a changing natural history of primary biliary cholangitis

140
A Changing Natural History of Primary Biliary Cholangitis and Its Influence on Risk Stratification by Carla Fiorella Murillo Perez A thesis submitted in conformity with the requirements for the degree of Master of Science Institute of Medical Science University of Toronto © Copyright by Carla Fiorella Murillo Perez (2018)

Upload: others

Post on 31-Dec-2021

1 views

Category:

Documents


0 download

TRANSCRIPT

A Changing Natural History of Primary Biliary Cholangitis and Its Influence on Risk Stratification

by

Carla Fiorella Murillo Perez

A thesis submitted in conformity with the requirements for the degree of Master of Science

Institute of Medical Science University of Toronto

© Copyright by Carla Fiorella Murillo Perez (2018)

ii

A changing natural history of primary biliary cholangitis and its

impact on risk stratification

Carla Fiorella Murillo Perez

Master of Science

Institute of Medical Science University of Toronto

2018

Abstract

We sought to describe temporal trends in the presenting characteristics and clinical course of

primary biliary cholangitis (PBC) from the 1970s to 2014 in a large international cohort of

patients. There was a 10-year increase in age at diagnosis and the proportion of patients

presenting with a mild biochemical and histological disease stage increased. Furthermore, recent

decades had improved decompensation and transplant-free survival rates. Since most patients

presented with normal bilirubin, we aimed to evaluate whether normal bilirubin is associated

with transplant-free survival. In patients with normal bilirubin at 1 year, the threshold with the

highest ability to predict liver transplantation or death was 0.6×ULN. Patients with normal

bilirubin, yet above this threshold, had a 2.1-fold increase in risk. In conclusion, the natural

history of PBC has changed over time and bilirubin levels ≤0.6×ULN are associated with the

lowest risk for liver transplantation or death in patients with PBC.

iii

Acknowledgments I would like to thank my family and friends for supporting and helping me on this journey

towards the completion of my Master’s.

I would like to express my gratitude towards my supervisor and mentors for their guidance,

advice, and support: Harry Janssen, Bettina Hansen, Jordan Feld, and Aliya Gulamhusein. It has

truly been a pleasure working with all of you.

I would also like to thank my international co-workers: Jorn Goet, Willem Lammers, Henk van

Buuren, Maren Harms, and Adriaan van de Meer who have collaborated with me.

I would also like to also acknowledge the MSc/PhD students at the Toronto Centre for Liver

Disease who have supported me: Seng Liem, Lisette Krassenburg, Mina Farag, Hooman

Zangneh, Surain Roberts, Jason Lau, and Hannah Choi.

I would like to thank the GLOBAL PBC Study Group members for welcoming me as a new

member and supporting my research.

iv

Table of Contents Acknowledgments.......................................................................................................................... iii

Table of Contents ........................................................................................................................... iv

List of Tables ............................................................................................................................... viii

List of Figures ..................................................................................................................................x

List of Abbreviations .................................................................................................................... xii

Chapter 1 : Literature Review ..........................................................................................................1

Primary Biliary Cholangitis ........................................................................................................1

1.1 Introduction ..........................................................................................................................1

1.2 Epidemiology .......................................................................................................................1

1.3 Etiology ................................................................................................................................3

1.3.1 Genetic factors .........................................................................................................3

1.3.2 Environmental factors ..............................................................................................5

1.4 Clinical Presentation and Symptoms ...................................................................................6

1.4.1 Associated disorders ................................................................................................6

1.4.2 Symptoms ................................................................................................................7

1.4.3 Symptom development and prognosis .....................................................................8

1.4.4 Ethnic differences in presentation and symptoms ...................................................9

1.5 Biochemistry, Serology and Histology ................................................................................9

1.5.1 Liver biochemistry ...................................................................................................9

1.5.2 Serology .................................................................................................................11

1.5.3 Histology ................................................................................................................14

1.6 Diagnosis............................................................................................................................16

1.7 Pathogenesis .......................................................................................................................16

1.7.1 Immunopathogenesis of PBC ................................................................................16

v

1.7.2 Loss of self-tolerance .............................................................................................18

1.8 Natural History and Prognosis ...........................................................................................18

1.9 Complications ....................................................................................................................19

1.9.1 Osteoporosis ...........................................................................................................19

1.9.2 Portal hypertension ................................................................................................20

1.9.3 HCC .......................................................................................................................21

1.9.4 Extrahepatic malignancies .....................................................................................21

1.9.5 Fat-soluble vitamin malabsorption ........................................................................21

1.10 Treatment ...........................................................................................................................22

1.10.1 Ursodeoxycholic acid.............................................................................................22

1.10.2 Adjuvant therapies .................................................................................................26

1.11 Predictors of Prognosis ......................................................................................................27

1.11.1 Prognostic models ..................................................................................................28

1.12 Liver Transplantation .........................................................................................................30

1.12.1 Recurrence of PBC after liver transplantation .......................................................31

1.13 Trends in PBC ....................................................................................................................31

Chapter 2 ........................................................................................................................................33

Aims and Hypothesis ................................................................................................................33

2.1 Study 1: Calendar Time Trends .........................................................................................33

2.1.1 Aims .......................................................................................................................33

2.1.2 Hypothesis..............................................................................................................33

2.1.3 Rationale for hypothesis ........................................................................................33

2.2 Study 2: Bilirubin Within the Normal Range ....................................................................34

2.2.1 Aims .......................................................................................................................34

2.2.2 Hypothesis..............................................................................................................34

2.2.3 Rationale for hypothesis ........................................................................................34

vi

Chapter 3 : Study 1 ........................................................................................................................36

Milder Disease Stage in Patients with Primary Biliary Bholangitis Over a 44-year Period: A Changing Natural History ..........................................................................................................36

3.1 Introduction ........................................................................................................................36

3.2 Patients and Methods .........................................................................................................37

3.2.1 Population and study design ..................................................................................37

3.2.2 Data collection .......................................................................................................38

3.2.3 Statistical analysis ..................................................................................................38

3.3 Results ................................................................................................................................39

3.3.1 Study population characteristics ............................................................................39

3.3.2 Age and sex trends .................................................................................................43

3.3.3 Liver biochemistry and serological status..............................................................44

3.3.4 Trends in biochemical and histological disease stage............................................46

3.3.5 Trends in UDCA-response rates ............................................................................46

3.3.6 Decompensation, HCC, and transplant-free survival ............................................49

3.4 Discussion ..........................................................................................................................51

3.5 Supplementary Tables and Figures ....................................................................................55

Chapter 4 : Study 2 ........................................................................................................................68

Bilirubin is Predictive of Transplant-free Survival Even Within the Normal Range in Patients with Primary Biliary Cholangitis .....................................................................................68

4.1 Introduction ........................................................................................................................68

4.2 Patients and Methods .........................................................................................................69

4.2.1 Population and study design ..................................................................................69

4.2.2 Data collection .......................................................................................................69

4.2.3 Statistical analysis ..................................................................................................70

4.3 Results ................................................................................................................................72

4.3.1 Study population characteristics ............................................................................72

vii

4.3.2 Normal bilirubin quartiles are associated with liver transplant-free survival ........74

4.3.3 Bilirubin threshold within the normal range ..........................................................75

4.3.4 The risk for liver transplantation or death increases at bilirubin levels of 0.6×ULN ................................................................................................................79

4.3.5 Patients who remain below 0.6×ULN over time have good long-term prognosis ................................................................................................................80

4.3.6 The proportion of patients with bilirubin ≤0.6×ULN increased over time ............80

4.4 Discussion ..........................................................................................................................82

4.5 Supplementary Tables and Figures ....................................................................................85

Chapter 5 ........................................................................................................................................91

General Discussion....................................................................................................................91

5.1 Calendar Time Trends........................................................................................................91

5.1.1 Discussion ..............................................................................................................91

5.1.2 Strengths and limitations........................................................................................96

5.1.3 Implications............................................................................................................97

5.2 Bilirubin Within the Normal Range ...................................................................................98

5.2.1 Discussion ..............................................................................................................98

5.2.2 Strengths and limitations......................................................................................100

5.2.3 Implications..........................................................................................................101

Chapter 6 ......................................................................................................................................103

Conclusions .............................................................................................................................103

Chapter 7 ......................................................................................................................................105

Future Directions .....................................................................................................................104

References ....................................................................................................................................105

Copyright Acknowledgements.....................................................................................................124

Appendix ......................................................................................................................................125

viii

List of Tables Table 1-1. Various response criteria to UDCA for PBC .............................................................. 25

Table 3-1. Demographic and clinical characteristics of PBC patients at study entry over calendar

time ............................................................................................................................................... 41

Table 3-2. Multivariable logistic regression for the attainment of biochemical response

according to Paris-Ia (n=2283) ...................................................................................................... 48

Table S3-1. Distribution of PBC patients across calendar time and center ................................. 55

Table S3-2. Calendar time trends in patients with a maximum lag of 2 years between diagnosis

and study entry .............................................................................................................................. 57

Table S3-3. Factorial ANOVA analysis of age at diagnosis over calendar time adjusting for sex

....................................................................................................................................................... 62

Table S3-4. Response rate in UDCA-treated patients according to various published criteria over

calendar time ................................................................................................................................. 63

Table S3-5. Response rate over calendar time in UDCA-treated patients who did not meet

criteria at baseline ......................................................................................................................... 64

Table S3-6. Multivariable Cox regression of 10-year hepatic decompensation (n=2962) .......... 65

Table S3-7. Multivariable Cox regression for 10-year HCC incidence (n=3963) ....................... 66

Table S3-8. Multivariable Cox regression analysis of 10-year transplant-free survival (n=3354)

....................................................................................................................................................... 67

Table 4-1. Characteristics of PBC patients in each normal bilirubin cohort ................................ 73

Table 4-2. Multivariable Cox regression analyses of various bilirubin thresholds in patients with

normal bilirubin at 1 year to evaluate performance for the prediction of liver transplantation and

death .............................................................................................................................................. 76

ix

Table S4-1. Multivariable Cox regression analyses of various bilirubin thresholds in patients

with normal bilirubin at time zero to evaluate performance for the prediction of liver

transplantation and death .............................................................................................................. 85

Table S4-2. Multivariable analysis of 0.6×ULN threshold at 1 year in various sub-groups ....... 87

x

List of Figures Figure 3-1. Age at diagnosis of PBC patients across different decades. ..................................... 43

Figure 3-2. Study entry characteristics associated with disease severity of patients diagnosed in

different decades.. ......................................................................................................................... 45

Figure 3-3. Response rates to ursodeoxycholic acid (UDCA) therapy over calendar time. ........ 47

Figure 3-4. Time-to-event analyses of decompensation, hepatocellular carcinoma (HCC), and

liver transplantation or death over calendar time. ......................................................................... 50

Figure S3-1. Mean age at diagnosis over calendar time stratified by A) Center (each line

corresponds to an individual center); B) Sex; and C) Biochemical disease stage. ....................... 60

Figure S3-2. Absolute number of patients according to age at diagnosis and over calendar time.

....................................................................................................................................................... 61

Figure 4-1. Transplant-free survival of the normal bilirubin quartiles in patients with normal

bilirubin at A) time zero and B) 1 year. ........................................................................................ 74

Figure 4-2. Transplant-free survival in patients with normal bilirubin (stratified by 0.6×ULN

threshold) and abnormal bilirubin. ................................................................................................ 77

Figure 4-3. Sub-group analyses based on the bilirubin threshold of 0.6×ULN in patients with

normal bilirubin at 1 year. ............................................................................................................. 78

Figure 4-4. The association between bilirubin levels (×ULN) and risk for liver transplantation or

death. ............................................................................................................................................. 79

Figure 4-5. Mean bilirubin levels over 5 years in patients with normal bilirubin at study entry

and stratified by outcome. ............................................................................................................. 81

Figure S4-1. Distribution of clinical events (liver transplantation, liver-related death, liver-

unrelated death) from the 10-year transplant-free survival rates associated with each bilirubin

group at A) time zero and B) 1 year. .............................................................................................86

xi

Figure S4-2. The association between bilirubin levels (mg/dL) and risk for liver transplantation

or death.......................................................................................................................................... 88

Figure S4-3. The association between bilirubin levels (×ULN) and risk for liver transplantation

(LT) or death. ................................................................................................................................ 89

Figure S4-4. The distribution of patients with bilirubin below and above 0.6×ULN in those with

normal bilirubin at baseline (n=2791)........................................................................................... 90

xii

List of Abbreviations AAR, AST/ALT ratio

AIH, autoimmune hepatitis

ALP, alkaline phosphatase

ALT, alanine aminotransferase

AMA, anti-mitochondrial antibody

ANA, antinuclear antibody

ANOVA, analysis of variance

APC, antigen presenting cell

APRI, AST to platelet ratio index

AST, aspartate aminotransferase

BCOADC, branched-chain oxo acid dehydrogenase complex

CI, confidence interval

ELISA, enzyme-linked immunosorbent assay

FDA, food and drug administration

FDR, first-degree relative

FXR, farnesoid X receptor

GGT, gamma-glutamyl transferase

HCC, hepatocellular carcinoma

HLA, human leukocyte antigen

HR; hazards ratio

HRQOL, health-related quality of life

IBD, inflammatory bowel disease

IIF, indirect immunofluorescence

IQR, interquartile range

xiii

LBR, lamin B receptor

LLN, lower limit of normal

MCMC, Markov chain Monte Carlo

MELD, model for end-stage liver disease

MHC, major histocompatibility complex

MMPT, mitochondrial membrane permeability transition

MMTV, mouse mammary tumor virus

MND, multiple nuclear dot

NHANES III, third national health and nutrition examination survey

NPC, nuclear pore complex

OADC, 2-oxo acid dehydrogenase complex

OCA, obeticholic acid

OGDC, oxoglutarate dehydrogenase complex

OR, odds ratio

PBC, primary biliary cholangitis

PDC, pyruvate dehydrogenase complex

PML, promyelocytic leukemia

POISE, PBC OCA international study of efficacy

PPAR, peroxisome proliferator-activated receptor

PSC, primary sclerosing cholangitis

PT, prothrombin time

RL/M, rim-like membranous

ROC, receiver operating characteristic

ROS, reactive oxygen species

SD, standard deviation

SMA, smooth muscle antibody

xiv

SMR, standard mortality ratio

TE, transient elastography

UDCA, ursodeoxycholic acid

UGT1A1, urine diphosphate glycosyltransferase 1A1

UK, United Kingdom

ULN, upper limit of normal

UTI, urinary tract infection

1

Chapter 1 : Literature Review

1 Primary Biliary Cholangitis

1.1 Introduction Autoimmune diseases constitute a diverse group of conditions that affect 5%-8% of the

population in which the immune system fails to distinguish between self and non-self antigens

(Fairweather & Rose, 2004). Some are organ-specific, while others can affect multiple systems.

Primary biliary cholangitis (PBC) is a chronic autoimmune disease that is characterized by

immune-mediated destruction of small and medium intrahepatic bile ducts. This process is slow

and progressive, thus, over time it may lead to fibrosis, cirrhosis, and liver failure. It is

considered a paradigmatic autoimmune disease due to its female predominance, its association

with other autoimmune diseases, and the presence of disease-specific anti-mitochondrial

antibodies (AMA) and antinuclear antibodies (ANA).

1.2 Epidemiology Like many other autoimmune diseases, PBC exhibits a typical female predominance and the

majority of female: male ratios have been reported to be 9/10:1. A systematic review that

assessed various epidemiological studies on PBC reported that the mean proportion of female

patients was 92% and ranged from 76% to 100% (Boonstra, Beuers, & Ponsioen, 2012).

Furthermore, recent reports suggest an increased proportion of males are affected by PBC, with

male: female ratios of 2.3:1, 4.2:1, and 6.2:1 in Lombardia, Denmark, and South Korea,

respectively (K. A. Kim et al., 2016; Lleo et al., 2016). However, studies from Finland and

Japan have not found evidence of changes in female: male ratios over time (Rautiainen et al.,

2007; Sakauchi, Oura, Ohnishi, & Mori, 2007). This may indicate that different populations have

varying male: female ratios, rather than there being a change over time.

PBC primarily affects middle-aged individuals and it is estimated that globally, 1 in 1,000

women over the age of 40 are affected by PBC (Hirschfield et al., 2017). The age at diagnosis of

PBC has been suggested to be increasing over time in a study of patients from Padova, Italy that

were diagnosed from 1973 to 2007, in which the mean age at diagnosis increased from 48 to 64

(Floreani et al., 2011). Even in symptomatic patients, an increase in median age has been

2

reported in a Japanese population from 59 years old in 1999 to 63 in 2004 (Sakauchi et al.,

2007).

Descriptive epidemiological studies that describe the incidence and prevalence of PBC in various

geographical regions are critical for understanding disease burden and gaining insight into the

etiology of PBC. Metcalf and James (1997) have described some criteria needed to conduct

proper descriptive epidemiological studies, which include strict inclusion criteria, multiple case

finding methods, and clear descriptions of disease onset, study period, geographical area, and the

population being studied. Although PBC affects people from all geographical regions and

ethnicities, it has an increased prevalence in Caucasian populations, especially those from

northern Europe, and the least prevalence in the Indian subcontinent and Africa (Howel et al.,

2000). Varying incidence and prevalence rates have been reported across different regions, in

which Northeast England, Iceland, and Minnesota have some of the highest prevalence rates,

whereas Australia and South Korea have some of the lowest prevalence rates (T. Baldursdottir et

al., 2012; James et al., 1999; K. A. Kim et al., 2016; W. R. Kim et al., 2000; Watson et al.,

1995).

The varying rates of incidence and prevalence according to geographical region may be due to

differences in genetics, ethnicity, and environmental factors. The contribution of genetics and

ethnicity was demonstrated in a study that compared the prevalence of PBC in Australia in

Australian-born individuals to that of migrant populations from Britain, Italy, and Greece. They

reported that the prevalence (per million) was higher in the migrant groups (Greece: 208, Italy:

200, and Britain: 141) compared to individuals that were born in Victoria, Australia (37) (Sood,

Gow, Christie, & Angus, 2004).

In an early review of descriptive epidemiologic studies for PBC, Metcalf and James (1997)

reported wide variations in incidence between regions from less than 25 cases per million

(Australia and Canada) to 200-252 cases per million (South Wales and Northeast England). They

also reported very few cases in sub-Saharan Africa and India (Metcalf & James, 1997). In a more

recent review of 24 studies published between 1972 and 2007, the annual incidence and

prevalence rates per 100,000 individuals ranged from 0.33-5.8 and 1.91-40.2, respectively

(Boonstra et al., 2012). Furthermore, all studies for which there were yearly prevalence rates

3

available for several consecutive years reported an increase in prevalence over time (Boonstra et

al., 2012). Conversely, temporal trends of incidence rates have been inconsistent, as some studies

report stable incidence rates and others report an increase over time (Boonstra et al., 2014;

McNally, James, Ducker, Norman, & James, 2014).

Rises in incidence and prevalence of PBC may be a surrogate of increased routine testing and

AMA testing as a result of improved disease awareness by physicians, which would suggest that

early epidemiological studies of PBC may have underestimated its prevalence (M. I. Prince &

James, 2003). On the contrary, if a true increase in incidence has occurred, it may be attributed to

an increased exposure to an environmental agent that triggers the disease or the aging population,

whom are inherently at risk for PBC (M. I. Prince & James, 2003). Furthermore, the increase in

prevalence of PBC can be attributed to the potential increase in survival of patients, an indication

of earlier diagnosis and improved care of PBC patients. Evidence for potentially improved

survival was observed in a population from Finland, in which there was an increase in the

median time from diagnosis to death from 1988 to 1999 (Rautiainen et al., 2007).

1.3 Etiology Although an exact cause for PBC is unknown, its development is speculated to be due to a

combination of both genetic and environmental factors. More specifically, genetics render an

individual susceptible to PBC, in whom the disease is triggered by exposure to an environmental

agent.

1.3.1 Genetic factors

Familial clustering and high concordance rates in monozygotic twins of 63% provide evidence to

suggest that genetic factors are implicated in the development of PBC (Hayase et al., 2005;

Selmi et al., 2004). There have been reports of PBC in sisters (Chohan, 1973), brothers (Bown,

Clark, & Doniach, 1975), and mother and daughter (Fagan, Williams, & Cox, 1977). The

prevalence rates of PBC in the family members of affected individuals are estimated to be

4%-9%. One particular study found estimates of 4282 per 100,000 (4.3%) in families compared

to 0.7-7.5 per 100,000 in the general population (Bach & Schaffner, 1994). Additionally, another

study reported that 5.9% of PBC patients had a family history of PBC (Hayase et al., 2005).

4

Unaffected relatives of patients with PBC still exhibit an increased frequency of extrahepatic

autoimmune diseases and immunological abnormalities, such as hypergammaglobulinemia

(increased IgM and IgA), and non-organ specific autoantibodies (ANA, smooth muscle

antibodies [SMA], and AMA) (Hayase et al., 2005). In a geographically-based cohort from the

United Kingdom (UK), 14% of first-degree relatives (FDR) of patients with PBC had an

autoimmune disease (Watt, James, & Jones, 2004). Additionally, both PBC patients and healthy

FDRs show abnormalities of in vitro suppressor cell function and T cell function (Miller,

Sepersky, Brown, Goldberg, & Kaplan, 1983). AMA positivity has been reported to be increased

in FDRs relative to age- and sex matched controls (Feizi, Naccarato, Sherlock, & Doniach,

1972; Lazaridis et al., 2007; Zografos et al., 2012). In one study, 7% of healthy relatives of

patients with PBC were AMA-positive, compared to none of the age- and sex- matched controls

(Feizi et al., 1972). In another study, the prevalence of AMA in FDRs and controls (age, sex,

and residence matched) was 13.1% and 1%, respectively (Lazaridis et al., 2007).

Genetic association studies of PBC have been primarily focused on genes from the major

histocompatibility complex (MHC)/human leukocyte antigen (HLA) region, which codes for 3

classes of proteins (class I, class II, and class III) that regulate the immune system. An

exploration into the potential genetic associations of PBC have been widely studied in many

populations and the only consistently reported genetic association has been with HLA-

DRB1*0801. Rates of this genetic variant have been shown to be increased in PBC patients

compared to population-specific healthy controls in Italy (12% vs 4%) and the UK (18% vs 6%)

(Donaldson et al., 2006).

A study on German PBC patients found no association with HLA class I, but they found an

increased association with HLA class II (HLA DRw8) and HLA class III (C4AQ0) in PBC

patients compared to healthy controls (Manns et al., 1991). In Danish patients, there have been

associations with B8, DR3, DQA1*0501, and DQB1*0201 (Morling et al., 1992). Other

associations in Caucasian populations have been with DR8 and DQB1*0402 (Gregory et al.,

1993; Underhill et al., 1992). Interestingly, there have also been reports of genetic loci that are

associated with a reduced risk for developing PBC, such DRB1*11 in an Italian population

(Invernizzi et al., 2003). The inconsistency of genetic association studies suggests that PBC is

5

genetically heterogeneous (Morling et al., 1992). Furthermore, variations between studies may

result from inherent problems in HLA serotyping and differences in methodology.

1.3.2 Environmental factors

Environmental factors are speculated to play a role in genetically susceptible individuals.

Although there have not been strong environmental factors associated with PBC, various

potential contributors have been reported, some of which are consistent across studies.

The female predominance of PBC has led to the hypothesis that reproductive factors may play a

role in the etiology of PBC. However, in a case-control study from the Netherlands, there were

no differences observed between PBC cases and controls in terms of age at menarche, age at first

pregnancy, and number of children (Boonstra et al., 2014). Smoking, tonsillectomy, and vaginal

or urinary tract infection (UTI) in females were some risk factors associated with PBC in a US

study (Parikh-Patel, Gold, Worman, Krivy, & Gershwin, 2001). Furthermore, in large case-

control studies, recurrent UTIs, active/past smoking, and the use of hormone replacement

therapy have been associated with an increased risk for PBC (Corpechot, Chrétien,

Chazouillères, & Poupon, 2010; Gershwin et al., 2005). Additional associations include the

frequent use of nail polish and hair dye (Gershwin et al., 2005; Mantaka et al., 2012; Prince,

Ducker, & James, 2010), while the use of oral contraceptives has been associated with a

decreased risk (Corpechot et al., 2010).

A smoking history is more common in patients with an advanced histological stage (III or IV) at

presentation, which suggests that smoking may accelerate the progression of PBC, independent

of its contribution to disease susceptibility (Corpechot et al., 2012; Zein et al., 2006). This

association was not related to an increase in histological inflammatory activity, severity of bile

duct damage, and biochemical and immunological markers of disease, which indicates that

tobacco smoking exerts direct, pro-fibrotic effects in PBC that are unrelated to immunity

(Corpechot et al., 2012).

Infectious agents have also been proposed to play a role in the development of PBC, such as

Chlamydia pneumoniae, whose antigens and RNA have been found in the liver tissue of PBC

patients (Abdulkarim et al., 2004). Additional infectious agents include Escherichia coli (E.

6

coli), mycobacteria, Novosphingobium aromaticivorans, Lactobacillus species, Helicobacter

pylori, viral infections (human retrovirus), and mouse mammary tumor virus (MMTV) (Mantaka

et al., 2012). Collectively, these studies indicate that exposure to multiple environmental factors

may contribute to PBC risk.

1.3.2.1 Geographical clustering

A phenomenon that supports the involvement of environmental factors in the etiology of PBC is

spatial clustering in specific locations that deviate from a random distribution, as has been found

in Northeast England and Alaska (Abu-Mouch et al., 2003; McNally et al., 2014; M. I. Prince et

al., 2001). In Northeast England, the risk for PBC was increased in geographical regions that had

higher levels of socioeconomic hardship, defined as those with overcrowded homes, an increased

proportion of households without cars, and higher levels of homes that were non-owned

(McNally et al., 2014). Furthermore, an increased prevalence of PBC patients has been reported

near toxic waste sites and among atomic bomb survivors from Hiroshima (Ala et al., 2006; Kita,

He, & Gershwin, 2004).

There is also evidence of space-time clustering, which is different from spatial clustering, and is

defined as an excess of cases within a small geographical region over a limited period of time

(McNally, Ducker, & James, 2009). This type of clustering was found in a population-based

study from Northeast England in which they also reported seasonal variation in the incidence of

PBC with a peak in diagnosis in June (McNally et al., 2009; McNally, James, Ducker, & James,

2011). The presence of this type of clustering may be indicative of a transient environmental

factor (infectious, air pollution, dietary factors) versus a static (non-infectious) factor (McNally

et al., 2009).

1.4 Clinical Presentation and Symptoms

1.4.1 Associated disorders

Patients with PBC have an increased prevalence of other autoimmune diseases (Gershwin et al.,

2005; Watt et al., 2004). In a geographically-based cohort from the UK, 53% of patients had at

least one autoimmune disease in addition to PBC (Watt et al., 2004). The most common

autoimmune disease associated with PBC is Sjogrens/sicca syndrome (69-81%), a disease that

7

affects the exocrine glands and leads to impaired glandular secretions and mucosa dryness

(Tsianos et al., 1990). However, they can also be affected by rheumatoid arthritis, complete or

incomplete CREST syndrome (calcinosis cutis, Raynaud syndrome, esophageal motility

disorder, sclerodactyly, telangiectasia), and thyroid disorders (Bittencourt et al., 2004; Parikh-

Patel et al., 2001; Reynolds, Denison, Frankl, Lieberman, & Peters, 1971; Siegel, Luthra,

Donlinger, Angulo, & Lindor, 2003; Tsianos et al., 1990)

1.4.2 Symptoms

Some of the symptoms that may be experienced by patients include fatigue, pruritus, pain in the

upper right quadrant, hyperlipidemia, keratoconjunctivitis, steatorrhea, and xerostomia (Siegel et

al., 2003). However, not all patients are affected in the same way as the presence and severity of

symptoms is heterogeneous. Furthermore, the symptoms affecting patients seem to have evolved

over time. Patients diagnosed in earlier times would frequently present with jaundice, which is

yellow discoloration of the skin, sclera, and mucous membranes that develops in late stages of

disease (Hirschfield et al., 2017). However, the proportion of asymptomatic patients at diagnosis

has reportedly been increasing over time, which implies an earlier diagnosis. In an early study,

13% of patients referred between 1955 and 1979 were asymptomatic at diagnosis, compared to

61% in prevalent cases from 1987 to 1994 (Mahl, Shockcor, & Boyer, 1994; M. Prince,

Chetwynd, Newman, Metcalf, & James, 2002). In Italy, there was an increase in the proportion

of asymptomatic patients at diagnosis from 25% to 46% from 1973 to 2007 (Floreani et al.,

2011). In Iceland, a decrease in the proportion of patients with symptoms at diagnosis was also

reported, from 57% in 1991-2000 to 36% in 2001-2010 (T. Baldursdottir et al., 2012). These

studies indicate that nowadays, up to 60% of patients are asymptomatic. Although asymptomatic

patients generally demonstrate a less advanced disease stage as indicated by biochemistry and

histology when compared to initially symptomatic patients, asymptomatic PBC does not

necessarily suggest early disease (M. Prince et al., 2002). There have been reports that 61% of

asymptomatic patients had a liver biopsy at baseline that demonstrated fibrosis or cirrhosis

(Balasubramaniam, Grambsch, Wiesner, Lindor, & Dickson, 1990). Furthermore, patients have

died prior to the development of any symptoms (M. Prince et al., 2002).

8

The most common symptoms at diagnosis are pruritus and fatigue, and least common are

hyperpigmentation, hepatomegaly, splenomegaly, and jaundice (M. Prince et al., 2002; Zein,

Angulo, & Lindor, 2003). Younger patients are more likely to report fatigue and pruritus

(Carbone et al., 2013). Fatigue, present in up to 50% of patients, is the most debilitating

symptom of PBC and imposes the greatest impact on quality of life, however, the degree of

fatigue does not correlate with any conventional parameters of disease severity, such as liver

histology or biochemistry (Cauch-Dudek, Abbey, Stewart, & Heathcote, 1998; Huet, Deslauriers,

Tran, Faucher, & Charbonneau, 2000; Newton et al., 2007; Newton, Gibson, Tomlinson, Wilton,

& Jones, 2006). In turn, fatigue does correlate with sleep quality (daytime somnolence) and is

associated with higher depression scores (Cauch-Dudek et al., 1998; Newton et al., 2006).

Patients with PBC not only experience decreased energy levels compared to an age- and sex-

matched control group, but they also demonstrate worse emotional reaction scores (R. E.

Poupon, Chrétien, Chazouillères, Poupon, & Chwalow, 2004). These findings emphasize the

importance of considering health-related quality of life (HRQOL) in PBC, defined as a patient’s

perception of their health status and the impact the disease poses on their life (R. E. Poupon et

al., 2004). PBC-40, a measure of HRQOL specifically for PBC was developed that allowed the

quantification of 6 domains that affect QOL: fatigue, emotional, social, cognitive function,

general symptoms, and itch (Jacoby et al., 2005). This measure was implemented in the UK-PBC

cohort and indicated that the majority (66%) of patients report good/neutral scores, but 34%

report poor scores (Dyson et al., 2016). Older age was associated with improved perceived QOL

for all symptom domains, except itch (Dyson et al., 2016).

1.4.3 Symptom development and prognosis

Although there are asymptomatic patients that don’t develop symptoms up to 10 years from

presentation, the majority will eventually develop symptoms during the course of disease and

experience progressive PBC (Long, Scheuer, & Sherlock, 1977; Mitchison et al., 1990; Springer,

Cauch-Dudek, O’Rourke, Wanless, & Heathcote, 1999). One or more symptoms developed in

89% of untreated patients during a median follow-up of 6.7 years (Balasubramaniam et al.,

1990). In another study, 50% of patients developed symptoms after 5 years and 95% after 20

years (M. I. Prince, Chetwynd, Craig, Metcalf, & James, 2004).

9

In older studies, the survival of asymptomatic patients was reported to be better than

symptomatic patients, but shorter than the general population (Mahl et al., 1994; Springer et al.,

1999). Once symptoms develop, however, there is no difference in the survival of patients who

developed symptoms during follow-up compared to patients who presented with symptoms

(Mahl et al., 1994; Mitchison et al., 1990). More recently, asymptomatic patients have shown to

have comparable survival to that of an age- and sex-matched population, especially if they

present at an early disease stage (Floreani et al., 2011; E. M. Kuiper et al., 2009).

