-167. [DOI: 10.3390/gastroent15010011] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
Abstract
Benign recurrent intrahepatic cholestasis (BRIC) stands as a rare genetic contributor to cholestasis, aligning itself within the spectrum of inherited intrahepatic cholestasis syndromes, such as progressive familial intrahepatic cholestasis (PFIC) and intrahepatic cholestasis of pregnancy. Manifesting in infancy or early adulthood, BRIC is marked by recurrent episodes of jaundice accompanied by intense pruritus, enduring from weeks to years across the lifespan. Normal gamma-glutamyl transferase (GGT) levels are a characteristic laboratory finding. Initially considered unlikely to progress to chronic liver disease or cirrhosis, some reports suggest BRIC may evolve into a continuous and progressive form of cholestasis. Moreover, these recurrent cholestatic episodes significantly impact quality of life, and certain mutations elevate the risk of hepatobiliary malignancy. Between episodes, histological findings of centrilobular cholestasis and abnormal laboratory parameters revert to normal, potentially obviating the need for liver biopsy. This review focuses on the genetic aspects of BRIC, its pathophysiology, clinical presentation, and prognosis. Additionally, it outlines triggering factors and available treatment options.
Collapse
Affiliation(s)
- Eleni V. Geladari
- 3rd Department of Internal Medicine & Liver Outpatient Clinic, Evangelismos General Hospital, 45-47 Ipsilantou Str., 10676 Athens, Greece
| | - Natalia G. Vallianou
- 1st Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou Str., 10676 Athens, Greece
| | - Evangelia Margellou
- 1st Department of Internal Medicine, Evangelismos General Hospital, 45-47 Ipsilantou Str., 10676 Athens, Greece
| | - Dimitris Kounatidis
- 2nd Department of Medicine & Research Laboratory, Medical School, National & Kapodistrian University of Athens, Hippokration Hospital, 114 Vasilissis Sofias Str., 11527 Athens, Greece
| | - Vassilios Sevastianos
- 3rd Department of Internal Medicine & Liver Outpatient Clinic, Evangelismos General Hospital, 45-47 Ipsilantou Str., 10676 Athens, Greece
| | - Alexandra Alexopoulou
- 2nd Department of Medicine & Research Laboratory, Medical School, National & Kapodistrian University of Athens, Hippokration Hospital, 114 Vasilissis Sofias Str., 11527 Athens, Greece
| |
Collapse
|
18
|
Quaglia A, Roberts EA, Torbenson M. Developmental and Inherited Liver Disease. MACSWEEN'S PATHOLOGY OF THE LIVER 2024:122-294. [DOI: 10.1016/b978-0-7020-8228-3.00003-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
19
|
Vasudevan AK, Shanmugam N, Rammohan A, Valamparampil JJ, Rinaldhy K, Menon J, Thambithurai R, Namasivayam S, Kaliamoorthy I, Rela M. Outcomes of pediatric liver transplantation for progressive familial intrahepatic cholestasis. Pediatr Transplant 2023; 27:e14600. [PMID: 37675889 DOI: 10.1111/petr.14600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/05/2023] [Accepted: 08/18/2023] [Indexed: 09/08/2023]
Abstract
BACKGROUND Progressive familial intrahepatic cholestasis (PFIC) is a heterogenous group of inherited hepatocellular disorders and the clinical aspects, role of liver transplantation (LT), and its outcomes remain largely unelucidated. We present our data of LT for each type of PFIC and compare their early, and long-term outcomes, highlighting their individual differences and management strategies. METHODS Prospectively collected data over a decade (2011-2022) of children with PFIC who underwent LT was analyzed. The groups (PFIC 1-4) were compared with regard to early and long-term outcomes including attainment of catch-up growth. RESULTS Of 60 children with PFIC who underwent LT, 13, 11, 31 & 5 were of PFIC 1, 2, 3 & 4, respectively. There were no significant differences in gender, PELD scores, BMI, type of grafts, cold and warm ischemia times, intraoperative blood loss, and morbidity among the groups. Post-LT chronic diarrhea was observed in 6 (46.1%) children with PFIC-I, and of them, 3 (23%) developed graft steatohepatitis. Three of these children underwent total internal biliary diversion (TIBD) and on 1-year follow-up, their graft steatosis resolved and they attained catch-up growth. Catch-up growth was significantly poorer in the PFIC1 group (44.4% vs. 88%, 90%, 100% p < .001). Overall 1- and 5-year patient survival of the four PFIC groups (1-4) were 69.2%, 81.8%, 96.8%, 100% & 69.2%, 81.8%, 96.8%, 100%, respectively. CONCLUSION Ours is the largest to-date series of LT for PFIC illustrating their short- and long-term outcomes. While the results for the whole cohort were excellent, those after LT for PFIC1 was relatively poorer as reflected by catch-up growth, graft steatosis, and post-LT diarrhea, which can be optimized by the addition of TIBD during LT.
Collapse
Affiliation(s)
- Anu K Vasudevan
- The Institute of Liver Disease and Transplantation, Dr.Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | - Naresh Shanmugam
- The Institute of Liver Disease and Transplantation, Dr.Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | - Ashwin Rammohan
- The Institute of Liver Disease and Transplantation, Dr.Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | - Joseph J Valamparampil
- The Institute of Liver Disease and Transplantation, Dr.Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | - Kshetra Rinaldhy
- The Institute of Liver Disease and Transplantation, Dr.Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | - Jagadeesh Menon
- The Institute of Liver Disease and Transplantation, Dr.Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | - Ravikumar Thambithurai
- The Institute of Liver Disease and Transplantation, Dr.Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | - Saravanapandian Namasivayam
- The Institute of Liver Disease and Transplantation, Dr.Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | - Ilankumaran Kaliamoorthy
- The Institute of Liver Disease and Transplantation, Dr.Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| | - Mohamed Rela
- The Institute of Liver Disease and Transplantation, Dr.Rela Institute & Medical Centre, Bharath Institute of Higher Education & Research, Chennai, India
| |
Collapse
|
20
|
Sokol RJ, Gonzales EM, Kamath BM, Baker A, Vig P, Mogul DB, Garner W, Hansen BE, Jacquemin E, Thompson RJ. Predictors of 6-year event-free survival in Alagille syndrome patients treated with maralixibat, an ileal bile acid transporter inhibitor. Hepatology 2023; 78:1698-1710. [PMID: 37278241 PMCID: PMC10653287 DOI: 10.1097/hep.0000000000000502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 05/14/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND AND AIMS Refractory pruritus and other complications of cholestasis are indications for liver transplantation (LT) in patients with Alagille syndrome (ALGS). We evaluated predictors of event-free survival and transplant-free survival in patients with ALGS treated with maralixibat (MRX), an ileal bile acid transporter inhibitor. APPROACH AND RESULTS We assessed patients with ALGS from 3 clinical trials of MRX with up to 6 years of follow-up. Event-free survival was defined as the absence of LT, surgical biliary diversion, hepatic decompensation, or death; transplant-free survival was the absence of LT or death. Forty-three potential predictors were evaluated, including age, pruritus (ItchRO[Obs] 0-4 scale), biochemistries, platelets, and serum bile acids. Harrell's concordance statistic assessed goodness-of-fit, and then, Cox proportional hazard models confirmed the statistical significance of the predictors identified. A further analysis was performed to identify cutoffs using a grid search. Seventy-six individuals met the criteria of receiving MRX for ≥48 weeks with laboratory values available at week 48 (W48). The median duration of MRX was 4.7 years (IQR: 1.6-5.8); 16 had events (10 LT, 3 decompensation, 2 death, and 1 surgical biliary diversion). The 6-year event-free survival improved with a clinically meaningful >1-point ItchRO(Obs) reduction from baseline to W48 (88% vs. 57%; p = 0.005), W48 bilirubin < 6.5 mg/dL (90% vs. 43%; p < 0.0001), and W48 serum bile acid < 200 µmol/L (85% vs. 49%; p = 0.001). These parameters were also predictive of 6-year transplant-free survival. CONCLUSIONS Improvement in pruritus by 48 weeks, and lower W48 bilirubin and serum bile acid levels were associated with fewer events. These data may help identify potential markers of disease progression for ALGS patients treated with MRX.
Collapse
Affiliation(s)
- Ronald J. Sokol
- Section of Pediatric Gastroenterology, Hepatology and Nutrition and the Digestive Health Institute, Children’s Hospital Colorado and University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Emmanuel M. Gonzales
- Department of Pediatric Hepatology and Liver Transplantation, National reference center for biliary atresia and genetic cholestasis (AVB-CG), FSMR FILFOIE, ERN RARE-LIVER, AP-HP, Université Paris-Saclay, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- Department of Pediatric Hepatology and Liver Transplantation, Inserm UMR_S 1193, Université Paris-Saclay, FHU Hepatinov, Orsay, France
| | - Binita M. Kamath
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada
| | - Alastair Baker
- Pediatric Liver Department, Pediatric Liver Centre, King’s College Hospital, London, UK
| | - Pamela Vig
- Department of Scientific and Medical Affairs, Mirum Pharmaceuticals Inc., Foster City, California, USA
| | - Douglas B. Mogul
- Department of Epidemiology, Erasmus MC, Rotterdam, The Netherlands
| | - Will Garner
- Department of Scientific and Medical Affairs, Mirum Pharmaceuticals Inc., Foster City, California, USA
| | | | - Emmanuel Jacquemin
- Department of Pediatric Hepatology and Liver Transplantation, National reference center for biliary atresia and genetic cholestasis (AVB-CG), FSMR FILFOIE, ERN RARE-LIVER, AP-HP, Université Paris-Saclay, Bicêtre Hospital, Le Kremlin-Bicêtre, France
- Department of Pediatric Hepatology and Liver Transplantation, Inserm UMR_S 1193, Université Paris-Saclay, FHU Hepatinov, Orsay, France
| | - Richard J. Thompson
- Department of Inflammation Biology, Institute of Liver Studies, King’s College London, London, UK
| |
Collapse
|
21
|
Ayoub MD, Bakhsh AA, Vandriel SM, Keitel V, Kamath BM. Management of adults with Alagille syndrome. Hepatol Int 2023; 17:1098-1112. [PMID: 37584849 PMCID: PMC10522532 DOI: 10.1007/s12072-023-10578-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/30/2023] [Indexed: 08/17/2023]
Abstract
Alagille syndrome (ALGS) is a complex rare genetic disorder that involves multiple organ systems and is historically regarded as a disease of childhood. Since it is inherited in an autosomal dominant manner in 40% of patients, it carries many implications for genetic counselling of patients and screening of family members. In addition, the considerable variable expression and absence of a clear genotype-phenotype correlation, results in a diverse range of clinical manifestations, even in affected individuals within the same family. With recent therapeutic advancements in cholestasis treatment and the improved survival rates with liver transplantation (LT), many patients with ALGS survive into adulthood. Although LT is curative for liver disease secondary to ALGS, complications secondary to extrahepatic involvement remain problematic lifelong. This review is aimed at providing a comprehensive review of ALGS to adult clinicians who will take over the medical care of these patients following transition, with particular focus on certain aspects of the condition that require lifelong surveillance. We also provide a diagnostic framework for adult patients with suspected ALGS and highlight key aspects to consider when determining eligibility for LT in patients with this syndrome.
