1
|
Caddeo A, Romeo S. Precision medicine and nucleotide-based therapeutics to treat steatotic liver disease. Clin Mol Hepatol 2025; 31:S76-S93. [PMID: 39103998 PMCID: PMC11925435 DOI: 10.3350/cmh.2024.0438] [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: 06/08/2024] [Revised: 07/31/2024] [Accepted: 08/04/2024] [Indexed: 08/07/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) is a complex multifactorial disease and becoming the leading cause of liver-related morbidity and mortality. MASLD spans from isolated steatosis to metabolic dysfunction-associated steatohepatitis (MASH), that may progress to cirrhosis and hepatocellular carcinoma (HCC). Genetic, metabolic, and environmental factors strongly contribute to the heterogeneity of MASLD. Lifestyle intervention and weight loss represent a viable treatment for MASLD. Moreover, Resmetirom, a thyroid hormone beta receptor agonist, has recently been approved for MASLD treatment. However, most individuals treated did not respond to this therapeutic, suggesting the need for a more tailored approach to treat MASLD. Oligonucleotide-based therapies, namely small-interfering RNA (siRNA) and antisense oligonucleotide (ASO), have been recently developed to tackle MASLD by reducing the expression of genes influencing MASH progression, such as PNPLA3 and HSD17B13. Here, we review the latest progress made in the synthesis and development of oligonucleotide-based agents targeting genetic determinants of MASH.
Collapse
Affiliation(s)
- Andrea Caddeo
- Department of Biomedical Sciences, Unit of Oncology and Molecular Pathology, University of Cagliari, Cagliari, Italy
| | - Stefano Romeo
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
- Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
- Department of Cardiology, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Medicine, Endocrinology (H7) Karolinska Institute and Hospital, Huddinge, Stockholm, Sweden
| |
Collapse
|
2
|
Innes H, Morgan MY, Hampe J, Stickel F, Buch S. The rs72613567:TA polymorphism in HSD17B13 is associated with survival benefit after development of hepatocellular carcinoma. Aliment Pharmacol Ther 2023; 58:623-631. [PMID: 37470344 DOI: 10.1111/apt.17638] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/08/2023] [Accepted: 06/27/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND The influence of genetic factors on survival following a diagnosis of hepatocellular carcinoma (HCC) remains unclear. AIM To assess whether genetic polymorphisms influencing the susceptibility to develop HCC are also associated with HCC prognosis. METHODS We included United Kingdom Biobank (UKB) participants diagnosed with HCC after study enrolment. The primary outcome was all-cause mortality. Patients were followed from the date of HCC diagnosis to death or the registry completion date. Five HCC susceptibility loci were investigated: rs738409 (PNPLA3), rs58542926 (TM6SF2); rs72613567 (HSD17B13); rs2242652 (TERT) and rs708113 (WNT3A). The associations between these genetic variants and HCC mortality risk were assessed using Cox regression, adjusted for age, sex, ethnicity, aetiology, severity of the underlying liver disease and receipt of curative HCC treatment. RESULTS The final sample included 439 patients; 74% had either non-alcoholic fatty liver disease or alcohol-related liver disease. There were 321 deaths during a mean follow-up of 1.9 years per participant. Kaplan-Meier survival estimates at 1, 3 and 5 years were 53.2%, 31.2% and 22.6% respectively. In multivariate analysis, rs72613567:TA (HSD17B13) was the only genetic susceptibility variant significantly associated with all-cause mortality risk (aHR: 0.74; 95% CI: 0.61-0.90; p = 0.003). Other associated factors were Baveno stage 3-4 (aHR: 1.65; 95% CI: 1.05-2.59; p = 0.03) and HCC treatment with curative intent (aHR: 0.25; 95% CI: 0.17-0.37; p < 0.001). CONCLUSIONS The rs72613567:TA polymorphism in HSD17B13 is not only associated with a reduction in the risk of developing HCC but with a survival benefit in HCC once established. Therapeutic inhibition of HSD17B13 may augment survival in individuals with HCC.