Several attempts have been made to predict which patients may develop symptoms and who will

remain symptom-free based on liver biochemistry or histology, but these did not yield any

significant factors that could predict symptom development (Long et al., 1977; Mahl et al., 1994;

Springer et al., 1999). However, a recent study of asymptomatic ursodeoxycholic acid (UDCA)-

treated patients reported that response to UDCA at 6 months, defined by normalization or a

decrease in gamma-glutamyl transferase (GGT) greater than 70%, was an independent predictor

of symptom development (Azemoto et al., 2009).

1.4.4 Ethnic differences in presentation and symptoms

There is variability in disease severity at presentation of patients according to ethnicity. Non-

Caucasians are more likely to present with a more severe case of PBC, as suggested by lower

activity levels, more severe pruritus, and a greater incidence of hepatic complications (ascites,

hepatic encephalopathy, variceal bleeding) (Levy et al., 2014; Peters et al., 2007). Furthermore,

Hispanics are less likely to respond to treatment when defined as alkaline phosphatase (ALP)

<2× the upper limit of normal (ULN) at 1 year, in which the response rate of Hispanic patients

was 60% vs 88% for non-Hispanics (Levy et al., 2014).

1.5 Biochemistry, Serology and Histology

1.5.1 Liver biochemistry

Alterations in liver biochemistry can be used for the diagnosis of PBC and to help establish the

stage of disease, as well as its progression (Hirschfield et al., 2017). Early biochemical markers

of cholestasis are ALP and GGT, of which an elevation in GGT can be identified prior to an

elevation in ALP (K. D. Lindor et al., 2009; N. Suzuki et al., 2006; Zein et al., 2003). The

10

magnitude of elevation of ALP strongly correlates with the severity of ductopenia and

inflammation and is associated with disease progression (Hirschfield et al., 2017; K. D. Lindor et

al., 2009). There may also be mildly elevated transaminases (alanine aminotransferase [ALT]

and aspartate aminotransferase [AST]) at presentation. Although these are not diagnostic of PBC,

they reflect the extent of liver parenchyma inflammation and necrosis (Hirschfield et al., 2017;

K. D. Lindor et al., 2009; Zein et al., 2003). An increase in immunoglobulin (Ig) concentrations,

particularly IgG and IgM, is also observed in PBC, but they don’t correlate with the duration of

symptoms, degree of jaundice, ALP, or histology (Hirschfield et al., 2017; K. D. Lindor et al.,

2009; MacSween, Horne, Moffat, & Hughes, 1972). Increases in conjugated bilirubin, and

alterations in prothrombin time (PT) and serum albumin are not manifested in PBC until later

stages of disease (K. D. Lindor et al., 2009). Hyperbilirubinemia reflects the severity of

ductopenia and biliary piecemeal necrosis (K. D. Lindor et al., 2009). PBC, as with other

cholestatic diseases, is associated with elevations in serum cholesterol and may result in the

development of xanthomas and xanthelasmas, yellow colored nodules on the surface of the skin

owing to the deposit of cholesterol under the skin. However, hypercholesterolemia is not

associated with an increase in cardiovascular risk or mortality (Cash et al., 2010).

There are three distinct biochemical disease stages based on albumin and bilirubin at study entry.

These biochemical parameters were chosen based on the finding that bilirubin and albumin were

the prognostic factors consistently associated with survival. An early disease is defined as both

normal albumin and bilirubin, moderately advanced is defined as normal bilirubin or albumin,

and advanced is defined as abnormal bilirubin and albumin (ter Borg, Schalm, Hansen, & van

Buuren, 2006).

1.5.1.1 Metabolism of bilirubin

Bilirubin is different from the other liver biochemistry parameters that assess liver function

because its homeostasis is a complex process that entails various enzymes and transporters.

Unlike aminotransferases, for example, whose serum concentrations are dictated by their release

from hepatocytes, serum bilirubin concentrations are dictated by various factors. Bilirubin is

derived primarily from heme produced as a result of red blood cell turnover. Heme oxygenases

cleave heme and produce biliverdin, which is subsequently reduced by biliverdin reductase to

11

bilirubin (Levitt & Levitt, 2014). The production of bilirubin is beneficial since it is a free radical

scavenger that also possesses antioxidant and anti-inflammatory properties. After production,

bilirubin covalently bound to albumin is transported to the liver (delta bilirubin), where it is

rendered water-soluble through conjugation with glucuronic acid to be excreted by the liver with

bile. Conjugation of bilirubin is carried out by urine diphosphate glycosyltransferase 1A1

(UGT1A1). Total serum bilirubin is a composite of unconjugated, delta, and conjugated

bilirubin. In a healthy individual, measurements with high-performance liquid chromatography

of conjugated bilirubin indicate that it comprises 3% of total bilirubin (Levitt & Levitt, 2014).

1.5.1.1.1 Bilirubin in PBC

In PBC, chronic cholestasis results in the accumulation of conjugated bilirubin within the liver

and causes leakage back to the circulation, thereby increasing conjugated bilirubin levels.

Therefore, unlike healthy individuals whose total bilirubin is primarily composed of conjugated

bilirubin, the total bilirubin of PBC patients is predominantly comprised of conjugated bilirubin

with a proportion above 70% (Levitt & Levitt, 2014).

1.5.2 Serology

1.5.2.1 Anti-mitochondrial antibodies

The main serologic hallmark of PBC is positivity for AMA, which are present in 90-95% of

patients and can be detected before any liver-related symptoms appear (Zein et al., 2003). Thus,

AMA-positivity without evidence of biochemical cholestasis may indicate the subsequent

development of PBC (Lazaridis et al., 2007). Although AMA is the characteristic antibody for

PBC, it is not specific for PBC since 2% of patients with primary sclerosing cholangitis (PSC)

are also positive for AMA (Zein et al., 2003). The majority of early studies failed to demonstrate

a prognostic value of AMA since there were no correlations between AMA (IgG and IgM) and

histologic stage or biochemical variables (Mutimer et al., 1989). However, in a recent study,

positivity for AMA (IgG or IgA) at baseline and increased titers during follow-up were

associated with biochemically and histologically advanced disease (Gatselis et al., 2013). AMA-

negative patients, whom constitute 5-10% of patients with PBC, were initially regarded as

having a different clinical entity, yet they exhibit the same clinical, histological, and biochemical

12

features of PBC, including after treatment with UDCA, as AMA-positive patients (Invernizzi et

al., 1997; W. R. Kim et al., 1997).

The autoantigen of AMA is localized to the inner mitochondrial membrane and corresponds to

the M2 family of autoantigens (2-oxo acid dehydrogenase complex [OADC]), for which there is

cross-reactivity due to structural homology (Dähnrich et al., 2009; Flannery et al., 1989;

Mutimer et al., 1989). This functionally-related family of enzymes consists of pyruvate

dehydrogenase complex (PDC), branched-chain oxo acid dehydrogenase complex (BCOADC),

and oxoglutarate dehydrogenase complex (OGDC); each complex consists of multiple copies of

three enzymes (E1-E3) (Mutimer et al., 1989). AMA mainly recognize the E2 subunits of

OADC, of which PDC-E2 is the main autoantigen, since 80-90% of sera react with PDC-E2 and

approximately 10% only react to BCOADC-E2 and/or OGDC-E2 (Dähnrich et al., 2009; Gabeta

et al., 2007). E2 is a dihydrolipoamide acetyltransferase that is highly conserved across species

(Mutimer et al., 1989; Van de Water et al., 1989). The autoepitope of the autoantigen is a 20

amino acid peptide that corresponds to the lipoic acid binding site of PDC-E2 (Van de Water,

Gershwin, Leung, Ansari, & Coppel, 1988). The presence of the lipoyl residue, which is bound

covalently to a lysine residue, is crucial for an effective recognition by AMA since there is a

higher relative affinity for the lipoylated form (Quinn et al., 1993). Meanwhile, the E1 and E3

subunits of OADC are recognized at less frequent rates (Dähnrich et al., 2009).

1.5.2.2 Detection of anti-mitochondrial antibodies

Indirect immunofluorescence (IIF) on rodent multiorgan substrates was the gold standard method

for detecting AMAs, but this method is time-consuming, labor-intensive, and observer-

dependent (Dähnrich et al., 2009; Gabeta et al., 2007; Muratori et al., 2004). In recent years there

has been a shift towards enzyme-linked immunosorbent assay (ELISA) and western

immunoblots as the first-line assays for the detection of AMA because these approaches yield

greater sensitivity and specificity as compared to IIF (Muratori et al., 2004; Van de Water et al.,

1989). A meta-analysis reported that the sensitivity and specificity of ELISA was 84.5% and

97.8%, respectively (S. Hu, Zhao, Wang, & Chen, 2014). Furthermore, these methods are

considered superior because of their objectivity, rapid speed, and semi-automation (Gabeta et al.,

2007; Van de Water et al., 1989).

13

MIT3 is a more recently developed ELISA assay that includes the epitopes from BCOADC-E2

and OGDC-E2 substrates in addition to PDC-E2 to increase the specificity and sensitivity of

AMA detection as compared to IIF and conventional anti-M2 ELISA (Gabeta et al., 2007).

Furthermore, in order to be able to detect antibodies against PDC-E1, a hybrid MIT3 clone was

constructed and mixed with a native PDC antigen (Dähnrich et al., 2009). This newly developed

anti-MIT3/PDC ELISA was compared to the conventional anti-PDC ELISA, anti-MIT3, and IIF,

of which anti-MIT3/PDC showed best diagnostic sensitivity.

1.5.2.3 Antinuclear antibodies

In addition to AMA, patients with PBC may also exhibit positivity for other serum

autoantibodies such as ANA and SMA. However, these additional autoantibodies are more

frequently observed in AMA-negative patients than AMA-positive patients (71% vs 31%)

(Invernizzi et al., 1997). ANAs are directed against proteins of the nuclear envelope and are

observed in 30-50% of patients (C. J. Hu et al., 2012; Shimoda et al., 2003; Wesierska-Gadek,

Hohenuer, Hitchman, & Penner, 1996). The specificity for ANA has been shown to be 99%, and

thus ANA positivity would strongly suggest a diagnosis of PBC, irrespective of AMA status

(Granito et al., 2006).

There are two distinct ANA patterns that are detectable by IIF, multiple nuclear dot (MND) and

Rim-like/membranous (RL/M) patterns. MND is characterized by staining of 3-20 dots of

variable size distributed throughout the cell nucleus and sparing nucleoli. Sp100 and

promyelocytic leukemia antigen (PML) give rise to this type of staining and can be found

simultaneously in 90% of patients. Anti-Sp100 can be observed in 20-40% of patients and has

a specificity of 97% and sensitivity of 30% (Shimoda et al., 2003).

The nuclear pore complex (NPC), a system that mediates molecular trafficking between the

nucleus and cytoplasm contains ANA antigens that give rise to RL/M staining, namely gp210,

lamin B receptor [LBR], and nucleoporin p62. These have high specificity for PBC of over 99%

(Granito et al., 2012; Muratori et al., 2003). Out of NPC antigens, the majority of patients (10-

40%) have autoantibodies against gp210, an integral glycoprotein, while p62 and LBR are

observed less frequently (Shimoda et al., 2003; Wesierska-Gadek et al., 1996).

14

ANAs have been associated with disease severity and can be considered a marker of poor

prognosis. PBC patients with ANA positivity tend to have a more severe biochemical and

histological disease compared to those seronegative, particularly those with IgG3 isotype

(Rigopoulou et al., 2005). Furthermore, bilirubin increased above 2mg/dL in anti-NPC positive

patients at increased rates compared to anti-NPC negative patients (26% vs 5%) (Wesierska-

Gadek et al., 2006). Specifically, anti-gp210 antibodies are more frequent in patients with more

pronounced cholestasis and impaired liver function and are associated with more severe interface

hepatitis and lobular inflammation (Gatselis et al., 2013; C. J. Hu et al., 2012; Muratori et al.,

2003; Nakamura et al., 2007). Positivity for anti-sp100 at baseline was also associated with

biochemically and histologically advanced disease, with decreases in anti-sp100 being reportedly

associated with improved Mayo risk scores and response according to Barcelona criteria

(Gatselis et al., 2013).

Additional autoantibodies other than AMA and ANA include anti-lymphocytotoxic, anti-

thyroglobulin, and anti-centromere (Miller et al., 1983; Nakamura et al., 2007; Watt et al., 2004).

Anti-centromere antibodies have been identified as a risk factor for the development of

esophageal varices or HCC without jaundice and may be a surrogate for a higher propensity to

develop portal hypertension. Additionally, anti-centromere antibodies are associated with more

severe ductular proliferation (Nakamura et al., 2007).

1.5.3 Histology

PBC is histologically characterized by portal inflammation and immune-mediated destruction of

intrahepatic small- and medium-sized bile ducts. There are four histologic stages associated with

PBC that have been described by Ludwig et al. and Scheuer (Ludwig, Dickson, & McDonald,

1978; Scheuer, 1967). Stage 1 is defined as the florid bile duct lesion (granulomatous destruction

of interlobular bile ducts although granulomas not always observed) restricted to the portal area

Stage 2 is periportal hepatitis and ductular proliferation. Stage 3 is characterized by septal

fibrosis or bridging necrosis, while stage 4 is cirrhosis. The majority of patients without effective

therapy will progress histologically within 2 years, as progression was observed in 62% of

patients with stage I or II, and 50% of patients in stage III (Locke, Therneau, Ludwig, Dickson,

15

& Lindor, 1996). Only a minority of precirrhotic patients (20%) showed histological stability,

while histological regression was only observed in 2% of patients.

Liver biopsies are no longer a mandatory requirement for the diagnosis of PBC and are therefore

not routinely performed. They are only beneficial for the diagnosis of PBC in AMA-negative

patients or a minority of AMA-positive patients who don’t demonstrate a cholestatic biochemical

profile (Zein et al., 2003). Some of the drawbacks for routinely performing a liver biopsy include

its invasive nature, high cost, interobserver and intraobserver bias, and the small risk for

complications, such as post biopsy pain, bleeding, bile duct injury, or penetration of the

abdominal viscera (Alempijevic et al., 2009; Su et al., 2009). The incidence of complications and

mortality after a liver biopsy has been reported to be 0.3% and 0.018%, respectively.

The prognostic information rendered by histological staging can be particularly important.

Therefore, non-invasive tools that can assess the degree of liver fibrosis have been sought to

replace liver biopsies and include biochemical markers and transient elastography. AST/ALT

ratio (AAR) is a potential biochemical marker that can evaluate fibrosis and is significantly

higher in patients with advanced fibrosis, yet its correlation with fibrosis has been variable with

reported receiver operating characteristic (ROC) values of 0.66 and 0.85 (Alempijevic et al.,

2009; Su et al., 2009). Furthermore, AST to platelet ratio index (APRI) is a non-invasive marker

that can capture liver fibrosis and portal hypertension (Trivedi et al., 2014). APRI > 0.54 at

baseline and 1 year have been independently associated with transplant-free survival. A more

promising way of evaluating liver fibrosis is the use of transient elastography (TE), a non-

invasive and safe technique that measures liver stiffness (Gómez-Dominguez et al., 2008). This

technique utilizes ultrasounds of 5MHz and low-frequency elastic waves to create an elastic

shear wave that propagates within the liver tissue, while a pulse echo ultrasound simultaneously

measures the velocity of the shear wave. In this context, the velocity is directly related to the

stiffness of the liver and high velocities indicate high liver stiffness. A significant correlation

between histological fibrosis stage and liver stiffness has been found (Gómez-Dominguez et al.,

2008).

16

1.6 Diagnosis The diagnosis of PBC is based on two of the following criteria: 1) biochemical evidence of

cholestasis with an elevated ALP; 2) presence of AMA at titers above 1:40; 3) Histopathologic

evidence of non-suppurative cholangitis and destruction of small- or medium-sized bile ducts if

biopsy is performed (K. D. Lindor et al., 2009). Interestingly, prior to the development of any

clinical or biochemical indication for PBC, AMA-positive patients whose histology is

compatible with PBC are affected but seem to have a slow progression. This was supported by a

study that reported 76% of patients eventually developed symptoms of PBC and 83% developed

cholestatic liver function tests (Metcalf et al., 1996).

1.7 Pathogenesis

1.7.1 Immunopathogenesis of PBC

Abnormalities in humoral and cell-mediated immunity, the presence of autoantibodies, abnormal

complement activation and clearance, abnormal results of in vitro immune function tests, and a

greater association with other autoimmune diseases suggests that the immune system plays a role

in the pathogenesis of PBC (Miller et al., 1983; Nakanuma, 1993). The infiltration of plasma

cells and lymphocytes into the portal tracts of patients with PBC suggests that there is an intense

inflammatory reaction.

1.7.1.1 The pathogenic role of T cells

The destruction of biliary epithelial cells is thought to be mediated by liver-infiltrating

autoreactive T cells (Löhr et al., 1993). Activated T cells with high cytotoxic activity including

CD8+ T cells and CD4+ T cells are the predominant infiltrates surrounding the portal tracts (Kita

et al., 2004; Löhr et al., 1993; Nakanuma, 1993). The major autoreactive antigen of these T cells

is also PDC-E2, similar to AMA. The immunodominant epitope of MHC class II-restricted

CD4+ T cells specific for PDC-E2 has been identified as amino acid residues 163-176 of PDC-

E2 (Kita et al., 2002; Shimoda et al., 2003), while the epitope for MHC class I-restricted CD8+ T

cells lies within amino acids 159-167 (Kita et al., 2002). These PDC-E2-specific T cells are

detected in the peripheral blood, liver, and portal lymph nodes of patients with PBC (Kita et al.,

2002; Shimoda et al., 2003). However, it seems that they are predominantly located in the liver

17

of PBC patients, as there is a 10-fold increase in the frequency of CD8+ T cells specific for

PDC2-E2 in the liver compared to peripheral blood (Kita et al., 2002).

Further evidence to support the immunologic role of T cells is the high expression of HLA class

II on bile duct epithelium (Ballardini et al., 1984; Löhr et al., 1993; Underhill et al., 1992). HLA

class II are cell surface glycoproteins that play an important role in presenting antigens to

regulate immunologic reactions, and their expression is normally restricted to antigen presenting

cells (APCs) (Ballardini et al., 1984; Underhill et al., 1992). It is speculated that this promotes

autorecognition because it may enable the bile duct epithelial cells to present self-antigens to T-

lymphocytes.

Furthermore, there is an increased expression of PDC-E2 on biliary epithelial cells (Shimoda et

al., 2003). It has been suggested that CD4+ helper T cells and the expression of MHC class II

antigens on biliary epithelial cells may be particularly important in the early stages of PBC

(Nakanuma, 1993).

1.7.1.2 The pathogenic role of AMA

Although AMA is present at high titers in the majority of patients, its pathogenic role remains

enigmatic. Evidence that suggests AMA contributes to the pathogenesis of PBC is their ability

to inhibit the enzymatic activity of PDC in vitro and the ability of IgA specific AMA to

undergo transcytosis in biliary epithelial cells that potentially predisposes cells for apoptosis

(Löhr et al., 1993; Matsumura et al., 2004). Although B cells can be found in the bile ducts of

patients, these may not play a relevant role in the pathogenesis of PBC, as they are relatively

scarce in the liver and neither the presence of AMA nor titers correlate with recurrence after

liver transplantation (Nakanuma, 1993; Neuberger, 2003).

1.7.1.3 Non-immune mediated mechanisms

The immunologic destruction of bile ducts results in decreased bile secretion, and therefore there

is retention of endogenous toxic primary bile acids and copper in the liver, which can result in

hepatocellular damage (Aboutwerat et al., 2003; Crosignani et al., 1991). It has also been

suggested that the generation of reactive oxygen species (ROS), such as superoxide anion and

18

hydroxyl radical may play role. Oxidant stress seems to be a significant feature of early stage

PBC (Aboutwerat et al., 2003).

1.7.2 Loss of self-tolerance

Although the exact process by which self PDC-E2 becomes antigenic is unknown, there are

several mechanisms that have been suggested to play a role based on experimental evidence,

including molecular mimicry, self-alteration of PDC-E2 by xenobiotics, and intact immunogenic

epitopes released from apoptotic biliary epithelial cells (M M Kaplan & Gershwin, 2005;

Lazaridis et al., 2007). The most common mechanism described is molecular mimicry between a

self-antigen and an exogenous bacterial or virologic antigen. This mechanism signifies that an

infectious agent is responsible for the initiation of PBC and seems plausible considering the

highly conserved nature of PDC-E2, especially the inner lipoyl domain. Some evidence for this

mechanism includes cross-reactivity between human PDC-E2 and bacterial E. coli E2 (Bogdanos

et al., 2004). The second mechanism by which self-tolerance may be lost is the replacement of

the lipoic acid bound to E2 with a chemical xenobiotic mimic. 107 potential xenobiotic mimics

were coupled to the lysine residue of PDC-E2 and tested against sera from PBC patients for

immunoglobulin reactivity (Amano et al., 2005). Nine of these xenobiotic mimics were found to

yield higher reactivity with PBC sera compared to control sera, and when compared to the native

lipoylated peptide. One particular xenobiotic that exhibited high reactivity with sera from PBC

patients was 2-octynoic acid-PDC-E2, a chemical that can modify PDC in vivo and is widely

used in cosmetics (Amano et al., 2005). Another study also identified 2-octynoic acid as a high

affinity reactant specifically for AMA (Rieger et al., 2006).

1.8 Natural History and Prognosis PBC develops over the course of many years and the rate of progression varies from one patient

to another. There are three irreversible stages: 1) cirrhosis; 2) a terminal phase when serum

bilirubin levels reach 6mg/dL; 3) death unless orthotopic liver transplantation is performed

(Corpechot et al., 2005; Hirschfield et al., 2017).

The survival of patients with PBC has been shown in multiple populations to be poorer than the

age- and sex-matched population (Corpechot et al., 2005; Krzeski et al., 1999; Myers et al.,

19

2009; M. Prince et al., 2002; ter Borg et al., 2006). In a UK study of 930 people with PBC whose

survival was compared to 9202 controls, there was a 2.7-fold increase in adjusted mortality for

the PBC cohort compared to the general population (Jackson et al., 2007). A study from

Northeast England reported the standard mortality ratio (SMR) of prevalent cases between 1987

and 1994 with PBC to be 2.87, and the 10-year survival to be 45% (M. Prince et al., 2002).

Although the majority of deaths are expected to be due to liver disease, a significant proportion

of mortality in these patients was a result of non-liver related deaths (SMR=1.73 when excluded

liver-related deaths), which suggests that PBC patients may be at an increased risk of death from

causes not directly related to the development of advanced liver disease (M. Prince et al., 2002).

A Canadian population-based study of patients diagnosed between 1996 and 2002 reported the

same SMR of 2.87, but reported that the 10-year transplant-free survival rate was 68% (Myers et

al., 2009).

Treatment with UDCA is expected to have an impact on survival since these patients show a less

pronounced increase in mortality with a probability of survival of 65% at 20 years (Floreani et

al., 2011; Jackson et al., 2007). However, the transplant-free survival of UDCA-treated patients

remains lower than an age- and sex-matched control (Corpechot et al., 2008). Some UDCA-

treated patients may still progress toward cirrhosis and end-stage liver disease, as the incidence

of cirrhosis after 5 years from stages I-III was 4%, 12%, and 59%, respectively (Corpechot,

Carrat, Poupon, & Poupon, 2002). Nonetheless, a similar survival to the control population may

be achieved if UDCA is given at early histological stages (I or II) (Corpechot et al., 2005).

1.9 Complications

1.9.1 Osteoporosis Osteoporosis is a progressive systemic skeletal disease whose main features include low bone

mass and deterioration of bone tissues. Although elderly women are naturally prone to

osteoporosis, untreated women with PBC have been shown to lose bone mass at double the rate

compared to age- and sex-matched controls (Mounach et al., 2008). Furthermore, they have an

increased risk for developing osteoporosis as well as a 2-fold increase in the risk for any type of

fracture (Mounach et al., 2008; Solaymani-Dodaran, Card, Aithal, & West, 2006). The

prevalence of osteoporosis was 51.5% in PBC vs 22.7% in a healthy control group (Mounach et

20

al., 2008). Osteoporosis has the potential to greatly impact morbidity, quality of life and even

survival of patient because they are more susceptible to spontaneous or low-trauma fracturing

due to bone fragility.

1.9.2 Portal hypertension

About a third of patients with PBC will develop portal hypertension, which can manifest into

esophageal varices. In a prospective study that screened 265 patients annually for esophageal

varices by endoscopy for a median of 5.6 years, 31% developed esophageal varices, of whom

48% experienced one or more episodes of bleeding (Gores et al., 1989). In another study, portal

hypertension was associated with the development of esophageal varices in up to 35% of patients

during a 4-year period (K D Lindor, Jorgensen, Therneau, Malinchoc, & Dickson, 1997).

Although the use of beta blockers can reduce the risk of the first occurrence of variceal bleeding

and subsequent episodes, the development of esophageal varices is associated with a higher

mortality risk and thus, patients at risk should be closely monitored (Angulo, Lindor, et al., 1999;

Floreani et al., 2011; Gores et al., 1989).

A platelet count of less than 140,000 cu mm and a Mayo risk score ≥4.5 have been reported to be

independent predictors of the development of varices (Levy et al., 2007). After developing

varices, 1- and 3-year survival estimates have been reported to be 83% and 59%, respectively

(Gores et al., 1989).

Portal hypertension can occur even during early stages of disease as a result of significant portal

tract inflammation causing portal venous compression, perisinusoidal fibrosis, and nodular

regenerative hyperplasia. However, they are most common in cirrhotic patients, of whom 50%

are affected by this complication and have a 2-15% annual incidence of bleeding (D’Amico &

Luca, 1997).

Other complications include ascites, the retention of fluid in the abdomen as a result of sodium

retention, and hepatic encephalopathy, a reversible state in which cognitive function is impaired

(Krige & Beckingham, 2001).

21

1.9.3 HCC

One of the least common complications of PBC is the development of hepatocellular carcinoma

(HCC), which affects 0.7-3.6% of patients followed for 3.6-6.8 years (Shibuya et al., 2002; A.

Suzuki et al., 2007). Patients with an advanced histological disease are affected at higher rates of

5.9%-11.1% and therefore HCC surveillance is recommended in patients with cirrhosis (Deutsch,

Papatheodoridis, Tzakou, & Hadziyannis, 2008; Shibuya et al., 2002; Silveira, Suzuki, & Lindor,

2008; A. Suzuki et al., 2007). Older age, male sex, history of blood transfusions, and any signs of

portal hypertension have been associated with the development of HCC (Shibuya et al., 2002; A.

Suzuki et al., 2007; Trivedi et al., 2016). Although UDCA treatment is not associated with HCC

development, non-response to UDCA treatment has been described as a significant predictor for

HCC development (E. M. M. Kuiper, Hansen, Adang, et al., 2010; Trivedi et al., 2016). The

impact of HCC on survival is unclear since an earlier study suggested that HCC was not

associated with survival, however, HCC development has been recently associated with worse

transplant-free survival and overall survival in a globally representative cohort (Shibuya et al.,

2002; Trivedi et al., 2016).

1.9.4 Extrahepatic malignancies

Patients with PBC also tend to be at an increased risk for extrahepatic malignancies that exceeds

the risk for HCC development (Deutsch et al., 2008; Liang, Yang, & Zhong, 2012). In a study of

Greek patients, 10.8% had malignancies, of which 3.8% were attributed to HCC and 7% were

extrahepatic malignancies. The 10-year risk for HCC and extrahepatic malignancies was 4% and

13%, respectively (Deutsch et al., 2008).

1.9.5 Fat-soluble vitamin malabsorption

Deficiencies in fat-soluble vitamins, such as A, D, E, K have been well documented in patients

with PBC (Muñoz, Heubi, Balistreri, & Maddrey, 1989; Phillips, Angulo, Petterson, & Lindor,

2001). Due to cholestasis, an inadequate quantity of bile salts is delivered to the intestinal

lumen and therefore results in fat-soluble vitamin malabsorption and deficiency

(Phillips et al., 2001). The mechanism of vitamin E deficiency is related to gastrointestinal

malabsorption of vitamin D (Muñoz et al., 1989). The proportion of patients with vitamin A, D,

22

E, K deficiency was reported as 33.5%, 13.2%, 1.9%, and 7.8%, respectively (Phillips et al.,

2001).

1.10 Treatment

1.10.1 Ursodeoxycholic acid

The standard treatment for PBC is UDCA, which is required as life-long treatment (U. Leuschner

et al., 1989). UDCA (3α, 7β-dihydroxy-5β-cholanic cid) is normally found endogenously but

makes up only 3% of the total bile acid pool (Paumgartner & Beuers, 2002). The off-label use of

UDCA began in the late 1980s, as suggested in a study that reported that all patients that were

diagnosed from 1988 onward were all treated with UDCA (Floreani et al., 2011). However,

UDCA did not gain Food and Drug Administration (FDA) approval until 1997. The dosage of

UDCA administered is important and dosages of 13-15mg/kg per are normally recommended. A

comparison of 3 dosages (5-7mg/kg, 13-15mg/kg, and 23-25mg/kg) showed that UDCA dosages

of 13-15mg/kg and 23-25mg/kg yield greater improvements in ALP, AST, and Mayo scores

when compared to the low dosage. However, the high dose group was not superior to the

standard dose group of 13-15mg/kg (Angulo, Dickson, et al., 1999). Furthermore, in a Dutch

study that assessed the treatment regimen of UDCA, inappropriate dosages at a median of

9.33mg/kg were mostly administered to patients diagnosed before 1999 (Lammers et al., 2016).

35% of non-responders that were initially given inappropriate dosages and in whom the dosage

was increased became responders after 2 years from the dosage change.

Several large randomized, double-blind, placebo-controlled trials have consistently shown that

UDCA improves liver biochemistry parameters, such as ALP, AST, ALT, bilirubin, cholesterol

and IgM, as early as 3 months from the start of treatment (Battezzati et al., 1993; Heathcote et

al., 1994; K. D. Lindor et al., 1994; R. E. Poupon, Poupon, & Balkau, 1994). In the Canadian

multicenter trial of 222 patients treated with 14mg/kg for 2 years, UDCA was shown to improve

some histological features, but did not have an impact on symptoms, liver transplantation, or

death (Heathcote et al., 1994). In the French multicenter trial of patients treated with UDCA at a

dosage of 13-15mg/kg for 4 years, three endpoints were evaluated: 1) progression as defined by

hyperbilirubinemia, ascites, variceal bleeding, or encephalopathy; 2) liver transplantation or

referral; and 3) transplant-free survival. This study showed that long-term UDCA slows

23

progression and reduces the need for liver transplantation (R. E. Poupon et al., 1994). The

American trial reported that UDCA delayed progression of disease, but failed to show an effect

on symptoms, histology, and liver transplantation or survival (K. D. Lindor et al., 1994). The

inconsistency in survival benefit results led to a combined analysis of data from these three trials

to determine the effect of UDCA after 4 years of treatment. The study concluded that long-term

UDCA improves transplant-free survival in patients with moderate or severe disease, as defined

with bilirubin 1.4-3.5mg/dL or >3.5mg/dL, and histological stage IV (R. E. Poupon et al., 1997).

Although some studies have reported that UDCA does not have an impact on liver

transplantation or death, multiple studies have confirmed the beneficial effect of UDCA on liver

transplantation and survival (K. D. Lindor, Therneau, Jorgensen, Malinchoc, & Dickson, 1996;

Parés et al., 2000; R. E. Poupon, Bonnand, Chrétien, & Poupon, 1999; Shi et al., 2006).

UDCA delays histological progression, particularly in those with early stage PBC (Parés et al.,

2000; Shi et al., 2006). Additionally, UDCA has been shown to reduce the risk of developing

esophageal varices in patients with PBC, as the 4-year risk for developing new varices was 16%

with UDCA and 58% in the placebo group (K. D. Lindor et al., 1997). Conversely, a meta-

analysis reported that UDCA does not seem to exert any significant effects on pruritus and

fatigue (Shi et al., 2006).

