risk factors associated to endoscopic retrograde

141
Department of Gastrointestinal Surgery, Abdominal Centre Helsinki University Hospital Helsinki, Finland Doctoral Program in Clinical Research University of Helsinki Helsinki, Finland RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE CHOLANGIOPANCREATOGRAPHY RELATED COMPLICATIONS Shamel Ismail ACADEMIC DISSERTATION To be presented, with the permission of the Faculty of Medicine of the University of Helsinki, for public examination in Lecture room 2, Haartman Institute, on 18 December 2020, at 12 noon. Helsinki 2020

Upload: others

Post on 05-Jun-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

Department of Gastrointestinal Surgery, Abdominal Centre

Helsinki University Hospital

Helsinki, Finland

Doctoral Program in Clinical Research

University of Helsinki

Helsinki, Finland

RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE CHOLANGIOPANCREATOGRAPHY RELATED COMPLICATIONS

Shamel Ismail

ACADEMIC DISSERTATION

To be presented, with the permission of the Faculty of Medicine of the University of Helsinki, for public examination in

Lecture room 2, Haartman Institute, on 18 December 2020, at 12 noon.

Helsinki 2020

Page 2: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

SUPERVISORS:

Docent Leena Kylänpää, M.D., Ph.D.

Department of Gastrointestinal Surgery, Abdominal Centre

Helsinki University Hospital and University of Helsinki

Helsinki, Finland

Docent Marianne Udd, M.D., Ph.D.

Department of Gastrointestinal Surgery, Abdominal Centre

Helsinki University Hospital and University of Helsinki

Helsinki, Finland

REVIEWERS: Professor Juha Saarnio M.D., Ph.D.

Department of Surgery

Cancer and Translational Medicine Research Unit

University of Oulu and Oulu University Hospital

Oulu, Finland

Professor Tuomo Rantanen M.D., Ph.D.

Faculty of Health Sciences

University of Eastern Finland

Kuopio, Finland

OPPONENT: Docent Urban Arnelo M.D., Ph.D.

Department of Surgery, Umeå University Hospital, Umeå, and

Division of Surgery, CLINTEC, Karolinska Institutet, Stockholm, Sweden

ISBN 978-951-51-6883-2 (paperback)

ISBN 978-951-51-6884-9 (PDF)

http://ethesis.helsinki.fi

Unigrafia Oy

Helsinki 2020

Page 3: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

2

To Omar and Sami

Page 4: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

3

Table of Contents

ORIGINAL PUBLICATIONS ........................................................................................................................................... 6

1. ABSTRACT ............................................................................................................................................................... 7

2. ABBREVIATIONS ..................................................................................................................................................... 9

3. INTRODUCTION .................................................................................................................................................... 11

4. LITERATURE REVIEW ............................................................................................................................................. 12

4.1 INDICATIONS OF ERCP ................................................................................................................................................. 12

4.1.1 Primary sclerosing cholangitis (PSC) ............................................................................................................... 13

4.1.2 Familial adenomatous polyposis (FAP) ........................................................................................................... 14

4.2 BILIARY CANNULATION .................................................................................................................................................. 14

4.2.1 Primary technique ........................................................................................................................................... 14

4.2.2 Standard cannula or sphincterotome with a guidewire or contrast ............................................................... 14

4.2.3 Pancreatic guidewire-assisted biliary cannulation (PGW) .............................................................................. 15

4.2.4 Precut papillotomy .......................................................................................................................................... 16

4.2.5 Transpancreatic biliary sphincterotomy (TPBS) .............................................................................................. 17

4.2.6 The pancreatic duct stent technique ............................................................................................................... 18

4.2.7 Papillectomy as a cannulation technique ........................................................................................................ 18

4.3 PROCEDURES DURING ERCP .......................................................................................................................................... 18

4.3.1 Biliary procedures ............................................................................................................................................ 18

4.3.2 Pancreatic procedures ..................................................................................................................................... 20

4.3.3 Cannulation and Sphincterotomy of minor papilla ......................................................................................... 20

4.3.4 Papillectomy to treat ampullary adenomas .................................................................................................... 21

4.4 CLASSIFICATION OF ERCP COMPLICATIONS ....................................................................................................................... 22

4.5 POST ERCP PANCREATITIS (PEP) ................................................................................................................................... 24

4.5.1 Incidence ......................................................................................................................................................... 24

4.5.2 Pathophysiology .............................................................................................................................................. 24

4.5.3 Operator-related risk factors .......................................................................................................................... 24

Page 5: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

4

4.5.4 Patient related risk factors .............................................................................................................................. 25

4.5.5 Procedure-related risk factors ......................................................................................................................... 25

4.5.6 Prevention ....................................................................................................................................................... 26

4.6 TECHNIQUE MODIFICATION TO PREVENT PEP .................................................................................................................... 26

4.6.1 Guidewire vs. contrast into the pancreatic duct ............................................................................................. 26

4.6.2 Contrast medium ............................................................................................................................................. 27

4.6.3 Difficult cannulation ........................................................................................................................................ 27

4.6.4 Intraoperative CBD cannulation ...................................................................................................................... 28

4.6.5 Pancreatic stent placement for PEP prophylaxis ............................................................................................. 29

4.7 MEDICATIONS AND HYDRATION TO PREVENT PEP .............................................................................................................. 30

4.7.1 Nonsteroidal anti-inflammatory drugs (NSAIDs) ............................................................................................ 30

4.7.2 Somatostatin and octreotide .......................................................................................................................... 30

4.7.3 Other medications ........................................................................................................................................... 31

4.7.4 Hydration ........................................................................................................................................................ 32

4.8 BLEEDING................................................................................................................................................................... 33

4.8.1 An endoscopic papillary balloon dilatation (EPBD) as an alternative to sphincterotomy ............................... 34

4.9 CHOLANGITIS .............................................................................................................................................................. 35

4.10 PERFORATION ........................................................................................................................................................... 36

4.11 STENT RELATED COMPLICATIONS ................................................................................................................................... 39

4.12 PERFORMANCE MEASURES (PMS) FOR ERCP ................................................................................................................. 39

4.13 ROUTINE PREOPERATIVE LABORATORY TESTING (RPLT) ..................................................................................................... 40

5. AIMS OF THE STUDY ............................................................................................................................................. 42

6. MATERIALS AND METHODS .................................................................................................................................. 43

6.1 STUDY I ..................................................................................................................................................................... 43

6.2 STUDY II .................................................................................................................................................................... 44

6.3 STUDY III ................................................................................................................................................................... 46

6.4 STUDY IV ................................................................................................................................................................... 47

6.5 STATISTICAL ANALYSIS ................................................................................................................................................... 48

7. RESULTS ................................................................................................................................................................ 50

Page 6: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

5

7.1 STUDY I ..................................................................................................................................................................... 50

7.1.1 Cannulation success rate ................................................................................................................................. 50

7.1.2 PEP incidence, predicting and protective factors ............................................................................................ 50

7.1.3 Other complications ........................................................................................................................................ 51

7.2 STUDY II .................................................................................................................................................................... 52

7.2.1 Non FAP benign papillary tumours.................................................................................................................. 52

7.2.2 FAP related papillary tumours......................................................................................................................... 54

7.2.3 Ampullary cancer ............................................................................................................................................ 54

7.3 STUDY III ................................................................................................................................................................... 55

7.3.1 Primary and rescue cannulation methods ....................................................................................................... 55

7.3.2 PEP .................................................................................................................................................................. 57

7.3.3 Difficult cannulation ........................................................................................................................................ 57

7.3.4 TPBS as a rescue method ................................................................................................................................ 58

7.3.5 Other complications ........................................................................................................................................ 58

7.4 STUDY IV ................................................................................................................................................................... 58

7.4.1 Post-ERCP complications ................................................................................................................................. 59

7.4.2 Complications due to sedation ........................................................................................................................ 60

8. DISCUSSION .......................................................................................................................................................... 63

8.1 ERCP FOR PSC PATIENTS (I) .......................................................................................................................................... 63

8.2 ENDOSCOPIC PAPILLECTOMY (II) ..................................................................................................................................... 66

8.3 THE ROLE OF TPBS IN DIFFICULT CANNULATIONS (III) ......................................................................................................... 67

8.4 RPLT FOR ERCP (IV) ................................................................................................................................................... 69

8.5 LIMITATIONS OF THE STUDIES ......................................................................................................................................... 70

9. FUTURE ASPECTS .................................................................................................................................................. 71

10. SUMMARY AND CONCLUSIONS .......................................................................................................................... 73

11. ACKNOWLEDGEMENTS ....................................................................................................................................... 74

12. REFERENCES ........................................................................................................................................................ 76

13. ORIGINAL PUBLICATIONS .................................................................................................................................. 108

Page 7: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

6

Original publications

1. Ismail, S., Kylänpää, L., Mustonen, H., Halttunen, J., Lindström, O. K., Jokelainen, K., Färkkilä, M. A. (2012). Risk factors for complications of ERCP in primary sclerosing cholangitis. Endoscopy, 44(12), 1133-1138. https://doi.org/10.1055/s-0032-1325677

2. Ismail, S., Udd, M., Järvinen, H., Halttunen, J., & Kylänpää, M-L. (2014). Endoscopic papillectomy, single-centre experience. Surgical Endoscopy, 28(11), 3234-3239. https://doi.org/10.1007/s00464-014-3596-5

3. Ismail, S., Udd, M., Lindström, O., Rainio, M., Halttunen, J., & Kylänpää, L. (2019). Criteria for difficult biliary cannulation: start to count. European Journal of Gastroenterology and Hepatology, 31(10), 1200-1205. https://doi.org/10.1097/MEG.0000000000001515

4. Jokelainen, J., Ismail, S., Kylänpää, L., Udd, M., Mustonen, H., Lindström, O., & Pöyhiä, R. (2019). Effect and Predictive Value of Routine Preoperative Laboratory Testing for Endoscopic Retrograde Cholangiopancreatography. Scandinavian Journal of Surgery. https://doi.org/10.1177/1457496918822616

Page 8: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

7

1. Abstract

Endoscopic retrograde cholangiopancreatography (ERCP) is a minimally invasive procedure

for biliary and pancreatic disorders. It has evolved to predominantly therapeutic procedure

with the diagnosis of primary sclerosing cholangitis (PSC) being one of the main exceptions.

ERCP has the high potential for complications, like post ERCP pancreatitis (PEP), bleeding,

cholangitis and perforation. Therefore, the data concerning the risk factors of ERCP is

essential. The roles of preoperative laboratory testing and the new criteria of difficult biliary

cannulation are poorly studied. ERCP plays also the major role in the endoscopic removal of

papillary tumours (European Society of Gastrointestinal Endoscopy, European Association

for the Study of the Liver, 2017).

The main purpose of this study is to assess the safety of ERCP for different groups of

patients, and the adverse effects that can follow. Additionally, the study examines the effect

of using different tools during the procedure on the complication rate and the means to

minimize the complication rates in the future. The main complications after ERCP are post

ERCP pancreatitis (PEP), bleeding, cholangitis and perforation.

The risk factors of ERCP complications in 441 consecutive PCS patients were retrospectively

analysed. The outcome of 61 endoscopic papillectomies was retrospectively studied in study

II. In the prospective study (III) of 821 consecutive patients with native papilla were evaluated

the complication rates in difficult cannulation cases. To evaluate the relationship between

ERCP complications and the need for routine preoperative laboratory tests (RPLT) before

the ERCP procedure, we conducted prospective study (IV) with 1196 patients.

In PSC patients (study I) the PEP and cholangitis rates were 7 and 1.4 %, respectively. PEP

predictive factors are female sex (Odds ratio 2.6, p=0.015), and a guide wire slipping in the

pancreatic duct (PD) (Odds ratio 8.2, p<0.01). The risk of PEP is directly proportional to the

number of times the guide wire slips into the pancreatic duct. A previous endoscopic biliary

sphincterotomy (EST) was found to be a protective factor from PEP (Odds ratio 0.28, p=

0.02). In benign tumours of the papilla (study II), the recurrence rate after resection was

Page 9: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

8

25.5% (study II). Altogether 5 (9.5%) patients underwent a pancreaticoduodenectomy, and

46 out of 51 (90.5%) were treated endoscopically. Obstruction of the bile duct and jaundice

as a manifestation were risk factors for malignancy (p<0.001). PEP occurred in 6 cases

(9.8%). After a benign tumour resection and placing a stent, the PEP rate decreased

(p=0.045). The bleeding rate was 18%. In study (III), when the primary cannulation method

used to cannulate the bile duct succeeded, the PEP rate was 2.3%, but when the surgeon

used advanced methods to achieve the cannulation, the PEP rate increased up to 13.5%.

The primary cannulation success rate was 79% in the absence of difficult cannulation criteria

compared to 20.4% when at least one criteria was present (p<0.001). Broad RPLT could not

predict the adverse effects of ERCP in study (IV).

In conclusion, the new criteria of difficult biliary cannulation are important because the

complication risk increases with the complexity of cannulation. Endoscopic papillectomy for

benign tumours is feasible and relatively safe. The role of RPLT in safety of ERCP is minimal.

Page 10: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

9

2. Abbreviations

ASA American Society of Anaesthesiologists

ASGE American Society for Gastrointestinal Endoscopy

BBC Basic blood count

CBD Common bile duct

CI Confidence interval

CT Computed tomography

DGW Double guide wire

EPBD Endoscopic papillary balloon dilation

EPLBD Endoscopic papillary large-balloon dilation

ERC Endoscopic retrograde cholangiography

ERP Endoscopic retrograde pancreatography

ERCP Endoscopic retrograde cholangiopancreatography

EST Endoscopic sphincterotomy

ESGE European Society of Gastrointestinal Endoscopy

EUS Endoscopic ultrasound

FAP Familial adenomatous polyposis

ICU Intensive care unit

IPMN Intraductal papillary mucinous neoplasia

LRS Lactated Ringer’s solution

MRCP Magnetic resonance cholangiopancreatography

NSAID Nonsteroidal anti-inflammatory drug

NSS Normal saline solution

PD Pancreatic duct

PEP Post-ERCP pancreatitis

P-INR International normalized ratio

Page 11: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

10

PSC Primary sclerosing cholangitis

P-TT Thromboplastin time

PM Performance measures

RCT Randomized control trial

RPLT Routine preoperative laboratory testing

SEMS Self-expandable metal stent

SGW Single guidewire technique

SOD Sphincter of Oddi dysfunction

TPBS Transpancreatic biliary sphincterotomy

Page 12: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

11

3. Introduction

Endoscopic retrograde cholangiopancreatography (ERCP) is an endoscopic technique in

which a side-viewing upper endoscope is guided into the duodenum, allowing for instruments

to be passed into the bile and pancreatic ducts. Then a contrast injection is applied permitting

radiologic visualization and allowing for a variety of therapeutic interventions. ERCP is a

complex endoscopic procedure since it requires specialized equipment and requires a steep

learning curve to develop proficiency in its use. Its benefits in the minimally invasive

management of biliary and pancreatic disorders are challenged by a higher potential of

serious complications than any other standard endoscopic technique. ERCP is used primarily

to diagnose and treat conditions of the bile ducts and main pancreatic duct,

including gallstones, strictures, and leaks as result of surgery or trauma. In the past ERCP

was performed for diagnostic and therapeutic reasons, but now the ERCP is used rarely for

diagnostic reasons in the presence of safer and relatively non-invasive investigations such

as magnetic resonance cholangiopancreatography (MRCP) and endoscopic ultrasound. In

cases of primary sclerosing cholangitis (PSC), brush cytology is an important method used

to predict timing for transplantation (Vannas et al., 2017).

Page 13: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

12

4. Literature review

4.1 Indications of ERCP

Choledocholithiasis is the most common reason for undertaking ERCP (Wang, X. et al.,

2017). In pancreatic cancer, biliary obstruction is another common indication for the use of

ERCP (Gohil et al., 2017). When surgery is delayed, patients with pancreatic cancer and with

jaundice complications need preoperative biliary drainage (Singhirunnusorn et al., 2013).

ERCP is used in biliary strictures to obtain brush cytology and intraductal biopsies

(Navaneethan, Njei et al., 2015), and in the diagnosis and management of

cholangiocarcinoma (Fogel et al., 2006; Lindberg et al., 2006).

ERCP is essential in the evaluation and treatment of benign biliary strictures, postoperative

biliary strictures and anastomotic strictures after liver transplantation (Morelli et al., 2003;

Shah et al., 2004; Verdonk et al., 2006; Chang et al., 2016). ERCP should be performed as

first-line therapy for postoperative biliary leakage (Luigiano et al., 2016; Rainio et al., 2018).

Patients with acute biliary pancreatitis and concomitant cholangitis or biliary obstruction need

urgent ERCP (Tse et al., 2012; ASGE Standards of Practice Committee et al., 2019). ERCP

is used in the treatment of pancreatic pseudocysts and pancreatic fluid collections (Lehman,

2002; Tyberg et al., 2016). Additionally, ERCP is used in the treatment of symptomatic

dominant pancreatic duct strictures (Seza et al., 2011), and in case of pancreatic duct

leakage (Kim, S. et al., 2017). ERCP can be used in the treatment of pancreatic stones

following the extracorporeal shock wave lithotripsy in the case of large stones, or without for

small stones (Tandan et al., 2016). In patients with main pancreatic duct intraductal papillary

mucinous neoplasia (IPMN), ERCP is used for cytology (Yoshioka et al., 2016). ERCP with

empiric biliary or pancreatic sphincterotomy is an alternative in evaluation of idiopathic acute

recurrent pancreatitis (suspected type 2 pancreatic SOD) when the findings on an

Page 14: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

13

endoscopic ultrasound (EUS) or MRCP are normal and do not suggest suspected biliary

stones, sludge, or chronic pancreatitis (Rehman et al. 2013).

It is generally recommended that all patients not undergo routine preprocedure laboratory

testing, chest radiography, or electrocardiography prior to endoscopic procedures. Instead,

preprocedure testing should be used selectively based on the patient's medical history,

physical examination findings, and procedural risk factors (ASGE Standards of Practice

Committee et al., 2014).

We typically obtain laboratory tests prior to the procedure if an intervention is anticipated

(such as sphincterotomy) and blood tests have not been obtained as part of the patient's prior

evaluation. Such tests include a complete blood count and prothrombin time. Also, pregnancy

test for potential pregnancy, coagulation studies for patients with active bleeding and with an

increased risk of bleeding due to medication use, and in patients with prolonged biliary

obstruction. Chest radiograph for patients with new respiratory symptoms or decompensated

heart failure.

4.1.1 Primary sclerosing cholangitis (PSC)

Ampullary adenomas, can occur sporadically or in the context of genetic syndromes such as

FAP. Autopsy series has estimated the prevalence of sporadic ampullary adenoma to be

between 0,04 and 0,12% (Park, S. W. et al., 2000). Benign tumours of the ampulla of Vater

are rare and include lipomas, lymphangiomas, haemangiomas, leiomyofibromas, and

neurogenic tumours. Villous and tubulovillous adenomas, however, remain the most common

such benign lesions (Culver et al., 2011). The frequency of a malignant lesion in an adenoma

of the papilla figures around 11% (Alali et al., 2020).

PSC is a chronic, inflammatory bile duct disease. It may lead to stenosis of the internal and

external bile ducts and as it progresses it blocks the flow of bile and leads to cirrhosis of the

liver. PSC patients usually have underlying inflammatory bowel disease in 7-13% of cases.

Due to increased risk of cholangiocarcinoma (Boberg et al., 2002) the dysplasia surveillance

Page 15: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

14

using ERC and brush cytology essential to early detection of malignancy (Boyd et al., 2017;

Vannas et al., 2017).

4.1.2 Familial adenomatous polyposis (FAP)

FAP is an autosomal dominant inherited disease, in which hundreds or thousands of

adenomas develop in the colorectum, with a 100 percent risk of developing colorectal cancer.

The incidence of disease is about 2-3 per 100,000 inhabitants (Jarvinen, 1992; Bjork et al.,

1999). The incidence of ampullary tumours in FAP patients is 200-300 fold higher compared

to the general population, with a cumulative risk of up to 10% for carcinoma (Gluck et al.,

2015). The endoscopic papillectomy for benign adenomas is increasingly being performed

(Park, S. W. et al., 2000).

4.2 Biliary cannulation

4.2.1 Primary technique

Standard cannulas with or without a guidewire are limited in their ability to vary the angle of

approach to the papilla. A standard sphincterotome was originally used to allow variable

upward angulation for biliary cannulation. It was bowed to enter the biliary orifice, then

relaxed to achieve deep cannulation (Rossos et al., 1993).

4.2.2 Standard cannula or sphincterotome with a guidewire or contrast

The primary technique to commence ERCP is cannulation with a sphincterotome and

guidewire. Two methods of guidewire assisted cannulation have been used. In the first

method, the sphincterotome is inserted into the papillary orifice and the guidewire is

advanced under fluoroscopic guidance until it is observed to enter the bile duct. In the second

method, the guidewire is advanced 1-2 mm beyond the tip of the cannula and directly pushed

into the papillary orifice in the axis of the common bile duct under fluoroscopic guidance.

Contrast injection is not routinely administered prior to the advancement of the guidewire into

the common bile duct (CBD).

Page 16: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

15

For the contrast assisted biliary cannulation, the tip of the cannula or the sphincterotome is

inserted into the papillary orifice at the 11-o’clock position and a small volume of contrast is

injected under fluoroscopic guidance to define the anatomy of the CBD.

In 3 meta-analyses that include 4, 7 and 12 randomized control trials (RCT) comparing

contrast assisted and guidewire-assisted biliary cannulation, all favoured the guidewire

technique in terms of its significantly higher success rate. In the meta-analysis by Cheung et

al. the success rate of the guidewire-assisted technique compared to the contrast assisted

technique was 89% vs. 78%, respectively, RR1.19 (95% CI, 1.05-1.35), and in the meta-

analysis by Tse et al. the success rate was 83.6% vs. 77.3%, RR1.07 (95% CI, 1.00-1.15)

(Cheung et al., 2009; Tse et al., 2013).

4.2.3 Pancreatic guidewire-assisted biliary cannulation (PGW)

When the primary cannulation technique fails to achieve cannulation, rescue techniques are

used. PGW is used when the wire unintentionally enters the pancreatic duct (PD). The wire

that has been unintentionally placed in the PD is left in place and then the CBD is cannulated

by using a cannula/sphincterotome with a contrast medium injection. This technique is called

single guidewire technique (SGW). Alternatively, the procedure can be carried out by using

an additional guidewire, which is known as the double guidewire technique (DGW). The wire

in the PD straightens the PD and the common channel and stabilises the papilla and

increases the chances of cannulating the CBD (Freeman et al., 2005).

In one RCT that compared the DGW and persistent standard cannulation technique in difficult

cannulations found a significantly higher success cannulation rate in the DGW group (93%

vs. 54%, P<0.05), with no difference in PEP or other complications (Maeda et al., 2003;

Gronroos et al., 2011). Another RCT comparing DGW to the precut technique found a

significantly higher PEP rate in the DGW group (38% vs. 11%, p=0.01), although there was

no difference in successful cannulation rates (Yoo et al., 2013).

Page 17: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

16

In a systematic review of 7 RCTs with a total of 577 patients, comparing to other persistent

conventional contrast or guidewire assisted cannulations, the DGW technique was

associated with increased risk of PEP and there was no significant difference in CBD

cannulation success, or in the risk of other ERCP complications (Tse et al., 2017). However,

not all the studies in this meta-analysis were blinded. Additionally, most patients included in

the studies had no NSAID medication administered, and no prophylactic pancreatic stent

was applied. In one RCT the placing of a prophylactic pancreatic stent after DGW cannulation

significantly reduced the risk of PEP (Ito et al., 2010). In another retrospective study, in which

prophylactic pancreatic stenting was tried after every DGW-assisted attempt to cannulate the

CBD, applying the stent was successful in 86% of patients and the PEP incidence rate in this

group was significantly lower compared to the group in which the stenting failed (5% vs. 30%,

p =0.0073) (Ito et al., 2014). DGW is a rescue technique and should be reserved for cases

with difficult cannulation.

4.2.4 Precut papillotomy

Precut papillotomy is a commonly used technique to gain access to the CBD in difficult cases

(Liu, Y. et al., 2012). In one method (the conventional precut technique) a needle knife is

used to access the biliary orifice and a cut is made in the biliary orientation towards 11 o’clock.

Once the bile duct is visualized, cannulation is done through this opening using a needle

knife catheter, sphincterotome, or regular catheter. In the fistulotomy method, the needle

knife accesses the bile duct superior to the orifice in the 11 o’clock direction. A meta-analysis

of 6 RCTs with a total of 966 patients showed a significantly lower rate of PEP in the early

precut group than in the persistent attempt group (OR 0.47, 95% CI 0-24-0.91) (Cennamo et

al., 2010). Another meta-analysis that included 7 RCTs and a total of 1,039 patients, showed

a tendency of a decreased rate of PEP after an early precut, but this was not statistically

significant (3.9 % in the precut sphincterotomy vs. 6.1 % in the persistent attempts group;

OR 0.58, 95 % CI 0.32-1.05; P = 0.08) (Navaneethan et al., 2014). In a prospective,

multicentre trial, 375 patients were randomized in two groups: the early precut group and the

Page 18: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

17

late precut group. The group with an early precut was found to have a lower PEP rate

compared to the late precut group at 5.4% (10/185) vs. 14.1% (19/135) (Mariani et al., 2016).

A meta-analysis of six RCTs examined two precut techniques. Four RCTs used the

conventional precut method, whereas two RCTs used needle knife fistulotomy. The meta-

analysis showed that fistulotomy significantly reduced the odds of PEP (OR = 0.27, 95 % CI

0.09-0.82, P = 0.02). The overall cannulation rates were comparable between both precut

techniques (OR = 0.90, 95% CI 0.59-1.37, P = 0.62) (Choudhary et al., 2014). Both

techniques are safe and successful but should be performed by experts in high volume

centres.

4.2.5 Transpancreatic biliary sphincterotomy (TPBS)

Transpancreatic biliary sphincterotomy introduced by John S. Goff in 1995, is gaining

popularity as a rescue technique for difficult cannulations (Goff, 1995). After a guidewire

accidentally goes into PD, pancreatic sphincterotomy is done towards 11 o’clock. The

purpose is to make an incision through the septum between the pancreatic and biliary duct,

which exposes the bile duct orifice (Ahmed et al., 2017). This technique requires special

expertise. Three RCTs with a total of 364 patients and two prospective studies with more

than 300 patients suggest that the rate of successful biliary cannulation following TPBS

ranges from 85% to 100% (median 92.9%), and the rate of adverse events ranges from 3.5%

to 20.5% (median 13.1%), with PEP occurring in 3.5% to 22.4% of cases (median 10.4%)

(Catalano et al., 2004; Kahaleh et al., 2004; Yoo et al., 2013; Zang et al., 2014; Lee, Y. J. et

al., 2015). A meta-analysis that included five prospective and eight retrospective studies

comparing TPBS to the precut technique in terms of the success rate and complications

found that the success rate in the precut group was significantly lower (OR 0.50, P= 0.046),

and the bleeding rate was higher (OR 2.24, P= 0.02), with no difference in the PEP rate (OR

0.79, P = 0.24; RR 0.80, P = 0.19) (Pecsi et al., 2017).

Page 19: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

18

4.2.6 The pancreatic duct stent technique

The pancreatic duct stent technique is an adjunct to the double wire guided method where a

single stent (usually 5Fr x 2-8cm) is placed after an unintentional PD cannulation and then

the BD cannulation is carried out using a guide wire technique. The stent aids in adjusting

the orientation of the bile duct by straightening the distal choledochus to facilitate the biliary

cannulation. In a retrospective study of native papilla, pancreatic stent placement facilitated

bile duct cannulation in 78.9% (60/76) of cases when cannulation by the standard method

was not achieved (Davee et al., 2012).

4.2.7 Papillectomy as a cannulation technique

Farrell et al. described a novel technique in the case of a bulky ampulla. The large size of an

ampulla because of a tumour or overgrowth makes the cannulation of the CBD very difficult

to achieve. In this case, a polypectomy snare and diathermy in a coagulation current were

used to remove the bulky papilla, then the cannulation can be resumed (Farrell et al., 1996).

Ten patients with failed primary cannulations were included in the study. The cannulation

succeeded in all cases. One case of bleeding occurred, and there was no PEP or cholangitis.

However, a larger study is required to compare this technique to other cannulation methods

to assess its effectiveness and safety.

4.3 Procedures during ERCP

4.3.1 Biliary procedures

Biliary sphincterotomy is instrumental for removing bile duct stones, stricture dilation and for

the treatment of papillary stenosis, and in some cases when stent placement is needed

(Foutch, 1991).

Stone extraction from the CBD is the most common procedure during ERCP. In this

procedure a cannula or a sphincterotome is inserted into the CBD and a contrast medium is

injected to visualize the stones. In the case of a CBD stone, biliary sphincterotomy must be

Page 20: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

19

done first. Then a balloon catheter or wire basket is used to extract the stone. Large stones

may require fragmentation before extraction. Dilatation and stent placement procedures are

used in cases of stenosis and obstruction of the CBD (Wang et al., 2017).

Using balloons, 6-10mm in diameter to dilatate the papilla up to the distal diameter of CBD,

with or without minor endoscopic sphincterotomy (EST) is a good alternative to EST alone

for patients with coagulopathy and for patients with altered anatomy. In a systematic review

and meta-analysis of 7 RCTs, a total of 369 patients underwent EPBD without EST, and 367

patients underwent EPBD with EST. There were no significant changes in the cannulation

success rate (OR 0.69, 95% CI= 0.44-10.19, p= 0.11), or in the PEP rate (OR 0.88, 95%

CI=0.43-1.78, p=0.72) (Liu, P. et al., 2019).

The only indication for an endoscopic papillary large-balloon dilation (EPLBD) is the

extraction of large choledochal stones. A systematic review of 15 RCTs involving 1,768

patients demonstrated significantly higher rates of PEP with EPLBD of an intact biliary

sphincter performed, compared with biliary sphincterotomy (RR 1.96, 95% CI, 1.34-2.89)

(Weinberg et al., 2006).

A papillectomy is possible to do endoscopically, but has an increased risk of PEP, which can

be reduced by PD stenting (Harewood et al., 2005).

In the case of biliary malignancy, brush cytology of the bile duct is the method of choice.

According to a recent study, the specificity of brush cytology is 90%, and the sensitivity is

56% (Geramizadeh et al., 2018). In the case of PSC, brush cytology is an important method

to determine the timing for transplantation (Vannas et al., 2017).

With per oral cholangioscopy it is possible to carry out manoeuvres under direct visualization

of the CBD. Cholangioscopy with or without directed lithotripsy can clear CBD stones with a

success rate of 73%-93% (Draganov et al., 2011; Kalaitzakis et al., 2012). Cholangioscopy

is also used for targeted biopsies of the bile and pancreatic ducts after undiagnostic brush

cytology or/and a biopsy during ERCP (Parsi, 2014; Navaneethan, Hasan et al., 2015).

Page 21: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

20

The complication rate of cholangioscopy ranges between none up to 7.7% including

bleeding, pain and perforation of the bile duct (Lee, Y. N. et al., 2012; Farnik et al., 2014;

Korrapati et al., 2016). A large nationwide study of ERCP procedures with and without

cholangioscopy, showed that the rate of adverse events of ERCP with cholangioscopy was

higher than without it. The PEP rate in this study was 7.4% vs. 3.9% (P =0.003) respectively,

and the cholangitis rate was 4.4% vs. 2.7% (P=0.0332), respectively (Lubbe et al., 2015).

In patients with an altered anatomy (Roux-en-Y total gasterectomy, Billroth II,

pancreaticoduodenectomy, gastrojejunostomy with duodenal obstruction,

hepaticojejunostomy, gastric bypass), an enteroscopy-assisted ERCP (DB-ERCP) is the

recommended procedure. The cannulation success rate using DB-ERCP is up to 97% and

the cholangiogram success rate is 98% according to one study (Shimatani et al., 2009). In

another study of 712 patients with a Billroth II reconstruction, the biliopancreatic duct

cannulation was successful in 93.8% (Bove et al., 2015) of the cases.

4.3.2 Pancreatic procedures

Pancreatic sphincterotomy is used for further manipulations, such as stent placement in the

PD or for better access to the PD in cases of stricture. In cases of papilla stenosis, and in

cases such as the management of pancreas divisum related strictures, the cutting of the

minor papilla will ensure the normal flow of the pancreatic fluid (Sherman et al., 2001; Chacko

et al., 2008 Clarke et al., 2012). Chronic pancreatitis patients, with manifestations such as

pseudocysts and pancreatic fistulas can be treated by PD stenting (Larsen et al., 2014).

4.3.3 Cannulation and Sphincterotomy of minor papilla

In some patients with pancreas divisum, the identification of the minor papilla can be difficult.

As the pancreatic juice secretion increases, the orifice enlarges making the cannulation easy.

A multi-centre randomized controlled trial showed significantly higher successful cannulation

rates after a porcine secretin injection compared to a placebo (89.3% vs. 6.3%, P<0.001)

(Devereaux et al., 2003). A spray of methylene blue in the area the papilla minor is suspected

Page 22: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

21

to be makes the identification of the papilla easier (Park, S. H. et al., 2003). In the case of

incomplete pancreatic divisum, an injection of a mixture of contrast media and methylene

blue into the major papilla can help to identify the minor papilla (Park et al., 2003).

There are two technical options for carrying out a pancreatic sphincterotomy of the minor

papilla. The first is a needle-knife cut over a plastic stent and the second is the standard pull-

type cut with a sphincterotome. In the retrospective study by Attwell et al. 133 patients

underwent needle-knife cut surgery and 51 patients underwent pull-type cut surgery. The

median follow-up for the needle-knife group was 6 years. Papillary stenosis for this group

was a rate of 24%. The median follow-up for the pull-type cut group was 5 years, with a

stenosis rate of 20%. The overall complication rates were similar in both groups (8.3% vs.

7.8%) (Attwell et al., 2006). Another technique described in a study by Maple et al. is wire

assisted access sphincterotomy. In this technique, the dorsal duct is deeply cannulated with

a guidewire, then the minor papilla cut is performed using a needle-knife, by cutting away

from the guidewire. This technique was compared with a pull-type cut with no significant

difference in complications (Maple et al., 2009).

4.3.4 Papillectomy to treat ampullary adenomas

A papillectomy is used to treat benign tumours of the papilla because it results in less

morbidity compared to surgery. Additionally, papillectomy is used to achieve a more accurate

diagnosis of the malignancy of ampullary tumours. As in colon cancer, the duodenal

adenoma tend to develop cancer in 30% of cases (Seifert et al., 1992). Although biopsies of

the papilla tumours are easy to obtain during an endoscopy, the accuracy of the biopsies in

cases of carcinoma within the adenoma are still low at 14%-66% (Hwang et al., 2008; Carter

et al., 2009), making the papillectomy of a whole tumour more accurate in order to achieve

a malignant diagnosis. However, in distinct malignant tumours surgery is the gold standard

(Irani et al., 2009). In a systematic review, and meta-analysis of five retrospectives, cohort

studies (446 patients) showed that an endoscopic resection was contraindicated when

malignancy manifestations were presented, such as ulcerations, duodenal infiltrations, lateral

Page 23: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

22

spread, polyps sized more than 4 cm or an intraductal extension more than 1 cm (Mendonca

et al., 2016). The complication rate after a papillectomy ranges between 8% and 35% (Ito et

al., 2012; Moon et al., 2014).

4.4 Classification of ERCP complications

Two classifications are available to define the ERCP complications. The first one is the

consensus definition and grading by Cotton et al (Cotton et al., 1991) (Table 1). Another

classification is the revised Atlanta consensus classification of pancreatitis, which grades

pancreatitis into mild with no organ failure and local complications (acute fluid collection

around the pancreas, pseudocyst, acute necrotic collection, pleural effusion), and moderate

with transient organ failure that resolves in less than 48h, with or without local complications,

and severe pancreatitis with single or multiple persistent organ failure for more than 48h

(Banks et al., 2013).

Page 24: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

23

Table 1. Consensus definition and grading of ERCP complications

Mild Moderate Severe Pancreatitis Clinical pancreatitis with

blood amylase levels more the 3 times normal and admission time up to 2-3 days

hospitalization time 4-10 days

Hospitalization time more than 10 days, pseudocyst, intervention needed

Bleeding Haemoglobin levels drop less than 3g, no transfusion needed

Transfusion needed (less than 4 units), but no invasive intervention needed (angiography, surgery)

Transfusion (5 units or more) and invasive intervention needed

Perforation Slight leak of contrast or fluid treated by fluids or suction for 3 days or less

Perforation treated medically for 4-10 days

Medical treatment for more than 10 days or percutaneous, surgical intervention needed

Infection Fever more than 38°c for 24-48h

Fever with more than 3 days hospitalisation or percutaneous intervention needed

Septic shock or surgery

(Cotton et al., 1991).

PEP is the most common and serious complication of ERCP with an incidence of 3.5% -9.7%

(Banks et al., 2013). Bleeding usually occurs as a result of sphincterotomy, with an incidence

of 0.3% - 9.6% (Cotton et al., 2010). Cholangitis is the most common infection complication

associated with ERCP. The incidence ranges between 0.5% and 3% (Masci et al., 2001;

Andriulli et al., 2007; Cotton et al., 2009). The incidence of duodenal perforation during the

ERCP is up to 0.6% (Masci et al., 2001; Cotton et al., 2009). Cardiopulmonary complications

are usually related to sedation during ERCP and include aspiration, hypoxia, cardiac

dysrhythmia and hypotension. The incidence of these complications ranges from 0.07% to

2.4% (Glomsaker et al., 2013; Jokelainen et al., 2017).

Page 25: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

24

4.5 Post ERCP pancreatitis (PEP)

4.5.1 Incidence

In a systematic review of 21 prospective studies with 16,855 patients, PEP was the most

frequent adverse event following ERCP with an incidence of 3.47% (95% CI, 3.19%, 3.75%).

Of these 44.8% were graded mild, 43.8% moderate and 11.4% were graded severe.

Altogether 3% of patients with PEP died (Andriulli et al., 2007). In another systematic review

that included 13,296 patients, the incidence of PEP was 9.7% (95% CI, 8.6%-10.7%), of

which 5.7% of the cases were mild, 2.6% were moderate and 0.5% were severe. In high-risk

patients and procedures (e.g. involving pancreatic sphincterotomy or precut sphincterotomy,

SOD, or a difficult cannulation), the PEP incidence increased to 14.7% (95% CI, 11.8%-

17.7%), of which 8.6% of the cases were mild, 3.9% were moderate and 0.8% were severe.

The mortality rate was 0.2% (Kochar et al., 2015).

4.5.2 Pathophysiology

The mechanism that causes PEP is still poorly known. Mechanical injury of the papilla during

instrumentation, or hydrostatic injury from the contrast injection into the PD may be a reason.

Chemical and allergic injuries may occur from injections of contrast medium into the PD. A

thermal injury may occur from using electrocautery, which can cause an edema and

obstruction of the PD which disturbs the pancreatic secretion flow. An infection theory was

also suggested. These injuries can lead to the premature intracellular activation of proteolytic

enzymes inside acinar cells triggering an inflammatory cascade, resulting in the

autodigestion of the pancreatic tissue (Freeman et al., 2004; Cheng et al., 2006).