Collapse
Affiliation(s)
- Hamish Innes
- School of Health and Life Sciences, Glasgow Caledonian University, Glasgow, UK
- Public Health Scotland, Glasgow, UK
- Division of Epidemiology and Public Health, University of Nottingham, Nottingham, UK
| | - Marsha Y Morgan
- Division of Medicine, UCL Institute for Liver & Digestive Health, Royal Free Campus, University College London, London, UK
| | - Jochen Hampe
- Medical Department, University Hospital Dresden, TUD Dresden University of Technology, Dresden, Germany
| | - Felix Stickel
- Department of Gastroenterology and Hepatology, University Hospital of Zurich, Zurich, Switzerland
| | - Stephan Buch
- Medical Department, University Hospital Dresden, TUD Dresden University of Technology, Dresden, Germany
| |
Collapse
|
3
|
Tavaglione F, Jamialahmadi O, Valenti L, Romeo S. Fatty liver disease genetic risk variants and interference on sex hormones. Liver Int 2023; 43:958-961. [PMID: 37161848 DOI: 10.1111/liv.15562] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 03/10/2023] [Indexed: 05/11/2023]
Affiliation(s)
- Federica Tavaglione
- Clinical Medicine and Hepatology Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Oveis Jamialahmadi
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
| | - Luca Valenti
- Department of Transfusion Medicine, Precision Medicine, Biological Resource Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, Wallenberg Laboratory, University of Gothenburg, Gothenburg, Sweden
- Cardiology Department, Sahlgrenska University Hospital, Gothenburg, Sweden
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, Catanzaro, Italy
| |
Collapse
|
4
|
Smyk W, Papapostoli I, Żorniak M, Sklavounos P, Blukacz Ł, Madej P, Koutsou A, Weber SN, Friesenhahn-Ochs B, Cebula M, Bosowska J, Solomayer EF, Hartleb M, Milkiewicz P, Lammert F, Stokes CS, Krawczyk M. Liver phenotypes in PCOS: Analysis of exogenous and inherited risk factors for liver injury in two European cohorts. Liver Int 2023; 43:1080-1088. [PMID: 36683562 DOI: 10.1111/liv.15527] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 01/17/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023]
Abstract
BACKGROUND & AIMS Fatty liver disease (FLD) is common in women with polycystic ovary syndrome (PCOS). Here, we use non-invasive tests to quantify liver injury in women with PCOS and analyse whether FLD-associated genetic variants contribute to liver phenotypes in PCOS. METHODS Prospectively, we recruited women with PCOS and controls at two university centres in Germany and Poland. Alcohol abuse was regarded as an exclusion criterion. Genotyping of variants associated with FLD was performed using TaqMan assays. Liver stiffness measurements (LSM), controlled attenuation parameters (CAP) and non-invasive HSI, FLI, FIB-4 scores were determined to assess hepatic steatosis and fibrosis. RESULTS A total of 42 German (age range 18-53 years) and 143 Polish (age range 18-40 years) women with PCOS, as well as 245 German and 289 Polish controls were recruited. In contrast to Polish patients, Germans were older, presented with more severe metabolic profiles and had significantly higher LSM (median 5.9 kPa vs. 3.8 kPa). In the German cohort, carriers of the PNPLA3 p.I148M risk variant had an increased LSM (p = .01). In the Polish cohort, the minor MTARC1 allele was linked with significantly lower serum aminotransferases activities, whereas the HSD17B13 polymorphism was associated with lower concentrations of 17-OH progesterone, total testosterone, and androstenedione (all p < .05). CONCLUSIONS FLD is common in women with PCOS. Its extent is modulated by both genetic and metabolic risk factors. Genotyping of variants associated with FLD might help to stratify the risk of liver disease progression in women suffering from PCOS.
