Next Article in Journal
Diffusion–Based Virtual MR Elastography of the Liver: Can It Be Extended beyond Liver Fibrosis?
Previous Article in Journal
Patient–Ventilator Dyssynchrony in Critically Ill Patients
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Real-World Management of Patients with Primary Biliary Cholangitis—A Retrospective Study from a Tertiary Medical Center in Israel

1
Center for Autoimmune Liver Diseases, Department of Gastroenterology, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
2
Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(19), 4551; https://doi.org/10.3390/jcm10194551
Submission received: 31 August 2021 / Revised: 22 September 2021 / Accepted: 27 September 2021 / Published: 30 September 2021
(This article belongs to the Section Gastroenterology & Hepatopancreatobiliary Medicine)

Abstract

:
Background: Primary biliary cholangitis (PBC) is a rare autoimmune liver disease with variation in prevalence, phenotype and prognosis across different geographical regions. Little is known about PBC in Israel. Our aim was to characterize the demography, clinical presentation, treatment patterns and prognosis in a cohort of PBC patients followed in a referral center in central Israel. Methods: Clinical, demographic and laboratory data were collected from the medical records of PBC patients followed at Tel Aviv Medical Center in the years 2003–2020. Results: We have identified 189 patients with a confirmed diagnosis of PBC; 92.6% were female and the mean age at diagnosis was 54.7 years. Thirty-nine percent were diagnosed with another autoimmune disease and 5.9% were diagnosed with a PBC-AIH (autoimmune hepatitis) variant syndrome. Ninety-six percent were treated with ursodeoxycholic acid (UDCA) at a mean dose of 13.3 mg/kg. A total of 28.1% were found with inadequate response to UDCA according to the Toronto criteria, and 53% of the UDCA non-responders were treated with bezafibrate. Younger age at diagnosis, higher baseline levels of alkaline phosphatase (ALP) and gamma-glutamyltransferase (GGT), AIH-PBC variant and positive anti-smooth muscle antibodies (ASMA) were associated with an inadequate UDCA response. In a multivariable analysis, higher ALP at diagnosis (OR = 1.92 CI 1.11–3.20 per 50-unit change, p = 0.018) and ASMA (OR = 27.6 CI 2.58–295, p = 0.006) independently predicted inadequate UDCA response. Higher alanine transaminase (ALT), ALP and GGT, lower albumin, younger age at diagnosis and pruritus conferred an increased risk for disease progression. Conclusions: Disease characteristics, treatment patterns, response to therapy and prognosis of a PBC patient cohort in a tertiary center in central Israel were revealed. The results highlight the importance of risk stratification in PBC, specifically in younger patients, those presenting with a high level of liver enzymes and in ASMA-positive patients with an assumed diagnosis of the AIH-PBC variant.

1. Introduction

Primary biliary cholangitis (PBC) is a chronic autoimmune liver disease of the small intrahepatic bile ducts. The disease typically affects middle aged women, with a female to male ratio of up to 10:1 [1], although a higher prevalence in males and younger age at diagnosis has been recently reported [2,3]. PBC is characterized by a cholestatic pattern of serum liver enzymes, the presence of anti-mitochondrial antibodies (AMA) and/or the PBC specific anti-nuclear antibodies (ANA) (anti-sp100 and anti-gp210), and typical histological features [4]. Without treatment, PBC may progress to cirrhosis and hepatic failure, and eventually, may require liver transplantation.
Ursodeoxycholic acid (UDCA) therapy was shown to improve liver-transplant-free survival and is considered the cornerstone of treatment [5,6]. Until recently, UDCA was the only therapeutic option for PBC patients; however, several new treatment options have recently been added to the treatment armamentarium as adjuncts to UDCA, in patients with inadequate response. The availability of these second line drugs, obeticholic acid and fibrates, varies greatly between different geographical regions.
Several biochemical UDCA response criteria have been developed in order to predict prognosis, either binary (Paris I, Paris II, Rotterdam, Toronto or Barcelona) or continuous (GLOBE score [7] and the UK-PBC risk score [8]). The biochemical response rates to UDCA vary widely according to the exact criteria and the population [9,10]. Notably, liver-transplantation-free survival is markedly decreased in patients with poor UDCA response [8] and current guidelines emphasize the importance of timely assessment of patients at high risk without adequate response to first line treatment [11].
PBC is considered a rare liver disease; however, its incidence rate is increasing worldwide [12]. In 2018, its prevalence in the US was reported to be 29.3 per 100,000. In a recent meta-analysis, the prevalence and annual incidence in Europe from 2000 through to 2020 were 22.3 and 1.87 per 100,000, respectively [13]. In Israel, the epidemiological data are limited. In 2012, Delgado et al. reported a prevalence of 25.5 cases per 100,000 in southern Israel [14]. Population-based studies showed a wide range of incidence and prevalence of PBC throughout the world [15]. Recently, a latitudinal geoepidemiological pattern of occurrence has been proposed, with a higher prevalence in northern countries [16]. Interestingly, the rates of non-response to UDCA also varied in different studies depending on the geographical region and the study population [17]. For example, the inadequate response rate according to the Toronto criteria were found to be as high as 38.6% in a single center in England and 33% in a population study in Iceland [18,19], while in a single center in China and in a multicenter study in Italy, lower non-responder rates of 17% and 20.1%, were reported, respectively [17,20].
The aim of the present study was to characterize the PBC population in a large tertiary referral medical center in Tel Aviv, Israel. We aimed to characterize disease phenotype including demographic data, disease-related symptoms and severity, laboratory parameters, presence of concomitant medical disorders, treatment patterns, treatment response rates and prognosis. Additionally, we evaluated associations of biochemical and demographical variables and UDCA response, and investigated prognostic factors for clinical endpoint of advanced liver disease and its complications.