Collapse
Affiliation(s)
- Mohammed D Ayoub
- Department of Pediatrics, Faculty of Medicine, Rabigh Branch, King Abdulaziz University, Jeddah, Saudi Arabia
- Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, University of Toronto, 555 University Ave, Toronto, ON, M5G 1X8, Canada
| | - Ahmad A Bakhsh
- Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, University of Toronto, 555 University Ave, Toronto, ON, M5G 1X8, Canada
- Department of Pediatrics, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Shannon M Vandriel
- Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, University of Toronto, 555 University Ave, Toronto, ON, M5G 1X8, Canada
| | - Verena Keitel
- Department of Gastroenterology, Hepatology and Infectious Diseases, Faculty of Medicine, Otto Von Guericke University Magdeburg, Magdeburg, Germany
| | - Binita M Kamath
- Division of Gastroenterology, Hepatology, and Nutrition, The Hospital for Sick Children, University of Toronto, 555 University Ave, Toronto, ON, M5G 1X8, Canada.
| |
Collapse
|
22
|
Gonzales E, Gardin A, Almes M, Darmellah-Remil A, Seguin H, Mussini C, Franchi-Abella S, Duché M, Ackermann O, Thébaut A, Habes D, Hermeziu B, Lapalus M, Falguières T, Combal JP, Benichou B, Valero S, Davit-Spraul A, Jacquemin E. Outcomes of 38 patients with PFIC3: Impact of genotype and of response to ursodeoxycholic acid therapy. JHEP Rep 2023; 5:100844. [PMID: 37701337 PMCID: PMC10494458 DOI: 10.1016/j.jhepr.2023.100844] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/16/2023] [Accepted: 06/22/2023] [Indexed: 09/14/2023] Open
Abstract
Background & Aims Progressive familial intrahepatic cholestasis type 3 (PFIC3) is a rare liver disease caused by biallelic variations in ABCB4. Data reporting on the impact of genotype and of response to ursodeoxycholic acid (UDCA) therapy on long-term outcomes are scarce. Methods We retrospectively describe a cohort of 38 patients with PFIC3 with a median age at last follow-up of 19.5 years (range 3.8-53.8). Results Twenty patients presented with symptoms before 1 year of age. Thirty-one patients received ursodeoxycholic acid (UDCA) therapy resulting in serum liver test improvement in 20. Twenty-seven patients had cirrhosis at a median age of 8.1 years of whom 18 received a liver transplant at a median age of 8.5 years. Patients carrying at least one missense variation were more likely to present with positive (normal or decreased) canalicular MDR3 expression in the native liver and had prolonged native liver survival (NLS; median 12.4 years [range 3.8-53.8]). In contrast, in patients with severe genotypes (no missense variation), there was no detectable canalicular MDR3 expression, symptom onset and cirrhosis occurred earlier, and all underwent liver transplantation (at a median age of 6.7 years [range 2.3-10.3]). The latter group was refractory to UDCA treatment, whereas 87% of patients with at least one missense variation displayed an improvement in liver biochemistry in response to UDCA. Biliary phospholipid levels over 6.9% of total biliary lipid levels predicted response to UDCA. Response to UDCA predicted NLS. Conclusions Patients carrying at least one missense variation, with positive canalicular expression of MDR3 and a biliary phospholipid level over 6.9% of total biliary lipid levels were more likely to respond to UDCA and to exhibit prolonged NLS. Impact and implications In this study, data show that genotype and response to ursodeoxycholic acid therapy predicted native liver survival in patients with PFIC3 (progressive familial intrahepatic cholestasis type 3). Patients carrying at least one missense variation, with positive (decreased or normal) immuno-staining for canalicular MDR3, and a biliary phospholipid level over 6.9% of total biliary lipids were more likely to respond to ursodeoxycholic acid therapy and to exhibit prolonged native liver survival.
Collapse
Affiliation(s)
- Emmanuel Gonzales
- Pediatric Hepatology and Liver Transplantation, National Reference Centre for Biliary Atresia and Genetic Cholestasis, FILFOIE, ERN RARE LIVER, France
- Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | - Antoine Gardin
- Pediatric Hepatology and Liver Transplantation, National Reference Centre for Biliary Atresia and Genetic Cholestasis, FILFOIE, ERN RARE LIVER, France
- Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | - Marion Almes
- Pediatric Hepatology and Liver Transplantation, National Reference Centre for Biliary Atresia and Genetic Cholestasis, FILFOIE, ERN RARE LIVER, France
- Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | - Amaria Darmellah-Remil
- Pediatric Hepatology and Liver Transplantation, National Reference Centre for Biliary Atresia and Genetic Cholestasis, FILFOIE, ERN RARE LIVER, France
| | - Hanh Seguin
- Pediatric Hepatology and Liver Transplantation, National Reference Centre for Biliary Atresia and Genetic Cholestasis, FILFOIE, ERN RARE LIVER, France
| | - Charlotte Mussini
- Pathology, Bicêtre Hospital, Assistance Publique – Hôpitaux de Paris, University Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Stéphanie Franchi-Abella
- Pediatric Radiology, Bicêtre Hospital, Assistance Publique – Hôpitaux de Paris, University Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Mathieu Duché
- Pediatric Hepatology and Liver Transplantation, National Reference Centre for Biliary Atresia and Genetic Cholestasis, FILFOIE, ERN RARE LIVER, France
- Pediatric Radiology, Bicêtre Hospital, Assistance Publique – Hôpitaux de Paris, University Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Oanez Ackermann
- Pediatric Hepatology and Liver Transplantation, National Reference Centre for Biliary Atresia and Genetic Cholestasis, FILFOIE, ERN RARE LIVER, France
- Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | - Alice Thébaut
- Pediatric Hepatology and Liver Transplantation, National Reference Centre for Biliary Atresia and Genetic Cholestasis, FILFOIE, ERN RARE LIVER, France
- Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | - Dalila Habes
- Pediatric Hepatology and Liver Transplantation, National Reference Centre for Biliary Atresia and Genetic Cholestasis, FILFOIE, ERN RARE LIVER, France
- Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | - Bogdan Hermeziu
- Pediatric Hepatology and Liver Transplantation, National Reference Centre for Biliary Atresia and Genetic Cholestasis, FILFOIE, ERN RARE LIVER, France
- Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | - Martine Lapalus
- Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | | | | | | | | | - Anne Davit-Spraul
- Biochemistry; Bicêtre Hospital, Assistance Publique – Hôpitaux de Paris, University Paris-Saclay, Le Kremlin-Bicêtre, France
- Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| | - Emmanuel Jacquemin
- Pediatric Hepatology and Liver Transplantation, National Reference Centre for Biliary Atresia and Genetic Cholestasis, FILFOIE, ERN RARE LIVER, France
- Inserm U1193, Hepatinov, University Paris-Saclay, Orsay, France
| |
Collapse
|
23
|
Thompson RJ, Artan R, Baumann U, Calvo PL, Czubkowski P, Dalgic B, D’Antiga L, Di Giorgio A, Durmaz Ö, Gonzalès E, Grammatikopoulos T, Gupte G, Hardikar W, Houwen RH, Kamath BM, Karpen SJ, Lacaille F, Lachaux A, Lainka E, Loomes KM, Mack CL, Mattsson JP, McKiernan P, Ni Q, Özen H, Rajwal SR, Roquelaure B, Shteyer E, Sokal E, Sokol RJ, Soufi N, Sturm E, Tessier ME, van der Woerd WL, Verkade HJ, Vittorio JM, Wallefors T, Warholic N, Yu Q, Horn P, Kjems L. Interim results from an ongoing, open-label, single-arm trial of odevixibat in progressive familial intrahepatic cholestasis. JHEP Rep 2023; 5:100782. [PMID: 37456676 PMCID: PMC10338319 DOI: 10.1016/j.jhepr.2023.100782] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 03/28/2023] [Accepted: 04/16/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND & AIMS PEDFIC 2, an ongoing, open-label, 72-week study, evaluates odevixibat, an ileal bile acid transporter inhibitor, in patients with progressive familial intrahepatic cholestasis. METHODS PEDFIC 2 enrolled and dosed 69 patients across two cohorts; all received odevixibat 120 μg/kg per day. Cohort 1 comprised children from PEDFIC 1, and cohort 2 comprised new patients (any age). We report data through 15 July 2020, with Week 24 of PEDFIC 2 the main time point analysed. This represents up to 48 weeks of cumulative exposure for patients treated with odevixibat from the 24-week PEDFIC 1 study (cohort 1A) and up to 24 weeks of treatment for those who initiated odevixibat in PEDFIC 2 (patients who received placebo in PEDFIC 1 [cohort 1B] or cohort 2 patients). Primary endpoints for this prespecified interim analysis were change from baseline to Weeks 22-24 in serum bile acids (sBAs) and proportion of positive pruritus assessments (≥1-point drop from PEDFIC 2 baseline in pruritus on a 0-4 scale or score ≤1) over the 24-week period. Safety monitoring included evaluating treatment-emergent adverse events (TEAEs). RESULTS In cohort 1A, mean change from PEDFIC 1 baseline to Weeks 22-24 of PEDFIC 2 in sBAs was -201 μmol/L (p <0.0001). For cohort 1B and cohort 2, mean changes from odevixibat initiation to weeks 22-24 in sBAs were -144 and -104 μmol/L, respectively. The proportion of positive pruritus assessments in the first 24-week period of PEDFIC 2 was 33%, 56%, and 62% in cohorts 1A, 1B, and 2, respectively. Most TEAEs were mild or moderate. No drug-related serious TEAEs occurred. CONCLUSIONS Odevixibat in patients with progressive familial intrahepatic cholestasis was generally well tolerated and associated with sustained reductions in sBAs and pruritus. CLINICAL TRIALS REGISTRATION This study is registered at ClinicalTrials.gov (NCT03659916). IMPACT AND IMPLICATIONS Disrupted bile flow is a hallmark feature of patients with progressive familial intrahepatic cholestasis and can result in build-up of bile constituents in the liver with spill over into the bloodstream; other effects that patients can experience include extremely itchy skin, and because not enough bile reaches the gut, patients can have problems digesting food, which may lead to poor growth. Odevixibat is an orally administered medication that shunts bile acids away from the liver. The current study, called PEDFIC 2, suggested that odevixibat can improve the problematic signs and symptoms of progressive familial intrahepatic cholestasis and was generally safe for patients.