Collapse
Affiliation(s)
- Wiktor Smyk
- Department of Gastroenterology and Hepatology, Medical University of Gdansk, Gdansk, Poland
| | - Ifigeneia Papapostoli
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Michał Żorniak
- Department of Gastroenterology, Hepatology and Oncology, Center for Postgraduate Medical Education, Warsaw, Poland.,Department of Oncological Gastroenterology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Panagiotis Sklavounos
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Łukasz Blukacz
- Department of Gynecological Endocrinology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Paweł Madej
- Department of Gynecological Endocrinology, Faculty of Medical Sciences in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Andreani Koutsou
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Susanne N Weber
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Bettina Friesenhahn-Ochs
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Maciej Cebula
- Department of Radiology and Nuclear Medicine, Faculty of Medicine in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Joanna Bosowska
- Department of Radiology and Nuclear Medicine, Faculty of Medicine in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Erich-Franz Solomayer
- Department for Gynecology, Obstetrics and Reproductive Medicine, Saarland University Medical Center, Saarland University, Homburg, Germany
| | - Marek Hartleb
- Department of Gastroenterology, Faculty of Medicine in Katowice, Medical University of Silesia in Katowice, Katowice, Poland
| | - Piotr Milkiewicz
- Liver and Internal Medicine Unit, Medical University of Warsaw, Warsaw, Poland.,Translational Medicine Group, Pomeranian Medical University, Szczecin, Poland
| | - Frank Lammert
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany.,Hannover Medical School, Hannover, Germany
| | - Caroline S Stokes
- Food and Health Research Group, Faculty of Life Sciences, Humboldt-Universität zu Berlin, Berlin, Germany.,Department of Molecular Toxicology, German Institute of Human Nutrition, Potsdam-Rehbrücke, Germany
| | - Marcin Krawczyk
- Department of Medicine II, Saarland University Medical Center, Saarland University, Homburg, Germany.,Laboratory of Metabolic Liver Diseases, Department of General, Transplant and Liver Surgery, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
5
|
Sakai N, Kamimura K, Miyamoto H, Ko M, Nagoya T, Setsu T, Sakamaki A, Yokoo T, Kamimura H, Soki H, Tokunaga A, Inamine T, Nakashima M, Enomoto H, Kousaka K, Tachiki H, Ohyama K, Terai S. Letrozole ameliorates liver fibrosis through the inhibition of the CTGF pathway and 17β-hydroxysteroid dehydrogenase 13 expression. J Gastroenterol 2023; 58:53-68. [PMID: 36301364 DOI: 10.1007/s00535-022-01929-w] [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: 10/07/2021] [Accepted: 09/21/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND To establish a treatment option for liver fibrosis, the possibility of the drug repurposing theory was investigated, with a focus on the off-target effects of active pharmaceutical ingredients. METHODS First, several active pharmaceutical ingredients were screened for their effects on the gene expression in the hepatocytes using chimeric mice with humanized hepatocytes. As per the gene expression-based screening assay for 36 medications, we assessed the mechanism of the antifibrotic effect of letrozole, a third-generation aromatase inhibitor, in mouse models of liver fibrosis induced by carbon tetrachloride (CCl4) and a methionine choline-deficient (MCD) diet. We assessed liver histology, serum biochemical markers, and fibrosis-related gene and protein expressions in the hepatocytes. RESULTS A gene expression-based screening assay revealed that letrozole had a modifying effect on fibrosis-related gene expression in the hepatocytes, including YAP, CTGF, TGF-β, and CYP26A1. Letrozole was administered to mouse models of CCl4- and MCD-induced liver fibrosis and it ameliorated the liver fibrosis. The mechanisms involved the inhibition of the Yap-Ctgf profibrotic pathway following a decrease in retinoic acid levels in the hepatocytes caused by suppression of the hepatic retinol dehydrogenase, Hsd17b13 and activation of the retinoic acid hydrogenase, Cyp26a1. CONCLUSIONS Letrozole slowed the progression of liver fibrosis by inhibiting the Yap-Ctgf pathway. The mechanisms involved the modification of the Hsd17b13 and Cyp26a1 expressions led to the suppression of retinoic acid in the hepatocytes, which contributed to the activation of Yap-Ctgf pathway. Because of its off-target effect, letrozole could be repurposed for the treatment of liver fibrosis. The third-generation aromatase inhibitor letrozole ameliorated liver fibrosis by suppressing the Yap-Ctgf pathway by partially modifying the Hsd17b13 and Cyp26a1 expressions, which reduced the retinoic acid level in the hepatocytes. The gene expression analysis using chimeric mice with humanized liver revealed that the mechanisms are letrozole specific and, therefore, may be repurposed for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Norihiro Sakai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | - Kenya Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan.