1.10.1.1 Mechanism of action of UDCA

In PBC, there is impaired hepatic clearance of bile acids, which leads to the accumulation of

endogenous hydrophobic bile acids in the liver and serum. The accumulation of bile acids in the

liver can lead to hepatocellular toxicity. Although a clear mechanism of action for UDCA is

unclear, it is likely multifactorial and experimental evidence suggests that there are three major

mechanisms of action that can be attributed to UDCA. First, UDCA protects cholangiocytes

against the cytotoxicity of hydrophobic bile acids by reducing the cytotoxicity of bile and

possibly reducing the concentration of hydrophobic bile acids in cholangiocytes (Paumgartner &

Beuers, 2002). UDCA has fewer hepatotoxic properties compared to endogenous bile acids and

treatment with UDCA leads to this hydrophilic acid becoming the predominant bile acid in the

pool (40-50%), while endogenous bile acids such as cholic acid are decreased, thus increasing

the degree of hydrophilicity of the bile acid pool (Crosignani et al., 1991; U. Leuschner et al.,

24

1989; K. D. Lindor et al., 1994; Stiehl et al., 1990). Second, UDCA can stimulate hepatobiliary

secretion. Third, it can prevent bile acid-induced apoptosis of hepatocytes by inhibiting the

mitochondrial membrane permeability transition (MMPT), and possibly stimulating the survival

pathway (Paumgartner & Beuers, 2002). Lastly, UDCA has been shown to decrease HLA class I

display by hepatocytes and reduce inflammatory cytokine production (K. Lindor, 2007; K. D.

Lindor et al., 1994).

1.10.1.2 Response to UDCA

UDCA has an impact on liver biochemistry, thus biochemical response based on liver

biochemistry can be a useful tool for the prediction of long-term outcomes. There have been

several criteria developed on the basis of liver biochemistry to determine response to treatment.

The Barcelona criteria was defined as an ALP decrease greater than 40% of baseline values or

normal levels after 1 year of treatment. Patients who met these criteria were considered

responders and had a survival similar to that of the control population, but survival differed in

non-responders (Parés, Caballería, & Rodés, 2006).

Paris-I response was established as ALP ≤3×ULN, AST ≤2×ULN, and bilirubin ≤1mg/dL after 1

year. Patients that met these criteria had improved survival and a 1-year transplant-free survival

rate of 90%, as compared to 51% in non-responders. Paris-I criteria showed to be superior than

Barcelona criteria in discriminating patient survival in this cohort, as Barcelona responders had a

1-year survival of 79% and 53% for non-responders (Corpechot et al., 2008). An absence of

response according to Paris-I criteria was an independent predictor of death or liver

transplantation, independent of other baseline predictive factors such as bilirubin, histologic

stage, and interface hepatitis. Sex and age have been reported to be independent predictors for

response to UDCA according to Paris-I criteria, in which men and younger patients are less

likely to respond to treatment (Carbone et al., 2013).

The Rotterdam criteria for response was based on changes in bilirubin and albumin after

treatment with UDCA. Response was defined as normal bilirubin and albumin after 1 year of

treatment, given one of these parameters was abnormal or both were abnormal at study entry (E.

M. Kuiper et al., 2009).

25

The Toronto criteria was established based on the risk for progressive liver damage. Histological

progression by a one-stage increase during extended follow-up was associated with an absence in

response when defined as ALP>1.67×ULN after 2 years. Additionally, a two-stage increase was

associated with an absence in response when defined as ALP>1.76×ULN after 2 years (Kumagi

et al., 2010).

Distinct response criteria were established for patients with an early histological stage or when

early PBC was defined as normal albumin and bilirubin. The best criteria in these patients,

named Paris-II, was determined as ALP≤1.5×ULN, AST≤1.5×ULN, and normal bilirubin after 1

year of treatment (Corpechot, Chazouillères, & Poupon, 2011).

Although the majority of the criteria were initially established to be determined at 1 year, with

the exception of Toronto criteria, it has been suggested that response can be applied at 6 months.

In a Chinese population, response at 6 months showed the same or higher positive predictive

value, as well as similar negative predictive values when compared to response at 1 year (Zhang

et al., 2013). These findings are important because an earlier determination of response would

allow earlier identification of patients in need of additional therapies.

Table 1-1. Various response criteria to UDCA for PBC

Response Response criteria

Barcelona (Parés et al., 2006) ALP decrease > 40% baseline values or normal levels

after 1 year of treatment

Paris-I (Corpechot et al., 2008) ALP ≤3×ULN, AST≤2×ULN, and bilirubin ≤1mg/dL

after 1 year of treatment

Rotterdam (E. M. Kuiper et al.,

2009)

Normal bilirubin and albumin after 1 year of treatment,

given one of these parameters was abnormal or both

were abnormal at baseline

Toronto (Kumagi et al., 2010) ALP<1.67×ULN after 2 years of treatment

Paris-II (Corpechot et al., 2011) ALP≤1.5×ULN, AST≤1.5×ULN, and normal bilirubin

after 1 year of treatment

26

1.10.2 Adjuvant therapies

Although UDCA is effective at reducing liver biochemistry and delaying histological

progression, 30-40% of patients still do not respond to UDCA and are at risk for complications

of PBC. Therefore, there is still a need for adjuvant therapies to UDCA for the treatment of PBC.

1.10.2.1 Obeticholic acid

Farnesoid X receptor (FXR) is a nuclear hormone receptor that regulates the expression of genes

involved in bile acids homeostasis, of which the most potent endogenous FXR agonist is

chenodeoxycholic acid (Pellicciari et al., 2002). Obeticholic acid (OCA) is a semi-synthetic

analogue of chenodeoxycholic acid that selectively activates FXR, however, it exerts a 100×

greater potency in the activation of FXR compared to chenodeoxycholic acid (Pellicciari et al.,

2002). OCA gained FDA approval in 2016 and has been the first therapeutic agent to gain FDA

approval for the treatment of PBC since the introduction of UDCA. The results from the PBC

OCA International Study of Efficacy (POISE) phase III trial, a 12-month, double-blind trial,

were pivotal for its approval (Nevens et al., 2016). This study assessed the efficacy of OCA as

monotherapy or as adjuvant therapy to UDCA in 216 patients that had an inadequate response to

UDCA monotherapy. It included patients whose ALP was ≥1.67×ULN or whose bilirubin was

abnormal. Three groups, corresponding to placebo, 5-10mg, and 10mg were compared. The end-

point was a composite of ALP <1.67×ULN and normal bilirubin with an ALP reduction of at

least 15% from baseline. The end-point was reached at similar rates for the 5mg and 5-10mg

groups (46% and 47%) yet was only achieved in 10% of patients receiving placebo. One

common side effect of OCA is pruritus and appears to be dose-dependent (Hirschfield et al.,

2015; Nevens et al., 2016). The long-term survival benefit of OCA remains to be determined.

1.10.2.2 Fibrates

Fibrates represent another agent with therapeutic potential in PBC that act as ligands for the

nuclear receptor peroxisome proliferator-activated receptor (PPAR), which exists in three

isoforms: PPARα, PPARγ, and PPARβ/δ (Tyagi, Gupta, Saini, Kaushal, & Sharma, 2011).

Bezafibrate is a non-selective PPAR-agonist that has recently shown favorable results in the first

large, randomized controlled trial to assess the efficacy of bezafibrate as adjuvant therapy to

UDCA. Patients without response to UDCA according to Paris-II criteria were randomized to

27

receive bezafibrate at 400mg/day or placebo. Out of the patients whom received bezafibrate,

30% achieved normalization of ALP, bilirubin, aminotransferases, albumin, and PT after 2 years,

while 67% of patients achieved ALP normalization. Conversely, none of the patients in the

placebo group achieved normalization of any of the biochemical parameters. Furthermore, it was

shown to improve pruritus and prevent the progression of liver stiffness (Corpechot et al., 2017).

1.10.2.3 Immunosuppressive agents

Due to the presumed autoimmune nature of PBC, several immunosuppressants and

immunomodulatory agents have been evaluated for the treatment of PBC, most of which were

tested in addition to UDCA. Randomized controlled trials have been conducted for methotrexate

(Combes et al., 2005), cyclosporine (Wiesner et al., 1990), colchicine (Almasio et al., 2000),

azathioprine (Gong, Christensen, & Gluud, 2007), and corticosteroids (M. Leuschner et al.,

1996). However, these trials have not been successful, as either the therapeutic agents led to

major side effects or did not show efficacy at improving liver biochemistry, histology, or

survival. For example, the addition of methotrexate to patients already receiving UDCA for 48

weeks led to toxicity and was not associated with added benefit in terms of symptoms,

biochemistry, or histology (González-Koch, Brahm, Antezana, Smok, & Cumsille, 1997).

1.11 Predictors of Prognosis Since PBC is a slowly progressing disease, predictors of prognosis are essential to assess disease

progression and predict survival of patients, and thus have been extensively studied in the

literature. These can be used to determine the optimal time of referral for liver transplantation,

when a patient requires additional therapies, and as end-points in clinical trials (Mayo et al.,

2008). Liver biopsy was previously regarded as the only reliable means to assess prognosis in

all stages of disease. Indeed, an advanced histological stage has consistently been associated

with an increased risk for liver transplantation or death (Christensen et al., 1980; Floreani et al.,

2011; Mahl et al., 1994; Trivedi et al., 2014). However, there has been an increased emphasis on

non-invasive surrogate markers because they can be easily attained, performed frequently, and

do not pose the same risks as liver biopsies.

28

Bilirubin has been established as a main predictor of prognosis prior to the introduction of

UDCA. In untreated patients, a rapid rise in bilirubin was consistently observed prior to a

patient’s death (Shapiro, Smith, & Schaffner, 1979). The prognostic value of bilirubin is

maintained in treated patients and treated patients who had normalization of bilirubin had

improved survival (Bonnand, Heathcote, Lindor, & Poupon, 1999). Elevated bilirubin has been

consistently associated with an increased risk for liver transplantation or death (Christensen et

al., 1980; Lammers et al., 2014; R. E. Poupon et al., 1994; Trivedi et al., 2014). Out of multiple

thresholds, the optimal threshold of bilirubin for determining risk of liver transplantation and

death has been established to be 1×ULN (Lammers et al., 2014).

Another liver parameter associated with transplant-free survival is ALP, in which ALP <2×ULN

was found to correlate with transplant-free survival, independent of time. Furthermore, absolute

levels of ALP after 1 year predict transplant-free survival better than a percentage change. A

combination of ALP and bilirubin increases the predictive value of these biochemical parameters

(Lammers et al., 2014). Additional biochemical factors associated with improved prognosis have

been AAR and APRI. An AAR ≤2 is associated with improved prognosis, while APRI>0.54 is

associated with worse prognosis (Su et al., 2009; Trivedi et al., 2014).

Demographic factors such as age and sex are also associated with prognosis in PBC patients, in

which male sex and older age at diagnosis are independently associated with increased mortality

(Mahl et al., 1994; Myers et al., 2009). The impact of age on mortality, however, is different

when comparing that of PBC patients to a general population. In a study of asymptomatic

patients, although the mortality rate of elderly patients above 55 years old was higher than that of

younger patients, it was not different from an age- and gender-matched population (Kubota et al.,

2009). This was attributed to an excess of deaths due to other causes in the elderly, particularly

malignancies, while younger patients were more likely to die as a result of liver failure (Kubota

et al., 2009).

1.11.1 Prognostic models

Several prognostic models have been developed for PBC to predict survival. The Mayo model is

one of the earliest prognostic models developed to predict survival and was cross-validated in

various populations. It was derived from 312 untreated patients seen at the Mayo clinic from

29

1974 to 1984 and includes age, serum bilirubin, albumin, PT, and severity of edema to estimate

survival up to 7 years (Dickson, Grambsch, Fleming, Fisher, & Langworthy, 1989). It was

subsequently updated to predict short-term survival at 2 years at any time during disease from

variables measured at the latest patient visit (Murtaugh et al., 1994). Some more recent models

include the UK-PBC risk score and the GLOBE score. The UK-PBC research group derived a

prognostic model to predict 5-, 10-, and 15-year survival in 1916 UDCA-treated patients that was

then validated in 1249 patients, namely the UK-PBC risk score. This model includes albumin

and platelet count from baseline, and bilirubin, transaminases, and ALP after 1 year of UDCA

therapy (Carbone et al., 2016). The GLOBE score was developed in a globally representative

population of 4119 UDCA-treated patients to predict whether the transplant-free survival of

patients with PBC differs from that of a control population. The model includes age, bilirubin,

albumin, ALP, and platelet count collected at 1 year (equation 1). However, it can also be used

with values collected from 2-5 years. The GLOBE score can be used as response criteria when

comparing it to age-specific thresholds. If the GLOBE score surpasses the age-specific threshold,

the patient is considered a non-responder and their survival deviates from that of the general

population. The age groups <45, 45-52, 52-58, 58-66, and >66 years are linked with the age-

specific GLOBE score thresholds of -0.52, 0.01, 0.60, 1.01 and 1.69, respectively. Indeed, the

GLOBE score showed to be superior in the discrimination of patients at risk for liver

transplantation or death when compared to Barcelona, Paris-I, Rotterdam, Toronto, and Paris-II

criteria (Lammers et al., 2015). These GLOBE score was validated and proved to be accurate

prognostic predictors in a Chinese population of PBC patients (Yang et al., 2017).

Equation 1: GLOBE score calculation

0.044378 × age at the start of UDCA therapy + 0.93982 × LN(bilirubin [×ULN] at 1 year of

follow-up) + 0.335648 × LN(ALP [×ULN] at 1 year of follow-up) – 2.266708 × albumin

(×LLN) at 1 year of follow-up – 0.002581 × platelet count (×109/L) at 1 year of follow-up +

1.216865.

30

1.12 Liver Transplantation Liver transplantation is the sole treatment option in patients with end-stage PBC and grants

improved patient survival and quality of life (Hubscher et al., 1993; Markus et al., 1989). The

survival rates at 1, 5, and 10 years after liver transplantation from a center in the UK have been

reported to be 83%, 78%, and 67% (Liermann Garcia, Evangelista Garcia, McMaster, &

Neuberger, 2001). The disease severity of PBC prior to liver transplantation also seems to have

an impact on survival post-transplantation, as the survival of patients with an earlier disease

stage has been reported to be 88% at 5 years (W. R. Kim et al., 1998). The status of patients at

the time of transplantation seem to have improved over time as indicated by lower ALP and

bilirubin levels, and less advanced disease stage (E. M. M. Kuiper, Hansen, Metselaar, et al.,

2010; Liermann Garcia et al., 2001). Furthermore, liver transplantations have been conducted at

increasingly succeeding rates because there have been improvements in rejection rates and the

management of complications after liver transplantation, such as acute rejection and infections

(W. R. Kim et al., 1998).

These findings suggest that patients who undergo liver transplantation for PBC nowadays have a

good prognosis. However, the proportion and absolute number of patients who are transplanted

for PBC has reportedly decreased in the US and Europe, which indicates a reduced

transplantation burden for PBC (E. M. M. Kuiper, Hansen, Metselaar, et al., 2010; Lee et al.,

2007; Liermann Garcia et al., 2001). In a single center from the UK, the proportion of patients

grafted for PBC has decreased from 35% in 1990 to 21% in 1999 (Liermann Garcia et al., 2001).

In the US, the number of absolute liver transplantations for PBC has decreased an average of 5.4

cases per year from 1995 to 2006 despite an average increase of 249 transplants per year.

Meanwhile, the number of transplantation for PSC in the US did not change (Lee et al., 2007).

Indications for liver transplantation include liver failure, complications of cirrhosis, rising

bilirubin above 3-5mg/dL, model for end-stage liver disease (MELD) score above 15, and poor

quality of life due to intractable lethargy or pruritus (Hirschfield et al., 2017; Liermann Garcia et

al., 2001). There seems to be a trend towards considering transplantation in those without end-

stage liver disease but who experience intractable symptoms (Pells et al., 2013). Although liver

31

transplantation may be beneficial for pruritus, its impact on systemic symptoms such as fatigue

and cognitive impairment may be limited (Pells et al., 2013).

1.12.1 Recurrence of PBC after liver transplantation

There is evidence to indicate that recurrent PBC is possible, in which the diagnosis is made on

the basis of histological features consistent with a florid duct lesion (Hubscher et al., 1993;

Liermann Garcia et al., 2001; Sylvestre, Batts, Burgart, Poterucha, & Wiesner, 2003).

Histological evidence is required to diagnose recurrent PBC because AMA persists after liver

transplantation and liver biochemistry may be normal (Hirschfield et al., 2017). Recurrence rates

have been variable. In an early study, 8% of patients were diagnosed with recurrent PBC 2-6

years after liver transplantation despite treatment with an immunosuppression regimen that

included cyclosporine, prednisone, and azathioprine (Hubscher et al., 1993). In a UK study,

recurrent PBC was found in 17% of patients at a mean time of 36 months (Liermann Garcia et

al., 2001). The Mayo clinic reported the same recurrence rate of 17% in their population with a

mean time to recurrence of 3.7 years (Sylvestre et al., 2003). Furthermore, a study that included

patients from French and Swiss centers from 1988 to 2010 reported recurrence rates of 53% and

demonstrated that recurrent PBC can be progressive, as 15% of patients progressed to cirrhosis

(Bosch et al., 2015). Although these studies suggest that recurrence of PBC is not uncommon, its

diagnosis does not seem to have a significant impact on re-transplantation or survival (Bosch et

al., 2015; Charatcharoenwitthaya et al., 2007).

Pre- and post-transplantation factors that may be able to predict recurrence of PBC have been

evaluated, but the only risk factor reported has been the use of tracolimus, a calcineurin inhibitor

used in the immunosuppressive regimen after liver transplantation (Liermann Garcia et al.,

2001). Whereas, preventative UDCA has been associated with a decreased risk of recurrence

(Bosch et al., 2015).

1.13 Trends in PBC Although limited, there have been some studies on the temporal trends for PBC. One study from

Finland reported trends over a 12-year period and compared four 3-year periods (1988-1990,

1991-1993, 1994-1997, 1997-1999). They reported no difference in the proportion of male to

32

female patients or median age of the population. However, they suggested there was an

improvement in survival, and the hazard ratio (HR) for all-cause mortality per a 10-year increase

in year of diagnosis was 0.6 (Rautiainen et al., 2007). A Canadian study that evaluated patients

diagnosed from 1996 to 2002 did not find the year of diagnosis was a significant predictor for

mortality (Myers et al., 2009). In a Japanese population of symptomatic patients from 1999 to

2004, there was no change in the male to female ratio (9:1), yet they noted an increase in

median age from 59 years in 1999 to 63 years in 2004, and a decrease in bilirubin, GGT, total

cholesterol and IgM levels (Sakauchi et al., 2007). Interestingly, other autoimmune diseases

were more frequently observed in 2004. A study from Iceland reported a decrease in the

proportion of symptomatic patients at diagnosis from 57% in 1991-2000 to 36% 2001-2010 but

did not find a difference in the proportion of patients who present with an advanced histological

stage (III-IV), whom accounted for 28% (T. R. Baldursdottir et al., 2012).

33

Chapter 2

2 Aims and hypothesis

2.1 Study 1: Calendar Time Trends

2.1.1 Aims

The aim of the first study is to describe the temporal trends in patient and disease characteristics

at presentation with PBC as well as the treatment regimen and clinical outcomes of patients

diagnosed from 1970 to 2014 in a globally representative population. The following time cohorts

with respect to year of diagnosis will be compared: 1970-1979, 1980-1989, 1990-1999, 2000-

2009, and ≥2010. In order to determine whether the natural history of PBC has changed, we will

evaluate whether there have been changes in age at diagnosis, female: male ratio, AMA

serological status, histological stage, and biochemical stage. In addition, changes in treatment

regimen will be assessed in terms of use of UDCA, dosage, and time from diagnosis to the start

of treatment. Response to UDCA will also be assessed. Lastly, to determine whether clinical

outcomes have improved in recent decades, we will examine hepatic decompensation, HCC, and

transplant-free survival rates over the respective time cohorts. Overall, these specific research

questions will aid in determining whether the natural history of PBC has changed over the course

of multiple decades and suggest putative factors that may play a role.

2.1.2 Hypothesis

We hypothesize that the natural history of patients diagnosed in earlier decades is significantly

different from those presenting in more recent decades, consistent with a milder disease stage at

presentation. Furthermore, we hypothesize that recent decades are associated with an improved

treatment regimen and a decrease in clinical outcomes.

2.1.3 Rationale for hypothesis

There have been multiple studies reporting an increase in prevalence of PBC. Furthermore, the

proportion of asymptomatic patients has increased, and the number of liver transplantations

conducted for PBC over the years has decreased. These previous findings collectively suggest

that PBC is being readily detected at increased rates and more importantly at an earlier stage

before symptoms commence. These observations can be attributed to an increased awareness of

34

PBC by physicians and more sensitive and readily available AMA testing. The decrease in liver

transplantations indicates that there has been an improvement in the prognosis of patients with

PBC.

2.2 Study 2: Bilirubin Within the Normal Range

2.2.1 Aims

The aim of the second study is to determine whether bilirubin within the normal range is

predictive of transplant-free survival and if attaining bilirubin levels below the upper limit of

normal yields additional benefit for patients. Therefore, we will first evaluate whether bilirubin

quartiles are associated with distinct transplant-free survival rates. If so, our aim is to establish

the optimal threshold of bilirubin with the best performance in predicting liver transplantation or

death. To validate and confirm the predictive ability of the bilirubin threshold, it will be tested in

various subgroups and at multiple time points.

2.2.2 Hypothesis

We hypothesize that bilirubin levels below the upper limit of normal can be used to predict the

risk for liver transplantation or death in patients with PBC.

2.2.3 Rationale for hypothesis

Bilirubin is one of the strongest predictors for transplant-free survival. Although bilirubin is not

markedly elevated until later stages of disease, minimal changes in bilirubin may be associated

with prognosis. It seems that the majority of patients, even those who are included in clinical

trials to receive additional therapy, have normal bilirubin, as only 6-10% of patients included in

the OCA trial had abnormal bilirubin levels (Nevens et al., 2016). Furthermore, OCA was found

to further decrease their bilirubin even if within the normal range.

In a healthy population from the United States, the modal bilirubin value was 0.4mg/dL (Zucker,

Horn, & Sherman, 2004). Although the ULN for bilirubin varies by laboratory, most are

determined to be from 1.0-1.2mg/dL (Levitt & Levitt, 2014). Therefore, to reach the ULN, an

increase of approximately 0.6-0.8mg/dL in total bilirubin primarily driven by conjugated

35

bilirubin would be required. This suggests a large increase before abnormal bilirubin is detected

and thus the ULN may not be a sensitive way to determine absence in risk for a poor outcome.

36

Chapter 3 : Study 1

Milder Disease Stage in Patients with Primary Biliary 3Cholangitis Over a 44-Year Period: A Changing Natural History

The contents in this chapter have been published:

Murillo Perez, C. F., Goet, J. C., Lammers, W. J., Gulamhusein, A., van Buuren, H. R.,

Ponsioen, C. Y., …. Hansen, B. E. (2018). Milder disease stage in patients with primary biliary

cholangitis over a 44-year period: A changing natural history. Hepatology, 67(5), 1920–1930.

3.1 Introduction Primary biliary cholangitis (PBC) is a chronic autoimmune liver disease characterized by

inflammation and destruction of the small intralobular bile ducts (Kaplan & Gershwin, 2005; K.

D. Lindor et al., 2009; R. Poupon, 2010). The disease mainly affects middle-aged women and

has a slow, progressive course that may lead to fibrosis, cirrhosis, and liver failure requiring liver

transplantation. The standard treatment for PBC is ursodeoxycholic acid (UDCA) as its long-

term use improves liver biochemistry, delays histological progression, and may improve

transplant-free survival (Corpechot et al., 2002; R. E. Poupon et al., 1997; Pratt, 2016). However,

up to 40% of patients can have an inadequate response to UDCA that is associated with reduced

transplant-free survival (Corpechot et al., 2008; E. M. Kuiper et al., 2009; Parés et al., 2006;

Pratt, 2016).

PBC is a rare disease with multiple studies reporting an increase in its incidence and prevalence

in recent years (Boonstra et al., 2012, 2014; Eriksson & Lindgren, 1984; James et al., 1999;

Metcalf, Bhopal, Gray, Howel, & James, 1997; Myers et al., 2009; Myszor & James, 1990; Pla et

al., 2007; Remmel, Remmel, Uibo, & Salupere, 1995). In a systematic review conducted by

Boonstra et al. (2012) the incidence of PBC varied from 0.33 to 5.8 per 100,000/year, yet its

temporal trends are conflicting as some studies suggest an increase (Boonstra et al., 2014;

Metcalf et al., 1997) while others do not substantiate this finding (T. R. Baldursdottir et al., 2012;

McNally et al., 2014). The prevalence ranged from 1.91 to 40.2 per 100,000 and all investigated

studies reported an increase (Boonstra et al., 2012). An increase in prevalence impacts how PBC

contributes to the health care system and may be a result of multiple societal and disease factors.

37

It is important to note that initial reports of an increasing prevalence began during the off-label

use of UDCA period, which suggests that the increased prevalence in the UDCA-era may be due

to prolonged survival (James et al., 1999; Metcalf et al., 1997; Myszor & James, 1990).

Correspondingly, the absolute number of liver transplantations for PBC has decreased in Europe

and the United States since the introduction of UDCA in the early 1990s (Gross & Odin, 2008;

Hirschfield et al., 2015; E. M. Kuiper et al., 2009; Lee et al., 2007; K. D. Lindor et al., 2009).

In addition to epidemiological changes, the clinical presentation of PBC has also changed over

the years. Whereas most patients presented with an advanced histological stage in earlier

decades, nowadays most patients present during an asymptomatic stage (Christensen et al., 1980;

Locke et al., 1996). Therefore, the underlying assumption that PBC, as a disease, is a static entity

may not be accurate. We used a representative large cohort of patients with PBC to assess how

disease presentation and prognosis have changed over the last nearly 50 years. In doing so, we

provide long-term insight into the changing nature of PBC in clinical practice.

3.2 Patients and Methods

3.2.1 Population and study design

This was a retrospective study based on patient data retrieved from the Global PBC Study Group

database, of which characteristics have been described in previous publications (Lammers et al.,

2015, 2014). The database comprises long-term follow-up cohorts from 17 centers across North

America and Europe. UDCA-treated and non-treated patients aged ≥18 with an established PBC

diagnosis from 1970 to 2014, according to internationally accepted guidelines, were included in

the study (Hirschfield et al., 2017; K. D. Lindor et al., 2009; Nguyen, Juran, & Lazaridis, 2010).

Patients with either a short follow-up (<6 months), an unknown date of important clinical events,

an overlap syndrome, or another concomitant liver disease were excluded. Completeness and

accuracy of the database was established through visits to individual centers. This study was

conducted in accordance with the 1975 Declaration of Helsinki. The protocol was approved by

the institutional research board of the corresponding center and at all participating centers as per

local regulations.

38

3.2.2 Data collection

In the established database, study entry (baseline) was the date of UDCA therapy initiation or the

date of the first visit for non-treated patients. The following demographic and clinical data were

available at study entry: sex, date of birth, date of diagnosis, anti-mitochondrial antibody (AMA)

serological status, liver histology, biochemical disease stage, and UDCA therapy (if received and

dosage). In addition, the following laboratory values were available at study entry and every 6-12

months until the end of follow-up: alkaline phosphatase, aspartate aminotransferase, alanine

aminotransferase, total bilirubin, albumin, and platelet count. Histology was considered if the

liver biopsy was completed within 24 months of diagnosis date and dichotomized according to

Ludwig et al. (Ludwig et al., 1978) and Scheuer’s (Scheuer, 1967) classification; specifically, as

mild (stage I and II) and advanced (stage III and IV). The Rotterdam criteria were used to

determine patients’ biochemical stage. According to these criteria, mild stage is defined as

normal bilirubin and albumin, moderate stage is defined as abnormal bilirubin or albumin, and

advanced stage is defined as abnormal bilirubin and albumin (E. M. Kuiper et al., 2009; ter Borg

et al., 2006). Baseline aspartate aminotransferase/platelet ratio index, an independent predictor of

transplant-free survival, was calculated to stratify patients at risk of liver transplantation and

death based on a threshold of 0.54 (Trivedi et al., 2014). The first occurrence of hepatic

decompensation (ascites, variceal bleeding, or hepatic encephalopathy), hepatocellular

carcinoma (HCC), liver transplantation, or all-cause mortality were also retrieved.

In patients that received therapy, biochemical response to UDCA was determined according to

Barcelona (Parés et al., 2006), Paris-I (Corpechot et al., 2008), Rotterdam (E. M. Kuiper et al.,

2009), Toronto (Kumagi et al., 2010), and Paris-II criteria (Corpechot et al., 2011). In addition,

the GLOBE score was compared to age-specific thresholds to determine UDCA-response

(Lammers et al., 2015). Patients were considered responders if their GLOBE score did not

surpass their age-specific threshold.

3.2.3 Statistical analysis

Patients diagnosed between 1970 and 2014 were divided into five cohorts according to their year

of diagnosis: 1970-1979, 1980-1989, 1990-1999, 2000-2009, and ≥2010. To compare patient and

disease characteristics across the five cohorts, we conducted Chi-square tests for categorical

39

variables and analyses of variance for continuous data. A P-value less than 0.05 was considered

significant for all statistical analyses. Significant results were further analyzed to correct for any

possible confounding variables and to assess the influence of other explanatory variables on the

outcome measure. A multivariable logistic regression was applied to binary outcomes, such as

biochemical response to UDCA, biochemical disease stage (moderate and advanced disease

stage grouped as advanced), and histological stage (odds ratio [OR] with 95% confidence

interval [CI]).

For time-to-event analyses, patients diagnosed from 2010 onward were excluded due to a shorter

follow-up period than the other cohorts. Patients without an event and those who were lost to

follow-up were censored at their last visit. The rates of hepatic decompensation, HCC, and liver

transplant-free survival were assessed by Kaplan-Meier estimates and compared across decades

using the log-rank test. If decompensation occurred within the first year of study entry, the

patient was excluded from the time-to-event analysis for decompensation. Transplant-free

survival was compared across decades in the PBC population and within each decade to an age-

and gender-matched Dutch population. These outcomes were also estimated by Cox proportional

hazards’ modelling (hazards ratio [HR] with 95% CI).

Demographic and clinical characteristics are presented as count (percentage) for categorical data

and mean ± standard deviation (SD) for continuous variables. Laboratory values are presented as

median (interquartile range [IQR]). Data that were not normally distributed were log transformed

for the analyses. All analyses were two-sided and were performed using IBM SPSS Statistics for

Windows, version 24.0 (IBM Corp., Armonk, NY).

3.3 Results

3.3.1 Study population characteristics

A total of 4805 PBC patients, diagnosed between 1970 and 2014, were included and divided into

five cohorts according to their year of diagnosis (Table 3-1, Table S3-1). 143 patients were

diagnosed from 1970 to 1979, 858 patients from 1980 to 1989, 1754 patients from 1990 to 1999,

1815 patients from 2000 to 2009, and 235 patients from 2010 onward. The characteristics of

each cohort are presented in Table 3-1. The median follow-up for the five respective cohorts

40

were: 6.7 years (IQR 3.0-14.3), 8.9 years (IQR 4.0-14.7), 10.0 years (IQR 6.0-13.9), 5.6 years

(IQR 3.4-8.3), and 1.6 years (IQR 1.0-2.1). The mean time from diagnosis to study entry was

variable for each cohort: 11.1 years (SD 7.0) for the 1970s, 5.1 years (SD 4.5) for the 1980s, 1.4

years (SD 2.3) for the 1990s, 0.4 years (SD 1.1) for the 2000s, and 0.1 years (SD 0.2) from 2010

onward. To consider this variation, all analyses were repeated in a sub-group of patients

(n=3518) with a maximum two-year lag between diagnosis and study entry, which included

14%, 29%, 76%, 93%, and 100% of patients from the main analysis in each respective cohort

(Table S3-2).