Collapse
Affiliation(s)
| | - Reha Artan
- Department of Pediatric Gastroenterology, Akdeniz University, Antalya, Turkey
| | - Ulrich Baumann
- Pediatric Gastroenterology and Hepatology, Hannover Medical School, Hannover, Germany
| | - Pier Luigi Calvo
- Pediatric Gastroenterology Unit, Regina Margherita Children’s Hospital, Azienda Ospedaliera-Città della Salute e della Scienza di Torino, Turin, Italy
| | - Piotr Czubkowski
- Department of Gastroenterology, Hepatology, Nutritional Disorders, and Pediatrics, The Children’s Memorial Health Institute, Warsaw, Poland
| | - Buket Dalgic
- Department of Pediatric Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Lorenzo D’Antiga
- Pediatric Hepatology, Gastroenterology, and Transplantation, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Angelo Di Giorgio
- Pediatric Hepatology, Gastroenterology, and Transplantation, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Özlem Durmaz
- Istanbul University, Istanbul Faculty of Medicine, Istanbul, Turkey
| | - Emmanuel Gonzalès
- Hépatologie et Transplantation Hépatique Pédiatriques, Centre de Référence de l’Atrésie des Voies Biliaires et des Cholestases Génétiques, FSMR FILFOIE, ERN RARE LIVER, Hôpital Bicêtre, AP-HP, Université Paris-Saclay, Hépatinov, Inserm U 1193, Paris, France
| | - Tassos Grammatikopoulos
- Institute of Liver Studies, King’s College London, London, UK
- Pediatric Liver, GI, and Nutrition Center and MowatLabs, King’s College Hospital NHS Trust, London, UK
| | - Girish Gupte
- Liver Unit and Small Bowel Transplantation, Birmingham Women’s and Children’s NHS Foundation Trust, Birmingham, UK
| | - Winita Hardikar
- Department of Gastroenterology, Royal Children's Hospital, Melbourne, Australia
| | - Roderick H.J. Houwen
- Department of Pediatric Gastroenterology at the Wilhelmina Children’s Hospital and University Medical Center, Utrecht, The Netherlands
| | - Binita M. Kamath
- Division of Gastroenterology, Hepatology, and Nutrition, Hospital for Sick Children and the University of Toronto, Toronto, ON, Canada
| | - Saul J. Karpen
- Pediatrics Department, Emory University School of Medicine, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Florence Lacaille
- Pediatric Gastroenterology-Hepatology-Nutrition Unit, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Alain Lachaux
- Hospices Civils de Lyon, Hôpital Femme-Mère-Enfant, Service D’hépatogastoentérologie et Nutrition Pédiatrique, Lyon, France
| | - Elke Lainka
- Department of Pediatric Gastroenterology, Hepatology, and Liver Transplantation, University Children’s Hospital, Essen, Germany
| | - Kathleen M. Loomes
- Department of Pediatrics, Division of Gastroenterology, Hepatology and Nutrition, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Cara L. Mack
- Pediatric Gastroenterology, Hepatology, & Nutrition, Children’s Hospital of Wisconsin, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Patrick McKiernan
- Liver Unit and Small Bowel Transplantation, Birmingham Women’s and Children’s NHS Foundation Trust, Birmingham, UK
| | | | - Hasan Özen
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Sanjay R. Rajwal
- Children’s Liver Unit, Leeds Teaching Hospitals NHS Trust, Leeds Children’s Hospital, Leeds, UK
| | | | - Eyal Shteyer
- Faculty of Medicine, Hebrew University of Jerusalem, Juliet Keidan Department of Pediatric Gastroenterology, Shaare Zedek Medical Center, Jerusalem, Israel
| | - Etienne Sokal
- Université Catholique de Louvain, Cliniques St Luc, Brussels, Belgium
| | - Ronald J. Sokol
- University of Colorado School of Medicine, Children’s Hospital Colorado, Aurora, CO, USA
| | - Nisreen Soufi
- Pediatrics Department, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Ekkehard Sturm
- Pediatric Gastroenterology and Hepatology, University Children’s Hospital Tübingen, Tübingen, Germany
| | - Mary Elizabeth Tessier
- Department of Pediatrics, Section of Pediatric Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine/Texas Children’s Hospital, Houston, TX, USA
| | - Wendy L. van der Woerd
- Department of Pediatric Gastroenterology at the Wilhelmina Children’s Hospital and University Medical Center, Utrecht, The Netherlands
| | - Henkjan J. Verkade
- Department of Pediatrics, University of Groningen, Beatrix Children’s Hospital/University Medical Center Groningen, Groningen, The Netherlands
| | - Jennifer M. Vittorio
- Department of Surgery, Center for Liver Disease and Transplantation, Columbia University Medical Center, New York, NY, USA
| | | | | | - Qifeng Yu
- Albireo Pharma, Inc., Boston, MA, USA
| | | | | |
Collapse
|
24
|
Rodrigo M, Dong X, Chien D, Karnsakul W. Cholestatic Pruritus in Children: Conventional Therapies and Beyond. BIOLOGY 2023; 12:biology12050756. [PMID: 37237568 DOI: 10.3390/biology12050756] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/12/2023] [Accepted: 05/18/2023] [Indexed: 05/28/2023]
Abstract
Pruritus in the setting of cholestatic liver disease is difficult to treat and occurs in patients ranging in age from infancy to adulthood. Likely multifactorial in etiology, this symptom often involves multimodal therapy targeting several pathways and mechanisms proposed in the underlying etiology of cholestatic pruritus. Many patients in both the pediatric and adult populations continue to experience unrelenting pruritus despite maximal conventional therapy. Options are further limited in treating pediatric patients due to sparse data regarding medication safety and efficacy in younger patients. Conventional therapies for the treatment of cholestatic pruritus in children include ursodeoxycholic acid, cholestyramine, hydroxyzine, and rifampin. Certain therapies are more routinely used in the adult populations but with limited data available for use in child and adolescent patients, including opioid antagonists and selective serotonin reuptake inhibitors. Recently, ileal bile acid transport inhibitors have been shown to alleviate pruritus in many children with Alagille syndrome and progressive familial intrahepatic cholestasis and is an additional therapy available for consideration for these patients. Ultimately, surgical options such as biliary diversion or liver transplantation are considered in specific circumstances when medical therapies have been exhausted and pruritus remains debilitating. While further investigation regarding underlying etiologies and effective therapies are needed to better understand itch pathogenesis and treatment in pediatric cholestasis, current considerations beyond conventional management include the use of opioid antagonists, selective serotonin reuptake inhibitors, ileal bile acid transport inhibitors, and surgical intervention.
Collapse
Affiliation(s)
- Minna Rodrigo
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xinzhong Dong
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Daphne Chien
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Wikrom Karnsakul
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
25
|
Levien TL, Baker DE. Formulary Drug Review: Odevixibat. Hosp Pharm 2023; 58:125-133. [PMID: 36890950 PMCID: PMC9986570 DOI: 10.1177/00185787211069036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Each month, subscribers to The Formulary Monograph Service receive 5 to 6 well-documented monographs on drugs that are newly released or are in late phase 3 trials. The monographs are targeted to Pharmacy & Therapeutics Committees. Subscribers also receive monthly 1-page summary monographs on agents that are useful for agendas and pharmacy/nursing in-services. A comprehensive target drug utilization evaluation/medication use evaluation (DUE/MUE) is also provided each month. With a subscription, the monographs are available online to subscribers. Monographs can be customized to meet the needs of a facility. Through the cooperation of The Formulary, Hospital Pharmacy publishes selected reviews in this column. For more information about The Formulary Monograph Service, contact Wolters Kluwer customer service at 866-397-3433.
Collapse
|
26
|
Porwal M, Kumar A, Rastogi V, Maheshwari KK, Verma A. Odevixibat: A Review of a Bioactive Compound for the Treatment of Pruritus Approved by the FDA. Curr Drug Res Rev 2023; 16:CDRR-EPUB-130058. [PMID: 36892028 DOI: 10.2174/2589977515666230308125238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 12/16/2022] [Accepted: 01/11/2023] [Indexed: 03/10/2023]
Abstract
Odevixibat is synthesized through chemical modification of Benzothiazepine's structure. It is a tiny chemical that inhibits the ileal bile acid transporter and is used to treat a variety of cholestatic illnesses, including progressive familial intrahepatic cholestasis (PFIC). For cholestatic pruritus and liver disease development, bile acid transporter inhibition is a unique treatment strategy. Odevixibat reduces enteric bile acid reuptake. Oral odevixibat was also studied in children with cholestatic liver disease. Odevixibat received its first approval in the European Union (EU) in July 2021 for the treatment of PFIC in patients aged 6 months, followed by approval in the USA in August 2021 for the treatment of pruritus in PFIC patients aged 3 months. Bile acids in the distal ileum can be reabsorbed by the ileal sodium/bile acid cotransporter, a transport glycoprotein. Odevixibat is a sodium/bile acid co-transporter reversible inhibitor. An average 3 mg once-daily dose of odevixibat for a week resulted in a 56% reduction in the area under the curve of bile acid. A daily dose of 1.5 mg resulted in a 43% decrease in the area under the curve for bile acid. Odevixibat is also being evaluated in many countries for the treatment of other cholestatic illnesses, including Alagille syndrome and biliary atresia. This article reviews the updated information on odevixibat with respect to its clinical pharmacology, mechanism of action, pharmacokinetics, pharmacodynamics, metabolism, drug-drug interactions, pre-clinical studies, and clinical trials.
Collapse
Affiliation(s)
- Mayur Porwal
- Department of Pharmacology, Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, 244001, Uttar Pradesh, India
| | - Arvind Kumar
- Drug Design Laboratory, School of Pharmaceutical Sciences, IFTM University, Moradabad, 244001, Uttar Pradesh, India
| | - Vaibhav Rastogi
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, 244001, Uttar Pradesh, India
| | | | - Anurag Verma
- Teerthanker Mahaveer College of Pharmacy, Teerthanker Mahaveer University, Moradabad, 244001, Uttar Pradesh, India
| |
Collapse
|
27
|
Nevens F, Trauner M, Manns MP. Primary biliary cholangitis as a roadmap for the development of novel treatments for cholestatic liver diseases †. J Hepatol 2023; 78:430-441. [PMID: 36272496 DOI: 10.1016/j.jhep.2022.10.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 10/07/2022] [Accepted: 10/07/2022] [Indexed: 11/16/2022]
Abstract
The discovery of nuclear receptors and transporters has contributed to the development of new drugs for the treatment of cholestatic liver diseases. Particular progress has been made in the development of second-line therapies for PBC. These new drugs can be separated into compounds primarily targeting cholestasis, molecules targeting fibrogenesis and molecules with immune-mediated action. Finally, drugs aimed at symptom relief (pruritus and fatigue) are also under investigation. Obeticholic acid is currently the only approved second-line therapy for PBC. Drugs in the late phase of clinical development include peroxisome proliferator-activated receptor agonists, norursodeoxycholic acid and NADPH oxidase 1/4 inhibitors.
Collapse
Affiliation(s)
- Frederik Nevens
- Department of Gastroenterology and Hepatology, University Hospital KU Leuven, Belgium; Centre of ERN RARE-LIVER.
| | - Michael Trauner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Austria; Centre of ERN RARE-LIVER
| | - Michael P Manns
- Hannover Medical School, Hannover, Germany; Centre of ERN RARE-LIVER
| |
Collapse
|
28
|
Ebel NH, Goldstein A, Howard R, Mogul DB, Marden JR, Anderson A, Gaburo K, Kirson N, Rosenthal P. Health Care Resource Utilization by Patients with Alagille Syndrome. J Pediatr 2023; 253:144-151.e1. [PMID: 36179890 DOI: 10.1016/j.jpeds.2022.09.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/08/2022] [Accepted: 09/22/2022] [Indexed: 10/14/2022]
Abstract
OBJECTIVE To assess and characterize health care resource utilization (HRU) in children with the rare, genetic, multisystem disorder, Alagille syndrome. STUDY DESIGN This retrospective analysis reviewed commercially insured and Medicaid-insured claims from October 1, 2015 to December 31, 2019 to assess HRU in patients with Alagille syndrome. As there is no specific International Classification ofDiseases-10 code for Alagille syndrome, patients were identified using the following algorithm: ≥1 claim with diagnosis code Q44.7 (other congenital malformations of the liver); <18 years of age, with no history of biliary atresia (International Classification ofDiseases-10 code: Q44.2); and ≥6 months of insurance eligibility prior to diagnosis. HRU was summarized per patient per year over all available claims postdiagnosis. RESULTS A total of 171 commercially insured and 215 Medicaid-insured patients with Alagille syndrome were available for analysis. Annually, commercially insured and Medicaid-insured patients averaged 31 medical visits (range, 1.5-237) and 48 medical visits (range, 0.7-690), respectively. The most common visits were outpatient with the majority encompassing lab/imaging and primary care visits (commercially insured: 21 [range, 0.0-183]; Medicaid-insured: 26 [range, 0.0-609]). Inpatient visits were the highest driver of costs in both the commercial and Medicaid populations. CONCLUSIONS Patients with Alagille syndrome have a substantial HRU burden driven largely by numerous outpatient visits and costly inpatient stays. Given the complexity and variability of Alagille syndrome presentation, patients may benefit from multidisciplinary and subspecialized care.
