Skip to main content
Erschienen in: Hepatology International 5/2022

Open Access 03.06.2022 | Original Article

Epidemiology and outcomes of primary sclerosing cholangitis: an Australian multicentre retrospective cohort study

verfasst von: Natassia Tan, N. Ngu, T. Worland, T. Lee, T. Abrahams, K. Pandya, E. Freeman, N. Hannah, K. Gazelakis, R. G. Madden, K. D. Lynch, Z. Valaydon, S. Sood, A. Dev, S. Bell, A. Thompson, J. Ding, A. J. Nicoll, K. Liu, P. Gow, J. Lubel, W. Kemp, S. K. Roberts, A. Majeed, the Melbourne Liver Group

Erschienen in: Hepatology International | Ausgabe 5/2022

Abstract

Background and aims

Little is known regarding the epidemiology and outcomes of patients with primary sclerosing cholangitis (PSC) in Australia. We, therefore, evaluated the epidemiology and clinical outcomes of PSC in a large cohort of Australian patients and compared these to the general population.

Methods

We conducted a multicentre, retrospective cohort study of PSC patients at nine tertiary liver centers across three Australian states, including two liver transplant centers.

Results

A total of 413 PSC patients with 3,285 person-years of follow-up were included. Three hundred and seventy-one (90%) patients had large duct PSC and 294 (71%) had associated inflammatory bowel disease. A total of 168 (41%) patients developed cirrhosis (including 34 at the time of PSC diagnosis) after a median of 15.8 (95% CI 12.4, NA) years. The composite endpoint of death or liver transplantation occurred in 49 (12%) and 78 (19%) patients, respectively, with a median transplant-free survival of 13.4 (95% CI 12.2–15) years. Compared to the general population, PSC accounted for a 240-fold increased risk of development of cholangiocarcinoma (CCA) and CCA-related death. CCA risk was increased with older age of PSC diagnosis, presence of dominant stricture and colectomy. Compared to same-aged counterparts in the general population, PSC patients who were diagnosed at an older age or with longer disease duration had reduced relative survival.

Conclusion

In this large retrospective cohort study of PSC patients in Australia, increased age and time from diagnosis was associated with increased mortality and morbidity particularly from CCA and development of cirrhosis, necessitating need for liver transplant.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1007/​s12072-022-10356-1.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
ALP
Alkaline phosphatase
ALT
Alanine aminotransferase
AST
Aspartate aminotransferase
CA 19-9
Carbohydrate antigen 19-9
CCA
Cholangiocarcinoma
CI
Confidence interval
CRC
Colorectal carcinoma
HCC
Hepatocellular carcinoma
HR
Hazard ratio
IBD
Inflammatory bowel disease
IBDU
Inflammatory bowel disease—unclassified
INR
International normalized ratio
IQR
Interquartile range
LT
Liver transplantation
PSC
Primary sclerosing cholangitis
SIR
Standardized incidence ratio
SMR
Standardized mortality ratio
UDCA
Ursodeoxycholic acid

Introduction

Primary sclerosing cholangitis (PSC) is an orphan, cholestatic liver disease characterized by chronic inflammation of the intra- and extra-hepatic biliary tree with multi-focal areas of stricturing [1]. It has a strong association with hepatobiliary cancers and inflammatory bowel disease (IBD) with a heightened risk of colorectal cancer (CRC) in this subgroup [2, 3]. There is no proven effective medical therapy which alters the natural history of PSC [1]. Liver transplantation (LT) is an effective intervention for patients who develop refractory complications or end-stage liver disease; however, disease recurrence can occur in up to 25–30% of grafts [3, 4].
There is a paucity of Australasian literature on PSC, with only two studies published from Australia [5, 6] and one from New Zealand [7]. Two studies highlighted the increased risk of PSC-related outcomes in patients with concomitant IBD [5, 7], but also demonstrated variability in median transplant-free survival. To address the limited Australian epidemiological and outcome data on PSC, we aimed to conduct a multicentre study to accurately describe the epidemiology, clinical characteristics, and outcomes in patients with PSC in Australia. We also described the relative survival of PSC patients compared to the general population.

Methods

Study population

We performed a retrospective cohort study of patients diagnosed with PSC from 1st January 2000 to 30th March 2021 at 9 tertiary academic liver centers across Australia in Victoria, New South Wales, and South Australia, including two transplant centers and seven non-transplant tertiary referral centers.

Inclusion and exclusion criteria

We included all adult patients with a confirmed diagnosis of PSC based on the presence of established cholangiographic or histological features of large or small duct PSC coupled with a cholestatic biochemical profile. Patients with secondary causes of sclerosing cholangitis as per recognized international guidelines were excluded [810]. Patients with concomitant liver diseases were not excluded for the purpose of this study.

Case ascertainment

Case ascertainment was achieved at a local level with a combination of pre-existing or prospectively collected databases in addition to computerized search of patient diagnoses through electronic medical records, radiology, and histology records using the terms—“primary sclerosing cholangitis” and “sclerosing cholangitis”. Pre- and post-liver transplant patient registries were also reviewed at the two transplant centers. All individual medical records were reviewed at a local level to confirm a diagnosis of PSC and accuracy of data collection, including removal of duplicate patient entries when referred for LT.

Data collection

Clinical demographics and characteristics collected included patient sex, date of birth, date of PSC diagnosis, ethnicity, phenotype, ursodeoxycholic acid (UDCA) use, IBD type, presence of cirrhosis with or without portal hypertension, decompensation events (development of ascites, hepatic encephalopathy, or variceal bleeding), LT indication and date, malignancy type and date of diagnosis.