2. Materials and Methods

This retrospective, single-center study included patients who were diagnosed with PBC according to ICD-9 coding (The International Classification of Diseases, ICD-9 code 571.6) and began follow-up in the outpatient liver clinic at Tel Aviv Sourasky Medical Center, during the period of January 2003–May 2020. Clinical, demographic and laboratory data were collected from electronic medical records. PBC diagnosis was defined according to European Association for the Study of the Liver (EASL) guidelines [11]. Two senior hepatologists with specific expertise in autoimmune and cholestatic liver diseases evaluated the data independently and decided which patients were correctly diagnosed with PBC and the PBC-AIH (autoimmune hepatitis) overlap variant, and who to exclude (non-agreement was discussed and agreed by consensus). Patients not meeting the diagnostic criteria or with an alternative diagnosis were excluded. Clinical endpoint was defined as any of the following clinical events: diagnosis of liver cirrhosis, liver transplantation, hepatocellular carcinoma, liver-related death, variceal bleeding, spontaneous bacterial peritonitis, hepatic encephalopathy, ascites, and hepatorenal syndrome. The degree of fatigue and pruritus was assessed by the treating physician during the outpatient clinic visit, and classified as severe or mild to moderate. Histological reports of liver biopsies were reviewed and assessed for the presence of interface hepatitis and the fibrosis stage. Advanced fibrosis was defined as Ludwig’s stages 3 or 4 (bridging fibrosis or cirrhosis).
The study was approved by the Sourasky Medical Center’s institutional ethics committee in keeping with the principles of the Declaration of Helsinki, approval number 0163-20. An informed consent waiver was granted by the IRB committee as the data were recorded anonymously.
Statistical Analysis: Data are presented as means and standard deviations for continuous variables. For analysis of UDCA response, we estimated the odds ratios (OR) by conducting a univariable logistic regression and, in addition, we conducted a stepwise logistic regression for multivariable analysis with entry probability of 0.1 and removal probability of 0.15. For analysis of clinical endpoint, we estimated the hazard ratios (HR) with the use of Cox regression models. In addition, comparing the UDCA responders and the UDCA non-responders groups, we applied the log-rank test and the Cox model and presented it in a Kaplan–Meier curve. p-value < 0.05 was considered statistically significant. We used MATLAB software, version R2020b (MathWorks, Natick, MA, USA), and IBM SPSS version 27 (IBM, Armonk, NY, USA) for all the statistical analyses.

3. Results

3.1. Patients and Disease Characteristics

A total of 215 patients with a diagnosis of PBC according to ICD-9 coding were found in a digital database search for patients who visited the outpatient liver clinic of the gastroenterology department in Tel Aviv Medical Center between January 2003 and May 2020. Revision of patients’ medical records found 26 patients without a probable diagnosis of PBC according to EASL guidelines [11] or with an alternative diagnosis; thus, a total of 189 patients with PBC were included for further analysis (Figure 1).
Among patients, 175 (92.6% of the cohort) were females and the average age at diagnosis was 54.7 years (±13.0). In total, 86.5% of the patients (147/170) were AMA-positive, 75.5% had at least one positive test for anti-nuclear antibodies (ANA) and 13.5% for anti-smooth muscle antibodies (ASMA). In total, 72.3% of the patients (125/173) were Ashkenazi Jews, and 24.9% (43/173) were Sephardi Jews; 39.2% of the patients (73/186) were diagnosed with at least one other autoimmune disease, most commonly hypothyroidism (19.9% of patients). The AIH-PBC overlap variant syndrome was diagnosed in 11 patients (5.9%). Twenty-eight patients (15.1%) had a history of cholecystitis or cholecystectomy (Table 1).
Most of the patients (96.3%, 182/189) were treated with UDCA with a mean dose of 13.3 (±4.47) mg UDCA/kg BW/d. Regarding the use of additional medications for PBC, 18.5% of the patients (35/189) were treated with bezafibrate, 13.8% (26/189) were treated with corticosteroids (prednisone or budesonide), and 2.65% (5/189) with obeticholic acid. Assessment of disease progression in the entire cohort revealed that 17.5% (33/189) of the patients reached an adverse clinical endpoint. Of those, 15.9% (30/189) developed cirrhosis, 5.3% (10/189) ascites, 3.7% (7/189) hepatic encephalopathy, 3.2% (6/189) variceal bleeding, 1.6% (3/189) hepato-renal syndrome, 0.5% (1/189) spontaneous bacterial peritonitis, and 0.5% (1/189) liver-related death. 7 patients, comprising 3.7% of the cohort, underwent liver transplantation.