Collapse
Affiliation(s)
- Noelle H Ebel
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Stanford University, Stanford, CA.
| | | | | | | | | | | | | | | | - Philip Rosenthal
- Division of Pediatric Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, University of California San Francisco (UCSF), San Francisco, CA
| |
Collapse
|
29
|
Gambella A, Kalantari S, Cadamuro M, Quaglia M, Delvecchio M, Fabris L, Pinon M. The Landscape of HNF1B Deficiency: A Syndrome Not Yet Fully Explored. Cells 2023; 12:cells12020307. [PMID: 36672242 PMCID: PMC9856658 DOI: 10.3390/cells12020307] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/15/2023] Open
Abstract
The hepatocyte nuclear factor 1β (HNF1B) gene is involved in the development of specialized epithelia of several organs during the early and late phases of embryogenesis, performing its function mainly by regulating the cell cycle and apoptosis pathways. The first pathogenic variant of HNF1B (namely, R177X) was reported in 1997 and is associated with the maturity-onset diabetes of the young. Since then, more than 230 different HNF1B variants have been reported, revealing a multifaceted syndrome with complex and heterogenous genetic, pathologic, and clinical profiles, mainly affecting the pediatric population. The pancreas and kidneys are the most frequently affected organs, resulting in diabetes, renal cysts, and a decrease in renal function, leading, in 2001, to the definition of HNF1B deficiency syndrome, including renal cysts and diabetes. However, several other organs and systems have since emerged as being affected by HNF1B defect, while diabetes and renal cysts are not always present. Especially, liver involvement has generally been overlooked but recently emerged as particularly relevant (mostly showing chronically elevated liver enzymes) and with a putative relation with tumor development, thus requiring a more granular analysis. Nowadays, HNF1B-associated disease has been recognized as a clinical entity with a broader and more variable multisystem phenotype, but the reasons for the phenotypic heterogeneity are still poorly understood. In this review, we aimed to describe the multifaceted nature of HNF1B deficiency in the pediatric and adult populations: we analyzed the genetic, phenotypic, and clinical features of this complex and misdiagnosed syndrome, covering the most frequent, unusual, and recently identified traits.
Collapse
Affiliation(s)
- Alessandro Gambella
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
- Division of Liver and Transplant Pathology, University of Pittsburgh, Pittsburgh, PA 15232, USA
| | - Silvia Kalantari
- Department of Medical Sciences, University of Turin, 10126 Turin, Italy
| | | | - Marco Quaglia
- Department of Translational Medicine, University of Piemonte Orientale, 28100 Novara, Italy
| | - Maurizio Delvecchio
- Metabolic Disease and Genetics Unit, Giovanni XXIII Children’s Hospital, AOU Policlinico di Bari, 70124 Bari, Italy
- Correspondence:
| | - Luca Fabris
- Department of Molecular Medicine, University of Padova, 35121 Padua, Italy
- Liver Center, Digestive Disease Section, Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Michele Pinon
- Pediatric Gastroenterology Unit, Regina Margherita Children’s Hospital, AOU Città della Salute e della Scienza di Torino, 10126 Turin, Italy
| |
Collapse
|
30
|
Mechanisms of pruritus in cholestasis: understanding and treating the itch. Nat Rev Gastroenterol Hepatol 2023; 20:26-36. [PMID: 36307649 DOI: 10.1038/s41575-022-00687-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2022] [Indexed: 02/01/2023]
Abstract
Pruritus in cholestatic liver diseases can be a major burden and dramatically impair the quality of life of those affected. Here, we provide an update on the latest insights into the molecular pathogenesis of and novel therapeutic approaches for cholestasis-associated itch. Endogenous and exogenous small-molecule pruritogen candidates bind to their receptors on unmyelinated itch C-fibres in the skin. Candidate pruritogens in cholestasis include certain lysophospholipids and sulfated progesterone metabolites, among others, whereas total bile acid or bilirubin conjugates seem unlikely to have a dominant role in the pathogenesis of cholestasis-associated pruritus. Transmission of itch signals via primary, secondary and tertiary itch neurons to the postcentral gyrus and activation of scratch responses offer various targets for therapeutic intervention. At present, evidence-based treatment options for pruritus in fibrosing cholangiopathies, such as primary biliary cholangitis and primary sclerosing cholangitis, are the peroxisome proliferator-associated receptor (PPAR) agonist bezafibrate and the pregnane X receptor (PXR) agonist rifampicin. In pruritus of intrahepatic cholestasis of pregnancy, ursodeoxycholic acid is recommended and might be supported in the third trimester by rifampicin if needed. Alternatively, non-absorbable anion exchange resins, such as cholestyramine, can be administered, albeit with poor trial evidence. Liver transplantation for intolerable refractory pruritus has become an extremely rare therapeutic strategy.
Collapse
|
31
|
Kamath BM, Goldstein A, Howard R, Garner W, Vig P, Marden JR, Billmyer E, Anderson A, Kirson N, Jacquemin E, Gonzales E. Maralixibat Treatment Response in Alagille Syndrome is Associated with Improved Health-Related Quality of Life. J Pediatr 2023; 252:68-75.e5. [PMID: 36096175 DOI: 10.1016/j.jpeds.2022.09.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/08/2022] [Accepted: 09/06/2022] [Indexed: 11/15/2022]
Abstract
OBJECTIVE The objective of this study was to assess the impact of treatment response to the ileal bile acid transporter inhibitor maralixibat on health-related quality of life (HRQoL) in children with Alagille syndrome. STUDY DESIGN This analysis used data from the ICONIC trial, a phase 2 study with a 4-week double-blind, placebo-controlled, randomized drug withdrawal period in children with Alagille syndrome with moderate-to-severe pruritus. Clinically meaningful treatment response to maralixibat was defined a priori as a ≥1-point reduction in the Itch-Reported Outcome (Observer) score, from baseline to week 48. HRQoL was assessed using the Pediatric Quality of Life Inventory Generic Core, Family Impact, and Multidimensional Fatigue scale scores, which were collected via the caregiver. The minimal clinically important difference for HRQoL ranged from 4 to 5 points, depending on the scale. RESULTS Twenty of the 27 patients (74%) included in this analysis achieved an Itch-Reported Outcome (Observer) treatment response at week 48. The mean (SD) change in Multidimensional Fatigue score was +25.8 (23.0) for responders vs -3.1 (19.8) for nonresponders (P = .03). Smaller and non-statistically significant mean changes were observed for the Pediatric Quality of Life Inventory Generic Core and Family Impact scores. Controlling for baseline Family Impact score, responders' Family Impact scores increased an average of 16.9 points over 48 weeks compared with non-responders (P = .05). Smaller and non-statistically significant point estimates were observed for the Pediatric Quality of Life Inventory Generic Core and Multidimensional Fatigue scores. CONCLUSION The significant improvements in pruritus seen with maralixibat at week 48 of the ICONIC study are clinically meaningful and are associated with improved HRQoL. TRIAL REGISTRATION ClinicalTrials.gov: NCT02160782.
Collapse
Affiliation(s)
- Binita M Kamath
- Division of Gastroenterology, Hepatology and Nutrition, The Hospital for Sick Children and University of Toronto, Toronto, Canada.
| | | | | | - Will Garner
- Mirum Pharmaceuticals, Inc., Foster City, CA
| | - Pamela Vig
- Mirum Pharmaceuticals, Inc., Foster City, CA
| | | | | | | | | | - Emmanuel Jacquemin
- Hépatologie et Transplantation Hépatique Pédiatriques, Centre de référence national de l'atrésie des voies biliaires et des cholestases génétiques, FSMR FILFOIE, Hôpital Bicêtre, AP-HP, Université Paris-Saclay, Kremlin-Bicêtre; INSERM UMR-1193, Hepatinov, Université Paris-Saclay, Orsay, France; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| | - Emmanuel Gonzales
- Hépatologie et Transplantation Hépatique Pédiatriques, Centre de référence national de l'atrésie des voies biliaires et des cholestases génétiques, FSMR FILFOIE, Hôpital Bicêtre, AP-HP, Université Paris-Saclay, Kremlin-Bicêtre; INSERM UMR-1193, Hepatinov, Université Paris-Saclay, Orsay, France; European Reference Network on Hepatological Diseases (ERN RARE-LIVER), Hamburg, Germany
| |
Collapse
|
32
|
Bedoyan SM, Lovell OT, Horslen SP, Squires JE. Odevixibat: a promising new treatment for progressive familial intrahepatic cholestasis. Expert Opin Pharmacother 2022; 23:1771-1779. [PMID: 36278881 PMCID: PMC10074157 DOI: 10.1080/14656566.2022.2140040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 10/21/2022] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Progressive familial intrahepatic cholestasis (PFIC) refers to a group of heterogeneous, mostly autosomal recessive disorders resulting from the inability to properly form and excrete bile from hepatocytes. The resulting shared phenotype is one of hepatocellular cholestasis. Clinical management targeting refractory itch and surgical interventions to interrupt the enterohepatic circulation are often pursued with variable efficacy. Recent development of the family of IBAT inhibitor therapeutics has introduced a novel tool in the armamentarium for the treatment of PFIC. AREAS COVERED Data from Phase 3 and 3 clinical trials were reviewed. The primary endpoints in most studies included effect on pruritus, serum bile acid levels, and quality of life metrics, with the duration of the study ranging between 24 and 72 weeks. Most common adverse events included diarrhea, vomiting, and elevation in transaminases. EXPERT OPINION IBAT inhibition with therapeutics such as odevibixat have shown that it is well-tolerated and efficacious in mitigating itch and reducing serum bile acid levels. While the few early published trials with odevixibat have shown good efficacy, what remains to be seen is long-term, sustainable improvement and if or how these medications will supplement or replace the current medical and surgical therapies available for managing PFIC disorders.