4.5.3 Operator-related risk factors

Operator-related risk factors, including prior experience, case volume, and trainee

participation, have been hypothesized to influence the risk of PEP, but this has been difficult

to evaluate because of confounding variables including the complexity of ERCP at high-

Page 26: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

25

volume centres versus low-volume centres. In one prospective study, a total of 3,635

procedures were included, 2,838 (78%) ERCPs were performed in the 11 high-volume

centres (a median of 257 each) and 797 ERCPs were carried out in low-volume centres (a

median of 45 each). Overall, 3,331 ERCPs were carried out by expert operators and 304 by

less-skilled operators. There were significantly more grade 3 difficult procedures in the high-

volume centres than in the low-volume ones (P<0.0001). PEP occurred in 137 patients

(3.8%); the rates did not differ statistically between the high- and low-volume centres (3.9%

vs. 3.1%) and expert and non-expert operators (3.8% vs. 5.5%). However, in the high-volume

centres, there were 25% more patients with patient- and procedure-related risk factors, and

the PEP rate was one-third higher among non-expert operators (Testoni, P. A. et al., 2010).

4.5.4 Patient related risk factors

Patients-related factors include prior PEP, female sex, younger age, normal serum bilirubin

levels and history of acute recurrent pancreatitis and suspected SOD (Freeman et al., 1996;

Cotton et al., 2009; Moffatt et al., 2011; Ding, X. et al., 2015; Chen, J. J. et al., 2014). The

pancreas divisum is an anatomical variation, and by itself is not an independent risk factor

for PEP. However, minor papilla manipulations and sphincterotomies increase the PEP rate

(Moffatt et al., 2011).

4.5.5 Procedure-related risk factors

A difficult cannulation increases the risk of PEP. A difficult cannulation is defined as a

situation, where the procedure performing person, using his regularly used cannulation

technique fails to achieve a BD cannulation in 5 minutes from the first moment that the

guidewire touches the papilla, or after 5 attempts to cannulate the CBD, or after one

unintentional passage or contrast injection into the pancreatic duct (Halttunen et al., 2014).

The above definition of a difficult cannulation has been adopted by the European Society of

Gastrointestinal Endoscopy (ESGE) in their guideline (Testoni, Pier Alberto et al., 2016).

Before that, there was no consensus definition, and different studies adopted different

Page 27: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

26

definitions of a difficult cannulation, which made the comparison of the studies very difficult.

In an easy cannulation, when in less than 5 attempts the cannulation of the BD is achieved,

the PEP rate is as low as 3%. In difficult cannulations the PEP rate increases up to 13-15%

(Freeman et al., 1996; Vandervoort et al., 2002). A pancreatic injection is another risk factor

for PEP (Ding et al., 2015).

4.5.6 Prevention

The indication of ERCP must be clear and the patients must be selected to prevent ERCP

complications. An ERCP should be performed on high-risk patients only after careful

consideration. Some of the high-risk groups are the SOD type III patients (patients with a

functional obstruction of the pancreaticobiliary flow at the level of the Oddi, with symptoms

such as right upper quadrant pain, dilated CBD and abnormality in liver enzymes in SOD

type I, pain and only one objective finding for SOD type II, and only pain in SOD type III). The

procedure should not be offered routinely to these patients (Cotton et al., 2014). There is a

surgical saying that the best way to avoid PEP is not to do an ERCP.

4.6 Technique modification to prevent PEP

4.6.1 Guidewire vs. contrast into the pancreatic duct

The guidewire assisted cannulation technique has less PEP compared to the contrast

assisted cannulation technique. In 2009 two meta-analysis of RCTs were published. In the

first, seven RCTs were included. The use of a guidewire in cannulation significantly reduced

the PEP compared to the contrast assisted cannulation group 3.2% vs. 8.7% (relative risk

RR 0.38%, 95% CI 0.19 – 0.76) (Cheung et al., 2009). The second meta-analysis of five

RCTs showed that the wire-guided technique reduced the PEP rate compared to the contrast

assisted group (OR 0.23, 95% CI, 0.13-0.43) (Cennamo et al., 2009). Two RCTs were later

published but did not confirm the above meta-analysis (Kawakami et al., 2012; Kobayashi et

al., 2013). However, in 2013, another meta-analysis was published that confirmed the

conclusions in Cheung et al. and Cennamo et al.’s meta-analyses. In a large systematic

Page 28: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

27

review and meta-analysis by Tse et al. (2013) that included 12 RCTs, a total of 3,450 patients,

showed that the wire-guided technique significantly reduced PEP risks compared with the

contrast-assisted cannulation technique (RR 0.51, 95 % CI 0.32 - 0.82). In addition, the wire-

guided technique had greater primary success rates in CBD cannulations (RR 1.07, 95 % CI

1.00-1.15), fewer precut sphincterotomies (RR 0.75%, 95% CI 0.60-0.95), and no increase

in other ERCP related complications (Tse et al., 2013).

4.6.2 Contrast medium

Injecting a contrast medium into the PD must be avoided, however, if it is required, the dose

should be as low as possible. In the large meta-analysis by Masci et al. that included 15

prospective trials, the injection of a contrast medium into the PD was an independent risk

factor for PEP (RR 2.2, 95% CI 1.60-3.01) (Masci et al., 2003). In another large retrospective

study that included 14,331 ERCPs, the patients were divided into four groups: the first group

contained patients with a CBD injection alone, the second group were patients who had an

injection into the head part of the PD, the third group had an injection into the body part and

the fourth group had an injection up to the tail part of the PD. The results showed that the

risk of PEP was significantly higher in the last three groups compared to the first one (6.9%

vs. 0.8%, P<0.001) (Cheon et al., 2007).

4.6.3 Difficult cannulation

It is well documented that the PEP incidence increases significantly after a difficult

cannulation. The ESGE guideline definition of a difficult cannulation is based on prospective

study by Halttunen et al. and was published in 2014. Altogether 907 cannulations were

performed by expert operators in Scandinavian healthcare centres. The primary cannulation

was achieved in 75% of the patients, with a median time of 0.88 minute (53 sec) and with

maximum of two attempts and no PD passage or injection. The overall cannulation success

was 97.4%, and the PEP rate was 5.3%. The median time for successful cannulation was

1.55 minutes (0.02-94.2 minutes). If the primary cannulation succeeded, the PEP rate was

Page 29: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

28

2.8%. After crossover to the second cannulation using different cannulation technique

depending on the choice of the operator, the PEP rate increased up to 11.5%. If the

cannulation was achieved within the first 5 minutes, the PEP rate was 2.6%. For procedures

that lasted longer than 5 minutes, the PEP rate was 11.8%. When the cannulation was

achieved in one attempt, the PEP rate was 0.6%, with two attempts 3.1%, with three to four

6.1%, and with five and more 11.9%. With one accidental PD passage the PEP rate was

3.7%, with two, it was 13.1% (Halttunen et al., 2014). Usually when the cannulation appeared

to be difficult, the operator chose to continue the cannulation with standard methods or

crossed over to another technique such as DGW, pre-cut techniques or TPBS. Another thing

to do if the cannulation fails, is to try later after 24-48 hours, or to ask a more experienced

surgeon to do the cannulation.

4.6.4 Intraoperative CBD cannulation

While the operator tries to achieve a cannulation of the CBD, the guidewire can cause injuries

to the papilla that increases the PEP incidence. Therefore, as few cannulation attempts as

possible are recommended. In a systematic review and meta-analysis by Wang et al. that

included five RCTs and a total of 631 patient with gallbladder and suspected CBD stones,

preoperative vs. intraoperative endoscopic sphincterotomies (rendezvous technique) were

compared. The meta-analysis showed that the PEP incidence was higher in the preoperative

endoscopic sphincterotomy group compared to intraoperative group (RR 2.27, 95 % CI 1.18-

4.40, p = 0.01), with no difference in the success rate of CDB stone clearance (RR 0.96, 95

% CI 0.91-1.01, p = 0.13) (Wang, B. et al., 2013). A prospective nationwide study from

Sweden came to the same conclusion that the intraoperative endoscopic technique reduced

the risk of PEP from 3.6% to 2.2% (OR 0.5, 95% CI 0.2-0.9, P=0.02 (Swahn et al., 2013).

Theoretically, every procedure that facilitates CBD access without causing any trauma to the

papilla, and without cannulation or opacification of the PD, should lower the PEP risk.

According to Swahn et al., the rendezvous technique provides cannulation over a guidewire

during laparoscopic cholecystectomy, avoiding trauma of the papilla, and unintentional PD

Page 30: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

29

cannulation. However, intraoperative ERCP during a laparoscopic cholecystectomy prolongs

the operation time and may cause logistical problems.

4.6.5 Pancreatic stent placement for PEP prophylaxis

Two meta-analyses and controlled trials showed that using a pancreatic stent reduces the

PEP rate and severity (Andriulli et al., 2007; Mazaki et al., 2014). A meta-analysis by Mazki

et al. included 14 RCTs, with a total of 1,541 patients who were divided into two groups: the

first group had prophylactic pancreatic stents, and the second had none. The results showed

a significant reduction of PEP in the group with pancreatic stents (RR 0.39 95 % CI 0.29-

0.53, P < 0.001). A nationwide, register-based study of 43,545 ERCPs performed in Sweden

between 2006 and 2014 showed that the patients who received a pancreatic stent ≤ 5 Fr in

diameter had four times more PEP compared to patients who received pancreatic stent > 5

Fr in diameter (OR 3.58; 95% CI 1.40-11.07) (Olsson et al., 2017). In a study by Ito et al. 70

patients who had undergone a DGW assisted cannulation were randomized either to receive

prophylactic pancreatic stenting or not. Pancreatic stenting attempts were successful for 91%

of the patients and the stent had migrated at the end of procedure in 3% of the patients. The

frequency of PEP in the stent group was significantly lower than in the unstented group (3%

vs. 23%, p < 0.05) (Ito et al., 2014).To get the best results and prevent PEP, the stents must

remain in place 12-24 hours according to one randomized, prospective study (Cha et al.,

2013). However, placing a pancreatic stent can be challenging, and in case of failure the PEP

rate increases. A retrospective study which included 146 difficult cannulations showed that

the pancreatic stenting after DGW was successful only in 72% of the cases. The failed

pancreatic stenting cases had significantly higher rates of PEP (30% vs. 5%, P=0.0073) (Ito

et al., 2014). In their meta-analysis Wang et.al. included 23 retrospective cohort studies and

2 RCTs. A sensitivity analysis of the data showed that pancreatic stents significantly reduced

the PEP risk after a papillectomy (OR 0.44, 95% CI, 0.24-0.80, p = 0.007) (Wang, Y. et al.,

2019). Prophylactic pancreatic stenting can be useful in difficult cannulations and for high

risk patients (Berry et al., 2019). The recent ESGE guideline recommends using prophylactic

Page 31: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

30

pancreatic stents in selected patients at high risk of PEP (unintentional guidewire insertion

into to the PD, DGW cannulation) (Dumonceau et al., 2020). Phillip et al. in their RCT (167

patients) showed that pancreatic stents significantly reduced the PEP in unselected patients

when an inadvertent cannulation of the PD occurred (OR 0.43; 95% Cl 0.19-0.98, P=0.04)

(Phillip et al., 2019).

4.7 Medications and hydration to prevent PEP

4.7.1 Nonsteroidal anti-inflammatory drugs (NSAIDs)

In a recent meta-analysis of 16 RCTs (6,458 patients), PEP occurred in 170 of 3,226 (5%)

patients in the NSAID group and in 317 of 3,232 (9.9%) patients in the placebo group. A dose

of 100mg of diclofenac or indomethacin was administered rectally immediately before or after

the procedure, and in one RCT an NSAID was administered during the procedure. NSAID

administration significantly reduced the PEP rate (RR = 0.55; 95%, 0.42-0.71; P<0.01). A

subgroup analysis showed that diclofenac was superior to indomethacin in preventing PEP

(RR = 0.32; 95%, 0.19–0.56, P < 0.01). Additionally, the NSAIDs administered rectally post-

ERCP were superior to NSAIDs that were administered pre-ERCP (RR = 0.39; 95% CI, 0.24–

0.63, P < 0.01) (Hou et al., 2017). However, in a multicentre, single-blinded, randomized

controlled trial of 2,600 patients undergoing ERCP, the PEP rate in the group with

indomethacin administered before the procedure was 4%, and in the other group with

indomethacin administered after the ERCP the PEP rate was 8% (RR 0.47; 95% CI, 0.34-

0.66; P< 0.0001) (Luo et al., 2016). Orally and intramuscularly administered diclofenac had

no effect in preventing PEP (Cheon et al., 2007; Park, S. W. et al., 2015). In high-volume,

low-PEP risk ERCP units, diclofenac seems to have no beneficial effect (Rainio et al., 2017).

4.7.2 Somatostatin and octreotide

A meta-analysis of 18 RCTs, and a total of 3,171 patients that compared octreotide vs. a

placebo found no significant difference in the PEP incidence (OR 0.77, 95% CI 0.56-1.05).

However, in a sub-group analysis of 6 RCTs (1470 patients), the dose of octreotide was

Page 32: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

31

higher than 0.5 mg compared to the other 12 RCTs. With a high dose the OR of PEP dropped

to 0.45 (95% CI 0.28-0.73; NNT=25). When it came to giving the medication before or after

the procedure, the results were inconclusive (Zhang, Y. et al., 2009). In a more recent meta-

analysis of 11 RCTs, a total of 4,192 patients were divided into a somatostatin group and a

placebo group. In the somatostatin group, a decrease was observed in the incidence of PEP

and hyperamylasaemia (RR = 0.63, 95% CI, 0.40-0.98; P = 0.04) (RR = 0.75, 95% CI, 0.66-

0.84; P < 0.001) regardless of the way that the medication was administered (single bolus,

long-term infusion, or high dosage). When a subgroup analysis was performed according to

the area (6 RCTs in Asia and 5 RCTs in Europe), the six Asian studies showed a significant

decrease of PEP, compared to the studies from Europe (RR = 0.48, 95% CI 0.34-0.69; P <

0.001) (RR = 0.77, 95% CI, 0.37-1.61; P = 0.49), respectively (Hu et al., 2016). The results

in the above-mentioned studies are contradictory. Furthermore the use of somatostatin and

octreotide never became a common practice.

4.7.3 Other medications

A meta-analysis of six randomized controlled trials involving 2,448 patients, four out of six

trials administered corticosteroids intravenously, and the remaining two trials gave them

orally showed that the use of prophylactic corticosteroids did not prevent PEP (OR 1.13: 95%

CI 0.88-1.46) (Bai et al., 2008).

ERCP may activate the proteolytic enzymes of the pancreas, which may contribute to PEP.

Protease inhibitors can prevent enzyme activation and may reduce the PEP rate. According

to a meta-analysis of 18 RCTs evaluating the efficacy of protease inhibitors in reducing PEP,

when ulinastatin was administered in a high dose (equal or > 150,000 unit), the PEP risk

decreased (OR 0.39, 95% CI 0.19-0.81; NNT 6). Additionally, gabexate was effective in high

doses (equal or >150,000 units), and the PEP risk decreased (OR 0.44, 95% CI 0.25-0.79;

NNT7) (Zhang, Z. F. et al., 2010). However, in 2 other meta-analyses the administration of

ulinastatin and gabexate was not associated with a PEP decrease when they were

Page 33: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

32

administered in doses of less than 15,000 units (Chen et al., 2010; Seta et al., 2011). The

use of protease inhibitors has not become a common practice.

There is no proof that the antibiotics are effective in PEP prophylaxis. In a systematic review

and network meta-analysis of 99 RCTs (25,313 patients), antibiotics were ranked in in fourth

position among 16 drugs for their effect in PEP prophylaxis (OR 0.46; 95% CI 0.15-1.07; NNT

21) (Akshintala et al., 2013). However, the difference between antibiotics and a placebo was

not statistically significant. Only 254 patients were included in the treatment arms of four

RCTs included in the meta-analysis (not 1,082 as stated in the meta-analysis), and two of

these RCTs did not have PEP prophylaxis as their primary study endpoint.

In patients with contraindications for NSAID and for aggressive hydration, the ESGE

guideline recommends using 5mg of sublingual glyceryl trinitrate before ERCP to prevent

PEP (Dumonceau et al., 2020). A meta-analysis of 11 RCTs (2,095 patients) showed that

the overall PEP incidence decreased in patients who had 2-5mg of glyceryl trinitrate (RR

0.67, 95%Cl 0.52-0.87) (Ding, J. et al., 2013).

4.7.4 Hydration

In patients with contraindications for NSAIDs, aggressive hydration during and after ERCP

may reduce the PEP rate. Contraindications to hydration are patients >80 years old, patients

with heart failure, advanced renal failure, and patients with advanced chronic obstructive

pulmonary disease. The amount of fluid used in hydration was 35-45ml/kg administered over

a period of 8–10h (Dumonceau et al., 2020). In a meta- analysis of three RCTs with a total

of 722 patients, the aggressively hydrated group had a lower incidence of PEP overall (OR

= 0.29, 95% CI 0.16-0.53), and a lower incidence of moderate and severe PEP (OR = 0.16,

95% Cl 0.03-0.96) (Wu et al., 2017). Another meta-analysis of 7 RCTs with a total of 1,047

patients showed that aggressive hydration using lactate Ringer’s solution (LRS) reduced the

PEP rates compared to a standard hydration group (OR = 0.47, 95% Cl 0.30-0.72, P=0.0006)

(Zhang, Z, et al., 2017). A recent RCT with a total of 395 patients reported a significant

Page 34: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

33

decrease in PEP rates in an LRS group compared to a standard hydration group (3.0% vs.

11.6%, P=0.03) (Park, C. H. et al., 2018).

4.8 Bleeding

Bleeding is the second most common, serious complication of ERCP, with an estimated rate

of 0.3-2% depending on the definition adopted in the study. Bleeding after ERCP usually

occurs after biliary or/and pancreatic sphincterotomy (Freeman et al., 2001; Cotton et al.,

2009). The classification and grading of bleeding are set in the consensus document by

Cotton et al. based on the number of transfused blood units, and the need for angiographic

or surgical intervention. Mild bleeding is an event when clinical evidence of bleeding occurs

(melena, hematemesis) and the haemoglobin drops by less than 3g/dl, and there is no need

for a transfusion. Moderate bleeding is a situation where ≤ 4 units are needed for transfusion,

but there no need for angiographic or surgical intervention. In the severe form of bleeding ≥

4 transfusion units are needed, or angiographic or/and surgical intervention is involved

(Cotton et al., 1991) (Table 1). In a prospective study by Freeman et al., 2,347 patients

underwent ERCP, mainly for the removal of stones (85%). The incidence of bleeding was

2% (mild 0.6%, moderate 0.9%, severe 0.5%). Death from bleeding occurred in 2 cases with

Child-Pugh class C cirrhosis. The independent risk factors for bleeding included the presence

of coagulopathy (OR 3.32, P<0.001), active cholangitis (OR 2.59, P<0.001), anticoagulant

therapy during the next 3 days after ERCP (OR 5.11, P<0.001), and an endoscopist with one

or fewer ERCPs per week (OR 2.17, P=0.002). The use of Aspirin or NSAIDs were not risk

factors in a multivariable analysis (Freeman, 1997). Avoiding the risk factors mentioned will

probably reduce the risk of bleeding. In a multicentre, prospective study with 2,013 patients,

and a total of 2,444 procedures, the bleeding rate was 1.22%, and no death cases from

bleeding occurred. The only significant risk factors for bleeding were precut sphincterotomy

and a stenosis of the orifice of the papilla (Masci et al., 2001). A meta-analysis of four RCTs

shows that the bleeding risk is significantly higher in the pure cut group (28.9%, 95% CI 16.3-

41.4), compared to mixed current (9.4%, 95% CI 2.1%-16.8%) (Verma et al., 2007).

Page 35: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

34

4.8.1 An endoscopic papillary balloon dilatation (EPBD) as an alternative to

sphincterotomy

The bleeding rate can be minimized by avoiding EST. EPBD is an alternative to EST when

there is no contraindication for EPBD. An EPBD preserves the function of the sphincter of

Oddi and prevents duodenobiliary reflux and the bacterial colonization of the bile duct, which

can cause stone recurrence and cholangitis (Isayama et al., 2003; Tsai et al., 2018). In a

meta-analysis by Liu et al. including 10 RCTs with a total of 1,451 ERCPs, the success rates

of stone removal in sphincterotomy and EPBD groups were similar (94.6% vs. 95.9%; OR

0.99, 95% CI 0.98-1.01). However, the EPBD group more frequently required mechanical

lithotripsy (35.0% vs. 26.2%; OR 1.31, 95% CI 1.13-1.51). Bleeding was significantly lower

in the EPBD group (0.1% vs. 4.2%, P <0.00001), but the PEP rate was higher (9.4% vs.

3.3%, P<0.00001) (Liu et al., 2012).

Another Meta-analysis with 14 RCTs (1,975 patients), showed that the bleeding rate was

significantly lower in an EPBD group comparing to a sphincterotomy group (0.2% vs. 3.4%,

P<0.01), and the PEP rate was higher (9.1% vs. 3.4%, P<0.0001). There were fewer

complete stone removals in the EPBD group (92.4% vs. 95.1%, OR 0.64, 95% CI 0.24-0.96),

and more frequent use of stone extraction baskets (OR 1.91, 95% CI 1.41-2.59), especially

when the stone diameter was larger than 8 mm. The stone recurrence rate in the EPBD group

was less than in the sphincterotomy group (6.6% vs.12.7%, OR 0.48, 95% CI 0.26-0.90)

(Zhao et al., 2013). A third meta-analysis separately compared four RCTs (923 patients) that

used balloon dilation for less than 1 minute (short EPBD) vs. sphincterotomy, and seven

RCTs that used balloon dilation for more than 1 minute (long EPBD) vs. sphincterotomy. The

overall adverse events rate was higher for the short EPBD group compared to long EPBD

group (OR 1.71, 95% CI 0.67-4.35) (OR 0.61, 95% CI 0.36-1.04), respectively. Compared to

EST group the PEP rate was higher in the short EPBD group (OR 3.87, 95% CI 1.08-13.84),

while the PEP rate was not higher in the long EPBD group (OR 1.14, 95% CI 0.56-2.35) (Liao

et al., 2012). In the above-mentioned meta-analysis, a study that compared 1-minute EPBD

Page 36: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

35

vs. 5 minutes EPBD showed that the PEP rates were lower in the 5 minutes EPBD group

(4.8% vs.15.1%, P=0.038) (Liao et al., 2010).

4.9 Cholangitis

Cholangitis is the most common infectious post-ERCP adverse event. The definition of

cholangitis is based on the consensus definition by Cotton et al. In mild cholangitis, patients

have a fever of >38ºC for 24 to 48 hours. In moderate cholangitis febrile or septic illness

requires more than three days of hospital treatment or endoscopic or percutaneous

intervention. In severe cholangitis, septic shock emerges, or surgery is required (Cotton et

al., 1991) (Table 1). The symptoms of cholangitis are upper abdominal pain, fever, and

jaundice. The most common cause is a failure of complete drainage of the obstructed and

infected biliary system. The incidence of cholangitis ranges between 0.5-3% (Andriulli et al.,

2007; Cotton et al., 2009). In a malignant hilar obstruction of the bile duct, and in cases of

PSC, the risk of cholangitis rises because of the difficulty to achieve complete drainage

(Rerknimitr et al., 2002; Paik et al., 2009). In their study Freeman et al. identified three

significant risk factors in a univariate analysis: combined percutaneous-endoscopic

procedures (P< 0.001), malignant strictures stenting (P<0.001), failed biliary access or

drainage (P<0.001) (Freeman et al., 1996).

Prophylactic antibiotics must be considered before ERCP when bile duct obstruction is

suspected or incomplete biliary drainage is expected, and when extracorporeal shock wave

lithotripsy is used to clear a CBD stone (Manes et al., 2019). According to ESGE guidelines,

prophylactic antibiotics must be given before ERCP to all patients with biliary occlusion and

anticipated incomplete biliary drainage, also to patients with pancreatic pseudocysts, and

patients with a history of previous cholangitis (Dumonceau et al., 2020). In a meta-analysis

that included nine RCTs with a total of 1,573 patients, the fixed-effect meta-analyses

significantly favoured the use of prophylactic antibiotics in preventing cholangitis (RR 0.54,

95% CI 0.33-0.91), septicaemia (RR 0.35, 95% CI 0.11-1.11), and bacteraemia (RR 0.50,

Page 37: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

36

95% CI 0.33-0.78) (Brand et al., 2010). In cases of malignancy, the unilateral drainage of the

biliary tree has higher rates of cholangitis compared to bilateral drainage (De Palma et al.,

2001), so it is recommended to use non-invasive imaging (MRCP) to plan selective guidewire

access before ERCP (Freeman et al., 2003). In cases of large stones in the bile duct, if stone

fragment clearance is not achieved, it is recommended to place a stent in the biliary tract to

reduce the cholangitis rate. Cholangitis can also be a delayed complication after a plastic or

metallic stent placement (Sawas et al., 2015). The stent obstruction can occur from sludge,

the growth of tumour tissue through the stent, migration of the stent, or from stone fragments.

The selection of the stent (metal, plastic) must depend on the aetiology and location of the

stricture, the patient survival, and the response to prior therapy (ASGE Technology

Assessment Committee et al., 2013). In cases of a malignant extrahepatic obstruction, the

ESGE recommends using a self-expandable metal stent to prevent the stent obstruction and

the possible cholangitis that can occur due to obstruction, which can delay the adjuvant

treatment later on (Dumonceau et al., 2018). Other infectious events such as cholecystitis,

duodenoscope-related transmission of infection, infective endocarditis, are less frequently

observed (Rahman et al., 2019).

4.10 Perforation

The incidence of perforation during ERCP is approximately 0.08 to 0.6% (Masci et al., 2001;

Cotton et al., 2009). A summary of 21 prospective studies with a total of 16,855 patients who

underwent ERCP, showed that the incidence rate of perforation was 0.6% (101 patients),

with 10 deaths (0.06%) (Andriulli et al., 2007).

Perforation is defined as the presence of air or contrast in the retroperitoneal space or in the

abdominal cavity that appears after the procedure. In the mild (defined) form of perforation,

a slight leak of contrast is observed and should be treated for 3 days conservatively. The

moderate (defined) form should be treated for 4-10 days conservatively, and the severe form

occurs when treated for more than 10 days and an intervention is needed (percutaneous or

Page 38: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

37

surgical) (Cotton et al., 1991) (Table 1). Another classification that is based on the location

and the mechanism of the perforation is widely used (Stapfer et al., 2000) and divides

perforation into four types: A type I perforation is duodenal wall perforation caused usually

by the duodenoscope. A type II perforation is a retroperitoneal periampullary perforation after

biliary or pancreatic sphincterotomy and a type III is a perforation of the bile or pancreatic

duct caused by instrumentation, which can be noticed when the guidewire or contrast leak

out from the duct. A type IV perforation is when retroperitoneal air alone is the only

manifestation of complication.

In a retrospective study of 9,880 ERCPs, the perforation rate was 0.4% (44 patients), and

type II was the most common perforation 68% (30 patients) (Polydorou et al., 2011). Vezakes

et al. reviewed 18 mainly retrospective studies, performed in the 2000-2014 period, which

addressed only post-ERCP perforation. A total of 142,847 patients were included. The overall

incidence of perforation was 0.39% (95% CI 0.34-0.69), from which type I perforation

occurred in 25% of the cases, type II in 46% and type III in 22% of cases. The overall mortality

was 7.8% (95% CI 3.80-13.07) (Vezakis et al., 2015). The risk factors of perforation were

identified in a large retrospective study that included 9,314 procedures. Perforation occurred

in 33 cases. The risk factors for perforation according to a univariate analysis were EST (OR

9.0, 95% CI 3.2-28.1), SOD (OR 3.8, 95% CI 1.4-11.0), and dilated CBD (OR 4.07, 95% CI

1.63-10.18, P=0.003). In a multivariate analysis, other predictive factors were the duration of

the procedure (OR 1.021, 95% CI 1.006-1.036) and biliary stricture dilation (OR 7.2, 95% CI

1.84-28.11) (Enns et al., 2002).

The location of the perforation was in the periampullary area in 65% of the cases, in the bile

ducts in 25% of cases, and in the PD in 1.4% of cases (Paspatis et al., 2014).

Symptoms of perforation are severe epigastric and back pain, epigastric tenderness, fever

and tachycardia. Additionally, subcutaneous emphysema can occur. When retroperitoneal

air is present (Type IV) in the CT scan, but there are no symptoms, the patient needs no

Page 39: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

38

further intervention (Vezakis et al., 2015). This kind of finding in the absence of symptoms

can be found in 13% - 29% of patients who have undergone EST (Genzlinger et al., 1999).

The diagnosis of perforation can be made during the ERCP. In a review of 437 cases from

15 studies, the diagnosis was made during the ERCP in 73% of cases (Vezakis et al., 2015).

In cases with a type I perforation, the diagnosis is usually obvious with symptoms of

peritonitis. In cases with severe pain after ERCP, a differential diagnosis between PEP and

perforation should be made. When perforation is suspected, a CT scan with oral contrast

should be considered (de Vries et al., 1997). If the perforation is discovered during the

procedure or within 12 h after the procedure, and the patient’s condition is stable, endoscopic

treatment should be considered using a stent. In type II cases, biliary drainage is essential

to prevent leakage of bile into the perforation site. In one study, 5 out of 13 cases of type II

perforation were managed successfully using a plastic biliary stent or percutaneous

transhepatic biliary drainage (Enns et al., 2002). In another study, 12 out of 30 cases had

nasobiliary drainage (Alfieri et al., 2013). Other studies reported the advantage of using the

fully covered self-expandable metallic stents which cover the laceration and permit free flow

of the bile into the duodenum (Park, W. Y. et al., 2012; Lee, S. M. et al., 2014; Odemis et al.,

2016). Vezakis et al. reviewed 11 studies with a total of 142,847 patients. After initial non-

surgical treatment for type II and type III patients, surgery was required later in 29/137 (21%)

cases for type II patients, with an overall mortality of 9.4%. The mortality rate for those

patients who underwent surgery was 38%. In surgery it is important to close the perforation,

and if necessary also carry out bile and gastric fluid diversion. For type III patients, non-

surgical treatment was successful for all of them (Vezakis et al., 2015). In another study, non-

surgical treatment was successful in 93% of type II and type III patients with a mortality rate

of 0.6% (Machado, 2012).

Page 40: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

39

4.11 Stent related complications

The most common complications of stenting are occlusion and migration of the stent. The

migration rate is 5-10% and occurs predominantly distally (Bagul, P. et al., 2010). Distally

migrated stents rarely cause a problem, whereas proximally (into the duct) migrated stents

can be challenging to remove, and in a few cases surgical intervention may be needed. The

occlusion rate of migrated stents can be as high as 30-40% (Ferreira et al., 2010; Decker et

al., 2011). Although no equipment has been specially designed for stent retrieval

endoscopically, devices such as polypectomy snares, stone extraction baskets, dilating

balloons and foreign body retrieval forceps have been used to extract stents. Peroral

cholangioscopy and pancreatoscopy devices (biopsy forceps, snares and baskets) are

valuable tools when plastic stents are migrated intraductally. Uncovered self-expandable

metal stents (SEMSs) migrate less frequently (< 1%) because of the large diameter and the

tissue growth through the stent (Yang et al., 2009). In cases of malignant BD occlusion, the

use of SEMS results in longer stent patency, compared to plastic stents (Almadi et al., 2017;

Moole et al., 2017). Antimigration stent designs, such flared ends and anchoring flaps, have

been created to limit the migration of the covered SEMS. In pilot studies, antimigration stents

have yielded promising results in patients with benign strictures (Park, D. H. et al., 2011;

Hamada et al., 2015; Minaga et al., 2016).

4.12 Performance measures (PMs) for ERCP

PMs are the measures that should be followed to minimise the adverse effects of the

procedure and make it safer to the patient. These measures according to the ESGE are the

following:

1. A prophylactic antibiotic should be given only to selective patients, e.g.

immunosuppressive patients, or patients with hilar tumour or PSC. Routine antibiotic use

is not recommended (Dumonceau et al., 2020). A systematic review of nine RCTs

concluded that prophylactic antibiotics decreased the post ERCP cholangitis rate in cases

Page 41: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

40

where an ERCP had failed to relieve the obstruction of the BD, but if the biliary obstruction

had been relieved there was no significant decrease in the cholangitis rate (Brand et al.,

2010), meaning that relieving the obstruction is the reason why patients did not have

cholangitis and did not require the use of antibiotics.

2. The successful cannulation of the BD should be achieved using all available tools and

techniques. Failure in BD cannulation increases the rate of adverse events (Bailey et al.,

2008).

3. In cases of biliary obstruction below the hilum, a successful stent placement reduces the

rate of adverse events (van Berkel et al., 2004).

4. For BD stones <10mm in size, the clearance rate should be at least 90%. Using the

available tools such as balloon/basket extractors and balloon dilatators for the papilla

increases the stone clearance success rate (Kuo et al., 2012; Oppong, Romagnuolo, &

Cotton, 2012). The cholangitis rate increases in cases of incomplete stone clearance.

5. PEP is the most common and serious complication; therefore, the PEP rate is a good

quality indicator of ERCP. According to the ESGE, the PEP rate should be below 10%

(Domagk et al., 2018). A systematic review of RCTs documented an overall PEP rate to

be 9.7% in low risk patients, and 14.7% for high risk patients (Kochar et al., 2015). Using

a rectally administered NSAID before the ERCP and placing a prophylactic stent in the

PD in high risk patients, reduces the risk of PEP.

4.13 Routine preoperative laboratory testing (RPLT)

Many studies have shown that RPLT is outdated, because of the small impact on the

perioperative outcomes. In one trial, 1,061 patients who underwent various ambulatory

surgeries were randomized into non RPLT and indicated RPLT groups. The RPLT test

battery included a complete blood count, electrolytes, blood glucose, creatinine, an

electrocardiogram, and chest radiograph according to the Ontario Preoperative Testing Grid.

There was no difference in the perioperative adverse effects, or in adverse effects over the

next 30 days after the procedure (Chung e al., 2009). From a national surgical quality

Page 42: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

41

improvement programme, patients who underwent elective hernia repair, were collected

(N=73,596). For 63.8% of the patients, preoperative testing was performed. Of these

patients, at least one abnormality was recorded in 61.6% of the patients. In patients with no

solid indication for testing, 54% of them had at least one test. Neither the results of testing,

nor abnormal results were correlated with postoperative outcomes (Benarroch-Gampel et al.,

2012). However, for a selected group of patients with underlying diseases and for patients

with risk factors that can affect the procedure, selective testing is appropriate (Czoski-Murray

et al., 2012). For patients with chronic diseases (e.g., complicated diabetes mellitus, heart

failure, chronic kidney disease, liver disease), measuring of the electrolytes and creatinine

levels should be considered. A complete blood count is recommended for patients with

probable anemia (e.g., chronic kidney and liver diseases, chronic inflammatory conditions),

or for patients with clinical manifestations of anemia, or when there is a high risk of bleeding

during the procedure (Committee on Standards and Practice Parameters et al., 2012; O'Neill

et al., 2016). Coagulation testing should be reserved for patients with haematopoiesis

conditions, and patients on anticoagulants, or patients with history of underlying coagulation

disorders (Chee et al., 2008: ASGE Standards of Practice Committee et al., 2014).

Numerous studies and guidelines outline lack of evidence for benefit in routine preoperative

testing (e.g., chest X-ray, echocardiogram) in low risk surgical patients. Economic analyses

suggest significant potential cost savings from implementation of guidelines (Chung et al.,

2009; Benarroch-Gampel et al., 2012).

Page 43: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

42

5. Aims of the study

The influencing factors of complications of ERCP were evaluated. The individual aims of the

study were the followings:

1. To evaluate the risk factors of complications of ERCP in patients with primary

sclerosing cholangitis (PSC).

2. To evaluate the outcomes, effectiveness and safety of the endoscopic resection of

papillary benign and malignant tumours.

3. To evaluate the adverse effects of biliary cannulation in patients with a native papilla.

4. To evaluate the association of Routine Preoperative Laboratory Testing (RLPTs) to

the risk factors of complications in ERCP.

Page 44: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

43

6. Materials and Methods

All studies were designed in the Helsinki University Hospital, Abdominal Centre, in the

Department of Endoscopy at Meilahti Hospital. Studies I and II are retrospective studies.

Studies III and IV are prospective studies. The approval of the studies came from the local

Ethics Committee.

Prior to the procedure, 100 mg of diclofenac was given rectally to patients without

contraindications for diclofenac, which has been the general practice since September 2013.

The patients were in the prone position. All the patients were consciously sedated by an

anaesthesiologist. The ERCP related adverse events definition and grading were based on

the consensus criteria developed by Cotton et al. (Cotton et al., 1991).

6.1 Study I

In between 2007-2009, in the Helsinki University Central Hospital (known as the Helsinki

University Hospital, Abdominal Centre since 2015) 3,019 ERCPs were performed, 443 of

these (14.7%) with indications of PSC. In two patients (0.5%) the cannulation failed. The

primary tools used for the ERCPs conducted were a papillotomy knife (Jagtome RX; Boston

Scientific, Miami, Florida, USA) and a 0.035-in, 450 cm guide wire (Jagwire; Boston

Scientific) to gain access to the biliary duct. In cases of failed cannulations using the primary

tools, and in which the guide wire slipped unintentionally into the PD, a pancreatic

sphincterotomy was performed using the papillotomy knife over the guide wire. If cannulation

of the BD was still not achieved, then an oblique cut was made using a needle knife towards

10 o’clock, starting from the upper end of the pancreatic sphincterotomy cut. Once the

cannulation of the BD was achieved, a biliary sphincterotomy was performed in the same

direction. A successful cannulation was confirmed with fluoroscopy. Then a balloon catheter

was inserted into the CBD and the intrahepatic bile tree was filled with contrast. There was

no contrast filling of the PD. Balloon occlusion enhances the visualization of the bile ducts.

Page 45: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

44

The strictures of the bile ducts were dilated when indicated, and brush cytology was collected

from the extra- and intrahepatic bile ducts, using 1-4 brushes. All ERCPs were performed by

a specialist. PSC imaging findings were scored according to the study by Ponsioen et al.

(Ponsioen et al., 2002).

The plasma amylase was measured in some cases before the procedure, and for all cases

4-6 hours after the procedure, and after 24 hours if the patient stayed overnight in the

hospital. The ERCP complications, PEP, cholangitis, bleeding, and perforation were

evaluated. The characteristics of the PSC patients are shown in Table 2.

Table 2. Characteristics of 441 patients undergoing ERCP for PSC Risk factors No. of patients Patient related factors Sex F/M 203/238 Age, years >50 115 >30-50 197 ≤30 129 Previous PEP Yes/No 23/418 Previous biliary sphincterotomy Yes/No 147/294 Radiological PSC score 3 89 2 77 1 44 0 227 (ERCP) endoscopic retrograde cholangiopancreatography, (PEP) post ERCP pancreatitis. (PSC) Primary sclerosing cholangitis.

6.2 Study II

Between 2000 and 2012, 61 papillectomies on tumours of the Vater of papilla were

performed. In 35 patients, the tumours were benign without FAP. Sixteen patients with FAP

had benign tumor and 10 patients had malignant tumor of the ampulla. The procedures were

performed by two experts with accumulated expertise of over 200 annual ERCPs. The

characteristics of the patients and tumours are presented in Table 3.

Page 46: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

45

First, using a sphincterotome and guide wire the PD was cannulated and a small amount of

methylene blue was injected to mark the orifice and the end of the PD. Using a polypectomy

snare, the tumour was grasped and resected using a blended electrosurgical current (50-60

J). The goal was to perform an en-block resection of the tumour, but when necessary, a

piecemeal resection was performed. After the resection, a 5-7 Fr, 3-7 cm long plastic

pancreatic stent was inserted. The control duodenoscopy was planned after 1, 3 and 6

months. In the first control, the pancreatic stent was removed if still in place.