3.2. Response to UDCA Treatment

Patients with a follow-up shorter than 12 months were excluded (Figure 1). Biochemical response to UDCA was evaluated according to accepted criteria (Table 2).
UDCA non-response rates were found in the range of 16.7–40.6% of the patients according to different dichotomous criteria (Table 2). As the Toronto criteria are based solely on ALP level, we had more patients with available information for this criterion and, thus, we chose this criterion for further statistical evaluations. Within the 128 patients included in the analysis, the median follow-up time since PBC diagnosis was 7 years with an interquartile range of 10 years. Univariable analysis for association of baseline disease parameters with UDCA non-response (Toronto criteria) revealed that patients with younger age and higher baseline ALP and GGT at diagnosis were less likely to respond to UDCA therapy with an OR = 0.95 CI 0.92–0.99 per year, p = 0.009, OR = 1.38 CI 1.15–1.66 for a 50-unit change, p < 0.001 and OR = 1.02 CI 1–1.04 for a 10-unit change, p = 0.03, respectively (Table 3).
The PBC-AIH variant syndrome was diagnosed in 11 patients (5.9% of the cohort) and ASMA was found positive in 17 patients (13.5% of the cohort; Table 1); both were associated with reduced response rates to UDCA therapy with an OR of 4.78 CI 1.08–21.2, p = 0.039, and OR = 8 CI 1.92–33.26, p = 0.004, respectively, (Table 3). Liver histology results were available for 52 patients. The presence of interface hepatitis was associated with reduced response rates to UDCA therapy with an OR of 12 CI 1.43–100.8, p = 0.022, while advanced fibrosis was not statistically significantly associated with response rates to UDCA (Table 3). No significant association with UDCA response rates was found for gender, ethnicity, symptoms of fatigue or pruritus, presence of other autoimmune diseases and serology of AMA or ANA. In a multivariable analysis, higher ALP at diagnosis (OR = 1.92 CI 1.11–3.20 per 50-unit change, p = 0.018) and ASMA (OR = 27.6 CI 2.58–295, p = 0.006) independently predicted inadequate UDCA response.

3.3. Risk Factors Associated with Disease Progression

One hundred and twenty-eight patients had a follow up period of at least 12 months and information regarding response to UDCA therapy (Figure 1); of these, 23 reached a clinical endpoint. Overall, 21 patients developed liver cirrhosis, 9 of them with decompensation. Of note, three patients were already diagnosed with liver cirrhosis at PBC diagnosis. As expected, UDCA non-responders were more likely to reach a clinical endpoint with 31.4% versus 10%, in UDCA non-responder versus responders, respectively (p = 0.033 log rank test) (Figure 2).
Univariable Cox regression analysis revealed an increased risk for reaching a clinical endpoint in patients with baseline higher ALT (HR 1.17 CI 1.02–1.35 per 10-unit change, p = 0.026), ALP (HR 1.28 CI 1.04–1.58 per 50-unit change, p = 0.022) and GGT (HR 1.03 CI 1.00–1.06 per 10-unit change, p = 0.021), as well as in patients with lower serum albumin (HR 0.93 CI 0.86–1.00, p = 0.044) and in patients with younger age at diagnosis (HR 0.96 CI 0.93–1.00, p = 0.046) (Table 4).
Interestingly, patients who suffered from pruritus had an increased risk of reaching a clinical endpoint (HR 3.45 CI 1.29–9.22, p = 0.014). Gender, fatigue, and the presence of other liver diseases or other autoimmune diseases were not significantly associated with early clinical endpoint. Due to lack of statistical power, we could not calculate odds ratios for histological parameters. Multivariable analysis did not identify statistically significant parameters associated with adverse clinical outcome.