Collapse
Affiliation(s)
- Sarah M. Bedoyan
- Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Olya T. Lovell
- Department of Pharmacy, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Simon P. Horslen
- Division of Gastroenterology and Hepatology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - James E. Squires
- Division of Gastroenterology and Hepatology, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
33
|
Gwaltney C, Ivanescu C, Karlsson L, Warholic N, Kjems L, Horn P. Validation of the PRUCISION Instruments in Pediatric Patients with Progressive Familial Intrahepatic Cholestasis. Adv Ther 2022; 39:5105-5125. [PMID: 36066745 PMCID: PMC9525250 DOI: 10.1007/s12325-022-02262-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 07/06/2022] [Indexed: 01/30/2023]
Abstract
INTRODUCTION Patients with cholestatic liver disease, including progressive familial intrahepatic cholestasis (PFIC) and Alagille syndrome, may have debilitating pruritus, and reducing pruritus is a key therapeutic goal. However, few instruments are available that adequately measure pruritus in pediatric patients with cholestatic liver disease. The objectives of the current study were to establish the measurement properties of the novel PRUCISION patient-reported outcome (PRO) and observer-reported outcome (ObsRO) instruments and to estimate a threshold for clinically meaningful change in pruritus score. METHODS The PRO/ObsRO instruments are completed twice daily via electronic diary and include 5-point pictorial responses to assess pruritus. Sleep disturbance and tiredness were quantified using 5-point pictorial responses, yes/no responses, and numerical ratings. Data from PEDFIC 1 (NCT03566238), a phase 3 study evaluating odevixibat efficacy and safety in children with PFIC, were used to assess the psychometric properties of these instruments. Quantitative assessments included evaluation of test-retest reliability, determination of construct validity via convergent and known-group validity analyses, and characterization of sensitivity to change. A threshold for within-patient meaningful change from baseline to week 24 was determined using blinded data from PEDFIC 1 and distribution- and anchor-based analyses. RESULTS Because the majority of patients in PEDFIC 1 were aged < 8 years (n = 52/62) and thus too young to complete the PRO instrument, which was intended for patients aged ≥ 8 years, the small sample size of patients who completed the PRO precluded a full psychometric analysis of the PRO instrument. The ObsRO was completed by a caregiver of every patient in PEDFIC 1. The ObsRO instrument had acceptable test-retest reliability based on intraclass correlation values (most > 0.75). Convergent validity analyses revealed moderate-to-strong correlations (r ≥ 0.3) between baseline ObsRO pruritus scores and baseline Global Impression of Symptoms (GIS) items. In known-groups validity analyses, there were significant differences between baseline groups defined by the GIS for ObsRO pruritus scores and for some sleep disturbance scores. Week 24 ObsRO scores were in the expected direction in groups defined by the Global Impression of Change scale (i.e., improved or not improved); many mean differences between these groups were significant. Sensitivity to change for the ObsRO PRUCISION instrument was also demonstrated by moderate-to-strong Pearson correlations between change from baseline to weeks 21-24 in ObsRO scores and GIS items (r ≥ 0.3). Based on these analyses, a within-patient change of -1.00 from baseline in ObsRO pruritus score was determined to be clinically meaningful. CONCLUSION The PRUCISION ObsRO instrument is reliable, valid, and sensitive to change, supporting its use as a tool to measure pruritus and sleep disturbance in patients with PFIC and other pediatric cholestatic liver diseases.
Collapse
Affiliation(s)
- Chad Gwaltney
- Gwaltney Consulting, 1 Bucks Trail, Westerly, RI, 02891, USA.
| | | | | | | | | | | |
Collapse
|
34
|
Thompson RJ, Arnell H, Artan R, Baumann U, Calvo PL, Czubkowski P, Dalgic B, D'Antiga L, Durmaz Ö, Fischler B, Gonzalès E, Grammatikopoulos T, Gupte G, Hardikar W, Houwen RHJ, Kamath BM, Karpen SJ, Kjems L, Lacaille F, Lachaux A, Lainka E, Mack CL, Mattsson JP, McKiernan P, Özen H, Rajwal SR, Roquelaure B, Shagrani M, Shteyer E, Soufi N, Sturm E, Tessier ME, Verkade HJ, Horn P. Odevixibat treatment in progressive familial intrahepatic cholestasis: a randomised, placebo-controlled, phase 3 trial. Lancet Gastroenterol Hepatol 2022; 7:830-842. [PMID: 35780807 DOI: 10.1016/s2468-1253(22)00093-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Progressive familial intrahepatic cholestasis (PFIC) is a group of inherited paediatric liver diseases resulting from mutations in genes that impact bile secretion. We aimed to evaluate the effects of odevixibat, an ileal bile acid transporter inhibitor, versus placebo in children with PFIC. METHODS Patients eligible for this 24-week, randomised, double-blind, completed, phase 3 study were paediatric outpatients diagnosed with PFIC1 or PFIC2 who had pruritus and elevated serum bile acids at screening. Patients were randomly assigned (1:1:1) using an interactive web-based system to once a day oral placebo, odevixibat 40 μg/kg, or odevixibat 120 μg/kg. Randomisation was done in a block size of six and stratified by PFIC type and patient age; patients, clinicians, and study staff were blinded to treatment allocation. Patients were enrolled at one of 33 global sites. Two primary endpoints were evaluated: proportion of positive pruritus assessments (PPAs; ie, scratching score of ≤1 or ≥1-point decrease as assessed by caregivers using the Albireo observer-reported outcome [ObsRO] PRUCISION instrument) over 24 weeks, and proportion of patients with serum bile acid response (ie, serum bile acids reduced by ≥70% from baseline or concentrations of ≤70 μmol/L) at week 24. Efficacy and safety were analysed in randomly allocated patients who received one or more doses of study drug. This study is registered with ClinicalTrials.gov, NCT03566238. FINDINGS Between June 21, 2018, and Feb 10, 2020, 62 patients (median age 3·2 [range 0·5-15·9] years) were randomly allocated to placebo (n=20), odevixibat 40 μg/kg per day (n=23), or odevixibat 120 μg/kg per day (n=19). Model-adjusted (least squares) mean proportion of PPAs was significantly higher with odevixibat versus placebo (55% [SE 8] in the combined odevixibat group [58% in the 40 μg/kg per day group and 52% in the 120 μg/kg per day group] vs 30% [SE 9] in the placebo group; model-adjusted mean difference 25·0% [95% CI 8·5-41·5]; p=0·0038). The percentage of patients with serum bile acid response was also significantly higher with odevixibat versus placebo (14 [33%] of 42 patients in the combined odevixibat group [10 in the 40 μg/kg per day group and four in the 120 μg/kg per day group] vs none of 20 in the placebo group; adjusting for stratification factor [PFIC type], the proportion difference was 30·7% [95% CI 12·6-48·8; p=0·0030]). The most common treatment-emergent adverse events (TEAEs) were diarrhoea or frequent bowel movements (13 [31%] of 42 for odevixibat vs two [10%] of 20 for placebo) and fever (12 [29%] of 42 vs five [25%] of 20); serious TEAEs occurred in three (7%) of 42 odevixibat-treated patients and in five (25%) of 20 placebo-treated patients. INTERPRETATION In children with PFIC, odevixibat effectively reduced pruritus and serum bile acids versus placebo and was generally well tolerated. Odevixibat, administered as once a day oral capsules, is a non-surgical, pharmacological option to interrupt the enterohepatic circulation in patients with PFIC. FUNDING Albireo Pharma.
Collapse
Affiliation(s)
| | - Henrik Arnell
- Department of Women's and Children's Health, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Reha Artan
- Department of Paediatric Gastroenterology, Akdeniz University, Antalya, Turkey
| | - Ulrich Baumann
- Paediatric Gastroenterology and Hepatology, Hannover Medical School, Hannover, Germany
| | - Pier Luigi Calvo
- Paediatric Gastroenterology Unit, Regina Margherita Children's Hospital, Azienda Ospedaliera Città della Salute e della Scienza di Torino, Turin, Italy
| | - Piotr Czubkowski
- Department of Gastroenterology, Hepatology, Nutritional Disorders and Paediatrics, The Children's Memorial Health Institute, Warsaw, Poland
| | - Buket Dalgic
- Department of Paediatric Gastroenterology, Gazi University Faculty of Medicine, Ankara, Turkey
| | - Lorenzo D'Antiga
- Department of Paediatric Hepatology, Gastroenterology, and Transplantation, Azienda Ospedaliera Papa Giovanni XXIII, Bergamo, Italy
| | - Özlem Durmaz
- Istanbul University Istanbul Faculty of Medicine, Department of Paediatric Gastroenterology and Hepatology, Istanbul, Turkey
| | - Björn Fischler
- Department of Paediatrics, CLINTEC, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Emmanuel Gonzalès
- Hépatologie et Transplantation Hépatique Pédiatriques, Centre de Référence de l'Atrésie des Voies Biliaires et des Cholestases Génétiques, FSMR FILFOIE, ERN RARE LIVER, Hôpital Bicêtre, AP-HP, Université Paris-Saclay, Hépatinov, Inserm U1193, Paris, France
| | - Tassos Grammatikopoulos
- Institute of Liver Studies, King's College London, London, UK; Paediatric Liver, GI and Nutrition Centre and MowatLabs, King's College Hospital NHS Trust, London, UK
| | - Girish Gupte
- Liver Unit and Small Bowel Transplantation, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Winita Hardikar
- Department of Gastroenterology, Royal Children's Hospital, Melbourne, VIC, Australia
| | - Roderick H J Houwen
- Department of Paediatric Gastroenterology, Wilhelmina Children's Hospital and University Medical Centre, Utrecht, Netherlands
| | - Binita M Kamath
- Department of Gastroenterology, Hepatology and Nutrition, Hospital for Sick Children and the University of Toronto, Toronto, ON, Canada
| | - Saul J Karpen
- Paediatrics Department, Emory University School of Medicine, Children's Healthcare of Atlanta, Atlanta, GA, USA
| | | | - Florence Lacaille
- Paediatric Gastroenterology-Hepatology-Nutrition Unit, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Alain Lachaux
- Hospices Civils de Lyon, Hôpital Femme-Mère-Enfant, Service D'hépatogastoentérologie et Nutrition Pédiatrique, Lyon, France
| | - Elke Lainka
- Children's Hospital, Department of Paediatric Gastroenterology, Hepatology, and Transplant Medicine, University Duisburg-Essen, Essen, Germany
| | - Cara L Mack
- Paediatrics Department, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Patrick McKiernan
- Liver Unit and Small Bowel Transplantation, Birmingham Women's and Children's NHS Foundation Trust, Birmingham, UK
| | - Hasan Özen
- Division of Paediatric Gastroenterology, Hepatology, and Nutrition, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Sanjay R Rajwal
- Children's Liver Unit, Leeds Teaching Hospitals NHS Trust, Leeds Children's Hospital, Leeds, UK
| | - Bertrand Roquelaure
- APHM, Service de Pédiatrie Multidisciplinaire, Hôpital de la Timone Enfants, Marseille, France
| | - Mohammad Shagrani
- Department of Liver and SB Transplant and Hepatobiliary-Paediatric Surgery, King Faisal Specialist Hospital and Research Centre-Organ Transplant Centre and College Of Medicine-Alfaisal University, Riyadh, Saudi Arabia
| | - Eyal Shteyer
- Faculty of Medicine, Hebrew University of Jerusalem, Juliet Keidan Department of Paediatric Gastroenterology, Shaare Zedek Medical Centre, Jerusalem, Israel
| | - Nisreen Soufi
- Paediatrics Department, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Ekkehard Sturm
- Paediatric Gastroenterology and Hepatology, University Children's Hospital Tübingen, Tübingen, Germany
| | - Mary Elizabeth Tessier
- Department of Paediatrics, Section of Paediatric Gastroenterology, Hepatology, and Nutrition, Baylor College of Medicine-Texas Children's Hospital, Houston, TX, USA
| | - Henkjan J Verkade
- Department of Paediatrics, University of Groningen, Beatrix Children's Hospital-University Medical Centre Groningen, Groningen, Netherlands
| | | |
Collapse
|
35
|
Shneider BL, Kamath BM, Magee JC, Goodrich NP, Loomes KM, Ye W, Spino C, Alonso EM, Molleston JP, Bezerra JA, Wang KS, Karpen SJ, Horslen SP, Guthery SL, Rosenthal P, Squires RH, Sokol RJ, for the Childhood Liver Disease Research Network (ChiLDReN). Use of funded multicenter prospective longitudinal databases to inform clinical trials in rare diseases-Examination of cholestatic liver disease in Alagille syndrome. Hepatol Commun 2022; 6:1910-1921. [PMID: 35506349 PMCID: PMC9315119 DOI: 10.1002/hep4.1970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 01/28/2023] Open
Abstract
The conduct of long-term conventional randomized clinical trials in rare diseases is very difficult, making evidenced-based drug development problematic. As a result, real-world data/evidence are being used more frequently to assess new therapeutic approaches in orphan diseases. In this investigation, inclusion and exclusion criteria from a published trial of maralixibat in Alagille syndrome (ALGS, ITCH NCT02057692) were applied to a prospective longitudinal cohort of children with cholestasis (LOGIC NCT00571272) to derive contextual comparator data for evolving clinical trials of intestinal bile acid transport inhibitors in ALGS. A natural history/clinical care cohort of 59 participants who met adapted inclusion and exclusion criteria of ITCH was identified from 252 LOGIC participants with ALGS with their native liver. Frequency weighting was used to match the age distribution of ITCH and yielded a cohort (Alagille Syndrome Natural History [ALGS NH]) that was very similar to the baseline status of ITCH participants. During a 2-year prospective follow-up there was a significant reduction in pruritus in the weighted ALGS NH cohort as assessed by the clinician scratch score (-1.43 [0.28] -1.99, -0.87; mean [SEM] 95% confidence interval). During the same time period, the total bilirubin, albumin, and alanine aminotransferase levels were unchanged, whereas platelet count dropped significantly (-65.2 [16.2] -98.3, -32.1). Weighted survival with native liver was 91% at 2 years in the ALGS NH. These investigations provide valuable real-world data that can serve as contextual comparators to current clinical trials, especially those without control populations, and highlight the value and importance of funded multicenter, prospective, natural history studies.