Table 3. Characteristics of patients and tumours

Benign tumour, FAP Cancer No FAP. n = 35 n = 16 n = 10 Male/female 24/11 9/7 5/5 ASA I 7 0 0 II 10 10 1 III 17 6 7 IV 1 0 2 ERCP Indications Incidental 18 16 0 Jaundice 7 0 9 Anaemia/bleeding 6 0 1 Pancreatitis 4 0 0 Abdominal pain 0 0 0 Tumour size in cm <1 1 0 0 1-2 23 15 5 2-4 7 1 5 >4 2 0 0 Unknown 2 0 0 Histologic diagnosis before resection Low grade dysplasia 24 13 2 High grade dysplasia 8 3 4 Adenocarcinoma 0 0 1

Histologic diagnosis after resection Low grade dysplasia 18 13 0 High grade dysplasia 14 13 0 Adenocarcinoma 0 0 10 Other 3 0 0 ASA: American Society of Anaesthesiology physical status classification. FAP: Familial adenomatous polyposis.

Page 47: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

46

6.3 Study III

From March 2011 to September 2014, 1,626 patients with native papilla underwent ERCP.

291 patients were excluded because the primary target was to cannulate the PD. Another

620 patients without cannulation time records were also excluded. A total of 821 patients

were included in the study. The characteristics of the patients are presented in Table 4.

Table 4. Patient characteristics No of patients 821 Age (years) (median, range) 69 (8-102) Male/Female 437/384 BMI; kg/m2 (median, range) 25.7 (12.1-51.9) ASA I 51 (6.2%) II 159 (19.4%) III 424 (51.6%) IV 187 (22.8%) Procedure indication Stone 445 (54%) Stricture 310 (38%) Post cholecystectomy 33 (4%) Post liver transplant 19 (2.3%) Other 14 (1.7%) NSAID prophylaxis 256 (31%) Duodenal diverticulum 93 (11%) Biliary pancreatitis prior to ERCP 78 (9.5%) (BMI) Body mass index; (ASA) American Society of Anaesthesiology physical status classification. (NSAID) non-steroidal anti-inflammatory drug; (ERCP) endoscopic retrograde cholangiopancreatography

The primary cannulation tools were a sphincterotome (Ultratome Boston Scientific), and a

straight wire (Hydrosteer T Jude Medical). The cannulation time was defined as the time

between the first contact and the deep cannulation of the CBD, and this time was recorded.

Cannulation attempts were recorded, and every loss of contact and repositioning were

counted as a new attempt. Passages of the guide wire and injected contrast into the PD were

also counted. In case of difficult cannulations, TPBS was performed if the guide wire was in

the PD. In the TPBS, a pancreatic sphincterotomy incision was made toward the CBD

through the septum between the CBD and PD. If the CBD was still not accessible a needle

Page 48: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

47

knife was used to continue the incision from the upper end of the pancreatic sphincterotomy

cut towards 10 o’clock. A difficult cannulation was defined according to the ESGE guidelines

(Testoni et al., 2016). Major adverse effects such as PEP, bleeding, cholangitis, and

perforation were evaluated.

6.4 Study IV

From March 2012 to February 2013, 1,228 patients underwent ERCP. 17 ERCPs performed

on patients under 18 years old were excluded from the study. Another 15 ERCPs performed

in the operation theatre were also excluded. The remaining 1,196 ERCPs were included. The

characteristics of the patients are shown in Table 5.

253 ERCPs were performed by two expert gastroenterologists when PSC was suspected or

was the indication for ERCP. The other 943 ERCPs were performed by 4 expert

gastrointestinal surgeons. Blood was collected for RPLT before the procedure. The RPLT

included basic blood count (BBC), electrolytes (P-NA, P-K), thromboplastin time (P-TT),

international normalized ratio (P-INR) and the plasma total amylase or pancreatic amylase.

4-6 hours after the procedure, the plasma amylase was collected again. If the patient stayed

in the hospital overnight, the amylase was collected again after 24 hours. Indications for the

procedure are shown in Table 5.

Page 49: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

48

Table 5. Patients characteristics and indications for ERCP Patients and ERCP Number of ERCP (%) Indications Male/Female 529 (55%)/427 (45%) Median age; years 59 Median BMI; Kg/m2 24.6 Emergency procedures 299 (25%) ASA I 35 (2.9%) II 417 (34.8%) III 582 (48.7%) IV 160 (13.4%) Comorbidities Diabetes 256 (21.4%) Cardio- and cerebrovascular disease 663 (55.5%) COPD 43 (3.5%) Other lung diseases 70 (5.9%) Kidney failure 29 (2.4%) Liver diseases 331 (27.7%) Endocrinal diseases 41 (3.4%) Malignancy 89 (7.4%) Other 391 (32.7%) ERCP Indications Choledocholithiasis 254 (21%) Biliary Stricture 401 (34%) Primary Sclerosing Cholangitis 251 (21%) Chronic pancreatitis 123(10%) Pancreatic pseudocysts 77 (6%) Miscellaneous 41 (3%) ASA: American Society of Anaesthesiologists physical status classification. COPD: Chronic Obstructive Pulmonary Disease.

6.5 Statistical analysis

I. In the first study, the data was analysed using SPSS v 15.0 (IBM Corporation, Somers,

New Work, USA) and by Statexact v. 4.0 (Cytel Software Corporation, Cambridge,

Massachusetts, USA). To test for differences between categorical variables, Fisher’s exact

test was used. A logistic regression was used to calculate odds ratios and P values

(WALD𝑥2) for nominal and ordinal variables. To test for an association between the incidence

of PEP and an ordered categorical variable, the Cochran-Armitage trend test was used. A

nonparametric Wilcoxon-Mann-Whitney test was used to compare differences between

continuous and ordinal variables. The P value was considered statistically significant when it

Page 50: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

49

was below 0.05. Professional statistician applied statistics in studies I and IV, and the

researcher applied statistics in studies II and III.

II. The statistical analysis in the second study was performed using IBM SPSS statistics

(version 19.0, Chicago, Illinois, USA) and the results were presented as the median and

range. A Mann-Whitney U test and Pearson’s 𝑥2 test were used to compare the results

between groups. P values below 0.05 were considered significant.

III. SPSS v 15.0 (IBM Corporation, Somers, NY) was used for the data analysis. A chi-square

test was used to test for the difference between categorical variables. A nonparametric

Wilcoxon-Mann-Whitney test was used to compare differences between continuous and

ordinal variables. P values < 0.05 were regarded as statistically significant.

IV. The statistical analysis in Study IV was generated using IBM SPSS Statistics 21

(International Business Machines Corporation, Endicott, NY, USA). The results were

reported as medians and interquartile ranges, or the number of patients and percentages. P

values <0.05 were regarded as statistically significant.

Page 51: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

50

7. Results

7.1 Study I

After an ERCP the median length of stay in hospital was 1 day (range, 0-28).

7.1.1 Cannulation success rate

The success rate of the primary CBD cannulation was in 389/441 patients (88.2%). 147

patients (37.8%) had a prior biliary sphincterotomy, which made the cannulation easier. For

patients with native papilla, the primary cannulation rate was 82.3%, whereas for those with

a previous biliary sphincterotomy it was 100% (P<0.001). For 52 patients, the primary

cannulation failed, and in order to achieve the cannulation, TPBS was performed. In 11 cases

of these, a further cut using a needle knife was needed.

7.1.2 PEP incidence, predicting and protective factors

31 patients developed PEP (7%). In 18 cases the disease was mild, in 10 moderate and in 3

severe. One patient with severe PEP was treated in the intensive care unit for 3 days. No

cases of necrotizing pancreatitis were recorded. All patients were treated conservatively.

Patients with PEP, stayed in the hospital for a median of 4 days (range, 2-28). The evaluated

risk factors for PEP are presented in Table 6. From the patient-related factors, the female

sex was a risk factor (OR 2.6, P=0.015). The PEP rate in patients with native papilla was

9.2%. In patients, with a previously performed biliary sphincterotomy, the PEP rate

decreased to 2.7% (P=0.019), being therefore a protecting factor against PEP (OR 0.28,

P=0.02). Technique-related factors results showed that the accidental passage of the guide

wire into the PD was a risk factor (OR 8.2, P<0.01). With no accidental passage of the guide

wire into the PD, the rate of PEP was 2.6%. When the guide wire strayed into the PD twice,

the PEP rate was 20%, and if this occurred five times, the PEP incidence increased to 31.6%.

Contrast medium was injected into the PD in 15 patients. Only two of them developed PEP,

Page 52: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

51

which is not statistically significant increase when compared to patients with no contrast in

the PD. The PEP rate increased when more complicated techniques had to be used to

achieve the CBD cannulation. When the cannulation was achieved without biliary

sphincterotomy, the PEP rate was 1.4%. When biliary sphincterotomy was performed alone,

the PEP rate was 6.8%. In cases of TPBS performed in order to gain access into the CBD,

the PEP rate increased to 25%, and with a combination of biliary sphincterotomy, the PEP

rate rose to 27%. In 9 cases with difficult cannulation where TPBS was performed in

combination with precut and biliary sphincterotomy, the PEP rate was 55.6%. The median

extrahepatic PSC score was lower in patients for whom TPBS was necessary (0, 0-6)

compared to the other patients without needing TPBS (1, 0-6, P<0.001).

7.1.3 Other complications

No bleeding cases after sphincterotomy were recorded. Six cases of cholangitis were

recorded (1.4%), one mild and five with a moderate form of disease. The patients that

developed cholangitis had the same procedure duration as others (median 40 min, range, 8-

50min) (27 min; range, 10-96min; P=0.096). All the cholangitis patients had a previous biliary

sphincterotomy. Cholangitis developed for 2% of patients with a previous biliary

sphincterotomy, versus 1% of patients without a biliary sphincterotomy (P=0.405). Three

perforations (0.6%) were recorded, one caused by the guide wire and the others occurred

after the sphincterotomy. All cases of perforation were treated conservatively in 5, 6 and 7

days, respectively. No deaths were recorded due to the procedure. The risk factors and

incidence of PEP are shown in Table 6.

Page 53: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

52

Table 6. Risk factors and incidence of PEP in 441 patients undergoing ERCP for PSC

Risk factors No. PEP cases/ Odds ratio/ P value No. of patients (%) 95% CI Patient related factors Sex Female 21/203(10.3) 2.63(1.21-5.73) 0.015 Male 10/238(4.2) Age in years >50 7/115(6.1) 0.94(0.57-1.53) 0.799 30-50 15/197(7.6) ≤30 9/129(7.0) Previous PEP Yes 3/23(13.0) 2.09(0.59-7.46) 0.256 No 28/418(6.7) Previous biliary sphincterotomy Yes 4/147(2.7) 0.28(0.10-0.81) 0.010 No 27/294(9.2) PSC total radiological score 3 4/89(4.5%) 0.70(0.49-1.00) 0.053 2 3/77(3.9) 1 1/44(2.3) 0 22/227(9.7) Technique related factors Wire unintentionally in pancreatic duct Yes 23/129(17.8) 8.25(3.58-18.99) <0.001 No 8/312(2.6) Bile duct balloon dilation Yes 4/80(5.0) 0.65(0.22-1.92) 0.436 No 27/361(7.5) Duration of ERCP >30 min 13/138(9.4) 1.54(0.90-2.64) 0.120 20-30 min 14/185(7.6) ≤20 min 3/85(3.5) Precut + biliary and pancreatic 5/9(55.6) 87.5(15.4-498.5) <0.001 EST Biliary and pancreatic EST 10/37(27.0) 25.9(6.7-100.1) <0.001 Pancreatic EST alone 1/4(25.0) 23.3(1.9-294.1) 0.015 Biliary EST alone 12/176(6.8) 5.1(1.4-18.4) 0.012 Cannulation alone 3/213(1.4) (ERCP) endoscopic retrograde cholangiopancreatography, (PEP) post ERCP pancreatitis, (PSC) primary sclerosing cholangitis, (95% CI) 95% confidence Interval, (EST) endoscopic sphincterotomy.

7.2 Study II

7.2.1 Non FAP benign papillary tumours

Of the non FAP benign papillary tumours 18 out of 35 (51%) were found accidentally during

gastroscopy (14 patients), computed tomography (2 patients), or magnetic resonance

imaging (2 patients). The investigations started with seven patients (20%) because of

jaundice, in six patients because of bleeding episodes or anaemia, and for four patients

because of acute pancreatitis with an unknown aetiology. The concordance between a pre-

resection biopsy and resected specimen was 24 of 32 (57%), see Table 7.

Page 54: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

53

Table 7. The ERCP procedures and the histologic diagnosis before and after the resection

Benign tumour, No FAP, n = 35

FAP n = 16

Cancer n =10

Histological diagnosis before resection Low grade dysplasia 24 13 2

High grade dysplasia 8 3 4 Adenocarcinoma 0 0 1 Histological diagnosis after resection Low grade dysplasia 18 13 0 High grade dysplasia 14 3 0 Adenocarcinoma 0 0 10 Other * 3 0 0 Procedures done during ERCP Biliary sphincterotomy 23 13 8 Pancreatic sphincterotomy 19 8 4 Pancreatic stenting 16 12 4 Coagulation of margins 23 15 9 *One amyloidosis, one paraganglioma, one inflammatory changes

An endoscopic ultrasound was performed in 12 cases before the papillectomy. Pancreatic

stenting was attempted for 17 patients and succeeded in 16 cases (94%). For 10 patients, a

pancreatic sphincterotomy was considered sufficient and prophylactic stenting was not

attempted. A biliary sphincterotomy was performed on 23 patients (66%). To prevent

bleeding after the papillectomy, resection margins were coagulated in 23 patients. PEP

developed in four patients (11%), three patients with a mild form of disease, and one with a

moderate form. Six patients suffered from bleeding and were treated endoscopically. Two

patients with a retroperitoneal perforation were treated conservatively. One patient

developed all three complications and was also treated conservatively. The median follow-

up was 14 months (range 1-91). During the follow-up, residual adenomas were found in ten

patients (29%), four of them underwent a pancreaticoduodenectomy. In patients who

underwent surgery, the pathologic report revealed an adenoma with high-grade dysplasia in

two patients, carcinoma in one patient, and no neoplasia in one patient, even if the pre-

operation biopsies reported a case of in-situ carcinoma.

Page 55: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

54

7.2.2 FAP related papillary tumours

The papillary adenomas in FAP patients were discovered during the routine follow-up

endoscopy. The concordance between the pre-resection biopsy and the post-resection

specimen was 14 out of 16 (88%). No EUS was planned before the papillectomy. The use of

contrast was necessary in four patients during the procedure. In 12 out of 16 patients (75%)

methylene blue was inserted into the PD. Pancreatic stenting was successful in 12 out of 14

patients (86%), while in one case the stent was placed in the minor papilla. In one patient, a

pancreatic sphincterotomy was considered to be sufficient. A biliary sphincterotomy was

done for 13 out of 16 patients (81%). Two patients developed a moderate form of PEP and

were treated conservatively. Three patients had bleeding and were treated endoscopically.

The median follow-up time was 37 months (range 1-63). Three residual adenomas were

found during the follow-up. Two of these were minor lesions and were treated with

coagulation and biopsy. One patient underwent a pancreaticoduodenectomy, which revealed

a high-grade adenoma in the specimen. In one patient with residual multiple duodenal

adenomas, a pancreaticoduodenectomy was performed 4 years later, which revealed one

high grade and three low grade adenomas.

7.2.3 Ampullary cancer

The primary symptom of ampullary cancer was jaundice in nine patients and bleeding in one

patient. A CT scan or MRI was done for all patients and revealed a bile duct dilation for all

the patients and PD dilation for six of them. In two patients, methylene blue was inserted into

the PD. Pancreatic stenting was attempted in five patients and was successful in four (85%).

In two patients, a wide pancreatic sphincterotomy was carried out and stenting was not

necessary. A biliary sphincterotomy was performed for eight patients. In this group of patients

there was no PEP and no perforation. In two patients (20%) bleeding occurred. One patient

was treated endoscopically, and another needed a laparotomy and duodenotomy. During the

follow-up, three patients developed residual cancer (4 mm, 1 cm, 3 cm), and all of them

underwent a pancreaticoduodenectomy. A patient with a 3 cm tumour had also 6/16 lymph

Page 56: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

55

node metastasis and died 14 months after the operation. The two other patients that

underwent surgery, have been followed up for 6 and 8.5 years without any sign of

malignancy. Five out of seven non-operated patients died 9-24 months later from ampullary

cancer, and one died due to pneumonia. The only patient that is still alive, is being monitored.

7.3 Study III

7.3.1 Primary and rescue cannulation methods

For primary cannulation in the native papilla, a sphincterotome with a straight guide wire was

used in all but in one case in which a regular cannula alone was used. After the primary

cannulation method, the success rate was 73%. After the second rescue cannulation method,

the success rate was 91%, after the third 98.7%, and after the fourth it was 99.1%. In 7

patients (0.91%) cannulation failed. The median success time was 1.1 minutes (range 0.02-

19.67), with a median of 2 (range 0-15) cannulation attempts. An intrusion into PD was rare

(median 0, range 0-8), and the injection of contrast into the PD was also rare (median 0,

range 0-1). 80% of all successful primary cannulations were achieved in 3.5 minutes, and

91% in 5 minutes. When the endoscopist found that the primary cannulation was not going

to be successful, he adapted a rescue cannulation method in a median time of 4.8 minutes

(range 0.3-28). After 5 minutes, only 9% of the primary cannulations succeeded. The most

common rescue cannulation methods were TPBS alone, or combined with needle knife

precut, and sphincterotome precut. In total, within 10 minutes, 85% (691 patients) of the

successful cannulations were performed. Details of the ERCP and adverse events are shown

in Table 8. For the rescue methods used in cases of failed primary cannulation methods, see

Tables 9, 10, and 11.

Page 57: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

56

Table 8. ERCP details, adverse events and mortality ERCP details Procedure length, min (median, range) 18 (5-110) Biliary sphincterotomy 803 (97.8%) Biliary cytology 209 (25.5%) Biliary duct dilatation 77 (9.4%) Common bile duct stone extraction 358 (43.8%) Biliary stent placement 286 (34.8%) Pancreatic sphincterotomy 145 (17.7%) Prophylactic pancreatic stent 5 (0.6%) Adverse events Post-ERCP pancreatitis 33 (4%) Mild 19 (45%) Moderate 20 (48%) Severe 3 (7%) Bleeding 20 (2.4%) Perforation 4 (0.5%) Cholangitis 10 (1.2%) 30-day mortality 42 (5%) ERCP: endoscopic retrograde cholangiopancreatography

Table 9. Rescue methods used in the second attempt, when the first attempt methods failed

The second cannulation Total Success Failure Attempt methods n=222 n=144 (65%) n=77 (35%) No second attempt 1 TPBS 156 107 (69%) 49 (31%) Sphincterotome precut 44 29 (66%) 15 (34%) Needle knife precut 13 7 (54%) 6 (46%) Double wire technique 5 1 (20%) 4 (80%) Pancreatic stent 2 0 2 (100%) Regular cannula alone 1 0 1 (100%)

TPBS: Transpancreatic biliary sphincterotomy.

Table 10. Rescue methods used in the third attempt, when the second attempt methods failed

The third cannulation Total Success Failure Attempt methods n=74 n=67 (91%) n=7 (9%) No third attempt 3 TPBS + NK 49 44 (90%) 5 (10%) TPBS 8 7 (88%) 1 (12%) NK 8 8 (100%) 0 Papillectomy 4 4 (100%) 0 Double wire technique 2 1 1 Sphincterotome precut 1 1 0 Sphincterotome with straight wire 1 1 0 Sphincterotome with J-tip wire 1 1 0

TPBS: Transpancreatic biliary sphincterotomy, NK: Needle knife precut.

Page 58: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

57

Table 11. Rescue methods used in the fourth attempt, when the third attempt methods failed

The fourth cannulation Total Success Failure Attempt methods n=4 n=4 n=0 Papillectomy 3 3 0 Sphincterotome with a straight wire 1 1 0

7.3.2 PEP

A total of 33 patients (4.0%) developed PEP. When the primary cannulation succeeded, only

14 patients (2.3%) had PEP. After using the second cannulation method, 9 patients (6.3%)

developed PEP, and after further cannulation methods the PEP increased up to 13.5% (10

patients). When the successful cannulation was achieved within 5 minutes of the beginning

of the procedure, the PEP rate was 2.1% (12 patients), but if the cannulation was prolonged

for more than 5 minutes, the PEP rate increased up to 8.7% (21 patients), p<0.001.

In patients who required a maximum of 5 attempts to achieve the successful cannulation, the

PEP rate was 2.1% (12 patients), with more than 5 attempts, the PEP increased to 8.8% (21

patients), p<0.001. If the guide wire slipped into the PD once, the PEP rate was 3.3% (18

patients), for more than once, the PEP was 6.4% (15 patients), p=0.048.

Contrast was injected into the PD in 29 patients (3.5%) and the PEP rate for this group was

10% (n=3), compared to 3.8% (n=30) in patients with no contrast injection, p=0.106. The

presence of a trainee did not affect the PEP rate, with 4 cases of PEP (5.8%) vs. 29 cases

without PEP (3.9%), p= 0.35.

7.3.3 Difficult cannulation

A total of 599 patients had a successful primary cannulation, and 477 of this group (79.6%)

had an easy cannulation with the absence of any of the difficult cannulation criteria mentioned

in the ESGE guidelines (Testoni et al., 2016). 311 patients out of 821 (37.9%) had at least

one difficult cannulation criteria (cannulation >5 minutes in 152 patients, more than five

cannulation attempts in 141 patients, and unintended PD passages in 234 patients). For

Page 59: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

58

these patients, the PEP rate was 7.1% (n=22), and if no criteria presented, the PEP rate was

2.2% (n=11), p=0.001. With only one difficult cannulation criteria presented, the PEP rate

was 6.9% (n=11), and with the presence of all criteria, the PEP rate was 12.3% (n=8).

7.3.4 TPBS as a rescue method

A TPBS was the preferred rescue method in 156 cases (70.6%), whereas other rescue

methods were used in 65 cases (29.4%). When a TPBS ± needle knife precut was used as

rescue method the PEP rate was 8.9% (n = 15), and when other rescue methods were used,

the PEP rate was 2.8% (n = 18), p =0.001.

7.3.5 Other complications

49 patients had adverse events. 33 patients (4%) had PEP, 20 patients (2.4%) had bleeding,

four patients (0.5%) had perforations, and 10 patients (1.2%) had cholangitis. In cases of

difficult cannulations, the rate of complications was 9.3% (n=29), whereas in the easy

cannulation group the complication rate was 3.9% (n=20), p=0.002. After excluding PEP,

there were no more significant differences in complications between the two groups 11

(3.5%) vs. 12 (2.4%), p=0.384.

7.4 Study IV

In 33 cases out of 1196, the cannulation failed. This was because the papilla could not be

reached due to previous surgery (gastric resection, pancreaticoduodenectomy) in 5 cases, a

proximal obstruction to the papilla in six cases, edema of the descending duodenum in eight

cases, and in 13 cases, a deep cannulation of the bile duct could not be achieved. In one

case the ERCP was not performed because of the patient's allergic reaction to sedatives.

Procedures done during the ERCP are listed in Table 12.

Page 60: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

59

Table 12. Procedures during the ERCP

ERCP procedures Number of ERCPs (n=1196)

Biliary cytology 416 (35%) Biliary sphincterotomy 562 (47%) Stone extraction from the CBD 345 (29%) Biliary dilatation 164 (14%) Biliary stent 357 (30%) Pancreatic sphincterotomy 207 (17%) Pancreatic cytology 35 (3%) Pancreatic dilatation 101 (8%) Pancreatic stent 216 (18%) Pseudocyst gastrostomy or 24 (2%) duodenostomy Per oral cholangioscopy 21 (2%) Double balloon ERCP 8 (1%) ERCP: Endoscopic retrograde cholangiopancreatography. CBD: Common bile duct.

7.4.1 Post-ERCP complications

PEP developed in 43 (3.6%) patients. The ERCP-related and sedation related complications

are listed in Table 13, and a univariate and multivariate analysis of PEP risk factors are listed

in Table 14.

Table 13. ERCP related and sedation related adverse events in 1196 ERCPs performed to 956 patients

Adverse events Count (%)

ERCP-related 102 (8.5) Pancreatitis 43 (3.6) Mild 24 (2) Moderate 14 (1.2) Severe 5 (0.4) Bleeding 17 (1.4) Cholangitis 17 (1.4) Perforation 6 (0.5) Miscellaneous 19 (1.6) Sedation-related 214 (17.9) Respiratory depression 128 (10.7) Cardiovascular depression 86 (7.2)

Page 61: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

60

Table 14. Univariate and multivariate analysis of PEP risk factors

Univariate Odds p-value 95% CI Diabetes, insulin treatment 0.140 (0.19-1024) 0.053 Diabetes, oral medication 0.216 (0.030-1.587) 0.132 Hypertension 0.431 (0.198-0.937) 0.034 Cardiovascular disease 0.625 (0.243-1.607) 0.329 Opioid use 1.494 (0.615-3.632) 0.376 Anticoagulant use 0.695 (0.290-1.669) 0.416 Smoker 0.481 (0.169-1.372) 0.171 Hospitalized patient 0.770 (0.362-1.636) 0.497 Outpatient case 1.336 (0.629-2.839) 0.451 P-tromb < 150 0.154 (0.021-1.125) 0.065 P-TT <70% 0.206 (0.049-0.860) 0.206 Abnormal ECG 0.596 (0.291-1.222) 0.158 Hyponatremia 0.506 (0.211-1.212) 0.127 Hypokalemia 1.837 (0.547-6.162) 0.325 Hyperkalemia 3.837 (0.468-32.400) 0.208 ASA 1-2 vs 3,4,5 0.51 (0.28-0.95) 0.033 Age 0.97 (0.96-0.99) 0.004 Age > 60 years 0.44 (0.23-0.86) 0.016 Female vs male 0.70 (0.37-1.32) 0.264 Anaemia 0.67 (0.34-1.29) 0.231 Multivariate analysis Odds 95% CI P-value

Age > 60 years old 0.525 (0.261-1.054) 0.070 Female vs male 0.714 (0.370-1.375) 0.313 Diabetes, insulin treatment 0.138 (0.019-1.020) 0.052 P-TT<70% 0.244 (0.57-1.037) 0.056

ECG: Electrocardiogram, ASA American Society of Anaesthesiologists.

The bleeding and perforation cases were treated conservatively. Nineteen miscellaneous

complications were reported, e.g. pseudocyst infection, stent migration, peptic ulcer bleeding,

air embolism. Two deaths happened the same day that the ERCP was performed, one due

to an air embolism, and another due to a myocardial infarction. Eleven deaths happened

within one week after ERCP, and 44 deaths during the next 30 days after the procedure.

7.4.2 Complications due to sedation

Respiratory depression occurred in 128 patients (11%). During the procedure, mask

ventilation was needed 20 times (1.7% of patients), and the ERCP was paused. Three

patients were intubated because of resuscitation due to an air embolism, second due to a

laryngobronchospasm and third due to septic cholecystitis.

Page 62: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

61

There was no association between respiratory depression and the RPLT in the multivariate

analysis (Table 15).

Table 15. Univariate and multivariate analysis of respiratory depression risk factors

Univariate Odds ratio P-Value 95% CI Diabetes insulin treatment 1.477 (0.916-2.380) 0.110 Hypertension 1.139 (0.778-1.668) 0.502 Cardiovascular disease 1.721 (1.117-2.651) 0.014 COPD 0.405 (0.097-1.695) 0.216 Kidney failure 0.609 (0.143-2.590) 0.502 Endocrinal disease 1.753 (0.760-4.044) 0.188 Opioid use 0.688 (0351-1.350) 0.277 Anticoagulant use 1.210 (0.769-1.904) 0.409 Hospitalized patient 0.760 (0.488-1.183) 0.223 Outpatient case 1.518 (0.957-2.407) 0.076 P-TT <70% 1.264 (0.812-1.967) 0.300 P-INR >2 0.648 (0.219-1.914) 0.432 Abnormal ECG 1.229 (0.841-1797) 0.287 ASA 1-2 vs 3-5 1.32 (0.98-1.95) 0.169 Age >60 1.13 (0.78-1.63) 0.506 Female vs male 1.27 (0.88-1.83) 0.205 Anemia 0.44 (0.29-0.68) <0.001 Multivariate all patients Cardiovascular disease 2.07 (129-3.34) 0.003 Anemia 0.39 (0.25-0.61) 0.000 Patients without cardiovascular disease Anemia 0.26 (0.14-0.48) 0.000 Patients with cardiovascular disease Female vs male 1.34 (0.59-3.02) 0.483

COPD: Chronic obstructive pulmonary disease, ECG: Electrocardiogram, ASA American Society of Anaesthesiologists.

In this multivariate analysis, an interaction between anemia and cardiovascular disease was

found in the statistical model. Upon further investigation, anemia was found to be a stronger

protective factor if the patient had no cardiovascular disease (OR 0.26, p<0.001).

In 86 patients (7.2%) cardiovascular depression occurred. The multivariate analysis showed

that cardiovascular depression was increased in cases of hyponatremia, thrombocytopenia,

or in patients over 60 years old. In this statistical model, an association was found between

Page 63: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

62

gender and hyponatremia, in which hyponatremia was associated with cardiovascular

depression slightly more in men than in women (Table 16).

Table 16. Univariate and multivariate analysis of cardiovascular depression risk factors Univariate Odds ratio P-value 95%CI Diabetes insulin treatment 1.316 (0.735-2.357) 0.355 Diabetes oral medication 1.432 (0.736-2.781) 0.291 Hypertension 1.363 (0.871-2.134) 0.175 Cardiovascular disease 2.854 (1.780-4.576) 0.000 Cerebrovascular disease 2.183 (0.630-7.566) 0.218 Atherosclerosis 3.664 (1.542-8.704) 0.003 COPD 3.219 (1.441-7.190) 0.004 Kidney failure 2.874 (1.065-7.760) 0.037 Endocrinal disease 1.835 (0.700-4.816) 0.217 Opioid use 1.56 (0.648-2.434) 0.500 Anticoagulant use 2.162 (1.335-3.502) 0.002 Benzodiazepam use Chemotherapy< 30days ago Smoker 1.585 (0.874-2.874) 0.130 Hospitalized patient 1.516 (0.925-2.414) 0.080 Outpatient case 0.706 (0.441-1.129) 0.146 P-Tromb <150% 2.194 (1.289-3.734) 0.004 P-TT <70% 2.151 (1.325-3.492) 0.002 P-INR >2 0.367 (0.123-1.096) 0.072 Abnormal ECG 2.121 (1.365-3.297) 0.001 Hyponatremia 2.641 (1.684-4.142) 0.000 Hypernatremia Hypokalemia 1.522 (0.587-3.947) 0.388 Hyperkalemia ASA 1-2 vs 3-5 3.70 (2.02-6.75) 0.000 Age >60 4.06 (2.41-6.85) 0.000 Anemia 1.89 (1.22-2.94) 0.005 Multivariate analysis of all patients Age >60yrs old 3.34 (1.95-5.72) 0.000 Female vs male 0.84 (0.53-1.34) 0.468 Atherosclerosis 2.41 (0.99-5.89) 0.053 P-tromb <150 1.87 (1.08-3.25) 0.025 Hyponatremia 2.22 (1.40-3.52) 0.001 Male patients Age >60yrs old 4.79 (2.23-10.30) 0.000 Atherosclerosis 2.58 (0.87-7.70) 0.088 P-Tromb <150 1.69 (0.81-3.53) 0.164 Hyponatremia 3.66 (1.98-6.75) 0.000 Female patients Age >60yrs old 2.14 (1.00-460) 0.050 Atherosclerosis 2.53 (0.50-12.89) 0.263 P-tromb <150 2.14 (0.92-4.99) 0.078

A split analysis was performed according to gender because a significant interaction was found between gender and hyponatremia.

Page 64: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

63

8. Discussion

In the last years, ERCP has evolved from a diagnostic to a predominantly therapeutic

procedure, and the use of this tool seems to be increasing. ERCP is still one of the most

technically complicated and challenging procedures since it requires a steep learning curve

to increase the success of the cannulation rate and to reduce the adverse events of the

procedure. PEP is the most common and severe complication of an ERCP. Other adverse

events include bleeding, cholangitis, and perforation.

8.1 ERCP for PSC patients (I)

For PSC patients the incidence of developing abdominal malignancies is high compared to

the general population. The lifetime incidence of cholangiocarcinoma in PSC patients is 20%,

while for hepatocellular carcinoma the lifetime incidence is 0.3%-2.8%, and for gallbladder

carcinoma it is 1-3.5% (Fung et al., 2019). In fact, 40%-50% of the mortality in PSC patients

occurs due to malignancies (Folseraas et al., 2016). ERCP is a sensitive and specific tool for

primary diagnosis and follow up of the progression of PSC. In the Helsinki University Hospital,

a PSC diagnosis is always confirmed by an ERCP and liver biopsy. In patients without PSC,

the adverse events associated with ERC have been reported to range from 4% to 16% (Etzel

et al., 2008; Bangarulingam et al, 2009; Cotton et al., 2009). The overall adverse events for

PSC patients have varied in different studies between 1.8% up to 18.4 {{261 European

Society of Gastrointestinal Endoscopy 2017}}. In our study, the overall complication rate was

9%.

Some studies that compare complication rates between PSC and non-PSC patients have

shown that the two groups have differed neither in the overall complication rate, nor in the

risks of PEP, perforation, or bleeding (Etzel et al., 2008; Bangarulingam et al., 2009). In their

study Bangarulingam et al. reported a higher rate of cholangitis in the PSC group (4% vs.

0.2%, p 0.0002). Additionally, the ERCP duration was longer for PSC patients who developed

Page 65: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

64

cholangitis. The differences between the reported complication rates can be explained by the

number of interventions carried out during the ERCP procedure. In our study, the cholangitis

rate was 1.4%, and no association was found between the procedure time and the

development of cholangitis. A previous biliary sphincterotomy did not affect the cholangitis

rate. The regular use of prophylactic antibiotics can explain these findings in our study. There

were no procedure-related deaths in this study, unlike in other studies that reported a

mortality rate of 0.4% (Freeman et al., 1996). The fact that the PSC patients were usually

young and had no severe comorbidities, may explain the absence of deaths in our study. For

instance, in the study by Freeman et al., half of the deceased were over 70 years old, and

the other half had severe comorbidities. Additionally, in our study, we could not find any

association between young age and PEP, which was reported in other studies (Freeman et

al., 1996; Wang, P. et al., 2009).

In our unit, the PEP rate for patients with native papilla was 4.8% for non-PSC patients, and

7% for PSC patients with native papilla. This is can be explained because the balloon

catheter insertion and several brushings and dilations, that take place in an ERC for PSC

patients are known to increase the overall risk of complications. The PEP rate in our study of

7% is slightly less than in previous studies (Etzel et al., 2008; Bangarulingam et al., 2009),

which can be attributed to the guidewire technique we used instead of a contrast injection.

Navaneethan et al. reported a rate of PEP as low as 1.2% and explained the likely reason to

be routine stenting of the PD after cannulation or injection in the PD (Navaneethan,

Jegadeesan et al., 2015), which we did not do routinely in our unit. The history of PEP was

not a risk factor in the present study. Female sex was associated with an increased risk of

PEP, which has also been reported in previous studies (Cotton et al., 1991; Wang et al.,

2009). However, there are studies that have found no association between sex and an

increased risk of PEP (Cheng et al., 2006; Etzel et al., 2008). An uninitiated wire insertion

into the PD increased the PEP. Having had a previous papillotomy was a protective factor

against PEP, which could probably be attributed to the ease of cannulating the bile duct,

Page 66: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

65

which reduces the mechanical irritation of the papilla. The accidental insertion of the

guidewire into the PD has been reported in many studies to be a risk factor for PEP (Cheng

et al., 2006; Cotton et al., 2009; Halttunen et al., 2014). However, one study has reported

that this is not a risk factor (Lella et al., 2004). The difference between the surveys can be

attributed to the difference in the PEP definition. In the study by Artifon et al., an amylase

level of five times the normal level was necessary for a PEP diagnosis, whereas in our study

it was three times the normal level. Still, in the study by Atrifon et al., an accidental guidewire

insertion into the PD did not cause PEP (Artifon et al., 2007).

The precut technique is a valuable method which can increase the cannulation success rates

up to 95% (Siege et al., 1989), but many studies have reported that a precut increased the

PEP rates (Freeman et al., 1996; Loperfido et al., 1998; Testoni et al., 2010). However, recent

meta-analyses showed that when an early precut was performed, the PEP rates were

significantly lower than in the late precut group (Cennamo et al., 2010; Mariani et al., 2016).

In our study, the precut technique was used in the most difficult cannulation cases. A previous

study from our unit showed that TPBS was safe, with a high cannulation success rate (97%)

compared to precut rates (71%), with no difference in the complication rates (Halttunen et

al., 2009). An additional precut after TPBS was needed in 42% of the cases in our previous

study (Halttunen et al., 2009), but in the present study it was 21%. The difference in the need

for a precut procedure after TPBS in the two studies could stem from the fact that in the

present study all the patients had PSC. However, the PSC score was no higher in the group

that needed TPBS. Thus, the difficulty of BD access cannot be entirely explained by the

stricture of the extrahepatic bile duct. A biliary sphincterotomy was associated with a 6.8%

risk of PEP (Freeman et al., 1996). A previously performed sphincterotomy was a significant

protective factor against PEP in our study. For this reason, we recommend to perform a

biliary sphincterotomy for PSC patients if repeat procedures are expected.

Page 67: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

66

8.2 Endoscopic papillectomy (II)

Jaundice and bile duct dilation are definite risk factors of malignant periampullary tumors, yet

in the present study, for benign tumours, jaundice was also present in 14% of patients.

Patients with cancer that survived were those who were operated and had no lymph node

metastasis. It is very hard with cancer patients to estimate complete endoscopic removal,

and the case in this study is a good example. With clean findings after a papillectomy, still, a

4 mm cancer was found in the pancreaticoduodenectomy specimen. A previous study

showed that pre-resection biopsies were unreliable in many cases (Irani et al., 2009). In the

present study, we came to the same conclusion since the biopsies revealed cancer in only

one case out of 10 cancer cases, and the papillectomy specimen was needed for diagnosis.

In the case of a papillary carcinoma, pancreaticoduodenectomy is the treatment of choice. In

this study, we saw that in spite of the follow-up, a small amount of cancer could be missed;

for this reason, many experts consider even high-grade adenomas to be an indication of

surgery (Beger et al., 1999; Kim, J. H. et al., 2009).

The frequency of ampullary tumours in FAP patients is 200-300 times higher compared to

the general population, with a risk of carcinoma up to 10% (Gluck et al., 2015). In the present

study, during the follow-up, none of the FAP patients developed cancer, and the concordance

between pre-resection and papillectomy specimens was up to 88%. The endoscopic

surveillance of FAP patients can delay the surgery, which can help patients with a high risk

of malignancy. With the lack of randomized controlled studies, the timing of the surgery is

still not clear. Additionally, adenomas can develop after surgery, which makes the decision

whether to operate or not, more complicated. In the present study, the PEP rate after a

papillectomy was 9.8% with mild and moderate forms. The use of a prophylactic stent

reduced the PEP rate, which was confirmed by previous research (Napoleon et al., 2013),

but locating the PD for stenting can be challenging. Some operators use a guidewire or

contrast to determine the PD. In our institution, we use methylene blue to identify the PD,

which is a safe technique (Catalano et al., 2004; Udd et al., 2010). Some prefer to perform a

Page 68: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

67

pancreatic papillotomy before a papillectomy to ensure pancreatic stenting afterwards

(Testoni, P. A., 2007). In this series, when a pancreatic papillotomy was performed without a

stent, the risk of PEP increased noticeably. By changing our policy and using prophylactic

stents every time after the PD was reached, we should be able to reduce the PEP rates. Non-

steroidal anti-inflammatory drugs (NSAIDs) can be more effective in preventing PEP than the

prophylactic stents (Akbar et al., 2013). Additional studies are needed to show whether both

NSAIDs and PD stents work synergistically or not, and until then, both can be used.