4. Discussion

PBC is an autoimmune liver disease with a reported geographical variability in prevalence, disease phenotype and response to treatment. Moreover, second line treatments beyond UDCA have emerged in recent years but still greatly vary between countries depending on local health regulations, drug availability and insurance coverage. Little is known about PBC in Israel and here, we present data on two decades of follow up of PBC patients in a tertiary liver center in central Israel.
Patients’ characteristics and baseline lab values in our study (Table 1) represent a typical PBC cohort with predominant female gender, cholestatic pattern of liver enzymes and a normal mean value of total bilirubin, albumin and platelets. Interestingly, we have found that about three-quarters of the cohort had positive ANA, while a recent study that included 434 PBC patients showed a lower ANA prevalence of about 55% [21]. The higher prevalence of ANA in our study may characterize the specific Jewish Israeli population in our study. Alternatively, this difference could be related to the fact that many of the patients in our cohort had blood tests performed in several different labs in primary care settings and in hospitals that use different techniques for ANA detection.
Biochemical non-response to UDCA is a well-known predictor of long-term histological progression [10]. UDCA response rates in our cohort were found to be generally similar to previous reports, yet higher in comparison to recent similar studies [21,22]. According to the Barcelona criterion, we have documented a non-response rate of 40.6%, while multicenter data from Portugal and Germany reported rates of 30.7% and 38.7%, respectively [21,22]. In addition, when assessing response rates according to the Toronto criterion, we found a non-response rate of 28.1%, while a recently published German study found a non-response rate of only 16.2% according to this criterion [22]. These higher percentages of UDCA non-response in our study could be attributed to the accumulation of difficult to treat patients in a referral tertiary center [17].
Eleven patients, representing 5.9% of the cohort, were diagnosed with the AIH-PBC variant syndrome. Interestingly, we found that the presence of PBC-AIH variant syndrome as well as the presence of ASMA conferred an increased risk of UDCA non-response with HRs of 4.8 and 8, respectively. Moreover, positive ASMA predicted lower response rates to UDCA independently in a multivariable analysis. These findings are in agreement with a recent study demonstrating reduced response rates to UDCA in PBC patients with autoimmune hepatitis features [23]. Of note, a previous report showed that the PBC-AIH variant was associated with higher rates of cirrhosis complications when compared to PBC or AIH alone [24]. Our findings suggest that these patients are at increased risk for complications and may benefit from additive therapeutic strategies.
We found that young age at diagnosis was a major risk factor for UDCA non-response. This association, which probably represents a more aggressive disease course in the younger population, has been described recently in large PBC cohorts [9,25,26]. In addition, in partial agreement with previous reports, we found ALP and GGT levels at baseline to predict inadequate UDCA response [21,26,27,28]. Liver biopsy is not mandatory for the diagnosis or follow up in PBC [11]; nevertheless, histological data were available in this study for 52 out of the 125 patients included in the analysis. Interface hepatitis, but not advanced fibrosis, was found to be associated with UDCA non-response. In fact, the absence of interface hepatitis was found to be a strong predictor of biochemical response to UDCA as only 1 out of 16 patients without interface hepatitis failed to respond to UDCA treatment, thus corroborating a previous study that addressed this issue [29].
Regarding risk factors for disease prognosis, we found younger age at diagnosis, high levels of ALT, GGT and ALP and low level of albumin at baseline to be associated with disease progression; all were shown in previous studies to predict a worse prognosis in PBC patients [7,8,30]. In addition, we found pruritus to be associated with reaching of a clinical endpoint. Notably, a few patients in our cohort underwent liver transplantation due to severe pruritus, without end-stage liver disease.
This study has several limitations. First, it is retrospective and prone to limitations associated with this design. Second, we reviewed the medical records of patients within the PBC clinic at a single referral hospital in Tel Aviv; therefore, our cohort may not represent PBC patients in the general Israeli population. Third, the number of patients with documented adverse clinical events was relatively small, thus hampering the statistical power of the study, especially the multivariable analysis.
In conclusion, we describe a large cohort of PBC patients in a referral liver center in central Israel. An extensive analysis of disease characteristics, management patterns, response to therapy and prognosis has been explored. The data revealed baseline parameters associated with inadequate response to therapy and prognosis, further strengthening the importance of risk stratification in the management of patients with PBC.

Author Contributions

Conceptualization, E.Z. and E.Y.; methodology, E.Y. and E.Z.; Statistical analysis, M.L.; data curation, E.Y and I.I.; writing—original draft preparation, E.Y. and E.Z.; writing—review and editing, O.S., I.H. and E.Z.; supervision, E.Z.; project administration, E.Z. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board of Sourasky Medical Center, approval number 0163-20, date of approval 27 August 2020.

Informed Consent Statement

Informed consent waiver was granted by the IRB committee as the data were recorded anonymously.

Data Availability Statement

The data presented in this study are available on request.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

PBCPrimary Biliary Cholangitis
AIHAutoimmune hepatitis
ALPAlkaline phosphates
GGTGamma glutamyl transferase
ALTAlanine transaminase
ASTAspartate transaminase
AMAAnti-mitochondrial antibodies
ANAAnti-nuclear antibodies
ASMAAnti-smooth muscle antibodies
UDCAUrsodeoxycholic acid
ULNUpper limit of normal
OROdds ratio
HRHazard ratio
EASLEuropean Association for the Study of the Liver