Collapse
Affiliation(s)
| | | | | | | | - Kathleen M. Loomes
- Division of Gastroenterology, Hepatology and NutritionThe Children’s Hospital of Philadelphia and Department of PediatricsPerelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Wen Ye
- University of MichiganAnn ArborMichiganUSA
| | | | - Estella M. Alonso
- Division of Gastroenterology, Hepatology, and NutritionAnn & Robert H. Lurie Children’s Hospital of ChicagoNorthwestern University Feinberg School of MedicineChicagoIllinoisUSA
| | - Jean P. Molleston
- Division of Gastroenterology, Hepatology, and NutritionDepartment of PediatricsRiley Hospital for Children, Indiana UniversityIndianapolisIndianaUSA
| | | | | | - Saul J. Karpen
- Division of Gastroenterology, Hepatology, and NutritionDepartment of PediatricsChildren’s Healthcare of Atlanta and Emory University School of MedicineAtlantaGeorgiaUSA
| | - Simon P. Horslen
- Department of PediatricsSeattle Children’s HospitalUniversity of Washington School of MedicineSeattleWAUSA
| | - Stephen L. Guthery
- Division of Gastroenterology, Hepatology and NutritionDepartment of PediatricsUniversity of Utah, and Intermountain Primary Children’s HospitalSalt Lake CityUtahUSA
| | - Philip Rosenthal
- Department of PediatricsUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Robert H. Squires
- Division of Gastroenterology, Hepatology, and NutritionDepartment of PediatricsUniversity of PittsburghSchool of Medicine and Children’s Hospital of Pittsburgh of University of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Ronald J. Sokol
- Department of Pediatrics‐Gastroenterology, Hepatology and NutritionUniversity of Colorado School of Medicine and Children’s Hospital ColoradoAuroraColoradoUSA
| | | |
Collapse
|
36
|
Shneider BL, Spino CA, Kamath BM, Magee JC, Ignacio RV, Huang S, Horslen SP, Molleston JP, Miethke AG, Kohli R, Leung DH, Jensen MK, Loomes KM, Karpen SJ, Mack C, Rosenthal P, Squires RH, Baker A, Rajwal S, Kelly D, Sokol RJ, Thompson RJ. Impact of long-term administration of maralixibat on children with cholestasis secondary to Alagille syndrome. Hepatol Commun 2022; 6:1922-1933. [PMID: 35672955 PMCID: PMC9315125 DOI: 10.1002/hep4.1992] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 04/03/2022] [Indexed: 11/10/2022] Open
Abstract
There is growing interest in, but limited data about, intestinal bile acid transport inhibitors as treatment for cholestatic liver disease. The current analyses combine two similar randomized placebo-controlled trials with subsequent extension phases investigating the impact of maralixibat in children with severe cholestasis secondary to Alagille Syndrome (n = 57). The primary outcomes were measures of pruritus (ItchRO[Obs]) and clinician scratch scale (CSS), both increasing in severity from 0 to 4) and quality of life (QoL) (Parent PedsQL and Multidimensional Fatigue Scale module [MFS] scaled 0-100 with increased QoL) at week 48 of the extension phase relative to the baseline of the placebo-controlled trials (week 13). Secondary assessments included other clinical and biochemical parameters assessed in participants at week 72 or end of treatment (after week 48). At week 48, statistically and clinically significant least square mean (95% CI) improvements in pruritus and QoL were observed (ItchRO[Obs] -1.59 [-1.81, -1.36], CSS -1.36 [-1.67, -1.05], PedsQL +10.17 [4.48, 15.86], and multidimension fatigue [MFS] +13.97 [7.85, 20.08]). At week 48, serum bile acids, platelet count, and cholesterol decreased, whereas alanine aminotransferase (ALT) increased and total bilirubin (TB) and albumin were stable. Changes were durable at week 72 and end of treatment. There were no deaths; 2 participants underwent liver transplantation. Study drug was discontinued in 9 participants after treatment-emergent adverse events, 6 of which were events of increased ALT or TB. Conclusion: Maralixibat administration was associated with marked improvement in pruritus and QoL. Interpretation of these findings is complicated by the complex natural history of severe cholestasis in Alagille syndrome.
Collapse
Affiliation(s)
- Benjamin L Shneider
- Division of Gastroenterology, Hepatology, and NutritionDepartment of PediatricsBaylor College of Medicine and Texas Children's HospitalHoustonTexasUSA
| | | | | | | | | | | | - Simon P Horslen
- Department of PediatricsSeattle Children's HospitalUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - Jean P Molleston
- Division of Gastroenterology, Hepatology, and NutritionDepartment of PediatricsRiley Hospital for ChildrenIndiana UniversityIndianapolisIndianaUSA
| | - Alexander G Miethke
- Division of Gastroenterology, Hepatology, and NutritionDepartment of PediatricsUniversity of CincinnatiCincinnatiOhioUSA
| | - Rohit Kohli
- Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA.,Liver Transplant ProgramChildren's Hospital-Los AngelesLos AngelesCaliforniaUSA
| | - Daniel H Leung
- Division of Gastroenterology, Hepatology, and NutritionDepartment of PediatricsBaylor College of Medicine and Texas Children's HospitalHoustonTexasUSA
| | - M Kyle Jensen
- Department of PediatricsUniversity of UtahSalt Lake CityUtahUSA
| | - Kathleen M Loomes
- Division of Gastroenterology, Hepatology, and NutritionThe Children's Hospital of Philadelphia and Department of Pediatrics Perelman School of Medicine at the University of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Saul J Karpen
- Division of Gastroenterology, Hepatology, and NutritionDepartment of PediatricsChildren's Healthcare of Atlanta and Emory University School of MedicineAtlantaGeorgiaUSA
| | - Cara Mack
- Department of Pediatrics-Gastroenterology, Hepatology, and NutritionUniversity of Colorado School of Medicine and Children's Hospital ColoradoAuroraColoradoUSA
| | - Philip Rosenthal
- Department of PediatricsUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Robert H Squires
- Division of Gastroenterology, Hepatology, and NutritionDepartment of PediatricsUniversity of Pittsburgh, School of Medicine and Children's Hospital of Pittsburgh of University of Pittsburgh Medical CenterPittsburghPennsylvaniaUSA
| | - Alastair Baker
- Department of Child HealthThe Pediatric Liver CenterKing's College HospitalLondonUK
| | | | - Deirdre Kelly
- Liver UnitBirmingham Women's & Children's HospitalBirminghamUK
| | - Ronald J Sokol
- Department of PediatricsUniversity of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Richard J Thompson
- Department of Child HealthThe Pediatric Liver CenterKing's College HospitalLondonUK
| | | |
Collapse
|
37
|
Heo YA. Odevixibat in progressive familial intrahepatic cholestasis: a profile of its use. DRUGS & THERAPY PERSPECTIVES 2022. [DOI: 10.1007/s40267-022-00926-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
38
|
Bing H, Li YL, Li D, Zhang C, Chang B. Case Report: A Rare Heterozygous ATP8B1 Mutation in a BRIC1 Patient: Haploinsufficiency? Front Med (Lausanne) 2022; 9:897108. [PMID: 35783636 PMCID: PMC9243653 DOI: 10.3389/fmed.2022.897108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 06/01/2022] [Indexed: 11/25/2022] Open
Abstract
Benign recurrent intrahepatic cholestasis (BRIC) is an autosomal recessive disorder characterized by recurrent cholestasis. ATPase class I, type 8B, member 1 (ATP8B1) encodes familial intrahepatic cholestasis 1 (FIC1), which acts as a phosphatidylserine reversing enzyme in the tubule membrane of hepatocytes to mediate the inward translocation of phosphatidylserine (PS). At present, dozens of ATP8B1 pathogenic mutations have been identified that mainly cause BRIC1 and progressive familial intrahepatic cholestasis 1 (PFIC1). The diagnosis of BRIC1 is based on symptoms, laboratory tests, imaging, liver histology, and genetic testing. BRIC1 treatment seeks to prevent recurrence and reduce disease severity. At present, the main treatment methods include ursodeoxycholic acid (UDCA), rifampin, cholestyramine and haemofiltration, and endoscopic nasobiliary drainage (ENBD). Here, we report a 17-year-old patient with cholestasis who has a rare heterozygous ATP8B1 gene mutation (p.T888K). The patient was treated with UDCA, glucocorticoids and haemofiltration, after which bilirubin levels gradually returned to normal. This case was thought to be caused by an ATP8B1 heterozygous mutation, which may be related to haploinsufficiency (HI).