According to Napoleon et al., post-papillectomy adverse events include PEP (5-19%),

perforation (0-5%), bleeding (0-18%) and cholangitis (0-4%) (Napoleon et al., 2013). We had

a relatively high overall complication rate of 24.6%. Of these PEP accounted for 9.8%,

bleeding for 18%, perforation for 3.3%, and there was no cholangitis. All but one patient was

treated conservatively. Three deaths which were non-procedure related were reported. A

high bleeding percentage can be from our policy to perform a duodenoscopy as soon as any

suspicion appears. Coagulation of the resection margins after the papillectomy did not

prevent bleeding. However, this may be due to the retrospective nature of the study. Using

haemostatic agents in the papilla area can be dangerous, if they obstruct the pancreatic

orifice. Using haemostatic clips after pancreatic stenting could be one option to reduce the

bleeding rate.

8.3 The role of TPBS in difficult cannulations (III)

The guidewire-assisted cannulation technique was shown to be associated with greater

primary cannulation success and reduced PEP risk compared to contrast assisted

cannulations in one large systematic review (Tse et al., 2012). In the present study, the

difficult cannulation rate was 37%. As a first rescue cannulation technique, we used the TPBS

technique with a final success rate of 99% and a PEP rate of 4%. The high final success rate

can be attributed to our policy of attempting to obey the 5-5-2 rule and move to the rescue

method quickly as it was in the present study, in a median of 4.8 minutes.

Page 69: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

68

In a multicentre prospective study by Wang et al., the guidewire passage to the PD was an

independent risk factor for PEP (Wang et al., 2009). In the present study, the risk of PEP was

6.4% if the wire slipped more than once into the pancreatic duct. According to one meta-

analysis, a contrast injection into the pancreatic duct is a risk factor for PEP (Ding et al.,

2015), unlike in our study, which could not confirm the same. This can be attributed to the

fact that we used guide wire cannulation and hardly used contrast injections because usually

it is easy to assess the location of the guidewire using only a fluoroscope.

TPBS was first described by Goff et al. (Goff, 1999). It was described as a safe and effective

method for gaining access to the CBD in difficult cannulations, with a success rate range

between 86-100%, and acceptable PEP rates (3.5-10.4%) (Goff, 1999; Weber et al., 2008;

Halttunen et al., 2009; Kim, S. J. et al., 2015). In our previous study, the success rate of

TPBS was 97% in difficult cannulations. The PEP rate was 9.3%, and when an additional

needle-knife cut was performed, the PEP rate was 8.2%. When only a needle-knife cut was

performed, the PEP rate was 5.1%. In a previous study from our unit, the BD cannulation

success rate of the needle knife group was lower compared to the TPBS group (71.3% vs.

97.3%, p<0.001) (Halttunen et al., 2009).

DGW cannulation is another rescue technique that needs additional guidewire in the PD to

straighten the BD, to make the cannulation of the BD easier. In cases of a tight tumour

stricture, the additional guidewire may hamper the cannulation and even block access.

Although when the first guidewire is placed already in the PD, the second guidewire can still

easily enter the PD. In a study comparing DGW and TPBS techniques as rescue methods,

the cannulation time for the DGW group was 7.8 +/- 1.7 minutes, and for the TPBS group it

was 3.7 +/-2.3 minutes (p<0.001), and the complication rate was 2.19% vs. 7.04%, (p=0.04)

respectively. It is noteworthy that the primary cannulations in this study succeeded in only

23.8% of the cases (Huang et al., 2015), which differs from other studies with primary

successful cannulation rates of 75-90% (Ito et al., 2014; Kim et al., 2015) . Another study that

compared DGW with needle knife techniques after the failure of a primary cannulation, found

Page 70: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

69

that the success rate was 44.8% in the DGW group and 79.1% in the needle knife group.

The cannulation time was 312 seconds vs. 257 seconds, and the PEP rate was 14.9% vs.

4.5%, respectively (Kim et al., 2015). It is essential to register the data during the ERCP,

concerning the cannulation time, the number of attempts and the number of passages into

the PD, to monitor the rate of difficult cannulation criteria in clinical practice.

8.4 RPLT for ERCP (IV)

The average utilization of ERCP increased over time up to 105 ERCPs /100,000 person/year

driven by therapeutic procedures (Coelho-Prabhu et al., 2013). The need for ERCP will most

likely rise more in the future. This means that the cost of the preoperative testing will also

rise. In our institution, an RPTL package costs about 41€ per one elective patient, and 66€

in emergency cases. Many studies previously showed an excessive RPLT to have no true

effect on clinical decisions (Pasternak, 2004; Czoski-Murray et al., 2012; ASGE Standards

of Practice Committee et al., 2014). Although the majority of these studies focused on RPTLs

for patients undergoing surgery, we can assume that ERCP as the less invasive procedure

needs even fewer RPTLs than surgical patients. The rate of the adverse events in the present

study was similar to other studies (Cohen et al., 1996; Andriulli et al., 2007). We found that

none of the RPTLs that were used were directly associated with any complications of ERCP.

Still an association was found with thrombocytopenia, hyponatremia, and cardiovascular

depression. However, the reason for this association is unclear, and further studies are

needed. Usually, hyponatremia and thrombocytopenia are causes of other health-related

issues, which most likely is the reason for hypotension during sedation. Correcting these

values probably would not alleviate the problem. Still, a clinical assessment of the patient

before the procedure may inform the anaesthesiologist of the cardiovascular depression risk,

even without RPTLs. This does not mean that we should abandon the preoperative testing

completely, but to focus on patients that would benefit from them. In the present study,

anemia was found to be a protective factor for respiratory depression caused by sedation,

especially for patients without cardiovascular disease, which the sensitization to hypoxemia

Page 71: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

70

that anemia causes could explain. However, the clinical utilization of this finding is difficult,

since anemia is an issue to be avoided in clinical practice.

8.5 Limitations of the studies

In study I, we included a large cohort of PSC patients who underwent ERCP for primary

diagnosis, surveillance of progression, and therapeutic purposes. For PSC cases,

prophylactic pancreatic stents were not used routinely in difficult cannulations. In high-risk

patients, prophylactic placement of a pancreatic stent decreases the risk of PEP. As in our

series there was a high PEP rate in patients with the most difficult cannulations, placement

of a pancreatic stent could be beneficial in such cases.

For papillectomy cases, a prophylactic pancreatic stent was used more often, but not for all

patients (Study II). Additionally, we did not have specific instructions on when we crossed

over to the rescue method in Study III. The use of diclofenac before the ERCP began in

September 2013 during the data collection phase of the studies. In cases of difficult

procedures, the change to a more experienced endoscopist was not documented.

Additionally, we did not collect the difficult cannulation criteria for every patient because of

the busy daily practice. We rarely used DGW as a rescue method because it has been quite

unreliable in our hands. Instead, we used TPBS as the first rescue method.

In Study IV the relatively small number of complications made it difficult to draw reliable

conclusions. However, the study is still a good foundation for additional studies on the

subject.

Page 72: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

71

9. Future aspects

Although advances have been made, we still need to reduce the rate of PEP. The

combination between more than one prophylactic procedure (e.g. diclofenac, pancreatic

stent, aggressive hydration) can reduce the rate of PEP. In our institution we are going to

begin a study in which we compare the using of diclofenac suppository prior to native papilla

ERC in the same time with using of pancreatic stent after PD sphincterotomy, and compare

it to using of only diclofenac, and compare the impact on PEP developing.

Avoidance of unnecessary ERCP is the best way to reduce the number of complications.

Professional practice guidelines inform the appropriateness of preoperative noninvasive

imaging or preoperative ERCP, respectively, before cholecystectomy based on pretest

probability of retained BD stone. Institutional adoption of a standard algorithm for

management of such patients may minimize redundant investigations, including ERCP.

Compared with cholangiography, EUS is highly sensitive and specific in the diagnosis of

choledocholithiasis, and has predominant role in defining pancreaticobiliary lesions because

of its relative safety and its superiority over ERCP in tissue acquisition. Endoscopists

performing ERCP may also be skilled in EUS. It would also be beneficial to have randomized,

controlled trials comparing ERCP- vs. EUS -facilitated biliary drainage in patients with

unresectable malignant obstructive jaundice.

Duodenoscopes should be redesigned to eliminate or minimize infections resulting from

difficult to clean elevators. Transmission of multi- drug resistant organisms by

duodenoscopes is not novel occurrence, with prior reports including transmission of

Pseudomonas aeruginosa and Klebsiella pneumoniae occurring in the setting of known

significant breach in endoscope reprocessing.

It would be great to expand the capabilities of ERCP to allow for something to be placed in

the pancreatic duct that could dissolve stones instead of using electrohydraulic, laser, or

extracorporeal shockwave lithotripsy. The potential injection of litholytic agents into the

Page 73: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

72

pancreas for chronic calcific pancreatitis, and directed infusion of chemotherapeutic or

immunomodulatory agents into the pancreaticobiliary tree may be a potential future scenario.

Page 74: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

73

10. Summary and conclusions

The following conclusions can be drawn from this study:

1. ERC for PSC patients is accompanied with a fairly low risk of severe adverse events and

no mortality. For PSC patients undergoing dysplasia surveillance with repeated ERC, we

recommend biliary sphincterotomy during the first ERC to prevent PEP after the next

procedures. Biliary sphincterotomy is not associated with ERCP-associated cholangitis.

2. For benign tumours, endoscopic ampullectomy is a relatively safe and effective

treatment. Jaundice and bile duct dilation are risk factors for malignancy. Although

residual adenomas occur frequently, they can be treated endoscopically or

conservatively. Malignant tumors should be treated surgically. Prophylactic pancreatic

stenting reduces the risk of PEP.

3. To monitor difficult cannulation cases, the cannulation attempts, and cannulation time

must be recorded; also, the number of guidewire passages into the PD must be counted.

This will help in the timing of crossover to another cannulation method. TPBS is an

efficient and safe rescue method.

4. Routine RPLTs often fail to predict adverse effects of ERCP. The RPLTs that should be

considered before ERCP are: a basic blood count, creatinine, P-NA, P-K, in patients with

ASA 4-5, and with coronary artery disease, atherosclerosis, kidney failure, or patients

consuming diuretics. If the patient takes oral anticoagulants, then P-INR must be

checked. Additional RPLTs should be considered according to the patient's health status.

For young, healthy patients RPLTs are not needed.

Page 75: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

74

11. Acknowledgements

This study was carried out at the Department of Surgery, University of Helsinki, and Helsinki University

Hospital, during the years of 2000-2020.

I want to express my sincere gratitude and respect to the academic head of the department, Professor

Pauli Puolakkainen, for the opportunity to carry out this thesis and for creating an inspiring and

supportive academic environment to work.

I owe my deepest gratitude to my supervisors Docent Leena Kylänpää and Docent Marianne Udd, for

not losing faith in me, for always being available and tirelessly helping me through the demanding

phases in the process. My first steps in the field of clinical research were not smooth, without their

never-ending optimism and vast knowledge in the clinical field, it would have been impossible to

accomplish this thesis. I truly admire both of them.

I sincerely thank my reviewers Professor Tuomo Rantanen and Professor Juha Saarnio for their

excellent and constructive comments. I also thank Docent Urban Arnelo for agreeing to be my

opponent. My present superiors, Docent Esko Kemppainen, Docent Leena Halme, are sincerely

acknowledged for their kind support.

All of my co-authors are thanked for their invaluable help: Professor Martti Färkkilä, Professor Heikki

Järvinen, Docent Jorma Halttunen, Docent Kalle Jokelainen, Docent Reino Pöyhiä, Dr Outi Lindström,

Dr Mia Rainio and Dr Jarno Jokelainen. I am deeply indebted for Harri Mustonen for his help with

statistical analysis.

I warmly thank Elina Aspiala for her kindness and for the effort she took to make the thesis appear in

this way. I also warmly thank Docent Arno Nordin for his niceness and support.

My deepest thanks to my lovely wife Nina and our dear children Omar and Sami. You are the most

important people in my life.

Helsinki, December 2020

Shamel

Page 76: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

75

Page 77: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

76

12. References

Ahmed, A. M., & Wilcox, C. M. (2017). Endoscopic retrograde cholangiopancreatography

cannulation in the difficult duct: Which technique should you pull out of your bag? Digestive

Endoscopy, 29(2), 149-151.

Akbar, A., Abu Dayyeh, B. K., Baron, T. H., Wang, Z., Altayar, O., & Murad, M. H. (2013). Rectal

nonsteroidal anti-inflammatory drugs are superior to pancreatic duct stents in preventing

pancreatitis after endoscopic retrograde cholangiopancreatography: A network meta-analysis.

Clinical Gastroenterology and Hepatology: The Official Clinical Practice Journal of the

American Gastroenterological Association, 11(7), 778-783. doi:10.1016/j.cgh.2012.12.043

[doi]

Akshintala, V. S., Hutfless, S. M., Colantuoni, E., Kim, K. J., Khashab, M. A., Li, T., Singh, V. K.

(2013). Systematic review with network meta-analysis: Pharmacological prophylaxis against

post-ERCP pancreatitis. Alimentary Pharmacology & Therapeutics, 38(11-12), 1325-1337.

doi:10.1111/apt.12534 [doi]

Alfieri, S., Rosa, F., Cina, C., Tortorelli, A. P., Tringali, A., Perri, V., Doglietto, G. B. (2013).

Management of duodeno-pancreato-biliary perforations after ERCP: Outcomes from an Italian

tertiary referral centre. Surgical Endoscopy, 27(6), 2005-2012. doi:10.1007/s00464-012-2702-

9 [doi]

Almadi, M. A., Barkun, A., & Martel, M. (2017). Plastic vs. self-expandable metal stents for

palliation in malignant biliary obstruction: A series of meta-analyses. The American Journal of

Gastroenterology, 112(2), 260-273. doi:10.1038/ajg.2016.512 [doi]

Page 78: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

77

Andriulli, A., Loperfido, S., Napolitano, G., Niro, G., Valvano, M. R., Spirito, F., Forlano, R. (2007).

Incidence rates of post-ERCP complications: A systematic survey of prospective studies. The

American Journal of Gastroenterology, 102(8), 1781-1788. doi:AJG1279 [pii]

Artifon, E. L., Sakai, P., Cunha, J. E., Halwan, B., Ishioka, S., & Kumar, A. (2007). Guidewire

cannulation reduces risk of post-ERCP pancreatitis and facilitates bile duct cannulation. The

American Journal of Gastroenterology, 102(10), 2147-2153. doi:AJG1378 [pii]

ASGE Standards of Practice Committee, Buxbaum, J. L., Abbas Fehmi, S. M., Sultan, S.,

Fishman, D. S., Qumseya, B. J., Wani, S. B. (2019). ASGE guideline on the role of endoscopy

in the evaluation and management of choledocholithiasis. Gastrointestinal Endoscopy, 89(6),

1075-1105.e15. doi:S0016-5107(18)33162-6 [pii]

ASGE Standards of Practice Committee, Pasha, S. F., Acosta, R., Chandrasekhara, V., Chathadi,

K. V., Eloubeidi, M. A., Cash, B. (2014). Routine laboratory testing before endoscopic

procedures. Gastrointestinal Endoscopy, 80(1), 28-33. doi:10.1016/j.gie.2014.01.019 [doi]

ASGE Technology Assessment Committee, Pfau, P. R., Pleskow, D. K., Banerjee, S., Barth, B. A.,

Bhat, Y. M., Rodriguez, S. A. (2013). Pancreatic and biliary stents. Gastrointestinal

Endoscopy, 77(3), 319-327. doi:10.1016/j.gie.2012.09.026 [doi]

Attwell, A., Borak, G., Hawes, R., Cotton, P., & Romagnuolo, J. (2006). Endoscopic pancreatic

sphincterotomy for pancreas divisum by using a needle-knife or standard pull-type technique:

Safety and reintervention rates. Gastrointestinal Endoscopy, 64(5), 705-711. doi:S0016-

5107(06)00558-X [pii]

Bagul, A., Pollard, C., & Dennison, A. R. (2010). A review of problems following insertion of biliary

stents illustrated by an unusual complication. Annals of the Royal College of Surgeons of

England, 92(4), W27-31. doi:10.1308/147870810X12659688852239 [doi]

Page 79: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

78

Bai, Y., Gao, J., Shi, X., Zou, D., & Li, Z. (2008). Prophylactic corticosteroids do not prevent post-

ERCP pancreatitis: A meta-analysis of randomized controlled trials. Pancreatology: Official

Journal of the International Association of Pancreatology (IAP) [Et Al.], 8(4-5), 504-509.

doi:10.1159/000151999 [doi]

Bailey, A. A., Bourke, M. J., Williams, S. J., Walsh, P. R., Murray, M. A., Lee, E. Y., Lynch, P. M.

(2008). A prospective randomized trial of cannulation technique in ERCP: Effects on technical

success and post-ERCP pancreatitis. Endoscopy, 40(4), 296-301. doi:10.1055/s-2007-995566

[doi]

Bangarulingam, S. Y., Gossard, A. A., Petersen, B. T., Ott, B. J., & Lindor, K. D. (2009).

Complications of endoscopic retrograde cholangiopancreatography in primary sclerosing

cholangitis. The American Journal of Gastroenterology, 104(4), 855-860.

doi:10.1038/ajg.2008.161 [doi]

Banks, P. A., Bollen, T. L., Dervenis, C., Gooszen, H. G., Johnson, C. D., Sarr, M. G., Acute

Pancreatitis Classification Working Group. (2013). Classification of acute pancreatitis--2012:

Revision of the Atlanta classification and definitions by international consensus. Gut, 62(1),

102-111. doi:10.1136/gutjnl-2012-302779 [doi]

Beger, H. G., Treitschke, F., Gansauge, F., Harada, N., Hiki, N., & Mattfeldt, T. (1999). Tumour of

the ampulla of Vater: Experience with local or radical resection in 171 consecutively treated

patients. Archives of Surgery (Chicago, Ill.: 1960), 134(5), 526-532.

Benarroch-Gampel, J., Sheffield, K. M., Duncan, C. B., Brown, K. M., Han, Y., Townsend, C. M., &

Riall, T. S. (2012). Preoperative laboratory testing in patients undergoing elective, low-risk

ambulatory surgery. Annals of Surgery, 256(3), 518-528.

doi:10.1097/SLA.0b013e318265bcdb [doi]

Page 80: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

79

Berry, R., Han, J. Y., & Tabibian, J. H. (2019). Difficult biliary cannulation: Historical perspective,

practical updates, and guide for the endoscopist. World Journal of Gastrointestinal

Endoscopy, 11(1), 5-21. doi:10.4253/wjge.v11.i1.5 [doi]

Bjork, J., Akerbrant, H., Iselius, L., Alm, T., & Hultcrantz, R. (1999). Epidemiology of familial

adenomatous polyposis in Sweden: Changes over time and differences in phenotype between

males and females. Scandinavian Journal of Gastroenterology, 34(12), 1230-1235.

doi:10.1080/003655299750024751 [doi]

Boberg, K. M., Bergquist, A., Mitchell, S., Pares, A., Rosina, F., Broome, U., Schrumpf, E. (2002).

Cholangiocarcinoma in primary sclerosing cholangitis: Risk factors and clinical presentation.

Scandinavian Journal of Gastroenterology, 37(10), 1205-1211.

doi:10.1080/003655202760373434 [doi]

Bove, V., Tringali, A., Familiari, P., Gigante, G., Boskoski, I., Perri, V., Costamagna, G. (2015).

ERCP in patients with prior Billroth II gastrectomy: Report of 30 years' experience.

Endoscopy, 47(7), 611-616. doi:10.1055/s-0034-1391567 [doi]

Boyd, S., Mustonen, H., Tenca, A., Jokelainen, K., Arola, J., & Farkkila, M. A. (2017). Surveillance

of primary sclerosing cholangitis with ERC and brush cytology: Risk factors for

cholangiocarcinoma. Scandinavian Journal of Gastroenterology, 52(2), 242-249.

doi:10.1080/00365521.2016.1250281 [doi]

Brand, M., Bizos, D., & O'Farrell, P. (2010). Antibiotic prophylaxis for patients undergoing elective

endoscopic retrograde cholangiopancreatography. The Cochrane Database of Systematic

Reviews, (10):CD007345. doi(10), CD007345. doi:10.1002/14651858.CD007345.pub2 [doi]

Carter, J. T., Grenert, J. P., Rubenstein, L., Stewart, L., & Way, L. W. (2009). Neuroendocrine

tumours of the ampulla of Vater: Biological behavior and surgical management. Archives of

Surgery (Chicago, Ill.: 1960), 144(6), 527-531. doi:10.1001/archsurg.2009.80 [doi]

Page 81: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

80

Catalano, M. F., Linder, J. D., Chak, A., Sivak, M. V., Raijman, I., Geenen, J. E., & Howell, D. A.

(2004). Endoscopic management of adenoma of the major duodenal papilla. Gastrointestinal

Endoscopy, 59(2), 225-232. doi:S0016510703023666 [pii]

Catalano, M. F., Linder, J. D., & Geenen, J. E. (2004). Endoscopic transpancreatic papillary

septotomy for inaccessible obstructed bile ducts: Comparison with standard pre-cut

papillotomy. Gastrointestinal Endoscopy, 60(4), 557-561. doi:S0016510704018772 [pii]

Cennamo, V., Fuccio, L., Zagari, R. M., Eusebi, L. H., Ceroni, L., Laterza, L., Bazzoli, F. (2009).

Can a wire-guided cannulation technique increase bile duct cannulation rate and prevent post-

ERCP pancreatitis?: A meta-analysis of randomized controlled trials. The American Journal of

Gastroenterology, 104(9), 2343-2350. doi:10.1038/ajg.2009.269 [doi]

Cennamo, V., Fuccio, L., Zagari, R. M., Eusebi, L. H., Ceroni, L., Laterza, L., Bazzoli, F. (2010).

Can early precut implementation reduce endoscopic retrograde cholangiopancreatography-

related complication risk? Meta-analysis of randomized controlled trials. Endoscopy, 42(5),

381-388. doi:10.1055/s-0029-1243992 [doi]

Cha, S. W., Leung, W. D., Lehman, G. A., Watkins, J. L., McHenry, L., Fogel, E. L., & Sherman, S.

(2013). Does leaving a main pancreatic duct stent in place reduce the incidence of precut

biliary sphincterotomy-associated pancreatitis? A randomized, prospective study.

Gastrointestinal Endoscopy, 77(2), 209-216. doi:10.1016/j.gie.2012.08.022 [doi]

Chacko, L. N., Chen, Y. K., & Shah, R. J. (2008). Clinical outcomes and nonendoscopic

interventions after minor papilla endotherapy in patients with symptomatic pancreas divisum.

Gastrointestinal Endoscopy, 68(4), 667-673. doi:10.1016/j.gie.2008.01.025 [doi]

Chang, J. H., Lee, I., Choi, M. G., & Han, S. W. (2016). Current diagnosis and treatment of benign

biliary strictures after living donor liver transplantation. World Journal of Gastroenterology,

22(4), 1593-1606. doi:10.3748/wjg.v22.i4.1593 [doi]

Page 82: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

81

Chee, Y. L., Crawford, J. C., Watson, H. G., & Greaves, M. (2008). Guidelines on the assessment

of bleeding risk prior to surgery or invasive procedures. British Committee for Standards in

Haematology. British Journal of Haematology, 140(5), 496-504. doi:10.1111/j.1365-

2141.2007.06968.x [doi]

Chen, J. J., Wang, X. M., Liu, X. Q., Li, W., Dong, M., Suo, Z. W., Li, Y. (2014). Risk factors for

post-ERCP pancreatitis: A systematic review of clinical trials with a large sample size in the

past 10 years. European Journal of Medical Research, 19, 26-783X. doi:10.1186/2047-783X-

19-26 [doi]

Chen, S., Shi, H., Zou, X., & Luo, H. (2010). Role of ulinastatin in preventing post-endoscopic

retrograde cholangiopancreatography pancreatitis: The emperor's new clothes or Aladdin's

magic lamp? Pancreas, 39(8), 1231-1237. doi:10.1097/MPA.0b013e3181dc67e7 [doi]

Cheng, C. L., Sherman, S., Watkins, J. L., Barnett, J., Freeman, M., Geenen, J., Lehman, G. A.

(2006). Risk factors for post-ERCP pancreatitis: A prospective multicentre study. The

American Journal of Gastroenterology, 101(1), 139-147. doi:AJG380 [pii]

Cheon, Y. K., Cho, K. B., Watkins, J. L., McHenry, L., Fogel, E. L., Sherman, S., & Lehman, G. A.

(2007). Frequency and severity of post-ERCP pancreatitis correlated with extent of pancreatic

ductal opacification. Gastrointestinal Endoscopy, 65(3), 385-393. doi:S0016-5107(06)03123-3

[pii]

Cheung, J., Tsoi, K. K., Quan, W. L., Lau, J. Y., & Sung, J. J. (2009). Guidewire versus

conventional contrast cannulation of the common bile duct for the prevention of post-ERCP

pancreatitis: A systematic review and meta-analysis. Gastrointestinal Endoscopy, 70(6), 1211-

1219. doi:10.1016/j.gie.2009.08.007 [doi]

Choudhary, A., Winn, J., Siddique, S., Arif, M., Arif, Z., Hammoud, G. M., Bechtold, M. L. (2014).

Effect of precut sphincterotomy on post-endoscopic retrograde cholangiopancreatography

Page 83: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

82

pancreatitis: A systematic review and meta-analysis. World Journal of Gastroenterology,

20(14), 4093-4101. doi:10.3748/wjg.v20.i14.4093 [doi]

Chung, F., Yuan, H., Yin, L., Vairavanathan, S., & Wong, D. T. (2009). Elimination of preoperative

testing in ambulatory surgery. Anesthesia and Analgesia, 108(2), 467-475.

doi:10.1213/ane.0b013e318176bc19 [doi]

Clarke, B., Slivka, A., Tomizawa, Y., Sanders, M., Papachristou, G. I., Whitcomb, D. C., & Yadav,

D. (2012). Endoscopic therapy is effective for patients with chronic pancreatitis. Clinical

Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American

Gastroenterological Association, 10(7), 795-802. doi:10.1016/j.cgh.2011.12.040 [doi]

Coelho-Prabhu, N., Shah, N. D., Van Houten, H., Kamath, P. S., & Baron, T. H. (2013).

Endoscopic retrograde cholangiopancreatography: Utilisation and outcomes in a 10-year

population-based cohort. BMJ Open, 3(5), 10.1136/bmjopen-2013. doi:10.1136/bmjopen-

2013-002689 [doi]

Cohen, S. A., Siegel, J. H., & Kasmin, F. E. (1996). Complications of diagnostic and therapeutic

ERCP. Abdominal Imaging, 21(5), 385-394.

Committee on Standards and Practice Parameters, Apfelbaum, J. L., Connis, R. T., Nickinovich, D.

G., American Society of Anesthesiologists Task Force on Preanesthesia Evaluation,

Pasternak, L. R., Twersky, R. S. (2012). Practice advisory for preanesthesia evaluation: An

updated report by the American Society of Anesthesiologists Task Force on Preanesthesia

evaluation. Anesthesiology, 116(3), 522-538. doi:10.1097/ALN.0b013e31823c1067 [doi]

Cotton, P. B., Durkalski, V., Romagnuolo, J., Pauls, Q., Fogel, E., Tarnasky, P., Robuck, P. (2014).

Effect of endoscopic sphincterotomy for suspected sphincter of Oddi dysfunction on pain-

related disability following cholecystectomy: The EPISOD randomized clinical trial. Jama,

311(20), 2101-2109. doi:10.1001/jama.2014.5220 [doi]

Page 84: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

83

Cotton, P. B., Eisen, G. M., Aabakken, L., Baron, T. H., Hutter, M. M., Jacobson, B. C., Vargo, J. J.

(2010). A lexicon for endoscopic adverse events: Report of an ASGE workshop.

Gastrointestinal Endoscopy, 71(3), 446-454. doi:10.1016/j.gie.2009.10.027 [doi]

Cotton, P. B., Garrow, D. A., Gallagher, J., & Romagnuolo, J. (2009). Risk factors for complications

after ERCP: A multivariate analysis of 11,497 procedures over 12 years. Gastrointestinal

Endoscopy, 70(1), 80-88. doi:10.1016/j.gie.2008.10.039 [doi]

Cotton, P. B., Lehman, G., Vennes, J., Geenen, J. E., Russell, R. C. G., Meyers, W. C., Nickl, N.

(1991). Endoscopic sphincterotomy complications and their management: An attempt at

consensus. Gastrointestinal Endoscopy, 37(3), 383-393.

Czoski-Murray, C., Lloyd Jones, M., McCabe, C., Claxton, K., Oluboyede, Y., Roberts, J., Fleming,

T. (2012a). What is the value of routinely testing full blood count, electrolytes and urea, and

pulmonary function tests before elective surgery in patients with no apparent clinical indication

and in subgroups of patients with common comorbidities: A systematic review of the clinical

and cost-effective literature. Health Technology Assessment (Winchester, England), 16(50), i-

xvi, 1. doi:10.3310/hta16500 [doi]

Davee, T., Garcia, J. A., & Baron, T. H. (2012). Precut sphincterotomy for selective biliary duct

cannulation during endoscopic retrograde cholangiopancreatography. Annals of

Gastroenterology, 25(4), 291-302.

De Palma, G. D., Galloro, G., Siciliano, S., Iovino, P., & Catanzano, C. (2001). Unilateral versus

bilateral endoscopic hepatic duct drainage in patients with malignant hilar biliary obstruction:

Results of a prospective, randomized, and controlled study. Gastrointestinal Endoscopy,

53(6), 547-553. doi:S0016-5107(01)70250-7 [pii]

de Vries, J. H., Duijm, L. E., Dekker, W., Guit, G. L., Ferwerda, J., & Scholten, E. T. (1997). CT

before and after ERCP: Detection of pancreatic pseudotumour, asymptomatic retroperitoneal

Page 85: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

84

perforation, and duodenal diverticulum. Gastrointestinal Endoscopy, 45(3), 231-235.

doi:S0016-5107(97)70264-5 [pii]

Decker, C., Christein, J. D., Phadnis, M. A., Wilcox, C. M., & Varadarajulu, S. (2011). Biliary metal

stents are superior to plastic stents for preoperative biliary decompression in pancreatic

cancer. Surgical Endoscopy, 25(7), 2364-2367. doi:10.1007/s00464-010-1552-6 [doi]

Devereaux, B. M., Fein, S., Purich, E., Trout, J. R., Lehman, G. A., Fogel, E. L., Sherman, S.

(2003). A new synthetic porcine secretin for facilitation of cannulation of the dorsal pancreatic

duct at ERCP in patients with pancreas divisum: A multicentre, randomized, double-blind

comparative study. Gastrointestinal Endoscopy, 57(6), 643-647. doi:10.1067/mge.2003.195

[doi]

Ding, J., Jin, X., Pan, Y., Liu, S., & Li, Y. (2013). Glyceryl trinitrate for prevention of post-ERCP

pancreatitis and improve the rate of cannulation: A meta-analysis of prospective, randomized,

controlled trials. PloS One, 8(10), e75645. doi:10.1371/journal.pone.0075645 [doi]

Ding, X., Zhang, F., & Wang, Y. (2015). Risk factors for post-ERCP pancreatitis: A systematic

review and meta-analysis. The Surgeon: Journal of the Royal Colleges of Surgeons of

Edinburgh and Ireland, 13(4), 218-229. doi:10.1016/j.surge.2014.11.005 [doi]

Domagk, D., Oppong, K. W., Aabakken, L., Czako, L., Gyokeres, T., Manes, G., Rutter, M. D.

(2018). Performance measures for ERCP and endoscopic ultrasound: A European Society of

Gastrointestinal Endoscopy (ESGE) quality improvement initiative. Endoscopy, 50(11), 1116-

1127. doi:10.1055/a-0749-8767 [doi]

Draganov, P. V., Lin, T., Chauhan, S., Wagh, M. S., Hou, W., & Forsmark, C. E. (2011).

Prospective evaluation of the clinical utility of ERCP-guided cholangiopancreatoscopy with a

new direct visualization system. Gastrointestinal Endoscopy, 73(5), 971-979.

doi:10.1016/j.gie.2011.01.003 [doi]

Page 86: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

85

Dumonceau, J. M., Kapral, C., Aabakken, L., Papanikolaou, I. S., Tringali, A., Vanbiervliet, G., van

Hooft, J. E. (2020). ERCP-related adverse events: European Society of Gastrointestinal

Endoscopy (ESGE) guideline. Endoscopy, 52(2), 127-149. doi:10.1055/a-1075-4080 [doi]

Dumonceau, J. M., Tringali, A., Papanikolaou, I. S., Blero, D., Mangiavillano, B., Schmidt, A., van

Hooft, J. E. (2018). Endoscopic biliary stenting: Indications, choice of stents, and results:

European Society of Gastrointestinal Endoscopy (ESGE) clinical guideline - updated October

2017. Endoscopy, 50(9), 910-930. doi:10.1055/a-0659-9864 [doi]

Enns, R., Eloubeidi, M. A., Mergener, K., Jowell, P. S., Branch, M. S., Pappas, T. M., & Baillie, J.

(2002). ERCP-related perforations: Risk factors and management. Endoscopy, 34(4), 293-

298. doi:10.1055/s-2002-23650 [doi]

Etzel, J. P., Eng, S. C., Ko, C. W., Lee, S. D., Saunders, M. D., Tung, B. Y., Kowdley, K. V. (2008).

Complications after ERCP in patients with primary sclerosing cholangitis. Gastrointestinal

Endoscopy, 67(4), 643-648. doi:S0016-5107(07)02442-X [pii]

European Society of Gastrointestinal Endoscopy, European Association for the Study of the Liver

Electronic address: easloffice@easloffice eu, & European Association for the Study of the

Liver. (2017). Role of endoscopy in primary sclerosing cholangitis: European Society of

Gastrointestinal Endoscopy (ESGE) and European Association for the Study of the Liver

(EASL) clinical guideline. Journal of Hepatology, 66(6), 1265-1281. doi:S0168-

8278(17)30108-3 [pii]

Farnik, H., Weigt, J., Malfertheiner, P., Grutzmann, A., Gossner, L., Friedrich-Rust, M., Albert, J. G.

(2014). A multicentre study on the role of direct retrograde cholangioscopy in patients with

inconclusive endoscopic retrograde cholangiography. Endoscopy, 46(1), 16-21. doi:10.1055/s-

0033-1359043 [doi]

Page 87: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

86

Farrell, R. J., Khan, M. I., Noonan, N., O'Byrne, K., & Keeling, P. W. (1996). Endoscopic

papillectomy: A novel approach to difficult cannulation. Gut, 39(1), 36-38.

doi:10.1136/gut.39.1.36 [doi]

Ferreira, L. E., & Baron, T. H. (2010). Endoscopic stenting for palliation of malignant biliary

obstruction. Expert Review of Medical Devices, 7(5), 681-691. doi:10.1586/erd.10.36 [doi]

Fogel, E. L., deBellis, M., McHenry, L., Watkins, J. L., Chappo, J., Cramer, H., Lehman, G. A.

(2006). Effectiveness of a new long cytology brush in the evaluation of malignant biliary

obstruction: A prospective study. Gastrointestinal Endoscopy, 63(1), 71-77. doi:S0016-

5107(05)02748-3 [pii]

Folseraas, T., & Boberg, K. M. (2016). Cancer risk and surveillance in primary sclerosing

cholangitis. Clinics in Liver Disease, 20(1), 79-98. doi:10.1016/j.cld.2015.08.014 [doi]

Foutch, P. G. (1991). Endoscopic management of large common duct stones. The American

Journal of Gastroenterology, 86(11), 1561-1565.

Freeman, M. L. (1997). Complications of endoscopic biliary sphincterotomy: A review. Endoscopy,

29(4), 288-297. doi:10.1055/s-2007-1004193

Freeman, M. L., DiSario, J. A., Nelson, D. B., Fennerty, M. B., Lee, J. G., Bjorkman, D. J., Roel, J.

P. (2001). Risk factors for post-ERCP pancreatitis: A prospective, multicentre study.

Gastrointestinal Endoscopy, 54(4), 425-434. doi:S0016-5107(01)70200-3 [pii]

Freeman, M. L., & Guda, N. M. (2004). Prevention of post-ERCP pancreatitis: A comprehensive

review. Gastrointestinal Endoscopy, 59(7), 845-864. doi:S0016-5107(04)00353-0 [pii]

Freeman, M. L., & Guda, N. M. (2005). ERCP cannulation: A review of reported techniques.

Gastrointestinal Endoscopy, 61(1), 112-125. doi:S0016-5107(04)02463-0 [pii]

Page 88: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

87

Freeman, M. L., Nelson, D. B., Sherman, S., Haber, G. B., Herman, M. E., Dorsher, P. J., Logan,

G. M. (1996). Complications of endoscopic biliary sphincterotomy. New England Journal of

Medicine, 335(13), 909-918.

Freeman, M. L., & Overby, C. (2003). Selective MRCP and CT-targeted drainage of malignant hilar

biliary obstruction with self-expanding metallic stents. Gastrointestinal Endoscopy, 58(1), 41-

49. doi:10.1067/mge.2003.292 [doi]

Fung, B. M., Lindor, K. D., & Tabibian, J. H. (2019). Cancer risk in primary sclerosing cholangitis:

Epidemiology, prevention, and surveillance strategies. World Journal of Gastroenterology,

25(6), 659-671. doi:10.3748/wjg.v25.i6.659 [doi]

Genzlinger, J. L., McPhee, M. S., Fisher, J. K., Jacob, K. M., & Helzberg, J. H. (1999). Significance

of retroperitoneal air after endoscopic retrograde cholangiopancreatography with

sphincterotomy. The American Journal of Gastroenterology, 94(5), 1267-1270. doi:S0002-

9270(99)00051-9 [pii]

Geramizadeh, B., Moughali, M., Shahim-Aein, A., Memari, S., Ghetmiri, Z., Taghavi, A., & Bagheri

Lankarani, K. (2018). False negative and false positive rates in common bile duct brushing

cytology, a single centre experience. Gastroenterology and Hepatology from Bed to Bench,

11(4), 296-300.

Glomsaker, T., Hoff, G., Kvaloy, J. T., Soreide, K., Aabakken, L., Soreide, J. A., & Norwegian

Gastronet ERCP Group. (2013). Patterns and predictive factors of complications after

endoscopic retrograde cholangiopancreatography. The British Journal of Surgery, 100(3),

373-380. doi:10.1002/bjs.8992 [doi]

Gluck, N., Strul, H., Rozner, G., Leshno, M., & Santo, E. (2015). Endoscopy and EUS are key for

effective surveillance and management of duodenal adenomas in familial adenomatous

polyposis. Gastrointestinal Endoscopy, 81(4), 960-966. doi:10.1016/j.gie.2014.08.029 [doi]

Page 89: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

88

Goff, J. S. (1995). Common bile duct pre-cut sphincterotomy: Transpancreatic sphincter approach.

Gastrointestinal Endoscopy, 41(5), 502-505. doi:S0016-5107(05)80011-2 [pii]

Goff, J. S. (1999). Long-term experience with the transpancreatic sphincter pre-cut approach to

biliary sphincterotomy. Gastrointestinal Endoscopy, 50(5), 642-645. doi:S0016-

5107(99)80012-1 [pii]

Gohil, V. B., & Klapman, J. B. (2017). Endoscopic palliation of pancreatic cancer. Current

Treatment Options in Gastroenterology, doi:10.1007/s11938-017-0145-z [doi]

Gronroos, J. M., Vihervaara, H., Gullichsen, R., Laine, S., Karvonen, J., & Salminen, P. (2011).