References

  1. Kaplan, M.M.; Gershwin, M.E. Primary biliary cirrhosis. N. Engl. J. Med. 2005, 353, 1261–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Fan, X.; Wang, T.; Shen, Y.; Xi, X.; Yang, L. Underestimated Male Prevalence of Primary Biliary Cholangitis in China: Results of a 16-year cohort study involving 769 patients. Sci. Rep. 2017, 7, 6560. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Lleo, A.; Jepsen, P.; Morenghi, E.; Carbone, M.; Moroni, L.; Battezzati, P.M.; Podda, M.; Mackay, I.R.; Gershwin, M.E.; Invernizzi, P. Evolving Trends in Female to Male Incidence and Male Mortality of Primary Biliary Cholangitis. Sci. Rep. 2016, 6, 25906. [Google Scholar] [CrossRef] [PubMed]
  4. Sebode, M.; Weiler-Normann, C.; Liwinski, T.; Schramm, C. Autoantibodies in Autoimmune Liver Disease—Clinical and Diagnostic Relevance. Front. Immunol. 2018, 9, 609. [Google Scholar] [CrossRef] [Green Version]
  5. Harms, M.H.; van Buuren, H.R.; Corpechot, C.; Thorburn, D.; Janssen, H.L.; Lindor, K.D.; Hirschfield, G.M.; Parés, A.; Floreani, A.; Mayo, M.J.; et al. Ursodeoxycholic acid therapy and liver transplant-free survival in patients with primary biliary cholangitis. J. Hepatol. 2019, 71, 357–365. [Google Scholar] [CrossRef]
  6. Poupon, R.E.; Lindor, K.D.; Cauch-Dudek, K.; Dickson, E.R.; Heathcote, E.J. Combined analysis of randomized controlled trials of ursodeoxycholic acid in primary biliary cirrhosis. Gastroenterology 1997, 113, 884–890. [Google Scholar] [CrossRef]
  7. Lammers, W.; Hirschfield, G.; Corpechot, C.; Nevens, F.; Lindor, K.D.; Janssen, H.L.; Floreani, A.; Ponsioen, C.Y.; Mayo, M.J.; Invernizzi, P.; et al. Development and Validation of a Scoring System to Predict Outcomes of Patients With Primary Biliary Cirrhosis Receiving Ursodeoxycholic Acid Therapy. Gastroenterology 2015, 149, 1804–1812. [Google Scholar] [CrossRef]
  8. Carbone, M.; Sharp, S.J.; Flack, S.; Paximadas, D.; Spiess, K.; Adgey, C.; Griffiths, L.; Lim, R.; Trembling, P.; Williamson, K.; et al. The UK-PBC risk scores: Derivation and validation of a scoring system for long-term prediction of end-stage liver disease in primary biliary cholangitis. Hepatology 2016, 63, 930–950. [Google Scholar] [CrossRef]
  9. Carbone, M.; Mells, G.F.; Pells, G.; Dawwas, M.F.; Newton, J.L.; Heneghan, M.A.; Neuberger, J.M.; Day, D.B.; Ducker, S.J.; Sandford, R.N.; et al. Sex and Age Are Determinants of the Clinical Phenotype of Primary Biliary Cirrhosis and Response to Ursodeoxycholic Acid. Gastroenterology 2013, 144, 560–569. [Google Scholar] [CrossRef]
  10. Kumagi, T.; Guindi, M.; Fischer, S.E.; Arenovich, T.; Abdalian, R.; Coltescu, C.; E Heathcote, J.; Hirschfield, G. Baseline Ductopenia and Treatment Response Predict Long-Term Histological Progression in Primary Biliary Cirrhosis. Am. J. Gastroenterol. 2010, 105, 2186–2194. [Google Scholar] [CrossRef]
  11. European Association for the Study of the Liver. Clinical Practice Guidelines: The diagnosis and management of patients with primary biliary cholangitis. J. Hepatol. 2017, 67, 145–172. [Google Scholar] [CrossRef]
  12. Galoosian, A.; Hanlon, C.; Zhang, J.; Holt, E.W.; Yimam, K.K. Clinical Updates in Primary Biliary Cholangitis: Trends, Epidemiology, Diagnostics, and New Therapeutic Approaches. J. Clin. Transl. Hepatol. 2020, 8, 49–60. [Google Scholar] [CrossRef]
  13. Gazda, J.; Drazilova, S.; Janicko, M.; Jarcuska, P. The Epidemiology of Primary Biliary Cholangitis in European Countries: A Systematic Review and Meta-Analysis. Can. J. Gastroenterol. Hepatol. 2021, 2021, 9151525. [Google Scholar] [CrossRef]
  14. Delgado, J.-S.; Vodonos, A.; Delgado, B.; Jotkowitz, A.; Rosenthal, A.; Fich, A.; Novack, V. Primary biliary cirrhosis in Southern Israel: A 20 year follow up study. Eur. J. Intern. Med. 2012, 23, e193–e198. [Google Scholar] [CrossRef]
  15. Lleo, A.; Colapietro, F. Changes in the Epidemiology of Primary Biliary Cholangitis. Clin. Liver Dis. 2018, 22, 429–441. [Google Scholar] [CrossRef]
  16. Zhang, H.; Carbone, M.; Lleo, A.; Invernizzi, P. Geoepidemiology, Genetic and Environmental Risk Factors for PBC. Dig. Dis. 2015, 33, 94–101. [Google Scholar] [CrossRef]