- Department of General Medicine, Niigata University School of Medicine, Niigata, Niigata, 951-8510, Japan.
| | - Hirotaka Miyamoto
- Department of Pharmaceutics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, 852-8588, Japan
| | - Masayoshi Ko
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | - Takuro Nagoya
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | - Toru Setsu
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | - Akira Sakamaki
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | - Takeshi Yokoo
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | - Hiroteru Kamimura
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| | - Hiroyuki Soki
- Unit of Medical Pharmacy, Department of Pharmacy Practice, Nagasaki University, Nagasaki, Nagasaki, 852-8588, Japan
| | - Ayako Tokunaga
- Department of Pharmaceutics, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki, Nagasaki, 852-8588, Japan
| | - Tatsuo Inamine
- Department of Pharmacotherapeutics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Nagasaki, 852-8588, Japan
- Organization for Research Promotion, University of the Ryukyus, Nishihara-Cho, Okinawa, 903-0213, Japan
| | - Mikiro Nakashima
- Unit of Medical Pharmacy, Department of Pharmacy Practice, Nagasaki University, Nagasaki, Nagasaki, 852-8588, Japan
| | - Hatsune Enomoto
- Scientific Research and Business Development Department, Towa Pharmaceutical Co., Ltd., Kadoma, Osaka, 571-8580, Japan
| | - Kazuki Kousaka
- Scientific Research and Business Development Department, Towa Pharmaceutical Co., Ltd., Kadoma, Osaka, 571-8580, Japan
| | - Hidehisa Tachiki
- Scientific Research and Business Development Department, Towa Pharmaceutical Co., Ltd., Kadoma, Osaka, 571-8580, Japan
| | - Kaname Ohyama
- Unit of Medical Pharmacy, Department of Pharmacy Practice, Nagasaki University, Nagasaki, Nagasaki, 852-8588, Japan
- Department of Hospital Pharmacy, Nagasaki University Hospital, Nagasaki, Nagasaki, 852-8501, Japan
| | - Shuji Terai
- Division of Gastroenterology and Hepatology, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Niigata, 951-8510, Japan
| |
Collapse
|
6
|
Riccio S, Melone R, Vitulano C, Guida P, Maddaluno I, Guarino S, Marzuillo P, Miraglia del Giudice E, Di Sessa A. Advances in pediatric non-alcoholic fatty liver disease: From genetics to lipidomics. World J Clin Pediatr 2022; 11:221-238. [PMID: 35663007 PMCID: PMC9134151 DOI: 10.5409/wjcp.v11.i3.221] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/05/2021] [Accepted: 04/02/2022] [Indexed: 02/06/2023] Open
Abstract
As a result of the obesity epidemic, non-alcoholic fatty liver disease (NAFLD) represents a global medical concern in childhood with a closely related increased cardiometabolic risk. Knowledge on NAFLD pathophysiology has been largely expanded over the last decades. Besides the well-known key NAFLD genes (including the I148M variant of the PNPLA3 gene, the E167K allele of the TM6SF2, the GCKR gene, the MBOAT7-TMC4 rs641738 variant, and the rs72613567:TA variant in the HSD17B13 gene), an intriguing pathogenic role has also been demonstrated for the gut microbiota. More interestingly, evidence has added new factors involved in the "multiple hits" theory. In particular, omics determinants have been highlighted as potential innovative markers for NAFLD diagnosis and treatment. In fact, different branches of omics including metabolomics, lipidomics (in particular sphingolipids and ceramides), transcriptomics (including micro RNAs), epigenomics (such as DNA methylation), proteomics, and glycomics represent the most attractive pathogenic elements in NAFLD development, by providing insightful perspectives in this field. In this perspective, we aimed to provide a comprehensive overview of NAFLD pathophysiology in children, from the oldest pathogenic elements (including genetics) to the newest intriguing perspectives (such as omics branches).