Double-guidewire-assisted biliary cannulation: Experiences from a single tertiary referral

centre. Surgical Endoscopy, 25(5), 1599-1602. doi:10.1007/s00464-010-1458-3 [doi]

Halttunen, J., Keranen, I., Udd, M., & Kylanpaa, L. (2009). Pancreatic sphincterotomy versus

needle knife precut in difficult biliary cannulation. Surgical Endoscopy, 23(4), 745-749.

doi:10.1007/s00464-008-0056-0 [doi]

Halttunen, J., Meisner, S., Aabakken, L., Arnelo, U., Gronroos, J., Hauge, T., Lohr, J. M. (2014).

Difficult cannulation as defined by a prospective study of the Scandinavian Association for

Digestive Endoscopy (SADE) in 907 ERCPs. Scandinavian Journal of Gastroenterology,

49(6), 752-758. doi:10.3109/00365521.2014.894120 [doi]

Hamada, T., Isayama, H., Nakai, Y., Togawa, O., Kogure, H., Takahara, N., Koike, K. (2015).

Antireflux metal stent with an antimigration system for distal malignant biliary obstruction: A

feasibility pilot study. Surgical Laparoscopy, Endoscopy & Percutaneous Techniques, 25(3),

212-217. doi:10.1097/SLE.0000000000000149 [doi]

Harewood, G. C., Pochron, N. L., & Gostout, C. J. (2005). Prospective, randomized, controlled trial

of prophylactic pancreatic stent placement for endoscopic snare excision of the duodenal

ampulla. Gastrointestinal Endoscopy, 62(3), 367-370. doi:S0016-5107(05)01883-3 [pii]

Page 90: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

89

Hou, Y. C., Hu, Q., Huang, J., Fang, J. Y., & Xiong, H. (2017). Efficacy and safety of rectal

nonsteroidal anti-inflammatory drugs for prophylaxis against post-ERCP pancreatitis: A

systematic review and meta-analysis. Scientific Reports, 7, 46650. doi:10.1038/srep46650

[doi]

Hu, J., Li, P. L., Zhang, T., Chen, J. P., Hu, Y. J., Yu, Z., Tong, X. F. (2016). Role of somatostatin

in preventing post-endoscopic retrograde cholangiopancreatography (ERCP) pancreatitis: An

update meta-analysis. Frontiers in Pharmacology, 7, 489. doi:10.3389/fphar.2016.00489 [doi]

Huang, L., Yu, Q., Zhang, Q., Liu, J., & Wang, Z. (2015). Comparison between double-guidewire

technique and transpancreatic sphincterotomy technique for difficult biliary cannulation.

Digestive Endoscopy, 27(3), 381-387.

Hwang, S., Lee, S. G., Lee, Y. J., Han, D. J., Kim, S. C., Kwon, S. H., Yu, E. S. (2008). Radical

surgical resection for carcinoid tumours of the ampulla. Journal of Gastrointestinal Surgery:

Official Journal of the Society for Surgery of the Alimentary Tract, 12(4), 713-717.

doi:10.1007/s11605-007-0389-3 [doi]

Irani, S., Arai, A., Ayub, K., Biehl, T., Brandabur, J. J., Dorer, R., Kozarek, R. A. (2009).

Papillectomy for ampullary neoplasm: Results of a single referral centre over a 10-year period.

Gastrointestinal Endoscopy, 70(5), 923-932. doi:10.1016/j.gie.2009.04.015 [doi]

Isayama, H., Komatsu, Y., Inoue, Y., Toda, N., Shiratori, Y., Tsujino, T., Omata, M. (2003).

Preserved function of the Oddi sphincter after endoscopic papillary balloon dilation. Hepato-

Gastroenterology, 50(54), 1787-1791.

Ito, K., Fujita, N., Noda, Y., Kobayashi, G., Obana, T., Horaguchi, J., Yamashita, Y. (2012). Impact

of technical modification of endoscopic papillectomy for ampullary neoplasm on the

occurrence of complications. Digestive Endoscopy: Official Journal of the Japan

Page 91: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

90

Gastroenterological Endoscopy Society, 24(1), 30-35. doi:10.1111/j.1443-1661.2011.01161.x

[doi]

Ito, K., Fujita, N., Noda, Y., Kobayashi, G., Obana, T., Horaguchi, J., Ogawa, T. (2010). Can

pancreatic duct stenting prevent post-ERCP pancreatitis in patients who undergo pancreatic

duct guidewire placement for achieving selective biliary cannulation? A prospective

randomized controlled trial. Journal of Gastroenterology, 45(11), 1183-1191.

doi:10.1007/s00535-010-0268-7 [doi]

Ito, K., Horaguchi, J., Fujita, N., Noda, Y., Kobayashi, G., Koshita, S., Hashimoto, S. (2014).

Clinical usefulness of double-guidewire technique for difficult biliary cannulation in endoscopic

retrograde cholangiopancreatography. Digestive Endoscopy: Official Journal of the Japan

Gastroenterological Endoscopy Society, 26(3), 442-449. doi:10.1111/den.12158 [doi]

Jarvinen, H. J. (1992). Epidemiology of familial adenomatous polyposis in Finland: Impact of family

screening on the colorectal cancer rate and survival. Gut, 33(3), 357-360.

doi:10.1136/gut.33.3.357 [doi]

Jokelainen, J., Udd, M., Kylanpaa, L., Mustonen, H., Halttunen, J., Lindstrom, O., & Poyhia, R.

(2017). How patient-controlled sedation is adopted in clinical practice of sedation for

endoscopic retrograde cholangiopancreatography? A prospective study of 1196 cases().

Scandinavian Journal of Gastroenterology, 52(2), 166-172.

doi:10.1080/00365521.2016.1242024 [doi]

Kahaleh, M., Tokar, J., Mullick, T., Bickston, S. J., & Yeaton, P. (2004). Prospective evaluation of

pancreatic sphincterotomy as a precut technique for biliary cannulation. Clinical

Gastroenterology and Hepatology: The Official Clinical Practice Journal of the American

Gastroenterological Association, 2(11), 971-977. doi:S1542356504004847 [pii]

Page 92: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

91

Kalaitzakis, E., Webster, G. J., Oppong, K. W., Kallis, Y., Vlavianos, P., Huggett, M., Sturgess, R.

(2012). Diagnostic and therapeutic utility of single-operator peroral cholangioscopy for

indeterminate biliary lesions and bile duct stones. European Journal of Gastroenterology &

Hepatology, 24(6), 656-664. doi:10.1097/MEG.0b013e3283526fa1 [doi]

Kawakami, H., Maguchi, H., Mukai, T., Hayashi, T., Sasaki, T., Isayama, H., Japan Bile Duct

Cannulation Study Group. (2012). A multicentre, prospective, randomized study of selective

bile duct cannulation performed by multiple endoscopists: The BIDMEN study.

Gastrointestinal Endoscopy, 75(2), 362-72, 372.e1. doi:10.1016/j.gie.2011.10.012 [doi]

Kim, C. W., Chang, J. H., Kim, T. H., & Han, S. W. (2015). Sequential double-guidewire technique

and transpancreatic precut sphincterotomy for difficult biliary cannulation. Saudi Journal of

Gastroenterology, 21(1), 18-24.

Kim, J. H., Kim, J. H., Han, J. H., Yoo, B. M., Kim, M. W., & Kim, W. H. (2009). Is endoscopic

papillectomy safe for ampullary adenomas with high-grade dysplasia? Annals of Surgical

Oncology, 16(9), 2547-2554. doi:10.1245/s10434-009-0509-2 [doi]

Kim, S. J., Kang, D. H., Kim, H. W., Choi, C. W., Park, S. B., Song, B. J., & Hong, Y. M. (2015).

Needle-knife fistulotomy vs double-guidewire technique in patients with repetitive unintentional

pancreatic cannulations. World Journal of Gastroenterology, 21(19), 5918-5925.

doi:10.3748/wjg.v21.i19.5918 [doi]

Kim, S., Kim, J. W., Jung, P. Y., Kwon, H. Y., Shim, H., Jang, J. Y., & Bae, K. S. (2017). Diagnostic

and therapeutic role of endoscopic retrograde pancreatography in the management of

traumatic pancreatic duct injury patients: Single centre experience for 34 years. International

Journal of Surgery (London, England), 42, 152-157. doi:S1743-9191(17)30274-1 [pii]

Kobayashi, G., Fujita, N., Imaizumi, K., Irisawa, A., Suzuki, M., Murakami, A., Yoneyama, K.

(2013). Wire-guided biliary cannulation technique does not reduce the risk of post-ERCP

Page 93: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

92

pancreatitis: Multicentre randomized controlled trial. Digestive Endoscopy: Official Journal of

the Japan Gastroenterological Endoscopy Society, 25(3), 295-302. doi:10.1111/j.1443-

1661.2012.01372.x [doi]

Kochar, B., Akshintala, V. S., Afghani, E., Elmunzer, B. J., Kim, K. J., Lennon, A. M., Singh, V. K.

(2015). Incidence, severity, and mortality of post-ERCP pancreatitis: A systematic review by

using randomized, controlled trials. Gastrointestinal Endoscopy, 81(1), 143-149.e9.

doi:10.1016/j.gie.2014.06.045 [doi]

Korrapati, P., Ciolino, J., Wani, S., Shah, J., Watson, R., Muthusamy, V. R., Komanduri, S. (2016).

The efficacy of peroral cholangioscopy for difficult bile duct stones and indeterminate

strictures: A systematic review and meta-analysis. Endoscopy International Open, 4(3), E263-

75. doi:10.1055/s-0042-100194 [doi]

Kuo, C. M., Chiu, Y. C., Changchien, C. S., Tai, W. C., Chuah, S. K., Hu, T. H., Kuo, C. H. (2012).

Endoscopic papillary balloon dilation for removal of bile duct stones: Evaluation of outcomes

and complications in 298 patients. Journal of Clinical Gastroenterology, 46(10), 860-864.

doi:10.1097/MCG.0b013e3182617a42 [doi]

Lahoti, S., Catalano, M. F., Geenen, J. E., & Schmalz, M. J. (1998). Endoscopic retrieval of

proximally migrated biliary and pancreatic stents: Experience of a large referral centre.

Gastrointestinal Endoscopy, 47(6), 486-491. doi:S0016-5107(98)70249-4 [pii]

Larsen, M., & Kozarek, R. (2014). Management of pancreatic ductal leaks and fistulae. Journal of

Gastroenterology and Hepatology, 29(7), 1360-1370. doi:10.1111/jgh.12574 [doi]

Lee, S. M., & Cho, K. B. (2014). Value of temporary stents for the management of perivaterian

perforation during endoscopic retrograde cholangiopancreatography. World Journal of Clinical

Cases, 2(11), 689-697. doi:10.12998/wjcc.v2.i11.689 [doi]

Page 94: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

93

Lee, Y. J., Park, Y. K., Lee, M. J., Lee, K. T., Lee, K. H., & Lee, J. K. (2015). Different strategies for

transpancreatic septotomy and needle knife infundibulotomy due to the presence of

unintended pancreatic cannulation in difficult biliary cannulation. Gut and Liver, 9(4), 534-539.

doi:10.5009/gnl14223 [doi]

Lee, Y. N., Moon, J. H., Choi, H. J., Min, S. K., Kim, H. I., Lee, T. H., Kim, S. J. (2012). Direct

peroral cholangioscopy using an ultraslim upper endoscope for management of residual

stones after mechanical lithotripsy for retained common bile duct stones. Endoscopy, 44(9),

819-824. doi:10.1055/s-0032-1309880 [doi]

Lehman, G. A. (2002). Role of ERCP and other endoscopic modalities in chronic pancreatitis.

Gastrointestinal Endoscopy, 56(6 Suppl.), S237-40. doi:10.1067/mge.2002.129008 [doi]

Lella, F., Bagnolo, F., Colombo, E., & Bonassi, U. (2004). A simple way of avoiding post-ERCP

pancreatitis. Gastrointestinal Endoscopy, 59(7), 830-834. doi:S0016-5107(04)00363-3 [pii]

Liao, W. C., Lee, C. T., Chang, C. Y., Leung, J. W., Chen, J. H., Tsai, M. C., Wang, H. P. (2010).

Randomized trial of 1-minute versus 5-minute endoscopic balloon dilation for extraction of bile

duct stones. Gastrointestinal Endoscopy, 72(6), 1154-1162. doi:10.1016/j.gie.2010.07.009

[doi]

Liao, W. C., Tu, Y. K., Wu, M. S., Wang, H. P., Lin, J. T., Leung, J. W., & Chien, K. L. (2012).

Balloon dilation with adequate duration is safer than sphincterotomy for extracting bile duct

stones: A systematic review and meta-analyses. Clinical Gastroenterology and Hepatology:

The Official Clinical Practice Journal of the American Gastroenterological Association, 10(10),

1101-1109. doi:10.1016/j.cgh.2012.05.017 [doi]

Lindberg, B., Enochsson, L., Tribukait, B., Arnelo, U., & Bergquist, A. (2006). Diagnostic and

prognostic implications of DNA ploidy and S-phase evaluation in the assessment of

malignancy in biliary strictures. Endoscopy, 38(6), 561-565. doi:10.1055/s-2006-925387 [doi]

Page 95: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

94

Liu, P., Lin, H., Chen, Y., Wu, Y. S., Tang, M., & Lai, L. (2019). Comparison of endoscopic

papillary large balloon dilation with and without a prior endoscopic sphincterotomy for the

treatment of patients with large and/or multiple common bile duct stones: A systematic review

and meta-analysis. Therapeutics and Clinical Risk Management, 15, 91-101.

doi:10.2147/TCRM.S182615 [doi]

Liu, Y., Su, P., Lin, S., Xiao, K., Chen, P., An, S., Bai, Y. (2012). Endoscopic papillary balloon

dilatation versus endoscopic sphincterotomy in the treatment for choledocholithiasis: A meta-

analysis. Journal of Gastroenterology and Hepatology, 27(3), 464-471. doi:10.1111/j.1440-

1746.2011.06912.x [doi]

Loperfido, S., Angelini, G., Benedetti, G., Chilovi, F., Costan, F., De Berardinis, F., Fratton, A.

(1998). Major early complications from diagnostic and therapeutic ERCP: A prospective

multicentre study. Gastrointestinal Endoscopy, 48(1), 1-10. doi:S0016-5107(98)70121-X [pii]

Lubbe, J., Arnelo, U., Lundell, L., Swahn, F., Tornqvist, B., Jonas, E., Enochsson, L. (2015).

ERCP-guided cholangioscopy using a single-use system: Nationwide register-based study of

its use in clinical practice. Endoscopy, 47(9), 802-807. doi:10.1055/s-0034-1391990 [doi]

Luigiano, C., Iabichino, G., Mangiavillano, B., Eusebi, L. H., Arena, M., Consolo, P., Pellicano, R.

(2016). Endoscopic management of bile duct injury after hepatobiliary tract surgery: A

comprehensive review. Minerva Chirurgica, 71(6), 398-406. doi:R06Y9999N00A16090201 [pii]

Luo, H., Zhao, L., Leung, J., Zhang, R., Liu, Z., Wang, X., Fan, D. (2016). Routine pre-procedural

rectal indometacin versus selective post-procedural rectal indometacin to prevent pancreatitis

in patients undergoing endoscopic retrograde cholangiopancreatography: A multicentre,

single-blinded, randomised controlled trial. Lancet (London, England), 387(10035), 2293-

2301. doi:S0140-6736(16)30310-5 [pii]

Page 96: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

95

Machado, N. O. (2012). Management of duodenal perforation post-endoscopic retrograde

cholangiopancreatography. when and whom to operate and what factors determine the

outcome? A review article. JOP: Journal of the Pancreas, 13(1), 18-25.

Maeda, S., Hayashi, H., Hosokawa, O., Dohden, K., Hattori, M., Morita, M., Tatsumi, S. (2003).

Prospective randomized pilot trial of selective biliary cannulation using pancreatic guide-wire

placement. Endoscopy, 35(9), 721-724. doi:10.1055/s-2003-41576 [doi]

Manes, G., Paspatis, G., Aabakken, L., Anderloni, A., Arvanitakis, M., Ah-Soune, P., van Hooft, J.

E. (2019). Endoscopic management of common bile duct stones: European Society of

Gastrointestinal Endoscopy (ESGE) guideline. Endoscopy, 51(5), 472-491. doi:10.1055/a-

0862-0346 [doi]

Maple, J. T., Keswani, R. N., Edmundowicz, S. A., Jonnalagadda, S., & Azar, R. R. (2009). Wire-

assisted access sphincterotomy of the minor papilla. Gastrointestinal Endoscopy, 69(1), 47-

54. doi:10.1016/j.gie.2008.04.010 [doi]

Mariani, A., Di Leo, M., Giardullo, N., Giussani, A., Marini, M., Buffoli, F., Testoni, P. A. (2016).

Early precut sphincterotomy for difficult biliary access to reduce post-ERCP pancreatitis: A

randomized trial. Endoscopy, 48(6), 530-535. doi:10.1055/s-0042-102250 [doi]

Masci, E., Mariani, A., Curioni, S., & Testoni, P. A. (2003). Risk factors for pancreatitis following

endoscopic retrograde cholangiopancreatography: A meta-analysis. Endoscopy, 35(10), 830-

834. doi:10.1055/s-2003-42614 [doi]

Masci, E., Toti, G., Mariani, A., Curioni, S., Lomazzi, A., Dinelli, M., Testoni, P. A. (2001).

Complications of diagnostic and therapeutic ERCP: A prospective multicentre study. The

American Journal of Gastroenterology, 96(2), 417-423. doi:S000292700002387X [pii]

Page 97: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

96

Mazaki, T., Mado, K., Masuda, H., & Shiono, M. (2014). Prophylactic pancreatic stent placement

and post-ERCP pancreatitis: An updated meta-analysis. Journal of Gastroenterology, 49(2),

343-355.

Mendonca, E. Q., Bernardo, W. M., Moura, E. G., Chaves, D. M., Kondo, A., Pu, L. Z., & Baracat,

F. I. (2016). Endoscopic versus surgical treatment of ampullary adenomas: A systematic

review and meta-analysis. Clinics (Sao Paulo, Brazil), 71(1), 28-35.

doi:10.6061/clinics/2016(01)06 [doi]

Minaga, K., Kitano, M., Imai, H., Harwani, Y., Yamao, K., Kamata, K., Kudo, M. (2016). Evaluation

of anti-migration properties of biliary covered self-expandable metal stents. World Journal of

Gastroenterology, 22(30), 6917-6924. doi:10.3748/wjg.v22.i30.6917 [doi]

Moffatt, D. C., Cote, G. A., Avula, H., Watkins, J. L., McHenry, L., Sherman, S., Fogel, E. L. (2011).

Risk factors for ERCP-related complications in patients with pancreas divisum: A retrospective

study. Gastrointestinal Endoscopy, 73(5), 963-970. doi:10.1016/j.gie.2010.12.035 [doi]

Moole, H., Jaeger, A., Cashman, M., Volmar, F. H., Dhillon, S., Bechtold, M. L., & Puli, S. R.

(2017). Are self-expandable metal stents superior to plastic stents in palliating malignant distal

biliary strictures? A meta-analysis and systematic review. Medical Journal, Armed Forces

India, 73(1), 42-48. doi:10.1016/j.mjafi.2016.08.014 [doi]

Moon, J. H., Choi, H. J., & Lee, Y. N. (2014). Current status of endoscopic papillectomy for

ampullary tumours. Gut and Liver, 8(6), 598-604. doi:10.5009/gnl14099 [doi]

Morelli, J., Mulcahy, H. E., Willner, I. R., Cunningham, J. T., & Draganov, P. (2003). Long-term

outcomes for patients with post-liver transplant anastomotic biliary strictures treated by

endoscopic stent placement. Gastrointestinal Endoscopy, 58(3), 374-379. doi:S0016-

5107(03)00011-7 [pii]

Page 98: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

97

Napoleon, B., Alvarez-Sanchez, M. V., Leclercq, P., Mion, F., Pialat, J., Gincul, R., Scoazec, J. Y.

(2013). Systematic pancreatic stenting after endoscopic snare papillectomy may reduce the

risk of postinterventional pancreatitis. Surgical Endoscopy, 27(9), 3377-3387.

doi:10.1007/s00464-013-2920-9 [doi]

Navaneethan, U., Hasan, M. K., Lourdusamy, V., Njei, B., Varadarajulu, S., & Hawes, R. H. (2015).

Single-operator cholangioscopy and targeted biopsies in the diagnosis of indeterminate biliary

strictures: A systematic review. Gastrointestinal Endoscopy, 82(4), 608-14.e2.

doi:10.1016/j.gie.2015.04.030 [doi]

Navaneethan, U., Jegadeesan, R., Nayak, S., Lourdusamy, V., Sanaka, M. R., Vargo, J. J., &

Parsi, M. A. (2015). ERCP-related adverse events in patients with primary sclerosing

cholangitis. Gastrointestinal Endoscopy, 81(2), 410-419. doi:10.1016/j.gie.2014.06.030 [doi]

Navaneethan, U., Konjeti, R., Venkatesh, P. G., Sanaka, M. R., & Parsi, M. A. (2014). Early precut

sphincterotomy and the risk of endoscopic retrograde cholangiopancreatography related

complications: An updated meta-analysis. World Journal of Gastrointestinal Endoscopy, 6(5),

200-208. doi:10.4253/wjge.v6.i5.200 [doi]

Navaneethan, U., Njei, B., Lourdusamy, V., Konjeti, R., Vargo, J. J., & Parsi, M. A. (2015).

Comparative effectiveness of biliary brush cytology and intraductal biopsy for detection of

malignant biliary strictures: A systematic review and meta-analysis. Gastrointestinal

Endoscopy, 81(1), 168-176. doi:10.1016/j.gie.2014.09.017 [doi]

Odemis, B., Oztas, E., Kuzu, U. B., Parlak, E., Disibeyaz, S., Torun, S., & Kayacetin, E. (2016).

Can a fully covered self-expandable metallic stent be used temporarily for the management of

duodenal retroperitoneal perforation during ERCP as a part of conservative therapy? Surgical

Laparoscopy, Endoscopy & Percutaneous Techniques, 26(1), e9-e17.

doi:10.1097/SLE.0000000000000240 [doi]

Page 99: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

98

Olsson, G., Lubbe, J., Arnelo, U., Jonas, E., Tornqvist, B., Lundell, L., & Enochsson, L. (2017). The

impact of prophylactic pancreatic stenting on post-ERCP pancreatitis: A nationwide, register-

based study. United European Gastroenterology Journal, 5(1), 111-118.

doi:10.1177/2050640616645434 [doi]

O'Neill, F., Carter, E., Pink, N., & Smith, I. (2016). Routine preoperative tests for elective surgery:

Summary of updated NICE guidance. BMJ (Clinical Research Ed.), 354, i3292.

doi:10.1136/bmj.i3292 [doi]

Oppong, K. W., Romagnuolo, J., & Cotton, P. B. (2012). The ERCP quality network benchmarking

project: A preliminary comparison of practice in UK and USA. Frontline Gastroenterology,

3(3), 157-161. doi:10.1136/flgastro-2011-100099 [doi]

Paik, W. H., Park, Y. S., Hwang, J. H., Lee, S. H., Yoon, C. J., Kang, S. G., Yoon, Y. B. (2009).

Palliative treatment with self-expandable metallic stents in patients with advanced type III or IV

hilar cholangiocarcinoma: A percutaneous versus endoscopic approach. Gastrointestinal

Endoscopy, 69(1), 55-62. doi:10.1016/j.gie.2008.04.005 [doi]

Parc, Y., Mabrut, J. Y., Shields, C., & Mallorca Group. (2011). Surgical management of the

duodenal manifestations of familial adenomatous polyposis. The British Journal of Surgery,

98(4), 480-484. doi:10.1002/bjs.7374 [doi]

Park, C. H., Paik, W. H., Park, E. T., Shim, C. S., Lee, T. Y., Kang, C., Lee, J. K. (2018).

Aggressive intravenous hydration with lactated ringer's solution for prevention of post-ERCP

pancreatitis: A prospective randomized multicentre clinical trial. Endoscopy, 50(4), 378-385.

doi:10.1055/s-0043-122386 [doi]

Park, D. H., Lee, S. S., Lee, T. H., Ryu, C. H., Kim, H. J., Seo, D. W., Kim, S. J. (2011). Anchoring

flap versus flared end, fully covered self-expandable metal stents to prevent migration in

Page 100: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

99

patients with benign biliary strictures: A multicentre, prospective, comparative pilot study (with

videos). Gastrointestinal Endoscopy, 73(1), 64-70. doi:10.1016/j.gie.2010.09.039 [doi]

Park, S. H., de Bellis, M., McHenry, L., Fogel, E. L., Lazzell, L., Bucksot, L., Lehman, G. A. (2003).

Use of methylene blue to identify the minor papilla or its orifice in patients with pancreas

divisum. Gastrointestinal Endoscopy, 57(3), 358-363. doi:10.1067/mge.2003.110 [doi]

Park, S. W., Chung, M. J., Oh, T. G., Park, J. Y., Bang, S., Park, S. W., & Song, S. Y. (2015).

Intramuscular diclofenac for the prevention of post-ERCP pancreatitis: A randomized trial.

Endoscopy, 47(1), 33-39. doi:10.1055/s-0034-1390743 [doi]

Park, S. W., Song, S. Y., Chung, J. B., Lee, S. K., Moon, Y. M., Kang, J. K., & Park, I. S. (2000).

Endoscopic snare resection for tumours of the ampulla of Vater. Yonsei Medical Journal,

41(2), 213-218. doi:10.3349/ymj.2000.41.2.213 [doi]

Park, W. Y., Cho, K. B., Kim, E. S., & Park, K. S. (2012). A case of ampullary perforation treated

with a temporally covered metal stent. Clinical Endoscopy, 45(2), 177-180.

doi:10.5946/ce.2012.45.2.177 [doi]

Parsi, M. A. (2014). Direct peroral cholangioscopy. World Journal of Gastrointestinal Endoscopy,

6(1), 1-5. doi:10.4253/wjge.v6.i1.1 [doi]

Paspatis, G. A., Dumonceau, J. M., Barthet, M., Meisner, S., Repici, A., Saunders, B. P., Hassan,

C. (2014). Diagnosis and management of iatrogenic endoscopic perforations: European

Society of Gastrointestinal Endoscopy (ESGE) position statement. Endoscopy, 46(8), 693-

711. doi:10.1055/s-0034-1377531 [doi]

Pasternak, L. R. (2004). Preoperative laboratory testing: General issues and considerations.

Anesthesiology Clinics of North America, 22(1), 13-25. doi:10.1016/S0889-8537(03)00115-9

[doi]

Page 101: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

100

Pecsi, D., Farkas, N., Hegyi, P., Balasko, M., Czimmer, J., Garami, A., Vincze, A. (2017).

Transpancreatic sphincterotomy has a higher cannulation success rate than needle-knife

precut papillotomy - a meta-analysis. Endoscopy, 49(9), 874-887.

Phillip, V., Pukitis, A., Epstein, A., Hapfelmeier, A., Haf, D., Schwab, M., Algul, H. (2019).

Pancreatic stenting to prevent post-ERCP pancreatitis: A randomized multicentre trial.

Endoscopy International Open, 7(7), E860-E868. doi:10.1055/a-0886-6384 [doi]

Polydorou, A., Vezakis, A., Fragulidis, G., Katsarelias, D., Vagianos, C., & Polymeneas, G. (2011).

A tailored approach to the management of perforations following endoscopic retrograde

cholangiopancreatography and sphincterotomy. Journal of Gastrointestinal Surgery: Official

Journal of the Society for Surgery of the Alimentary Tract, 15(12), 2211-2217.

doi:10.1007/s11605-011-1723-3 [doi]

Ponsioen, C. Y., Vrouenraets, S. M., Prawirodirdjo, W., Rajaram, R., Rauws, E. A., Mulder, C. J.,

Tytgat, G. N. (2002). Natural history of primary sclerosing cholangitis and prognostic value of

cholangiography in a Dutch population. Gut, 51(4), 562-566.

Rahman, M. R., Perisetti, A., Coman, R., Bansal, P., Chhabra, R., & Goyal, H. (2019).

Duodenoscope-associated infections: Update on an emerging problem. Digestive Diseases

and Sciences, 64(6), 1409-1418. doi:10.1007/s10620-018-5431-7 [doi]

Rainio, M., Lindstrom, O., Udd, M., Haapamaki, C., Nordin, A., & Kylanpaa, L. (2018). Endoscopic

therapy of biliary injury after cholecystectomy. Digestive Diseases and Sciences, 63(2), 474-

480. doi:10.1007/s10620-017-4768-7 [doi]

Rainio, M., Lindstrom, O., Udd, M., Louhimo, J., & Kylanpaa, L. (2017). Diclofenac does not reduce

the risk of post-endoscopic retrograde cholangiopancreatography pancreatitis in low-risk units.

Journal of Gastrointestinal Surgery: Official Journal of the Society for Surgery of the

Alimentary Tract, 21(8), 1270-1277. doi:10.1007/s11605-017-3412-3 [doi]

Page 102: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

101

Rehman, A., Affronti, J., & Rao, S. (2013). Sphincter of Oddi dysfunction: An evidence-based

review. Expert Review of Gastroenterology & Hepatology, 7(8), 713-722.

doi:10.1586/17474124.2013.849197 [doi]

Rerknimitr, R., Fogel, E. L., Kalayci, C., Esber, E., Lehman, G. A., & Sherman, S. (2002).

Microbiology of bile in patients with cholangitis or cholestasis with and without plastic biliary

endoprosthesis. Gastrointestinal Endoscopy, 56(6), 885-889. doi:10.1067/mge.2002.129604

[doi]

Rossos, P. G., Kortan, P., & Haber, G. (1993). Selective common bile duct cannulation can be

simplified by the use of a standard papillotome. Gastrointestinal Endoscopy, 39(1), 67-69.

doi:S0016-5107(93)70015-2 [pii]

Sawas, T., Al Halabi, S., Parsi, M. A., & Vargo, J. J. (2015). Self-expandable metal stents versus

plastic stents for malignant biliary obstruction: A meta-analysis. Gastrointestinal Endoscopy,

82(2), 256-267.e7. doi:10.1016/j.gie.2015.03.1980 [doi]

Seifert, E., Schulte, F., & Stolte, M. (1992). Adenoma and carcinoma of the duodenum and papilla

of Vater: A clinicopathologic study. The American Journal of Gastroenterology, 87(1), 37-42.

Seta, T., & Noguchi, Y. (2011). Protease inhibitors for preventing complications associated with

ERCP: An updated meta-analysis. Gastrointestinal Endoscopy, 73(4), 700-706.e1.

doi:10.1016/j.gie.2010.09.022 [doi]

Seza, K., Yamaguchi, T., Ishihara, T., Tadenema, H., Tawada, K., Saisho, H., & Yokosuka, O.

(2011). A long-term controlled trial of endoscopic pancreatic stenting for treatment of main

pancreatic duct stricture in chronic pancreatitis. Hepato-Gastroenterology, 58(112), 2128-

2131. doi:10.5754/hge09346 [doi]

Shah, J. N., Ahmad, N. A., Shetty, K., Kochman, M. L., Long, W. B., Brensinger, C. M., Ginsberg,

G. G. (2004). Endoscopic management of biliary complications after adult living donor liver

Page 103: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

102

transplantation. The American Journal of Gastroenterology, 99(7), 1291-1295.

doi:10.1111/j.1572-0241.2004.30775.x [doi]

Shao, L. M., Chen, Q. Y., Chen, M. Y., & Cai, J. T. (2009). Can wire-guided cannulation reduce the

risk of post-endoscopic retrograde cholangiopancreatography pancreatitis? A meta-analysis of

randomized controlled trials. Journal of Gastroenterology and Hepatology, 24(11), 1710-1715.

doi:10.1111/j.1440-1746.2009.06010.x [doi]

Shapiro, A. M., Scudamore, C. H., July, L. V., Buczkowski, A. K., Chung, S. W., Gul, S., &

Patterson, E. J. (1999). Calcific intra-pancreatic embedding of a pancreatic stent necessitating

surgical removal--a danger of chronic endoscopic retrograde pancreatic stent placement.

Gastrointestinal Endoscopy, 50(6), 860-862. doi:S0016-5107(99)70178-1 [pii]

Sherman, S., & Lehman, G. A. (2001). Sphincter of Oddi dysfunction: Diagnosis and treatment.

JOP: Journal of the Pancreas, 2(6), 382-400. doi:v02i06a05 [pii]

Shimatani, M., Matsushita, M., Takaoka, M., Koyabu, M., Ikeura, T., Kato, K., Okazaki, K. (2009).

Effective "short" double-balloon enteroscope for diagnostic and therapeutic ERCP in patients

with altered gastrointestinal anatomy: A large case series. Endoscopy, 41(10), 849-854.

doi:10.1055/s-0029-1215108 [doi]

Siegel, J. H., Ben-Zvi, J. S., & Pullano, W. (1989). The needle knife: A valuable tool in diagnostic

and therapeutic ERCP. Gastrointestinal Endoscopy, 35(6), 499-503. doi:S0016-

5107(89)72897-2 [pii]

Singhirunnusorn, J., Roger, L., Chopin-Laly, X., Lepilliez, V., Ponchon, T., & Adham, M. (2013).

Value of preoperative biliary drainage in a consecutive series of resectable periampullary

lesions. from randomized studies to real medical practice. Langenbeck's Archives of Surgery,

398(2), 295-302. doi:10.1007/s00423-012-1000-2 [doi]

Page 104: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

103

Stapfer, M., Selby, R. R., Stain, S. C., Katkhouda, N., Parekh, D., Jabbour, N., & Garry, D. (2000).

Management of duodenal perforation after endoscopic retrograde cholangiopancreatography

and sphincterotomy. Annals of Surgery, 232(2), 191-198.

Swahn, F., Nilsson, M., Arnelo, U., Lohr, M., Persson, G., & Enochsson, L. (2013). Rendezvous

cannulation technique reduces post-ERCP pancreatitis: A prospective nationwide study of

12,718 ERCP procedures. The American Journal of Gastroenterology, 108(4), 552-559.

doi:10.1038/ajg.2012.470 [doi]

Tandan, M., Talukdar, R., & Reddy, D. N. (2016). Management of pancreatic calculi: An update.

Gut and Liver, 10(6), 873-880. doi:10.5009/gnl15555 [doi]

Testoni, P. A. (2007). Endoscopic pancreatic duct stent placement for inflammatory pancreatic

diseases. World Journal of Gastroenterology, 13(45), 5971-5978.

doi:10.3748/wjg.v13.45.5971 [doi]

Testoni, P. A., Mariani, A., Giussani, A., Vailati, C., Masci, E., Macarri, G., SEIFRED Group.

(2010). Risk factors for post-ERCP pancreatitis in high- and low-volume centres and among

expert and non-expert operators: A prospective multicentre study. The American Journal of

Gastroenterology, 105(8), 1753-1761. doi:10.1038/ajg.2010.136 [doi]

Testoni, P. A., Mariani, A., Aabakken, L., Arvanitakis, M., Bories, E., Costamagna, G., Williams, E.

J. (2016). Papillary cannulation and sphincterotomy techniques at ERCP: European Society of

Gastrointestinal Endoscopy (ESGE) clinical guideline. Endoscopy, 48(7), 657-683.

Tsai, T. J., Lin, C. K., Lai, K. H., Chan, H. H., Wang, E. M., Tsai, W. L., Hsu, P. I. (2018). Does

preserved sphincter of Oddi function prevent common bile duct stones recurrence in patients

after endoscopic papillary balloon dilation? Journal of the Chinese Medical Association:

JCMA, 81(4), 311-315. doi:S1726-4901(18)30027-3 [pii]

Page 105: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

104

Tse, F., & Yuan, Y. (2012). Early routine endoscopic retrograde cholangiopancreatography

strategy versus early conservative management strategy in acute gallstone pancreatitis. The

Cochrane Database of Systematic Reviews, (5):CD009779. doi(5), CD009779.

doi:10.1002/14651858.CD009779.pub2 [doi]

Tse, F., Yuan, Y., Moayyedi, P., & Leontiadis, G. I. (2013). Guide wire-assisted cannulation for the

prevention of post-ERCP pancreatitis: A systematic review and meta-analysis. Endoscopy,

45(8), 605-618.

Tse, F., Yuan, Y., Moayyedi, P., Leontiadis, G. I., & Barkun, A. N. (2017). Double-guidewire

technique in difficult biliary cannulation for the prevention of post-ERCP pancreatitis: A

systematic review and meta-analysis. Endoscopy, 49(1), 15-26. doi:10.1055/s-0042-119035

[doi]

Tyberg, A., Karia, K., Gabr, M., Desai, A., Doshi, R., Gaidhane, M., Kahaleh, M. (2016).

Management of pancreatic fluid collections: A comprehensive review of the literature. World

Journal of Gastroenterology, 22(7), 2256-2270. doi:10.3748/wjg.v22.i7.2256 [doi]

Udd, M., Kylanpaa, L., & Halttunen, J. (2010). Management of difficult bile duct cannulation in

ERCP. World Journal of Gastrointestinal Endoscopy, 2(3), 97-103. doi:10.4253/wjge.v2.i3.97

[doi]

van Berkel, A. M., Huibregtse, I. L., Bergman, J. J., Rauws, E. A., Bruno, M. J., & Huibregtse, K.

(2004). A prospective randomized trial of Tannenbaum-type Teflon-coated stents versus

polyethylene stents for distal malignant biliary obstruction. European Journal of

Gastroenterology & Hepatology, 16(2), 213-217. doi:00042737-200402000-00015 [pii]

Vandervoort, J., Soetikno, R. M., Tham, T. C., Wong, R. C., Ferrari, A. P., Montes, H., Carr-Locke,

D. L. (2002). Risk factors for complications after performance of ERCP. Gastrointestinal

Endoscopy, 56(5), 652-656. doi:10.1067/mge.2002.129086 [doi]

Page 106: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

105

Vannas, M. J., Boyd, S., Farkkila, M. A., Arola, J., & Isoniemi, H. (2017). Value of brush cytology

for optimal timing of liver transplantation in primary sclerosing cholangitis. Liver International:

Official Journal of the International Association for the Study of the Liver, 37(5), 735-742.

doi:10.1111/liv.13276 [doi]

Verdonk, R. C., Buis, C. I., Porte, R. J., van der Jagt, E J, Limburg, A. J., van den Berg, A P,

Haagsma, E. B. (2006). Anastomotic biliary strictures after liver transplantation: Causes and

consequences. Liver Transplantation: Official Publication of the American Association for the

Study of Liver Diseases and the International Liver Transplantation Society, 12(5), 726-735.

doi:10.1002/lt.20714 [doi]

Verma, D., Kapadia, A., & Adler, D. G. (2007). Pure versus mixed electrosurgical current for

endoscopic biliary sphincterotomy: A meta-analysis of adverse outcomes. Gastrointestinal

Endoscopy, 66(2), 283-290. doi:S0016-5107(07)00032-6 [pii]

Vezakis, A., Fragulidis, G., & Polydorou, A. (2015). Endoscopic retrograde

cholangiopancreatography-related perforations: Diagnosis and management. World Journal of

Gastrointestinal Endoscopy, 7(14), 1135-1141. doi:10.4253/wjge.v7.i14.1135 [doi]

Wang, B., Guo, Z., Liu, Z., Wang, Y., Si, Y., Zhu, Y., & Jin, M. (2013). Preoperative versus

intraoperative endoscopic sphincterotomy in patients with gallbladder and suspected common

bile duct stones: System review and meta-analysis. Surgical Endoscopy, 27(7), 2454-2465.

doi:10.1007/s00464-012-2757-7 [doi]

Wang, P., Li, Z. S., Liu, F., Ren, X., Lu, N. H., Fan, Z. N., Zhi, F. C. (2009). Risk factors for ERCP-

related complications: A prospective multicentre study. The American Journal of

Gastroenterology, 104(1), 31-40. doi:10.1038/ajg.2008.5 [doi]

Wang, X., Dai, C., Jiang, Z., Zhao, L., Wang, M., Ma, L., Fan, Z. (2017). Endoscopic retrograde

cholangiopancreatography versus laparoscopic exploration for common bile duct stones in

Page 107: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

106

post-cholecystectomy patients: A retrospective study. Oncotarget, 8(47), 82114-82122.

doi:10.18632/oncotarget.18839 [doi]

Wang, Y., Qi, M., Hao, Y., & Hong, J. (2019). The efficacy of prophylactic pancreatic stents against

complications of post-endoscopic papillectomy or endoscopic ampullectomy: A systematic

review and meta-analysis. Therapeutic Advances in Gastroenterology, 12,

1756284819855342. doi:10.1177/1756284819855342 [doi]

Weber, A., Roesch, T., Pointner, S., Born, P., Neu, B., Meining, A., Prinz, C. (2008).