  17. Vespasiani-Gentilucci, U.; Rosina, F.; Pace-Palitti, V.; Sacco, R.; Pellicelli, A.; Chessa, L.; De Vincentis, A.; Barlattani, M.; Barlattani, A.; Feletti, V.; et al. Rate of non-response to ursodeoxycholic acid in a large real-world cohort of primary biliary cholangitis patients in Italy. Scand. J. Gastroenterol. 2019, 54, 1274–1282. [Google Scholar] [CrossRef]
  18. Örnolfsson, K.T.; Lund, S.H.; Olafsson, S.; Bergmann, O.M.; Björnsson, E.S. Biochemical response to ursodeoxycholic acid among PBC patients: A nationwide population-based study. Scand. J. Gastroenterol. 2019, 54, 609–616. [Google Scholar] [CrossRef]
  19. Trivedi, P.J.; Bruns, T.; Cheung, A.; Li, K.-K.; Kittler, C.; Kumagi, T.; Shah, H.; Corbett, C.; Al-Harthy, N.; Acarsu, U.; et al. Optimising risk stratification in primary biliary cirrhosis: AST/platelet ratio index predicts outcome independent of ursodeoxycholic acid response. J. Hepatol. 2014, 60, 1249–1258. [Google Scholar] [CrossRef]
  20. Yang, F.; Yang, Y.; Wang, Q.; Wang, Z.; Miao, Q.; Xiao, X.; Wei, Y.; Bian, Z.; Sheng, L.; Chen, X.; et al. The risk predictive values of UK-PBC and GLOBE scoring system in Chinese patients with primary biliary cholangitis: The additional effect of anti-gp210. Aliment. Pharmacol. Ther. 2017, 45, 733–743. [Google Scholar] [CrossRef]
  21. Cortez-Pinto, H.; Liberal, R.; Lopes, S.; Machado, M.V.; Carvalho, J.; Dias, T.; Santos, A.; Agostinho, C.; Figueiredo, P.; Loureiro, R.; et al. Predictors for incomplete response to ursodeoxycholic acid in primary biliary cholangitis. Data from a national registry of liver disease. United Eur. Gastroenterol. J. 2021, 9, 699–706. [Google Scholar] [CrossRef]
  22. Wilde, A.-C.; Lieb, C.; Leicht, E.; Greverath, L.; Steinhagen, L.; de Chamorro, N.W.; Petersen, J.; Hofmann, W.; Hinrichsen, H.; Heyne, R.; et al. Real-World Clinical Management of Patients with Primary Biliary Cholangitis—A Retrospective Multicenter Study from Germany. J. Clin. Med. 2021, 10, 1061. [Google Scholar] [CrossRef]
  23. Wen, M.; Men, R.; Fan, X.; Shen, Y.; Ni, P.; Hu, Z.; Yang, L. Worse Response to Ursodeoxycholic Acid in Primary Biliary Cholangitis Patients with Autoimmune Hepatitis Features. Dig. Dis. 2021, 39, 366–374. [Google Scholar] [CrossRef]
  24. Jiang, Y.; Xu, B.-H.; Rodgers, B.; Pyrsopoulos, N. Characteristics and Inpatient Outcomes of Primary Biliary Cholangitis and Autoimmune Hepatitis Overlap Syndrome. J. Clin. Transl. Hepatol. 2021, 9, 392–398. [Google Scholar] [CrossRef]
  25. Carbone, M.; Nardi, A.; Flack, S.; Carpino, G.; Varvaropoulou, N.; Gavrila, C.; Spicer, A.; Badrock, J.; Bernuzzi, F.; Cardinale, V.; et al. Pretreatment prediction of response to ursodeoxycholic acid in primary biliary cholangitis: Development and validation of the UDCA Response Score. Lancet Gastroenterol. Hepatol. 2018, 3, 626–634. [Google Scholar] [CrossRef] [Green Version]
  26. Cheung, A.C.; Lammers, W.; Perez, C.F.M.; van Buuren, H.R.; Gulamhusein, A.; Trivedi, P.J.; Lazaridis, K.N.; Ponsioen, C.Y.; Floreani, A.; Hirschfield, G.M.; et al. Effects of Age and Sex of Response to Ursodeoxycholic Acid and Transplant-free Survival in Patients With Primary Biliary Cholangitis. Clin. Gastroenterol. Hepatol. 2019, 17, 2076–2084. [Google Scholar] [CrossRef]
  27. Floreani, A.; Mangini, C.; Reig, A.; Franceschet, I.; Cazzagon, N.; Perini, L.; Caballeria, L.; Cocchio, S.; Baldo, V.; Pares, A. Thyroid Dysfunction in Primary Biliary Cholangitis: A Comparative Study at Two European Centers. Am. J. Gastroenterol. 2017, 112, 114–119. [Google Scholar] [CrossRef]
  28. Gazda, J.; Drazilova, S.; Janicko, M.; Grgurevic, I.; Kanizaj, T.F.; Koller, T.; Bodorovska, B.; Bozin, T.; Mijic, M.; Rob, Z.; et al. Prognostic Factors in Primary Biliary Cholangitis: A Retrospective Study of Joint Slovak and Croatian Cohort of 249 Patients. J. Pers. Med. 2021, 11, 495. [Google Scholar] [CrossRef]
  29. Corpechot, C.; Abenavoli, L.; Rabahi, N.; Chrétien, Y.; Andréani, T.; Johanet, C.; Chazouillères, O.; Poupon, R. Biochemical response to ursodeoxycholic acid and long-term prognosis in primary biliary cirrhosis. Hepatology 2008, 48, 871–877. [Google Scholar] [CrossRef]