Collapse
Affiliation(s)
- Simona Riccio
- Department of Woman, Child, General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Rosa Melone
- Department of Woman, Child, General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Caterina Vitulano
- Department of Woman, Child, General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Pierfrancesco Guida
- Department of Woman, Child, General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Ivan Maddaluno
- Department of Woman, Child, General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Stefano Guarino
- Department of Woman, Child, General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Pierluigi Marzuillo
- Department of Woman, Child, General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Emanuele Miraglia del Giudice
- Department of Woman, Child, General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Anna Di Sessa
- Department of Woman, Child, General and Specialized Surgery, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| |
Collapse
|
7
|
Tavaglione F, Targher G, Valenti L, Romeo S. Human and molecular genetics shed lights on fatty liver disease and diabetes conundrum. Endocrinol Diabetes Metab 2020; 3:e00179. [PMID: 33102799 PMCID: PMC7576307 DOI: 10.1002/edm2.179] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/28/2020] [Accepted: 08/01/2020] [Indexed: 12/13/2022] Open
Abstract
The causal role of abdominal overweight/obesity, insulin resistance and type 2 diabetes (T2D) on the risk of fatty liver disease (FLD) has robustly been proven. A consensus of experts has recently proposed the novel definition of 'metabolic dysfunction-associated fatty liver disease, MAFLD' instead of 'nonalcoholic fatty liver disease, NAFLD', emphasizing the central role of dysmetabolism in the disease pathogenesis. Conversely, a direct and independent contribution of FLD per se on risk of developing T2D is still a controversial topic. When dealing with FLD as a potential risk factor for T2D, it is straightforward to think of hepatic insulin resistance as the most relevant underlying mechanism. Emerging evidence supports genetic determinants of FLD (eg PNPLA3, TM6SF2, MBOAT7, GCKR, HSD17B13) as determinants of insulin resistance and T2D. However, recent studies highlighted that the key molecular mechanism of dysmetabolism is not fat accumulation per se but the degree of hepatic fibrosis (excess liver fat content-lipotoxicity), leading to reduced insulin clearance, insulin resistance and T2D. A consequence of these findings is that drugs that will ameliorate liver fat accumulation and fibrosis in principle may also exert a beneficial effect on insulin resistance and risk of T2D in individuals with FLD. Finally, initial findings show that these genetic factors might be directly implicated in modulating pancreatic beta-cell function, although future studies are needed to fully understand this relationship.