Transpancreatic precut sphincterotomy for cannulation of inaccessible common bile duct: A

safe and successful technique. Pancreas, 36(2), 187-191.

doi:10.1097/MPA.0b013e31815ac54c [doi]

Weinberg, B. M., Shindy, W., & Lo, S. (2006). Endoscopic balloon sphincter dilation

(sphincteroplasty) versus sphincterotomy for common bile duct stones. The Cochrane

Database of Systematic Reviews, (4)(4), CD004890. doi:10.1002/14651858.CD004890.pub2

[doi]

Wu, D., Wan, J., Xia, L., Chen, J., Zhu, Y., & Lu, N. (2017). The efficiency of aggressive hydration

with lactated ringer solution for the prevention of post-ERCP pancreatitis: A systematic review

and meta-analysis. Journal of Clinical Gastroenterology, 51(8), e68-e76.

doi:10.1097/MCG.0000000000000856 [doi]

Yang, K. Y., Ryu, J. K., Seo, J. K., Woo, S. M., Park, J. K., Kim, Y. T., & Yoon, Y. B. (2009). A

comparison of the Niti-D biliary uncovered stent and the uncovered Wallstent in malignant

biliary obstruction. Gastrointestinal Endoscopy, 70(1), 45-51. doi:10.1016/j.gie.2008.10.029

[doi]

Page 108: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

107

Yoo, Y. W., Cha, S. W., Lee, W. C., Kim, S. H., Kim, A., & Cho, Y. D. (2013). Double guidewire

technique vs transpancreatic precut sphincterotomy in difficult biliary cannulation. World

Journal of Gastroenterology: WJG, 19(1), 108-114. doi:10.3748/wjg.v19.i1.108 [doi]

Yoshioka, T., Shigekawa, M., Yamai, T., Suda, T., Kegasawa, T., Iwahashi, K., Takehara, T.

(2016). The safety and benefit of pancreatic juice cytology under ERCP in IPMN patients.

Pancreatology: Official Journal of the International Association of Pancreatology (IAP).[Et Al.],

16(6), 1020-1027. doi:S1424-3903(16)31177-2 [pii]

Zang, Jinfeng, Z., Chi, G., & Junye. (2014). Guidewire-assisted transpancreatic sphincterotomy for

difficult biliary cannulation: A prospective randomized controlled trial. Surgical Laparoscopy,

Endoscopy & Percutaneous Techniques, 24(5), 429-433.

Zhang, Y., Chen, Q. B., Gao, Z. Y., & Xie, W. F. (2009). Meta-analysis: Octreotide prevents post-

ERCP pancreatitis, but only at sufficient doses. Alimentary Pharmacology & Therapeutics,

29(11), 1155-1164. doi:10.1111/j.1365-2036.2009.03991.x [doi]

Zhang, Z. F., Duan, Z. J., Wang, L. X., Zhao, G., & Deng, W. G. (2017). Aggressive hydration with

lactated ringer solution in prevention of postendoscopic retrograde cholangiopancreatography

pancreatitis: A meta-analysis of randomized controlled trials. Journal of Clinical

Gastroenterology, 51(3), e17-e26. doi:10.1097/MCG.0000000000000781 [doi]

Zhang, Z. F., Yang, N., Zhao, G., Zhu, L., Zhu, Y., & Wang, L. X. (2010). Preventive effect of

ulinastatin and gabexate mesylate on post-endoscopic retrograde cholangiopancreatography

pancreatitis. Chinese Medical Journal, 123(18), 2600-2606.

Zhao, H. C., He, L., Zhou, D. C., Geng, X. P., & Pan, F. M. (2013). Meta-analysis comparison of

endoscopic papillary balloon dilatation and endoscopic sphincteropapillotomy. World Journal of

Gastroenterology, 19(24), 3883-3891. doi:10.3748/wjg.v19.i24.3883 [doi]

Page 109: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE
Page 110: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

I

Page 111: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE
Page 112: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

Risk factors for complications of ERCP in primarysclerosing cholangitis

Authors S. Ismail1, L. Kylänpää1, H. Mustonen1, J. Halttunen1, O. Lindström1, K. Jokelainen2, M. Udd1, M. Färkkilä2,3

Institutions 1 Department of Gastrointestinal and General Surgery, Helsinki University Central Hospital, Helsinki, Finland2 Department of Gastroenterology, Helsinki University Central Hospital, Helsinki, Finland3 Helsinki University, Institute of Clinical Medicine, Department of Medicine, Clinic of Gastroenterology, Helsinki, Finland

submitted14. December 2011accepted after revision18. June 2012

BibliographyDOI http://dx.doi.org/10.1055/s-0032-1325677Published online: 29.10.2012Endoscopy 2012; 44: 1133–1138© Georg Thieme Verlag KGStuttgart · New YorkISSN 0013-726X

Corresponding authorL. Kylänpää, MD, PhDDepartment of Gastrointestinaland General SurgeryHelsinki University CentralHospitalHaartmaninkatu 4PL34000029 [email protected]

Original article 1133

Introduction!

Primary sclerosing cholangitis (PSC) is a chronicprogressive disease of the biliary tract character-ized by diffuse inflammation and fibrosis andstrictures of both intrahepatic and extrahepaticbile ducts [1, 2]. The disease is slowly progressiveand may eventually lead to cirrhosis, but the mostfeared complication is cholangiocarcinoma. Non-invasive diagnostic methods such as magneticresonance cholangiography (MRC) have largelysuperseded diagnostic endoscopic retrogradecholangiopancreatography (ERCP) for many indi-cations. However, diagnosis of PSC in its earlyphase, especially when intrahepatic, is impossiblewith even the best MRC. ERCP done with the bal-loon occlusion technique using adequate contrastfilling is still the gold standard for diagnosing PSC,and is the only option for collecting cytologicalmaterial from bile ducts for dysplasia screeningand surveillance. Early diagnosis of PSC is essen-

tial, as it has great impact on both the surveillanceof the liver disease and the associated colitis. Atpresent, detection of biliary dysplasia with ERCPis the only way of diagnosing cholangiocarcinomain its early, curable stage. Using DNA flow cyto-metry combined with brush cytology with Papa-nicolaou stain, we have previously shown in con-secutive patients with PSC that an aneuploid DNAcontent was found in 19% of patients and cellssuspected of malignancy in 21% during dysplasiasurveillance in PSC [3]. The brush cytology andbiopsies obtained during ERCP in combinationwith advanced cytology techniques, such as fluor-escence in situ hybridization (FISH), are the mostsensitivemethods for early detection of the devel-opment of cholangiocarcinoma [4]. At our unit,repeated ERCP procedures are used for the dila-tion of dominant strictures and for dysplasia sur-veillance of the biliary tract in patients with PSC.The reported incidence of complications afterERCP in patients with PSC ranges from 4% to

Ismail S et al. Risk factors for complications of ERCP in primary sclerosing cholangitis… Endoscopy 2012; 44: 1133–1138

Background and study aims: Endoscopic retro-grade cholangiographic pancreatography (ERCP)is the most accurate technique for surveillance ofpatients with primary sclerosing cholangitis(PSC). Our aim was to evaluate risk factors forcomplications of ERCP in patients with PSC.Patients and methods: In 2007–2009 we per-formed 441 ERCPs in patients with PSC. The pri-mary tools for ERCP were a guide wire and papil-lotomy knife to gain access into the biliary duct. Ifthe primary cannulation failed, and the wire wentonly into the pancreatic duct, pancreatic sphinc-terotomy was performed. If necessary, a furtheroblique cut with a needle knife was done in orderto expose the biliary duct.Results: Primary cannulation was successful in389 patients (88.2%). Of these, 147 (37.8%) hadhad biliary sphincterotomy performed previous-ly. In the group with failed primary cannulation,access into the biliary duct was achieved after

pancreatic sphincterotomy in 52 patients. In 11of these, a further cut with a needle knife was per-formed. Post-ERCP pancreatitis (PEP) was diag-nosed in 31 patients (7.0%). Factors predictingPEP were female sex (odds ratio [OR] 2.6, P=0.015) and a guide wire in the pancreatic duct(OR 8.2, P<0.01). Previous biliary sphincterotomywas a protective factor (OR 0.28, P=0.02). The riskof PEP increased with the number of times thewire accidentally passed into the pancreatic duct(P<0.001). Cholangitis developed in 6 patients(1.4%).Conclusions: In patients with PSC the incidence ofERCP complications remained relatively low. Thecomplication risk increased with the complexityof cannulation. In a patient with PSC in whom fol-low-up ERCP is planned, biliary sphincterotomyshould be considered, as it may protect againstPEP.

Page 113: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

15.9% [5–9], post-ERCP pancreatitis (PEP) being the most fre-quent and dreaded complication. Its prevalence ranges from 1%to 7% [4], average 3.47% (CI 3.19%–3.75%), in a large systematicsurvey of prospective studies involving 16 855 patients [7].Known patient-related risk factors for PEP include younger age,female sex, suspected sphincter of Oddi dysfunction (SOD), andprevious PEP. Difficult cannulation, needle knife precut, frequentcontrast injections into the pancreatic duct, and pancreaticsphincterotomy are the most commonly reported procedure-related factors. Although in most cases PEP is mild, 20% of pa-tients with PEP suffer from a moderate or severe form of the dis-ease, with prolonged hospitalization and even PEP-related organfailure and death. The other ERCP-related complications are cho-langitis, bleeding, and retroperitoneal duodenal perforation,usually following therapeutic procedures. Understanding whatfactors increase the complication rates could help to reducethem to the minimum. Patients with PSC undergo repeatedERCP examinations with multiple therapeutic interventions dur-ing the procedures, such as brushing, biopsies, and dilations, andso form a special group of patients at high risk of complications ofERCP. The aim of the present study was to identify patient- andprocedure-related risk factors for complications of ERCP in pa-tients with PSC.

Patients and methods!

In the years 2007–2009, 3019 ERCPs were performed in HelsinkiUniversity Central Hospital. In 443 of these (14.7%) the indicationfor ERCP was PSC. The patient records were retrospectively re-viewed. In two patients (0.5%) biliary cannulation had failed. Nei-ther of them developed PEP and the biliary cannulation succeed-ed in the second ERCP session. For these patients, the data of thesecond, successful cannulation are included in the analysis. Thepatient group was made up of 441 consecutive patients withPSC. PSC imaging findings were scored according to Ponsioen etal. [10]. All the procedures were performed by an expert (M.F.),who performs all ERCPs for patients with PSC at Helsinki Univer-sity Hospital. All patients received oral levofloxacillin 500mg as aprophylactic antibiotic, within 1 hour prior to ERCP.The primary tools for ERCP were a papillotomy knife (JagtomeRX; Boston Scientific, Miami, Florida, USA) and a 0.035-in, 450-cm guide wire (Jagwire; Boston Scientific) to gain access into thebiliary duct. If the primary cannulation failed, and the guide wireslipped only into the pancreatic duct, pancreatic sphincterotomywas performed with the papillotomy knife over the guide wire. Ifnecessary, a further oblique cut with a needle knife towards 10o’clock starting from the upper end of the pancreatic sphincterot-omy cut was performed in order to gain biliary access. After suc-cessful biliary cannulation a sphincterotomy was performed alsoin that direction. The presence of the guide wire in the pancreaticduct was assessed by the position in fluoroscopy; no contrast wasinjected into the pancreatic duct for the purpose. After cannula-tion of the biliary tract with the papillotomy knife and guidewire, a balloon catheter was inserted into the common hepaticduct and the intrahepatic bile ducts were filled with contrastusing the balloon occlusion technique to ensure adequate visua-lization of the bile ducts. In all ERCP procedures brush cytologyspecimens were collected from intrahepatic and extrahepaticbile ducts using between one and four brushes. In addition, dila-tion of dominant hilar and extrahepatic strictures was performedwhen indicated: i. e., in the presence of pruritus, cholestasis, sig-

nificant prestenotic dilatation, or inability to pass the cytologybrush.Plasma total amylase or pancreatic amylase was measured beforeand 4–6 hours after ERCP. If the patient stayed overnight in thehospital, plasma amylase was checked on the following morning.If a complication occurred, the patient stayed in the hospital untilhe or she recovered.Themain aimwas to evaluate the frequency of complications, i. e.,PEP, cholangitis, bleeding, and perforation. PEP was defined asplasma amylase elevated to three times the upper reference limitor more, and the presence of abdominal pain persisting 24 hoursafter the procedure. Cholangitis was defined as fever requiringintravenous or intramuscular antibiotics occurring within 2days after ERCP. Bleeding was defined as a need for repeat endos-copy due to melena or blood transfusion within 1 week afterERCP. Perforation was diagnosed as an extravasation of contrastduring ERCP or retroperitoneal air on computed tomographyafter the procedure. The severity of the complications was de-fined according to consensus criteria [11].Five patient-related risk factors were recorded: sex, age, historyof previous PEP, radiological scoring of PSC, and previously per-formed biliary sphincterotomy. Six procedure-related risk factorswere included: the number of accidental passes of the guide wireinto the pancreatic duct, pancreatic sphincterotomy, biliarysphincterotomy, precut with a needle knife, balloon dilation ofthe biliary tract, and the duration of ERCP.Data are presented as the median (range) or number of patientsand percentages. Odds ratios are presented with 95%Wald confi-dence intervals (CI). The data were analyzed using SPSS v. 15.0(IBM Corporation, Somers, New York USA) and by Statexact v.4.0 (Cytel Software Corporation, Cambridge, Massachusetts,USA). Fisher’s exact test was used to test for differences betweencategorical variables. Logistic regression was used to calculateodds ratios and P values (Wald χ2) for nominal and ordinal vari-ables. Multivariate logistic regressionwas used to obtain adjustedodds ratios. Age and sex were included in the model as well asrisk factors with a univariate P value of less than 0.1.The risk fac-tors in●" Table1 and contrast agent in the pancreatic duct wereconsidered as risk factors for multivariate analysis. Interactionterms were considered. The Cochran–Armitage trend test wasused to test for an association between the incidence of PEP andan ordered categorical variable. The nonparametric Wilcoxon–Mann–Whitney test was used to compare differences in contin-uous and ordinal variables. Probabilities were regarded as statis-tically significant when they were below the 0.05 level.

Results!

Themedian age of the patients was 38 years (range, 5–80); 46.0%were females. Themedian lengthof hospital staywas 1day (range,0–28) after ERCP. Primary cannulationwas successful in 389/441patients (88.2%). Of these, 147 (37.8%) had undergone a prior bili-ary sphincterotomy. In patients with a naïve papilla, the primarycannulation rate was 82.3%, whereas in those with a previoussphincterotomy it was 100% (P<0.001). In the group with failedcannulation, access to the biliary duct was achieved in 52 cases byfirst performingpancreatic sphincterotomy.Of these, a further cutwith a needle knifewas performed in 11 cases.PEP was seen in 31 patients (7.0%). It was graded as mild in 18,moderate in 10, and severe in 3.One patient with severe PEPwas treated in the intensive care unit for 3 days. No case of necro-

Ismail S et al. Risk factors for complications of ERCP in primary sclerosing cholangitis… Endoscopy 2012; 44: 1133–1138

Original article1134

Page 114: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

tizing pancreatitis was diagnosed. All the patients with PEP weretreated conservatively. The median length of hospital stay for PEPpatients was 4 days (range, 2–28 days). Evaluated risk factors forPEP are presented in●" Table1. Factors predicting PEP were fe-male sex (OR 2.6, P=0.015) and accidental passage of the guidewire into the pancreatic duct (OR 8.2, P<0.01). In cases wherethe pancreatic duct remained untouched, the incidence of PEPwas 2.6%. If the wire was twice passed accidentally into the pan-creatic duct, the incidencewas 20%, and if five times it was 31.6%(●" Fig.1; P<0.001 for a trend). Contrast agent was seen in thepancreatic duct in 15 patients. Two of them developed PEP, whichwas not a statistically significant figure (P=0.342).The PEP rate for patients with naïve papillas was 9.2%, whereasfor those with previous sphincterotomy it was 2.7% (P=0.01).Previously performed biliary sphincterotomy was a protectivefactor against PEP (OR 0.28, P=0.02). The risk for PEP increasedas more complicated techniques had to be used for successful ac-cess to the biliary tract. If only cannulation was performed, with-out sphincterotomy, the risk of PEP was 1.4%. If biliary sphincter-otomy was performed alone, the risk for PEP was 6.8%. Whenpancreatic sphincterotomy had to be performed in order to gainaccess into the biliary tract, the PEP ratewas 25%, andwith an ad-ditional biliary sphincterotomy the rate was 27%. In the caseswhere cannulation was most difficult (pancreatic sphincterot-omy, precut, and biliary sphincterotomy, n=9), the rate of PEP

Table 1 Risk factors and incidence of post-ERCP pancreatitis in 441 patients undergoing ERCP for primary sclerosing cholangitis.

Variable No.of PEP cases/No.of patients (%) Odds ratio 95% confidence interval P

SexFemaleMale

21 /203 (10.3)10 /238 (4.2)

2.63 1.21–5.73 0.015

Age, years> 50> 30–50≤30

7 /115 (6.1)15 /197 (7.6)9 /129 (7.0)

0.94 0.57–1.53 0.799

Previous PEPYesNo

3 /23 (13.0)28 /418 (6.7)

2.09 0.59–7.46 0.256

Previous biliary sphincterotomyYesNo

4 /147 (2.7)27 /294 (9.2)

0.28 0.10–0.81 0.010

Wire accidentally in pancreatic ductYesNo

23 /129 (17.8)8 /312 (2.6)

8.25 3.58–18.99 < 0.001

Bile duct balloon dilationYesNo

4 /80 (5.0)27 /361 (7.5)

0.65 0.22–1.92 0.436

Duration of ERCP, minutes> 30> 20–30≤20

13 /138 (9.4)14 /185 (7.6)3 /85 (3.5)

1.54 0.90–2.64 0.120

Radiological PSC score3210

4 /89 (4.5 )3 /77 (3.9)1 /44 (2.3)

22 /227 (9.7)

0.70 0.49–1.00 0.053

TechniquePrecut + biliary and pancreatic sphinc.Biliary and pancreatic sphinc.Pancreatic sphinc.Biliary sphinc.Cannulation

5 /9 (55.6)10 /37 (27.0)1 /4 (25.0)

12 /176 (6.8)3 /213 (1.4)

87.525.923.35.1

15.4–498.56.7–100.11.9–294.11.4–18.4

< 0.001< 0.0010.0150.012

ERCP, endoscopic retrograde cholangiopancreatography; PEP, post-ERCP pancreatitis; sphinc., sphincterotomy.For odds ratios, the last category is used as a reference category.

50

45

40

35

30

25

20

15

10

5

0

Post

ERC

P pa

ncre

atiti

s rat

e (%

)

N = 312

0

N = 33

2–3No of accidental passes of guide wire in pancreatic duct

N = 30

4–6

N = 43

1

Fig.1 The risk of pancreatitis after endoscopic retrograde cholangiopan-creatography increases as the number of times the wire accidentally passesinto the pancreatic duct increases (P<0.001, Cochran–Armitage trendtest; data are presented with exact 95% confidence intervals). N, totalnumber of patients in each group.

Ismail S et al. Risk factors for complications of ERCP in primary sclerosing cholangitis… Endoscopy 2012; 44: 1133–1138

Original article 1135

Page 115: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

rose as high as 55.6%. Intrahepatic, extrahepatic, and total PSCscores were slightly lower in patients who developed PEP thanin others (●" Table2). The extrahepatic score was lower in pa-tients in whom performance of a pancreatic sphincterotomywas necessary (0, 0-6) than in other patients (1, 0-6, P<0,001).Patient age and history of PEP, dilation of biliary tract, and dura-tion of ERCP each had no influence on the development of PEP(●" Table1). In multivariate analysis the risk for PEP increasedwith the complication of technique (●" Table3). Another factorpredicting PEP was female sex. Patients in whom biliary sphinc-terotomy had previously been performed always had easy cannu-lations (85% with cannulation only and the rest with biliarysphincterotomy), and so their risk of PEP was not different fromthat of the reference group (reference group: male, age≤30 years,cannulation, no wire in pancreas, radiological score=0).There were no cases of post-sphincterotomy bleeding. Six pa-tients (1.4%) suffered from cholangitis after ERCP. One of themhad mild and the others moderate disease. Patients who devel-oped cholangitis did not have a longer duration of ERCP (median,40 min; range, 18–50min) than the rest of the patients (median,27 min; range, 10–96 min; P=0.096). Three of the six patientswith cholangitis had had a previous biliary sphincterotomy. Cho-langitis developed in 2% of patients with sphincterotomy and in1% of those without (P=0.405). So, there is no evidence that pre-vious biliary sphincterotomy increases the risk of ascending cho-langitis.There were three perforations (0.6%), one due to guide wire per-foration and two related to the sphincterotomy. None of themwas life threatening, and the patients were all successfully treat-ed conservatively. The three patients with a perforation stayed inthe hospital for 5, 6, and 7 days respectively. There were no pro-cedure-related deaths.

Discussion!

ERCP performed with the balloon occlusion technique remainsthe most sensitive and specific method for primary diagnosisand follow-up of the progression of PSC. Diagnosis of small-ductPSC cannot be done without high-quality ERCP. Moreover, for de-tection and surveillance of biliary dysplasia and dilation of domi-nant strictures, ERC is an irreplaceable diagnostic and therapeu-tic tool. However, the risks related to the procedure must alwaysbe weighed up against the benefits for the patient. The compli-cation rate associated with ERCP has been reported to rangefrom 4% to 15.9% in patients without PSC, and up to 14% in pa-tients with PSC [12–14]. Some studies comparing complicationsin PSC and non-PSC patients show that neither the overall com-plication rate nor the risks of PEP, perforation, and bleeding differbetween the two groups [8, 12]. In the authors’ unit, the PEP ratein naïve papillas is 4.8% in indications other than PSC, whichtends to be somewhat lower than in patients with PSC with a na-ïve papilla (9.2%). In a recent study, the incidence of post-ERCPcholangitis was reported to be higher in patients with PSC thanin the control group of non-PSC patients [8]. The study also foundthe duration of ERCP to be longer in those patients who devel-oped cholangitis. One explanatory factor for the differences be-tween the reported complication rates is the number of interven-tions carried out during the ERCP procedure. Cholangitis was nota common complication in the present study and no associationwas found between a prolonged procedure time and the develop-ment of cholangitis, neither did previous biliary sphincterotomyincrease the risk of ERCP-associated cholangitis. These findingsmay partly be explained by the prophylactic antibiotic regularlyused in this study.In the present study, we included a large cohort of patients withPSC who underwent ERCP for primary diagnosis, therapeuticpurposes, or surveillance of disease progression and biliary dys-plasia. In our unit, a diagnosis of PSC is always confirmed by ERCPand liver biopsy. All patients who are potential candidates for liv-

Table 2 Scoring of primary scle-rosing cholangitis (PSC) and inci-dence of post-ERCP pancreatitis in441 patients.

1

Intrahepatic score Extrahepatic score Total score N

Median Range Median Range Median Range

Min. Max. Min. Max. Min. Max.

No PEP 3 0 6 1 0 6 4 0 11 410PEP 2 0 5 0 0 6 2 0 10 31P 0.022 0.030 0.005

ERCP, endoscopic retrograde cholangiopancreatography; PEP, post-ERCP pancreatitis.1 Scoring according to Ponsioen et al. [10].

Table 3 Multivariate analysis ofrisk factors of post-ERCP pancrea-titis in 441 patients undergoingERCP for primary sclerosing cho-langitis.

Variable Odds ratio 95% confidence interval P

Sex, female/male 2.4 1.0–5.8 0.046Age, years, > 50 / > 30–50 / ≤30 0.9 0.5–1.6 0.726TechniquePrecut + biliary and pancreatic sphinc.Biliary and pancreatic sphinc.Pancreatic sphinc.Biliary sphinc.Cannulation (reference category)

46.313.112.94.7

5.0–430.01.9–90.30.8–221.51.05–21.15

0.0010.0090.0780.046

No. of times wire in pancreas, 5–0 (0 reference category) 1.2 0.97–1.61 0.088Previous biliary sphincterotomy, yes/no 1.5 0.4–5.9 0.561Radiological score PSC, 3 /2 /1 /0 0.85 0.58–1.26 0.417

ERCP, endoscopic retrograde cholangiopancreatography; sphinc., sphincterotomy.For odds ratios, the last category is used as a reference category. No significant interactions were found.

Ismail S et al. Risk factors for complications of ERCP in primary sclerosing cholangitis… Endoscopy 2012; 44: 1133–1138

Original article1136

Page 116: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

er transplantation undergo regular screening and surveillance forbiliary dysplasia with repeated ERCPs and multiple brushings ofintrahepatic and extrahepatic bile ducts. With this strategy, ana-lyzing DNA ploidy and Papanicolaou stain from brush cytology,we have been able to demonstrate a sensitivity of 72% and a spe-cificity of 82%, with a positive predictive value of 86% and a neg-ative predictive value of 67% for biliary malignancy [3].However, multiple interventions in bile ducts have been shownto increase the complication rate [8]. Since most patients areasymptomatic when they come to ERCP, safety and technical suc-cess must be optimized. In the present study, the success rate ofbiliary cannulation was high.

Complication rate and mortalityThe overall complication rate of 9% after ERCP is a little less thanin some previously published reports of patients with PSC [8, 12],which may be due to the guide wire cannulation technique weuse. On the other hand, the patients underwent multiple proce-dures during the ERCP, including balloon catheter insertion andseveral brush instruments and dilations, which are known to in-crease the overall risk of complications. All the complicationscaused prolonged hospitalization, but it was possible to treatthem conservatively. There were no procedure-related deaths.In one study with a large number of ERCP patients, the overallmortality rate was 0.4%, and half of the deceased were over 70years of age and another half had serious co-morbidities [6].Most patients with PSC are young and have no serious coexistingmedical condition, which explains the absence of mortality.Probably for the same reason, we could not find any associationbetween age and risk of PEP, although there are studies reportingthat young age is associated with a high PEP rate [6,15,16].

PancreatitisPEP occurred in 31 cases, in three of which it was graded as se-vere. Unlike in some other studies [17,18], in the present studya history of PEP was not a risk factor for new PEP. The PEP rate infemale patients was more than twice that in male patients. Fe-male sex has been related to increased risk of post-ERCP compli-cations [11,16,19–21]. However, there are many studies whichfind no association between sex and increased complications[5, 8,15,18,22]. Accidental wire insertion into the pancreaticduct was a risk factor for PEP, and the risk increased with multi-ple passes. Previous papillotomy was a protective factor for PEP;this can be explained by the fact that cannulation is technicallyeasier and also that accidental slipping of the wire into the pan-creatic duct is less probable. These results relating to the techni-cal part of the procedure suggest that mechanical irritation of thepapilla has a significant effect on the risk of PEP.Many studies show that contrast agent in the pancreatic duct in-creases the risk of PEP [5,15,17,18,23]. In the technique of pan-creatic sphincterotomy, the position of the wire in the pancreaticduct is usually clearly distinguishable without contrast injectionunder fluoroscopy. However, in our previous study, the presenceof contrast agent in the pancreatic duct was not a risk factor forPEP [9]. In the study of Lella et al. [24], accidental insertion ofthe guide wire into the main pancreatic duct a median of twotimes happened in 82/200 patients (41%) without any adverseeffect. The difference in these results may be due to differencesin PEP diagnosis: an amylase level five times the upper referencelimit was needed to diagnose PEP in their study compared withthree times the upper reference limit in the present study. Still,

in the study of Artifon et al. [23], accidental guide wire insertioninto the pancreatic duct did not cause PEP.Precutting is a valuable tool which increases the success rate oftherapeutic ERCP to over 90–95% [25,26]. In many studies, pre-cutting has been related to an increased complication rate [6,18,22]. Freeman et al. reported that precutting increased the risk ofPEP [6], while Loperfido et al. showed it to increase the risk ofduodenal perforation [22]. There are, however, other studiesshowing that this technique is nomore harmful than convention-al sphincterotomy [8,26–29]. In our data set precutting was per-formed only if cannulation was unsuccessful after pancreaticsphincterotomy. So, in nearly all cases where precut was used,the wire had slipped into the pancreatic duct many times, andalso pancreatic and biliary sphincterotomy were performed,forming the most difficult cannulation cases of the series– thusexplaining the high PEP rate. In high-risk patients, prophylacticplacement of a pancreatic stent decreases the risk of PEP [30]. Asin our series there was a high PEP rate in patients with the mostdifficult cannulations, placement of a pancreatic stent could bebeneficial in such cases.We have previously compared pancreaticsphincterotomywith the precut technique in difficult biliary can-nulations [9]. In our experience, pancreatic sphincterotomy issafe and results in a high cannulation success rate. Catalano et al.also found the complication rate to be lower with pancreaticsphincterotomy than with the precut technique [31]. In our pre-vious study [9], the need for the additional precut after pan-creatic sphincterotomy was 42%, while in the present study itwas 21%. Cannulation technique may vary according to individ-ual endoscopists. The dissimilarity may also be due to the factthat in the present study the patient group consisted only ofthose with PSC. However, the extrahepatic PSC score was nohigher in patients requiring pancreatic sphincterotomy than inthose who did not. Thus, a stricture of the extrahepatic bile ductdoes not completely explain the difficulty of biliary access.Difficult cannulation increased the risk of PEP [6,17,19]. Biliarysphincterotomy has also been associated with an increased riskof PEP; in one large prospective study the odds ratio (OR) for PEPwas 1.21 (range, 0.91–1.60) [6]. In our study, biliary sphincterot-omy was associated with a 6.8% risk of PEP, but in consecutiveERCP procedures a previously performed biliary sphincterotomywas a significantly protective factor (OR 0.28, P=0.01). For thisreason, we recommend that biliary sphincterotomy be per-formed for patients with PSC in whom long-term follow-up withERCP is planned.In conclusion, PSC has previously been shown to increase the riskof cholangiocarcinoma by up to 161 times. As the development ofcholangiocarcinoma is unpredictable on the basis of clinicalsymptoms, ERCP, or magnetic resonance cholangiographic find-ings, ERCP with brush cytology is an indispensable diagnosticand therapeutic tool for detecting biliary dysplasia in patientswith PSC with fairly low risk of severe complications and no mor-tality. We recommend endoscopic sphincterotomy to preventPEP in patients with PSC undergoing dysplasia surveillance withrepeated ERCPs. Endoscopic sphincterotomy is not associatedwith an increased risk of ERCP-associated cholangitis.

Competing interests: None

Ismail S et al. Risk factors for complications of ERCP in primary sclerosing cholangitis… Endoscopy 2012; 44: 1133–1138

Original article 1137

Page 117: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

References1 Chapman R, Fevery J, Kalloo A et al. Diagnosis and management of pri-

mary sclerosing cholangitis. Hepatology 2010; 51: 660–6782 Rasmussen HH, Fallingborg JF, Mortensen PB et al. Hepatobiliary dys-

function and primary sclerosing cholangitis in patients with Crohn’sdisease. Scand J Gastroenterol 1997; 32: 604–610

3 Halme L, Arola J, Numminen K et al. Biliary dysplasia in patients withprimary sclerosing cholangitis: additional value of DNA ploidity. LiverInt 2012; 32: 783–789

4 Moreno Luna LE, Gores GJ. Advances in the diagnosis of cholangio-car-cinoma in patients with primary sclerosing cholangitis. Liver Transpl2006; 12 : 02 15–19

5 Cotton PB, Garrow DA, Gallagher J et al. Risk factors for complicationsafter ERCP: a multivariate analysis of 11,497 procedures over 12 years.Gastrointest Endosc 2009; 70: 80–88

6 Freeman ML, Nelson DB, Sherman S et al. Complications of endoscopicbiliary sphincterotomy. N Engl J Med 1996; 335: 909–918

7 Andriulli A, Loperfido S, Napolitano G et al. Incidence rates of post-ERCPcomplications: a systematic survey of prospective studies. Am J Gas-troenterol 2007; 102: 1781–1788

8 Bangarulingam SY, Gossard AA, Petersen BT et al. Complications of en-doscopic retrograde cholangiopancreatography in primary sclerosingcholangitis. Am J Gastroenterol 2009; 104: 855–860

9 Halttunen J, Keränen I, Udd M et al. Pancreatic sphincterotomy versusneedle knife precut in difficult biliary cannulation. Surg Endosc 2009;23: 745–749

10 Ponsioen C, Vrouenraets S, Prawirodirdjo W et al. Natural history of pri-mary sclerosing cholangitis and prognostic value of cholangiographyin a Dutch population. Gut 2002; 51: 562–566

11 Cotton PB, Lehman G, Vennes J et al. Endoscopic sphincterotomy com-plications and their management: an attempt at consensus. Gastroin-test Endosc 1991; 37: 383–393

12 Etzel JP, Eng SC, Ko CW et al. Complications of ERCP in patients with pri-mary sclerosing cholangitis. Gastrointest Endosc 2008; 67: 643–648

13 Lee JG, Schutz SM, England RE et al. Endoscopic therapy of sclerosingcholangitis. Hepatology 1995; 21: 661–667

14 Ponsioen CY, Lam K, vanMilligen deWit AW et al. Four years’ experiencewith short term stenting in primary sclerosing cholangitis. Am J Gas-troenterol 1999; 94: 2403–2407

15 Cheng CL, Sherman S, Watkins JL et al. Risk factors for post-ERCP pan-creatitis: a prospective multicenter study. Am J Gastroenterol 2006;101: 139–147

16 Wang P, Li ZS, Liu F et al. Risk factors for ERCP-related complications: aprospective multicenter study. Am J Gastroenterol 2009; 104: 31–40

17 Vandervoort J, Soetikno RM, Tham TC et al. Risk factors for complica-tions after performance of ERCP. Gastrointest Endosc 2002; 56: 652–656

18 Testoni PA, Mariani A, Giussani A et al. Risk factors for post-ERCP pan-creatitis in high- and low-volume centers and among expert and non-expert operators: a prospectivemulticenter study. Am J Gastro-enterol2010; 105: 1753–1761

19 Freeman ML, DiSario JA, Nelson DB et al. Risk factors for post-ERCP pan-creatitis: a prospective, multicenter study. Gastrointest Endosc 2001;54: 425–434

20 Masci E, Toti G, Mariani A et al. Complications of diagnostic and thera-peutic ERCP: a prospective multicenter study. Am J Gastroenterol2001; 96: 417–423

21 Lee TH, Park do H, Park JY et al. Can wire-guided cannulation preventpost-ERCP pancreatitis? A prospective randomized trial GastrointestEndosc 2009; 69: 444–449

22 Loperfido S, Angelini G, Benedetti G et al. Major early complicationsfrom diagnostic and therapeutic ERCP: a prospective multicenterstudy. Gastrointest Endosc 1998; 48: 1–10

23 Artifon EL, Sakai P, Cunha JE et al. Guidewire cannulation reduces riskof post-ERCP pancreatitis and facilitates bile duct cannulation. Am JGastroenterol 2007; 102: 2147–2153

24 Lella F, Bagnolo F, Colombo E et al. A simple way of avoiding post-ERCPpancreatitis. Gastrointest Endosc 2004; 59: 830–834

25 Siegel JH, Ben-Zvi JS, Pullano W. The needle-knife: a valuable tool in di-agnostic and therapeutic ERCP. Gastrointest Endosc 1989; 35: 499–503

26 Bruins Slot W, Schoeman MN, Disario JA. Needle-knife sphincterotomyas a precut procedure: a retrospective evaluation of efficacy and com-plications. Endoscopy 1996; 28: 334–339

27 Foutch PG. A prospective assessment of results for needle knife papil-lotomy and standard endoscopic sphincterotomy. Gastrointest Endosc1995; 41: 25–32

28 Rabenstein T, Ruppert T, Muehldorfer S et al. The precut technique is asafe procedure and does not increase the overall complication risk ofsphincterotomy [abstract]. Gastrointest Endosc 1995; 41: 412

29 Binmoeller KF, Seifert H, Gerke H et al. Papillary roof incision using theErlangen-type precut papillotome to achieve bile duct cannulation.Gastrointest Endosc 1996; 44: 689–695

30 Choudhary A, Bechtold ML, Arif M et al. Pancreatic stent for prophylaxisagainst post-ERCP pancreatitis: A meta-analysis and systematic re-view. Gastrointest Endosc 2011; 73: 275–282

31 CatalanoMF, Linder JD, Chak A et al. Endoscopicmanagement of adeno-ma of the major duodenal papilla. Gastrointest Endosc 2004; 59: 225–232

Ismail S et al. Risk factors for complications of ERCP in primary sclerosing cholangitis… Endoscopy 2012; 44: 1133–1138

Original article1138

Page 118: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

II

Page 119: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE
Page 120: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

Endoscopic papillectomy, single-centre experience

Shamel Ismail • Udd Marianne • Jarvinen Heikki •

Halttunen Jorma • Kylanpaa Leena

Received: 3 January 2014 / Accepted: 29 April 2014! Springer Science+Business Media New York 2014

AbstractBackground Endoscopic removal of benign tumours of

papilla is increasing. Our aim was to evaluate the outcome

of endoscopic resection of papillary tumours.Methods In the years 2000–2012, 61 papillectomies were

performed in Helsinki University Central Hospital. The

cases were analysed retrospectively.Results There were 35 patients with benign tumour of

papilla without familial adenomatous polyposis (FAP), 16

patients with FAP and 10 patients with ampullary cancer.Jaundice and bile duct dilation were risk factors for malig-

nancy (p \ 0.001). In benign tumours, the recurrence rate was

25.5 %. In 5/51 benign tumour cases (9.8 %), a pancreatico-duodenectomy was performed. The remaining cases were

treated endoscopically. Neither tumour size, resection in one

piece or piecemeal technique, nor coagulation of resectionmargins had an effect on the development of residual tumour.

The total complication rate was 24.6 %. Pancreatitis devel-

oped in six patients (9.8 %, 3 mild and 3 moderate). In benigntumour cases, pancreatic stent decreased pancreatitis rate

(p = 0.045). In cases where only a pancreatic sphincterotomywas performed, the risk of pancreatitis was high 4/7 (57 %).

Bleeding was the most common complication (18 %). Only

one patient was operated due to complication, a post-papil-lectomy bleeding. In six out of seven non-operated cancer

patients, the disease progressed.

Conclusion Endoscopic papillectomy is an effectiveprocedure for treating benign papillary tumours. Jaundice

and bile duct dilation are more common in malignant

tumours. Pancreatic stent decreases the risk of post-papil-lectomy pancreatitis. Pancreatic sphincterotomy without

stenting carries a high risk of pancreatitis. For papillary

cancer, surgery is recommended.