Collapse
Affiliation(s)
- Hao Bing
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
- Department of Gastroenterology, Shengjing Hospital Affiliated by China Medical University, Shenyang, China
| | - Yi-Ling Li
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Dan Li
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Chen Zhang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Bing Chang
- Department of Gastroenterology, First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Bing Chang,
| |
Collapse
|
39
|
Ibrahim SH, Kamath BM, Loomes KM, Karpen SJ. Cholestatic liver diseases of genetic etiology: Advances and controversies. Hepatology 2022; 75:1627-1646. [PMID: 35229330 DOI: 10.1002/hep.32437] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 12/14/2022]
Abstract
With the application of modern investigative technologies, cholestatic liver diseases of genetic etiology are increasingly identified as the root cause of previously designated "idiopathic" adult and pediatric liver diseases. Here, we review advances in the field enhanced by a deeper understanding of the phenotypes associated with specific gene defects that lead to cholestatic liver diseases. There are evolving areas for clinicians in the current era specifically regarding the role for biopsy and opportunities for a "sequencing first" approach. Risk stratification based on the severity of the genetic defect holds promise to guide the decision to pursue primary liver transplantation versus medical therapy or nontransplant surgery, as well as early screening for HCC. In the present era, the expanding toolbox of recently approved therapies for hepatologists has real potential to help many of our patients with genetic causes of cholestasis. In addition, there are promising agents under study in the pipeline. Relevant to the current era, there are still gaps in knowledge of causation and pathogenesis and lack of fully accepted biomarkers of disease progression and pruritus. We discuss strategies to overcome the challenges of genotype-phenotype correlation and draw attention to the extrahepatic manifestations of these diseases. Finally, with attention to identifying causes and treatments of genetic cholestatic disorders, we anticipate a vibrant future of this dynamic field which builds upon current and future therapies, real-world evaluations of individual and combined therapeutics, and the potential incorporation of effective gene editing and gene additive technologies.
Collapse
Affiliation(s)
- Samar H Ibrahim
- Division of Pediatric GastroenterologyMayo ClinicRochesterMinnesotaUSA
| | - Binita M Kamath
- The Hospital for Sick ChildrenUniversity of TorontoTorontoOntarioCanada
| | - Kathleen M Loomes
- The Children's Hospital of Philadelphia and Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Saul J Karpen
- Emory University School of Medicine and Children's Healthcare of AtlantaAtlantaGeorgiaUSA
| |
Collapse
|
40
|
Martínez-García J, Molina A, González-Aseguinolaza G, Weber ND, Smerdou C. Gene Therapy for Acquired and Genetic Cholestasis. Biomedicines 2022; 10:biomedicines10061238. [PMID: 35740260 PMCID: PMC9220166 DOI: 10.3390/biomedicines10061238] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/16/2022] Open
Abstract
Cholestatic diseases can be caused by the dysfunction of transporters involved in hepatobiliary circulation. Although pharmacological treatments constitute the current standard of care for these diseases, none are curative, with liver transplantation being the only long-term solution for severe cholestasis, albeit with many disadvantages. Liver-directed gene therapy has shown promising results in clinical trials for genetic diseases, and it could constitute a potential new therapeutic approach for cholestatic diseases. Many preclinical gene therapy studies have shown positive results in animal models of both acquired and genetic cholestasis. The delivery of genes that reduce apoptosis or fibrosis or improve bile flow has shown therapeutic effects in rodents in which cholestasis was induced by drugs or bile duct ligation. Most studies targeting inherited cholestasis, such as progressive familial intrahepatic cholestasis (PFIC), have focused on supplementing a correct version of a mutated gene to the liver using viral or non-viral vectors in order to achieve expression of the therapeutic protein. These strategies have generated promising results in treating PFIC3 in mouse models of the disease. However, important challenges remain in translating this therapy to the clinic, as well as in developing gene therapy strategies for other types of acquired and genetic cholestasis.
Collapse
Affiliation(s)
- Javier Martínez-García
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, 31008 Pamplona, Spain; (J.M.-G.); (A.M.); (G.G.-A.)
| | - Angie Molina
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, 31008 Pamplona, Spain; (J.M.-G.); (A.M.); (G.G.-A.)
| | - Gloria González-Aseguinolaza
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, 31008 Pamplona, Spain; (J.M.-G.); (A.M.); (G.G.-A.)
- Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
- Vivet Therapeutics S.L., 31008 Pamplona, Spain
| | - Nicholas D. Weber
- Vivet Therapeutics S.L., 31008 Pamplona, Spain
- Correspondence: (N.D.W.); (C.S.); Tel.: +34-948194700 (N.D.W. & C.S.)
| | - Cristian Smerdou
- Division of Gene Therapy and Regulation of Gene Expression, Cima Universidad de Navarra, 31008 Pamplona, Spain; (J.M.-G.); (A.M.); (G.G.-A.)
- Instituto de Investigación Sanitaria de Navarra (IdISNA), 31008 Pamplona, Spain
- Correspondence: (N.D.W.); (C.S.); Tel.: +34-948194700 (N.D.W. & C.S.)
| |
Collapse
|
41
|
Ileal Bile Acid Transporter Inhibition Reduces Post-Transplant Diarrhea and Growth Failure in FIC1 Disease—A Case Report. CHILDREN 2022; 9:children9050669. [PMID: 35626847 PMCID: PMC9139332 DOI: 10.3390/children9050669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/07/2022] [Accepted: 05/03/2022] [Indexed: 12/12/2022]
Abstract
Familial intrahepatic cholestasis 1 (FIC1) disease is a genetic disorder characterized by hepatic and gastrointestinal disease due to ATP8B1 deficiency, often requiring liver transplantation (LT). Extrahepatic symptoms, such as diarrhea, malabsorption, and failure to thrive, do not improve and instead may be aggravated after LT. We describe a patient with FIC1 disease who underwent LT at 2 years, 8 months of age. After LT, the child developed severe refractory diarrhea and failed to thrive. The response to bile acid resins was unsatisfactory, and the parents declined our recommendation for partial external biliary diversion (PEBD). Quality of life was extremely impaired, especially due to severe diarrhea, making school attendance impossible. Attempting to reduce the total bile acids, we initiated off-label use of the ileal bile acid transporter (IBAT) inhibitor Elobixibat (Goofice™), later converted to Odevixibat (Bylvay™). After six months of treatment, the patient showed less stool output, increased weight and height, and improved physical energy levels. The child could now pursue higher undergraduate education. In our patient with FIC1 disease, the use of IBAT inhibitors was effective in treating chronic diarrhea and failure to thrive. This approach is novel; further investigations are needed to clarify the exact mode of action in this condition.
Collapse
|
42
|
Tanimizu N. The neonatal liver: Normal development and response to injury and disease. Semin Fetal Neonatal Med 2022; 27:101229. [PMID: 33745829 DOI: 10.1016/j.siny.2021.101229] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The liver emerges from the ventral foregut endoderm around 3 weeks in human and 1 week in mice after fertilization. The fetal liver works as a hematopoietic organ and then develops functions required for performing various metabolic reactions in late fetal and neonatal periods. In parallel with functional differentiation, the liver establishes three dimensional tissue structures. In particular, establishment of the bile excretion system consisting of bile canaliculi of hepatocytes and bile ducts of cholangiocytes is critical to maintain healthy tissue status. This is because hepatocytes produce bile as they functionally mature, and if allowed to remain within the liver tissue can lead to cytotoxicity. In this review, we focus on epithelial tissue morphogenesis in the perinatal period and cholestatic liver diseases caused by abnormal development of the biliary system.
Collapse
Affiliation(s)
- Naoki Tanimizu
- Department of Tissue Development and Regeneration, Research Institute for Frontier Medicine, Sapporo Medical University School of Medicine, S-1, W-17, Chuo-ku, Sapporo, 060-8556, Japan.
| |
Collapse
|
43
|
Alam S, Lal BB. Recent updates on progressive familial intrahepatic cholestasis types 1, 2 and 3: Outcome and therapeutic strategies. World J Hepatol 2022; 14:98-118. [PMID: 35126842 PMCID: PMC8790387 DOI: 10.4254/wjh.v14.i1.98] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/17/2021] [Accepted: 11/30/2021] [Indexed: 02/06/2023] Open
Abstract
Recent evidence points towards the role of genotype to understand the phenotype, predict the natural course and long term outcome of patients with progressive familial intrahepatic cholestasis (PFIC). Expanded role of the heterozygous transporter defects presenting late needs to be suspected and identified. Treatment of pruritus, nutritional rehabilitation, prevention of fibrosis progression and liver transplantation (LT) in those with end stage liver disease form the crux of the treatment. LT in PFIC has its own unique issues like high rates of intractable diarrhoea, growth failure; steatohepatitis and graft failure in PFIC1 and antibody-mediated bile salt export pump deficiency in PFIC2. Drugs inhibiting apical sodium-dependent bile transporter and adenovirus-associated vector mediated gene therapy hold promise for future.
Collapse
Affiliation(s)
- Seema Alam
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Bikrant Bihari Lal
- Department of Pediatric Hepatology, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| |
Collapse
|
44
|
Henkel SAF, Salgado CM, Reyes-Mugica M, Soltys KA, Strauss K, Mazariegos GV, Squires RH, McKiernan PJ, Zhang X, Squires JE. Long-term liver transplant outcomes for progressive familial intrahepatic cholestasis type 1: The Pittsburgh experience. Pediatr Transplant 2021; 25:e14108. [PMID: 34339082 DOI: 10.1111/petr.14108] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/26/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Progressive familial intrahepatic cholestasis type 1 (PFIC1) arises from biallelic variants in the ATP8B1 gene that annul FIC1 activity, resulting in progressive liver disease. Liver transplant (LT) is indicated in refractory disease; however, post-LT complications including worsening diarrhea and steatohepatitis progressing to fibrosis with graft loss have been reported. We aim to describe long-term outcomes of PFIC1 LT recipients at our center, focusing on the histological changes of the allografts. METHODS We assessed 7 PFIC1 patients post-LT at the Children's Hospital of Pittsburgh (CHP). All pre-transplant, explant, and sequential post-transplant pathology samples were reviewed. Continuous data are presented as the mean ± SD. We compared the pre- and post-transplant height and weight z-scores using Wilcoxon signed-rank test. RESULTS Seven (29% male) patients with PFIC1 received a LT (n = 6) or had post-LT care (n = 1) at CHP. Six had confirmed or suspected identical genetic. At a mean follow-up of 10.9 years, both patient survival and graft survival were 100%. Diarrhea persisted (n = 3) or newly developed (n = 4) in all patients after LT contributing to ongoing growth failure, with mean z-scores -2.63 (weight) and -2.98 (height) at follow-up. Histologically, allograft steatosis was common but was not accompanied by significant inflammation, ballooning, or fibrosis. CONCLUSION We show that extrahepatic disease persists and near-universal allograft steatosis occurs. However, at a mean follow-up period of over 10 years, no patients developed steatohepatitis or significant fibrosis, and both patient survival and graft survival are excellent.