Collapse
Affiliation(s)
- Federica Tavaglione
- Clinical Medicine and Hepatology UnitDepartment of Internal Medicine and GeriatricsCampus Bio‐Medico UniversityRomeItaly
- Department of Molecular and Clinical MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Giovanni Targher
- Section of Endocrinology, Diabetes and MetabolismDepartment of MedicineUniversity and Azienda Ospedaliera Universitaria Integrata of VeronaVeronaItaly
| | - Luca Valenti
- Department of Pathophysiology and TransplantationUniversità degli Studi di MilanoMilanoItaly
- Translational MedicineDepartment of Transfusion Medicine and HematologyFondazione IRCCS Ca’ Granda Ospedale Maggiore PoliclinicoMilanoItaly
| | - Stefano Romeo
- Department of Molecular and Clinical MedicineSahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Clinical Nutrition UnitDepartment of Medical and Surgical ScienceMagna Graecia UniversityCatanzaroItaly
- Department of CardiologySahlgrenska University HospitalGothenburgSweden
| |
Collapse
|
8
|
Díaz-González Á, Sapena V, Boix L, Brunet M, Torres F, LLarch N, Samper E, Millán O, Corominas J, Iserte G, Sanduzzi-Zamparelli M, da Fonseca LG, Darnell A, Belmonte E, Forner A, Ayuso C, Bruix J, Reig M. Pharmacokinetics and pharmacogenetics of sorafenib in patients with hepatocellular carcinoma: Implications for combination trials. Liver Int 2020; 40:2476-2488. [PMID: 33021346 DOI: 10.1111/liv.14587] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 06/07/2020] [Accepted: 06/09/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND & AIMS Sorafenib and lenvatinib are the first-line treatments approved in hepatocellular carcinoma (HCC), but information is lacking about the relationships between their pharmacokinetics, patients pharmacogenetic profiles, adverse events (AE) and overall survival. We aimed to elucidate these relationships of tyrosine Kinase Inhibitors, such as sorafenib, in order to improve the design of trials testing it in combination with checkpoint inhibitors. METHODS We assessed the pharmacokinetics of sorafenib and its N-oxide metabolite at day-0, day-7, day-30, day-60, day-90, day-120, day-150 and day-180 and nine single-nucleotide polymorphisms (SNP) in five genes related to sorafenib metabolism/transport to identify the best point for starting the combination between tyrosine kinases and checkpoint inhibitors. RESULTS We prospectively included 49 patients (96% cirrhotic, 37% hepatitis-C, 82% Child-Pugh-A and 59% BCLC-C). Pharmacokinetic values peaked at day-7 and progressively declined until day-60. In the 16 patients without further dose modifications after day-60, pharmacokinetic values remained stable through day-180 (sorafenib P = .90; N-oxide P = .93). Pharmacokinetic values were higher in patients with early dermatological adverse events and lower in patients with early diarrhoea. Sorafenib and N-oxide pharmacokinetic values varied linearly with different alleles of MRP2*3972. CONCLUSIONS Sorafenib's pharmacokinetics is heterogeneous across HCC patients. This heterogeneity affects adverse events development and must be taken into account in setting the dose and timing of its combination with checkpoint inhibitors.
Collapse
Affiliation(s)
- Álvaro Díaz-González
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Víctor Sapena
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Loreto Boix
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Mercè Brunet
- Pharmacology and Toxicology Laboratory, Biochemistry and Molecular Genetics Department, Biomedical Diagnostic Center, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Ferrán Torres
- Medical Statistics Core Facility, IDIBAPS. Hospital Clínic de Barcelona. Biostatistics Unit, Faculty of Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Neus LLarch
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Esther Samper
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Olga Millán
- Pharmacology and Toxicology Laboratory, Biochemistry and Molecular Genetics Department, Biomedical Diagnostic Center, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Josep Corominas
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Gemma Iserte
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Marco Sanduzzi-Zamparelli
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Leonardo G da Fonseca
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Anna Darnell