Keywords Ampulla of Vater ! Papillectomy ! Adenoma !Carcinoma ! Familial adenomatous polyposis

The most common benign lesions in the ampulla of Vater

are villous and tubulovillous adenomas [1, 2]. Benign

ampullary adenomas have premalignant potential [3].These adenomas may occur sporadically or in the setting of

familial adenomatous polyposis (FAP). Autopsy series

have estimated the prevalence of ampullary adenoma to bebetween 0.04 and 0.12 % [4, 5]. In FAP, duodenal ade-

nomas occur in nearly all patients with time, the most

commonly at the area of the papilla [6, 7]. With theincreasing use of endoscopy, especially endoscopic retro-

grade cholangiopancreatography (ERCP), tumours of the

papilla are being progressively more recognised.The treatment of the adenomas of the major duodenal

papilla can be either surgical (pancreaticoduodenectomy or

local excision) or endoscopic resection. To eliminate the riskof histologic progression and to exclude foci of carcinoma

within the adenoma, pancreaticoduodenectomy remains the

only choice of treatment, but is associated with significantmorbidity and mortality. In pancreaticoduodenectomy, a

perioperative mortality rate of between 2 and 5 % and a sig-

nificant rate of postoperative complications (up to 41 %) havebeen reported [8, 9]. Local excision has lower mortality and

morbidity rates, but is associated with the risk of recurrence

[10, 11]. Currently, endoscopic papillectomy with curativeintent is increasingly performed for benign papillary tumours.

A systematic review estimated that success rates for

S. Ismail ! U. Marianne ! J. Heikki ! H. Jorma ! K. Leena (&)Department of Gastrointestinal and General Surgery, HelsinkiUniversity Central Hospital, P.O. Box 340, 00029 HUS Helsinki,Finlande-mail: [email protected]

123

Surg Endosc

DOI 10.1007/s00464-014-3596-5

and Other Interventional Techniques

Page 121: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

endoscopic removal of ampullary adenomas range from 46 to

92 %, while recurrence rates range from 0 to 33 % [12].This study evaluates the safety of endoscopic resection

of papilla in benign and malignant tumours, and patient

outcomes under surveillance.

Patients and methods

In the years 2000–2012, a total of 61 papillectomies for

tumours of the papilla of Vater were performed at HelsinkiUniversity Central Hospital, in the Department of Gastro-

intestinal and General Surgery. All the procedures were

performed by the two experts who perform over 200ERCPs annually. All the patients received 500 mg of oral

levofloxacillin as a prophylactic antibiotic within 1 h prior

to the ERCP. The patient material consisted of 35 patientswith a benign papilla tumour without FAP, 16 patients with

FAP and 10 patients with ampullary cancer. No formal

Research Ethics Board approval was applied due to theretrospective nature of the study. However, institutional

research permission was granted for the study protocol.

Papillectomy

Papillectomy was performed under fluoroscopic guidanceand with a side-viewing duodenoscope. The patient was in

a prone position. An anaesthesiologist sedated and moni-tored the patient throughout the procedure.

The pancreatic duct was identified and cannulated with a

sphincterotome and a guide wire. Using the sphinctero-tome, a minimal dose of methylene blue was injected into

the pancreatic duct to mark the orifice and end of the duct.

The tumour was grasped with a polypectomy snare andresected using blended electrosurgical current (50–60 J).

En bloc resection was attempted, and piecemeal resection

was performed when necessary. After the papillectomy, theaim was to insert a 5–7 Fr, 3–7 cm pancreatic stent. A

pathologist examined the resected specimen.

Follow-up

Control examinations using a side-viewing endoscope after1, 3 and 6 months were planned. Complications were divi-

ded into pancreatitis, bleeding, perforation and cholangitis.

The severity of complications was graded by the consensusdefinition by Cotton et al. [13]. Data about the patients were

collected retrospectively from the patient files.

Statistical analysis

Statistical analysis was performed using IBM SPSS Sta-tistics (version 19.0, Chicago, Ilinois, USA).The results are

presented as median and range. Comparison of the results

between groups was made using the Mann–Whitney U testand Pearson’s v2 test, as appropriate. A p value below 0.05

was considered significant.

Results

Patient characteristics are presented in Table 1. Lesion size

was determined by pathologic reports. The median age ofthe cancer patients was higher (77 years, 67–91) than the

age the patients with benign lesions (58 years, 28–79,

p \ 0.001). The ampullary cancer patients had jaundicemore often than the patients with benign tumours (90 vs.

14 %, p \ 0.001). Similarly, bile duct dilation was more

frequent in the cancer patients (100 vs. 27 %, p \ 0.001).The median hospital length of stay was 1 day (0–45).

Benign papillary tumours: no FAP

Of benign papillary adenomas in patients without FAP, 18

of 35 (51 %) were found incidentally in gastroscopy(n = 14), computed tomography (CT, n = 2) or magnetic

resonance imaging (MRI, n = 2). Seven of the patients

(20 %) had jaundice as a symptom. Six patients underwentinvestigations due to bleeding episodes or anaemia. Four

patients had ERCP due to acute pancreatitis with unknown

aetiology. The concordance between pre-resection biopsyand the resected specimen was 24 of 32 (75 %, Table 1).

Endoscopic ultrasound (EUS) was performed on 12

patients before papillectomy. In ERCP, contrast was nec-essary in only 10 patients, while the position of the wire in

pancreatic duct could be confirmed in fluoroscopy. During

the endoscopy, methylene blue was inserted into the pan-creatic duct in 6 out of 35 patients. Pancreatic stenting was

attempted for 17 patients. For 10 patients, stenting was not

attempted because pancreatic endoscopic papillotomy(EPT) was considered sufficient. Thus, pancreatic stenting

was successful in 16 of 17 patients (94 %). EPT for the bile

duct was performed for 23 patients (66 %). After thepapillectomy, resection margins were coagulated in 23

patients in order to prevent bleeding.

Post-papillectomy complications developed in ninepatients (25.7 %, Table 1). Post-ERCP pancreatitis devel-

oped for four patients (11 %, 3 mild cases, 1 moderate). Six

patients had bleeding and were treated endoscopically.There were two retroperitoneal perforations which were

treated conservatively. One patient with a large adenoma

had all the three complications.The follow-up time was 14 months (range 1–91). Ten

patients (29 %) had residual adenomas. Four of them

received pancreaticoduodenectomy, which revealed anadenoma with high-grade dysplasia in two patients and a

Surg Endosc

123

Page 122: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

carcinoma in one patient. One patient had a carcinoma

in situ in follow-up biopsies, but in the pancreaticoduo-denectomy specimen there was no neoplasm. The rest of

the residual cases could be treated endoscopically.

FAP

All papillary adenomas in FAP patients were found inroutine endoscopic follow-up (Table 1). The concordance

between pre-resection biopsy and the resected specimenwas 14/16 (88 %). EUS was not planned before papillec-

tomy. During the procedure, contrast was necessary in only

four patients. Methylene blue was inserted into the pan-creatic duct in 12 of 16 patients (75 %). Pancreatic stenting

was attempted for 14 of 16 patients. For one patient, pan-

creatic EPT only was considered sufficient. Thus, pancre-atic stenting was successful in 12 of 14 patients (86 %).

One of them had the stent placed in the minor papilla. A

bile duct EPT was done for 13 of 16 patients (81 %,Table 1).

Four patients (25 %) developed complications. Post-

ERCP pancreatitis with moderate severity developed intwo patients. Three patients (19 %) had a bleeding com-

plication which could be treated endoscopically. The fol-

low-up time is 37 months (range 1–63). Three patients(19 %) had residual adenomas. In two of them they were

minor lesions, which could be treated with biopsy forceps

and coagulation. A pancreaticoduodenectomy was per-formed on the third patient, which revealed an adenoma

with high-grade dysplasia in the specimen. One of the FAP

patients had a pancreaticoduodenectomy 4 years after thepapillectomy because of multiple duodenal adenomas

which were distant from the papilla (1 high grade and 3 low

grade).

Ampullary cancer

Of the patients, nine out of ten had jaundice as the primary

symptom. One patient had a gastroscopy because of a

bleeding episode. The pre-resection biopsies revealedcancer in only one patient. In four patients, the result was

an adenoma with high-grade dysplasia, and two patients

had low-grade dysplasia (Table 1). EUS was performed forfour patients before the papillectomy. In three of them, the

tumour was considered to be resectable endoscopically. In

one patient, the tumour infiltrated through the duodenalwall but the biopsies revealed only adenoma with high-

grade dysplasia and not cancer. CT or MRI scans were

performed on all the patients before papillectomy. Bileduct dilation was present in all the cases and dilation of the

pancreatic duct was ascertained in six of them. During the

Table 1 Characteristics of the patients and the procedure

Benign tumour,no FAP

FAP Cancer

n = 35 n = 16 n = 10

Male/female 24/11 9/7 5/5

ASA

I 7 0 0

II 10 10 1

III 17 6 7

IV 1 0 2

Concomitant diseases

Coronary heart disease 8 0 3

Pulmonary disease 8 1 3

Diabetes 6 0 0

Other 31 0 9

Indications of ERCP

Incidental finding/surveillance 18 16 0

Jaundice 7 0 9

GI bleeding or anaemia 6 0 1

Pancreatitis 4 0 0

Abdominal pain 0 0 0

The size of tumour

\1 cm 1 0 0

1–2 cm 23 15 5

2.1–4 cm 7 1 5

[4 cm 2 0 0

Unknown 2 0 0

Pre-resection histologic diagnosis

Low-grade dysplasia 24 13 2

High-grade dysplasia 8 3 4

Adenocarcinoma 0 0 1

Other 0 0 0

Post-resection histologic diagnosis

Low-grade dysplasia 18 13 0

High-grade dysplasia 14 3 0

Adenocarcinoma 0 0 10

Othera 3 0 0

Procedure

Biliary sphincterotomy 23 13 8

Pancreatic sphincterotomy 19 8 4

Pancreatic stenting 16 12 4

Coagulation of margins 23 15 9

Complications

Pancreatitis 4 2 0

Mild 3 0

Moderate 1 2

Bleeding 6 3 2

Extraperitoneal perforation 2 0 0

a One amyloidosis, one paraganglioma, one inflammatory changes

Surg Endosc

123

Page 123: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

procedure, methylene blue was inserted into the pancreatic

duct in two patients. Pancreatic stenting was not performedfor two patients because of wide pancreatic EPT. Pancre-

atic stenting was successful in four of the five cases in

which it was attempted (85 %). Biliary sphincterotomy wasperformed for eight patients.

There were no cases of post-papillectomy pancreatitis or

perforation. Two patients (20 %) developed bleeding as acomplication, despite coagulation during the papillectomy.

One was treated endoscopically, but the other needed alaparotomy and duodenotomy.

Three patients ended up undergoing a pancreaticoduo-

denectomy. In all of them, a residual cancer (4 mm, 1 cm,3 cm) was found in the resected specimen. The patient with

the 3 cm tumour also had 6/16 lymph node metastasis. The

patient with the 4 mm residual cancer had undergone acontrol duodenoscopy before the operation, which did not

reveal any tumour at the papillectomy site. The two

patients without lymph node metastasis, who were operatedon, have now been followed up for 6 and 8.5 years,

respectively, without any signs of malignancy. The patient

with lymph node metastasis died 14 months after theoperation due to cancer. Two out of 7 non-operated

patients needed endoscopic stenting due to biliary

obstruction. Six of the 7 non-operated patients died9–28 months after papillectomy. Three of them died due to

ampullary cancer and one due to pneumonia. The patient

who is still alive has now been followed up for 5 months.

Procedure and complications

In total, 15 patients out of 61 (24.6 %) had complications.

In benign tumours there were 13 patients out of 51

(25.5 %) with residual adenomas in follow-up. The numberwas the same regardless of whether the papillectomy was

performed en bloc or in piecemeal. The size or grade of the

adenoma did not have an effect on the development ofresidual adenoma, neither did coagulation of the resection

margins decrease the risk of residual adenomas (p = ns).

Post-papillectomy pancreatitis developed in 6 of 61patients (9.8 %). Contrast was inserted into the pancreatic

duct in only 18 patients (30 %), but the pancreatitis risk did

not increase for them (n = 1). Among patients with benigntumour, the patients with a prophylactic pancreatic stent

(n = 28) had only one case of pancreatitis. The patients

without a stent (n = 23) had five pancreatitis cases(p = 0.045). In cases where only pancreatic EPT was

performed without inserting a stent (n = 7), the rate of

pancreatitis was high (n = 4, 57 %, p = 0.002). Resectionmargins were coagulated in 47 patients (77 %), but this did

not prevent the risk of bleeding, since 10 out of 11 bleeding

complications happened in these cases.

Discussion

Cancer

Jaundice as a symptom and bile duct dilation are clear riskfactors of malignancy. Still, in the present series, in the

benign tumour cases, 14 % of patients presented jaundice

as a symptom. The only cases that survived the cancer werethose who were operated on and had no lymph node

metastasis. In cancer patients, especially, it is hard to

estimate complete endoscopic removal, which was under-lined in the case with a clean post-papillectomy finding in

the duodenoscopy but a 4 mm cancer in the pancreatico-

duodenectomy specimen. Of course, the most commonreason not to operate is that the patient is unfit for surgery.

Pre-resection biopsy has been shown to be unreliable in

many cases [14]. Similarly, in the present series, biopsiesrevealed cancer in only one patient and the papillectomy

specimen was needed for diagnosis. Pancreaticoduoden-

ectomy is the treatment of choice for papillary cancer.Many experts consider even the adenomas with high-grade

dysplasia to be an indication for surgery [10, 15, 16]. As

was shown here, even a thorough follow-up may miss asmall cancer. By radical surgery the outcome of the patient

is favourable. If the cancer diagnosis is made before pap-

illectomy, palliative biliary stenting would probably be anappropriate procedure for patients unfit for surgery. In

bleeding tumours, papillectomy could still be helpful.

FAP

In FAP patients, duodenal adenomas are common and they

often occur in the papilla [17]. Surveillance seems to be

beneficial, since none of the FAP patients in the presentseries developed cancer. In addition, the concordance

between pre-resection biopsies and papillectomy speci-

mens was high. By endoscopic it is possible to postponesurgery, which should be reserved for patients with a high

risk of malignancy. The timing of the operation is not clear,

since randomised controlled studies are lacking. The factthat adenomas also develop after surgery, especially near

hepaticojejunal anastomosis, makes the decision about

whether to operate even more complex.

Pancreatitis and pancreatic stent

The post-papillectomy pancreatitis rate in our series was

9.8 %, which is acceptable. There were no cases of the

severe form of disease. The use of a prophylactic pancre-atic stent decreased the risk of pancreatitis, as has also been

shown in previous studies [18–21]. Using the guide-wire

cannulation technique, injection of contrast into the pan-creatic duct is not necessary to ensure correct positioning

Surg Endosc

123

Page 124: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

of the guide wire. Some endoscopists perform pancreatic

EPT before a papillectomy to ensure pancreatic stentingafterwards. The technique using methylene blue minimises

the risk of not locating the pancreatic duct orifice. In the few

cases where the pancreatic duct was not found with standardtechniques, administering secretin to the patient was helpful.

In our unit, we use pancreatic sphincterotomy in difficult

biliary cannulation, and it has been shown to be a safetechnique [22, 23]. However, in the present series, pancre-

atic EPT without a pancreatic stent was not sufficient toprevent pancreatitis; on the contrary, the risk increased

markedly in papillectomy. By changing our policy to insert a

pancreatic stent every time when the pancreatic duct isreached, we should be able to reduce our pancreatitis risk to

a lower level. It seems that ampullary cancer patients have

minimal risk of pancreatitis, but the explanation for this isunknown. Recently, it was suggested that non-steroidal anti-

inflammatory drugs (NSAIDs) would be more efficient than

a pancreatic stent in preventing post-ERCP pancreatitis [24],but NSAIDs and pancreatic stenting could also act syner-

gistically [25]. While papillectomy is a high-risk procedure,

both NSAIDs and pancreatic stenting may be needed untilthere is stronger evidence for either of them from prospec-

tive randomised studies.

Complications

If endoscopic treatment has acceptable morbidity, mortal-ity and recurrence rates, the procedure could be considered

to be the treatment of choice for benign tumours of the

duodenum. Post-papillectomy complications include pan-creatitis (5–19 %), perforation 0–5 %, bleeding 0–18 %

and cholangitis 0–4 % [21]. We had 11 cases of bleeding

(18 %), six of pancreatitis (9.8 %), two perforations(3.3 %) and no cholangitis. Our total complication rate of

24.6 % of patients is relatively high. However, among

these there were three patients with mild pancreatitisrequiring only 2 days of hospitalisation. In addition, all but

one patient could be treated conservatively, and there were

no procedure-related deaths. In treating FAP patients inparticular with papillectomy, safety is the most important

factor to be considered. The complication rate of FAP

patients was no higher than in other benign tumours, whichdiffers from the study by Irani et al. [14], where the pan-

creatitis rate was significantly higher in FAP patients.

Bleeding

Post-papillectomy bleeding happened in 18 % of thepatients. One explanation for the high percentage may be that

our policy is to do a duodenoscopy quickly, if there is any

suspicion of bleeding. Interestingly, coagulation of theresection margins during the papillectomy did not prevent

bleeding. This may, however, be due to the retrospective

nature of the study. Perhaps the patients with the greatest riskof bleeding were coagulated. Still, it is hard to find a means of

reducing the risk of bleeding, since using haemostatic agents

next to the pancreatic duct seems to be a risky option. Hae-mostatic clips placed after pancreatic stenting could be one

option to reduce bleeding complications.

In summary, the endoscopic ampullectomy is a safe andeffective treatment for benign ampullary tumours and

should be considered as the initial intervention. Jaundiceand bile duct dilation are risk factors for malignancy.

Residual adenomas occur frequently but are mostly treated

endoscopically. The risk of complications is high, but theycan usually be treated endoscopically or conservatively.

Pancreatic stenting decreases the risk of pancreatitis. It is

recommended to treat cancer surgically whenever possible.

Disclosures Drs. Shamel I, Udd M, Jarvinen H, Halttunen J andKylanpaa L have no conflicts of interest or financial ties to disclose.

Funding The manuscript has no funding.

References

1. Park SW, Song SY, Chung JB, Lee SK, Moon YM, Kang JK,Park IS (2000) Endoscopic snare resection for tumors of theampulla of Vater. Yonsei Med J 41:213–218

2. Treitschke F, Beger HG (1999) Local resection of benign pe-riampullary tumors. Ann Oncol 10(Suppl 4):212–214

3. Park SH, Kim YI, Park YH, Kim SW, Kim KW, Kim YT, KimWH (2000) Clinicopathologic correlation of p53 protein over-expression in adenoma and carcinoma of the ampulla of Vater.World J Surg 24:54–59

4. Baker HL, Caldwell DW (1947) Lesions of the ampulla of Vater.Surgery 21:523–531

5. Sato T, Konishi K, Kimura H, Maeda K, Yabushita K, Tsuji M,Miwa A (1999) Adenoma and tiny carcinoma in adenoma of thepapilla of Vater—p53 and PCNA. Hepatogastroenterology46:1959–1962

6. Bulow S, Bjork J, Christensen I, Fausa O, Jarvinen H, MoesgaardF, Vasen HF, DAF Study Group (2004) Duodenal adenomatosisin familial adenomatous polyposis. Gut 53:381–386

7. Bulow S, Christensen I, Højen H, Bjork J, Elmberg M, JarvinenH, Lepisto A, Nieuwenhuis M, Vasen H (2012) Duodenal sur-veillance improves the prognosis after duodenal cancer infamilial adenomatous polyposis. Colorectal Dis 14:947–952

8. Yeo C, Cameron J, Sohn T, Lillemoe KD, Pitt HA, Talamini MA,Hruban RH, Ord SE, Sauter PK, Coleman J, Zahurak ML, Gro-chow LB, Abrams RA (1997) Six hundred fifty consecutivepancreaticoduodenectomies in the 1990s. Ann Surg 226:248–260

9. Lepisto A, Kiviluoto T, Halttunen J, Jarvinen H (2009) Surveil-lance and treatment of duodenal adenomatosis in familial ade-nomatous polyposis. Endoscopy 41:504–509

10. Galandiuk S, Hermann R, Jagelman D, Fazio V, Sivak M (1988)Villous tumors of the duodenum. Ann Surg 207:234–239

11. Bjork K, Davis C, Nagorney D, Mucha P Jr (1990) Duodenalvillous tumors. Arch Surg 125:961–965

12. Han J, Kim M (2006) Endoscopic papillectomy for adenomas ofthe major duodenal papilla (with video). Gastrointest Endosc63:292–301

Surg Endosc

123

Page 125: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

13. Cotton P, Lehman G, Vennes J, Geenen JE, Russell RC, MeyersWC, Liguory C, Nickl N (1991) Endoscopic sphincterotomycomplications and their management: an attempt at consensus.Gastrointest Endosc 37:383–393

14. Irani S, Arai A, Ayub K, Biehl T, Brandabur J, Dorer R, Gluck M,Jiranek G, Patterson D, Schembre D, Traverso L, Kozarek R (2009)Papillectomy for ampullary neoplasm: results of a single referralcenter over a 10-year period. Gastrointest Endosc 70:923–932

15. Beger HG, Treitschke F, Gansauge F, Harada N, Hiki N, Matt-feldt T (1999) Tumor of the ampulla of Vater: experience withlocal or radical resection in 171 consecutively treated patients.Arch Surg 134:526–532

16. Kim J, Kim J, Han J, Yoo B, Kim M, Kim W (2009) Is endo-scopic papillectomy safe for ampullary adenomas with high-grade dysplasia? Ann Surg Oncol 16:2547–2554

17. Parc Y, Mabrut J, Schields C, Mallorca Group (2011) Surgicalmanagement of the duodenal manifestations of familial ade-nomatous polyposis. Br J Surg 98:480–484

18. Singh P, Das A, Isenberg G, Wong R, Sivak M Jr, Agrawal D,Chak A (2004) Does prophylactic pancreatic stent placementreduce the risk of post-ERCP acute pancreatitis? A meta-analysisof controlled trials. Gastrointest Endosc 60:544–550

19. Harewood G, Pochron N, Gostout C (2005) Prospective, ran-domized, controlled trial of prophylactic pancreatic stent place-ment for endoscopic snare excision of the duodenal ampulla.Gastrointest Endosc 62:367–370

20. Yamao T, Isomoto H, Kohno S, Mizuta Y, Yamakawa M, NakaoK, Irie J (2010) Endoscopic snare papillectomy with biliary andpancreatic stent placement for tumors of the major duodenalpapilla. Surg Endosc 24:119–124

21. Napoleon B, Alvarez-Sanchez V, Leclercq P, Mion F, Pialat J,Gincul R, Ribeiro D, Cambou M, Lefort C, Rodrıguez-GirondoM, Scoazec J (2013) Systematic pancreatic stenting after endo-scopic snare papillectomy may reduce the risk of postinterven-tional pancreatitis. Surg Endosc 27:3377–3387

22. Catalano MF, Linder JD, Geenen JE (2004) Endoscopic trans-pancreatic papillary septotomy for inaccessible obstructed bileducts: comparison with standard pre-cut papillotomy. Gastroin-test Endosc 60:557–561

23. Udd M, Kylanpaa L, Halttunen J (2010) Management of difficultbile duct cannulation in ERCP. World J Gastrointest Endosc16:97–103

24. Akbar A, Abu D, Baron T, Wang Z, Altayar O, Murad M (2013)Rectal nonsteroidal anti-inflammatory drugs are superior topancreatic duct stents in preventing pancreatitis after endoscopicretrograde cholangiopancreatography: a network meta-analysis.Clin Gastroenterol Hepatol 11:778–783

25. Cheon Y (2013) Can postendoscopic retrograde cholangiopan-creatography pancreatitis be prevented by a pharmacologicalapproach? Korean J Intern Med 28:141–148

Surg Endosc

123

Page 126: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

III

Page 127: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE
Page 128: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

0954-691X Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved. DOI: 10.1097/MEG.0000000000001515 1200

Original article

Criteria for difficult biliary cannulation: start to countShamel Ismail, Marianne Udd, Outi Lindström, Mia Rainio, Jorma Halttunen and Leena Kylänpää

Introduction

The primary biliary cannulation with sphincterotome and guidewire or catheter fails in 15–35% of patients [1–3]. The risk of post-endoscopic retrograde cholangiopancrea-tography (ERCP) pancreatitis (PEP) varies between 3 and 15%, and is increased in high-risk patient groups [4, 5]. Difficult cannulation, needle-knife precut, frequent injec-tions into the pancreatic duct (PD), transpancreatic bil-iary sphincterotomy (TPBS) and balloon dilatation of the intact biliary sphincter are reported to be procedure-re-lated factors that are responsible for the PEP risk [6, 7].

The definition of difficult biliary cannulation [8] is more than five contacts with the papilla; more than 5 min cannulation time following touching of the papilla; more than one unintended PD guidewire passage. This defini-tion is based on a Nordic multicenter study [9].

Aim of the study is to confirm these criteria for difficult cannulation with regard to the PEP rate, and to study the final cannulation success, when primary cannulation meth-ods and advanced cannulation methods like TPBS with or without needle-knife precut sphincterotome precut (SPH),

needle-knife precut, double guidewire cannulation (DGW) and papillectomy were used.

Methods

Analysis of routinely collected data in the Helsinki University Hospital was undertaken during March 2011 to September 2014. Four gastrointestinal surgeons under-took 3272 ERCPs, including 1723 cases with native papilla. After every ERCP, an endoscopist had to fill an excel file with details of patient demographics, indication, cannulation details, performed procedures. We excluded 322 patients where the PD was the target and 580 patients without a complete recorded cannulation time and details. These included altogether 6–18 variables (method: cannu-lation time, number of papilla contacts, number of pancre-atic guidewire passages and contrast injections) depending on the needed cannulation methods to gain success. The number of patients with the recorded cannulation details increased from 37.8% in year 2011 to 69.3% in year 2014, P < 0.001. The study group comprised 821 patients. The study was approved by the hospital ethics committee.

Peroral levofloxacin 500  mg was administered. After September 2013, protocol to administer rectal diclofenac (100 mg) before ERCP was started, and during this period, 256 patients underwent ERCP. Of them, 181 (71%) had rectal diclofenac and 75 did not receive it (elevated cre-atinine: n = 42; allergy: n = 7; miscellaneous reasons: n = 9; reason not known: n = 17). All in all, 22.1% of the patients had rectal diclofenac. Patients were in left lat-eral decubitus position and an anesthesiologist provided conscious sedation with propofol. During perioperative period, patients received standard hydration. We did not have aggressive hydration policy [10].

European Journal of Gastroenterology & Hepatology, 2019 31:1200–1205

Keywords: adverse event, biliary sphincterotomy, difficult cannulation, ERCP, mortality, needle-knife precut, post-ERCP pancreatitis, sphincterotome precut, transpancreatic biliary sphincterotomy

Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland

Correspondence to Marianne Udd, MD, PhD, University of Helsinki and Helsinki University Hospital, P.O. Box 340, HUS 00029, Finland

Tel: +358504270182; fax: +3580947174688; e-mail: [email protected]

Received 10 April 2019 Accepted 18 June 2019

Objective In European Society of Gastrointestinal Endoscopy guidelines, biliary cannulation of native papilla is defined as difficult in the presence of >5 papilla contacts, >5 min cannulation time or >1 unintended pancreatic duct cannulation (5-5-2). The aim is to test 5-5-2-criteria in a single-center practice predicting the risk of post-endoscopic retrograde cholangiopancreatography pancreatitis (PEP), and to study the efficacy of transpancreatic biliary sphincterotomy (TPBS) as an advanced method for biliary cannulation.Methods Prospectively collected data of 821 patients with native papilla were analyzed. Primary cannulation was the first method chosen for cannulation (sphincterotome and a guidewire). Advanced cannulation method was endoscopist-chosen cannulation method after failed primary cannulation.Results Primary cannulation succeeded in 599 (73%) patients in a median of 2 min. TPBS ± needle knife resulted in a 90% success rate. The final cannulation success was 814 (99.1%) cases in a median of 5.3 min. PEP risk was 4.0%. When primary cannulation succeeded, the PEP rate was 2.3%. When advanced methods were needed, the PEP rate increased to 13.5%. Altogether 311 (37.9%) patients fulfilled at least one 5-5-2-criterion. In patients without 5-5-2-criteria, the primary cannulation succeeded in 79.6% (n = 477), compared to 20.4% (n = 122) with the criteria, P < 0.001, indicating the need to exchange the cannulation method instead of persistence. If all the 5-5-2-criteria were present, the risk of PEP was 12.7%.Conclusion The results support the use of the 5-5-2-criteria for difficult cannulation. TPBS is an effective advanced cannulation method with an acceptable complication rate. Eur J Gastroenterol Hepatol 31: 1200–1205Copyright © 2019 Wolters Kluwer Health, Inc. All rights reserved.

Page 129: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

www.eurojgh.com 1201Criteria for difficult biliary cannulation Ismail et al.

Definitions

The cannulation time was defined as the period between the first contact with the papilla and the deep biliary cannulation. An attempt was defined as each continu-ous contact with the papilla. Loss of contact and repo-sitioning counted as a new attempt. Contrast injections and passages with a guidewire into the PD were counted. Total procedure time was a period between insertion and removal of duodenoscope.

Primary cannulation was defined as the first cannu-lation method (cannulation with sphincterotome and a straight guidewire) as used. After primary cannulation, second, third and fourth methods were advanced can-nulation methods chosen by an endoscopist. Advanced cannulation methods were TPBS, SPH, needle knife (NK) precut, DGW and papillectomy. In TPBS, with the guide-wire in the PD, an incision is directed toward the bile duct through the septum between the two ducts. Then the bil-iary duct opens and cannulation can be attempted with sphincterotome and guidewire; or the wall of the duct becomes visible and can be opened with a NK and then, cannulation with sphincterotome and a guidewire is possi-ble [11, 12]. In sphincterotomy precut, papillary orifice is cut with sphincterotome inside papilla, then, cannulation could be attempted. In NK technique, papillary orifice was cut with NK until the underlying biliary sphincter visual-ized and cannulation can be attempted. In papillectomy, papillary mucosa is resected with snare to visualize bil-iary duct orifice to help cannulation of the biliary duct. In DGW technique, with the guidewire in the PD, biliary cannulation is attempted with the second guidewire. We did not routinely place prophylactic pancreatic stents after advanced cannulation.

Successful cannulation was defined as a biliary cannu-lation with the guidewire inside the common bile duct. Failed cannulation was a situation when no access to bil-iary duct was achieved during ERCP procedure. Difficult cannulation was defined according to the European Society of Gastrointestinal Endoscopy guidelines (5-5-2 criteria) [8]. Easy cannulation was defined as cannulation not fulfilling the difficult cannulation criteria. Adverse events were defined and graded according to the modified Cotton consensus [13].

Mild PEP was defined as a state with clinical pancreati-tis, amylase at least three times normal at more than 24 h after the procedure, requiring admission or prolongation of planned admission to 2–3 days, moderate pancreatitis as pancreatitis requiring hospitalization of 4–10 days and severe pancreatitis as pancreatitis requiring hospitaliza-tion for more than 10 days, or hemorrhagic pancreatitis, phlegmon, or pseudocyst, or intervention (percutaneous drainage or surgery). Bleeding was graded as mild (clinical bleeding was present with no need of transfusion), moder-ate [need of transfusion (4 units or less), no angiographic intervention or surgery] and severe (need of transfusion 5 units or more, or angiographic/surgical intervention). Perforation was graded as mild (slight leak of fluid or contrast, treatable by fluids and suction for 3 days or less), moderate (any definite perforation treated medically for 4–10 days) or severe (hospital stay for more than 10 days, or percutaneous or surgical intervention needed). Cholangitis was graded as mild (fever >38ºC, 24–48 h),

moderate (fever or sepsis requiring more than 3 days of hospital stay or endoscopic or percutaneous intervention) or severe (septic shock or surgery).

SPSS v 15.0 (IBM Corporation, Somers, New York) was used for data analysis. The χ2 test was used to test for a difference between categorical variables. A nonparamet-ric Wilcoxon-Mann–Whitney test was used to compare differences in continuous and ordinal variables. P-values <0.05 (two-sided) were regarded as being statistically significant.

Results

Cannulation details in 821 patients

Data of 821 patients with recorded cannulation time are presented in Table 1. The total cannulation time was median 2 min (range 0.02–89.5 min) in 814 patients with final cannulation success. The final cannulation success method was 99.1%. In seven (0.91%) patients, biliary cannulation failed. Of the successful primary cannula-tions, 49% could be performed in a minute and 91% in 5 min. The most common advanced cannulation method in 221 cases was TPBS in 70.6% (n = 156). SPH was the second most common in 20% (n = 44) of the cases. In total, TPBS was used in 76.5% (n = 169) of the cases. Flow chart of cannulation methods in 821 native papilla patients is presented in Fig. 1.

Table 1. Baseline characteristics of 821 patients and endoscopic retrograde cholangiopancreatography procedures

Age (years) (median, range) 69 (8–102)

Male/female 437 (53%)/384 (47%)BMI (kg/m2) (median, range) 25.7 (12.1–51.9)ASA class I–I 210 (25.6%)ASA class III–IV 611 (74.4%)Elective/emergency procedure 420 (51%)/401 (49%)Indication: stone 445 (54%)Indication: stricture 310 (38%)Indication: postcholecystectomy, liver transplant,

other66 (8%)

Diclofenac prophylaxis (starting Oct 2013) 181 (71%)Duodenal diverticula 93 (11%)Biliary pancreatitis before ERCP 78 (9.5%)Primary cannulation success 599 (73) Time for success (min) (median, range) 1.1 (0.02–19.67) Cannulation attempts (median, range) 2 (0–15) Unintended PD cannulations (median, range) 0 (0–8) Contrast injections to PD (median, range) 0 (0–1)Second method attempted after (min) (median, range) 4.8 (0.3–28)Cannulation time with the second method (min)

(median, range)4.2 (0.3–56)

Secondary cannulation success 144 Time for success with the second method (min)

(median, range)3 (0.33–56)

Third method attempted after (min) (median, range) 7 (1–56) Cannulation time with the third method; min

(median, range)5 (0.3–39.3)

Cannulation time, total min (median, range) 2 (0.02–89.5)Total procedure time (min) (median, range) 18 (5–110)Biliary sphincterotomy 803 (97.8%)Biliary cytology 209 (25.5%)Biliary duct dilatation 77 (9.4%)Common bile duct stone extraction 358 (43.6%)Biliary stent application 286 (34.8%)Pancreatic sphincterotomy 169 (20.5%)Protective pancreatic stent 5 (0.6 %)

ASA, Data of Physical Status Classification of the American Society of Anesthesiologists ASA class; ERCP, endoscopic retrograde cholangiopancreatography.

Page 130: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

1202 European Journal of Gastroenterology & Hepatology October 2019 • Volume 31 • Number 10

Difficult cannulation

The primary cannulation time was more than 5 min in 152 patients (18.5%), at least five cannulation attempts were needed in 141 (17.2%) patients and unintended pancreatic passages occurred in 234 (28.5%) patients. Altogether, 311 out of 821 (37.9%) patients met at least one 5-5-2-criterion. If there were no 5-5-2-criteria present, 2.2% (n = 11) had PEP, if 5-5-2-criteria were present, the risk was 7.1% (n = 22), P = 0.001. If one criterion was present, the PEP rate was 6.9% (n = 11) and if all the criteria were present, 12.3% (n = 8) suffered from PEP. Difficult cannulation criteria were present significantly more often with biliary stricture than with other indica-tions, 46.3% (n = 152) vs. 32.3% (n = 159); P < 0.001. Accordingly, primary cannulation failed significantly more often with this indication compared to other indications, 36.9% (n = 121) vs. 20.5% (n = 101); P < 0.001. The flow chart of 821 native papilla patients with easy and difficult cannulation is presented in Fig. 2.

Unsuccessful cannulation

In seven patients, access to biliary duct was not achieved. In them, cannulation was attempted for median of 48 min (range 1.00–81.00). There were three patients with com-mon bile duct (CBD) stones (one patient had successful repeat ERCP, one patient underwent repeat magnetic reso-nance cholangiopancreatography with finding of no CBD stones and one moribund patient did not undergo repeat ERCP) and four cases with malignant strictures (one patient had successful repeat ERCP and three patients had percutaneous transhepatic biliary drainage).

Post-ERCP pancreatitis

Thirty-three (4.0%) patients developed PEP. It was mild in 19, moderate in 20 and severe in three patients. When primary cannulation succeeded, 14 (2.3%) patients had PEP. After the second cannulation method, the PEP rate was 6.3% (nine patients) and after the third and following

Fig. 1. The flow chart of different cannulation methods in 821 native papilla patients. DGW, double guidewire cannulation; NK, needle knife precut; PD, pancreatic duct; SPH, sphincterotome precut; TPBS, transpancreatic biliary sphincterotomy.

Page 131: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

www.eurojgh.com 1203Criteria for difficult biliary cannulation Ismail et al.

cannulation methods, the risk of PEP was 13.5% (10 patients).

If the biliary duct was cannulated in 5 min, 2.1% (12 patients) had PEP, if it took more than 5 min, the PEP rate increased to 8.7% (21 patients), P < 0.001. The PEP risk was 2.1% (12 patients) if a maximum of five cannulation attempts was needed for biliary cannulation, but increased to 8.8% (21 patients) with additional attempts, P < 0.001. If PD was not cannulated at all, the PEP risk was 2.8% (13 patients). If PD was cannulated up to one (0–1) time, the PEP risk was 3.3% (18 patients) compared to PD cannu-lation more than once: 6.4% (15 patients); P = 0.048. In 29 (3.5%) patients, contrast was injected in to the PD. In them, the risk of PEP was 10% (n = 3) compared to 3.8% (n = 30), in patient without contrast injection, P = 0.106. The presence of a trainee did not affect the PEP rate, four patients (5.8%) with trainees vs. 29 patients (3.9%) with-out them, P = 0.35. In patients with biliary pancreatitis, there was no exacerbation of pancreatitis after ERCP.

With TPBS ± NK method, the PEP rate was 8.9%; n = 15, compared to situation when this was not used 2.8%, n = 18, P < 0.001.

After September 2013, the protocol to administer rectal diclofenac to prevent PEP was started. There was no sig-nificant difference in PEP rate without or with diclofenac, [28 (4.4%) vs. 5(2.8%); P = 0.31].

Other adverse events

A total of 49 patients (6%) had adverse events: PEP in 33 (4%) patients, bleeding in 20 (2.4%), perforation in four (0.5%) and cholangitis in 10 (1.2%) patients. In cases of difficult cannulation, the rate of adverse events was 9.3% (n = 29), but in cases of easy cannulation, the rate was 3.9% (n = 20), P = 0.002. After excluding PEP complication, there was no differences in complication rates between patients with 5-5-2-criteria and easy cannu-lation; 11 (3.5%) vs. 12 (2.4%), P = 0.384. TPBS did not

Patients

n = 821

Easy cannulation

n = 510

Difficult cannulation

n = 311

Primarycannulation

success477(93.5%)

Primary cannulation

failure33(6.5%)

Primary cannulation

success122(39.7%)

Primary cannulation

failure189(60.8%)

2nd methodsuccess

24(72.7%)

2nd methodfailure

8(14.3%)

2nd methodsuccess

141(74.6%)

2nd methodfailure

48(25.4%)

3rd methodsuccess

8(100%)

3rd methodfailure

0

3rd methodsuccess

38(79.1%)

3rd method failure4(8.3%)

4th method success4(100%)

No attempt, final failure

1(3%)

No attempt, final failure6(12.5%)

Fig. 2. The flow chart of 821 native papilla patients with easy and difficult cannulation.

Page 132: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

1204 European Journal of Gastroenterology & Hepatology October 2019 • Volume 31 • Number 10

adversely affect the rate of other complications, 3% (n = 5), versus 2.8% (n = 18) without the procedure, P = 0.89.

30-day mortality

Total 30-day mortality was 5% (n = 42). There was no difference in patients with and without PEP, (3/9% vs. 39/4.9%, P = 0.235) or in patients with difficult and easy cannulation (21/6.8% vs. 21/4.1%, P = 0.104); or when TPBS or other rescue methods were necessary [(13/8.3% vs. 4/6.2%); P = 0.58]. Deaths were mostly related to underlying disease (malignancy in 70.4%), and not directly to complications.