41

Table 3-1. Demographic and clinical characteristics of PBC patients at study entry over calendar time

Baseline characteristics 1970-1979 (n=143)

1980-1989 (n=858)

1990-1999 (n=1754)

2000-2009 (n=1815)

≥2010 (n=235)

P-value

Age at diagnosis, ya 46.9 (10.1) 50.1 (10.7) 52.8 (11.5) 55.0 (12.5) 57.0 (12.1) <0.001

Female 131 (91.6) 775 (90.3) 1593 (90.8) 1619 (89.2) 207 (88.1) 0.396

AMA-positiveᵇ

123/140 (87.9) 763/842 (90.6)

1565/1704

(91.8)

1599/1765

(90.6) 213/235 (90.6)

0.449

Laboratory valuesc

Serum ALP (×ULN) 2.99 (1.85-4.77) 3.20 (1.95-5.23) 2.03 (1.30-3.56) 1.79 (1.19-3.05) 1.55 (1.08-2.93) <0.001

Serum bilirubin (×ULN) 0.93 (0.60-2.1) 0.81 (0.52-1.30) 0.64 (0.47-1.00) 0.60 (0.41-0.95) 0.59 (0.41-1.0) <0.001

Serum AST (×ULN) 1.59 (1.06-2.32) 1.95 (1.20-2.77) 1.35 (0.87-2.20) 1.30 (0.90-2.00) 1.29 (0.85-2.07) <0.001

Serum ALT (×ULN) 1.30 (0.85-2.47) 2.00 (1.3-3.1) 1.66 (1.03-2.68) 1.42(0.90-2.27) 1.32 (0.75-2.38) <0.001

Serum albumin (×LLN) 1.11 (0.99-1.21) 1.16 (1.06-1.26) 1.14 (1.06-1.23) 1.14 (1.06-1.23) 1.14 (1.03-1.23) 0.005

Platelet count (×109/L) 194 (127-242.5) 224 (165-275) 238 (185-289) 258 (204-311) 237 (174.5-291) <0.001

APRI (>0.54)d 61 (76.3) 260 (69.0) 456 (52.3) 476 (47.4) 85 (54.1) <0.001

Biochemical disease stagee 121/143 627/859 985/1755 1073/1816 152/235 <0.001

Mild 50/121 (41.3) 370/627 (59.0) 711/985 (72.2) 757/1073 (70.5) 106/152 (69.7)

Moderately advanced 51/121 (42.1) 196/627 (31.3) 205/985 (20.8) 238/1073 (22.2) 27/152 (17.8)

Advanced 20/121 (16.5) 61/627 (9.7) 69/985 (7.0) 78/1073 (7.3) 19/152 (12.5)

Histological disease stagef 326/1001 948/1754 943/2050 <0.001

Mild (I or II) 197 (60.4) 634 (66.9) 721 (76.5)

42

Baseline characteristics 1970-1979 (n=143)

1980-1989 (n=858)

1990-1999 (n=1754)

2000-2009 (n=1815)

≥2010 (n=235)

P-value

Advanced (III or IV) 129 (39.6) 314 (33.1) 222 (23.5)

UDCA-treatedg 78/139 (56.1) 735/832 (88.3) 1605/1737

(92.4)

1563/1789

(87.4)

195/230 (84.8) <0.001

Data represented as mean (standard deviation), n (%), or median (interquartile range). Primary biliary cholangitis, PBC; AMA, antimitochondrial antibody; ALP, alkaline phosphatase; AST, aspartate aminotransferase; ULN, upper limit of normal; ALT, alanine aminotransferase; LLN, lower limit of normal; APRI, AST to platelet ratio index; UDCA, ursodeoxycholic acid. aAge at diagnosis not available for one patient in 2000-2009 cohort. bAMA status was available for 4686 (97.5%) patients. cALP, bilirubin, AST, and ALT were log transformed prior to analyses and availability for laboratory values is as follows: ALP: 3560 (74.1%); Bilirubin: 3595 (74.8%); AST: 3460 (72.0%); ALT: 3007 (62.6%); Albumin: 3039 (63.2%); Platelet count: 2769 (57.6%) dThe cut-point APRI >0.54 at baseline is predictive of liver transplantation or death (Trivedi et al., 2014) eBiochemical disease stage classification according to Rotterdam criteria (ter Borg et al., 2006) was available in 2958 (61.6%) patients. fHistological disease stage at diagnosis according to Ludwig et al. and Scheuer (Ludwig et al., 1978; Scheuer, 1967) classification was available in 2217 (46.1%) patients. gUDCA therapy status was available for 4727 patients (98.4%).

3.3.2 Age and sex trends

The mean age at diagnosis increased incrementally from 46.9 ± 10.1 years in the 1970s to 57.0 ±

12.1 years from 2010 onward (P < 0.001, Figure 3-1A). This trend was consistent across center,

sex, and biochemical disease stage (Figure S3-1A-C). The effect of calendar time on the

increase in age at diagnosis remained significant (P < 0.001) after correcting for sex (Table S3-

3). Furthermore, the age distribution of patients notably changed over the investigated decades (P

< 0.001, Figure 3-1B). The proportion of patients aged 50-59 years at diagnosis remained

relatively stable across the years, whereas the proportion of patients <50 years of age decreased

and patients ≥60 years of age increased. There was no significant temporal trend in the female to

male ratio, which remained approximately 9:1 (Table 3-1).

Figure 3-1. Age at diagnosis of PBC patients across different decades. A) Mean age (± standard

deviation) at diagnosis (dots) and estimated marginal means (squares) obtained after adjusting

for sex. B) The distribution of age groups over calendar time.

Year of diagnosis

Age

at d

iagn

osis

(yea

rs)

1970-1979 1980-1989 1990-1999 2000-2009 ≥ 201020

30

40

50

60

70

Raw dataEstimated marginal means

P < 0.001

Perc

enta

ge o

f pat

ient

s

1970-19791980-19891990-19992000-2009 ≥ 20100

20

40

60

80

100

<3030-3940-4950-5960-69≥70

Year of diagnosis

Age at diagnosis (years)

A) B)

43

44

3.3.3 Liver biochemistry and serological status

The proportion of patients that were AMA-positive did not significantly differ across the

investigated decades (Table 3-1). Median alkaline phosphatase and bilirubin values (× upper

limit of normal) at study entry decreased, while circulating platelet counts were noted to increase

(P < 0.001), which collectively suggests a less advanced disease stage. The proportion of patients

with alkaline phosphatase values below 2 × the upper limit of normal increased gradually from

30.0% in the 1970s to 63.1% from 2010 onward (P < 0.001) (Figure 3-2A). The proportion of

patients with normal serum bilirubin concentrations also increased from 51.1% in the 1970s to

77.6% in the 1990s, after which it remained relatively stable (P < 0.001) (Figure 3-2B).

Furthermore, a reduced percentage of patients with aspartate aminotransferase/platelet ratio

index >0.54 at study entry was observed (Table 3-1).

45

Figure 3-2. Study entry characteristics associated with disease severity of patients diagnosed in

different decades. A) Percentage of patients with alkaline phosphatase (ALP) above or below 2

times the upper limit of normal (×ULN). B) Percentage of patients with bilirubin above or below

1×ULN. C) Percentage of patients corresponding to each biochemical stage according to

Rotterdam criteria (ter Borg et al., 2006); mild (normal albumin and bilirubin), moderate

(abnormal albumin or bilirubin), advanced (abnormal albumin and bilirubin). D) Percentage of

patients corresponding to each histological stage at diagnosis according to Ludwig et al.’s (1978)

and Scheuer’s (1967) classification: mild (stage I and II) or advanced (stage III and IV).

Perc

enta

ge o

f pat

ient

s

1970-19791980-19891990-19992000-2009 ≥ 20100

20

40

60

80

100 Mild biochemical stageModerate biochemical stageAdvanced biochemical stage

Year of diagnosis

Year of diagnosis

Per

cent

age

of p

atie

nts

1970-19791980-19891990-19992000-2009 ≥ 20100

20

40

60

80

100 bilirubin≤1XULNbilirubin>1XULN

Perc

enta

ge o

f pat

ient

s

1970-19791980-19891990-19992000-2009 ≥ 20100

20

40

60

80

100

Year of diagnosis

ALP≤2xULNALP>2xULN

Perc

enta

ge o

f pat

ient

s

1970-1989 1990-1999 2000-20140

20

40

60

80

100 Mild histological stage (I or II)Advanced histological stage (III or IV)

Year of diagnosis

A) B)

C) D)

P < 0.001 P < 0.001

P < 0.001 P < 0.001

46

3.3.4 Trends in biochemical and histological disease stage

There was a gradual increase in the proportion of patients presenting with a mild biochemical

disease stage from the 1970s to 1990s, and remained stable thereafter (P < 0.001, Figure 3-2C).

In a multivariable logistic regression, calendar time was a significant predictor for biochemical

disease stage (P < 0.001) after adjusting for sex and age at diagnosis. Earlier decades were

associated with an advanced biochemical disease stage.

Out of 2831 patients who underwent liver biopsy at diagnosis, 2217 patients had histological

disease stage available and were included in a subgroup analysis that combined cohorts due to

the limited number of biopsies in the first and last cohorts. There were 326 biopsies from 1970-

1989, 948 biopsies from 1990-1999, and 943 from 2000-2014. The proportion of patients with a

mild histological stage (I or II) at diagnosis increased with time (Table 3-1, Figure 3-2D). In a

multivariable logistic regression, calendar time was a significant predictor for histological stage

after adjusting for sex and age at diagnosis (P < 0.001).

3.3.5 Trends in UDCA-response rates

The proportion of patients that ever received UDCA increased across the investigated decades (P

< 0.001, Table 3-1). In patients that received UDCA, the median number of years between

diagnosis and the start of UDCA therapy decreased across the respective cohorts (1970s to

≥2010): 12.6 years (IQR 10.6-16.1), 4.4 years (IQR 2.1-8.1), 0.23 years (IQR 0.0-2.0), 0.05

years (IQR 0.0-0.41), and 0.0 years (IQR 0.0-0.04). Additionally, the median initial dosage of

UDCA received by patients across the five respective cohorts increased: 9.4 mg/kg/day (IQR

8.5-11.0), 10.0 mg/kg/day (IQR 8.7-13.7), 12.2 mg/kg/day (IQR 9.2-14.7), 13.5 mg/kg/day (IQR

11.1-15.3), 13.3 mg/kg/day (IQR 11.1-15.1).

The proportion of UDCA-responders according to Paris-I, Toronto, Paris-II, Rotterdam, and

GLOBE score criteria increased over the investigated decades (P < 0.001), but not according to

Barcelona criteria (Figure 3-3, Table S3-4). Importantly, this trend remained true in patients

who did not meet the individual criteria at baseline (Table S3-5). In a multivariable logistic

regression, calendar time was not a significant predictor for UDCA-response according to Paris-I

criteria (Table 3-2). Response was associated with an increased age at diagnosis, and lower

alkaline phosphatase and bilirubin levels (P < 0.001). Additionally, calendar time was also not a

47

significant predictor for UDCA-response according to Toronto, Paris-II, Rotterdam, and GLOBE

score criteria.

Figure 3-3. Response rates to ursodeoxycholic acid (UDCA) therapy over calendar time.

Response was determined according to various published criteria: Barcelona, Paris-I, Rotterdam,

Toronto, Paris-II, and the GLOBE score (Corpechot et al., 2008, 2011; E. M. Kuiper et al., 2009;

Kumagi et al., 2010; Lammers et al., 2015; Parés et al., 2006). Response rates according to all

criteria were significantly different over calendar time (P < 0.001), except Barcelona criteria (P =

0.19).

Year of diagnosis

Perc

enta

ge o

f pat

ient

s

1970-19791980-19891990-19992000-2009 ≥20100

20

40

60

80

100

Paris-I

Paris-II

Rotterdam

Barcelona

GLOBE score

Toronto

Table 3-2. Multivariable logistic regression for the attainment of biochemical response

according to Paris-Ia (n=2283)

Variable OR 95% CI P-value

Male sex 0.90 0.63-1.29 0.58

Year of diagnosis 0.67

1970-1979 1.00

1980-1989 0.80 0.37-1.71 0.66

1990-1999 1.01 0.44-2.37 0.96

2000-2009 0.97 0.40-2.32 0.94

≥2010 0.92 0.33-2.57 0.88

Age at diagnosis 0.04

<30 1.00

30-39 1.29 0.53-3.15 0.57

40-49 1.41 0.60-3.33 0.44

50-59 1.95 0.82-4.59 0.13

60-69 2.06 0.86-4.96 0.11

≥70 2.06 0.82-5.21 0.13

Log bilirubin (×ULN) 0.01 0.01-0.02 <0.001

Log ALP (×ULN) 0.12 0.08-0.18 <0.001

Difference between diagnosis and study entry (years) 0.98 0.94-1.03 0.44

OR, odds ratio; CI, confidence interval; ULN, upper limit of normal; ALP, alkaline phosphatase. aResponse rate according to Paris-I is defined as: ALP ≤3 ×ULN, AST ≤2 ×ULN, and normal bilirubin after 1 year of UDCA therapy.

48

49

3.3.6 Decompensation, HCC, and transplant-free survival

The 10-year incidence rate of hepatic decompensation (ascites, variceal bleeding, or hepatic

encephalopathy, whichever came first) decreased over time: 18.5% in the 1970s, 13.7% in the

1980s, 8.5% in the 1990s, and 5.8% in the 2000s (Figure 3-4Ai). All pairwise comparisons were

significantly different, except the difference between the 1970s and 1980s cohorts (P = 0.45). In

a multivariable Cox regression, a temporal trend of lower decompensation risk was observed

after adjusting for sex and age at diagnosis (Figure 3-4Bi, Table S3-6, P = 0.07). Calendar time

as a continuous variable was a significant predictor for hepatic decompensation (HR per 10-year

increase: 0.57, 95% CI 0.44-0.75, P < 0.001).

The 10-year HCC incidence rates across the investigated decades were: 10.3%, 4.0%, 2.1%, and

2.3%, respectively (Figure 3-4Aii). The Kaplan-Meier estimate of cumulative HCC incidence

was significantly higher in the 1970s compared to the 1980s (P = 0.01), 1990s (P < 0.001), and

2000s (P < 0.001). In a multivariable Cox regression, calendar time was not a significant

predictor for HCC risk (P = 0.68) after adjusting for sex, age at diagnosis, and UDCA treatment

(Figure 3-4Bii, Table S3-7).

The 10-year liver-related death rate decreased from 1970-2009: 34.6%, 13.2%, 5.6%, and 6.4%

(P < 0.001). Furthermore, the 10-year transplant-free survival rate improved over the four

respective investigated decades: 48.4%, 68.7%, 79.7%, and 80.1% (Figure 3-4Aiii). There was a

significant difference in transplant-free survival between the 1970s and 1980s (P < 0.001), and

between the 1980s and 1990s (P < 0.001). However, the transplant-free survival rates between

the 1990s and 2000s were equivalent (P = 0.80). In a multivariable Cox regression, calendar time

remained an independent predictor of transplant-free survival, and earlier decades were

associated with an increased risk for liver transplantation and all-cause mortality (Figure 3-4Biii,

Table S3-8). Furthermore, the 10-year transplant-free survival of PBC patients has improved

even when compared to an age- and gender-matched general population (1970s: HR 4.38, 95%

CI 3.54-5.43, P < 0.001; 1980s: HR 2.90, 95% CI 2.60-3.24, P < 0.001; 1990s: HR 2.14, 95% CI

1.94-2.36, P < 0.001; 2000s: HR 1.93, 95% CI 1.69-2.21, P < 0.001).

50

Figure 3-4. Time-to-event analyses of decompensation, hepatocellular carcinoma (HCC), and

liver transplantation or death over calendar time. A) Kaplan-Meier (crude) and B) Multivariable

Cox regression (adjusted) estimates of i) cumulative incidence of decompensation, ii) cumulative

incidence of hepatocellular carcinoma (HCC), and iii) transplant-free survival.

Follow-up (years)

1086420

Cum

ulat

ive

inci

denc

e of

deco

mpe

nsat

ion

(%)

20

15

10

5

0

1970-1979-censored

2000-20091990-20001980-19891970-1979

Follow-up (years)1086420

Cum

ulat

ive

HC

C

inci

denc

e (%

)

15

10

5

0

1970-1979-censored

2000-20091990-20001980-19891970-1979

Follow-up (years)1086420

Tran

spla

nt-fr

ee

surv

ival

(%)

100

80

60

40

20

0

1970-1979-censored

2000-20091990-20001980-19891970-1979

Follow-up (years)1086420

Cum

ulat

ive

inci

denc

e of

de

com

pens

atio

n (%

)

20

15

10

5

0

Crude AdjustedA) B)i)

ii)

iii)

i)

No. at risk1970-19791980-19891990-19992000-2009

No. at risk1970-19791980-19891990-19992000-2009

No. at risk1970-19791980-19891990-19992000-2009

50492

1039810

46434936559

40367784348

34298637163

57574

11401046

59622

12021127

139790

15201666

112697

14101471

93574

12511121

74496

1110742

61420954451

49336788194

143858

17541815

117763

16411611

99641

14661242

78557

1315829

63472

1101502

51379876224

P < 0.001

P < 0.001

P < 0.001

P = 0.07

Follow-up (years)1086420

Tran

spla

nt-fr

ee

su

rviv

al (%

)

100

80

60

40

20

0

P < 0.001

iii)

Follow-up (years)1086420

Cum

ulat

ive

HC

C

inci

denc

e (%

)

5

4

3

2

1

0

ii)P = 0.68

51

3.4 Discussion In this study of a large, internationally representative cohort of PBC patients, we demonstrate

that patients diagnosed in recent decades are older and have a milder disease stage compared to

patients diagnosed in earlier decades. In addition, more patients respond favourably to UDCA

therapy and have improved transplant-free survival. To the best of our knowledge no previous

study has reported on these PBC trends. These results provide unique insight into the possible

changing natural history of PBC over the last five decades. It is noteworthy to mention that

similar results have been observed in a study from Sweden that included 246 patients diagnosed

with primary sclerosing cholangitis between 1984 and 2004. Bergquist et al. reported an increase

in age at diagnosis and lower frequency of symptoms in patients diagnosed after 1998

(Bergquist, Said, & Broomé, 2007).

Although some of the observed trends could be potentially attributed to more sensitive AMA

tests that detect the disease at an earlier stage, we speculate that any changes in AMA testing

have not had a major impact in the observed temporal trends. The conventional method of AMA

detection is indirect immunofluorescence, yet there has been an increase in ELISA-based assays

and immunoblotting that have led to greater sensitivity and specificity (Oertelt et al., 2007).

These improvements would translate to an increase in the proportion of AMA-positive patients,

however this has remained unchanged.

We demonstrate a 10-year increase in the mean age at diagnosis from 1970 to 2014. A similar

increase has been reported previously in the Canadian PBC population, in which prevalent cases

in 1996 had a median age of 53, whereas prevalent cases in 2002 had a median age of 57 (Myers

et al., 2009). These numbers coincide with the findings from our study, in which the mean age at

diagnosis in the 1990s and 2000s is 52.8 and 55.0 years, respectively. Furthermore, an increased

proportion of patients diagnosed in recent years are over 50 years of age and account for 71.5%

of patients diagnosed on 2010 and beyond. Comparable results were found within the UK-PBC

cohort, in which 75% of patients prevalent between 2008 and 2010 were over 50 years of age

(Carbone et al., 2013).

The increase in age may be attributed to the general aging of the population, as the median age in

Northern America and Europe has reportedly increased from 30 in 1970 to 40 in 2015 (United

52

Nations, Department of Economic and Social Affairs, 2017). This represents a 10-year increase

over a 45-year period, which is similar to the 10-year increase in age at diagnosis we observe

over a 44-year interval. Furthermore, the 34% absolute increase of PBC patients 50 years old and

above from 1970 to 2014 was greater than that of the general population, which was only 11%

(25% in 1970 to 36% in 2015) (United Nations, Department of Economic and Social Affairs,

2017). The increase in age may also be attributed to differences in the trigger for a PBC

diagnosis over the years. Although we are not able to assess the symptoms in our cohort, we

speculate that patients in recent decades are predominantly asymptomatic and are therefore

diagnosed when they see their physician to undergo routine testing of liver function, which

occurs more frequently in older individuals. Conversely, younger patients in earlier decades were

more likely to develop symptoms, which led to their diagnoses (Mahl et al., 1994; M. Prince et

al., 2002). Lastly, the increase in age may be disease-specific and represent a shift in the natural

history of PBC towards a new older at-risk population, considering the increase in age was

observed irrespective of biochemical disease stage. It can also be speculated that the later onset is

a result of a prolonged subclinical disease period and potentially a delayed exposure to an

unknown environmental trigger due to temporal changes in lifestyle.

An older age at diagnosis is clinically important because it has been associated with an increased

likelihood of meeting Paris-I criteria for response to UDCA (Carbone et al., 2013). Similarly, we

found an older age at diagnosis to be an independent predictor of Paris-I response, yet calendar

time was not a significant predictor. These results indicate the increase in age at diagnosis may

be an important factor contributing to the increase in UDCA-response rather than calendar time

itself. Furthermore, the low response rates observed in earlier decades can be a result of

inadequate UDCA dosages and the delay in treatment. The importance of an adequate UDCA

dosage of 13-15mg/kg per day has been emphasized in a study that found 40% of UDCA-non-

responders in whom the dosage was increased became responders (Angulo, Dickson, et al., 1999;

Lammers et al., 2016).

In recent decades, patients present at an older age, yet they have milder biochemical and

histological disease stage. Improved disease severity might be explained by an earlier detection

of PBC due to improved disease awareness leading to liver function tests and AMA assays

(European Association for the Study of the Liver, 2009; M. I. Prince et al., 2004). The

53

histological disease stage at diagnosis has important prognostic implications for UDCA-response

and survival. Advanced histological stages are associated with an increased risk of treatment

failure (Corpechot et al., 2008). In addition, the survival of UDCA-treated patients in stage I/II is

similar to that of an age- and sex-matched control population, while the probability of liver

transplantation or death is significantly increased in patients with advanced histological stages

(Corpechot et al., 2005).

Although a decrease in the number of liver transplantations for PBC has been reported over the

years (Lee et al., 2007), an improvement in transplant-free survival has not been previously

documented. In a Canadian population-based study of patients diagnosed between 1996 and

2002, Myers et al. did not observe a significant difference in survival according to year of

diagnosis (Myers et al., 2009). The lack of difference in survival may be attributed to the small

interval of study, which only spanned six years. The reported increase in median age of the

general population well reflects an increase in life expectancy over time (United Nations,

Department of Economic and Social Affairs, 2017); therefore transplant-free survival was

compared to that of the general population. Our study showed that transplant-free survival

improved over a 44-year period, even when compared to the general population, and supports its

potential role in the increased prevalence of PBC.

The inclusion of a large population of PBC patients from different geographical regions, long-

term follow-up, and broad study period are some of the strengths of our study. However, some

limitations need to be considered. First, the 1970s and 1980s cohorts were susceptible to a delay

in documentation since study entry can be many years after the date of diagnosis in these

cohorts. As such, the difference in years between these two dates was included in all

multivariable analyses and we assessed a sub-group of patients with a maximum two-year

difference. The same trends emerged in the sub-group analyses, thus excluding the possibility

that the delay in documentation is the reason for an advanced disease in the early cohorts.

Second, due to the retrospective nature of the study, biochemical data was not available for all

patients and thus response to UDCA could not be determined for all patients. To account for

missing laboratory values, all analyses were repeated in an imputed dataset and revealed similar

results. Lastly, the trends observed in our study cohort could not be assessed for correlations with

symptom profiles or various environmental factors previously associated with PBC, such as

54

smoking, age at first pregnancy, or the use of hormonal replacement therapy (Gershwin et al.,

2005). Even though the trends observed may be due to a selection of patients whose diagnosis is

triggered by symptoms or complications in earlier decades rather than routine liver function tests

as in recent decades, we describe the presenting characteristics of a typical PBC patient seen by

physicians and how they have changed over time. The observed temporal trends warrant further

investigation in other PBC populations to determine whether they are universally applicable and

to explore the potential influence of a changing environmental trigger.

In conclusion, we demonstrate a 10-year increase in age at diagnosis accompanied by milder

disease severity at presentation of PBC patients. These findings provide the most comprehensive

evidence of a changing natural history of PBC to date.

3.5 Supplementary Tables and Figures Table S3-1. Distribution of PBC patients across calendar time and center

Center 1970-1979 (n=143)

1980-1989 (n=858)

1990-1999 (n=1754)

2000-2009 (n=1815)

≥2010 (n=235)

Total N=4805

North Europe

Rotterdam, The Netherlands

(1973-2012)a 25 (17.5) 122 (14.2) 274 (15.6) 361 (19.9) 37 (15.7) 819

Leuven, Belgium (1974-2011)b 5 (3.5) 20 (2.3) 44 (2.5) 64 (3.5) 13 (5.5) 146

Ghent, Belgium (1991-2014)c 0 0 4 (0.2) 14 (0.8) 6 (2.6) 24

Paris, France (1974-2001)b 11 (7.7) 209 (24.4) 113 (6.4) 14 (0.8) 0 347

London, UK (1972-2007)b 11 (7.7) 31 (3.6) 68 (3.9) 26 (1.4) 0 136

Birmingham, UK (1972-2011)b 1 (0.7) 4 (0.5) 79 (4.5) 264 (14.5) 14 (6.0) 362

Jena, Germany (1979-2013)c 1 (0.7) 5 (0.6) 38 (2.2) 53 (2.9) 24 (10.2) 121

South Europe

Milan, Italy (1970-2005)b,d 71 (49.7) 217 (25.3) 183 (10.4) 62 (3.4) 0 533

Padua, Italy (1972-2012)b 3 (2.1) 38 (4.4) 102 (5.8) 99 (5.5) 28 (11.9) 270

Barcelona, Spain (1971-2005)b 3 (2.1) 51 (5.9) 147 (8.4) 68 (3.7) 0 269

Larissa, Greece (1991-2014)c 0 0 1 (0.1) 76 (4.2) 23 (9.8) 100

North America

Rochester, USA (1970-2012)b 2 (1.4) 11 (1.3) 245 (14) 352 (19.4) 69 (29.4) 679

55

56

Center 1970-1979 (n=143)

1980-1989 (n=858)

1990-1999 (n=1754)

2000-2009 (n=1815)

≥2010 (n=235)

Total N=4805

Toronto, Canada (1974-2010)b 9 (6.3) 87 (10.1) 229 (13.1) 257 (14.2) 1 (0.4) 583

Texas, USA (1977-2011)b 1 (0.7) 62 (7.2) 209 (11.9) 44 (2.4) 10 (4.3) 326

Edmonton, Canada (1989-2007)b 0 1 (0.1) 13 (0.7) 42 (2.3) 0 56

Seattle, USA (1995-2012)b 0 0 5 (0.3) 19 (1) 10 (4.3) 34

Data represented as n (% within corresponding decade). ᵃComprised of centers across the Netherlands (mainly secondary centers). bTertiary center. cSecondary center. dComprised of two centers.

57

Table S3-2. Calendar time trends in patients with a maximum lag of 2 years between diagnosis and study entry

Characteristics 1970-1979 (n=20)

1980-1989 (n=245)

1990-1999 (n=1331)

2000-2009 (n=1687)

≥2010 (n=235)

P-value

Age at diagnosis, ya 49.3 (12.9) 52.3 (11.7) 52.9 (11.6) 55.0 (12.6) 57.0 (12.1) ˂0.001

Female 18 (90) 220 (89.8) 1210 (90.9) 1509 (89.4) 207 (88.1) 0.60

AMA-positiveb 16 (84.2) 217 (90.0) 1190 (91.8) 1487 (90.7) 213 (90.6) 0.63

Laboratory valuesc

Serum ALP (×ULN) 3.05 (1.15-7.32) 3.76 (2.04-6.50) 2.14 (1.33-3.69) 1.83 (1.21-3.08) 1.55 (1.08-2.93) ˂0.001

Serum bilirubin (×ULN) 1.3 (0.59-4.56) 0.74 (0.47-1.27) 0.62 (0.47-1.00) 0.60 (0.41-0.97) 0.59 (0.41-1.00) 0.001

Serum AST (×ULN) 1.47 (0.91-1.80) 1.8 (1.13-2.6) 1.43 (0.94-2.27) 1.32 (0.92-2.03) 1.29 (0.85-2.07) ˂0.001

Serum ALT (×ULN) 0.98 (0.53-1.64) 1.95 (1.19-3.00) 1.71 (1.06-2.75) 1.46 (0.91-2.33) 1.32 (0.75-2.38) ˂0.001

Serum albumin (×LLN) 1.04 (0.92-1.15) 1.11 (1.03-1.25) 1.14 (1.06-1.23) 1.14 (1.06-1.23) 1.14 (1.03-1.23) 0.038

Platelet count (×109/L) 203 (187-244) 256 (194-305) 242 (190-295) 257 (204-310) 237 (175-291) 0.001

APRI (>0.54)d 10 (71.4) 56 (60.2) 359 (54.9) 454 (47.8) 85 (54.1) 0.009

Biochemical disease stagee ˂0.001

Mild 5 (27.8) 100 (59.2) 515 (72.4) 707 (70.2) 106 (69.7)

Moderately advanced 7 (38.9) 52 (30.8) 145 (20.4) 228 (22.6) 27 (17.8)

Advanced 6 (33.3) 17 (10.1) 51 (7.2) 72 (7.1) 19 (12.5)

UDCA-treatedg 0 172 (76.1) 1208 (91.8) 1447 (87.1) 195 (84.8) ˂0.001

UDCA dosage (mg/kg/day)h - 11.7 (3.9) 11.9 (3.5) 13.3 (3.3) 13.1 (3.1) ˂0.001

Response to UDCAi

58

Characteristics 1970-1979 (n=20)

1980-1989 (n=245)

1990-1999 (n=1331)

2000-2009 (n=1687)

≥2010 (n=235)

P-value

Toronto - 43/83 (51.8) 427/616 (69.3) 523/710 (73.7) 48/65 (73.8) ˂0.001

Paris-I - 61/113 (54.0) 610/837 (72.9) 725/977 (74.2) 107/149 (71.8) ˂0.001

Barcelona - 73/113 (64.6) 518/795 (65.2) 633/1058 (59.8) 100/155 (64.5) 0.12

Paris-II - 36/116 (31.0) 432/861 (50.2) 522/1038 (50.3) 81/155 (52.3) ˂0.001

Rotterdam - 87/106 (82.1) 424/516 (82.2) 561/668 (84.0) 90/111 (81.1) 0.79

GLOBE score - 27/40 (67.5) 162/211 (76.8) 360/435 (82.8) 67/88 (76.1) 0.047

Kaplan-Meier estimates (%)

10-year decompensation rate - 7.9 7.1 5.6 - 0.49

10-year HCC incidence rate - 3.0 1.2 2.4 - 0.16

10-year transplant-free

survival

40.1 72.0 87.6 87.1 - <0.001

10-year liver-related death 53.2 14.0 4.9 6.5 - <0.001

Data represented as mean (standard deviation), n (%), or median (interquartile range). AMA, antimitochondrial antibody; ALP, alkaline phosphatase; ULN, upper limit of normal; AST, aspartate aminotransferase; ALT, alanine aminotransferase; LLN, lower limit of normal; APRI, AST to platelet ratio index; UDCA, ursodeoxycholic acid; HCC, hepatocellular carcinoma. aAge at diagnosis not available for one patient in 2000-2009 cohort. bAMA status was available for 3430 (97.5%) patients. cALP, bilirubin, AST, and ALT were log transformed prior to analyses and availability for laboratory values is as follows: ALP: 2662 (75.7%); Bilirubin: 2586 (73.5%); AST: 2593 (73.7%); ALT: 2271 (64.6%); Albumin: 2123 (60.3%); Platelet count: 1998 (56.8%) dThe cut-point APRI >0.54 at baseline is predictive of liver transplantation or death (Trivedi et al., 2014). eBiochemical disease stage classification according to Rotterdam criteria (E. M. Kuiper et al., 2009) was available in 2057 (58.5%) patients.

59 fUDCA therapy status was available for 3452 patients (98.1%). gUDCA dosage was available for 1319 (43.6%) of UDCA-treated patients. hResponse was determined based on the availability of laboratory values at 1 year of UDCA therapy. Response according to Toronto criteria was calculated after 2 years of UDCA therapy.

60

Figure S3-1. Mean age at diagnosis over calendar time stratified by A) Center (each line corresponds to an individual center); B) Sex; and

C) Biochemical disease stage.

Year of diagnosis

Age

at d

iagn

osis

(yea

rs)

1970-19791980-19891990-19992000-2009 ≥20100

2020

30

40

50

60

70

Year of diagnosis

Age

at d

iagn

osis

(yea

rs)

1970-19791980-19891990-19992000-2009 ≥20100

4040

50

60

70

MaleFemale

Year of diagnosis

Age

at d

iagn

osis

(yea

rs)

1970-19791980-19891990-19992000-2009 ≥20100

4040

50

60

70

Mild biochemical stageModerate biochemical stageAdvanced biochemical stage

A) B) C)

61

Figure S3-2. Absolute number of patients according to age at diagnosis and over calendar time.

Year of diagnosis

Cou

nt n

umbe

r

1970-1979 1980-1989 1990-1999 2000-20090

200

400

600

800

<3030-3940-4950-5960-69≥70

Age at diagnosis(years)

62

Table S3-3. Factorial ANOVA analysis of age at diagnosis over calendar time adjusting for sex

N=4804 Beta

coefficient Lower

95% CI Upper

95% CI P-value Male 4.03 2.93 5.14 <0.001

Female 0.00

Year of diagnosis 1970-1980 -10.00 -12.43 -7.57 <0.001

Year of diagnosis 1980-1990 -6.83 -8.51 -5.14 <0.001

Year of diagnosis 1990-2000 -4.17 -5.76 -2.58 <0.001

Year of diagnosis 2000-2010 -2.00 -3.58 -0.41 0.014

Year of diagnosis ≥2010 0.00

ANOVA, analysis of variance; CI, Confidence interval.

63

Table S3-4. Response rate in UDCA-treated patients according to various published criteria over calendar time

Response criterionᵃ 1970-1979 (n=78)

1980-1989 (n=735)

1990-1999 (n=1605)

2000-2009 (n=1563)

≥2010 (n=195)

P-value

Barcelona 30/61 (49.2) 277/493 (56.2) 630/1062 (59.3) 674/1131 (59.6) 100/155 (64.9) 0.185

Paris-I 31/60 (51.7) 268/533 (50.3) 790/1121 (70.5) 773/1047 (73.8) 107/149 (71.8) <0.001

Rotterdam 37/57 (64.9) 358/479 (74.7) 595/746 (79.8) 601/716 (83.9) 90/111 (81.1) <0.001

Toronto 19/41 (46.3) 200/395 (50.6) 570/851 (67.0) 564/759 (74.3) 48/65 (73.8) <0.001

Paris-II 13/64 (20.3) 151/542 (27.9) 548/1157 (47.4) 563/1121 (50.2) 81/155 (52.3) <0.001

GLOBE scoreb 13/25 (52.0) 111/190 (58.4) 200/285 (70.2) 382/463 (82.5) 67/88 (76.1) <0.001

Data represented as n (%). UDCA, ursodeoxycholic acid. ᵃResponse was determined based on the availability of laboratory values at 1 year of UDCA therapy. Response according to Toronto criteria was calculated after 2 years of UDCA therapy. bResponse according to the GLOBE score was established when the calculated value did not surpass the age-specific threshold (Lammers et al., 2015).

64

Table S3-5. Response rate over calendar time in UDCA-treated patients who did not meet criteria at baseline

Response criterionᵃ 1970-1979 (n=78)

1980-1989 (n=735)

1990-1999 (n=1605)

2000-2009 (n=1563)

≥2010 (n=195)

P-value

Paris-I 12/40 (30.0) 122/344 (35.5) 202/436 (46.3) 215/410 (52.4) 28/58 (48.3) <0.001

Rotterdam 9/29 (31.0) 121/242 (50.0) 176/327 (53.8) 264/379 (69.7) 43/64 (67.2) <0.001

Toronto 12/34 (35.3) 115/284 (40.5) 209/395 (52.9) 209/352 (59.4) 12/24 (50.0) <0.001

Paris-II 10/57 (17.5) 106/448 (23.7) 235/695 (33.8) 274/705 (38.9) 36/93 (38.7) <0.001

Data represented as n (%). UDCA, ursodeoxycholic acid. ᵃResponse was determined based on the availability of laboratory values at 1 year of UDCA therapy. Response according to Toronto criteria was calculated after 2 years of UDCA therapy.

65

Table S3-6. Multivariable Cox regression of 10-year hepatic decompensation (n=2962)

Variable HR 95% CI P value

Year of diagnosis 0.07

1970-1979 1.00

1980-1989 1.32 0.61-2.87 0.48

1990-1999 1.11 0.46-2.70 0.81

2000-2009 0.73 0.28-1.87 0.51

Male sex 1.25 0.80-1.98 0.33

Age at diagnosis (years) 0.21

<30 1.00

30-39 1.35 0.40-4.50 0.63

40-49 1.84 0.58-5.87 0.30

50-59 1.82 0.57-5.82 0.31

60-69 2.00 0.61-6.61 0.25

>70 3.09 0.88-10.81 0.08

Difference between diagnosis and study entry (years) 1.08 1.04-1.13 <0.001

HR, Hazard ratio; CI, confidence interval.

66

Table S3-7. Multivariable Cox regression for 10-year HCC incidence (n=3963)

Variable HR 95% CI P value

Male sex 3.48 2.05-5.89 <0.001

UDCA 0.38 0.21-0.67 0.001

Year of diagnosis 0.68

1970-1979 1.00

1980-1989 1.19 0.49-2.89 0.70

1990-1999 1.51 0.51-4.48 0.46

2000-2009 1.88 0.58-6.06 0.29

Age at diagnosis (years) 0.11

30-39 1.00

40-49 0.27 0.10-0.75 0.01

50-59 0.53 0.28-1.02 0.06

60-69 1.63 0.34-1.15 0.13

≥70 0.62 0.23-1.68 0.35

Difference between diagnosis and study entry (years) 1.19 1.13-1.26 <0.001

HCC, hepatocellular carcinoma; HR, hazard ratio, CI, confidence interval, UDCA, ursodeoxycholic acid.

67

Table S3-8. Multivariable Cox regression analysis of 10-year transplant-free survival (n=3354)

Variable HR 95% CI P-valueMale sex 1.11 0.89-1.40 0.35

UDCA 0.55 0.45-0.68 <0.001

Year of diagnosis <0.001

1970-1979 1.00

1980-1989 1.14 0.81-1.60 0.45

1990-1999 0.72 0.49-1.06 0.10

2000-2009 0.60 0.40-0.89 0.01

Age at diagnosis <0.001

<30 1.00

30-39 1.45 0.58-3.63 0.42

40-49 2.31 0.95-5.63 0.07

50-59 2.34 0.96-5.71 0.06

60-69 4.46 1.82-10.89 0.001

>70 8.52 3.45-21.07 <0.001

Log bilirubin (×ULN) 12.8 10.6-15.4 <0.001

Difference between diagnosis and study entry (years) 1.06 1.03-1.08 <0.001

HR, hazard ratio; CI, confidence interval; UDCA, ursodeoxycholic acid; ULN, upper limit of normal.

68

Chapter 4 : Study 2

Bilirubin is Predictive of Transplant-Free Survival 4Even Within the Normal Range in Patients with Primary Biliary Cholangitis

4.1 Introduction Primary biliary cholangitis (PBC) is an autoimmune cholestatic liver disease that is characterized

by chronic non-suppurative inflammation of the small intrahepatic bile ducts (Kaplan &

Gershwin, 2016). The disease usually has a slow progressive course, which may eventually lead

to cirrhosis and ultimately liver failure or premature death in the absence of liver transplantation.

However, the prolonged number of years it may take for patients to develop such clinical

outcomes poses a significant obstacle in randomized controlled trials that aim to evaluate the

clinical benefit of therapeutic interventions. Due to these feasibility concerns, various surrogate

markers have been evaluated for their prognostic value on clinical outcomes (Lammers et al.,

2014). Such surrogate markers can allow the risk stratification of patients without the need for an

extended follow-up period and can be implemented by health care providers or in clinical trials

to promptly assess the need and benefit of a therapeutic agent.

It is widely established that bilirubin is an independent predictor of prognosis in both

ursodeoxycholic acid (UDCA)-treated and untreated patients with PBC (Bonnand et al., 1999;

Lammers et al., 2014; Shapiro et al., 1979). The normalization of bilirubin prompted by UDCA

has been associated with improved transplant-free survival (Bonnand et al., 1999). Furthermore,

bilirubin has been established as a surrogate endpoint that is “reasonably likely to predict clinical

benefit” and the threshold that best predicted liver transplant-free survival was reported to be the

upper limit of normal (ULN) (Lammers et al., 2014). Normal bilirubin is also a component of

multiple response criteria, such as the Rotterdam, Paris-I, and Paris-II criteria (Corpechot et al.,

2008, 2011; ter Borg et al., 2006). Abnormal bilirubin levels are observed during later stages of

PBC and are indicative of impaired liver function (Hirschfield et al., 2017). Over the past

decades, however, there has been an increase in the proportion of patients that present with

normal bilirubin levels over the years and this group now represents the majority of patients with

PBC (Murillo Perez et al., 2018). Since bilirubin is usually not elevated above the ULN until

69

later stages of the disease, it is considered to be an inadequate marker for risk stratification in

early stage PBC. The prognostic value of bilirubin below the ULN has not been previously

assessed. Thus, the aim of this study was to evaluate whether bilirubin levels within the normal

range (≤1×ULN) are associated with liver transplant-free survival in patients with PBC.

4.2 Patients and Methods

4.2.1 Population and study design

This was a retrospective study on the predictive value of normal bilirubin for liver transplant-free

survival. We utilized the GLOBAL PBC Study Group database, which includes long-term

follow-up data of PBC patients from 17 centers across Europe and North America. To evaluate

the association between normal bilirubin and liver transplant-free survival, we included UDCA-

treated and untreated patients diagnosed with PBC according to internationally accepted

guidelines and whose bilirubin levels were normal (≤1×ULN as defined by each local center) at

time zero or 1 year after study entry (Hirschfield et al., 2017; K. D. Lindor et al., 2009;

Reshetnyak, 2015). Those with short follow-up (<6 months), short-term treatment with UDCA

(discontinued), absent laboratory values, unknown dates of important clinical events, overt

overlapping features of autoimmune hepatitis (AIH), or other concomitant liver diseases were

excluded from the study. Patients were allocated to two independent cohorts based on the time

point(s) at which their bilirubin levels were normal (time zero and 1 year). The inclusion of

patients into each cohort is not mutually exclusive, as patients may have had normal bilirubin

levels at both time points. This study was conducted in accordance with the 1975 Declaration of

Helsinki. The protocol was approved by the institutional research board at all participating

centers as per local regulations.

4.2.2 Data collection

In the GLOBAL PBC Study Group database, time zero (study entry) is defined as the date

UDCA was initiated in treated patients and the date of the first visit in untreated patients. At

study entry, the following data were available: sex, age at diagnosis, anti-mitochondrial (AMA)

antibody serological status, liver histology, biochemical disease stage (according to Rotterdam

criteria [ter Borg et al., 2006]), and UDCA therapy. The following laboratory parameters were

collected every 6-12 months: total bilirubin, alkaline phosphatase (ALP), albumin, aspartate

70

aminotransferase, alanine aminotransferase, and platelet count. Histological data obtained from

liver biopsies conducted within 12 months of study entry were staged according to Ludwig et

al.’s and Scheuer’s criteria (Ludwig et al., 1978; Scheuer, 1967). The completeness and accuracy

of the data was established by visits to participating centers.

4.2.3 Statistical analysis

The primary endpoint was a composite of liver transplantation or all-cause mortality. Patients

without an event (liver transplantation or death) at the end of follow-up and those who were lost

to follow-up were censored at their last visit. The predictive value of normal bilirubin on the

primary endpoint was initially analyzed based on the bilirubin quartiles corresponding to each

cohort. The liver transplant-free survival rates across quartiles were estimated with a Kaplan-

Meier curve and compared with a log-rank test. Multivariable Cox proportional hazards’

regression (hazard ratio [HR] with 95% CI) analyses were performed to adjust for potential

confounding variables: age at study entry, sex, year of diagnosis, UDCA therapy, ALP, and

geographical region.

To test the hypothesis of a threshold and to determine the optimal threshold for bilirubin within

the normal range two approaches were followed: 1) bilirubin levels at baseline and 1 year of

follow-up were dichotomized according to various thresholds ranging from 0.3 to 0.9 ×ULN in

0.01 increments. Multivariable Cox proportional hazards’ regression analyses were employed to

estimate the risk for liver transplantation or death associated with each threshold. The C-statistic

was calculated to evaluate the performance of each threshold in predicting liver transplant-free

survival and the threshold with the best performance was determined by the highest C-statistic.

2) To assess bilirubin on a continuous spectrum and test the hypothesis that the predetermined

bilirubin threshold is the point at which the risk for liver transplantation or death increases,

bilirubin was inserted into the Cox regression as a restricted cubic spline function with five

knots. This analysis included patients with bilirubin levels above the ULN to illustrate how their

risk for a poor prognosis differs relative to those with bilirubin below the ULN. The restricted

spline function was repeated with crude bilirubin levels (mg/dL).

All analyses were adjusted for age at study entry, sex, year of diagnosis, UDCA therapy, ALP,

and geographical region. Laboratory data included in the multivariable model that were not

71

normally distributed were log transformed. Sensitivity analyses of the predetermined bilirubin

threshold by multivariable Cox regression were performed in additional sub-groups stratified by

the ULN of bilirubin (<1.2mg/dL and ≥1.2mg/dL), age at study entry (≤55 years and >55 years),

sex, treatment (UDCA-treated and UDCA-untreated), histological stage (I-II and III-IV), and

ALP (≤1.67×ULN and >1.67×ULN). Furthermore, sensitivity analyses were performed for

bilirubin at 2-4 years after the start of follow-up.

For illustrative purposes, Kaplan-Meier analyses were conducted to describe the liver transplant-

free survival rates associated with bilirubin levels at baseline and 1 year (normal bilirubin [≤/>

the threshold] and abnormal bilirubin). Patients with abnormal bilirubin were included for

reference purposes. The distribution of the clinical events (liver transplantation, liver-related

death, or liver-unrelated death) at 10 years within each bilirubin group was also evaluated.

An additional analysis was conducted in UDCA-treated patients whose baseline bilirubin levels

were above the predetermined threshold and stratified based on their bilirubin levels at 1 year. In

case of missing bilirubin at baseline or 1 year, the imputed laboratory data was used. Multiple

imputation with by the Markov chain Monte Carlo (MCMC) method for missing data and

Rubin’s rules were used to estimate bilirubin and its standard error (Rubin, 1996). Ten imputed

datasets based on the assumption that data were missing at random were created from iterations

to reduce sampling variability.

The pattern of bilirubin (mean and 95% CI) over the first 5 years was evaluated in patients with

normal bilirubin at time zero and stratified based on whether they experienced a late clinical

event (liver transplantation or death from 5-10 years) or no clinical event in the first 10 years of

follow-up. All patients included in the latter group had 10 years of follow-up. The imputed

dataset was used for this analysis.

A P-value less than 0.05 was considered statistically significant. All analyses were two-sided and

were performed using IBM SPSS Statistics for Windows, version 24.0 (IBM Corp., Armonk,

NY, USA) and SAS version 9.4 (SAS Institute Inc., Cary, NC, USA).

72

4.3 Results

4.3.1 Study population characteristics

A total of 3640 patients with normal bilirubin at baseline or one year after study entry were

included. Two normal bilirubin cohorts were constructed based on the time point(s) at which

their bilirubin levels were normal: time-zero cohort (n=2795) and 1-year cohort (n=2967). An

overlap of 2122 patients exists between these cohorts. There were 374 and 410 primary

endpoints according to each respective cohort. Patient characteristics per cohort are presented in

Table 4-1.

73

Table 4-1. Characteristics of PBC patients in each normal bilirubin cohort

Parameter Time zero cohort a, b

(n=2795) 1-year cohort

(n=2967)Follow-up time, y, median (IQR) 7.6 (4.0-12.1) 7.1 (3.5-11.2)

Age at study entry, mean ± SD 55.7 ± 11.9 55.0 ± 11.7

Female, no. (%) 2543 (91.0) 2718 (91.6)

AMA-positive, no. (%) 2502/2736 (91.4) 2649/2897 (91.4)

Year of diagnosis, median (range) 1999 (1961-2014) 1998 (1961-2013)

UDCA-treated, no. (%) 2336/2737 (85.3) 2652/2935 (90.4)

Laboratory parameters, median (IQR)c

Total bilirubin, ×ULN 0.53 (0.40-0.71) 0.50 (0.38-0.68)

ALP, ×ULN 1.84 (1.20-3.03) 1.25 (0.88-1.96)

Albumin, ×LLN 1.17 (1.09-1.26) 1.17 (1.09-1.26)

AST, ×ULN 1.20 (0.83-1.80) 0.84 (0.64-1.18)

ALT, ×ULN 1.45 (0.93-2.30) 0.83 (0.57-1.31)

Platelet count, 109/L 252 (202-304) 247 (197-300)

Bilirubin ULN (mg/dL), median (IQR)d 1.0 (1.0-1.2) 1.0 (1.0-1.2)

PBC, primary biliary cholangitis; IQR, interquartile range; SD, standard deviation; AMA, antimitochondrial antibody; UDCA, ursodeoxycholic acid; ULN, upper limit of normal; ALP, alkaline phosphatase; AST, aspartate aminotransferase; ALT, alanine aminotransferase. aHistological disease stage at study entry available for 1485 patients (53.1%)- stage I/II: 1111 patients (74.8%), stage III/IV: 374 patients (25.2%). bBiochemical disease stage at study entry available for 2259 patients (80.8%) – mild: 2094 (74.9%), moderate: 165 (5.9%), advanced: 0 patients. cLaboratory parameters other than bilirubin were not available for all patients:

Time zero cohort: ALP (n=2627), albumin (n=2259), AST (n=2531), ALT (n=2134), platelet count (n=2039). 1-year cohort: ALP (n=2824), albumin (n=2043), AST (n=2542), ALT (n=2309), plateletcount (n=1370).

dThe upper limit of normal for bilirubin was variable per center.

74

4.3.2 Normal bilirubin quartiles are associated with liver transplant-free survival

The quartiles in each individual cohort were formulated according to the following bilirubin

levels (median [IQR], ×ULN): 0.53 (0.40-0.71) and 0.50 (0.38-0.68), respectively. In Kaplan-

Meier analysis of patients that had normal bilirubin at time zero, the cumulative 10-year liver

transplant-free survival rate decreased with higher bilirubin quartiles and was 92.4%, 89.0%,

87.9%, 77.5% from quartiles 1-4 (Q1-Q4), respectively (Figure 4-1). In pairwise comparisons,

Q4 was significantly different from Q1-Q3 (all P < 0.0001). Similar results were obtained in the

Kaplan-Meier analysis of the 1-year cohort, in which the 10-year liver transplant-free survival

rates with increasing bilirubin quartiles were 91.3%, 91.4%, 86.2%, and 76.7%. Q3 and Q4 were

significantly different from one another and from the remaining quartiles (all P < 0.05). In

multivariable Cox regression analyses, normal bilirubin quartiles were a significant predictor for

transplant-free survival. In the time zero cohort, the risk for liver transplantation or death

increased with higher bilirubin quartiles: Q1 (reference), Q2 (HR 1.24, 95% CI 0.85-1.81, P =

0.26), Q3 (HR 1.28, 95% CI 0.90-1.83, P = 0.18), Q4 (HR 2.16, 95% CI 1.53-3.04, P < 0.0001).

A similar trend was observed in the 1-year cohort: Q1 (reference), Q2 (HR 0.98, 95% CI 0.68-

1.41, P = 0.91), Q3 (HR 1.43, 95% CI 1.01-2.01, P = 0.04), Q4 (HR 2.27, 95% CI 1.65-3.12, P <

0.0001).

Figure 4-1. Transplant-free survival of the normal bilirubin quartiles in patients with normal

bilirubin at A) time zero and B) 1 year.

Follow-up (years)

14121086420

Tran

plan

t-fre

e su

rviv

al (%

)

100

90

80

70

60

50

<25th percentile-censored

>75th percentile 50-75th percentile 25-50th percentile <25th percentile

Quartile 4 Quartile 3Quartile 2 Quartile 1

Follow-up (years)

11111000

Tran

spla

nt-fr

ee s

urvi

val (

%)

100

90

80

70

60

50

<0-25th percentile-censored

>75th percentile 50-75th percentile 25-50th percentile <25th percentile

1 3 5 7 9 11 13 15

A ) B )T im e 0 c o h o rt 1 -y e a r c o h o rt

75

4.3.3 Bilirubin threshold within the normal range

Upon exploration of the optimal threshold of bilirubin within the normal range, all bilirubin

thresholds (0.3-0.9×ULN) were significant predictors of liver transplant-free survival in that

patients with bilirubin above each threshold had an increased risk for liver transplantation or

death (Table 4-2 and Table S4-1). The bilirubin threshold at 1 year with the highest ability to

predict liver transplantation or death was 0.6×ULN (C-statistic 0.7394, 95% CI 0.7134-0.7655).

The 10-year liver transplant-free survival of patients with normal bilirubin ≤0.6×ULN, normal

bilirubin >0.6×ULN, and abnormal bilirubin 1 year were 90.7%, 78.0%, and 39.0%, respectively

(P < 0.0001) (Figure 4-2A). At baseline, the 10-year liver transplant-free survival rates were

89.4%, 81.9%, and 43.4% (P < 0.0001). We evaluated the distribution of clinical events from the

10-year liver transplant-free survival rates associated with each bilirubin group. Clinical events

in patients with bilirubin from 0.6-1.0×ULN were characterized by an increased proportion of

liver transplantations and liver-related deaths, alongside a decreased proportion of liver-unrelated

deaths compared to patients with bilirubin ≤0.6×ULN (Figure S4-1). In an analysis of UDCA-

treated patients with normal bilirubin levels >0.6×ULN at baseline (n=1318), a reduction in

bilirubin ≤0.6×ULN at 1 year was associated with prolonged liver transplant-free survival as

compared to stable bilirubin that remained above the threshold and abnormal bilirubin after 1

year (both P < 0.001) (Figure 4-2B). From these patients, the 10-year transplant-free survival of

patients ≤0.6×ULN, normal bilirubin >0.6×ULN, and abnormal bilirubin was 92.8%, 84.7%, and

62.1%.

76

Table 4-2. Multivariable Cox regression analyses of various bilirubin thresholds in patients with

normal bilirubin at 1 year to evaluate performance for the prediction of liver transplantation and

death

Bilirubin at 1 year (n=2793) Threshold

(×ULN) C-statistic (95%CI) HR (95% CI) P value No. of patients

≤/> threshold 0.30 0.7163 (0.6890-0.7437) 1.52 (1.02-2.27) 0.041 349/2444

0.40 0.7202 (0.6931-0.7472) 1.53 (1.17-2.02) 0.002 821/1972

0.50 0.7335 (0.7072-0.7598) 1.88 (1.51-2.34) <0.001 1435/1357

0.55 0.7330 (0.7068-0.7591) 1.93 (1.56-2.38) <0.001 1609/1184

0.59 0.7372 (0.7110-0.7633) 2.05 (1.67-2.52) <0.001 1766/1027

0.60 0.7394 (0.7134-0.7655) 2.13 (1.74-2.62) <0.001 1885/908

0.61 0.7393 (0.7131-0.7655) 2.11 (1.72-2.59) <0.001 1896/897

0.62 0.7365 (0.7101-0.7629) 2.05 (1.67-2.51) <0.001 1942/851

0.63 0.7363 (0.7099-0.7626) 2.06 (1.68-2.53) <0.001 1953/840

0.65 0.7333 (0.7067-0.7598) 1.97 (1.60-2.41) <0.001 2017/776

0.66 0.7335 (0.7070-0.7600) 1.97 (1.61-2.42) <0.001 2021/772

0.67 0.7347 (0.7082-0.7613) 2.00 (1.63-2.45) <0.001 2087/706

0.68 0.7338 (0.7073-0.7603) 1.98 (1.61-2.43) <0.001 2090/703

0.69 0.7331 (0.7066-0.7596) 1.99 (1.62-2.44) <0.001 2120/673

0.70 0.7332 (0.7068-0.7596) 2.00 (1.62-2.46) <0.001 2214/579

0.75 0.7340 (0.7072-0.7608) 2.07 (1.67-2.58) <0.001 2324/469

0.80 0.7305 (0.7036-0.7573) 2.19 (1.75-2.76) <0.001 2448/345

0.85 0.7263 (0.6992-0.7535) 2.01 (1.55-2.60) <0.001 2538/255

0.90 0.7193 (0.6919-0.7467) 1.79 (1.31-2.43) <0.001 2629/164

ULN, upper limit of normal; HR, hazard ratio; CI, confidence interval.

77

Figure 4-2. Transplant-free survival in patients with normal bilirubin (stratified by 0.6×ULN

threshold) and abnormal bilirubin. A) Kaplan-Meier estimates of transplant-free survival rates in

patients with normal bilirubin (stratified by 0.6×ULN threshold) and abnormal bilirubin at 1

year. B) Additional analysis of the transplant-free survival rates in UDCA-treated patients with

bilirubin levels >0.6×ULN at baseline.

13

Follow-up (years)

12840

Tran

spla

nt-fr

ee s

urvi

val (

%)

100

80

60

40

20

0Bilirubin at 1yr <=0.6-censoredAbnormal bilirubin Normal bilirubin (>0.6)Nomal bilirubin (<=0.6)

1 5 9

1 9 8 49 8 37 1 5

1 4 2 06 9 93 6 2

8 3 24 4 21 6 8

4 1 32 2 08 9

abc

Follow-up (years)

12840

Tran

spla

nt-fr

ee s

urvi

val (

%)

100

80

60

40

20

0

<=0.6-censoredAbnormal bilirubinBilirubin remained >0.6Bilirubin decreased (<0.6)

1-year bilirubin bilirubin in patients with normal bilirubin (>0.6) at time 0

1 5 9 13

5 4 56 6 31 1 0

4 1 14 9 57 5

2 7 63 3 84 4

1 5 71 6 22 4

abc

a

b

c

a

b

c

A ) B )

78

The threshold was evaluated in various sub-groups of patients that had normal bilirubin at 1 year,

all of which confirmed that patients with bilirubin ≤0.6×ULN have a decreased risk for liver

transplantation or death (Figure 4-3, Table S4-2). Importantly, the association with a reduced

risk remained when all patients with normal bilirubin in which the ULN was defined as

≥1.2mg/dL were excluded from the analyses (HR 2.06, 95% CI 1.58-2.67, P < 0.0001). The

threshold of 0.6×ULN did not reach statistical significance in males, however bilirubin levels

above the threshold were also associated with an increased risk in these patients.

Figure 4-3. Sub-group analyses based on the bilirubin threshold of 0.6×ULN in patients with

normal bilirubin at 1 year. Hazard ratio for liver transplantation or death (95% CI) obtained from

multivariable Cox regression analyses in patients with normal bilirubin in various sub-groups.

The hazard ratios correspond to bilirubin levels >0.6×ULN (versus bilirubin ≤0.6×ULN).

Transplant-free survival hazard ratio ofbilirubin >0.6×ULN at 1 year (95%CI)

0 1 2 3 4 5 6

ALP>1.67×ULN 912

ALP≤1.67×ULN 1870

Histological stage (III-IV) 321

Histological stage (I-II) 896

UDCA-treated 2527

UDCA-untreated 254

Male 229

Female 2561

Age at study entry >55 1379

Age at study entry ≤55 1395

ULN of bilirubin ≥1.2mg/dl 861

ULN of bilirubin <1.2mg/dl 1420

Entire cohort 2793

Sub-groups n=

79

4.3.4 The risk for liver transplantation or death increases at bilirubin levels of 0.6×ULN

We assessed bilirubin on a continuous spectrum with a restricted spline function to evaluate

whether the predetermined threshold is the point at which the transplant-free survival hazard

ratio increases. The reference in each cohort was the predetermined threshold of 0.6×ULN. In

both cohorts, the risk for liver transplantation or death remained stable below 0.6×ULN (Figure

4-4). However, beyond this threshold, a linear relationship was observed between bilirubin and

the risk for liver transplantation or death that continued past the normal range. The test for

curvature, which establishes a non-linear relationship, was significantly different for the baseline

cohort (P < 0.0001) and for the 1-year cohort it approached significance (P = 0.08). As a

sensitivity analysis, the restricted spline function analysis was repeated using crude bilirubin

levels (mg/dL) (Figure S4-2). The spline function analyses were repeated with normal bilirubin

levels at other time points (2-4 years), which suggested that the threshold can be applied after 1

year of follow-up as a similar trend is observed (Figure S4-3).

Figure 4-4. The association between bilirubin levels (×ULN) and risk for liver transplantation or

death. Hazard ratios and 95% CI were estimated by a restricted cubic spline function in A) the

time zero cohort and B) the 1-year cohort. The bilirubin reference in each cohort is 0.6×ULN.

80

4.3.5 Patients who remain below 0.6×ULN over time have good long-term prognosis

To assess how the trajectory of bilirubin over time may be related with the development of a

clinical event (liver transplantation or death), bilirubin levels over the course of 5 years were

evaluated in patients with normal bilirubin at time zero. The patients were stratified according to

whether they developed a late clinical event from 5-10 years (n=132) or did not develop a

clinical event in the first 10 years of follow-up (n=979). Patients who had no clinical event after

10 years of follow-up presented with a mean bilirubin level of 0.55×ULN (95% CI 0.54-0.56)

and demonstrated stable bilirubin levels (below 0.6×ULN) in the first five years (Figure 4-5). In

contrast, patients who reached a clinical endpoint presented with slightly higher mean bilirubin

levels (0.63×ULN, 95% CI 0.59-0.67, P < 0.001) and exhibited a gradual increase within the

normal range that precluded the occurrence liver transplantation or death.

4.3.6 The proportion of patients with bilirubin ≤0.6×ULN increased over time

The proportion of patients with normal bilirubin has increased over time. Therefore, we assessed

if this was applicable with our threshold of 0.6×ULN in patients with already normal bilirubin at

baseline. Even in this group, there was a gradual increase over time in those with bilirubin

≤0.6×ULN over time, whom accounted for 48% in the 1970s but 73% after 2010 (Figure S4-4).

This further supports the notion that bilirubin within the normal may not indicate an absence of

risk in patients with PBC.

81

Figure 4-5. Mean bilirubin levels over 5 years in patients with normal bilirubin at study entry

and stratified by outcome. Trajectory of the mean bilirubin levels (×ULN) and 95% CI over the

first 5 years depending on whether they experienced a late clinical event between 5 and 10 years

(n=132) or no event within the first 10 years of follow-up (n=979). Clinical event is defined as

liver transplantation or death. All patients without a clinical event had a follow-up of at least 10

years.

Time (years)

Bili

rubi

n (x

ULN

)

0 1 2 3 4 50.0

0.5

1.5Late event (5-10 years)No event at 10 years (follow-up to 10 years)

1

0.6

82

4.4 Discussion This study is the first to report that bilirubin levels within the normal range are associated with

the risk for liver transplantation or death in patients with PBC. We demonstrated that bilirubin

levels ≤0.6×ULN at baseline and 1 year were associated with a decreased risk for liver

transplantation or death compared to patients with bilirubin above this threshold and that a

reduction in bilirubin within the ULN after 1 year of UDCA therapy was associated with

prolonged liver transplant-free survival. While the risk for liver transplantation or death was

stable when bilirubin levels were below 0.6×ULN, beyond this threshold, a positive linear

relationship was observed between bilirubin and the risk for a clinical event. These results were

confirmed in several sub-groups of patients. Our findings suggest that the interpretation of not

being at risk if bilirubin is within the normal range needs to be revised. This might have

implications for the number of patients that are eligible for clinical trials and that would be able

to receive novel second-line therapeutic agents. Furthermore, the results presented in this study

raise the question of whether there may be a similar trend present in other laboratory parameters.

Although previous studies reported that the ULN of bilirubin was the most predictive for liver

transplant-free survival in patients with PBC and considered a reasonable threshold (Lammers et

al., 2014), we found that the risk for liver transplantation or death is already increased when

bilirubin levels were above 0.6×ULN. The current ULN of bilirubin represents the 97.5

percentile cut-off in the general population, yet this may not be the best approach to determine an

optimal threshold since levels below this threshold are not reflective of an absence of increased

risk (Zucker et al., 2004). In part, this might be explained by the high percentage of individuals

with Gilbert’s syndrome in the general population, which ranges from 3-10% (Bosma et al.,

1995). These patients experience hyperbilirubinemia due to a genetic deficiency in UGT1A1,

the enzyme responsible for glucuronidation of unconjugated bilirubin, also rendering it water-

soluble for excretion through bile. Additionally, the current ULN of bilirubin may be a

suboptimal threshold for risk stratification in PBC due to the female predominance of the

disease, while sex differences in bilirubin are present in the general population (Podda, Selmi,

Lleo, Moroni, & Invernizzi, 2013). An American study based on the Third National Health and

Nutrition Examination Survey (NHANES III) assessed serum bilirubin levels in 16,865 adults

from the general population and reported that mean serum bilirubin levels are significantly lower

in women (0.52 mg/dL ± 0.003) than men (0.72 mg/dL ± 0.004) (Zucker et al., 2004).

83

Consequently, the 97.5 percentile cut-off was 0.5 mg/dL higher in men. Other studies have

reported similar sex differences in bilirubin levels in the general population (Rosenthal, Pincus,

& Fink, 1984; White, Nelson, Pedersen, & Ash, 1981). Thus, the overall ULN of bilirubin may

be skewed to higher levels in PBC because of the inclusion of both men and women. These

considerations suggest that the ULN for bilirubin may need to be stratified by sex, as has been

previously implemented for aspartate aminotransferase (Prati et al., 2002; Terrault et al., 2016).

We found that the predictive value of the bilirubin threshold of 0.6×ULN was irrespective of age,

treatment with UDCA, histological stage, or ALP levels. Importantly, it remained significantly

predictive at various independent time points. Furthermore, in patients treated with UDCA that

had a bilirubin level above 0.6×ULN but below the ULN at initiation of treatment, we found that

a reduction below 0.6 was associated with a significantly prolonged liver transplant-free survival

as compared to remaining within the normal range or increasing to an abnormal bilirubin level.

This suggests that besides the predictive value of bilirubin within the normal range, a treatment-

induced reduction of bilirubin within the current normal range is beneficial for long-term

prognosis, which could have important implications for current patient care, but also for the

design and interpretation of future clinical trials of potential second-line therapies in PBC. While

recent clinical trials have often included normalization of bilirubin as a primary endpoint, it

might be preferable to aim for lower bilirubin levels (Corpechot et al., 2017; Nevens et al.,

2016).

The pattern of bilirubin within the current normal range over time may also be relevant, as there

was an overall increase of 0.41×ULN in mean bilirubin during the first 5 years of follow-up in

patients who eventually reached a clinical endpoint after extended follow-up. While rapid

increases in bilirubin have been shown to preclude death in untreated patients, these results

suggest that there is an association between the trajectory of bilirubin and clinical outcomes even

if within the normal range (Shapiro et al., 1979). The fact that the mean bilirubin levels of

patients who did not experience a clinical event remained below 0.6×ULN over time further

supports an incentive to aim for bilirubin levels below our proposed threshold of 0.6×ULN.

Further, our findings emphasize the importance of the continuous clinical evaluation of patients’

bilirubin levels even in those with early stage disease.

84

A robust analysis of the predictive value of bilirubin within the normal range would not be

possible without the large number of patients and extended follow-up available from the

GLOBAL PBC Study Group cohort. Furthermore, bilirubin was assessed at multiple

independent time points to confirm that bilirubin levels obtained during a random follow-up

assessment could also be utilized for risk stratification. Nonetheless, some study limitations

should be noted. Bilirubin was available as total bilirubin; therefore, we could not assess

conjugated bilirubin levels independently or the conjugated/unconjugated bilirubin ratio.

Whereas, total serum bilirubin levels in healthy patients are primarily composed of unconjugated

bilirubin, total bilirubin in patients with PBC is predominantly conjugated bilirubin that leaks

into the serum when it is unable to be excreted through bile (Levitt & Levitt, 2014). Since PBC

is characterized by elevations in conjugated bilirubin and an altered conjugated/unconjugated

bilirubin ratio, it might be of additional relevance to measure conjugated bilirubin in future

studies of bilirubin (Levitt & Levitt, 2014). Standard clinical laboratories measure bilirubin

fractions, such as direct and indirect, with the diazo (Van den Bergh) reaction. Both conjugated

and delta bilirubin react directly with the diazo reagent, while unconjugated bilirubin does not.

Therefore, direct bilirubin is not synonymous to conjugated bilirubin, while poses a problem for

measuring conjugated bilirubin independently. Although bilirubin was analyzed based on the

ULN defined by local centers, which ranged from 0.6-1.7mg/dL, sensitivity analyses were

performed to address this. The analyses with crude bilirubin levels (mg/dL) as well as the one

excluding patients with an ULN above 1.2mg/dL confirmed our initial findings and exclude the

possibility that patients with bilirubin levels above 0.6×ULN have worse liver transplant-free

survival due to the utilization of high ULNs.

In this multi-center international follow-up study of patients with PBC, bilirubin levels below the

current ULN were shown to be predictive of liver transplant-free survival and 0.6×ULN was

established as the threshold from which point on the risk for liver transplantation or death

increases. Additionally, reduction within the current normal range to below 0.6×ULN was

associated with prolonged transplant-free survival. Our proposed threshold of 0.6× the current

ULN of bilirubin may be a more sensitive reference to identify patients at risk for a poor

outcome and represent a threshold that increases the number of patients included in intervention

studies that may benefit from therapeutic agents.

85

4.5 Supplementary Tables and Figures Table S4-1. Multivariable Cox regression analyses of various bilirubin thresholds in patients

with normal bilirubin at time zero to evaluate performance for the prediction of liver

transplantation and death

Threshold (×ULN)

C-statistic (95%CI) HR (95% CI) P value No. of patients ≤/> threshold

0.30 0.7320 (0.7036-0.7604) 1.63 (1.02-2.60) 0.042 272/2282

0.40 0.7357 (0.7076-0.7638) 1.53 (1.12-2.10) 0.008 649/1905

0.50 0.7390 (0.7113-0.7666) 1.54 (1.22-1.95) <0.001 1191/1363

0.55 0.7378 (0.7098-0.7658) 1.47 (1.17-1.84) 0.001 1347/1207

0.60 0.7397 (0.7117-0.7676) 1.53 (1.23-1.90) <0.001 1620/934

0.65 0.7456 (0.7181-0.7730) 1.73 (1.39-2.15) <0.001 1732/822

0.66 0.7457 (0.7182-0.7732) 1.72 (1.39-2.14) <0.001 1736/818

0.67 0.7443 (0.7167-0.7719) 1.64 (1.32-2.03) <0.001 1789/765

0.68 0.7415 (0.7136-0.7693) 1.58 (1.27-1.97) <0.001 1800/754

0.69 0.7423 (0.7146-0.7701) 1.60 (1.29-1.99) <0.001 1824/730

0.70 0.7436 (0.7159-0.7713) 1.69 (1.35-2.10) <0.001 1910/644

0.75 0.7435 (0.7161-0.7709) 1.79 (1.43-2.25) <0.001 2041/513

0.80 0.7421 (0.7143-0.7700) 1.73 (1.35-2.21) <0.001 2187/367

0.85 0.7432 (0.7154-0.7709) 1.87 (1.44-2.42) <0.001 2278/276

0.90 0.7434 (0.7157-0.7710) 2.19 (1.61-2.98) <0.001 2392/162

ULN, upper limit of normal; HR, hazard ratio; CI, confidence interval.

86

Figure S4-1. Distribution of clinical events (liver transplantation, liver-related death, liver-

unrelated death) from the 10-year transplant-free survival rates associated with each bilirubin

group at A) time zero and B) 1 year.

Bilirubin (×ULN)

Perc

enta

ge o

f pat

ient

s

≤0.6 0.6-1.0 >1.00

20

40

60

80

100

Patients:10-year events:

n=1755 n=1040 n=1003n=119 n=136 n=441

Time zero cohort

Bilirubin (xULN)

Perc

enta

ge o

f pat

ient

s

≤0.6 0.6-1.0 >1.00

20

40

60

80

100

Patients:10-year events:

n=1984 n=983 n=715n=117 n=157 n=345

1-year cohortA) B)

Liver-unrelated deathLiver-related deathLiver transplantation

87

Table S4-2. Multivariable analysis of 0.6×ULN threshold at 1 year in various sub-groups

Sub-group Number of patients

HR (95% CI) P value

ULN <1.2 mg/dL 1420 2.06 (1.58-2.67) <0.001

ULN ≥1.2 mg/dL 861 3.71 (2.37-5.82) <0.001

Age at study entry ≤55 1395 1.86 (1.31-2.64) 0.001

Age at study entry >55 1379 2.30 (1.79-2.97) <0.001

Female 2561 2.25 (1.81-2.79) <0.001

Male 229 1.31 (0.68-2.49) 0.42

UDCA-untreated 254 1.97 (1.04-3.73) 0.038

UDCA-treated 2527 2.17 (1.74-2.70) <0.001

Histological stage (I-II)a 896 2.10 (1.28-3.47) 0.004

Histological stage (III-IV)a 321 2.01 (1.74-3.29) 0.006

ALP ≤1.67×ULN 1870 1.95 (1.49-2.56) <0.001

ALP >1.67×ULN 912 2.39 (1.74-3.29) <0.001

HR, hazard ratio; CI, confidence interval; ULN, upper limit of normal; UDCA, ursodeoxycholic acid; ALP, alkaline phosphatase. aHistological stage determined at study entry.

88

Figure S4-2. The association between bilirubin levels (mg/dL) and risk for liver transplantation

or death. Hazard ratios and 95% CI were estimated by a restricted cubic spline function in A) the

time zero cohort and B) the 1-year cohort. The bilirubin reference in each cohort is 0.65 mg/dL

and the test for curvature is significant in both cohorts (P < 0.0001 and P = 0.04).

89

Figure S4-3. The association between bilirubin levels (×ULN) and risk for liver transplantation

(LT) or death. Hazard ratios and 95% CI were estimated by a restricted cubic spline function at

2-4 years. The bilirubin reference in each cohort is 0.6×ULN.

90

Figure S4-4. The distribution of patients with bilirubin below and above 0.6×ULN in those with

normal bilirubin at baseline (n=2791).

Year of diagnosis

Perc

enta

ge o

f pat

ient

s

1970-19791980-19891990-19992000-2009 ≥20100

20

40

60

80

100

≤0.6×ULN

>0.6×ULN

Baseline bilirubin

P < 0.001

91

Chapter 5

General Discussion 5

5.1 Calendar Time Trends

5.1.1 Discussion

Insight into the temporal changes of clinical features at presentation of PBC patients has been

limited. Studies suggest that patients diagnosed in recent decades are predominantly

asymptomatic (T. R. Baldursdottir et al., 2012; Floreani et al., 2011). There are also reports of an

increase in age at diagnosis, and no difference in the female: male ratios (Floreani et al., 2011;

Rautiainen et al., 2007; Sakauchi et al., 2007). Whether there is a difference in the survival rates

of patients according to year of diagnosis is inconclusive. One study from Finland reported

improved survival and a Canadian study did not find year of diagnosis to be a significant

predictor for survival (Myers et al., 2009; Rautiainen et al., 2007).

We sought to describe patient and disease characteristics over a 44-year period in a globally

representative population. This study contains the most comprehensive evidence to date due to

its size, long-term follow-up, and the inclusion of multiple centers worldwide. The main findings

from this retrospective study indicated that the mean age at diagnosis increased by 2-3 years per

decade from 47 years to 57 years, the female to male ratio and AMA-positivity were unchanged,

the proportion of patients presenting with mild biochemical and histological disease stage

increased, more patients responded to UDCA, and there were lower decompensation rates and

higher transplant-free survival rates in more recent decades. These results support the hypothesis

that the natural of history of PBC has changed over time. The most plausible contributors to the

observed changes in the natural history of PBC are improved awareness, early diagnosis, and

availability of treatment. However, the possibility that these changes may also be due to

environmental factors cannot be excluded since multiple studies have indicated that the incidence

of PBC is increasing in various geographical regions (Al-Harthy & Kumagi, 2012; Floreani et

al., 2011).

92

5.1.1.1 Symptoms

A key difference in patients diagnosed in recent times compared to those diagnosed in earlier

times is symptomology, as previous studies have consistently reported a decrease in the

proportion of symptomatic patients (T. R. Baldursdottir et al., 2012; Floreani et al., 2011).

Symptoms are an important aspect to consider when assessing these temporal trends, as studies

show that asymptomatic PBC patients present with an earlier histological stage and improved

biochemical measures (Mitchison et al., 1990). Furthermore, symptomatic presentation may

predict an inadequate response to UDCA and poor prognosis (Jones, Al-Rifai, Frith, Patanwala,

& Newton, 2010; Quarneti et al., 2015). Although asymptomatic PBC is generally less severe at

diagnosis than symptomatic, being asymptomatic is not always synonymous with having an early

disease (Kumagi & Heathcote, 2008). Unfortunately, an evaluation of the changes in

symptomology was not possible because this information was not available in the GLOBAL

PBC database. Since most patients nowadays present without symptoms, we speculate that

patients diagnosed in recent decades from our cohort were predominantly asymptomatic.

5.1.1.2 AMA testing and diagnosis

One of the current diagnostic criteria for PBC is seropositivity for AMA. The conventional

method for AMA detection is IIF, yet there has been an increase in ELISA-based assays and

immunoblotting due to the identification of specific antigen reactivity (Oertelt et al., 2007).

These improvements have led to greater sensitivity and specificity when detecting AMA.

However, there is still heterogeneity in the results and antigenic epitopes tested across

laboratories. Furthermore, patients are generally screened for AMA after they demonstrate

cholestatic laboratory results, such as elevated ALP or GGT. Improvements in the sensitivity of

AMA tests would hypothetically translate to an increase in the proportion of AMA-positive

patients, however this has remained unchanged in our cohort. Therefore, we speculate that any

changes in the techniques used for AMA testing have not played a major role in the distinct

presentation of patients over calendar time. A significant role on the observed changes is

expected to result from increased physician awareness and routine testing of liver function in

recent years, which suggests that earlier cohorts were largely composed of patients that were

diagnosed due to symptoms/complications and only a minority of patients’ diagnosis were

prompted by routine testing.

93

5.1.1.3 Age relative to the general population

In line with our findings, there have been reports that the age at diagnosis has increased over

time in Italian and Japanese populations (Floreani et al., 2011; Sakauchi et al., 2007). Increased

routine testing in recent years may account for the increase in older patients being diagnosed

with PBC, as screening occurs more frequently in the elderly (Spalding & Sebesta, 2008). The

increase in age at diagnosis may potentially be associated with the increase in age seen in the

general population, therefore it is critical that the age data be compared to that of the general

population. In the PBC population, there was a gradual increase in the proportion of patients 50

years old and above at the time of diagnosis from 38% in the 1970s to 72% after 2010, an overall

increase of 34% in the investigated time frame. In the general population there has been an

overall increase of 11% in the proportion of individuals ≥50 years old, which increased from

25% in 1970 to 36% in 2015 (United Nations, Department of Economic and Social Affairs,

2017). When comparing age trends in PBC patients to that of the general population, the

percentage of patients ≥50 years old are overall higher in the PBC population, which may reflect

the restricted age spectrum observed in PBC since patients under 18 years old were excluded and

PBC typically affects middle-aged individuals. It is logical that there are higher proportions of

this age group in the PBC cohort compared to that of the general population. Yet, the increase of

this age group in the PBC population was greater compared to that of the general population

within the specified time frame. This suggests that there may be additional factors contributing to

the increase in age observed in PBC other than an aging population. Additional potential

contributors to the increase in age include a delayed trigger for PBC as a result of changes in

environmental factors or a longer incubation period from when there is AMA-positivity to the

development of abnormal liver biochemistry.

5.1.1.4 Biochemical and histological disease stage

Liver biochemistry and histological findings in the GLOBAL PBC cohort indicated that patients

presented at an earlier disease stage, which can also be attributed to improved patient care and an

earlier diagnosis of PBC. A study from Iceland compared the proportion of patients who

presented with an advanced histological stage (III-IV) in 1991-2000 to those from 2001-2010

and reported no difference over time, accounting for 28% of patients (T. R. Baldursdottir et al.,

2012). Interestingly, they did note a decrease in the proportion of asymptomatic patients, which

94

further supports the notion that symptoms may not directly correlate with histology. In contrast,

we noted patients with an advanced histological stage (III-IV) accounted for 33% in 1990-1999

and that there was a decrease to 23.5% in 2000-2014.

5.1.1.5 Treatment regimen with UDCA

The availability and timing of treatment for PBC can have a major impact on response to

treatment and the subsequent development of clinical outcomes. Although UDCA was used as

off-label therapy starting in the late 1980s, it did not gain FDA approval until 1997. UDCA is

the standard treatment for PBC because it has been shown to delay histological progression and

improve survival (Parés et al., 2000; Shi et al., 2006). The introduction of UDCA was likely a

major contributor for the decrease in the number of liver transplantations for PBC, compared to

the unchanged number of liver transplantations for PSC, a similar chronic disease to PBC for

which there is no available treatment (Lee et al., 2007). Even when UDCA was available for the

treatment of PBC, it seems that the treatment regimen was not optimal, as inappropriate dosages

were administered in earlier decades that deviated from the currently recommended dosage of

13-15mg/kg/day.

Another aspect in the treatment regimen of PBC that has greatly changed over time is timing to

treatment. Although there was a substantial portion (88%) of patients who were diagnosed in the

1980s that received UDCA, they still experienced worse response rates and transplant-free

survival rates compared to patients diagnosed in more recent decades. Time to treatment was a

critical factor that influenced their outcomes since they received UDCA many years after their

diagnosis. In our analysis, the time from diagnosis to study entry was a significant predictor for

hepatic decompensation, HCC development, and transplant-free survival. In the time between

their diagnosis and the start of treatment, the disease had an opportunity to progress and therefore

the introduction of UDCA at this more advanced disease stage would not be as beneficial had it

been administered at an earlier disease stage (Parés et al., 2000; Shi et al., 2006). The time lag to

treatment administration is the greatest in the earlier cohorts and is likely attributable to the lack

of availability of UDCA. Overall, this suggests that the treatment regimen of PBC with UDCA

has improved considerably by the usage of more appropriate dosages and a prompt initiation

after diagnosis.

95

5.1.1.6 Clinical outcomes

As mentioned, there has been an increase in median age and the proportion of individuals ≥50

years old in the general population, which reflects an increase in life expectancy (median life)

over time. We showed that transplant-free survival increased over calendar time in PBC patients,

as is also expected in the general population. Therefore, we compared transplant-free survival

over time with respect to an age-, gender-, and birth year-matched general population. These

results indicated that the survival of PBC patients has improved over time even when compared

to the general population. An increase in survival over calendar time, especially when compared

to the general population, has not been previously shown in a globally representative population.

A Canadian study failed to show an improvement in survival from 1996 to 2002 most likely due

to the short period analyzed (Myers et al., 2009). A study from Finland that evaluated survival

from 1988 to 1999 reported improved survival, in which the age-, gender- and study area-

adjusted HR for all-cause mortality was 0.6 per a 10-year increase in year (Rautiainen et al.,

2007). An improved transplant-free survival may be potentially associated with the strongly

documented increase in the prevalence of in various geographical regions.

There was no difference in HCC incidence after the 1980s, however there was a decrease in

decompensation over time. This can be attributed the overall low incidence of HCC when

compared to decompensation. Therefore, one is unable to capture any differences due to the

small number of events.

5.1.1.7 Trends in primary sclerosing cholangitis

It is relevant to assess how the natural history of another cholestatic liver disease may have

evolved over time as compared to PBC. A Swedish study compared the clinical presentation of

patients with PSC between 1984 and 2004 with an emphasis on patients diagnosed before 1998

and after 1998. In the later cohort of patients diagnosed after 1998, they found an older age at

diagnosis (41 years old vs 37 years old), a lower frequency of symptoms (47% vs 63%), and a

lower rate of inflammatory bowel disease (IBD) (Bergquist et al., 2007). The lower age in the

earlier cohort was partially attributed to the higher proportion of patients with IBD, since these

patients undergo regular clinical check-ups. These results suggest that an increase in age may not

be specific to PBC and that it may also be applicable to other liver diseases. Furthermore, it

96

indicates that overall health care has improved, and patients with various liver diseases are being

diagnosed at an earlier disease stage without symptoms. A key difference between these two

diseases is that PBC exhibits improved outcomes relative to PSC, as suggested by the stable

number in liver transplantations for PSC, compared to a decrease in liver transplantations for

PBC.

5.1.2 Strengths and limitations

5.1.2.1 Strengths

There are strengths associated with this study that entail the population in which it was

performed, and the methodology used. The first strength of the GLOBAL PBC cohort is that it is

a multicenter cohort that includes centers through Europe and North America. This makes the

study unique from other studies that have also evaluated changes in patient characteristic because

the changes can be extrapolated to various geographical regions, rather than be limited to a

certain geographical region. Furthermore, due to the inclusion of patients dating back to the

1960s and their long-term follow-up, an analysis of a broad time period was possible, as well as

their respective rates of complications and outcomes. The 44-year period that was assessed is not

only the longest time period assessed for such a descriptive study, but it also spans the time prior

to UDCA and after UDCA, which allowed us to gain insight into how the introduction of this

treatment affected its natural history.

The survival of the general population has undoubtedly increased over time. Therefore, a simple

description of changes over time would not have been sufficient because it is expected that

survival has improved over time. Therefore, it was critical for the survival to be compared to that

of the general population to adequately assess whether survival indeed changed over time.

5.1.2.2 Limitations

Most epidemiological studies of PBC have been conducted in patients from Western countries

and less so in other populations, such as Asian or Hispanic populations. One of the limitations of

this study is also the limited ethnicity of the population, as the only centers included were from

Europe and North America. Therefore, it is still uncertain whether these changes are applicable

to other ethnic populations or from other geographical locations, especially in populations that

97

have been reported to have a more severe disease stage, such as African American and Hispanic

populations (Peters et al., 2007). Similar results are to be expected, however, as a study on the

temporal trends of a symptomatic Japanese population also reported an increase in age and

improved liver biochemistry at study entry (Sakauchi et al., 2007).

Another limitation of this study is that the association of the observed changes could not be

correlated to symptoms or other environmental factors that have been previously reported to be

associated with PBC, such as smoking, UTIs, and hormonal replacement therapy because these

variables were absent from the GLOBAL PBC database. It would be of relevance to assess their

potential association because it is possible that evolving habits in terms of these environmental

factors may have contributed to a delay in PBC presentation and its associated milder disease

severity.

5.1.3 Implications

We have provided a description of the presenting disease characteristics of a typical PBC patient

seen by physicians during a specified time frame. This in itself is useful because it proves that

the disease is not static and has evolved over time. It also documents the influence that UDCA

has had on the natural history of PBC, as well as the improved health care over time. This is

reassurance that the improvement of care for patients with PBC through an early diagnosis and

timely access to treatment can translate into improved outcomes for patients. Furthermore, it

emphasizes that there are various factors affecting the outcome in patients, including diagnosis

year, availability to treatment, time to treatment, dosage of UDCA, as well as baseline disease

characteristics.

As the disease has proven to not be static and evolving, the prognostic parameters used in the

clinic should be in line with the patients of today. For example, the Mayo model was developed

in a time when patients were at a symptomatic and advanced stage without available treatment.

This model is not applicable in the PBC population of today and has been found to underestimate

the survival of patients.

98

Bilirubin is one of the major predictors for prognosis, it is included in various prognostic models

and response criteria, and the number of patients presenting with normal bilirubin increased over

time, thus this biochemical parameter was further evaluated in the second study.

5.2 Bilirubin Within the Normal Range

5.2.1 Discussion

Up to date, the majority of studies have made an emphasis on normal bilirubin, as indicated by

the inclusion of this criterion for various response criteria, such as Paris-I, Paris-II, and

Rotterdam. Indeed, a study that evaluated various thresholds of bilirubin reported that the best

threshold for predicting liver transplantation or death was at the ULN but did not assess

thresholds below the ULN (Lammers et al., 2014). Bilirubin is not abnormal until later stages of

disease and thus, the predictive value of bilirubin is thought to be limited during early stages of

disease to a small number of patients. In our first study, we showed that there has been an

increasing proportion of patients that have normal bilirubin at study entry, and presumably an

even higher number after 1 year of treatment because of the effect of UDCA on bilirubin. A

recent clinical trial on OCA showed that bilirubin decreased even within the normal range and it

is unknown whether this translates to an improved outcome. We sought to determine whether

bilirubin levels below the ULN could be predictive of transplant-free survival.

When we evaluated the predictive value of bilirubin below the ULN, a bilirubin threshold of

0.6×ULN was established as the optimal threshold. Patients with bilirubin levels below this

threshold possess the lowest risk for liver transplantation or death, whereas the risk increases

linearly above this threshold. Furthermore, we show that in patients who are above the threshold

and subsequently experience a decrease in bilirubin to below the threshold as a result of

treatment with UDCA, there is an improvement in transplant-free survival. It was also suggested

that increasing bilirubin levels over time above this threshold may indicate the future

development of a poor clinical outcome.

5.2.1.1 Sensitivity of ULN of bilirubin for predicting outcome

Our results indicate that the current ULN for bilirubin may not be a sensitive indicator of risk in

PBC. One potential reason for this finding is that PBC is a female predominant disease, yet the

99

ULN is determined in the general population, whereby male bilirubin values may skew the ULN

as they tend to have higher bilirubin levels than females. Furthermore, we speculate that it may

have to do with the relative increase in bilirubin required to reach the ULN, given the modal

bilirubin level in a healthy population was 0.4mg/dL (Zucker et al., 2004). The ULN is usually

between 1-1.2mg/dL, which suggests an increase of 0.6-0.8mgdL would be required.

5.2.1.2 Bilirubin in the general population

There is a positive relationship with bilirubin and the risk for liver transplantation and death

beyond the 0.6× ULN threshold in PBC. Interestingly, there appears to be a different relationship

with bilirubin in individuals without liver disease, in whom increased bilirubin levels have been

associated with a decreased incidence of cardiovascular, rectal cancer, and diabetes (Levitt &

Levitt, 2014; Zucker et al., 2004). In an NHANES study of 4303 individuals 60 years old and

above from 1999 to 2004, individuals with bilirubin between 0.1-0.4mg/dL had the highest

mortality rates. In comparison to those with bilirubin of 0.5-0.7mg/dL, they had a 1.36-fold

increase in risk. Although individuals with bilirubin ≥0.8mg/dL also had higher mortality rates, it

was not statistically significant (Ong et al., 2014). The anti-inflammatory and antioxidant

properties of bilirubin may account for these differences. Furthermore, it is important to keep in

mind that total bilirubin is primarily composed of unconjugated bilirubin in these individuals as

compared to PBC patients, in which elevated bilirubin would be primarily composed of

conjugated bilirubin.

5.2.1.3 Risk stratification in PBC

There are various liver biochemistry parameters that may be utilized in PBC for risk stratification

of which ALP and bilirubin are the primary parameters (Lammers et al., 2014). These are readily

available as they are usually routinely checked in patients, and thus represent a primary means

for risk stratification. However, established response criteria rely on these biochemical

parameters in a dichotomized form, which may lead to a loss in predictive ability. Therefore, risk

stratification in PBC has recently shifted towards prognostic models that utilize various liver

biochemistries to predict transplant-free survival such as the GLOBE score (Lammers et al.,

2015). This score was shown to be superior to other response criteria, potentially because

bilirubin in addition to the other biochemical variables included were input into this model as a

100

continuous variable and it includes a variety of prognostic variables for PBC. Response criteria

in which normal bilirubin was a component would lose the increased risk imposed on patients

with normal bilirubin whose levels were above 0.6×ULN.

Biochemical parameters and prognostic models that rely on these biochemical parameters are the

preferred method for risk stratification as compared to liver biopsies. Although histological

staging provides important prognostic information, it is no longer routinely performed due to its

invasive nature. Transient elastography is a promising tool to assess liver fibrosis by measuring

liver stiffness, yet this may not be the best manner to assess response to therapy after a short

period of time, as changes in fibrosis may not be observed until longer follow-up takes place.

Therefore, liver biochemistry parameters represent an important manner of assessing prognosis

as they are easily attainable and can provide long-term prognostic information (Lammers et al.,

2015).

5.2.2 Strengths and limitations

5.2.2.1 Strengths

The strengths of this study include the large number of patients from a globally representative

population and long-term follow-up. In terms of methodology, the predictive ability of bilirubin

was not only assessed at baseline but also up to 4 years of follow-up, which confirms that the

bilirubin threshold can be applied at various independent time points. This is important because

it allows the implementation of the threshold before treatment, and after therapy to assess

whether the patient requires additional therapy. Indeed, we showed that a reduction in bilirubin

by treatment with UDCA can translate into an improved prognosis. The applicability of the

threshold was found in multiple subgroups, stratified according to age, histological stage, ALP,

and UDCA treatment, which emphasizes that the threshold is generalizable. Although the

threshold was not statistically significant in males, we attribute it to a lack of power due to the

small sample size.

Another strength of this study is that the risk imposed by bilirubin levels above 0.6×ULN was

interpreted relative to that associated with bilirubin above the ULN. This allowed us to

demonstrate that there was a positive relationship between bilirubin and the risk for liver

transplantation or death after 0.6×ULN that extended beyond the ULN. We also underline that

101

there is not only an increased risk for liver transplantation or death above 0.6×ULN, but that

the events are primarily liver-related in this range. Additionally, we were able to assess trends

in bilirubin over 5 years and its relationship with clinical events due to the imputation of

bilirubin. Imputation is a way to deal with missing data by replacing the missing values with an

estimate that is then analyzed as if they were observed values. It provides an unbiased way to

deal with missing data for analysis.

5.2.2.2 Limitations

The study has some limitations. In PBC, conjugated bilirubin is the form that is mainly elevated

and thus it would be beneficial to measure this type of bilirubin to directly measure the effect of

cholestasis on bilirubin levels (Levitt & Levitt, 2014). However, conjugated bilirubin was not

available and only total bilirubin was available in the GLOBAL PBC database. This is due to

the fact that clinical laboratories can only measure direct and indirect bilirubin through the

diazo reaction, in which direct bilirubin does not only include conjugated bilirubin but also

delta bilirubin. Therefore, in order to measure the conjugated bilirubin fraction separately, one

would need to implement other methods, such as high/performance liquid chromatography or

direct spectrophotometry, which are not ideal for routine use.

Furthermore, bilirubin was assessed relative to the ULN of each center, which was variable

across each center. Patients from centers that have a higher ULN may have accounted for the

increased events observed above 0.6×ULN. However, this limitation was overcome by

excluding those with an ULN above 1.2mg/dL and assessing crude bilirubin levels measured in

mg/dL.

5.2.3 Implications

Since patients with bilirubin between 0.6×ULN and 1×ULN are already at an increased risk for

liver transplantation or death, future intervention studies that assess the benefit of therapeutic

drugs may also include these patients since they may be able to benefit from additional therapies.

One of the clinical trials for OCA specified that patients must either have ALP≥1.67×ULN or

abnormal bilirubin (up to 2×ULN) to be included in the study (Nevens et al., 2016). Whereas, the

BEZURSO trial of bezafibrate adjuvant to UDCA indicated that patients included must be non-

responders to Paris-II criteria which also includes abnormal bilirubin (Corpechot et al., 2017).

102

We believe that patients whose bilirubin levels are above the threshold of 0.6×ULN may still

benefit from these therapeutic trials, and if the threshold is implemented, it can lead to an

increase in the number of patients that will become eligible for second-line therapies. This is of

relevance because only a minority of patients that are included in these trials have an abnormal

bilirubin. For example, in the OCA trial, 6% of patients had an abnormal bilirubin in the 5-10mg

group and 10% in 10mg group of OCA (Nevens et al., 2016). Our findings also give insight into

the implications of changes in bilirubin seen in these trials. Depending on the randomization

group, patients included had a mean bilirubin level of 0.69mg/dL (placebo), 0.60mg/dL (OCA

5-10mg), and 0.66mg/dL (OCA 10mg) at baseline. Patients randomized to receive OCA

experienced decreases in bilirubin, whereas the placebo group experienced an increase. Our

results suggest that decreases in bilirubin in the range from 1×ULN to 0.6×ULN are still

beneficial irrespective of whether patients attain the 0.6×ULN threshold. Additionally, in the

BEZURSO trial, mean bilirubin at study entry was 0.8mg/dL after which the median decrease in

patients receiving bezafibrate and UDCA after 2 years was 14% (Corpechot et al., 2017). In

light of these results from clinical trials assessing the benefit of second-line therapies for PBC,

we believe that our study grants insight into how bilirubin changes within the normal range

observed as a result of treatment correlate with transplant-free survival that goes beyond a

normalization of bilirubin being associated with improved prognosis.

Our study also has the implication of early determination of patients that may be at risk for a

future outcome. Currently, patients are not considered at risk if their bilirubin is normal, and thus

may be overlooked until it surpasses the normal range. This emphasizes that patients who are

above the 0.6×ULN threshold but experience gradual increases in bilirubin that remain within the

normal range may be at risk for liver transplantation or death and should be closely monitored.

103

Chapter 6

Conclusions 6The natural history of PBC has proven to be evolving over time and patients diagnosed in recent

decades have an increased age at diagnosis, a predominantly mild disease stage at presentation,

and improved clinical outcomes. Furthermore, we established that patients with bilirubin

≤0.6×ULN have the lowest risk for liver transplantation or death, from which point onward there

is a linear increase in risk.

104

Chapter 7

Future Directions 7Our study provides the most comprehensive evidence to date for a changing natural history of

PBC. However, there are still some questions that need exploration. One potential research

question that can be addressed is whether the changes observed in this population are also

observed in other populations, such as African Americans and Hispanics, in whom PBC tends to

have a more advanced disease stage. Furthermore, the increase in age of patients is peculiar

given that they present with an earlier disease stage. Therefore, the potential influence of

changing environmental factors on the increase in age can be explored in future studies, such as

smoking.

The natural history of PBC has changed primarily due to the introduction of UDCA as the

majority of changes were observed thereafter. The introduction of a new treatment for PBC may

drive further changes. Therefore, it would be interesting to determine how the natural history of

PBC may change in the future with the introduction of OCA or other therapeutic agents and

whether it may impose greater improvements on transplant-free survival.

Bilirubin was found to be an independent predictor of transplant-free survival even within the

normal range. The fact that the normalization of bilirubin is not associated with an absence of

risk raises the question of whether the same may be true for other biochemical parameters such

as ALP. A future study may assess the predictive value of ALP below 1.67×ULN in UDCA-

treated patients, as current studies use this threshold for inclusion in clinical trials and as part of

the primary endpoint. An elevation of ALP is necessary for a diagnosis of PBC, therefore its

predictive ability below 1.67×ULN need be assessed starting after 1 year of treatment, at which

point decreases in ALP are observed.

It may be that bilirubin imposes different risks in the general population compared to PBC

patients. In the general population, it seems that low conjugated bilirubin levels are associated

with an increased risk for mortality. Therefore, it would be of interest to compare mortality of

patients with PBC to that of the general population while adjusting for bilirubin and stratifying

conjugated and unconjugated bilirubin to gain further insight into the role of bilirubin in PBC.

105

References Abdulkarim, A. S., Petrovic, L. M., Kim, W. R., Angulo, P., Lloyd, R. V, & Lindor, K. D.

(2004). Primary biliary cirrhosis: an infectious disease caused by Chlamydia pneumoniae? Journal of Hepatology, 40(3), 380–384.

Aboutwerat, A., Pemberton, P. W., Smith, A., Burrows, P. C., McMahon, R. F., Jain, S. K., & Warnes, T. W. (2003). Oxidant stress is a significant feature of primary biliary cirrhosis. Biochimica et Biophysica Acta, 163(2), 142–150.

Abu-Mouch, S., Selmi, C., Benson, G. D., Kenny, T., Invernizzi, P., Zuin, M., … Gershwin, M. E. (2003). Geographic clusters of primary biliary cirrhosis. Clinical and Developmental Immunology, 10(2–4), 127–131.

Al-Harthy, N., & Kumagi, T. (2012). Natural history and management of primary biliary cirrhosis. Hepatic Medicine: Evidence and Research, 4, 61–71.

Ala, A., Stanca, C. M., Bu-Ghanim, M., Ahmado, I., Branch, A. D., Schiano, T. D., … Bach, N. (2006). Increased prevalence of primary biliary cirrhosis near Superfund toxic waste sites. Hepatology, 43(3), 525–531.

Alempijevic, T., Krstic, M., Jesic, R., Jovanovic, I., Sokic Milutinovic, A., Kovacevic, N., … Popovic, D. (2009). Biochemical markers for non-invasive assessment of disease stage in patients with primary biliary cirrhosis. World Journal of Gastroenterology, 15(5), 591–594.

Almasio, P. L., Floreani, A., Chiaramonte, M., Provenzano, G., Battezzati, P., Crosignani, A., … Craxì, A. (2000). Multicentre randomized placebo-controlled trial of ursodeoxycholic acid with or without colchicine in symptomatic primary biliary cirrhosis. Alimentary Pharmacology & Therapeutics, 14(12), 1645–1652.

Amano, K., Leung, P. S., Rieger, R., Quan, C., Wang, X., Marik, J., … Gershwin, M. E. (2005). Chemical xenobiotics and mitochondrial autoantigens in primary biliary cirrhosis: identification of antibodies against a common environmental, cosmetic, and food additive, 2-octynoic acid. Journal of Immunology, 174(9), 5874–5883.

Angulo, P., Dickson, E. R., Therneau, T. M., Jorgensen, R. A., Smith, C., DeSotel, C. K., … Lindor, K. D. (1999). Comparison of three doses of ursodeoxycholic acid in the treatment of primary biliary cirrhosis: a randomized trial. Journal of Hepatology, 30(5), 830–835.

Angulo, P., Lindor, K. D., Therneau, T. M., Jorgensen, R. A., Malinchoc, M., Kamath, P. S., & Dickson, E. R. (1999). Utilization of the Mayo risk score in patients with primary biliary cirrhosis receiving ursodeoxycholic acid. Liver, 19(2), 115–121.

106

Azemoto, N., Abe, M., Murata, Y., Hiasa, Y., Hamada, M., Matsuura, B., & Onji, M. (2009). Early biochemical response to ursodeoxycholic acid predicts symptom development in patients with asymptomatic primary biliary cirrhosis. Journal of Gastroenterology, 44(6), 630–634.

Bach, N., & Schaffner, F. (1994). Familial primary biliary cirrhosis. Journal of Hepatology, 20(6), 698–701.

Balasubramaniam, K., Grambsch, P. M., Wiesner, R. H., Lindor, K. D., & Dickson, E. R. (1990). Diminished survival in asymptomatic primary biliary cirrhosis. A prospective study. Gastroenterology, 98(6), 1567–1571.

Baldursdottir, T., Bergmann, O. M., Jonasson, J. G., Ludviksson, B. R., Axelsson, T. A., & Björnsson, E. S. (2012). The epidemiology and natural history of primary biliary cirrhosis: a nationwide population-based study. European Journal of Gastroenterology and Hepatology, 24(7), 824–830.

Baldursdottir, T. R., Bergmann, O. M., Jonasson, J. G., Ludviksson, B. R., Axelsson, T. A., & Björnsson, E. S. (2012). The epidemiology and natural history of primary biliary cirrhosis: a nationwide population-based study. European Journal of Gastroenterology and Hepatology, 24(7), 824–830.

Ballardini, G., Mirakian, R., Bianchi, F. B., Pisi, E., Doniach, D., & Bottazzo, G. F. (1984). Aberrant expression of HLA-DR antigens on bileduct epithelium in primary biliary cirrhosis: relevance to pathogenesis. Lancet, 2(8410), 1009–1013.

Battezzati, P. M., Podda, M., Bianchi, F. B., Naccarato, R., Orlandi, F., Surrenti, C., … Manenti, F. (1993). Ursodeoxycholic acid for symptomatic primary biliary cirrhosis. Preliminary analysis of a double-blind multicenter trial. Italian Multicenter Group for the Study of UDCA in PBC. Journal of Hepatology, 17(3), 332–338.

Bergquist, A., Said, K., & Broomé, U. (2007). Changes over a 20-year period in the clinical presentation of primary sclerosing cholangitis in Sweden. Scandinavian Journal of Gastroenterology, 42(1), 88–93.

Bittencourt, P. L., Farias, A. Q., Abrantes-Lemos, C P Goncalves, L. L., Goncalves, P. L., Magalhães, E. P., Carrilho, F. J., … Cançado, E. L. (2004). Prevalence of immune disturbances and chronic liver disease in family members of patients with primary biliary cirrhosis. Journal of Gastroenterology and Hepatology, 19(8), 873–878.

Bogdanos, D. P., Baum, H., Grasso, A., Okamoto, M., Butler, P., Ma, Y., … Vergani, D. (2004). Microbial mimics are major targets of crossreactivity with human pyruvate dehydrogenase in primary biliary cirrhosis. Journal of Hepatology, 40(1), 31–39.

Bonnand, A. M., Heathcote, E. J., Lindor, K. D., & Poupon, R. E. (1999). Clinical significance of serum bilirubin levels under ursodeoxycholic acid therapy in patients with primary biliary cirrhosis. Hepatology, 29(1), 39–43.

107

Boonstra, K., Beuers, U., & Ponsioen, C. Y. (2012). Epidemiology of primary sclerosing cholangitis and primary biliary cirrhosis: a systematic review. Journal of Hepatology, 56(5), 1181–1188.

Boonstra, K., Kunst, A. E., Stadhouders, P. H., Tuynman, H. A., Poen, A. C., van Nieuwkerk, K. M., … Epi PSC PBC study group. (2014). Rising incidence and prevalence of primary biliary cirrhosis: a large population-based study. Liver International, 34(6), e31–e38.

Bosch, A., Dumortier, J., Maucort-Boulch, D., Scoazec, J. Y., Wendum, D., Conti, F., … Corpechot, C. (2015). Preventive administration of UDCA after liver transplantation for primary biliary cirrhosis is associated with a lower risk of disease recurrence. Journal of Hepatology, 63(6), 1449–1458.

Bosma, P. J., Roy Chowdhury, J., Bakker, C., Gantla, S., De Boer, A., Oostra, B. A., … Roy Chowdhury, N. (1995). The genetic basis of the reduced expression of bilirubin UDP-glucuronosyltransferase 1 in gilbert’s syndrome. New England Journal of Medicine, 333(18), 1171–1175.

Bown, R., Clark, M. L., & Doniach, D. (1975). Primary biliary cirrhosis in brothers. Postgraduate Medical Journal, 51(592), 110–115.

Carbone, M., Mells, G. F., Pells, G., Dawwas, M. F., Newton, J. L., Heneghan, M. A., … Jones, D. E. (2013). Sex and age are determinants of the clinical phenotype of primary biliary cirrhosis and response to ursodeoxycholic acid. Gastroenterology, 144(3), 560-569.e7; quiz e13-4.

Carbone, M., Sharp, S. J., Flack, S., Paximadas, D., Spiess, K., Adgey, C., … Kordul, J. (2016). 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, 63(3), 930–950.

Cash, W. J., McCance, D., Young, I. S., McEneny, J., Cadden, I. S., Mcdougall, N. I., & Callender, M. E. (2010). Primary biliary cirrhosis is associated with oxidative stress and endothelial dysfunction but not increased cardiovascular risk. Hepatology Research, 40, 1098–1106.

Cauch-Dudek, K., Abbey, S., Stewart, D. E., & Heathcote, E. J. (1998). Fatigue in primary biliary cirrhosis. Gut, 43(5), 705–710.

Charatcharoenwitthaya, P., Pimentel, S., Talwalkar, J. A., Enders, F. T., Lindor, K. D., Krom, R. A., & Wiesner, R. H. (2007). Long-term survival and impact of ursodeoxycholic acid treatment for recurrent primary biliary cirrhosis after liver transplantation. Liver Transplantation, 13(9), 1236–1245.

Chohan, M. R. (1973). Primary biliary cirrhosis in twin sisters. Gut, 14(3), 213–214.

108

Christensen, E., Crowe, J., Doniach, D., Popper, H., Ranek, L., Rodés, J., … Williams, R. (1980). Clinical pattern and course of disease in primary biliary cirrhosis based on an analysis of 236 patients. Gastroenterology, 78(2), 236–246.

Combes, B., Emerson, S. S., Flye, N. L., Munoz, S. J., Luketic, V. A., Mayo, M. J., … Hofmann, A. F. (2005). Methotrexate (MTX) plus ursodeoxycholic acid (UDCA) in the treatment of primary biliary cirrhosis. Hepatology, 42(5), 1184–1193.

Corpechot, C., Abenavoli, L., Rabahi, N., Chretien, Y., Andreani, T., Johanet, C., … Poupon, R. (2008). Biochemical response to ursodeoxycholic acid and long-term prognosis in primary biliary cirrhosis. Hepatology, 48(3), 871–877.

Corpechot, C., Carrat, F., Bahr, A., Chrétien, Y., Poupon, R.-E., & Poupon, R. (2005). The effect of ursodeoxycholic acid therapy on the natural course of primary biliary cirrhosis. Gastroenterology, 128(2), 297–303.

Corpechot, C., Carrat, F., Poupon, R., & Poupon, R. E. (2002). Primary biliary cirrhosis: incidence and predictive factors of cirrhosis development in ursodiol-treated patients. Gastroenterology, 122(3), 652–658.

Corpechot, C., Chazouillères, O., & Poupon, R. (2011). Early primary biliary cirrhosis: Biochemical response to treatment and prediction of long-term outcome. Journal of Hepatology, 55(6), 1361–1367.

Corpechot, C., Chazouillères, O., Rousseau, A., Guyader, D., Habersetzer, F., Mathurin, P., … Poupon, R. (2017). A 2-year multicenter, double-blind, randomized, placebo-controlled study of bezafibrate for the treatment of primary biliary cholangitis in patients with inadequate biochemical response to ursodeoxycholic acid therapy (Bezurso). Journal of Hepatology, 66(1), S89.

Corpechot, C., Chrétien, Y., Chazouillères, O., & Poupon, R. (2010). Demographic, lifestyle, medical and familial factors associated with primary biliary cirrhosis. Journal of Hepatology, 53(1), 162–169.

Corpechot, C., Gaouar, F., Chrétien, Y., Johanet, C., Chazouillères, O., & Poupon, R. (2012). Smoking as an independent risk factor of liver fibrosis in primary biliary cirrhosis. Journal of Hepatology, 56(1), 218–224.

Crosignani, A., Podda, M., Battezzati, P. M., Bertolini, E., Zuin, M., Watson, D., & Setchell, K. D. (1991). Changes in bile acid composition in patients with primary biliary cirrhosis induced by ursodeoxycholic acid administration. Hepatology, 14(6), 1000–1007.

D’Amico, G., & Luca, A. (1997). Natural history. Clinical-haemodynamic correlations. Prediction of the risk of bleeding. Bailliere’s Clinical Gastroenterology, 11(2), 243–256.

Dähnrich, C., Pares, A., Caballeria, L., Rosemann, A Schlumberger, W., Probst, C., Mytilinaiou, M., … Komorowski, L. (2009). New ELISA for detecting primary biliary cirrhosis-specific antimitochondrial antibodies. Clinical Chemistry, 55(5), 978–985.

109

Deutsch, M., Papatheodoridis, G. V, Tzakou, A., & Hadziyannis, S. J. (2008). Risk of hepatocellular carcinoma and extrahepatic malignancies in primary biliary cirrhosis. European Journal of Gastroenterology and Hepatology, 20(1), 5–9.

Dickson, E. R., Grambsch, P. M., Fleming, T. R., Fisher, L. D., & Langworthy, A. (1989). Prognosis in primary biliary cirrhosis: model for decision making. Hepatology, 10(1), 1–7.

Donaldson, P. T., Baragiotta, A., Heneghan, M. A., Floreani, A., Venturi, C., Underhill, J. A., … Bassendine, M. F. (2006). HLA class II alleles, genotypes, haplotypes, and amino acids in primary biliary cirrhosis: a large-scale study. Hepatology, 44(3), 667–674.

Dyson, J. K., Wilkinson, N., Jopson, L., Mells, G., Bathgate, A., Heneghan, M. A., … Jones, D. E. (2016). The inter-relationship of symptom severity and quality of life in 2055 patients with primary biliary cholangitis. Alimentary Pharmacology & Therapeutics, 44(10), 1039–1050.

Eriksson, S., & Lindgren, S. (1984). The prevalence and clinical spectrum of primary biliary cirrhosis in a defined population. Scandinavian Journal of Gastroenterology, 19(7), 971–976.

European Association for the Study of the Liver. (2009). EASL Clinical Practice Guidelines: management of cholestatic liver diseases. Journal of Hepatology, 51(2), 237–267.

Fagan, E., Williams, R., & Cox, S. (1977). Primary biliary cirrhosis in mother and daughter. British Medical Journal, 2(6096), 1195.

Fairweather, D., & Rose, N. R. (2004). Women and autoimmune diseases. Emerging Infectious Diseases, 10(11), 2005–2011.

Feizi, T., Naccarato, R., Sherlock, S., & Doniach, D. (1972). Mitochondrial and other tissue antibodies in relatives of patients with primary biliary cirrhosis. Clinical and Experimental Immunology, 10(4), 609–622.

Flannery, G. R., Burroughs, A. K., Butler, P., Chelliah, J., Hamilton-Miller, J., Brumfitt, W., & Baum, H. (1989). Antimitochondrial antibodies in primary biliary cirrhosis recognize both specific peptides and shared epitopes of the M2 family of antigens. Hepatology, 10(3), 370–374.

Floreani, A., Caroli, D., Variola, A., Rizzotto, E. R., Antoniazzi, S., Chiaramonte, M., … Baldo, V. (2011). A 35-year follow-up of a large cohort of patients with primary biliary cirrhosis seen at a single centre. Liver International, 31(3), 361–368.

Gabeta, S., Norman, G. L., Liaskos, C., Papamichalis, P. A., Zografos, T., Garagounis, A., … Dalekos, G. N. (2007). Diagnostic relevance and clinical significance of the new enhanced performance M2 (MIT3) ELISA for the detection of IgA and IgG antimitochondrial antibodies in primary biliary cirrhosis. Journal of Clinical Immunology, 27(4), 378–387.

110

Gatselis, N. K., Zachou, K., Norman, G. L., Gabeta, S., Papamichalis, P., Koukoulis, G. K., & Dalekos, G. N. (2013). Clinical significance of the fluctuation of primary biliary cirrhosis-related autoantibodies during the course of the disease. Autoimmunity, 46(7), 471–479.

Gershwin, M. E., Selmi, C., Worman, H. J., Gold, E. B., Watnik, M., Utts, J., … USA PBC Epidemiology Group. (2005). Risk factors and comorbidities in primary biliary cirrhosis: a controlled interview-based study of 1032 patients. Hepatology, 42(5), 1194–1202.

Gómez-Dominguez, E., Mendoza, J., García-Buey, L., Trapero, M., Gisbert, J. P., Jones, E. A., & Moreno-Otero, R. (2008). Transient elastography to assess hepatic fibrosis in primary biliary cirrhosis. Alimentary Pharmacology & Therapeutics, 27(5), 441–447.

Gong, Y., Christensen, E., & Gluud, C. (2007). Azathioprine for primary biliary cirrhosis. The Cochrane Database of Systematic Reviews, (3), CD006000.

González-Koch, A., Brahm, J., Antezana, C., Smok, G., & Cumsille, M. A. (1997). The combination of ursodeoxycholic acid and methotrexate for primary biliary cirrhosis is not better than ursodeoxycholic acid alone. Journal of Hepatology, 27(1), 143–149.

Gores, G. J., Wiesner, R. H., Dickson, E. R., Zinsmeister, A. R., Jorgensen, R. A., & Langworthy, A. (1989). Prospective evaluation of esophageal varices in primary biliary cirrhosis: development, natural history, and influence on survival. Gastroenterology, 96(6), 1552–1559.

Granito, A., Muratori, P., Muratori, L., Pappas, G., Cassani, F., Worthington, J., … Bianchi, F. B. (2006). Antinuclear antibodies giving the “multiple nuclear dots” or the “rim-like/membranous” patterns: diagnostic accuracy for primary biliary cirrhosis. Alimentary Pharmacology & Therapeutics, 24(11–12), 1575–1583.

Granito, A., Muratori, P., Quarneti, C., Pappas, G., Cicola, R., & Muratori, L. (2012). Antinuclear antibodies as ancillary markers in primary biliary cirrhosis. Expert Review of Molecular Diagnostics, 12(1), 65–74.

Gregory, W. L., Mehal, W., Dunn, A. N., Cavanagh, G., Chapman, R., Fleming, K. A., … Bassendine, M. F. (1993). Primary biliary cirrhosis: contribution of HLA class II allele DR8. The Quarterly Journal of Medicine, 86(6), 393–399.

Gross, R. G., & Odin, J. A. (2008). Recent advances in the epidemiology of primary biliary cirrhosis. Clinics in Liver Disease, 12(2), 289–303.

Hayase, Y., Iwasaki, S., Akisawa, N., Saibara, T., Kadokawa, Y., Omagari, K., … Onishi, S. (2005). Similar anti-mitochondrial antibody reactivity profiles in familial primary biliary cirrhosis. Hepatology Research, 33(1), 33–38.

Heathcote, E. J., Cauch-Dudek, K., Walker, V., Bailey, R. J., Blendis, L. M., Ghent, C. N., … Wanless, I. R. (1994). The Canadian multicenter double-blind randomized controlled trial of ursodeoxycholic acid in primary biliary cirrhosis. Hepatology, 19(5), 1149–1156.

111

Hirschfield, G. M., Beuers, U., Corpechot, C., Invernizzi, P., Jones, D., Marzioni, M., & Schramm, C. (2017). EASL Clinical Practice Guidelines : The diagnosis and management of patients with primary biliary cholangitis. Journal of Hepatology, 67(1), 145–172.

Hirschfield, G. M., Mason, A., Luketic, V., Lindor, K., Gordon, S. C., Mayo, M., … Shapiro, D. (2015). Efficacy of obeticholic acid in patients with primary biliary cirrhosis and inadequate response to ursodeoxycholic acid. Gastroenterology, 148(4), 751–761.e8.

Howel, D., Fischbacher, C. M., Bhopal, R. S., Gray, J., Metcalf, J. V, & James, O. F. (2000). An exploratory population-based case-control study of primary biliary cirrhosis. Hepatology, 31(5), 1055–1060.

Hu, C. J., Song, G., Huang, W., Liu, G.-Z., Deng, C.-W., Zeng, H.-P., … Li, Y.-Z. (2012). Identification of new autoantigens for primary biliary cirrhosis using human proteome microarrays. Molecular & Cellular Proteomics, 11(9), 669–680.

Hu, S., Zhao, F., Wang, Q., & Chen, W.-X. (2014). The accuracy of the anti-mitochondrial antibody and the M2 subtype test for diagnosis of primary biliary cirrhosis: A meta-analysis. Clinical Chemistry and Laboratory Medicine, 52(11), 1533–1542.

Hubscher, S. G., Elias, E., Buckels, J. A., Mayer, A. D., McMaster, P., & Neuberger, J. M. (1993). Primary biliary cirrhosis. Histological evidence of disease recurrence after liver transplantation. Journal of Hepatology, 18(2), 173–184.

Huet, P. M., Deslauriers, J., Tran, A., Faucher, C., & Charbonneau, J. (2000). Impact of fatigue on the quality of life of patients with primary biliary cirrhosis. American Journal of Gastroenterology, 95(3), 760–767.

Invernizzi, P., Battezzati, P. M., Crosignani, A., Perego, F., Poli, F., Morabito, A., … Podda, M. (2003). Peculiar HLA polymorphisms in Italian patients with primary biliary cirrhosis. Journal of Hepatology, 38(4), 401–406.

Invernizzi, P., Crosignani, A., Battezzati, P. M., Covini, G., De Valle, G., Larghi, A., … Podda, M. (1997). Comparison of the clinical features and clinical course of antimitochondrial antibody-positive and -negative primary biliary cirrhosis. Hepatology, 25(5), 1090–1095.

Jackson, H., Solaymani-Dodaran, M., Card, T. R., Aithal, G. P., Logan, R., & West, J. (2007). Influence of ursodeoxycholic acid on the mortality and malignancy associated with primary biliary cirrhosis: a population-based cohort study. Hepatology, 46(4), 1131–1137.

Jacoby, A., Rannard, A., Buck, D., Bhala, N., Newton, J. L., James, O. F., & Jones, D. E. (2005). Development, validation, and evaluation of the PBC-40, a disease specific health related quality of life measure for primary biliary cirrhosis. Gut, 54(11), 1622–1629.

James, O. F., Bhopal, R., Howel, D., Gray, J., Burt, A. D., & Metcalf, J. V. (1999). Primary biliary cirrhosis once rare, now common in the United Kingdom? Hepatology, 30(2), 390–394.

112

Jones, D. E., Al-Rifai, A., Frith, J., Patanwala, I., & Newton, J. L. (2010). The independent effects of fatigue and UDCA therapy on mortality in primary biliary cirrhosis: Results of a 9 year follow-up. Journal of Hepatology, 53(5), 911–917.

Kaplan, M. M., & Gershwin, M. E. (2005). Primary biliary cirrhosis. New England Journal of Medicine, 353(12), 1261–1273.

Kaplan, M. M., & Gershwin, M. E. (2016). Primary biliary cirrhosis. New England Journal of Medicine, 353(12), 1261–1273.

Kim, K. A., Ki, M., Choi, H. Y., Kim, B. H., Jang, E. S., & Jeong, S. H. (2016). Population-based epidemiology of primary biliary cirrhosis in South Korea. Alimentary Pharmacology and Therapeutics, 43(1), 154–162.

Kim, W. R., Lindor, K. D., Locke, G. R. 3rd, Therneau, T. M., Homburger, H. A., Batts, K. P., … Dickson, E. R. (2000). Epidemiology and natural history of primary biliary cirrhosis in a US community. Gastroenterology, 119(6), 1631–1636.

Kim, W. R., Poterucha, J. J., Jorgensen, R. A., Batts, K. P., Homburger, H. A., Dickson, E. R., … Lindor, K. D. (1997). Does antimitochondrial antibody status affect response to treatment in patients with primary biliary cirrhosis? Outcomes of ursodeoxycholic acid therapy and liver transplantation. Hepatology, 26(1), 22–26.

Kim, W. R., Wiesner, R. H., Therneau, T. M., Poterucha, J. J., Porayko, M. K., Evans, R. W., … Dickson, E. R. (1998). Optimal timing of liver transplantation for primary biliary cirrhosis. Hepatology, 28(1), 33–38.

Kita, H., He, X. S., & Gershwin, M. E. (2004). Autoimmunity and environmental factors in the pathogenesis of primary biliary cirrhosis. Annals of Medicine, 36(1), 72–80.

Kita, H., Matsumura, S., He, X. S., Ansari, A. A., Lian, Z. X., Van de Water, J., … Gershwin, M. E. (2002). Quantitative and functional analysis of PDC-E2-specific autoreactive cytotoxic T lymphocytes in primary biliary cirrhosis. The Journal of Clinical Investigation, 109(9), 1231–1240.

Krige, J. E., & Beckingham, I. J. (2001). ABC of diseases of liver, pancreas, and biliary system: portal hypertensions-2. Ascites, encephalopathy, and other conditions. BMJ, 322(7283), 416–418.

Krzeski, P., Zych, W., Kraszewska, E., Milewski, B., Butruk, E., & Habior, A. (1999). Is serum bilirubin concentration the only valid prognostic marker in primary biliary cirrhosis? Hepatology, 30(4), 865–869.

Kubota, J., Ikeda, F., Terada, R., Kobashi, H., Fujioka, S., Okamoto, R., … Yamamoto, K. (2009). Mortality rate of patients with asymptomatic primary biliary cirrhosis diagnosed at age 55 years or older is similar to that of the general population. Journal of Gastroenterology, 44(9), 1000–1006.

113

Kuiper, E. M., Hansen, B. E., de Vries, R. A., den Ouden-Muller, J. W., van Ditzhuijsen, T. J., Haagsma, E. B., … Dutch PBC Study Group. (2009). Improved prognosis of patients with primary biliary cirrhosis that have a biochemical response to ursodeoxycholic acid. Gastroenterology, 136(4), 1281–1287.

Kuiper, E. M. M., Hansen, B. E., Adang, R. P. R., van Nieuwkerk, C. M. J., Timmer, R., Drenth, J. P. H., … van Buuren, H. R. (2010). Relatively high risk for hepatocellular carcinoma in patients with primary biliary cirrhosis not responding to ursodeoxycholic acid. European Journal of Gastroenterology and Hepatology, 22, 1495–1502.

Kuiper, E. M. M., Hansen, B. E., Metselaar, H. J., Man, R. A. De, Haagsma, E. B., & Hoek, B. Van. (2010). Trends in liver transplantation for primary biliary cirrhosis in the Netherlands 1988-2008. BMC Gastroenterology, 10(1), 144.

Kumagi, T., Guindi, M., Fischer, S. E., Arenovich, T., Abdalian, R., Coltescu, C., … Hirschfield, G. M. (2010). Baseline ductopenia and treatment response predict long-term histological progression in primary biliary cirrhosis. American Journal of Gastroenterology, 105(10), 2186–2194.

Kumagi, T., & Heathcote, E. J. (2008). Primary biliary cirrhosis. Orphanet Journal of Rare Diseases, 3, 1.

Lammers, W. J., Hirschfield, G. M., Corpechot, C., Nevens, F., Lindor, K. D., Janssen, H. L. A., … van Buuren, H. R. (2015). Development and validation of a scoring system to predict outcomes of patients with primary biliary cirrhosis receiving ursodeoxycholic acid therapy. Gastroenterology, 149(7), 1804–1812.e4.

Lammers, W. J., Leeman, M., Ponsioen, C. I., Boonstra, K., van Erpecum, K J Wolfhagen, F H Kuyvenhoven, J. P., Vrolijk, J. M., … Dutch Multicenter PBC Study Group. (2016). How the concept of biochemical response influenced the management of primary biliary cholangitis over time. The Netherlands Journal of Medicine, 74(6), 240–246.

Lammers, W. J., van Buuren, H. R., Hirschfield, G. M., Janssen, H. L. A., Invernizzi, P., Mason, A. L., … Hansen, B. E. (2014). Levels of alkaline phosphatase and bilirubin are surrogate end points of outcomes of patients with primary biliary cirrhosis: An international follow-up study. Gastroenterology, 147(6), 1338–1349.e5.

Lazaridis, K. N., Juran, B. D., Boe, G. M., Slusser, J. P., de Andrade, M., Homburger, H. A., … Petersen, G. M. (2007). Increased prevalence of antimitochondrial antibodies in first-degree relatives of patients with primary biliary cirrhosis. Hepatology, 46(3), 785–792.

Lee, J., Belanger, A., Doucette, J. T., Stanca, C., Friedman, S., & Bach, N. (2007). Transplantation trends in primary biliary cirrhosis. Clinical Gastroenterology and Hepatology, 5(11), 1313–1315.

Leuschner, M., Güldütuna, S., You, T., Hübner, K., Bhatti, S., & Leuschner, U. (1996). Ursodeoxycholic acid and prednisolone versus ursodeoxycholic acid and placebo in the treatment of early stages of primary biliary cirrhosis. Journal of Hepatology, 25(1), 49–57.

114

Leuschner, U., Fischer, H., Kurtz, W., Güldütuna, S., Hübner, K., Hellstern, A., … Leuschner, M. (1989). Ursodeoxycholic acid in primary biliary cirrhosis: results of a controlled double-blind trial. Gastroenterology, 97(5), 1268–1274.

Levitt, D. G., & Levitt, M. D. (2014). Quantitative assessment of the multiple processes responsible for bilirubin homeostasis in health and disease. Clinical and Experimental Gastroenterology, 7, 307–328.

Levy, C., Naik, J., Giordano, C., Mandalia, A., O’Brien, C., Bhamidimarri, K. R., … Martin, P. (2014). Hispanics with primary biliary cirrhosis are more likely to have features of autoimmune hepatitis and reduced response to ursodeoxycholic acid than non-Hispanics. Clinical Gastroenterology and Hepatology, 12(8), 1398–1405.

Levy, C., Zein, C. O., Gomez, J., Soldevila-Pico, C., Firpi, R., Morelli, G., & Nelson, D. (2007). Prevalence and predictors of esophageal varices in patients with primary biliary cirrhosis. Clinical Gastroenterology and Hepatology, 5(7), 803–808.

Liang, Y., Yang, Z., & Zhong, R. (2012). Primary biliary cirrhosis and cancer risk: a systematic review and meta-analysis. Hepatology, 56(4), 1409–1417.

Liermann Garcia, R. F., Evangelista Garcia, C., McMaster, P., & Neuberger, J. (2001). Transplantation for primary biliary cirrhosis: retrospective analysis of 400 patients in a single center. Hepatology, 33(1), 22–27.

Lindor, K. (2007). Ursodeoxycholic acid for the treatment of primary biliary cirrhosis. New England Journal of Medicine, 357(15), 1524–1529.

Lindor, K. D., Dickson, E. R., Baldus, W. P., Jorgensen, R. A., Ludwig, J., Murtaugh, P. A., … Hofmann, A. F. (1994). Ursodeoxycholic acid in the treatment of primary biliary cirrhosis. Gastroenterology, 106(5), 1284–1290.

Lindor, K. D., Gershwin, M. E., Poupon, R., Kaplan, M., Bergasa, N. V., & Heathcote, E. J. (2009). Primary biliary cirrhosis. Hepatology, 50(1), 291–308.

Lindor, K. D., Jorgensen, R. A., Therneau, T. M., Malinchoc, M., & Dickson, E. R. (1997). Ursodeoxycholic acid delays the onset of esophageal varices in primary biliary cirrhosis. Mayo Clinic Proceedings, 72(12), 1137–1140.

Lindor, K. D., Therneau, T. M., Jorgensen, R. A., Malinchoc, M., & Dickson, E. R. (1996). Effects of ursodeoxycholic acid on survival in patients with primary biliary cirrhosis. Gastroenterology, 110(5), 1515–1518.

Lleo, A., Jepsen, P., Morenghi, E., Carbone, L., Moroni, M., Battezzati, P., … Invernizzi, P. (2016). Evolving trends in female to male incidence and male mortality of primary biliary cholangitis. Scientific Reports, 6, 25906.

Locke, G. R. 3rd, Therneau, T. M., Ludwig, J., Dickson, E. R., & Lindor, K. D. (1996). Time course of histological progression in primary biliary cirrhosis. Hepatology, 23(1), 52–56.

115

Löhr, H., Fleischer, B., Gerken, G., Yeaman, S. J., Meyer zum Büschenfelde, K., & Manns, M. (1993). Autoreactive liver-infiltrating T cells in primary biliary cirrhosis recognize inner mitochondrial epitopes and the pyruvate dehydrogenase complex. Journal of Hepatology, 18(3), 322–327.

Long, R. G., Scheuer, P. J., & Sherlock, S. (1977). Presentation and course of asymptomatic primary biliary cirrhosis. Gastroenterology, 72(6), 1204–1207.

Ludwig, J., Dickson, E. R., & McDonald, G. S. A. (1978). Staging of chronic nonsuppurative destructive cholangitis (Syndrome of primary biliary cirrhosis). Virchows Archiv A Pathological Anatomy and Histology, 379(2), 103–112.

MacSween, R. N., Horne, C. H., Moffat, A. J., & Hughes, H. M. (1972). Serum protein levels in primary biliary cirrhosis. Journal of Clinical Pathology, 25(9), 789–792.

Mahl, T. C., Shockcor, W., & Boyer, J. L. (1994). Primary biliary cirrhosis: survival of a large cohort of symptomatic and asymptomatic patients followed for 24 years. Journal of Hepatology, 20(6), 707–713.

Manns, M. P., Bremm, A., Schneider, P. M., Notghi, A., Gerken, G., Prager-Eberle, M Stradmann-Bellinghausen, B., … Rittner, C. (1991). HLA DRw8 and complement C4 deficiency as risk factors in primary biliary cirrhosis. Gastroenterology, 101(5), 1367–1373.

Mantaka, A., Koulentaki, M., Chlouverakis, G., Enele-Melono, J. M., Darivianaki, A., Tzardi, M., & Kouroumalis, E. A. (2012). Primary biliary cirrhosis in a genetically homogeneous population: disease associations and familial occurrence rates. BMC Gastroenterology Gastroenterology, 12, 110.

Markus, B. H., Dickson, E. R., Grambsch, P. M., Fleming, T. R., Mazzaferro, V., Klintmalm, G. B., … Starzl, T. E. (1989). Efficacy of liver transplantation in patients with primary biliary cirrhosis. New England Journal of Medicine, 320(26), 1709–1713.

Matsumura, S., Van De Water, J., Leung, P., Odin, J. A., Yamamoto, K., Gores, G. J., … Gershwin, M. E. (2004). Caspase induction by IgA antimitochondrial antibody: IgA-mediated biliary injury in primary biliary cirrhosis. Hepatology, 39(5), 1415–1422.

Mayo, M. J., Parkes, J., Adams-Huet, B., Combes, B., Mills, A. S., Markin, R. S., … Rosenberg, W. M. (2008). Prediction of clinical outcomes in primary biliary cirrhosis by serum enhanced liver fibrosis assay. Hepatology, 48(5), 1549–1557.

McNally, R. J., Ducker, S., & James, O. F. (2009). Are transient environmental agents involved in the cause of primary biliary cirrhosis? Evidence from space-time clustering analysis. Hepatology, 50(4), 1169–1174.

McNally, R. J., James, P. W., Ducker, S., & James, O. F. (2011). Seasonal variation in the patient diagnosis of primary biliary cirrhosis: further evidence for an environmental component to etiology. Hepatology, 54(6), 2099–2103.

116

McNally, R. J., James, P. W., Ducker, S., Norman, P. D., & James, O. F. (2014). No rise in incidence but geographical heterogeneity in the occurrence of primary biliary cirrhosis in North East England. American Journal of Epidemiology, 179(4), 492–498.

Metcalf, J., & James, O. (1997). The geoepidemiology of primary biliary cirrhosis. Seminars in Liver Disease, 17(1), 13–22.

Metcalf, J. V, Bhopal, R. S., Gray, J., Howel, D., & James, O. F. (1997). Incidence and prevalence of primary biliary cirrhosis in the city of Newcastle upon Tyne, England. International Journal of Epidemiology, 26(4), 830–836.

Metcalf, J. V, Mitchison, H. C., Palmer, J. M., Jones, D. E., Bassendine, M. F., & James, O. F. (1996). Natural history of early primary biliary cirrhosis. Lancet, 348(9039), 1399–1402.

Miller, K. B., Sepersky, R. A., Brown, K. M., Goldberg, M. J., & Kaplan, M. M. (1983). Genetic abnormalities of immunoregulation in primary biliary cirrhosis. The American Journal of Medicine, 75(1), 75–80.

Mitchison, H. C., Lucey, M. R., Kelly, P. J., Neuberger, J. M., Williams, R., & James, O. F. (1990). Symptom development and prognosis in primary biliary cirrhosis: a study in two centers. Gastroenterology, 99(3), 778–784.

Morling, N., Dalhoff, K., Fugger, L., Georgsen, J., Jakobsen, B., Ranek, L., … Svejgaard, A. (1992). DNA polymorphism of HLA class II genes in primary biliary cirrhosis. Immunogenetics, 35(2), 112–116.

Mounach, A., Ouzzif, Z., Wariaghli, G., Achemlal, L., Benbaghdadi, I., Aouragh, A., … El Maghraoui, A. (2008). Primary biliary cirrhosis and osteoporosis: a case-control study. Journal of Bone and Mineral Metabolism, 26(4), 379–384.

Muñoz, S. J., Heubi, J. E., Balistreri, W. F., & Maddrey, W. C. (1989). Vitamin E deficiency in primary biliary cirrhosis: gastrointestinal malabsorption, frequency and relationship to other lipid-soluble vitamins. Hepatology, 9(4), 525–531.

Muratori, P., Muratori, L., Ferrari, R., Cassani, F., Bianchi, G., Lenzi, M., … Bianchi, F. B. (2003). Characterization and clinical impact of antinuclear antibodies in primary biliary cirrhosis. American Journal of Gastroenterology, 98(2), 431–437.

Muratori, P., Muratori, L., Gershwin, M. E., Czaja, A. J., Pappas, G., MacCariello, S., … Bianchi, F. B. (2004). “True” antimitochondrial antibody-negative primary biliary cirrhosis, low sensitivity of the routine assays, or both? Clinical and Experimental Immunology, 135(1), 154–158.

Murillo Perez, C. F., Goet, J. C., Lammers, W. J., Gulamhusein, A., van Buuren, H. R., Ponsioen, C. Y., … Hansen, B. E. (2018). Milder disease stage in patients with primary biliary cholangitis over a 44-year period: A changing natural history. Hepatology, 67(5), 1920–1930.

117

Murtaugh, P. A., Dickson, E. R., Van Dam, G. M., Malinchoc, M., Grambsch, P. M., Langworthy, A. L., & Gips, C. H. (1994). Primary biliary cirrhosis: prediction of short-term survival based on repeated patient visits. Hepatology, 20(1 Pt 1), 126–134.

Mutimer, D. J., Fussey, S. P., Yeaman, S. J., Kelly, P. J., James, O. F., & Bassendine, M. F. (1989). Frequency of IgG and IgM autoantibodies to four specific M2 mitochondrial autoantigens in primary biliary cirrhosis. Hepatology, 10(4), 403–407.

Myers, R. P., Shaheen, A. A., Fong, A., Burak, K. W., Wan, A., Swain, M. G., … Quan, H. (2009). Epidemiology and natural history of primary biliary cirrhosis in a Canadian health region: a population-based study. Hepatology, 50(6), 1884–1892.

Myszor, M., & James, O. F. (1990). The epidemiology of primary biliary cirrhosis in north-east England: an increasingly common disease? The Quarterly Journal of Medicine, 75(276), 377–385.

Nakamura, M., Kondo, H., Mori, T., Komori, A., Matsuyama, M., Ito, M., … Ishibashi, H. (2007). Anti-gp210 and anti-centromere antibodies are different risk factors for the progression of primary biliary cirrhosis. Hepatology, 45(1), 118–127.

Nakanuma, Y. (1993). Distribution of B lymphocytes in nonsuppurative cholangitis in primary biliary cirrhosis. Hepatology, 18(3), 570–575.

Neuberger, J. (2003). Liver transplantation for primary biliary cirrhosis. Autoimmunity Reviews, 2(1), 1–7.

Nevens, F., Andreone, P., Mazzella, G., Strasser, S. I., Bowlus, C., Invernizzi, P., … Shapiro, D. (2016). A placebo-controlled trial of obeticholic acid in primary biliary cholangitis. New England Journal of Medicine, 375(7), 631–643.

Newton, J. L., Davidson, A., Kerr, S., Bhala, N., Pairman, J., Burt, J., & Jones, D. E. (2007). Autonomic dysfunction in primary biliary cirrhosis correlates with fatigue severity. European Journal of Gastroenterology and Hepatology, 19(2), 125–132.

Newton, J. L., Gibson, G. J., Tomlinson, M., Wilton, K., & Jones, D. (2006). Fatigue in primary biliary cirrhosis is associated with excessive daytime somnolence. Hepatology, 44(1), 91–98.

Nguyen, D. L., Juran, B. D., & Lazaridis, K. N. (2010). Primary biliary cirrhosis. Best Practice and Research. Clinical Gasttroenterology, 24(5), 647–654.

Oertelt, S., Rieger, R., Selmi, C., Invernizzi, P., Ansari, A. A., Coppel, R. L., … Gershwin, M. E. (2007). A sensitive bead assay for antimitochondrial antibodies: Chipping away at AMA-negative primary biliary cirrhosis. Hepatology, 45(3), 659–665.

118

Ong, K. L., Allison, M. A., Cheung, B. M., Wu, B. J., Barter, P. J., & Rye, K. A. (2014). The relationship between total bilirubin levels and total mortality in older adults: the United States National Health and Nutrition Examination Survey (NHANES) 1999-2004. PLoS ONE, 9(4), e94479.

Parés, A., Caballería, L., & Rodés, J. (2006). Excellent long-term survival in patients with primary biliary cirrhosis and biochemical response to ursodeoxycholic acid. Gastroenterology, 130(3), 715–720.

Parés, A., Caballería, L., Rodés, J., Bruguera, M., Rodrigo, L., García-Plaza, A., … Velicia, R. (2000). Long-term effects of ursodeoxycholic acid in primary biliary cirrhosis: Results of a double-blind controlled multicentric trial. UDCA-Cooperative Group from the Spanish Association for the Study of the Liver. Journal of Hepatology, 32(4), 561–566.

Parikh-Patel, A., Gold, E. B., Worman, H., Krivy, K. E., & Gershwin, M. E. (2001). Risk factors for primary biliary cirrhosis in a cohort of patients from the United States. Hepatology, 33(1), 16–21.

Paumgartner, G., & Beuers, U. (2002). Ursodeoxycholic acid in cholestatic liver disease: mechanisms of action and therapeutic use revisited. Hepatology, 36(3), 525–531.

Pellicciari, R., Fiorucci, S., Camaioni, E., Clerici, C., Costantino, G., Maloney, P. R., … Willson, T. M. (2002). 6alpha-ethyl-chenodeoxycholic acid (6-ECDCA), a potent and selective FXR agonist endowed with anticholestatic activity. Journal of Medicinal Chemistry, 45(17), 3569–3572.

Pells, G., Mells, G. F., Carbone, M., Newton, J. L., Bathgate, A. J., Burroughs, A. K., … Jones, D. E. (2013). The impact of liver transplantation on the phenotype of primary biliary cirrhosis patients in the UK-PBC cohort. Journal of Hepatology, 59(1), 67–73.

Peters, M. G., Di Bisceglie, A. M., Kowdley, K. V, Flye, N. L., Luketic, V. A., Munoz, S. J., … PUMPS Group. (2007). Differences between Caucasian, African American, and Hispanic patients with primary biliary cirrhosis in the United States. Hepatology, 46(3), 769–775.

Phillips, J. R., Angulo, P., Petterson, T., & Lindor, K. D. (2001). Fat-soluble vitamin levels in patients with primary biliary cirrhosis. American Journal of Gastroenterology, 96(9), 2745–2750.

Pla, X., Vergara, M., Gil, M., Dalmau, B., Cisteró, B., Bella, R. M., & Real, J. (2007). Incidence, prevalence and clinical course of primary biliary cirrhosis in a Spanish community. European Journal of Gastroenterology and Hepatology, 19(10), 859–864.

Podda, M., Selmi, C., Lleo, A., Moroni, L., & Invernizzi, P. (2013). The limitations and hidden gems of the epidemiology of primary biliary cirrhosis. Journal of Autoimmunity, 46, 81–87.

Poupon, R. (2010). Primary biliary cirrhosis: a 2010 update. Journal of Hepatology, 52(5), 745–758.

119

Poupon, R. E., Bonnand, A. M., Chrétien, Y., & Poupon, R. (1999). Ten-year survival in ursodeoxycholic acid-treated patients with primary biliary cirrhosis. The UDCA-PBC Study Group. Hepatology, 29(6), 1668–1671.

Poupon, R. E., Chrétien, Y., Chazouillères, O., Poupon, R., & Chwalow, J. (2004). Quality of life in patients with primary biliary cirrhosis. Hepatology, 40(2), 489–494.

Poupon, R. E., Lindor, K. D., Cauch-Dudek, K., Dickson, E. R., Poupon, R., & Heathcote, E. J. (1997). Combined analysis of randomized controlled trials of ursodeoxycholic acid in primary biliary cirrhosis. Gastroenterology, 113(3), 884–890.

Poupon, R. E., Poupon, R., & Balkau, B. (1994). Ursodiol for the long-term treatment of primary biliary cirrhosis. The UDCA-PBC Study Group. New England Journal of Medicine, 330(19), 1342–1347.

Prati, D., Taioli, E., Zanella, A., Torre, E. Della, Butelli, S., Del Vecchio, E., … Sirchia, G. (2002). Updated definitions of healthy ranges for serum alanine aminotransferase levels. Annals of Internal Medicine, 137(1), 1–10.

Pratt, D. S. (2016). Primary biliary cholangitis--A new name and a new treatment. New England Journal of Medicine, 375(7), 685–687.

Prince, M., Chetwynd, A., Newman, W., Metcalf, J. V, & James, O. F. (2002). Survival and symptom progression in a geographically based cohort of patients with primary biliary cirrhosis: follow-up for up to 28 years. Gastroenterology, 123(4), 1044–1051.

Prince, M. I., Chetwynd, A., Craig, W. L., Metcalf, J. V, & James, O. F. (2004). Asymptomatic primary biliary cirrhosis: clinical features, prognosis, and symptom progression in a large population based cohort. Gut, 53(6), 865–870.

Prince, M. I., Chetwynd, A., Diggle, P., Jarner, M., Metcalf, J. V, & James, O. F. (2001). The geographical distribution of primary biliary cirrhosis in a well-defined cohort. Hepatology, 34(6), 1083–1088.

Prince, M. I., Ducker, S. J., & James, O. F. (2010). Case-control studies of risk factors for primary biliary cirrhosis in two United Kingdom populations. Gut, 59(4), 508–512.

Prince, M. I., & James, O. F. (2003). The epidemiology of primary biliary cirrhosis. Clinics in Liver Disease, 7(4), 795–819.

Quarneti, C., Muratori, P., Lalanne, C., Fabbri, A., Menichella, R., Granito, A., … Muratori, L. (2015). Fatigue and pruritus at onset identify a more aggressive subset of primary biliary cirrhosis. Liver International, 35(2), 636–641.

Quinn, J., Diamond, A. G., Palmer, J. M., Bassendine, M. F., James, O. F., & J, Y. S. (1993). Lipoylated and unlipoylated domains of human PDC-E2 as autoantigens in primary biliary cirrhosis: significance of lipoate attachment. Hepatology, 18(6), 1384–1391.

120

Rautiainen, H., Salomaa, V., Niemelå, S., Karvonen, A. L., Nurmi, H., Isoniemi, H., & Färkkilä, M. (2007). Prevalence and incidence of primary biliary cirrhosis are increasing in Finland. Scandinavian Journal of Gastroenterology, 42(11), 1343–1353.

Remmel, T., Remmel, H., Uibo, R., & Salupere, V. (1995). Primary biliary cirrhosis in Estonia. With special reference to incidence, prevalence, clinical features, and outcome. Scandinavian Journal of Gastroenterology, 30(4), 367–371.

Reshetnyak, V. I. (2015). Primary biliary cirrhosis : Clinical and laboratory criteria for its diagnosis. World Journal of Gastroenterology, 21(25), 7683–7708.

Reynolds, T. B., Denison, E. K., Frankl, H. D., Lieberman, F. L., & Peters, R. L. (1971). Primary biliary cirrhosis with scleroderma, Raynaud’s phenomenon and telangiectasia. New syndrome. The American Journal of Medicine, 50(3), 302–312.

Rieger, R., Leung, P. S., Jeddeloh, M. R., Kurth, M. J., Nantz, M. H., Lam, K. S., … Gershwin, M. E. (2006). Identification of 2-nonynoic acid, a cosmetic component, as a potential trigger of primary biliary cirrhosis. Journal of Autoimmunity, 27(1), 7–16.

Rigopoulou, E. I., Davies, E. T., Pares, A., Zachou, K., Liaskos, C., Bogdanos, D. P., … Vergani, D. (2005). Prevalence and clinical significance of isotype specific antinuclear antibodies in primary biliary cirrhosis. Gut, 54(4).

Rosenthal, P., Pincus, M., & Fink, D. (1984). Sex- and age-related differences in bilirubin concentrations in serum. Clinical Chemistry, 30(8), 1380–1382.

Rubin, D. B. (1996). Multiple imputation after 18+ years. Journal of the American Statitical Association, 91(434), 473–489.

Sakauchi, F., Oura, A., Ohnishi, H., & Mori, M. (2007). Comparison of the clinical features of Japanese patients with primary biliary cirrhosis in 1999 and 2004: utilization of clinical data when patients applied to receive public financial aid. Journal of Epidemiology, 17(6), 210–214.

Scheuer, P. (1967). Primary biliary cirrhosis. Proceedings of the Royal Society of Medicine, 60(12), 1257–1260.

Selmi, C., Mayo, M. J., Bach, N., Ishibashi, H., Invernizzi, P., Gish, R. G., … Gershwin, M. E. (2004). Primary biliary cirrhosis in monozygotic and dizygotic twins: genetics, epigenetics, and environment. Gastroenterology, 127(2), 485–492.

Shapiro, J. M., Smith, H., & Schaffner, F. (1979). Serum bilirubin: a prognostic factor in primary biliary cirrhosis. Gut, 20(2), 137–140.

Shi, J., Wu, C., Lin, Y., Chen, Y. X., Zhu, L., & Xie, W. F. (2006). Long-term effects of mid-dose ursodeoxycholic acid in primary biliary cirrhosis: a meta-analysis of randomized controlled trials. American Journal of Gastroenterology, 101(7), 1529–1538.

121

Shibuya, A., Tanaka, K., Miyakawa, H., Shibata, M., Takatori, M., Sekiyama, K., … Morizane, T. (2002). Hepatocellular carcinoma and survival in patients with primary biliary cirrhosis. Hepatology, 35(5), 1172–1178.

Shimoda, S., Nakamura, M., Ishibashi, H., Kawano, A., Kamihira, T., Sakamoto, N., … Harada, M. (2003). Molecular mimicry of mitochondrial and nuclear autoantigens in primary biliary cirrhosis. Gastroenterology, 124(7), 1915–1925.

Siegel, J. L., Luthra, H., Donlinger, J., Angulo, P., & Lindor, K. (2003). Association of primary biliary cirrhosis and rheumatoid arthritis. Journal of Clinical Rheumatology, 9(6), 340–343.

Silveira, M. G., Suzuki, A., & Lindor, K. D. (2008). Surveillance for hepatocellular carcinoma in patients with primary biliary cirrhosis. Hepatology, 48(4), 1149–1156.

Solaymani-Dodaran, M., Card, T. R., Aithal, G. P., & West, J. (2006). Fracture risk in people with primary biliary cirrhosis: a population-based cohort study. Gastroenterology, 131(6), 1752–1757.

Sood, S., Gow, P. J., Christie, J. M., & Angus, P. W. (2004). Epidemiology of primary biliary cirrhosis in Victoria, Australia: high prevalence in migrant populations. Gastroenterology, 127(2), 470–475.

Spalding, M. C., & Sebesta, S. C. (2008). Geriatric screening and preventive care. American Family Physician, 78(2), 206–215.

Springer, J., Cauch-Dudek, K., O’Rourke, K., Wanless, I. R., & Heathcote, E. J. (1999). Asymptomatic primary biliary cirrhosis: a study of its natural history and prognosis. American Journal of Gastroenterology, 94(1), 47–53.

Stiehl, A., Rudolph, G., Raedsch, R., Möller, B., Hopf, U., Lotterer, E., … Senn, M. (1990). Ursodeoxycholic acid-induced changes of plasma and urinary bile acids in patients with primary biliary cirrhosis. Hepatology, 12(3 Pt 1), 492–497.

Su, W. C., Chan, C. C., Hung, H. H., Huo, T. I., Huang, Y. H., Li, C. P., … Wu, J. C. (2009). Predictive value of aspartate aminotransferase to alanine aminotransferase ratio for hepatic fibrosis and clinical adverse outcomes in patients with primary biliary cirrhosis. Journal of Clinical Gastroenterology, 43(9), 876–883.

Suzuki, A., Lymp, J., Donlinger, J., Mendes, F., Angulo, P., & Lindor, K. (2007). Clinical predictors for hepatocellular carcinoma in patients with primary biliary cirrhosis. Clinical Gastroenterology and Hepatology, 5(2), 259–264.

Suzuki, N., Irie, M., Iwata, K., Nakane, H., Yoshikane, M., Koyama, Y., … Sakisaka, S. (2006). Altered expression of alkaline phosphatase (ALP) in the liver of primary biliary cirrhosis (PBC) patients. Hepatology Research, 35(1), 37–44.

122

Sylvestre, P. B., Batts, K. P., Burgart, L. J., Poterucha, J. J., & Wiesner, R. H. (2003). Recurrence of primary biliary cirrhosis after liver transplantation: Histologic estimate of incidence and natural history. Liver Transplantation, 9(10), 1086–1093.

ter Borg, P. C. J., Schalm, S. W., Hansen, B. E., & van Buuren, H. R. (2006). Prognosis of ursodeoxycholic acid-treated patients with primary biliary cirrhosis. Results of a 10-yr cohort study involving 297 patients. American Journal of Gastroenterology, 101(9), 2044–2050.

Terrault, N. A., Bzowej, N. H., Chang, K., Hwang, J. P., Jonas, M. M., & Murad, M. H. (2016). AASLD guidelines for treatment of chronic hepatitis B. Hepatology, 63(1), 261–283.

Trivedi, P. J., Bruns, T., Cheung, A., Li, K.-K., Kittler, C., Kumagi, T., … Hirschfield, G. M. (2014). Optimising risk stratification in primary biliary cirrhosis: AST/platelet ratio index predicts outcome independent of ursodeoxycholic acid response. Journal of Hepatology, 60(6), 1249–1258.

Trivedi, P. J., Lammers, W. J., van Buuren, H. R., Parés, A., Floreani, A., Janssen, H. L. A., … Hirschfield, G. M. (2016). Stratification of hepatocellular carcinoma risk in primary biliary cirrhosis: A multicentre international study. Gut, 65(2), 321–329.

Tsianos, E. V, Hoofnagle, J. H., Fox, P. C., Alspaugh, M., Jones, E. A., Schafer, D. F., & Moutsopoulos, H. M. (1990). Sjögren’s syndrome in patients with primary biliary cirrhosis. Hepatology, 11(5), 730–734.

Tyagi, S., Gupta, P., Saini, A. S., Kaushal, C., & Sharma, S. (2011). The peroxisome proliferator-activated receptor: A family of nuclear receptors role in various diseases. Journal of Advanced Pharmaceutical Technology and Research, 2(4), 236–240.

Underhill, J., Donaldson, P., Bray, G., Doherty, D., Portmann, B., & Williams, R. (1992). Susceptibility to primary biliary cirrhosis is associated with the HLA-DR8-DQB1*0402 haplotype. Hepatology, 16(6), 1404–1408.

United Nations, Department of Economic and Social Affairs, P. D. (2017). World population prospects: The 2017 revision, key findings and advance tables. Retrieved from https://esa.un.org/unpd/wpp/Publications/Files/WPP2017_KeyFindings.pdf

Van de Water, J., Cooper, A., Surh, C. D., Coppel, R., Danner, D., Ansari, A., … Gershwin, M. E. (1989). Detection of autoantibodies to recombinant mitochondrial proteins in patients with primary biliary cirrhosis. New England Journal of Medicine, 320(21), 1377–1380.

Van de Water, J., Gershwin, M. E., Leung, P., Ansari, A., & Coppel, R. L. (1988). The autoepitope of the 74-kD mitochondrial autoantigen of primary biliary cirrhosis corresponds to the functional site of dihydrolipoamide acetyltransferase. The Journal of Experimental Medicine, 167(6), 1791–1799.

123

Watson, R. G., Angus, P. W., Dewar, M., Goss, B., Sewell, R. B., & Smallwood, R. A. (1995). Low prevalence of primary biliary cirrhosis in Victoria, Australia. Melbourne Liver Group. Gut, 36(6), 927–930.

Watt, F. E., James, O. F., & Jones, D. E. (2004). Patterns of autoimmunity in primary biliary cirrhosis patients and their families: a population-based cohort study. QJM, 97(7), 397–406.

Wesierska-Gadek, J., Hohenuer, H., Hitchman, E., & Penner, E. (1996). Autoantibodies against nucleoporin p62 constitute a novel marker of primary biliary cirrhosis. Gastroenterology, 110(3), 840–847.

Wesierska-Gadek, J., Penner, E., Battezzati, P. M., Selmi, C., Zuin, M., Hitchman, E., … Invernizzi, P. (2006). Correlation of initial autoantibody profile and clinical outcome in primary biliary cirrhosis. Hepatology, 43(5), 1135–1144.

White, G. L., Nelson, J. A., Pedersen, D. M., & Ash, K. O. (1981). Fasting and gender (and altitude?) influence reference intervals for serum bilirubin in healthy adults. Clinical Chemistry, 27(6), 1140–1142.

Wiesner, R. H., Ludwig, J., Lindor, K. D., Jorgensen, R. A., Baldus, W. P., Homburger, H. A., & Dickson, E. R. (1990). A controlled trial of cyclosporine in the treatment of primary biliary cirrhosis. New England Journal of Medicine, 322(20), 1419–1424.

Yang, F., Yang, Y., Wang, Q., Wang, Z., Miao, Q., Xiao, X., … Ma, X. (2017). The risk predictive values of UK-PBC and GLOBE scoring system in Chinese patients with primary biliary cholangitis: the additional effect of anti-gp210. Alimentary Pharmacology & Therapeutics, 45(5), 733–743.

Zein, C. O., Angulo, P., & Lindor, K. D. (2003). When is liver biopsy needed in the diagnosis of primary biliary cirrhosis? Clinical Gastroenterology and Hepatology, 1(2), 89–95.

Zein, C. O., Beatty, K., Post, A. B., Logan, L., Debanne, S., & McCullough, A. J. (2006). Smoking and increased severity of hepatic fibrosis in primary biliary cirrhosis: A cross validated retrospective assessment. Hepatology, 44(6), 1564–1571.

Zhang, L. N., Shi, T. Y., Shi, X. H., Wang, L., Yang, Y. J., Liu, B., … Zhang, F. C. (2013). Early biochemical response to ursodeoxycholic acid and long-term prognosis of primary biliary cirrhosis: results of a 14-year cohort study. Hepatology, 58(1), 264–272.

Zografos, T. A., Gatselis, N., Zachou, K., Liaskos, C., Gabeta, S., Koukoulis, G. K., & Dalekos, G. N. (2012). Primary biliary cirrhosis-specific autoantibodies in first degree relatives of Greek primary biliary cirrhosis patients. World Journal of Gastroenterology, 18(34), 4721–4728.

Zucker, S. D., Horn, P. S., & Sherman, K. E. (2004). Serum bilirubin levels in the U.S. population: Gender effect and inverse correlation with colorectal cancer. Hepatology, 40(4), 827–835.

124

Copyright Acknowledgements

JOHN WILEY AND SONS LICENSE TERMS AND CONDITIONS

Jul 03, 2018

This Agreement between Ms. Carla Fiorella Murillo Perez ("You") and John Wiley and Sons ("John Wiley and Sons") consists of your license details and the terms and conditions provided by John Wiley and Sons and Copyright Clearance Center.

License Number 4353830876851

License date May 21, 2018

Licensed Content Publisher

John Wiley and Sons

Licensed Content Publication

Hepatology

Licensed Content Title Milder disease stage in patients with primary biliary cholangitis over a 44‐year period: A changing natural history

Licensed Content Author Carla F. Murillo Perez, Jorn C. Goet, Willem J. Lammers, et al

Licensed Content Date Apr 6, 2018

Licensed Content Volume 67

Licensed Content Issue 5

Licensed Content Pages 11

Type of use Dissertation/Thesis

Requestor type Author of this Wiley article

Format Print and electronic

Portion Full article

Will you be translating? No

Title of your thesis / dissertation

The changing natural history of primary biliary cholangitis and its influence on risk stratification

Expected completion date Sep 2018

Expected size (number of pages)

110

Publisher Tax ID EU826007151

125

Appendix

Figure. Flow chart depicting patient exclusion criteria for the GLOBAL PBC cohort.

GLOBAL PBCcohortN=6677

AIH or otherconcomintant liver

diseasen=193

Uncertain diagnosisn=44

No laboratory valuesn=3

Unknown follow-upor short follow-up

n=1068Short treatment

with UDCAn=268

Total patientsn=5101

126

Contributions Carla Fiorella Murillo Perez had access to the data and performed the majority of the statistical

analysis. Harry Janssen, Jordan Feld, and Bettina Hansen provided guidance throughout the

course of my research, from study design to the interpretation of results. Bettina Hansen also

provided assistance with statistical analyses. Jorn Goet, Willem Lammers, Henk van Buuren,

Maren Harms, and Adrian van der Meer provided feedback and aided with critical revision of the

study for important intellectual content.

Acquisition of data was a collective effort by the GLOBAL PBC Study group. Members of the

GLOBAL PBC group also aided with critical revision of the study for important intellectual

content and provided advice and suggestions.

This study was supported by unrestricted grants from Intercept Pharmaceuticals and was funded

by the Foundation for Liver and Gastrointestinal Research (a not-for-profit foundation) in

Rotterdam, the Netherlands and Toronto Centre for Liver Disease. The supporting parties had no

influence on the study design, data collection and analyses, or interpretation of the results.