- BCLC group. Radiology department, Hospital Clínic de Barcelona. IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Ernest Belmonte
- BCLC group. Radiology department, Hospital Clínic de Barcelona. IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Alejandro Forner
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - Carmen Ayuso
- BCLC group. Radiology department, Hospital Clínic de Barcelona. IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Jordi Bruix
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| | - María Reig
- BCLC group. Liver Unit, Hospital Clínic de Barcelona. IDIBAPS. CIBERehd, University of Barcelona, Barcelona, Spain
| |
Collapse
|
9
|
Raja AM, Ciociola E, Ahmad IN, Dar FS, Naqvi SMS, Moaeen-ud-Din M, Raja GK, Romeo S, Mancina RM. Genetic Susceptibility to Chronic Liver Disease in Individuals from Pakistan. Int J Mol Sci 2020; 21:ijms21103558. [PMID: 32443539 PMCID: PMC7278956 DOI: 10.3390/ijms21103558] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 05/14/2020] [Accepted: 05/15/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic liver disease, with viral or non-viral etiology, is endemic in many countries and is a growing burden in Asia. Among the Asian countries, Pakistan has the highest prevalence of chronic liver disease. Despite this, the genetic susceptibility to chronic liver disease in this country has not been investigated. We performed a comprehensive analysis of the most robustly associated common genetic variants influencing chronic liver disease in a cohort of individuals from Pakistan. A total of 587 subjects with chronic liver disease and 68 healthy control individuals were genotyped for the HSD17B13 rs7261356, MBOAT7 rs641738, GCKR rs1260326, PNPLA3 rs738409, TM6SF2 rs58542926 and PPP1R3B rs4841132 variants. The variants distribution between case and control group and their association with chronic liver disease were tested by chi-square and binary logistic analysis, respectively. We report for the first time that HSD17B13 variant results in a 50% reduced risk for chronic liver disease; while MBOAT7; GCKR and PNPLA3 variants increase this risk by more than 35% in Pakistani individuals. Our genetic analysis extends the protective role of the HSD17B13 variant against chronic liver disease and disease risk conferred by the MBOAT7; GCKR and PNPLA3 variants in the Pakistani population.
Collapse
Affiliation(s)
- Asad Mehmood Raja
- University Institute of Biochemistry and Biotechnology, Pir Mehr Ali Shah Arid Agriculture University Rawalpindi, Rawalpindi 46300, Pakistan; (A.M.R.); (S.M.S.N.); (G.K.R.)
| | - Ester Ciociola
- Department of Molecular and Clinical Medicine, The Sahlgrenska Academy at the University of Gothenburg, Wallenberg Laboratory, 413 45 Gothenburg, Sweden;
| | - Imran Nazir Ahmad
- Department of Pathology and Laboratory Medicine, Shifa International Hospitals Ltd., Islamabad 44790, Pakistan;
| | - Faisal Saud Dar
- Liver Transplantation, Hepatobiliary and Pancreatic Services Unit, Shifa International Hospitals Ltd., Islamabad 44790, Pakistan;
| | - Syed Muhammad Saqlan Naqvi
- University Institute of Biochemistry and Biotechnology, Pir Mehr Ali Shah Arid Agriculture University Rawalpindi, Rawalpindi 46300, Pakistan; (A.M.R.); (S.M.S.N.); (G.K.R.)
| | - Muhammad Moaeen-ud-Din
- Department of Animal Breeding and Genetics/National Center for Livestock Breeding, Genetics & Genomics, Pir Mehr Ali Shah Arid Agriculture University Rawalpindi, Rawalpindi 46300, Pakistan;
| | - Ghazala Kaukab Raja
- University Institute of Biochemistry and Biotechnology, Pir Mehr Ali Shah Arid Agriculture University Rawalpindi, Rawalpindi 46300, Pakistan; (A.M.R.); (S.M.S.N.); (G.K.R.)
| | - Stefano Romeo
- Department of Molecular and Clinical Medicine, The Sahlgrenska Academy at the University of Gothenburg, Wallenberg Laboratory, 413 45 Gothenburg, Sweden;
- Department of Cardiology, Sahlgrenska University Hospital, 413 45 Gothenburg, Sweden
- Clinical Nutrition Unit, Department of Medical and Surgical Sciences, University Magna Graecia, 88100 Catanzaro, Italy
- Correspondence: (S.R.); (R.M.M.); Tel.: +46-(0)313-426-735 (S.R.); +46-(0)31342186 (R.M.M.)
| | - Rosellina Margherita Mancina
- Department of Molecular and Clinical Medicine, The Sahlgrenska Academy at the University of Gothenburg, Wallenberg Laboratory, 413 45 Gothenburg, Sweden;
- Correspondence: (S.R.); (R.M.M.); Tel.: +46-(0)313-426-735 (S.R.); +46-(0)31342186 (R.M.M.)
| |
Collapse
|