Discussion

The study results support the use of the 5-5-2 criteria for difficult cannulation [9] as it predicts the risk of PEP. Our preferred advanced cannulation method was TPBS in 70.6% of the cases. Altogether 37% had difficult cannu-lation criteria in the present series, and advanced cannula-tion methods were used in 27% of the cases. TPBS as our preferred advanced cannulation method ended in a final success rate of over 99% and a PEP rate of 4%.

Transpancreatic biliary sphincterotomy

TPBS as a rescue method for deep biliary cannulation by Goff et al. [11] succeeds in 86–100% of the cases and has acceptable PEP rates (3.5–10.4%) [11, 12, 14–18]. Previously, in our unit, the biliary cannulation success was less common after a NK precut (71.3%) than after the TPBS (97.3%; P < 0.001) with PEP rates of 5.1% and 9.3%, respectively. When TPBS was combined to NK precut, the PEP rate was 8.2% [12]. In present series, the PEP rate with TPBS is similar, 8.9%.

Other rescue techniques

With TPBS, we had similarly high success rate (97.6%) as in the literature. In difficult cannulation, DGW and TPBS were compared in a prospective randomized study of 34 patients in each group. TPBS had higher success rate of 94% compared to 58% in DGW group. PEP rate in both groups were 2.9% [19]. When DGW technique and TPBS as a rescue method after a failed primary cannulation in 10 min were compared, the cannulation time was 7.8 ± 1.7 min and 3.7 ± 2.3 min, P < 0.001, respectively, with similar PEP risks, 1.8% [3]. The adverse events rate was 2.19% in the DGW group and 7.04% in the TPBS group, P = 0.04. In this study, however, primary cannulation suc-ceeded in only 23.8% of the 366 patients and is much lower than in other studies with 75–90% success rates of primary cannulation.

Early NK precut has also been effective. After an unsuc-cessful biliary cannulation in native papilla patients, the suc-cess rate with an early NK was 79.1%, and with the DGW technique, it was only 44.8%. The PEP rates were 4.5% and 14.9%, respectively [16]. We seldom used DGW technique. DGW cannulation needs an additional guidewire in the PD to straighten the biliary duct. Sometimes an additional guide-wire blocks the access, especially in cases of a tight tumor stricture. In DGW cannulation, an additional guidewire still easily enters the pancreatic and not biliary duct.

A competent ERCP-endoscopist should be able to can-nulate the duct of interest in at least 90% of procedures [20]. However, risk of PEP increases with repeated can-nulation attempts, when papilla is manipulated and espe-cially with multiple guidewire passages to PD. Keeping this in mind, endoscopist must be aware of the time spent to cannulate. The frequency of 5-5-2-criteria should be routinely recorded. It will assist in decision making when to change the cannulation method or endoscopist or post-pone ERCP. In the present study, when difficult criterion was fulfilled, primary method success rate was only 20.4%. Early exchange of cannulation method enhances cannula-tion success with lower PEP rates compared persistence in cannulation with sphincterotome and guidewire.

Limitations

We were not able to collect data of 5-5-2-criteria in con-secutive patients with native papilla due to busy clinical practice. Intention was to routinely collect cannulation data to guide clinical decision making. The ratio of patients with complete data increased during the study period. Cannulation time, pancreatic passages and papilla contacts were monitored with numerous patients. Aggressive hydra-tion with lactated Ringer solution decreased the incidence of PEP in a meta-analysis, but needs further evaluation [10]. We had standard hydration policy. The use of preven-tive techniques for PEP was not uniform, rectal non-ste-roidal anti-inflammatory drug administration to prevent PEP was started during the data collection. However, rec-tal diclofenac probably may have a limited impact in low PEP rate units. In our recent article, with unselected ERCP patients, PEP rate was similar, 2.8%, in 1000 historic con-trols without diclofenac prophylaxis and in 1000 patients with the prophylaxis. The rate in native papilla cases was 3.9% and 3.6%, accordingly [21]. Also, protective pancre-atic stenting was rare, and it was mainly used with papillec-tomy cases. Preventive pancreatic stents have been used to prevent PEP. In meta-analyses, PEP rate was 13.6–18.9% without protective pancreatic stents, but with the stent the PEP rate was 3.2–7% [22, 23]. Majority of the patients, however, had sphincter of Oddi dyskinesia (SOD) or other high PEP risk status as an ERCP indication. In our unit, SOD is an extremely rare indication for ERCP. In recent meta-analysis of 12 RCT and 1369 patients, prophylactic pancreatic stents significantly decreased the odds of PEP (odds ratio 0.28; 95% confidence interval: 0.18–0.42) [24]. In a Californian study of 1873 patients, overall biliary can-nulation success rate was 97% and advanced cannulation techniques (DGW, TPBS and NKS) were needed in 12%. Protective PD stents were placed with repeated PD cannu-lation and a policy of giving indomethacin suppositories with advanced cannulation techniques was started. Overall, PEP rate was only 0.4% [25]. Our main rescue method was TPBS and we seldom used other methods. In the future, these advanced methods should be compared in a prospec-tive randomized study.

Conclusion

The ERCP-endoscopist should monitor the difficult cannulation criteria in clinical practice. Risk of PEP increases when at least one criterion is present. Counting

Page 133: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

Copyright © 2019 Wolters Kluwer Health, Inc. Unauthorized reproduction of this article is prohibited.

www.eurojgh.com 1205Criteria for difficult biliary cannulation Ismail et al.

5-5-2-criteria will help in decision making when to change the cannulation method. TPBS seems to be an efficient res-cue method.

Acknowledgements

Conflicts of interest

There are no conflicts of interest.

References1 Nalankilli K, Kannuthurai S, Moss A. A modern approach to ERCP:

maintaining efficacy while optimising safety. Dig Endosc 2016; 28(Suppl 1):70–76.

2 Zhang QS, Han B, Xu JH, Gao P, Shen YC. Needle-knife papillotomy and fistulotomy improved the treatment outcome of patients with diffi-cult biliary cannulation. Surg Endosc 2016; 30:5506–5512.

3 Huang L, Yu QS, Zhang Q, Liu JD, Wang Z. Comparison between double-guidewire technique and transpancreatic sphincterotomy technique for difficult biliary cannulation. Dig Endosc 2015; 27: 381–387.

4 Cotton PB, Garrow DA, Gallagher J, Romagnuolo J. Risk factors for complications after ERCP: a multivariate analysis of 11,497 proce-dures over 12 years. Gastrointest Endosc 2009; 70:80–88.

5 Kochar B, Akshintala VS, Afghani E, Elmunzer BJ, Kim KJ, Lennon AM, et al. Incidence, severity, and mortality of post-ERCP pancre-atitis: a systematic review by using randomized, controlled trials. Gastrointest Endosc 2015; 81:143–149.e9.

6 Loperfido S, Angelini G, Benedetti G, Chilovi F, Costan F, De Berardinis F, et al. Major early complications from diagnostic and therapeutic ERCP: a prospective multicenter study. Gastrointest Endosc 1998; 48:1–10.

7 Williams EJ, Taylor S, Fairclough P, Hamlyn A, Logan RF, Martin D, et al. Risk factors for complication following ERCP; results of a large-scale, prospective multicenter study. Endoscopy 2007; 39: 793–801.

8 Testoni PA, Mariani A, Aabakken L, Arvanitakis M, Bories E, Costamagna G, et al. Papillary cannulation and sphincterotomy tech-niques at ERCP: European Society of Gastrointestinal Endoscopy (ESGE) clinical guideline. Endoscopy 2016; 48:657–683.

9 Halttunen J, Meisner S, Aabakken L, Arnelo U, Grönroos J, Hauge T, et al. Difficult cannulation as defined by a prospective study of the Scandinavian Association for Digestive Endoscopy (SADE) in 907 ercps. Scand J Gastroenterol 2014; 49:752–758.

10 Wu D, Wan J, Xia L, Chen J, Zhu Y, Lu N. The efficiency of aggres-sive hydration with lactated ringer solution for the prevention of post-ERCP pancreatitis: a systematic review and meta-analysis. J Clin Gastroenterol 2017; 51:e68–e76.

11 Goff JS. Long-term experience with the transpancreatic sphincter pre-cut approach to biliary sphincterotomy. Gastrointest Endosc 1999; 50:642–645.

12 Halttunen J, Keränen I, Udd M, Kylänpää L. Pancreatic sphincterot-omy versus needle knife precut in difficult biliary cannulation. Surg Endosc 2009; 23:745–749.

13 Cotton PB, Lehman G, Vennes J, Geenen JE, Russell RC, Meyers WC, et al. Endoscopic sphincterotomy complications and their management: an attempt at consensus. Gastrointest Endosc 1991; 37:383–393.

14 Catalano MF, Linder JD, Geenen JE. Endoscopic transpancreatic papillary septotomy for inaccessible obstructed bile ducts: compar-ison with standard pre-cut papillotomy. Gastrointest Endosc 2004; 60:557–561.

15 Weber A, Roesch T, Pointner S, Born P, Neu B, Meining A, et al. Transpancreatic precut sphincterotomy for cannulation of inaccessible common bile duct: a safe and successful technique. Pancreas 2008; 36:187–191.

16 Kim SJ, Kang DH, Kim HW, Choi CW, Park SB, Song BJ, Hong YM. Needle-knife fistulotomy vs double-guidewire technique in patients with repetitive unintentional pancreatic cannulations. World J Gastroenterol 2015; 21:5918–5925.

17 Kahaleh M, Tokar J, Mullick T, Bickston SJ, Yeaton P. Prospective eval-uation of pancreatic sphincterotomy as a precut technique for biliary cannulation. Clin Gastroenterol Hepatol 2004; 2:971–977.

18 Lin LF. Transpancreatic precut sphincterotomy for biliary access: the relation of sphincterotomy size to immediate success rate of biliary cannulation. Diagn Ther Endosc 2014; 2014:864082.

19 Sugiyama H, Tsuyuguchi T, Sakai Y, Mikata R, Yasui S, Watanabe Y, et al. Transpancreatic precut papillotomy versus double-guidewire tech-nique in difficult biliary cannulation: prospective randomized study. Endoscopy 2018; 50:33–39.

20 Baron TH, Petersen BT, Mergener K, Chak A, Cohen J, Deal SE, et al.; ASGE/ACG Taskforce on Quality in Endoscopy. Quality indi-cators for endoscopic retrograde cholangiopancreatography. Am J Gastroenterol 2006; 101:892–897.

21 Rainio M, Lindström O, Udd M, Louhimo J, Kylänpää L. Diclofenac does not reduce the risk of post-endoscopic retrograde cholangio-pancreatography pancreatitis in low-risk units. J Gastrointest Surg 2017; 21:1270–1277.

22 Choudhary A, Bechtold ML, Arif M, Szary NM, Puli SR, Othman MO, et al. Pancreatic stents for prophylaxis against post-ERCP pancreati-tis: a meta-analysis and systematic review. Gastrointest Endosc 2011; 73:275–282.

23 Mazaki T, Mado K, Masuda H, Shiono M. Prophylactic pancreatic stent placement and post-ERCP pancreatitis: an updated meta-anal-ysis. J Gastroenterol 2014; 49:343–355.

24 Vadala di Prampero SF, Faleschini G, Panic N, Bulajic M. Endoscopic and pharmacological treatment for prophylaxis against posten-doscopic retrograde cholangiopancreatography pancreatitis: a meta-analysis and systematic review. Eur J Gastroenterol Hepatol 2016; 28:1415–1424.

25 Huang C, Kung J, Liu Y, Tse A, Datta A, Singh I, et al. Use of dou-ble wire-guided technique and transpancreatic papillary septot-omy in difficult ERCP: 4-year experience. Endosc Int Open 2016; 4:E1107–E1110.

Page 134: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

IV

Page 135: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE
Page 136: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

https://doi.org/10.1177/1457496918822616

Scandinavian Journal of Surgery 1 –6 © The Finnish Surgical Society 2019Article reuse guidelines: sagepub.com/journals-permissionsDOI: 10.1177/1457496918822616journals.sagepub.com/home/sjs

SCANDINAVIANJOURNAL OF SURGERY

SJSEffEct And PrEdictivE vAluE Of rOutinE PrEOPErAtivE lAbOrAtOry tEsting fOr EndOscOPic rEtrOgrAdE chOlAngiOPAncrEAtOgrAPhy

J. Jokelainen1,2, s. ismail1,3, l. Kylänpää1,3, M. udd1,3, h. Mustonen1,3, O. lindström1,3, r. Pöyhiä1,4

1University of Helsinki, Helsinki, Finland2 Department of Anesthesia and Intensive Care Medicine, South Karelia Central Hospital, Lappeenranta, Finland

3 Department of Gastroenterological and General Surgery, Helsinki University Hospital, Helsinki, Finland 4Kauniala Hospital, Kauniainen, Finland

AbstrAct

Background and Aims: several studies and guidelines are questioning routine preoperative laboratory tests in surgical and endoscopic procedures. their effect in endoscopic retrograde cholangiopancreatography is not currently known. this study was carried out to evaluate the risk of adverse effects in endoscopic retrograde cholangiopancreatography and their association with preoperative lab tests.Materials and Methods: A single-center, prospective observational study on all 956 patients undergoing 1196 endoscopic retrograde cholangiopancreatographies in the Endoscopy unit of helsinki university central hospital from 1 March 2012 to 28 february 2013. routine preoperative laboratory test results (basic blood count, creatinine, potassium, sodium, international normalized ratio/thromboplastin time, and amylase), health status, medication, and demographic information of all patients were analyzed in relation to adverse effects related to endoscopic retrograde cholangiopancreatography and procedural sedation.Results: Multivariate analysis showed post–endoscopic retrograde cholangiopancreatography pancreatitis (43 cases, 3.6%) to have no association with abnormal routine preoperative laboratory tests. respiratory depression caused by sedation (128 cases, 11%) was not associated with abnormal routine preoperative laboratory tests, and anemia was found to be a slightly protecting factor. cardiovascular depression caused by sedation was associated

Correspondence:Jarno Jokelainen Department of Anesthesia and Intensive Care Medicine South Karelia Central Hospital Valto Käkelän katu 1 53130 Lappeenranta Finland Email: [email protected]

Original Research Article

Page 137: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

J. Jokelainen, et al.2

with thrombocytopenia (odds ratio = 1.87, p = 0.025) and, in male patients, hyponatremia (odds ratio = 3.66, p < 0.001). incidence of other adverse effects was too low for statistical analysis.Conclusion: routine universal preoperative lab testing was not found to be successful in predicting adverse effects in endoscopic retrograde cholangiopancreatography procedures. laboratory testing should be done focusing on each patient’s individual needs.

Key words: Cholangiopancreatography; endoscopic retrograde; pancreatitis; endoscopy; hematologic tests; adverse effects; anesthesia

InTRODUCTIOn

Routine preoperative laboratory testing (RPLT) is a practice of ordering a given set of laboratory tests to every patient before a procedure regardless of the health status of individual patients. Traditionally, rou-tine laboratory tests have been part of the protocol for endoscopic retrograde cholangiopancreatography (ERCP) in our institution. Several studies and guide-lines are questioning such policy for surgery and endoscopy (1–3). ERCP is one of the most technically demanding endoscopic procedures with a risk for seri-ous adverse effects (4). To our knowledge, no studies have been made to assess the effect of RPLT on patient outcomes and adverse effects for ERCP. Because of the policy of RPLT in our institution, we had a large data-base of preoperative laboratory results we could com-pare with adverse effects associated with ERCP.

This study was carried out in order to assess the association of RPLT before ERCP with adverse effects and mortality in the Endoscopic Unit of Meilahti Hospital, a tertiary university clinic where over 1200 ERCP procedures are performed annually.

MATERIAL AnD METHODS

This study was approved by the Institutional Ethics Committee of Helsinki University Central Hospital (Ethics Committee, Department of Surgery, Biomedicum Helsinki 2C, Tukholmankatu 8C, PL 705, 00029 Helsinki, Finland).

A total number of 1228 ERCPs were attempted at the hospital during the 12-month period from 1 March 2012 to 28 February 2013. All the adult patients who were treated in the Endoscopy Unit were included in the study.

The following data were registered on each patient: age, weight, height, American Society of Anesthesiology (ASA) physical status classification, RPLT results (basic blood count (BBC), electrolytes (P-na and P-K), throm-boplastin time (P-TT) and international normalized ratio (P-InR), amylase levels before the procedure and 4 and 24 h after the procedure), indications of ERCP, cannulation method, performed procedures, duration of the procedure, ERCP adverse events (pancreatitis, bleeding, perforation, etc.), sedation-related adverse effects (cardiovascular depression—defined as systolic blood pressure <90 mm Hg, respiratory depression—defined as oxygen saturation SpO2 < 90%, need for mask ventilation, need for intubation), comorbidities (diabetes treated with insulin, diabetes treated with

oral medication, electrocardiogram (ECG) analysis, hypertension, cardiovascular disease, cerebrovascular disease, atherosclerosis, chronic obstructive pulmo-nary disease, other pulmonary disease, kidney failure, hypothyreosis/other endocrinological disease, malig-nancy, bleeding disorders, other), regular medication (opioids, diuretics, beta blockers, other cardiological medication, corticosteroids, anticoagulants, benzodi-azepines, other psychopharmacological drugs, and chemotherapy within the last 30 days), smoking and alcohol consumption, and 1-day and 30-day mortality were recorded.

Primary end points were adverse effects related to ERCP, sedation, or both.

The results are reported as medians and interquar-tile ranges (median (IQR)) or number of patients and percentages. Logistic regression analysis was used to assess the risk of adverse effects. Forward stepping was used with p < 0.05 inclusion and p < 0.1 removal criteria (preselection from univariate with p < 0.1); however, age and gender were included in all the models. Statistical calculations were generated using IBM SPSS Statistics 21 (International Business Machines Corporation, Endicott, nY, USA).

RESULTS

We excluded 15 ERCPs performed in the operation theater and 17 ERCPs performed on patients under 18 years of age. In total, 956 patients with 1196 ERCPs were left in our analysis. Two gastroenterologists per-formed 253 ERCPs for cases with suspected or diag-nosed primary sclerosing cholangitis (PSC) and 4 gastrointestinal surgeons performed 943 ERCP proce-dures with other indications. All patients were in prone position during the procedure.

DEMOGRAPHICS

Median age of the 956 patients was 59.0 [25] years (range 18–100 years) and median body mass index (BMI) 24.8 [6.2] kg/m2, 529 (55%) were male and 427 (45%) female. The median length of the procedure was 23 [19] min. There were 299 (25%) procedures regarded as emergency ERCPs.

COMORBIDITIES

ASA classification of the patients was as follows (num-ber of patients (percentage)): ASA 1: 35 (2.9%), ASA 2:

Page 138: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

Routine preoperative laboratory testing for ERCP 3

417 (34.8%), ASA 3: 585 (48.7%), ASA 4: 160 (13.4%), and ASA 5: 2 (0.2%). Major comorbidities were as fol-lows: diabetes: 256 (21.4%), chronic hypertension 404 (33.8%), cardiovascular disease 204 (17.1%), and liver disease 331 (27.7%). Prevalence of other diseases was less than 10%. 191 (16.0%) of the patients were smok-ers.

ERCP DETAILS

ERCP was considered successful if the level of papilla or anastomosis was reached and the intended duct was successfully cannulated. Attempted ERCP was unsuccessful in 33 out of 1196 procedures. In 20 proce-dures, papilla was not reached or found (postopera-tive state (gastric resection, pancreaticoduodenectomy) (n = 5), obstruction proximal to papillary region (n = 6), edema of the descending duodenum (n = 8)) and one ERCP could not be performed due to patient’s allergy to sedatives. In 13 cases, papilla was reached, but the desired duct could not be cannulated. The endoscopic procedures included 1844 biliary tract interventions (biliary cytology, biliary sphincterotomy, biliary dila-tation, or stent application) and 503 pancreatic proce-dures (pancreatic cytology, pancreatic dilatation, or stent application). Majority of the patients (787, 82%) had only one ERCP, but 116 (12%) had two, 42 (4%) had three, and 11 (2%) had four or more ERCPs per-formed on them during the study period.

LABORATORY TEST RESULTS

Laboratory test results, listed here as median (IQR), followed by reference values were given as follows: hemoglobin (g/L): female 122 (20), 117–155, male 135 (29.5), 134–167. Platelet count (10E9/L): 243 (126), 150–360; potassium (P-K, mmol/L), 3.9 (0.53), 3.3–4.9; sodium (P-na): 139 (4), 137–145; thromboplastin time (P-TT, %): 95 (41), 70–130; and international normal-ized ratio (P-InR): 1.1 (0.2), 2–3 (patients on anticoag-ulants). Hypokalemia was found on 62 patients, most of which was mild and required no intervention. Hypokalemia was treated with intravenous potas-sium chloride before the procedure on 20 patients. P-InR or P-TT levels needed to be normalized with prothrombin complex concentrate on 44 patients before the procedure.

ADVERSE EFFECTS—ERCP

There were altogether 315 adverse events in 956 patients. Post-ERCP pancreatitis was defined as pain in the stomach, rise in the plasma amylase by three times the upper range limit, and the patient had to be hospitalized for 2 days because of pancreatitis. Pain in the stomach was experienced by 118 (9.9%) of patients. This was associated with already hospitalized patients (p = 0.012). Post-ERCP pancreatitis developed after 43 out of 1196 ERCPs, (3.6%). 24 of them were catego-rized as mild, 14 moderate, and 5 severe (5). Multivariate analysis (Table 1) showed that abnormal-ities in none of the RPLTs were associated with increased risk of post-ERCP pancreatitis.

Post-sphincterotomy bleeding (17 cases, 1.4%) and periampullary perforations (6 cases, 0.5%) were treated conservatively. no luminal perforation occurred. In addition, 12 patients (1%) with guide-wire-induced perforations received antibiotics. 17 (1.2%) patients had post-ERCP cholangitis, of which 2 patients (0.2%) were treated with antibiotics. In addi-tion, 19 miscellaneous adverse events (1.6%; pseudo-cyst infection, stent migration, stent rupture, peptic ulcer bleeding, air embolism, etc.) and 5 cardiopulmo-nary adverse events (0.4%; myocardial infarction and pulmonary embolism) occurred.

Two patients died the same day ERCP was per-formed (one due to air embolism caused by gas insuf-flation during endoscopy on a patient with a transplanted liver and one elderly patient because of acute myocardial infarction). In total, 10 patients (1%)

TABLE 1Univariate and multivariate analysis of risk factors of post-ERCP

pancreatitis.

p Odds ratio

95% CI

lower upper

UnivariateDiabetes, insulin treatment 0.053 0.140 0.019 1.024Diabetes, oral medication 0.132 0.216 0.030 1.587Hypertension 0.034 0.431 0.198 0.937Cardiovascular disease 0.329 0.625 0.243 1.607Opioid use 0.376 1.494 0.615 3.632Anticoagulant use 0.416 0.695 0.290 1.669Smoker 0.171 0.481 0.169 1.372Hospitalized patient 0.497 0.770 0.362 1.636Outpatient case 0.451 1.336 0.629 2.839P-Tromb < 150 0.065 0.154 0.021 1.125P-TT < 70% 0.030 0.206 0.049 0.860Abnormal ECG 0.158 0.596 0.291 1.222Hyponatremia 0.127 0.506 0.211 1.212Hypernatremia 0.647 0.626 0.084 4.657Hypokalemia 0.325 1.837 0.547 6.162Hyperkalemia 0.208 3.895 0.468 3.400Leukocytes/10 units 0.265 0.570 0.220 1.520Amylase (preoperative)a 0.357 1.43 0.67 3.05ALT/100 units 0.167 0.82 0.61 1.09ALP/100 units 0.177 0.91 0.78 1.05Bilirubine/100 units 0.857 0.97 0.74 1.29Bilirubine, conjugated/100 units 0.330 1.18 0.84 1.67ASA 1–2 versus 3–5 0.033 0.51 0.28 0.95Age 0.004 0.97 0.96 0.99Age > 60 years old 0.016 0.44 0.23 0.86Sex: female versus male 0.264 0.70 0.37 1.32Anemia 0.231 0.67 0.34 1.29Multivariate analysisAge > 60 years old 0.070 0.525 0.261 1.054Sex: female versus male 0.313 0.714 0.370 1.375Diabetes, insulin treatment 0.052 0.138 0.019 1.020P-TT < 70% 0.056 0.244 0.057 1.037

CI: confidence interval; ECG: electrocardiogram; ASA: American Society of Anesthesiology.aLog-transformed, pancreas-specific amylase.

Page 139: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

J. Jokelainen, et al.4

died within a week after the procedure, 8 patients with end stage cancer, 1 patient suffering from compli-cations of cholecystectomy died of pulmonary embo-lism 5 days after ERCP, and 1 elderly patient died of myocardial infarction on the third day after ERCP. The 30-day mortality was 44 (4.6%).

ADVERSE EFFECTS—SEDATIOn

Respiratory depression occurred with 11% of patients (128 of 1196). The multivariate analysis of respiratory depression (Table 2) showed no association with pre-operative laboratory tests. There was an interaction between anemia and cardiovascular disease in the sta-tistical model, and upon further analysis, anemia was found to be a stronger protective factor (p < 0.001, odds ratio (OR) = 0.26), if the patient had no cardiovas-cular disease.

Cardiovascular depression occurred in 7.2% of patients (86 of 1196). The multivariate analysis of car-diovascular depression (Table 3) showed that the risk of cardiovascular depression was increased if the patient had hyponatremia, thrombocytopenia, or was over 60 years old. There was an interaction between gender and hyponatremia in this statistical model, according to which hyponatremia was associated slightly more with cardiovascular depression on men than women.

In total, 20 (1.7%) patients had both cardiovascular and respiratory depression as defined in the study. The procedure had to be paused due to mask ventila-tion 20 times (1.7% of patients). 3 patients were intu-bated (one case of air embolism leading to resuscitation, one case of laryngobronchospasm, and one ICU patient with septic cholecystitis, already intubated upon arrival to endoscopy room). Other sedation-related adverse events were not observed. Statistical analysis of the risk of mask ventilation or intubation was not feasible due to the small number of cases.

DISCUSSIOn

It has been estimated that ERCP is needed in about 50–100 per 100,000 persons per year. As more thera-peutic options become available, the need will most likely rise (6, 7). As the need for ERCP rises, the eco-nomic burden of preoperative testing will also rise. In our institution, the RPLT package costs about US$49 (€41) on one elective patient. In case of emergency patients, the costs of same package are significantly higher, about US$79 (€66). If we had refrained from taking RPLTs from patients not considered to be at risk for adverse effects or abnormal laboratory test results based on clinical judgment, based on the data gath-ered for this study, we would not have taken RPLTs from 805 patients, which translates to savings of at least US$40,000 (€33,000), assuming they were elective procedures. Systematic preoperative laboratory screening will most likely lead to excessive testing without any true impact on clinical decisions as has been shown by numerous studies and guidelines (1–3, 8). While most such studies are focused on surgery, comparable results are also found on studies on endos-

TABLE 2Univariate and multivariate analysis of risk factors of respiratory

depression.

p Odds ratio 95% CI

Lower Upper

UnivariateDiabetes, insulin treatment 0.110 1.477 0.916 2.380Diabetes oral medication 0.830 1.069 0.581 1.965Hypertension 0.502 1.139 0.778 1.668Cardiovascular disease 0.014 1.721 1.117 2.651Cerebrovascular disease 0.669 1.308 0.382 4.484Atherosclerosis 0.800 1.147 0.397 3.316COPD 0.216 0.405 0.097 1.695Kidney failure 0.502 0.609 0.143 2.590Endocrinological disease 0.188 1.753 0.760 4.044Opioid use 0.277 0.688 0.351 1.350Anticoagulant use 0.409 1.210 0.769 1.904Benzodiazepine use 0.656 0.629 0.082 4.846Smoker 0.564 0.843 0.472 1.505Hospitalized patient 0.223 0.760 0.488 1.183Outpatient case 0.076 1.518 0.957 2.407Thrombocytopenia 0.855 0.950 0.546 1.651P-TT < 70% 0.300 1.264 0.812 1.967P-InR > 2 0.432 0.648 0.219 1.914Abnormal ECG 0.287 1.229 0.841 1.797Hyponatremia 0.747 0.931 0.601 1.441Hypernatremia 0.853 1.094 0.423 2.832Hypokalemia 0.691 1.194 0.498 2.867Hyperkalemia 0.870 1.192 0.145 9.772Leukocytes/10units 0.340 1.24 0.80 1.92Amylase (preoperative)a 0.030 0.59 0.36 0.95ALT/100 units 0.068 0.87 0.74 1.01ALP/100 units 0.276 0.96 0.90 1.03Bilirubine/100 units 0.240 0.89 0.74 1.08Bilirubine, conjugated/100 units

0.240 0.86 0.67 1.11

ASA 1–2 versus 3–5 0.169 1.32 0.89 1.95Age > 60 years old 0.506 1.13 0.78 1.63Sex: female versus male 0.205 1.27 0.88 1.83Anemia <0.001 0.44 0.29 0.68Multivariate, all patientsAge > 60 years old 0.686 1.09 0.72 1.64Sex: female versus male 0.840 1.04 0.71 1.53Cardiovascular disease 0.003 2.07 1.29 3.34Anemia 0.000 0.39 0.25 0.61Patients without cardiovascular diseaseAge > 60 years old 0.732 1.08 0.69 1.69Sex: female versus male 0.976 0.99 0.64 1.54Anemia 0.000 0.26 0.14 0.48Patients with cardiovascular diseaseAge > 60 years old 0.517 1.45 0.47 4.51Sex: female versus male

0.483 1.34 0.59 3.02

Anemia 0.706 0.86 0.38 1.92

CI: confidence interval; COPD: chronic obstructive pulmonary disease; ECG: electrocardiogram; ASA: American Society of Anesthesiology.A significant interaction was found in between cardiovascular disease and anemia; therefore, a split analysis was performed according to cardiovascular disease.aLog-transformed, pancreas-specific amylase.

Page 140: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

Routine preoperative laboratory testing for ERCP 5

copy, even though they are fewer in number. Also, one could assume that an endoscopic procedure such as ERCP would cause less stress on the patient and there-fore require even less preoperative routine testing than patients undergoing surgery. Some RPLT results did indeed have an impact on the treatment of these patients, such as correcting hypokalemia or coagula-tion levels with prothrombin complex concentrate. However, the vast majority with only a few excep-tions, these patients had a health condition or medica-tion that would have warranted laboratory testing regardless of RPLT.

The rate of adverse effects of ERCP (9, 10) and seda-tion (11, 12) in this study was similar to other studies on the subject. This would lend credence to the notion that the results of this study should be applicable out-side the Endoscopy Unit, in which this study was con-ducted.

There are some limitations to our study. First, being a single-center study, its applicability to other institu-tions with less experience with treating these patients may be limited. Also, the relatively small number of adverse effects prevents making solid conclusions on the matter, but does provide a foundation for possible further studies. There were 15 ERCPs performed in the operating theater instead of the Endoscopy Unit. The reason for this was because the patients were con-sidered too unstable for sedation and required gen-eral anesthesia and more stringent monitoring than was available in the endoscopy unit. naturally, this will create a possibility for bias, but the total number of these procedures is low, and the effect should be minimal.

We did not differentiate between acute and elective setting of ERCP. The rationale behind this was that RPLT is more relevant in the elective setting since patients having acute ERCP are already having labo-ratory testing done on them because of the underlying medical condition. When considering this—and the fact that patients receiving acute ERCP are more prone to adverse effects and abnormal laboratory test results—we should not expect the results to be biased against RPLT when considering all the patients as a single group, since RPLT is the focus of this study.

This study found that none of the RPLTs that were used were directly associated with any adverse effects for ERCP. There was an association with RPLT and adverse effects related to sedation with thrombocyto-penia and hyponatremia and cardiovascular depres-sion. However, the causal link between these findings is unclear and does warrant further studies. Both hyponatremia and thrombocytopenia are usually caused by some other health-related issues, and it is these root causes which are most likely responsible for hypotension during sedation. Simply correcting the values would most likely not alleviate the problem. Also, a clinical assessment of the patient before the procedure would probably inform the anesthesiolo-gist of the risk of cardiovascular depression even without the preoperative laboratory testing. Therefore, it would seem that the policy of RPLT is not positively affecting the patients’ treatment and care and is not economically sound. That is not to say preoperative testing should be abandoned completely, but should

TABLE 3Univariate and multivariate analysis of risk factors of cardiovascular

depression.

p Odds ratio

95% CI

Lower Upper

UnivariateDiabetes, insulin treatment 0.355 1.316 0.735 2.357Diabetes, oral medication 0.291 1.431 0.736 2.781Hypertension 0.175 1.363 0.871 2.134Cardiovascular disease 0.000 2.854 1.780 4.576Cerebrovascular disease 0.218 2.183 0.630 7.566Atherosclerosis 0.003 3.664 1.542 8.704COPD 0.004 3.219 1.441 7.190Kidney failure 0.037 2.874 1.065 7.760Endocrinological disease 0.217 1.835 0.700 4.816Opioid use 0.500 1.56 0.648 2.434Anticoagulant use 0.002 2.162 1.335 3.502Benzodiazepine use Chemotherapy < 30 days ago Smoker 0.130 1.585 0.874 2.874Hospitalized patient 0.080 1.516 0.952 2.414Outpatient case 0.146 0.706 0.441 1.129P-Tromb < 150 0.004 2.194 1.289 3.734P-TT < 70% 0.002 2.151 1.325 3.492P-InR > 2.0 0.072 0.367 0.123 1.096Abnormal ECG 0.001 2.121 1.365 3.297Hyponatremia 0.000 2.641 1.684 4.142Hypernatremia Hypokalemia 0.388 1.522 0.587 3.947Hyperkalemia Leukocytes/10 units 0.085 1.52 0.94 2.44Amylase (preoperative)a 0.443 0.81 0.48 1.38ALT/100 units 0.084 1.09 0.99 1.21ALP/100 units 0.435 1.03 0.96 1.09Bilirubine/100 units 0.000 1.49 1.30 1.71Bilirubine, conjugated/100 units

0.000 1.52 1.28 1.82

ASA 1–2 versus 3–5 0.000 3.70 2.02 6.75Age > 60 years old 0.000 4.06 2.41 6.85Sex: female versus male 0.805 0.95 0.61 1.47Anemia 0.005 1.89 1.22 2.94Multivariate analysis, all patientsAge > 60 years old 0.000 3.34 1.95 5.72Sex: female versus male 0.468 0.84 0.53 1.34Atherosclerosis 0.053 2.41 0.99 5.89P-Tromb < 150 0.025 1.87 1.08 3.25Hyponatremia 0.001 2.22 1.40 3.52Male patientsAge > 60 years old 0.000 4.79 2.23 10.30Atherosclerosis 0.088 2.58 0.87 7.70P-Tromb < 150 0.164 1.69 0.81 3.53Hyponatremia 0.000 3.66 1.98 6.75Female patientsAge > 60 years old 0.050 2.14 1.00 4.60Atherosclerosis 0.263 2.53 0.50 12.89P-Tromb < 150 0.078 2.14 0.92 4.99Hyponatremia 0.823 1.09 0.51 2.32

CI: confidence interval; COPD: chronic obstructive pulmonary disease; ECG: electrocardiogram; ASA: American Society of Anesthesiology.A significant interaction was found in between gender and hyponatremia; therefore, a split analysis was performed according to gender.aLog-transformed, pancreas-specific amylase.

Page 141: RISK FACTORS ASSOCIATED TO ENDOSCOPIC RETROGRADE

J. Jokelainen, et al.6

be focused on those patients that would benefit from them. Interestingly, anemia was found to be a protect-ing factor for respiratory depression caused by seda-tion, especially on patients without cardiovascular disease. It could be that anemia causes sensitization to hypoxemia and thus leads to pronounced reactivity and increased respiratory drive when the patient is sedated, and respiratory center is affected by the seda-tive agents. However, clinical utilization of this find-ing, should it prove to be factual in possible further studies, is somewhat difficult, since anemia is some-thing to be avoided when possible in clinical practice.

In conclusion, while it is not possible to give precise recommendations on RPLTs based on our study, we would venture to suggest the following general rules based on clinical experience and generally accepted med-ical practices as shown in Table 4. Other laboratory tests should be prescribed by the treating physician’s delibera-tion based on the patient’s individual health status.

ACKnOWLEDGEMEnTS

J.J. and S.I. contributed equally to this paper. This work was carried out in the Endoscopy Unit, Helsinki University Hospital, Helsinki, Finland.

DECLARATIOn OF COnFLICTInG InTERESTS

The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.

FUnDInG

The author(s) received no financial support for the research, authorship, and/or publication of this article.

REFEREnCES 1. Czoski-Murray C, Lloyd Jones M, McCabe C et al: What is the

value of routinely testing full blood count, electrolytes and

urea, and pulmonary function tests before elective surgery in patients with no apparent clinical indication and in subgroups of patients with common comorbidities: A systematic review of the clinical and cost-effective literature. Health Technol Assess 2012;16(50):1–159.

2. Pasha SF, Acosta R, Chandrasekhara V et  al: Routine labora-tory testing before endoscopic procedures. Gastrointest Endosc 2014;80(1):28–33.

3. Pasternak LR: Preoperative laboratory testing: General issues and considerations. Anesthesiol Clin n Am 2004;22(1):13–25.

4. Cotton PB, Garrow DA, Gallagher J et al: Risk factors for com-plications after ERCP: A multivariate analysis of 11,497 proce-dures over 12 years. Gastrointest Endosc 2009;70(1):80–88.

5. Cotton PB, Lehman G, Vennes J et al: Endoscopic sphincterot-omy complications and their management: An attempt at con-sensus. Gastrointest Endosc 1991;37(3):383–393.

6. Gear MW, Dent nA, Colin-Jones DG et  al: Future needs for ERCP: Incidence of conditions leading to bile duct obstruction and requirements for diagnostic and therapeutic biliary proce-dures. Gut 1990;31(10):1150–1155.

7. Coelho-Prabhu n, Shah nD, Van Houten H et  al: Endo-scopic retrograde cholangiopancreatography: Utilisation and outcomes in a 10-year population-based cohort. BMJ Open 2013;3(5): e002689.

8. Rotzen MF: A prospective evaluation of the value of preopera-tive laboratory testing for office anesthesia and sedation. J Oral Maxillofac Surg 1999;57(1):21–22.

9. Andriulli A, Loperfido S, napolitano G et al: Incidence rates of post-ERCP complications: A systematic survey of prospective studies. Am J Gastroenterol 2007;102(8):1781–1788.

10. Cohen SA, Siegel JH, Kasmin FE: Complications of diagnostic and therapeutic ERCP. Abdom Imaging 1996;21(5):385–394.

11. nilsson A, Grossmann B, Kullman E et  al: Sedation during endoscopic retrograde cholangiopancreatography: A rand-omized controlled study of patient-controlled propofol seda-tion and that given by a nurse anesthetist. Scand J Gastroenterol 2015;50(10):1285–1292.

12. Han SJ, Lee TH, Park S et  al: Efficacy of midazolam- versus propofol-based sedations by non-anesthesiologists during ther-apeutic endoscopic retrograde cholangiopancreatography in patients aged over 80 years. Dig Endosc 2017;29(3):369–376.

Received: February 21, 2018Accepted: December 4, 2018

TABLE 4.Suggested laboratory tests before ERCP.

Medical condition Suggested laboratory test

ASA 4–5 BBC, ECG, creatinine, P-na, P-KCoronary artery disease, atherosclerosis, kidney failure, use of diuretics BBC, ECG, creatinine, P-na, P-Koral anticoagulant use (warfarin), liver disorder P-InR

ASA: American Society of Anesthesiologists physical status classification, BBC: basic blood count, ECG: electrocardiogram, P-na: plasma sodium, P-K: plasma potassium; P-InR: international normalized ratio.