  30. Gerussi, A.; Bernasconi, D.P.; O’Donnell, S.E.; Lammers, W.J.; Van Buuren, H.; Hirschfield, G.; Janssen, H.; Corpechot, C.; Reig, A.; Pares, A.; et al. Measurement of Gamma Glutamyl Transferase to Determine Risk of Liver Transplantation or Death in Patients With Primary Biliary Cholangitis. Clin. Gastroenterol. Hepatol. 2021, 19, 1688–1697.e1614. [Google Scholar] [CrossRef]
Figure 1. Study flow diagram. ICD, The International Classification of Diseases; PBC, primary biliary cholangitis; TLVMC, Tel Aviv Sourasky Medical Center; UDCA, ursodeoxycholic acid; ALP, alkaline phosphatase; EASL, European Association for the Study of the Liver.
Figure 1. Study flow diagram. ICD, The International Classification of Diseases; PBC, primary biliary cholangitis; TLVMC, Tel Aviv Sourasky Medical Center; UDCA, ursodeoxycholic acid; ALP, alkaline phosphatase; EASL, European Association for the Study of the Liver.
Jcm 10 04551 g001
Figure 2. Kaplan–Meier survival curve to achieve any clinical endpoint in UDCA responder (blue line) versus non-responders (red line).
Figure 2. Kaplan–Meier survival curve to achieve any clinical endpoint in UDCA responder (blue line) versus non-responders (red line).
Jcm 10 04551 g002
Table 1. Patients, disease characteristics, laboratory values, treatment patterns and clinical endpoints.
Table 1. Patients, disease characteristics, laboratory values, treatment patterns and clinical endpoints.
Number of Patients189
DemographicsFemale gender, n (%)175 (92.6)
Age at diagnosis, years, mean (std)54.7 (13.0)
Ethnicity, n (%)
Jewish Ashkenazi125 (72.3)
Jewish non-Ashkenazi43 (24.9)
Other5 (2.9)
Smoking, n (%)31 (23.1)
SymptomsFatigue, n (%)
Severe fatigue, n (%)
80 (47.9)
28 (14.8)
Pruritus, n (%)
Severe pruritus, n (%)
56 (29.6)
21 (11.1)
Other liver diseasesNASH, n (%)37 (20.0)
Cholecystitis/Cholecystectomy, n (%)28 (15.1)
PBC-AIH variant, n (%)11 (5.9)
Chronic HBV, n (%)2 (1.1)
Other autoimmune diseasesAny extrahepatic autoimmune disease, n (%)73 (39.2)
Hypothyroidism, n (%)37 (19.9)
Celiac, n (%)3 (1.6)
Scleroderma, n (%)4 (2.2)
Sjogren, n (%)11 (5.9)
Rheumatoid arthritis, n (%)9 (4.8)
Psoriasis, n (%)4 (2.2)
Systemic lupus erythematosus, n (%)2 (1.1)
Ulcerative colitis, n (%)3 (1.6)
Immune thrombocytopenic purpura, n (%)4 (2.2)
Raynaud’s syndrome, n (%)3 (1.6)
Other autoimmune disease, n (%)8 (4.3)
Baseline Laboratory results (before UDCA) mean (std)Hemoglobin (g/L)13.0 (1.2)
Platelets (×109/L)237 (80)
Creatinine (mg/dL)0.78 (0.22)
Total bilirubin (mg/dL)0.74 (0.56)
AST (IU/L)59.2 (43.1)
ALT (IU/L)72.7 (66.8)
ALP (IU/L)280 (200)
GGT (IU/L)342 (339)
Albumin (g/L)40.3 (5.5)
IgM (mg/dL)418 (382)
Immune SerologyAMA, n (%)147 (86.5)
ANA, n (%)108 (75.5)
ASMA, n (%)17 (13.5)
Anti-gp210, n (%)9 (32.1)
Anti-sp100, n (%)8 (29.6)
Std, standard deviation; NASH, non-alcoholic steatohepatitis; PBC-AIH, primary biliary cholangitis-autoimmune hepatitis; HBV, hepatitis B virus; AST, aspartate transaminase; ALT, alanine transaminase; ALP, alkaline phosphatase; GGT, gamma-glutamyltransferase; IgM, immunoglobulin M; AMA, anti-mitochondrial antibodies; ANA, anti-nuclear antibodies; ASMA, anti-smooth muscle antibodies.
Table 2. UDCA non-response rates according to different criteria.
Table 2. UDCA non-response rates according to different criteria.
UDCA Biochemical Response Rates
CriteriaDefinition of Biochemical ResponsenNon-responder Rate
TorontoALP ≤ 1.67 × ULN12828.1%
BarcelonaALP normalization or decrease >40%6940.6%
Paris-IALP ≤ 3 × ULN and AST ≤ 2 × ULN and normal total bilirubin8617.4%
Paris-IIALP ≤ 1.5 × ULN and AST ≤ 1.5 × ULN and normal total bilirubin8638.4%
RotterdamAlbumin and total bilirubin normalization9616.7%
GLOBE scoreBelow age-specific threshold8117.3%
UK PBC scoreRisk of event in 5 years < 5%796.3%
UDCA, ursodeoxycholic acid; ULN, upper limit of normal.
Table 3. Univariable analysis for parameters associated with UDCA non-response using Toronto criteria.
Table 3. Univariable analysis for parameters associated with UDCA non-response using Toronto criteria.
ResponderNon-ResponderUnivariable
Analysis
Number of Patients, n (%)92 (71.9)36 (28.1)OR (95% CI)p-Value
DemographicsFemale gender, n (%)87 (94.6)32 (88.9)0.46 (0.12–1.82)0.268