Collapse
Affiliation(s)
- Sarah A F Henkel
- Department of Pediatric Gastroenterology and Hepatology, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Claudia M Salgado
- Department of Pathology, University of Pittsburgh, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Miguel Reyes-Mugica
- Department of Pathology, University of Pittsburgh, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Kyle A Soltys
- Hillman Center for Pediatric Transplantation, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Kevin Strauss
- Clinic for Special Children, Strasburg, PA, USA.,Department of Pediatrics, Penn Medicine-Lancaster General Hospital, Lancaster, PA, USA
| | - George V Mazariegos
- Hillman Center for Pediatric Transplantation, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Robert H Squires
- Department of Pediatric Gastroenterology and Hepatology, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick J McKiernan
- Department of Pediatric Gastroenterology and Hepatology, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Xingyu Zhang
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - James E Squires
- Department of Pediatric Gastroenterology and Hepatology, University of Pittsburgh School of Medicine, Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
45
|
Kriegermeier A, Taylor S. Apical sodium-dependent bile acid transporter inhibition in children with Alagille syndrome. Lancet 2021; 398:1544-1545. [PMID: 34755616 DOI: 10.1016/s0140-6736(21)01447-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 06/18/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Alyssa Kriegermeier
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Division of Gastroenterology, Hepatology and Nutrition at Ann & Robert H Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA.
| | - Sarah Taylor
- Department of Pediatrics, Northwestern University, Feinberg School of Medicine, Division of Gastroenterology, Hepatology and Nutrition at Ann & Robert H Lurie Children's Hospital of Chicago, Chicago, IL 60611, USA
| |
Collapse
|
46
|
Gonzales E, Hardikar W, Stormon M, Baker A, Hierro L, Gliwicz D, Lacaille F, Lachaux A, Sturm E, Setchell KDR, Kennedy C, Dorenbaum A, Steinmetz J, Desai NK, Wardle AJ, Garner W, Vig P, Jaecklin T, Sokal EM, Jacquemin E. Efficacy and safety of maralixibat treatment in patients with Alagille syndrome and cholestatic pruritus (ICONIC): a randomised phase 2 study. Lancet 2021; 398:1581-1592. [PMID: 34755627 DOI: 10.1016/s0140-6736(21)01256-3] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Revised: 05/05/2021] [Accepted: 05/27/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Alagille syndrome is a rare genetic disease that often presents with severe cholestasis and pruritus. There are no approved drugs for management. Maralixibat, an apical, sodium-dependent, bile acid transport inhibitor, prevents enterohepatic bile acid recirculation. We evaluated the safety and efficacy of maralixibat for children with cholestasis in Alagille syndrome. METHODS ICONIC was a placebo-controlled, randomised withdrawal period (RWD), phase 2b study with open-label extension in children (aged 1-18 years) with Alagille syndrome (NCT02160782). Eligible participants had more than three times the normal serum bile acid (sBA) levels and intractable pruritus. After 18 weeks of maralixibat 380 μg/kg once per day, participants were randomly assigned (1:1) to continue maralixibat or receive placebo for 4 weeks. Subsequently, all participants received open-label maralixibat until week 48. During the long-term extension (204 weeks reported), doses were increased up to 380 μg/kg twice per day. The primary endpoint was the mean sBA change during the RWD in participants with at least 50% sBA reduction by week 18. Cholestastic pruritus was assessed using observer-rated, patient-rated, and clinician-rated 0-4 scales. The safety population was defined as all participants who had received at least one dose of maralixibat. This trial was registered with ClinicalTrials.gov, NCT02160782, and is closed to recruitment. FINDINGS Between Oct 28, 2014, and Aug 14, 2015, 31 participants (mean age 5·4 years [SD 4·25]) were enrolled and 28 analysed at week 48. Of the 29 participants who entered the randomised drug withdrawal period, ten (34%) were female and 19 (66%) were male. In the RWD, participants switched to placebo had significant increases in sBA (94 μmol/L, 95% CI 23 to 164) and pruritus (1·7 points, 95% CI 1·2 to 2·2), whereas participants who continued maralixibat maintained treatment effect. This study met the primary endpoint (least square mean difference -117 μmol/L, 95% CI -232 to -2). From baseline to week 48, sBA (-96 μmol/L, -162 to -31) and pruritus (-1·6 pts, -2·1 to -1·1) improved. In participants who continued to week 204 (n=15) all improvements were maintained. Maralixibat was generally safe and well tolerated throughout. The most frequent adverse events were gastrointestinal related. Most adverse events were self-limiting in nature and mild-to-moderate in severity. INTERPRETATION In children with Alagille syndrome, maralixibat is, to our knowledge, the first agent to show durable and clinically meaningful improvements in cholestasis. Maralixibat might represent a new treatment paradigm for chronic cholestasis in Alagille syndrome. FUNDING Mirum Pharmaceuticals.
Collapse
Affiliation(s)
- Emmanuel Gonzales
- Hépatologie et Transplantation Hépatique Pédiatriques, Centre de référence de l'atrésie des voies biliaires et des cholestases génétiques, FSMR FILFOIE, ERN RARE LIVER, Hôpital Bicêtre, AP-HP, Université Paris-Saclay, Hépatinov, Inserm U 1193, Le Kremlin-Bicêtre, France.
| | - Winita Hardikar
- Department of Gastroenterology, The Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| | - Michael Stormon
- Department of Gastroenterology, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Alastair Baker
- Department of Child Health, The Paediatric Liver Centre, King's College Hospital, London, UK
| | - Loreto Hierro
- Servicio de Hepatologìa y Trasplante Pediátrico, Hospital Universitario La Paz, Madrid, Spain
| | - Dorota Gliwicz
- Department of Gastroenterology, Hepatology, Feeding Disorders and Pediatrics, Children's Memorial Health Institute, Warsaw, Poland
| | - Florence Lacaille
- Gastroenterology-Hepatology-Nutrition Unit, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Alain Lachaux
- Service de Gastroentérologie, Hépatologie et Nutrition Pédiatriques, Centre de référence de la maladie de Wilson et autres maladies rares liées au cuivre, Centre de référence de l'atrésie des voies biliaires et cholestases génétiques, Hôpital Femme-Mère-Enfant, Lyon, France
| | - Ekkehard Sturm
- Paediatric Gastroenterology/Hepatology, University Hospital for Children and Adolescents, Tübingen, Germany
| | - Kenneth D R Setchell
- Division of Pathology and Laboratory Medicine, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | - Alejandro Dorenbaum
- Pediatrics - Immunology and Allergy, Stanford Medical School, Stanford University, Palo Alto, CA, USA
| | | | | | | | | | - Pamela Vig
- Mirum Pharmaceuticals, Foster City, CA, USA
| | | | - Etienne M Sokal
- UCLouvain, Cliniques Universitaires Saint Luc, Service de Gastroentérologie Hépatologie Pédiatrique, Brussels, Belgium
| | - Emmanuel Jacquemin
- Hépatologie et Transplantation Hépatique Pédiatriques, Centre de référence de l'atrésie des voies biliaires et des cholestases génétiques, FSMR FILFOIE, ERN RARE LIVER, Hôpital Bicêtre, AP-HP, Université Paris-Saclay, Hépatinov, Inserm U 1193, Le Kremlin-Bicêtre, France
| |
Collapse
|
47
|
Felzen A, Verkade HJ. The spectrum of Progressive Familial Intrahepatic Cholestasis diseases: Update on pathophysiology and emerging treatments. Eur J Med Genet 2021; 64:104317. [PMID: 34478903 DOI: 10.1016/j.ejmg.2021.104317] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 08/11/2021] [Accepted: 08/22/2021] [Indexed: 02/07/2023]
Abstract
The Progressive Familial Intrahepatic Cholestasis (PFIC) disease spectrum encompasses a variety of genetic diseases that affect the bile production and the secretion of bile acids. Typically, the first presentation of these diseases is in early childhood, frequently followed by a severe course necessitating liver transplantation before adulthood. Except for transplantation, treatment modalities have been rather limited and frequently only aim at the symptoms of cholestasis, such as cholestatic pruritus. In recent years, progress has been made in understanding the pathophysiology of these diseases and new treatment modalities have been emerging. Herewith we summarize the latest developments in the field and formulate the current key questions and opportunities for further progress.
Collapse
Affiliation(s)
- Antonia Felzen
- Pediatric Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, the Netherlands
| | - Henkjan J Verkade
- Pediatric Gastroenterology and Hepatology, University Medical Center Groningen, University of Groningen, the Netherlands.
| |
Collapse
|
48
|
Wang K, Chan YC, So PK, Liu X, Feng L, Cheung WT, Lee SST, Au SWN. Structure of mouse cytosolic sulfotransferase SULT2A8 provides insight into sulfonation of 7α-hydroxyl bile acids. J Lipid Res 2021; 62:100074. [PMID: 33872606 PMCID: PMC8134075 DOI: 10.1016/j.jlr.2021.100074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/09/2021] [Accepted: 04/09/2021] [Indexed: 11/17/2022] Open
Abstract
Cytosolic sulfotransferases (SULTs) catalyze the transfer of a sulfonate group from the cofactor 3'-phosphoadenosine 5'-phosphosulfate to a hydroxyl (OH) containing substrate and play a critical role in the homeostasis of endogenous compounds, including hormones, neurotransmitters, and bile acids. In human, SULT2A1 sulfonates the 3-OH of bile acids; however, bile acid metabolism in mouse is dependent on a 7α-OH sulfonating SULT2A8 via unknown molecular mechanisms. In this study, the crystal structure of SULT2A8 in complex with adenosine 3',5'-diphosphate and cholic acid was resolved at a resolution of 2.5 Å. Structural comparison with human SULT2A1 reveals different conformations of substrate binding loops. In addition, SULT2A8 possesses a unique substrate binding mode that positions the target 7α-OH of the bile acid close to the catalytic site. Furthermore, mapping of the critical residues by mutagenesis and enzyme activity assays further highlighted the importance of Lys44 and His48 for enzyme catalysis and Glu237 in loop 3 on substrate binding and stabilization. In addition, limited proteolysis and thermal shift assays suggested that the cofactor and substrates have protective roles in stabilizing SULT2A8 protein. Together, the findings unveil the structural basis of bile acid sulfonation targeting 7α-OH and shed light on the functional diversity of bile acid metabolism across species.
Collapse
Affiliation(s)
- Kai Wang
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong.
| | - Yan-Chun Chan
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Pui-Kin So
- University Research Facility in Life Sciences, The Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Xing Liu
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Lu Feng
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Wing-Tai Cheung
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Susanna Sau-Tuen Lee
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Shannon Wing-Ngor Au
- Faculty of Science, School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong; Center for Protein Science and Crystallography, School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong.
| |
Collapse
|
49
|
Liver Steatosis and Diarrhea After Liver Transplantation for Progressive Familial Intrahepatic Cholestasis Type 1: Can Biliary Diversion Solve These Problems? J Pediatr Gastroenterol Nutr 2021; 72:341-342. [PMID: 33230072 DOI: 10.1097/mpg.0000000000002990] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
50
|
van de Peppel IP, Verkade HJ, Jonker JW. Metabolic consequences of ileal interruption of the enterohepatic circulation of bile acids. Am J Physiol Gastrointest Liver Physiol 2020; 319:G619-G625. [PMID: 32938201 DOI: 10.1152/ajpgi.00308.2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The enterohepatic circulation of bile acids comprises a tightly regulated process of hepatic bile acid secretion, intestinal reabsorption and transport back to the liver. Disruption of this process has significant consequences for gastrointestinal, liver and whole body homeostasis and therefore offers opportunities for therapeutic intervention. In this review we discuss the effects of (pharmacological) interruption of the enterohepatic circulation at different levels. Recently, several studies have been published on ileal interruption of the enterohepatic circulation of bile acids, targeting the apical-sodium dependent bile acid transporter (ASBT, SLC10A2), as therapy for various diseases. However, ambiguous results have been reported and in-depth mechanistic insights are lacking. Here we discuss these novel studies and review the current knowledge on the consequences of ASBT inhibition and its potential effects on physiology and metabolism.
Collapse
Affiliation(s)
- Ivo P van de Peppel
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Henkjan J Verkade
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Johan W Jonker
- Section of Molecular Metabolism and Nutrition, Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|