Definitions

Evidence for overlap syndrome with autoimmune hepatitis was based on biochemical or serological features with or without histological confirmation. IBD phenotypes of Crohn’s disease, ulcerative colitis or IBD-unclassified (IBDU) were determined via clinical documentation and biochemical, imaging, endoscopic and histological confirmation if available based on consensus guidelines [11]. The presence of cirrhosis and portal hypertension was based on radiological or histological findings if available coupled with supportive clinical, non-invasive testing with transient elastography and laboratory data [12].

Statistics

Non-parametric continuous variables were presented as medians and interquartile ranges (IQR) and analyzed with the Mann–Whitney U test for skewed distributions. Binomial data were presented as percentages and were compared using Chi-square or Fisher’s exact test. The Kaplan Meier method was used to analyze outcomes over time including cirrhosis, malignancy, LT, and death, and to estimate the probability and hazard rate of events over time. Cox proportional hazards model was used to analyze predictors of death or LT, and development of cholangiocarcinoma (CCA) via univariate and multivariate analysis. Patients who were diagnosed with cirrhosis at the time of or within 6 months of PSC diagnosis were not included in the expected survival or hazard rate analysis of cirrhosis.
Standardized incidence (SIR) and mortality ratios (SMR) were calculated with the indirect method using population data from the Australian Institute of Health and Welfare as the reference population. Relative survival is defined as the ratio of the observed all-cause survival among patients and the expected survival in a comparable group in the general population. Life tables for the general Australian population stratified by calendar year, age and sex were used to estimate the relative survival. Observed survival in individuals with PSC was calculated on 2-yearly intervals from the date of PSC diagnosis and compared to age and sex matched data from population life tables as described previously [13].
A two-sided p-value of < 0.05 was considered as statistically significant, and 95% confidence intervals are reported where appropriate. Data analysis was performed with SPSS, version 27.0 (IBM) and SAS software, version 9.4 (SAS Institute Inc., Cary, NC, US).

Results

Study population

A total of 413 PSC patients were included in the study with a median follow-up time of 7.1 (IQR 3.6–11.7) years, contributing to 3,285 person-years of follow-up time from diagnosis. The median age at PSC diagnosis was 37 (IQR 23–53) years, and 266 (64%) patients were male with the majority (88%) of patients being of European ancestry. Two hundred and nineteen (53%) patients were initially managed at a liver transplant tertiary center. Three hundred and seventy-one (90%) patients had a large duct phenotype at diagnosis, and 294 (71%) had associated IBD. Three (0.7%) patients had concomitant chronic hepatitis C. Two hundred and sixty-four (64%) patients were taking ursodeoxycholic acid (UDCA) at any stage during their follow-up. Further descriptions of the demographics and clinical characteristics of the overall cohort are presented in Table 1.
Table 1
Baseline clinical characteristics of the study cohort
 
Total
 
N = 413
Age at diagnosis
37 (23–53)
Male
266 (64.4)
Ethnicity
 
 European
360 (87.8)
 Asian (inc. East Asian)
27 (6.6)
 Middle Eastern
17 (4.1)
 Hispanic
4 (1.0)
 African
2 (0.5)
Transplant center
219 (53)
Phenotype
 
 Large duct
371 (90.3)
 Small duct
40 (9.7)
AIH overlap
58 (14.2)
Concomitant liver disease
 
 Chronic hepatitis C
3 (0.7%)
Dominant stricture
110 (27.4)
UDCAa
264 (63.9)
Liver function at diagnosis
 
 ALT (IU/L)
93 (51–168)
 AST (IU/L)
68 (40–128)
 ALP (IU/L)
282 (156–562)
 Bilirubin (μmol/L)
16 (9–33)
 Albumin (g/L)
37 (33–42)
 INR
1.0 (1.0–1.1)
IBD
294 (71.2)
IBD type
 
 Ulcerative colitis
213 (72.4)
 Crohn’s disease
73 (24.8)
IBDU
8 (2.7)
Results presented as median (interquartile range) for continuous variables and absolute numbers (percentages) for categorical variables
AIH autoimmune hepatitis, ALT alanine aminotransferase, AST aspartate aminotransferase, ALP alkaline phosphatase, INR international normalised ratio, IBD inflammatory bowel disease, IBDU inflammatory bowel disease unclassified, UDCA ursodeoxycholic acid
aUDCA use at any stage during data collection
The annual number of new PSC cases at the contributing centers in greater Melbourne increased from 7 in 2000 to a peak of 25 cases in 2017 but declined to 5 toward the end of the study period in 2020/2021. This corresponded to an average annual incidence of 0.27 cases per 100,000 person-year in greater Melbourne (Fig. 1). Towards the end of the study in 2019/2020, there was a total of 266 cases (165 males, 101 females) of PSC alive and managed in the contributing hospitals in greater Melbourne corresponding to an estimated prevalence of 5.9 cases per 100,000 inhabitants (males; 3.7:100,000, females; 2.2:100,000).

Outcomes

Development of cirrhosis

A total of 168 (41%) patients were diagnosed with cirrhosis in our cohort. Thirty-four (8.2%) of patients were found to be cirrhotic at time of PSC diagnosis. Patients with cirrhosis at the time of PSC diagnosis were less likely to have concomitant IBD compared to those who did not have cirrhosis at time of diagnosis (44% vs 74%, p < 0.001).
In those who were not cirrhotic at the time of PSC diagnosis, the median time to development of cirrhosis was 15.8 (95% CI 12.4, NA) years (Supp Fig. 1: Cirrhosis-free survival amongst non-cirrhotic patients at diagnosis). The 5-, 10- and 20-year probabilities of cirrhosis were 17.6%, 33.9% and 75.5%, respectively. A total of 135 (80%) cirrhotic patients had concomitant portal hypertension and 82 (49%) experienced a decompensating event.
The incidence rate of cirrhosis in our cohort was 4.9 (95% CI 4.1–5.8) per 100 patient-years. The risk of developing cirrhosis increased with time from PSC diagnosis (Supp Fig. 2: Risk of developing cirrhosis over time). Patients on UDCA were more likely to be diagnosed with cirrhosis (45% vs 33%, p = 0.015) but there was no influence on rates of decompensation (22.5% vs 17%, p = 0.197) compared to those not on UDCA. There was no difference in time to development of cirrhosis between patients on UDCA and those who were not (p = 0.731).

Development of malignancy

Fifty-eight (14%) patients developed malignancy after an estimated median of 6.3 (IQR 3.5–10.7) years from diagnosis (Table 2 and 3). Apart from CCA (n = 24), CRC (n = 12) and hepatocellular carcinoma (HCC) (n = 6), four patients were diagnosed with squamous cell carcinoma of the skin, two patients with extranodal marginal zone lymphoma, two patients with neuroendocrine tumors, and one case each of prostate cancer, post-transplant lymphoproliferative disorder, ovarian cancer, breast cancer, non-small cell lung cancer and an appendiceal mucinous neoplasm. Two patients had metastatic malignancy of unclear primary. There was no difference in UDCA use between patients who developed malignancy and those who did not. There was a higher proportion of cirrhotic patients (55.2% vs 44.8%, p = 0.021) in patients who developed malignancy.
Table 2
Observed outcomes in the study cohort
 
Total
 
N = 413
Cirrhosis
168 (40.7)
Cirrhosis at diagnosis
34 (8.2)
Portal hypertension
135 (32.6)
Hepatic decompensation
82 (19.8)
LT
78 (18.9)
 End-stage liver disease
62 (83.8)
 HCC
3 (4.1)
 Recurrent cholangitis
5 (6.8)
 Refractory pruritus
1 (1.4)
 CCA
1 (1.4)
 Other
2 (2.7)
Death
49 (11.9)
 End-stage liver disease
12 (31.6)
 CCA
15 (39.5)
 CRC
1 (2.6)
 HCC
1 (2.6)
 Other malignancy
5 (13.2)
 Cholangitis
1 (2.6)
 Other
3 (7.9)
Malignancy
58 (14)
 HCC
6 (1.5)
 CRC
12 (2.9)
 CCA
24 (5.8)
 Others
16 (3.8)
Results presented as absolute numbers (percentages) for categorical variables
LT liver transplantation, HCC hepatocellular carcinoma, CCA cholangiocarcinoma, CRC colorectal carcinoma
Table 3
Malignant outcomes in the PSC cohort
Malignancy
Observed events
Time (years) to malignancy median (IQR)
Annual risk (%)
Expected events
SIR (95% CI)
p-value
Follow-up time from cancer (P-Yr)
Cancer-related death
SMR (95% CI)
p-value
CCA
24
6.7 (4.5–11.0)
0.74
0.1
243.6 (146.2–341.1)
0.0001
205.7
15
242.1 (119.6–364.5)
0.0001
HCC
6
6.3 (5.5–11.1)
0.18
0.17
35.4 (7.1–63.7)
0.02
80.7
1
6.9 (0–20.4)
0.39
CRC
10a
7.1 (4.9–9.7)
0.31
1.95
5.1 (1.9–8.3)
0.01
133.8
1
1.3 (0–3.8)
0.82
CCA cholangiocarcinoma, CRC colorectal cancer, CI confidence interval, HCC hepatocellular carcinoma, P-Yr patient-years, SIR standardized incidence ratio, SMR standardized mortality ratio
aTwo cases not included in analysis as date of cancer diagnosis not obtained
Cholangiocarcinoma CCA was the most common malignancy diagnosed with 24 (5.8%) patients developing CCA after a median of 6.7 (IQR 4.5–11.0) years from PSC diagnosis. Two patients were diagnosed within the first year of PSC diagnosis. Two others were diagnosed in liver explant (4- and 8-year post-PSC diagnosis). The median age at CCA diagnosis was 52.5 (IQR 38–65) years, and the majority (70%) were males. PSC was associated with over 240-fold increased risk for CCA compared to the general population (Table 3), with the highest risk for CCA observed 7–11 years after the diagnosis of PSC (Supp Fig. 3: Risk of developing malignancy after PSC diagnosis). CCA in PSC was also associated with an over 240-fold risk for death with a median survival of only 7 months (Supp Fig. 4: Overall survival after diagnosis of CCA—all-cause mortality). On Cox proportional hazards multivariate analysis, the presence of dominant stricture (HR 3.33, 95% CI 1.40–7.88), history of colectomy (HR 2.95, 95% CI 1.29–6.76) and increased age at PSC diagnosis (HR 1.03, 95% CI 1.00–1.05) were independently associated with an increased risk of CCA (Supplementary Table 2).
Colorectal carcinoma Twelve (2.9%) patients developed colorectal carcinoma (CRC) after a median of 7.1 (IQR 4.9–9.7) years from PSC diagnosis. Ten (83%) of those who developed CRC had a pre-existing diagnosis of IBD. PSC was associated with a fivefold increased risk for CRC as compared with the general population, with the highest risk between 6 and 8 years after PSC diagnosis (Supp Fig. 3: Risk of developing malignancy after PSC diagnosis). However, only one patient died of CRC and observed mortality from CRC in PSC patients (Table 3) was comparable to that of CRC in the general population (SMR 1.3, 95% CI 0–3.8, p-value 0.82). One patient was diagnosed initially with CRC and subsequently CCA 7 years after.
Hepatocellular carcinoma Six (1.5%) patients developed hepatocellular carcinoma (HCC), all of whom were cirrhotic, after a median time from PSC diagnosis of 6.3 (5.5–11.1) years. One HCC-related death was observed in the cohort during a median follow-up of 4.3 (IQR 2.1–7.6) years after HCC diagnosis. PSC was associated with a 35-fold increased risk for HCC (Table 3), but comparable HCC-related mortality to that of the general population (SMR 6.9, 95% CI 0–20.4, p-value 0.39).

Survival after PSC diagnosis

Transplant-free survival

One hundred and twenty-three (30%) patients died or received a LT after an estimated median of 13.4 (95% CI 12.2–15) years from PSC diagnosis (Table 2, Fig. 2). The 5-, 10- and 20-year transplant-free survival rates were 88.9%, 69.7% and 23.7%, respectively. Patients on UDCA were more likely to have died (14.4% vs 7.4%, p = 0.03) but there was no difference in need for LT (20.5% vs 16.1%, p = 0.28).
The presence of cirrhosis, dominant stricture, CCA, increased age, alkaline phosphatase (ALP) and bilirubin levels at diagnosis were significant on univariate analysis for reduced transplant-free survival. After adjusting for the above variables, only cirrhosis (HR 6.49, 95% CI 2.90–14.6, p < 0.0001), CCA (HR 3.03, 95% CI 1.28–7.15, p = 0.011) and increased age at PSC diagnosis (HR 1.02, 95% CI 1.01–1.03, p = 0.003) were associated with reduced transplant-free survival on multivariate analysis (Supplementary Table 3).

All-cause mortality

Forty-nine (12%) patients died during the follow-up period. Median survival was unable to be estimated with Kaplan Meier analysis (Supp Fig. 5: All-cause mortality in the PSC cohort), and mean estimated survival was 17.6 (95% CI 16.8–18.5) years. The 5-, 10- and 20-year survival rates were 95.2%, 89.6% and 60.3%, respectively. The most common cause of death was CCA (n = 15, 39.5%), followed by end-stage liver disease (n = 12, 32%) and other malignancy in five (13%) patients. Further details of causes of death are illustrated in Table 2.

Liver transplantation

Seventy-eight (19%) patients received a LT during the follow-up period. Median time to LT was unable to be estimated with Kaplan Meier analysis (Supp Fig. 6: Cumulative risk of liver transplantation after PSC diagnosis), and mean time to LT was 15 (95% CI 14.1–15.9) years. The most common indication for LT was end-stage liver disease in 62 (84%) patients. LT for malignancy was performed in three (4%) patients for HCC and one (1%) for CCA. Further details of indications for LT in the cohort are illustrated in Table 2.

Relative survival compared to the general population

Relative survival of PSC patients compared to the Australian general population as stratified by the age of PSC diagnosis is presented in Supplementary Table 1 and Fig. 3. Patients in all age groups had comparable survival to the general population at time of PSC diagnosis; however, there was a trend towards reduced relative survival in PSC patients with increased time from diagnosis (Fig. 3). This reduction in relative survival was steeper and occurred sooner after diagnosis in patients who were diagnosed at older ages, especially in patients diagnosed above 60 years of age (Supplementary Table 1). Survival in patients diagnosed with PSC before the age of 30 dropped below that of the general population first after 12–14 years from PSC diagnosis. The corresponding time point for those diagnosed with PSC between the age of 30 and 59 years was 6–8 years after PSC diagnosis. For those aged above 60 at the time of PSC diagnosis, survival was lower than the general populations by 2–4 years after PSC diagnosis.

Discussion

Key findings

This is the largest published study of a Southern Hemisphere cohort of patients with PSC and demonstrates that the natural history of PSC patients in Australia is similar to that of their Northern Hemisphere counterparts. PSC remains an uncommon disease in Australia (266 patients in 2019/2020 managed in 7 Victorian hospitals with a catchment population of ~ 4.5 million). Thirty percent of the cohort required liver transplant or died at a median time from diagnosis of 13 years, with development of cirrhosis, CCA and increased age at PSC diagnosis being significantly associated with reduced transplant-free survival. Despite the increased risk of developing hepatobiliary and colorectal malignancy, only PSC-related CCA was associated with an over 240-fold increase in mortality compared to the general population with a median survival of only 7 months. The presence of a dominant stricture, history of colectomy and increased age at PSC diagnosis was significantly associated with increased risk of CCA development. Furthermore, we demonstrate a reduction in relative survival with increased age and time from diagnosis as compared to the Australian general population.

Comparison to previous studies

Our cohort draws similarities to other European populations with the majority being of male gender with concomitant IBD and a median age of diagnosis at 37 years [3, 1416]. Our estimated prevalence rate of 5.9 cases per 100,000 inhabitants is similar to the highest prevalence reported in a United Kingdom study [15]; however, this was significantly lower than the point prevalence of 11 cases per 100,000 inhabitants in Canterbury, New Zealand [7]. This difference in prevalence despite geographical proximity may be due to complete identification of all PSC patients under the care of a small group of gastroenterologists in Canterbury, reflecting a true population prevalence rate [7]. An alternative explanation would be an increased burden of PSC in association with predisposition towards autoimmune disease at higher latitudes [17].
Forty-one percent of our study cohort were cirrhotic, with 8% diagnosed with cirrhosis at time of PSC diagnosis. Patients who were cirrhotic at diagnosis were less likely to have concomitant IBD, which may reflect regular screening with liver function tests prompting earlier diagnosis in this cohort. Majority of cirrhotic patients had concomitant portal hypertension and almost 50% experienced at least one episode of hepatic decompensation. This seems to be higher compared to other cholestatic liver conditions such as primary biliary cholangitis, where less than 30% of patients develop liver failure 10 years after diagnosis [18]. Perhaps related to the lack of an effective medical therapy, PSC patients seem to develop cirrhosis and liver decompensation at higher rates compared to other liver conditions. The finding of increased proportion of cirrhosis in patients on UDCA therapy likely reflects a degree of selection bias rather than true influence of UDCA on outcomes, with patients of a more severe phenotype and/or higher ALP levels being more likely to be commenced on therapy. This may also explain the increased proportion of deaths in patients who took UDCA compared to those who did not.
The proportion of PSC patients that develop cirrhosis has been reported to develop at an incidence rate of 18.6 per 1000 person-years in a American population-based study [15], which is significantly lower than our incidence rate of 4 per 100 person-years. This may reflect an element of referral bias, as half of our patients were managed primarily at transplant centers. Also, the increased risk of developing cirrhosis early after diagnosis (Supp Fig. 2: Risk of developing cirrhosis over time) may reflect latent disease that had been undiagnosed for years in asymptomatic patients or be representative of patients with an aggressive disease phenotype.
CCA remains a significant cause of morbidity and mortality in PSC patients [2, 19, 20]. The proportion of patients who developed CCA (5.8%) is comparable to other studies with prevalence ranging from 2.5 to 12% [2, 14, 21, 22]. The median time from diagnosis to development of CCA in our cohort was 6.7 years, which mirrors the findings of a large population-based study in the Netherlands where median time to CCA was 6 years [20]. This is longer compared to previous studies where most cases of CCA were detected in the first year of PSC diagnosis [14, 20, 22]. The risk of CCA and CRC development compared to the general population are similar to other European cohorts at 240-fold and fivefold, respectively [14, 20]. Our findings of the presence of a dominant stricture and increased age of PSC diagnosis being risk factors for development of CCA have been previously reported in the literature [23, 24]. The increased association of CCA with colectomy has been demonstrated in other studies along with presence of IBD [25, 26], although the latter was not replicated in our findings. It is unclear the mechanisms that colectomy may confer increased risk of CCA, but this finding lends weight to the argument that the gut microbiome plays a pathogenetic role in disease development in PSC.
The reported transplant-free survival of 13 years in our Australian cohort is comparable to the largest international PSC cohort ever described—derived from 35 tertiary centers of over 7000 patients [19]. This also matched the estimated median transplant-free survival in transplant cohorts described by Boonstra et al. [20], of which the study also highlighted the greatly improved median transplant-free survival in the population-based cohort of 21.3 years in comparison to 13.2 years in the transplant center cohort. Increased age at PSC diagnosis, development of cirrhosis and CCA has also been found to be significant predictors of transplant-free survival in keeping with the current literature [21, 23, 27].
Another important observation from our study was that patients who are diagnosed at an older age do poorly compared to their same-aged counterparts in the general population. This effect is even seen in younger age groups, although it is more pronounced as patients are diagnosed at increasing age especially after 60 years. Overall, our findings do support the current literature that age of diagnosis has a significant impact on disease course [16, 19, 21], possibly related to an unique disease phenotype [23].
Our study highlights the difficulty and importance of diagnosing PSC early and suggests a role for screening higher risk individuals such as middle-aged males of European descent, particularly those with IBD. In addition, non-smokers may be at increased risk of developing PSC [28], including those with first-degree relatives with PSC [29]. Once the diagnosis of PSC is established, it is prudent to monitor for development of complications such as cirrhosis and CCA especially in older patients. Although there is lacking evidence on the benefit of surveillance of hepatobiliary malignancy in PSC, this is strongly recommended by international guidelines [9, 30] and screening (especially for CCA) may be warranted in all patients but perhaps more frequently in those with risk factors such as diagnosed at an older age, presence of a dominant stricture or history of colectomy.

Strengths and limitations

Our study strengths include being the largest multicentre study of PSC in the Southern Hemisphere and long duration of patient follow-up. We also included comprehensive clinical endpoints such as the development of cirrhosis, malignancy, survival and need for LT. This was compared to the general population to derive the impact of PSC-related complications on overall morbidity and mortality. Identified limitations include our study being a retrospective analysis, as we acknowledge the real-world challenges of complete data collection and potential bias in case acquisition. Clinical delays with PSC diagnosis due to the typical asymptomatic presentation of PSC [1] may have affected results of the survival analysis. This is a limitation with most retrospective studies on PSC as we cannot accurately determine its subclinical course. However, our study reflects real-world data that correlates with other PSC cohorts as previously described. Although we did not exclude patients with other liver diseases, this contributed to less than 1% of the cohort and by including these patients allowed for a greater estimate of the epidemiology of PSC in Victoria.
We estimated the prevalence and incidence of a Victorian tertiary cohort of PSC patients in greater Melbourne; however, this does not reflect a true population-based result due to possible incomplete case acquisition and referrals from other states (Tasmania) to our Victorian liver transplant center contributing to a wider catchment population than expected. We also possibly missed out on a small subgroup of patients who may be managed privately or regionally that may have milder phenotypes of disease with improved outcomes. We acknowledge that some patients in our cohort were included in previous Australian studies [5, 6].

Summary and conclusion

This study describes the demographics, clinical characteristics, and outcomes of the largest ever reported Australian cohort of PSC patients. We demonstrate a reduction in relative survival in patients diagnosed with older age with increased time from diagnosis, coupled with increased morbidity and mortality from development of CCA and cirrhosis. In addition, we found that presence of a dominant stricture and previous history of colectomy were risk factors for CCA.
Further prospective studies on PSC in Australia encompassing clinical data and serial prognostic markers are underway, which will allow us to further stratify high-risk patient groups and improve our understanding and management of PSC patients.

Declarations

Conflict of interest

Natassia Tan, N. Ngu, T. Worland, T. Lee, T. Abrahams, K. Pandya, E. Freeman, Hannah, K. Gazelakis, R. G. Madden, K. D. Lynch, Z. Valaydon, S. Sood, A. Dev, S. Bell, A. Thompson, J. Ding, A. J. Nicoll, K. Liu1, P. Gow, J. Lubel, W. Kemp, S. K. Roberts, A. Majeed have no relevant financial or non-financial conflicts of interests to disclose relevant to this study.

Ethics approval

The study was approved by the Alfred Hospital Human Research Ethics Committee in Melbourne (Project 353/19), Australia and performed in accordance with the ethical standards as laid down in the Helsinki Declaration of 1975, as revised in 2008.
Informed consent was not required from patients included in the study as all patient information was de-identified.
Informed consent was not required from patients included in the study as all patient information was de-identified.

Clinical trials registration

This study was not registered as a clinical trial.

Animal research

This article does not contain any studies with human or animal subjects.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

Anhänge

Supplementary Information

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Lazaridis KN, LaRusso NF. Primary sclerosing cholangitis. N Engl J Med 2016;375(12):1161–1170CrossRef Lazaridis KN, LaRusso NF. Primary sclerosing cholangitis. N Engl J Med 2016;375(12):1161–1170CrossRef
2.
Zurück zum Zitat Guerra I, Bujanda L, Castro J, Merino O, Tosca J, Camps B, et al. Clinical characteristics, associated malignancies and management of primary sclerosing cholangitis in inflammatory bowel disease patients: a multicentre retrospective cohort study. J Crohns Colitis 2019;13(12):1492–1500CrossRef Guerra I, Bujanda L, Castro J, Merino O, Tosca J, Camps B, et al. Clinical characteristics, associated malignancies and management of primary sclerosing cholangitis in inflammatory bowel disease patients: a multicentre retrospective cohort study. J Crohns Colitis 2019;13(12):1492–1500CrossRef
3.
Zurück zum Zitat Tabibian JH, Ali AH, Lindor KD. Primary sclerosing cholangitis, part 1: epidemiology, etiopathogenesis, clinical features, and treatment. Gastroenterol Hepatol (N Y) 2018;14(5):293–304 Tabibian JH, Ali AH, Lindor KD. Primary sclerosing cholangitis, part 1: epidemiology, etiopathogenesis, clinical features, and treatment. Gastroenterol Hepatol (N Y) 2018;14(5):293–304
4.
Zurück zum Zitat Gow PJ, Chapman RW. Liver transplantation for primary sclerosing cholangitis. Liver 2000;20(2):97–103CrossRef Gow PJ, Chapman RW. Liver transplantation for primary sclerosing cholangitis. Liver 2000;20(2):97–103CrossRef
5.
Zurück zum Zitat Liu K, Wang R, Kariyawasam V, Wells M, Strasser SI, McCaughan G, et al. Epidemiology and outcomes of primary sclerosing cholangitis with and without inflammatory bowel disease in an Australian cohort. Liver Int 2017;37(3):442–448CrossRef Liu K, Wang R, Kariyawasam V, Wells M, Strasser SI, McCaughan G, et al. Epidemiology and outcomes of primary sclerosing cholangitis with and without inflammatory bowel disease in an Australian cohort. Liver Int 2017;37(3):442–448CrossRef
6.
Zurück zum Zitat Freeman E, Majeed A, Kemp W, Roberts SK. Long-term outcomes of primary sclerosing cholangitis: an Australian non-transplant tertiary hospital perspective. Intern Med J 2019;49(3):323–327CrossRef Freeman E, Majeed A, Kemp W, Roberts SK. Long-term outcomes of primary sclerosing cholangitis: an Australian non-transplant tertiary hospital perspective. Intern Med J 2019;49(3):323–327CrossRef
7.
Zurück zum Zitat Ngu JH, Gearry RB, Wright AJ, Stedman CA. Inflammatory bowel disease is associated with poor outcomes of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol 2011;9(12):1092–1097 (quiz e135)CrossRef Ngu JH, Gearry RB, Wright AJ, Stedman CA. Inflammatory bowel disease is associated with poor outcomes of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol 2011;9(12):1092–1097 (quiz e135)CrossRef
8.
Zurück zum Zitat European Association for the Study of the L. EASL Clinical Practice Guidelines: management of cholestatic liver diseases. J Hepatol 2009;51(2):237–267CrossRef European Association for the Study of the L. EASL Clinical Practice Guidelines: management of cholestatic liver diseases. J Hepatol 2009;51(2):237–267CrossRef
9.
Zurück zum Zitat Chapman MH, Thorburn D, Hirschfield GM, Webster GGJ, Rushbrook SM, Alexander G, et al. British Society of Gastroenterology and UK-PSC guidelines for the diagnosis and management of primary sclerosing cholangitis. Gut 2019;68(8):1356–1378CrossRef Chapman MH, Thorburn D, Hirschfield GM, Webster GGJ, Rushbrook SM, Alexander G, et al. British Society of Gastroenterology and UK-PSC guidelines for the diagnosis and management of primary sclerosing cholangitis. Gut 2019;68(8):1356–1378CrossRef
10.
Zurück zum Zitat Lindor KD, Kowdley KV, Harrison ME, American College of G. ACG clinical guideline: primary sclerosing cholangitis. Am J Gastroenterol 2015;110(5):646–659 (quiz 60)CrossRef Lindor KD, Kowdley KV, Harrison ME, American College of G. ACG clinical guideline: primary sclerosing cholangitis. Am J Gastroenterol 2015;110(5):646–659 (quiz 60)CrossRef
11.
Zurück zum Zitat Maaser C, Sturm A, Vavricka SR, Kucharzik T, Fiorino G, Annese V, et al. ECCO-ESGAR guideline for diagnostic assessment in IBD part 1: initial diagnosis, monitoring of known IBD, detection of complications. J Crohns Colitis 2019;13(2):144–164CrossRef Maaser C, Sturm A, Vavricka SR, Kucharzik T, Fiorino G, Annese V, et al. ECCO-ESGAR guideline for diagnostic assessment in IBD part 1: initial diagnosis, monitoring of known IBD, detection of complications. J Crohns Colitis 2019;13(2):144–164CrossRef
12.
Zurück zum Zitat Schuppan D, Afdhal NH. Liver cirrhosis. Lancet 2008;371(9615):838–851CrossRef Schuppan D, Afdhal NH. Liver cirrhosis. Lancet 2008;371(9615):838–851CrossRef
13.
Zurück zum Zitat Dickman PW, Sloggett A, Hills M, Hakulinen T. Regression models for relative survival. Stat Med 2004;23(1):51–64CrossRef Dickman PW, Sloggett A, Hills M, Hakulinen T. Regression models for relative survival. Stat Med 2004;23(1):51–64CrossRef
14.
Zurück zum Zitat Barner-Rasmussen N, Pukkala E, Jussila A, Farkkila M. Epidemiology, risk of malignancy and patient survival in primary sclerosing cholangitis: a population-based study in Finland. Scand J Gastroenterol 2020;55(1):74–81CrossRef Barner-Rasmussen N, Pukkala E, Jussila A, Farkkila M. Epidemiology, risk of malignancy and patient survival in primary sclerosing cholangitis: a population-based study in Finland. Scand J Gastroenterol 2020;55(1):74–81CrossRef
15.
Zurück zum Zitat Liang H, Manne S, Shick J, Lissoos T, Dolin P. Incidence, prevalence, and natural history of primary sclerosing cholangitis in the United Kingdom. Medicine (Baltimore) 2017;96(24): e7116CrossRef Liang H, Manne S, Shick J, Lissoos T, Dolin P. Incidence, prevalence, and natural history of primary sclerosing cholangitis in the United Kingdom. Medicine (Baltimore) 2017;96(24): e7116CrossRef
16.
Zurück zum Zitat Wiesner RH, Grambsch PM, Dickson ER, Ludwig J, MacCarty RL, Hunter EB, et al. Primary sclerosing cholangitis: natural history, prognostic factors and survival analysis. Hepatology 1989;10(4):430–436CrossRef Wiesner RH, Grambsch PM, Dickson ER, Ludwig J, MacCarty RL, Hunter EB, et al. Primary sclerosing cholangitis: natural history, prognostic factors and survival analysis. Hepatology 1989;10(4):430–436CrossRef
17.
Zurück zum Zitat Tafur M, Cheung A, Menezes RJ, Feld J, Janssen H, Hirschfield GM, et al. Risk stratification in primary sclerosing cholangitis: comparison of biliary stricture severity on MRCP versus liver stiffness by MR elastography and vibration-controlled transient elastography. Eur Radiol 2020;30(7):3735–3747CrossRef Tafur M, Cheung A, Menezes RJ, Feld J, Janssen H, Hirschfield GM, et al. Risk stratification in primary sclerosing cholangitis: comparison of biliary stricture severity on MRCP versus liver stiffness by MR elastography and vibration-controlled transient elastography. Eur Radiol 2020;30(7):3735–3747CrossRef
18.
Zurück zum Zitat Prince M, Chetwynd A, Newman W, Metcalf JV, James OF. Survival and symptom progression in a geographically based cohort of patients with primary biliary cirrhosis: follow-up for up to 28 years. Gastroenterology 2002;123(4):1044–1051CrossRef Prince M, Chetwynd A, Newman W, Metcalf JV, James OF. Survival and symptom progression in a geographically based cohort of patients with primary biliary cirrhosis: follow-up for up to 28 years. Gastroenterology 2002;123(4):1044–1051CrossRef
19.
Zurück zum Zitat Weismuller TJ, Trivedi PJ, Bergquist A, Imam M, Lenzen H, Ponsioen CY, et al. Patient age, sex, and inflammatory bowel disease phenotype associate with course of primary sclerosing cholangitis. Gastroenterology 2017;152(8):1975–1984CrossRef Weismuller TJ, Trivedi PJ, Bergquist A, Imam M, Lenzen H, Ponsioen CY, et al. Patient age, sex, and inflammatory bowel disease phenotype associate with course of primary sclerosing cholangitis. Gastroenterology 2017;152(8):1975–1984CrossRef
20.
Zurück zum Zitat Boonstra K, Weersma RK, van Erpecum KJ, Rauws EA, Spanier BW, Poen AC, et al. Population-based epidemiology, malignancy risk, and outcome of primary sclerosing cholangitis. Hepatology 2013;58(6):2045–2055CrossRef Boonstra K, Weersma RK, van Erpecum KJ, Rauws EA, Spanier BW, Poen AC, et al. Population-based epidemiology, malignancy risk, and outcome of primary sclerosing cholangitis. Hepatology 2013;58(6):2045–2055CrossRef
21.
Zurück zum Zitat Farrant JM, Hayllar KM, Wilkinson ML, Karani J, Portmann BC, Westaby D, et al. Natural history and prognostic variables in primary sclerosing cholangitis. Gastroenterology 1991;100(6):1710–1717CrossRef Farrant JM, Hayllar KM, Wilkinson ML, Karani J, Portmann BC, Westaby D, et al. Natural history and prognostic variables in primary sclerosing cholangitis. Gastroenterology 1991;100(6):1710–1717CrossRef
22.
Zurück zum Zitat Boberg KM, Bergquist A, Mitchell S, Pares A, Rosina F, Broome U, et al. Cholangiocarcinoma in primary sclerosing cholangitis: risk factors and clinical presentation. Scand J Gastroenterol 2002;37(10):1205–1211CrossRef Boberg KM, Bergquist A, Mitchell S, Pares A, Rosina F, Broome U, et al. Cholangiocarcinoma in primary sclerosing cholangitis: risk factors and clinical presentation. Scand J Gastroenterol 2002;37(10):1205–1211CrossRef
23.
Zurück zum Zitat Rupp C, Rossler A, Zhou T, Rauber C, Friedrich K, Wannhoff A, et al. Impact of age at diagnosis on disease progression in patients with primary sclerosing cholangitis. United Eur Gastroenterol J 2018;6(2):255–262CrossRef Rupp C, Rossler A, Zhou T, Rauber C, Friedrich K, Wannhoff A, et al. Impact of age at diagnosis on disease progression in patients with primary sclerosing cholangitis. United Eur Gastroenterol J 2018;6(2):255–262CrossRef
24.
Zurück zum Zitat Chapman RW, Williamson KD. Are dominant strictures in primary sclerosing cholangitis a risk factor for cholangiocarcinoma? Curr Hepatol Rep 2017;16(2):124–129CrossRef Chapman RW, Williamson KD. Are dominant strictures in primary sclerosing cholangitis a risk factor for cholangiocarcinoma? Curr Hepatol Rep 2017;16(2):124–129CrossRef
25.
Zurück zum Zitat Tischendorf JJ, Meier PN, Strassburg CP, Klempnauer J, Hecker H, Manns MP, et al. Characterization and clinical course of hepatobiliary carcinoma in patients with primary sclerosing cholangitis. Scand J Gastroenterol 2006;41(10):1227–1234CrossRef Tischendorf JJ, Meier PN, Strassburg CP, Klempnauer J, Hecker H, Manns MP, et al. Characterization and clinical course of hepatobiliary carcinoma in patients with primary sclerosing cholangitis. Scand J Gastroenterol 2006;41(10):1227–1234CrossRef
26.
Zurück zum Zitat Burak K, Angulo P, Pasha TM, Egan K, Petz J, Lindor KD. Incidence and risk factors for cholangiocarcinoma in primary sclerosing cholangitis. Am J Gastroenterol 2004;99(3):523–526CrossRef Burak K, Angulo P, Pasha TM, Egan K, Petz J, Lindor KD. Incidence and risk factors for cholangiocarcinoma in primary sclerosing cholangitis. Am J Gastroenterol 2004;99(3):523–526CrossRef
28.
Zurück zum Zitat Boonstra K, de Vries EM, van Geloven N, van Erpecum KJ, Spanier M, Poen AC, et al. Risk factors for primary sclerosing cholangitis. Liver Int 2016;36(1):84–91CrossRef Boonstra K, de Vries EM, van Geloven N, van Erpecum KJ, Spanier M, Poen AC, et al. Risk factors for primary sclerosing cholangitis. Liver Int 2016;36(1):84–91CrossRef
29.
Zurück zum Zitat Bergquist A, Montgomery SM, Bahmanyar S, Olsson R, Danielsson A, Lindgren S, et al. Increased risk of primary sclerosing cholangitis and ulcerative colitis in first-degree relatives of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol 2008;6(8):939–943CrossRef Bergquist A, Montgomery SM, Bahmanyar S, Olsson R, Danielsson A, Lindgren S, et al. Increased risk of primary sclerosing cholangitis and ulcerative colitis in first-degree relatives of patients with primary sclerosing cholangitis. Clin Gastroenterol Hepatol 2008;6(8):939–943CrossRef
30.
Zurück zum Zitat Bowlus CL, Lim JK, Lindor KD. AGA clinical practice update on surveillance for hepatobiliary cancers in patients with primary sclerosing cholangitis: expert review. Clin Gastroenterol Hepatol 2019;17(12):2416–2422CrossRef Bowlus CL, Lim JK, Lindor KD. AGA clinical practice update on surveillance for hepatobiliary cancers in patients with primary sclerosing cholangitis: expert review. Clin Gastroenterol Hepatol 2019;17(12):2416–2422CrossRef
Metadaten
Titel
Epidemiology and outcomes of primary sclerosing cholangitis: an Australian multicentre retrospective cohort study
verfasst von
Natassia Tan
N. Ngu
T. Worland
T. Lee
T. Abrahams
K. Pandya
E. Freeman
N. Hannah
K. Gazelakis
R. G. Madden
K. D. Lynch
Z. Valaydon
S. Sood
A. Dev
S. Bell
A. Thompson
J. Ding
A. J. Nicoll
K. Liu
P. Gow
J. Lubel
W. Kemp
S. K. Roberts
A. Majeed
the Melbourne Liver Group
Publikationsdatum
03.06.2022
Verlag
Springer India
Erschienen in
Hepatology International / Ausgabe 5/2022
Print ISSN: 1936-0533
Elektronische ISSN: 1936-0541
DOI
https://doi.org/10.1007/s12072-022-10356-1

Weitere Artikel der Ausgabe 5/2022

Hepatology International 5/2022 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Echinokokkose medikamentös behandeln oder operieren?

06.05.2024 DCK 2024 Kongressbericht

Die Therapie von Echinokokkosen sollte immer in spezialisierten Zentren erfolgen. Eine symptomlose Echinokokkose kann – egal ob von Hunde- oder Fuchsbandwurm ausgelöst – konservativ erfolgen. Wenn eine Op. nötig ist, kann es sinnvoll sein, vorher Zysten zu leeren und zu desinfizieren. 

Umsetzung der POMGAT-Leitlinie läuft

03.05.2024 DCK 2024 Kongressbericht

Seit November 2023 gibt es evidenzbasierte Empfehlungen zum perioperativen Management bei gastrointestinalen Tumoren (POMGAT) auf S3-Niveau. Vieles wird schon entsprechend der Empfehlungen durchgeführt. Wo es im Alltag noch hapert, zeigt eine Umfrage in einem Klinikverbund.

Proximale Humerusfraktur: Auch 100-Jährige operieren?

01.05.2024 DCK 2024 Kongressbericht

Mit dem demographischen Wandel versorgt auch die Chirurgie immer mehr betagte Menschen. Von Entwicklungen wie Fast-Track können auch ältere Menschen profitieren und bei proximaler Humerusfraktur können selbst manche 100-Jährige noch sicher operiert werden.

Die „Zehn Gebote“ des Endokarditis-Managements

30.04.2024 Endokarditis Leitlinie kompakt

Worauf kommt es beim Management von Personen mit infektiöser Endokarditis an? Eine Kardiologin und ein Kardiologe fassen die zehn wichtigsten Punkte der neuen ESC-Leitlinie zusammen.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.