Age at diagnosis, years, mean (std)55.6 (12.6)48.8 (12.6)0.95 (0.92–0.99)0.009
Ethnicity, n (%)
Jewish Ashkenazi61 (71.8)26 (78.8)1.3 (0.5–3.38)0.588
Jewish non-Ashkenazi21 (24.7)7 (21.2)
Other3 (3.5)0 (0)
Smoking, n (%)13 (19.1)7 (28.0)1.49 (0.68–3.23)0.316
SymptomsFatigue, n (%)38 (43.7)22 (62.9)0.50 (0.22–1.10)0.083
Severe fatigue, n (%)13 (14.1)8 (22.2)1.37 (0.55–3.42)0.499
Pruritus, n (%)29 (32.6)15 (42.9)0.67 (0.30–1.46)0.311
Severe pruritus, n (%)9 (9.8)8 (22.2)2.22 (0.84–5.85)0.106
Other liver diseasesNASH, n (%)15 (16.7)10 (27.8)1.97 (0.79–4.93)0.145
Cholecystitis/Cholecystectomy, n (%)9 (10.0)8 (22.2)2.63 (0.93–7.49)0.069
PBC-AIH variant, n (%)3 (3.3)5 (13.9)4.78 (1.08–21.2)0.039
Chronic HBV, n (%)0 (0)1 (2.8)
Other AI diseasesExtrahepatic autoimmune disease, n (%)41 (45.1)13 (36.1)0.7 (0.32–1.56)0.385
Hypothyroidism, n (%)17 (18.7)4 (11.1)0.42 (0.13–1.33)0.141
Baseline
lab results
(before UDCA) mean (std)
Hemoglobin (g/L)12.9 (1.2)13.2 (0.9)1.19 (0.68–2.07)0.545
Platelets (× 109/L)236 (74)234 (98)1 (0.99–1.01)0.945
Creatinine (mg/dL)0.77 (0.23)0.76 (0.16)0.97 (0.03–31.15)0.984
Total bilirubin (mg/dL)0.62 (0.26)1.16 (1.02)5.84 (0.97–35.32)0.055
AST (IU/L) *50.4 (32.9)76.6 (63.6)1.13 (0.98–1.31)0.087
ALT (IU/L) *65.0 (69.4)101.1 (85.2)1.06 (0.98–1.14)0.142
ALP (IU/L) **222 (125)518 (309)1.38 (1.15–1.66)<0.001
GGT (IU/L) *275 (284)545 (479)1.02 (1–1.04)0.030
Albumin (g/L)41.2 (3.2)38.8 (10.8)1.02 (0.82–1.27)0.871
Immune SerologyAMA, n (%)75 (88.2)26 (78.8)0.5 (0.17–1.44)0.195
ANA, n (%)54 (74.0)25 (89.3)2.93 (0.79–10.83)0.107
ASMA, n (%)3 (4.9)8 (29.6)8 (1.92–33.26)0.004
Anti-gp210, n (%)2 (18.2)4 (44.4)
Anti-sp100, n (%)3 (27.2)2 (25.0)
Liver HistologyInterface Hepatitis, n (%)20 (57.1)16 (94.1)12 (1.43–100.8)0.022
Advanced Fibrosis, n (%)7 (21.2)5 (27.8)1.43 (0.38–5.38)0.598
* denotes OR for a 10-unit change ** denotes OR for a 50-unit change. Bold values indicate a statistical significance (p < 0.05).
Table 4. Univariable analysis for reaching a clinical endpoint.
Table 4. Univariable analysis for reaching a clinical endpoint.
HR (95% CI)p-Value
DemographicsFemale gender1.26 (0.17–9.55)0.826
Age at diagnosis0.96 (0.93–1.00)0.046
Ethnicity—Jewish Ashkenazi2.89 (0.66–12.64)0.159
Smoking0.59 (0.13–2.63)0.487
SymptomsFatigue1.37 (0.53–3.53)0.521
Severe fatigue0.90 (0.26–3.16)0.875
Pruritus3.45 (1.29–9.22)0.014
Severe pruritus4.29 (1.62–11.34)0.003
Other liver diseasesNASH1.72 (0.56–5.24)0.342
Cholecystitis/Cholecystectomy0.96 (0.22–4.24)0.957
PBC-AIH variant1.77 (0.50–6.23)0.374
Other AI diseasesExtrahepatic autoimmune disease0.89 (0.35–2.28)0.809
Hypothyroidism0.86 (0.28–2.60)0.790
Laboratory before UDCA Hemoglobin0.18 (0.02–1.40)0.101
Platelets0.99 (0.98–1.00)0.178
Creatinine16.21 (0.07–3710)0.315
Total bilirubin2.30 (0.89–5.96)0.086
AST *1.28 (0.94–1.74)0.115
ALT *1.17 (1.02–1.35)0.026
ALP **1.28 (1.04–1.58)0.022
GGT *1.03 (1.00–1.06)0.021
Albumin0.93 (0.86–1.00)0.044
Immune SerologyAMA5.40 (0.71–41.21)0.104
ANA2.42 (0.31–18.7)0.396
ASMA2.27 (0.60–8.61)0.227
* denotes OR for a 10-unit change ** denotes OR for a 50-unit change. Bold values indicate a statistical significance (p < 0.05).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Yehezkel, E.; Israel, I.; Houri, I.; Leshno, M.; Shibolet, O.; Zigmond, E. Real-World Management of Patients with Primary Biliary Cholangitis—A Retrospective Study from a Tertiary Medical Center in Israel. J. Clin. Med. 2021, 10, 4551. https://doi.org/10.3390/jcm10194551

AMA Style

Yehezkel E, Israel I, Houri I, Leshno M, Shibolet O, Zigmond E. Real-World Management of Patients with Primary Biliary Cholangitis—A Retrospective Study from a Tertiary Medical Center in Israel. Journal of Clinical Medicine. 2021; 10(19):4551. https://doi.org/10.3390/jcm10194551

Chicago/Turabian Style

Yehezkel, Eyal, Inbal Israel, Inbal Houri, Moshe Leshno, Oren Shibolet, and Ehud Zigmond. 2021. "Real-World Management of Patients with Primary Biliary Cholangitis—A Retrospective Study from a Tertiary Medical Center in Israel" Journal of Clinical Medicine 10, no. 19: 4551. https://doi.org/10.3390/jcm10194551

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop