Jjae 089

Download as pdf or txt
Download as pdf or txt
You are on page 1of 71

Manuscript Doi: 10.

1093/ecco-jcc/jjae089
ECCO Guidelines on Therapeutics in Crohn's Disease:

Surgical Treatment

Michel Adamina1,2, Silvia Minozzi, Janindra Warusavitarne, Christianne Buskens, Maria

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Chaparro, Bram Verstockt, Uri Kopylov, Manasi Agrawal, Mariangela Allocca, Raja Atreya,
Robert Battat, Dominik Bettenworth, Gabriele Bislenghi, Steven Ross Brown, Johan Burisch,
María José Casanova, Wladyslawa Czuber-Dochan, Joline de Groof, Alaa El-Hussuna,

t
ip
Pierre Ellul, Catarina Fidalgo, Gionata Fiorino, Javier Gisbert, João Guedelha Sabino, Jurij
Hanzel, Stefan Holubar, Marietta Iacucci, Nusrat Iqbal, Christina Kapizioni, Konstantinos

cr
Karmiris, Taku Kobayashi, Paulo Gustavo Kotze, Gaetano Luglio, Christian Maaser, Gordon
Moran, Nurulamin Noor, Konstantinos Papamichail, Georgios Peros, Catherine Reenaers,

us
Giuseppe Sica, Rotem Sigall-Boneh, Stephan R. Vavricka, Henit Yanai, Tim Raine, Hannah
Gordon, Pär Myrelid
an
Michel Adamina 1. Department of Surgery, Cantonal Hospital of Fribourg & Faculty of Science and
M

Medicine, University of Fribourg, Fribourg, Switzerland


2. Department of Visceral and Thoracic Surgery, Kantonsspital Winterthur,
Winterthur, Switzerland
d

Silvia Minozzi Department of Epidemiology, Lazio Regional Health Service, Rome, Italy
Janindra St Mark’s Hospital London; United Kingdom
e

Warusavitarne
Christianne Department of Surgery, Amsterdam UMC, Location VUMC, Amsterdam, The
pt

Johanna Buskens Netherlands


Maria Chaparro Gastroenterology Department. Hospital Universitario de La Princesa, Instituto de
ce

Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid


(UAM), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y
Digestivas (CIBEREHD), Madrid; Spain
Bram Verstockt Department Gastroenterology & Hepatology, University Hospitals Leuven, KU
Ac

Leuven and Dpt. Chronic Diseases and Metabolism, KU Leuven, Leuven, Belgium
Uri Kopylov Department of Gastroenterology, Sheba Medical Center, Ramat Gan, Israel
Henit Yanai IBD Center, Division of Gastroenterology, Rabin Medical Center, Petah Tikva;
Faculty of Medicine, Tel Aviv University, Tel Aviv; Israel
Stephan R. Department of Gastroenterology and Hepatology, University Hospital Zürich,
Vavricka Zürich, Switzerland
Rotem Sigall- Pediatric Gastroenterology and Nutrition Unit, The E. Wolfson Medical Center,
Boneh Holon, Israel.
Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology
Endocrinology and Metabolism, University of Amsterdam, Amsterdam, the
Netherlands
Giuseppe S. Sica Department of Surgery, Università Tor Vergata, Roma, Italy
© The Author(s) 2024. Published by Oxford University Press on behalf of European Crohn’s and
Colitis Organisation. All rights reserved. For permissions, please email:
[email protected]
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Catherine Gastroenterology Department, Chu Liege, Liege, Belgium
Reenaers
Georgios Peros Department of Surgery, Cantonal Hospital Winterthur, Winterthur, Switzerland
Konstantinos Center for Inflammatory Bowel Diseases, Division of Gastroenterology, Beth
Papamichael Israel Deaconess Medical Center, Harvard Medical School, Boston,
Massachusetts
Nurulamin Noor Department of Medicine, University of Cambridge, Cambridge, United Kingdom
Gordon William 1. National Institute of Health Research Nottingham Biomedical Research

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Moran Centre, University of Nottingham and Nottingham University Hospitals,
Nottingham
2. Translational Medical Sciences, School of Medicine, Faculty of Medicine
and Health Sciences, University of Nottingham, Nottingham. NG7 2UH. United

t
Kingdom

ip
Christian Maaser Outpatients Department of Gastroenterology, University Teaching Hospital
Lueneburg, Germany

cr
Gaetano Luglio Department of Public Health, University of Naples Federico II, Naples, Italy
Paulo Gustavo Health Sciences Postgraduate Program, Pontificia Universidade Católica do
Kotze Paraná (PUCPR), Curitiba, Brazil

us
Taku Kobayashi Center for Advanced IBD Research and Treatment, Kitasato University Kitasato
Institute Hospital, Tokyo, Japan
Konstantinos Department of Gastroenterology, Venizeleio General Hospital, Heraklion, Greece
an
Karmiris
Christina Department of Gastroenterology, Attikon University Hospital, Athens, Greece
Kapizioni
Nusrat Iqbal Department of Surgery, Worcestershire Acute Hospitals NHS Trust, Worcester,
M

UK.
Marietta Iacucci APC Microbiome Ireland, College of Medicine and Health, University College of
Cork, Cork, Ireland
d

Stefan Holubar Department of Colon & Rectal Surgery, Cleveland Clinic, Cleveland, Ohio, USA
Jurij Hanzel Department of Gastroenterology, University Medical Centre Ljubljana, Ljubljana;
e

Chair of Internal Medicine, Faculty of Medicine, University of Ljubljana,


Ljubljana, Slovenia
pt

João Guedelha Department of Gastroenterology and Hepatology, University Hospitals Leuven,


Sabino Leuven, Belgium
ce

Javier P. Gisbert Gastroenterology Department. Hospital Universitario de La Princesa, Instituto de


Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid
(UAM), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y
Digestivas (CIBEREHD), Madrid, Spain
Ac

Gionata Fiorino IBD Unit, San Camillo-Forlanini Hospital, Rome, Italy


Catarina Fidalgo Division of Gastroenterology, Hospital Beatriz Ângelo, Loures
Division of Gastroenterology, Hospital da Luz, Lisboa
Portugal
Pierre Ellul Division of Gastroenterology, Mater Dei Hospital, Malta
Alaa El-Hussuna OpenSourceResearch Organization (OSRC.Network), Aalborg, Denmark
Joline de Groof Colorectal Surgery, Royal Surrey NHS Foundation Trust, Guildford, UK
Wladyslawa Florence Nightingale Faculty of Nursing- Midwifery and Palliative Care, King’s
Czuber-Dochan College London, London, United Kingdom
María José Gastroenterology Department. Hospital Universitario de La Princesa, Instituto de
Casanova Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid
(UAM), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Digestivas (CIBEREHD), Madrid. Spain
Johan Burisch Gastrounit, Medical Division, Copenhagen University Hospital - Amager and
Hvidovre, Hvidovre; Copenhagen Center for Inflammatory Bowel Disease in
Children, Adolescents and Adults, Copenhagen University Hospital - Amager and
Hvidovre, Hvidovre, Denmark
Steven Ross Department of Surgery, Sheffield Teaching Hospitals, Sheffield UK
Brown
Gabriele Department of Abdominal Surgery, University Hospitals Leuven, Belgium

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Bislenghi
Dominik CED Schwerpunktpraxis, Münster and Medical Faculty of the University of
Bettenworth Münster, Münster, NRW, Germany
Robert Battat Division of Gastroenterology, Centre Hospitalier de l’Université de Montréal,

t
Montreal, Canada

ip
Raja Atreya First Department of Medicine, Friedrich-Alexander-University Erlangen-
Nürnberg, Erlangen, Germany.

cr
Mariangela IRCCS Hospital San Raffaele and University Vita-Salute San Raffaele,
Allocca Gastroenterology and Endoscopy, Milan, Italy
Manasi Agrawal 1. The Dr. Henry D. Janowitz Division of Gastroenterology, Icahn School of

us
Medicine at Mount Sinai, New York, New York, USA
2. Center for Molecular Prediction of Inflammatory Bowel Disease (PREDICT),
Department of Clinical Medicine, Aalborg University, Copenhagen, Denmark
an
Tim Raine Department of Gastroenterology, Addenbrooke’s Hospital, Cambridge University
Hospitals NHS Foundation Trust, Cambridge, United Kingdom
Hannah Gordon Translational Gastroenterology and Liver Unit, University of Oxford, Oxford, UK
Pär Myrelid Department of Surgery and Department of Biomedical and Clinical Sciences,
M

Linköping University, Linköping, Sweden


e d

Corresponding author: Prof. Dr. Michel Adamina, MD, PD, MSc, EMBA HSG, FEBS,
FASCRS, Chairman, Department of Surgery, Cantonal Hospital Fribourg, Chemin des
pt

Pensionnats 2/6, 1752 Villars-sur-Glâne, Switzerland. Tel. +41 26 306 25 10; Email
[email protected]
ce
Ac
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Abstract

This article is the second in a series of two publications on the European Crohn’s and Colitis
Organisation [ECCO] evidence-based consensus on the management of Crohn’s disease.
The first article covers medical management; the present article addresses surgical
management, including preoperative aspects and drug management before surgery. It also
provides technical advice for a variety of common clinical situations. Both articles together
represent the evidence-based recommendations of the ECCO for Crohn’s disease and an

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


update of prior ECCO guidelines.

Keywords: Crohn’s disease; Surgery; Inflammatory bowel disease (IBD)

t
ip
cr
us
an
M
e d
pt
ce
Ac
Manuscript Doi: 10.1093/ecco-jcc/jjae089
1. Introduction

The incidence and prevalence of Crohn’s disease [CD] is on the rise globally, with increases
in incidence ranging from 4–15% yearly over the last three decades.1 CD is a lifelong disease
and optimal management is multidisciplinary and interprofessional and has become
increasingly complex. Surgery is a major therapeutic avenue in this context. Indeed, half of

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


patients with CD undergo one or more operations during their lifetime. Patients with CD often
suffer from malnutrition, psychological comorbidities, and may have to accept and live with a
stoma.2-5 Many different medications and combinations thereof are reshaping clinical practice,

t
while refined surgical techniques, tailored approaches, and a wider acceptance of a surgical

ip
alternative benefit patients. Hence, the best possible outcomes are currently achieved within
dedicated expert centres providing personalized medicine.6-10 The European Crohn’s and

cr
Colitis Organisation [ECCO] provides an interdisciplinary framework with these evidence-
based guidelines to inform and guide practice and clinicians caring for patients with CD. The

us
present guidelines focus on surgery for CD, including pre- and perioperative aspects, and
provides technical advice for a variety of common clinical presentations. Further, ECCO
an
guidelines offer guidance on most aspects of interdisciplinary and interprofessional care for
CD in separate publications.11-16
M

2. Methods
d

A detailed description of the methodology used is presented in the supplementary materials.


e

This article is the second in a series of two publications on the ECCO evidence-based
pt

consensus on the management of CD. The first article covered medical management; 17 the
present article is focused on surgical management while covering both medical and surgical
ce

management of perianal CD. These two articles together represent the evidence-based
recommendations of the ECCO for CD and update prior guidelines published in 2020.18,19 The
Ac

present guidelines follow the GRADE methodology in terms of framing clinically relevant
questions to draw evidence-based statements and recommendations. However, due to the
peculiarities of the surgical literature, appraisal of the systematically researched literature was
conducted according to the Oxford Centre for Evidence-Based Medicine, which grades from
evidence level [EL]1: systematic review of randomized controlled trials to EL5: expert
opinion.20 This allowed us to formulate statements and practice recommendations that can
effectively inform and guide clinical management.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
3. Perianal Crohn’s disease

3.1. Medical approaches

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Statement 3.1 ECCO CD Treatment GL [2024]

We do not recommend use of antibiotics as monotherapy for treatment of complex

t
perianal fistulae in patients with CD [EL4]

ip
cr
Although antibiotics are widely used in the treatment of perianal CD, most available studies

us
are uncontrolled.21 To our knowledge, only one randomized controlled trial [RCT] compared
placebo with antibiotics in perianal fistulae [supplementary table 1]. Remission at week 10
was observed in 1/8 [12.5%] versus 3/17 [17.6%] patients treated with placebo or antibiotics,
an
respectively (relative risk [RR]: 1.41; 95% confidence interval [CI]: 0.17–11.54]. Complete
healing was observed in 3/10 [30%] patients treated with ciprofloxacin and 0/8 patients
M

treated with metronidazole.22 Uncontrolled data and data from studies on combination
therapy with anti-TNF suggest that ciprofloxacin can improve the efficacy of anti-TNF in the
short term with good safety but with no impact on longer-term healing rates.23,24 Importantly,
d

despite the lack of evidence to support their role as monotherapy in closing perianal fistulae,
e

antibiotics are indicated and recommended to treat and control perianal sepsis.
pt
ce

Statement 3.2 ECCO CD Treatment GL [2024]


Ac

We suggest against using thiopurines as monotherapy [azathioprine, mercaptopurine]


for treatment of complex perianal fistulae in patients with CD [EL3]

The effect of azathioprine [AZA] on fistula healing in perianal CD has been numerically
reported in RCTs in 18 patients only.25-28 A meta-analysis on this limited group of patients
demonstrated that AZA is not superior to placebo for fistula healing [RR: 2.00; 95% CI: 0.67–
5.93].29 Another study reported complete fistula closure in 9/29 [31%] fistulae during
Manuscript Doi: 10.1093/ecco-jcc/jjae089
mercaptopurine therapy, in contrast to 1/17 [6%] with placebo-treated fistulae30
[supplementary table 2]. Nevertheless, these data could not be incorporated in the pooled
analysis, as they were reported as number of fistulae closing rather than number of patients
who had complete fistulae closing. With the availability of effective anti-TNF agents, it seems
inappropriate to recommend any further randomized placebo-controlled trial studying the
efficacy of AZA in complex perianal fistulae.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Statement 3.3 ECCO CD Treatment GL [2024]

t
ip
We recommend infliximab for the induction and maintenance of remission in complex
perianal fistulae in CD [EL2]

cr
us
Infliximab was the first agent shown to be effective in a RCT for inducing closure of perianal
fistulae and for maintaining this response over 1 year. Complete response [defined as the
an
absence of any draining fistulae at two consecutive visits at least 4 weeks apart] was
observed in 4/31 [12.9%] placebo-treated patients versus 29/63 [46%] infliximab-treated
M

patients [RR: 3.57; 95% CI: 1.38–9.25].31 Subsequently, the ACCENT II trial evaluated the
efficacy of infliximab [5 mg/kg every 8 weeks] in a maintenance trial in 195 patients who had
a response [defined as a reduction of 50% of draining fistulae in two visits at least 4 weeks
d

apart] at week 14 after open-label induction treatment with infliximab. A complete response
e

was maintained until week 54 in 19/99 [19.2%] placebo-treated patients versus 33/96
pt

[34.4%] infliximab-treated patients [RR: 1.79; 95% CI: 1.10– 2.92].32 A recent meta-analysis
of the existing data revealed that infliximab was effective in inducing [RR: 3.57; 95%
ce

CI:1.38–9.25] and maintaining clinical fistula healing [RR: 1.79; 95% CI:1.10–2.92] 33 with no
significant risk of serious AEs as compared with placebo [RR: 1.31; 95% CI: 0.11–15.25,
Ac

supplementary figure 1]. A combined evaluation of both RCTs for safety revealed a risk of
serious AEs of 18.9% [33/175 patients] in the placebo groups versus 11.9% [24/201
patients] in the infliximab groups. Overall, the most recent meta-analysis (2023) provided low
certainty on clinical outcomes. Some retrospective data suggest that fistula healing is more
likely in patients with higher infliximab trough levels, suggesting the need for personalized
dosing in this setting.34-38
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Statement 3.4 ECCO CD Treatment GL [2024]

We suggest use of adalimumab for induction and maintenance of remission in


complex perianal fistulae in CD [EL3]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Fistula healing in the subgroup of patients with enterocutaneous or perianal fistulae [or both]
at baseline [n = 117] was a secondary endpoint of the CHARM double-blind, placebo-
controlled, randomized trial.39 A subsequent post-hoc analysis that focused specifically on

t
the efficacy of adalimumab over time in this subgroup confirmed the superiority of

ip
adalimumab over placebo [RR: 2.57; 95% CI: 1.13–5.84] for fistula healing after 56 weeks39
[supplementary table 3]. Data from CHARM combined with data from the open-label

cr
extension study ADHERE revealed that there was no significant increase in serious AEs for

us
patients treated with adalimumab [RR: 1.21; 95% CI: 0.43–3.38].40-43 Data were insufficient
to ascertain maintenance of fistula healing beyond 56 weeks, resolution of perianal sepsis,
stoma-free survival, and quality of life. In a retrospective multicentre analysis evaluating 46
an
patients [83% with complex fistula] naïve to anti-TNF therapy, 72% of patients responded to
adalimumab [54% remission, 18% partial response] at 6 months and 49% of patients
M

maintained response at 12 months [41% remission, 8% partial response].44 Additional data


suggested that adalimumab may have a role in patients who failed infliximab because of
immunogenicity [either primary non-responders or secondary loss-of-response]. The open-
d

label CHOICE trial indeed demonstrated that complete fistula healing [mainly perianal fistula]
e

could be achieved in 39% [34/88] of patients who initiated adalimumab after infliximab
pt

failure.42 This finding has also been reported in a limited case series.41 Some retrospective
data suggest that fistula healing is more likely in patients with higher adalimumab trough
ce

levels, suggesting the need for personalized dosing in this setting.35,37,40,45


Ac

Statement 3.5 ECCO CD Treatment GL [2024]

There is insufficient evidence to recommend use of certolizumab pegol as a treatment


for complex perianal fistulae in patients with CD [EL4]
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Certolizumab pegol [CZP], a pegylated humanized Fab' fragment that targets TNF-α, was
evaluated for treatment of CD in two RCTs [PRECISE 1 and PRECISE 2]. The PRECISE 1
study included 662 patients with moderate-to-severe CD who were randomly assigned to
receive either CZP 400 mg or placebo subcutaneously at weeks 0, 2, and 4, followed by
administration every 4 weeks up to week 26.46 Fistula closure was a secondary endpoint;
30% [14/46] of patients in the CZP group achieved closure versus 31% [19/61] in the

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


placebo group. According to this study, CZP did not show a significant benefit for fistula
closure.

t
The PRECISE 2 trial included 668 adults with moderate-to-severe CD47 and used the same

ip
induction therapy as in PRECISE 1. Patients with clinical response [reduction of ≥100 from
baseline score on the Crohn’s disease activity index] were randomly assigned to receive

cr
CZP 400 mg or placebo every 4 weeks through week 26. Among patients responding to
induction therapy with CZP, 28 of those randomized to CZP and 30 of those randomized to

us
placebo had draining fistulae at baseline. The primary endpoint of the fistula subanalysis
was fistula closure, defined as ≥50% closure at two consecutive post-baseline visits ≥3
an
weeks apart. At week 26, 54% [15/28] of CZP-treated patients achieved fistula closure [per
protocol] compared with 43% [13/30] of placebo-treated patients; the difference was not
M

statistically significant [p = 0.069]. At week 26, 36% of patients in the CZP group achieved
complete fistula closure compared with 17% in the placebo group [p = 0.038]. Among
patients who achieved the predefined criteria for fistula closure, there was a higher
d

numerical proportion of patients who received continuous treatment with CZP compared with
e

those who initially underwent induction therapy followed by placebo. However, these
pt

differences were not statistically significant for the small sample size analysed. Patients
randomized to CZP in the maintenance phase maintained a 50% fistula closure rate at week
ce

26 (11/15 [73%] patients vs 39% [5/15] patients; p = 0.069) and achieved 100% closure at
week 26 (10/15 [67%] patients vs 4/13 [31%] patients; p = 0.064). The results from these
post-hoc analyses suggest a possible effect of CZP in complex perianal fistulae in CD.
Ac

However, possibly due to limited sample size, the benefit of CZP over placebo was not
demonstrated.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Statement 3.6 ECCO CD Treatment GL [2024]

There is insufficient evidence to recommend use of vedolizumab for the treatment of


complex perianal fistulae in CD [EL4]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Vedolizumab [VDZ], a gut-selective α4β7 integrin antibody, was assessed for the treatment
of complex perianal fistulae in an exploratory analysis of data from the GEMINI 2 study. 48
GEMINI 2 was a phase 3, randomized, double-blind, placebo-controlled trial that consisted

t
of separate induction and maintenance phases. Following a 6-week induction period with

ip
VDZ, responders were randomly assigned to receive either placebo [VDZ/placebo group] or
VDZ [VDZ/VDZ group] and entered a maintenance phase. Fistula closure was defined as the

cr
absence of clinically draining fistulae at weeks 14 and 52. A total of 57 patients with draining

us
fistulae at the start of the maintenance period were included in the analysis; half of them
previously failed anti-TNF therapy. By week 14, 28% [11/39] of patients in the VDZ/VDZ
group and 11% [2/18] of patients in the VDZ/placebo group achieved fistula closure.
an
However, in a meta-analysis, maintenance with VDZ did not reach statistical significance
[RR: 2.54; 95% CI: 0.63–10.29; p = 0.19].49 At week 52, 31% in the VDZ/VDZ group and
M

11% in VDZ/placebo group had fistula closure. Despite the numerically greater proportion of
fistula healing observed in patients treated with VDZ, no statistically significant differences
were observed. This post hoc analysis has several limitations, including a small sample size
d

and inadequate statistical power. It is also biased by the induction phase with VDZ and lacks
e

a design specifically evaluating VDZ for fistula closure.


pt

A small clinical trial compared the efficacy of standard VDZ dosing versus standard dosing
plus an additional dose at week 10 in patients with ≥1 draining perianal fistula at baseline. 50
ce

Fistula closure was observed at week 30 in 12 [42.9%] patients (7 patients in the standard
and 5 patients in the additional VDZ dose group).
Ac

In summary, the available evidence is of low quality and insufficient to recommend VDZ for
complex perianal fistulae in patients with CD. However, VDZ could be considered in patients
refractory or intolerant to anti-TNF therapy. Further studies with appropriate design are
warranted to determine the benefit of VDZ in the treatment of complex perianal fistulae.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Statement 3.7 ECCO CD Treatment GL [2024]

There is insufficient evidence to recommend use of ustekinumab as a treatment for


complex perianal fistulae in CD [EL4]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


The sole study comparing ustekinumab to placebo in treating complex perianal fistulae was
a post hoc pooled analysis of data from the phase 2 CERTIFI and from the phase 3 UNITI-1
and UNITI-2 trials. This analysis provided information on the induction of fistula response

t
and remission rates.51 A total of 150 patients were treated with ustekinumab and 71 were

ip
treated with placebo. Due to the limited sample size, data from the final induction visit at
week 8 were aggregated across the three studies for evaluation. The analysis revealed a

cr
higher proportion of fistula closure after 8 weeks of treatment in the ustekinumab group

us
[24.7%] compared with the placebo group [14.1%], although the observed difference did not
reach statistical significance [p = 0.073]. This finding was confirmed in a meta-analysis [RR:
1.77; 95% CI: 0.93–3.37].49
an
In the maintenance phase, fistula response to treatment was assessed at week 22 and 44.
However, all patients included in the maintenance phase were either responders or non-
M

responders to induction with ustekinumab who were re-randomized to receive ustekinumab


or placebo, which may bias the results. Among patients in the maintenance phase, fistula
d

response at week 22 occurred in 9/19 [47%] patients in the ustekinumab group and in 6/20
e

[30%] patients in the placebo group of the CERTIFI study and in 12/15 [80%] and 5/11
[45.5%] patients, respectively, at week 44 in the IM-UNITI study. Despite the numerically
pt

higher proportion of fistula healing in patients treated with ustekinumab, no significant


differences were found. Moreover, being a post hoc analysis, fistula response or remission
ce

was a secondary outcome, making it an exploratory study with insufficient statistical power
and a small sample size. In a recent meta-analysis that included 25 studies [most of which
Ac

were observational studies and 20% of them being abstracts], 24.7% of patients achieved
clinical remission of complex perianal fistulae at weeks 8–12 and 41.9% at 12 months.52
Overall, there is insufficient evidence to recommend ustekinumab for treatment of complex
perianal fistulae in patients with CD. However, ustekinumab could be considered in patients
with perianal fistulae who are refractory or intolerant to anti-TNF agents. Further studies with
appropriate design are warranted to determine the benefit of ustekinumab in the treatment of
complex perianal fistulae.
Manuscript Doi: 10.1093/ecco-jcc/jjae089

Statement 3.8 ECCO CD Treatment GL [2024]

There is insufficient evidence to recommend use of upadacitinib for the treatment of


complex perianal fistulae in CD [EL4].

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Upadacitinib [UPA] is currently the only JAK inhibitor approved for CD. Patients with
moderate-to-severe CD were randomized to UPA 45 mg once daily or placebo for 12 weeks

t
ip
in two phase 3 induction trials. Patients who achieved clinical response after 12 weeks of
UPA therapy were randomly assigned to receive UPA 30 mg or 15 mg or placebo once daily

cr
for 52 weeks. Among 1021 enrolled patients, 143 patients had fistulae at baseline [124
patients had perianal fistulae, 19 had enterocutaneous fistulae]. Post hoc analyses

us
published as an abstract reported that in patients with draining fistulae at baseline, the
proportion of patients with ≥50% reduction in draining fistulae at week 12 was significantly
an
higher with UPA 45 mg compared with placebo (22/44 [50%] patients vs 3/22 [13.6%]
patients; p = 0.004).
M

Furthermore, complete resolution of draining fistulae at week 12 was also significantly higher
with UPA 45 mg than with placebo (21/44 [47.7%] patients vs 2/22 [9.1%] patients; p =
0.002). Numerically, a similar resolution pattern was seen in patients treated with either UPA
d

30 mg or 15 mg (1/11 [9.1%] and 3/17 [17.6%] patients, respectively vs 0/8 [0%] placebo-
e

treated patients). Closure of the external fistula opening at week 52 was higher with either
pt

UPA 30 mg or 15 mg (4/19 [21.1%] and 6/35 [17.1%] patients, respectively vs 0/25 [0%]
placebo-treated patients).53 Nevertheless, this post hoc analysis has several limitations,
ce

including small sample size and inadequate statistical power.

In summary, the available evidence is of low quality and insufficient to recommend UPA as
Ac

treatment for complex perianal fistulae in patients with CD. Further studies with appropriate
design are warranted to determine the benefit of UPA in the treatment of complex perianal
fistulae.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Statement 3.9 ECCO CD Treatment GL [2024]

There is lack of evidence to recommend use of risankizumab for the treatment of


complex perianal fistulae in CD [EL5]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


3.2. Surgical techniques

t
Statement 3.10 ECCO CD Treatment GL [2024]

ip
We recommend fistulotomy in carefully selected CD patients with a simple fistula in

cr
the absence of proctitis [EL4]

us
Studies on fistulotomy in CD are largely retrospective single-centre studies with specific
an
eligibility criteria, including Parks classification superficial, intersphincteric, or low
transsphincteric fistula,54-60 absence of proctitis,57,58 quiescent abdominal disease,61 and a
low number of daily bowel motions.57 Few studies have compared the outcomes of
M

fistulotomy in these select patients with alternative surgical procedures, which were mostly
performed in patients with more complex or high anal fistulae. Due to this selection bias,
d

these studies demonstrated improved healing and reduced recurrence rates in patients
e

undergoing fistulotomy when compared with sphincter-preserving procedures, seton


removal, and mesenchymal stem cells [MSC].55,58,60. In the largest studies, recurrence rates
pt

of 3–13% up to 1 year post-fistulotomy55,57,58,60 were reported. However, few studies provide


ce

robust data on continence and wound healing. Other reports present data from
heterogenous populations, including non-CD fistulae54,59 or those undergoing multiple
procedures prior to fistulotomy,62 highlighting the difficulty in drawing recommendations from
Ac

such data. Therefore, fistulotomy can only be recommended in simple, superficial, or low
anal fistulae with absence of proctitis and stable intestinal disease.

Statement 3.11 ECCO CD Treatment GL [2024]

We suggest advancement flap as a treatment option for selected patients with CD and
complex perianal fistulae in the absence of proctitis [EL4]
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Fistula closure can be achieved by raising a flap of mucosal tissue within the anus and lower
part of the rectum. The advancement [AF] flap is then used to cover the internal opening of
the fistula. CD patients with a single internal fistula opening and without proctitis or an anal
stenosis are eligible. A systematic review identified 11 retrospective studies that reported
data from 135 patients with CD perianal fistulae treated with an AF.63 The pooled success
rate was 66%. However, results were heterogeneous, probably due to varying definitions of

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


success and length of follow-up. In a more recent meta-analysis, Stellingwerf et al. observed
a weighted overall success rate of 61% in CD patients.64 Results were not significantly
different when compared with the success rate of ligation of the intersphincteric fistula tract

t
[LIFT] procedure.

ip
Additional prospective and retrospective series not included in the meta-analyses showed

cr
comparable clinical healing rates with AF, ranging from 47–90%65-68 and recurrence rates
around 15–20%. Two studies showed a higher clinical healing rate when AF was performed

us
in patients treated with anti-TNF/immunomodulators [75.0% vs 37.5%] and after seton
drainage.68 One study also showed a 100% success rate in diverted patients.66
an
The disadvantage of AF is risk of impaired continence. The systematic review showed an
acceptable postoperative incontinence rate, which was higher in AF when compared with the
LIFT procedure [7.8% vs 1.6%].64 However, most prospective series revealed a
M

postoperative higher incontinence rate of up to 20% following AF. Conversely, one


retrospective study reported a postoperative improvement in faecal continence.68
e d
pt

Statement 3.12 ECCO CD Treatment GL [2024]


ce

We recommend ligation of the intersphincteric fistula tract as a treatment option for


Ac

selected patients with CD and complex perianal fistulae [EL3]

LIFT aims to achieve fistula closure by ligation of the fistula tract in the intersphincteric
plane, close to the internal opening. A theoretical advantage of LIFT over AF in CD patients
is that it does not involve surgery of the [diseased] mucosa. Patients with a single, non-
branching fistula and a well epithelialized tract are preferably eligible.

Two systematic reviews and meta-analyses, both including approximately 1300 patients,
demonstrated a high clinical success rate of 77% and 69% [range 47–95%], respectively,
Manuscript Doi: 10.1093/ecco-jcc/jjae089
after a median follow-up of over 1 year.64,69 However, there was only a minority of patients
with CD in these studies, and these patients had a lower success rate of 53%. Included
studies were heterogeneous, with a wide range of outcomes and follow-up times, which
makes it difficult to draw firm conclusions. The described recurrence rates were low [1.6%]
and compared favourably to AF [7.8%].

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Two retrospective and one prospective study published after the aforementioned meta-
analyses reported results on an additional 95 patients with CD.68,70,71 Clinical closure rates
were comparable with the results previously published. However, data on recurrence were

t
only reported in one series, with a rate of 21%.70 Overall, this suggests a possible

ip
underreporting in the systematic reviews and meta-analyses. Another retrospective study
demonstrated that in patients with a [predominantly] fibrotic tract after LIFT at MRI, no

cr
reinterventions or recurrences were seen during long-term follow-up, which also emphasizes
the requirement of radiological healing to consider a patient healed.68

us
The only prospective series included 46 patients with a mean follow-up of 33 months and
demonstrated fistula healing in 65% of patients.71 Smoking at time of surgery was
an
significantly associated with failure (hazard ratio [HR] 3.2), and a trend was seen towards
increased failure in patients with active proctitis [HR 2.0]. No other factors [use of biologics,
M

prior seton drainage, type of fistula, previous repair attempts] appeared to influence LIFT
healing.
d

Postoperative complications after LIFT were seen in up to 14% of patients and were
e

predominantly wound dehiscence. Incontinence rates appeared to be lower when compared


pt

with AF. However, continence should be interpreted with caution as there is a risk of under-
reporting in the literature. The only retrospective series specifically examining postoperative
ce

incontinence in 37 patients demonstrated increased incontinence in 16% of patients after


LIFT, whereas 53% of patients operated with LIFT and 43% with AF reported a
postoperative improvement in faecal continence.68
Ac

Statement 3.13 ECCO CD Treatment GL [2024]

We recommend against the use of fibrin glue in the treatment of patients with
complex perianal CD fistulae [EL4]
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Fibrin glue for treatment of perianal CD fistulae was assessed in an open-label RCT with 71
patients randomized to instillation of fibrin glue into the fistula tract or no further treatment
after seton removal.72 This RCT demonstrated a significant difference in overall clinical
remission rate [38% for fibrin glue and 16% in the observation group; p = 0.04]. However,
the length of follow-up in this RCT was only 8 weeks and was insufficient for a definitive
judgement on the true success rate. The only retrospective series with adequate follow-up

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


time [5 years] suggested an acceptable healing rate of 45% at 1 year,73 but the single
predictor for complete clinical remission was combination with medical therapy. This series
also demonstrated a worrisome cumulative incidence of iterative anal surgery of 54% within

t
5 years, suggesting a high recurrence rate after fibrin glue. Despite the limited efficacy of

ip
fibrin glue in daily clinical practice, a uniform characteristic of all studies is the relatively good

cr
safety profile of this technique with no reported injury to the sphincter muscles.

us
an
Statement 3.14 ECCO CD Treatment GL [2024]

We recommend against anal fistula plug in the treatment of patients with complex
M

perianal CD fistulae (EL4)


e d

Use of a collagen anal fistula plug [AFP] in patients with perianal CD fistulae was assessed
in a single RCT including 106 patients, which compared AFP after seton removal with seton
pt

removal only.74 The fistula closure rate after 12 weeks in the AFP group was 33.3% in
ce

patients with complex fistulae and 30.7% in patients with simple fistulae as compared with
15.4% and 25.6% with seton removal alone, respectively. These differences were not
statistically significant. In addition, there was a trend towards more adverse events [AE] in
Ac

the AFP group [17% vs 8%; p = 0.07], although cumulative AE rates at 12 months follow-up
were similar.

A systematic review of 12 observational studies including 84 patients with CD demonstrated


an overall AFP success rate of 58%, with 14% recurrence after median follow-up of 9 [3–24]
months.75 However, there was no uniform definition for fistula closure or follow-up regimen.
The quality of evidence for this systematic review was low due to risk of bias and
imprecision. Use of an AFP in patients with CD appears to be relatively safe and may not
affect continence [limited data on continence reported].76 However, in studies using AFP for
Manuscript Doi: 10.1093/ecco-jcc/jjae089
cryptoglandular fistulae, the abscess formation/sepsis rate ranged from 4–29% and the plug
extrusion rate from 4–41%.77

Statement 3.15 ECCO CD Treatment GL [2024]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


There is insufficient evidence to recommend use of video-assisted anal fistula
treatment, fistula-tract laser closure, or over-the-scope clip for achieving healing in

t
complex perianal fistulae in CD [EL5]

ip
cr
The role of video-assisted anal fistula treatment [VAAFT] in the treatment of anal fistulae in
CD has been investigated only in small cohort studies. A first retrospective study including a

us
mixed population of 84 patients with cryptoglandular and CD fistulae [n = 11] with a limited
median follow-up of 8 months revealed a 27% healing rate in patients with CD.78 Data on
an
postoperative complications and risk of postoperative incontinence were lacking. A second
retrospective study reported an overall healing rate of 82% at 9 months follow-up.79
However, these results are difficult to interpret due to the very limited sample size of 11
M

patients and by the fact that internal opening closure was achieved by fashioning a rectal
advancement mucosal flap. Furthermore, in about 40% of patients, faecal diversion [FD] was
d

present at time of surgery. No patients experienced postoperative morbidity or postoperative


e

faecal incontinence. VAAFT was further evaluated in a retrospective analysis of


prospectively collected data of 25 patients with anal fistulae refractory to multiple previous
pt

surgeries and adequate medical treatment with biologics.80 Twenty-one of 25 patients [84%]
had a statistically significant improvement in a quality-of-life questionnaire before and 6
ce

weeks after surgery, in particular in both pain and discharge scores. Eighty-one percent
agreed that the procedure was the right decision and no patient regretted undergoing the
Ac

procedure. Reoperation was necessary in one patient [4%].

Fistula-tract laser closure [FiLaC] is a relatively new sphincter-preserving technique initially


reported in 2011. A systematic review published in 2022 identified six retrospective studies
investigating FiLAC as a treatment option for perianal CD on a total of 50 patients.81 There
was heterogeneity in length of follow-up, fistula characteristics, and outcomes reported. The
techniques used were only partially described, especially how to address internal opening[s]
of the fistula, and included technical variations. The pooled rate of primary healing among
Manuscript Doi: 10.1093/ecco-jcc/jjae089
the studies was 68% [95% CI: 53.0–84.0%]. No postoperative complications or faecal
incontinence was observed, although not all studies reported these outcomes.

The role of over-the-scope clip [OTSC] in the treatment of anal fistulae in CD has only been
investigated in several small observational case series, often with mixed populations; the
majority were cryptoglandular cases and fewer were CD-related fistula. Mennigen et al.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


reported a case series of 10 patients including data on 6 patients with CD.81 A total of 4/6
[66.7%] patients were on biologic therapy at the time of OTSC and all these patients
achieved fistula closure; only 1 patient not receiving biologics healed. Although no

t
postoperative morbidity or faecal incontinence was observed, the OTSC may be

ip
spontaneously passed [2/6, 33%] or need to be subsequently removed due to discomfort
[1/6, 16.7%]. A study by Prosst and Joos reported OTSC in 100 patients (11 had

cr
inflammatory bowel disease [IBD]) with a closure rate of 45% in IBD. 82 Overall, the OTSC

us
was spontaneously passed in 18 patients and appeared to be associated with a lower fistula
closure rate of 33% [6/18 patients]. The OTSC needed to be removed or operatively
explanted in 14 patients. No significant postoperative morbidity or faecal incontinence was
an
reported. Although OTSC appears to be safe and may result in fistula closure in some
patients, widespread adoption of this technique is currently limited by a paucity of data in
M

CD.
e d

Statement 3.16 ECCO CD Treatment GL [2024]


pt

We recommend against use of chronic seton treatment as the sole treatment for
ce

perianal CD fistulae other than as palliation [EL3]


Ac

We recommend against a cutting seton due to the risk of incontinence [EL5]

There are no RCTs or studies comparing seton drainage with no treatment for perianal CD
fistulae. Two systematic reviews, including 10 studies [n = 305 patients] on patients treated
solely with seton drainage, reported varying results.83,84 Complete closure rates ranged from
13.6–100% and recurrence rates from 0–83.3%. Timing of seton removal differed among
studies [range 3 weeks to 40 months]. Included studies were prospective and retrospective
cohort studies and case series and mostly of questionable quality.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Additionally, the PISA trial published in 2020 compared the following three treatment
strategies: long-term seton drainage alone, anti-TNF treatment, and surgical closure [the
latter two with prior seton drainage].85 The study was stopped by the data safety monitoring
board because of futility. Seton treatment was associated with the highest reintervention rate
[10/15 seton vs 6/15 anti-TNF vs 3/14 surgical closure patients; p = 0.02]. No substantial
differences in perianal disease activity and quality of life were observed between the groups.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Interestingly, in the accompanying PISA prospective registry, inferiority of chronic seton
treatment was not observed for any of these outcome measures. This study suggested that
chronic seton treatment should not be recommended as the sole treatment for perianal CD

t
fistulae.

ip
The cutting seton, in which a non-absorbable thread is inserted into the fistula tract and

cr
exteriorized through the anorectal canal with subsequent tightening, causing gradual cutting
through the anal sphincter, should not be used as many studies have shown associated

us
complications, including prolonged perianal pain and incontinence rates up to 58%.84
an
3.3. Combined approaches
M
d

Statement 3.17 ECCO CD Treatment GL [2024]


e

We recommend seton drainage preceding medical or surgical therapy for complex


pt

perianal CD fistulae [EL3]


ce

Combined anti-TNF therapy and seton removal could result in improved healing rates,
faster time to healing, longer time to relapse, and a reduced need for surgery than
Ac

either therapy alone [EL3]

There were no RCTs comparing medical or surgical therapy with or without preceding seton
drainage. Five systematic reviews were included.83,84,86-88 Most studies focused on anti-TNF
therapy. One of the largest systematic reviews [42 studies] included studies assessing anti-
TNF agents for perianal fistulae. In most studies, anti-TNF was combined with preceding
seton placement and it was suggested that combining seton drainage with an anti-TNF
agent was superior. These results are consistent with another large systematic review that
Manuscript Doi: 10.1093/ecco-jcc/jjae089
revealed that a combination of surgical treatment [including seton drainage] with medical
therapy [anti-TNF agents and immunomodulators] may have additional benefit on healing of
perianal CD fistulae compared with surgery or medical therapy alone.88 One study showed
that 75% of patients treated with anti-TNF therapy after prior seton placement healed
compared with 63% of patients without initial seton.89 Another study revealed that patients
with seton placement prior to anti-TNF therapy had a better initial response [100% vs 82.6%;

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


p = 0.014], lower recurrence rate [44% vs 79%; p = 0.001], and longer time to recurrence
[13.5 vs 3.6 months; p = 0.0001] compared with patients receiving infliximab alone.90
Additionally, patients with seton placement prior to anti-TNF therapy were less likely to

t
require hospitalization and had reduced healthcare costs.87 Studies have also shown shorter

ip
mean time to healing,91,92 longer time to relapse,92 and reduced need for repeat surgery93

cr
than with either therapy alone.

Timing of seton removal is largely variable and inconsistent between studies, ranging from

us
4–27 weeks post insertion.93,94 However, the heterogeneity and low quality of the mainly
retrospective studies included should be considered.
an
In most studies, seton drainage was performed prior to surgical closure in patients with
perianal CD fistulae. However, several small retrospective studies showed no association
M

between fistula healing rate after a LIFT procedure and prior seton placement or duration of
seton drainage prior to surgery.71,95
d

A recent retrospective study analysed medical and surgical therapies to identify the optimal
e

care strategy in 200 patients. Seton drainage prior to anti-TNF therapy alone did not
pt

significantly increase the fistula closure [HR: 1.15; 95% CI 0.61–2.32; p = 0.66]. The
combination of seton placement and anti-TNF therapy followed by fistula closure surgery
ce

within 52 weeks was the best management strategy for fistula healing in multivariate
analysis [p = 0.02]. Cumulative probabilities of fistula closure following the latter combined
approach were 43.8%, 82.2%, and 93.7% at 1, 3, and 5 years, respectively. Patients
Ac

concomitantly treated with a combination of anti-TNF therapy and immunosuppression at


surgery had the highest long-term closure rate.96

Importantly, especially in case of perianal sepsis, adequate seton drainage seems to be of


key importance to create optimal circumstances prior to starting medication or proceeding to
surgical closure.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Statement 3.18 ECCO CD Treatment GL [2024]

We recommend the combination of medical therapy with surgical fistula closure in


amenable patients with complex perianal fistulae, as surgical closure results in
improved long-term outcomes [EL3]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Two RCTs and one retrospective study investigated surgical closure of the fistula tract in
combination with medical therapy. A first multicentre RCT compared seton removal and

t
surgeon’s choice of closure with seton removal alone in patients treated with adalimumab.

ip
There was no difference in clinical closure at 12 months [surgery 56.3% vs control 65.4%; p
= 0.48] or in secondary outcomes measuring quality of life, continence, and AEs. Patients

cr
with surgical closure experienced longer disease duration and were more likely to have been

us
previously treated with infliximab, suggesting more aggressive disease. Most patients [79%]
were treated with fibrin glue with limited efficacy in perianal CD. In addition, the study was
underpowered and robust conclusions could not be drawn from these data.97
an
Ninety-four patients were enrolled [38 patients with surgical closure and 56 with anti-TNF
therapy] in the patient preference PISA II trial.98 At 18 months, radiological healing was
M

significantly more common after surgical closure (12/38 [32%] patients) than after anti-TNF
therapy (5/56 [9%] patients) [p = 0.005]. Clinical closure was not significantly different
d

between the two treatments [68% vs 52%, respectively; p = 0.076]. Fewer patients required
e

a reintervention and the perianal disease activity index was significantly lower after surgical
closure. Long-term results after a median follow-up of 5.7 years showed no recurrences in
pt

patients with radiological healing; recurrence was observed in 41% of patients with clinical
closure without radiological healing.99
ce

A retrospective study of 226 patients found no difference in healing when patients underwent
Ac

a variety of surgeries alone compared with those undergoing surgery with concurrent
infliximab [60% vs 59%, respectively]. Surgical procedures included seton drainage [50%],
fistulotomy [41%], fibrin glue [6%], advancement flap [2%], and collagen plug [1%]. However,
time to healing was 6.5 months after combination therapy [surgery and infliximab] and 12.1
months after surgery alone [p < 0.0001].91
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Statement 3.19 ECCO CD Treatment GL [2024]

There are conflicting data on allogenic adipose-derived stem cell therapy for the
induction and maintenance of remission in complex perianal fistulae in CD [EL5]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


The efficacy of MSC in treatment of perianal fistulae CD is mediated by anti-inflammatory
properties and by the capacity to engraft and transdifferentiate into healthy tissue.100
Allogenic MSC from adipose tissue (Cx601-darvadstrocel; Alofisel®) was assessed in a

t
phase 3 RCT that included 212 patients with refractory fistulizing perianal CD.101 At week 52,

ip
a significantly higher proportion of patients treated with darvadstrocel achieved combined
remission when compared with controls [56.3% vs 38.6%; 95% CI 4.2–31.2; p = 0.010].

cr
Combined remission was defined as closure of all treated external openings at clinical

us
examination and absence of collections >2 cm at MRI. A study extension including 40
patients was prospectively conducted through week 104.102 Clinical remission was reported
in 14/25 [56%] patients in the darvadstrocel group and 6/15 [40%] patients in the control
an
group, which was not statistically significant [95% CI: -15.5 to 47.5]. No serious AEs were
reported at week 52 or week 104. Due to the high cost of darvadstrocel, the costs and
M

potential benefits should be considered on a case-by-case basis of the clinical situation.

A meta-analysis published in 2018 that included three studies suggested that MSC of
d

different origin significantly improved healing of perianal fistulae when compared with control
e

at 6 to 24 weeks (odds ratio [OR]: 3.06; 95% CI: 1.05–8.90; p = 0.04) and numerically at 24
to 52 weeks [OR: 2.37; 95% CI: 0.90–6.25; p = 0.08].103 No significant increases in AEs [OR:
pt

1.07; 95% CI: 0.61–1.89; p = 0.81] were observed in treated patients. Limitations of the
available studies on MSC in perianal CD include heterogeneity in protocols [allogenic or
ce

autologous MSC, bone-marrow or adipose tissue-derived MSC], low number of patients,


varying definitions of fistula healing, and lack of consensus on definition of perianal fistula
Ac

healing in MRI. Further studies based on robust well-defined radiological targets are needed
to evaluate the role of MSC on the natural history of perianal fistulizing CD. Results from the
phase 3 RCT ADMIRE-CD II will provide additional information.104 Although the results of the
ADMIRE-CD II were not yet published at the time of writing the present guidelines, the
sponsor announced in a press release dated 17 October 2023 that the primary endpoint of
combined remission at 24 weeks in complex perianal CD fistulae treated with darvadstrocel
was not met. These inconclusive results were also presented at ECCO 2024 on 23 February
2024. The safety profile for darvadstrocel was consistent with prior studies and no new
Manuscript Doi: 10.1093/ecco-jcc/jjae089
safety signals were identified. The final results of ADMIRE-CD II will help position this
treatment in the management of complex perianal fistulae in CD.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Statement 3.20 ECCO CD Treatment GL [2024]

We suggest autologous adipose-derived stem cells may be used as a treatment


option in complex perianal CD [EL4]

t
ip
There is insufficient evidence to recommend use of platelet-derived factors or stromal

cr
vascular fraction in complex perianal CD [EL5]

us
Autologous stem cells [ASC] have the advantage of originating from the patient undergoing
an
treatment, as opposed to donor-based therapy, thus making ASC readily available and less
costly. ASC may be injected in a similar manner as allogenic MSC, mixed with fibrin glue, or
M

loaded onto a fistula plug.

The most recent systematic review summarizing results of four RCTs demonstrated
d

increased clinical healing rates of ASCs when compared with control patients treated with
fibrin glue alone [OR: 3.19; 95% CI: 1.05–9.65; p = 0.04].105 Unfortunately, it is difficult to
e

draw firm conclusion for patients with CD, as only 20 patients with CD were included in these
pt

studies and most patients had a short follow-up of only 8 weeks. There are no studies that
directly compared autologous to allogeneic stem cells for perianal CD fistulae.
ce

The best evidence on the use of ASCs for perianal CD fistulae comes from various
prospective case series including a total of 110 patients.106-111 Although treatment protocols
Ac

varied substantially, most involved curettage of the fistula tract, suturing of the internal
opening [with or without an advancement flap], and filling of the fistula tract with ASCs. Most
studies allowed a second injection of ASCs in patients with incomplete closure. Clinical
healing rates, defined as no suppuration from the external orifices, ranged from 33–91%.
However, most of these series lacked an adequate follow-up [range 2–12 months] with
recurrence rates rarely described. The largest study included 30 patients and showed a
closure rate of 83.3% with a recurrence rate of 33%.110
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Despite the additional requirement of harvesting cells via liposuction to obtain ASC, the
procedure appeared safe. The most common AEs were postoperative pain, abscess, or
bleeding.105 There were no significant differences in AEs when compared with the control
group [OR: 1.06; 95% CI: 0.71–1.59; p = 0.77].

There are also some studies that investigated the effects of injecting freshly collected

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


microfragmented autologous adipose tissue, platelet-derived growth factors, or stromal
vascular fraction into perianal CD fistulae.109,111,112 Feasibility was demonstrated in most
patients and results appeared comparable to ASCs, with clinical healing ranging from 38–

t
67%. Harvesting, preparation, and administration of these tissues are described as easy,

ip
inexpensive procedures with minimal AEs. Again, these series suffer from small patient
numbers and brief follow-up and lack description of recurrence rates. Further studies are

cr
required to define the true potential of these approaches.

us
an
Statement 3.21 ECCO CD Treatment GL [2024]
M

We suggest medical treatment in anogenital and rectogenital CD fistulae and


counselling for surgical closure in selected patients with CD [EL5]
e d

Anogenital and rectogenital fistula are complex and disabling conditions that are better
managed by an experienced multidisciplinary team. No RCTs or prospective studies were
pt

found that compared anti-TNF agents alone versus anti-TNF agents and surgery combined
ce

to treat these fistulae.

A post hoc analysis of the ACCENT II study identified 25 women with ano- or rectovaginal
Ac

fistulae.113 This study demonstrated that infliximab is more effective than placebo in
prolonged closure [defined as non-draining fistula at week 14]; 13/29 [44.8%] fistulae
responded to induction regimen with infliximab and were closed. From weeks 14 to 46,
among responders in the infliximab maintenance group, the proportion of rectovaginal
fistulae that closed ranged from 54.5–90.0% compared with 28.6– 42.9% in the placebo
group.

A French retrospective multicentric observational study including 131 consecutive patients


treated with anti‐TNF agents for 1 year found that 37% of patients had complete clinical
fistula closure, 22% had partial response, and 41% had no response.114 Complementary
Manuscript Doi: 10.1093/ecco-jcc/jjae089
surgery was allowed, including advancement flap [rectal, vaginal, or Martius flap], fibrin glue,
collagen plug, or gracilis muscle interposition and performed during the first year in 10
patients [8%], translating into a higher closure rate in multivariate analysis [adjusted RR:
2.02; 95% CI 1.25–3.26; p = 0.004]. A retrospective study of 166 patients who underwent
operations for anogenital fistulae revealed an overall fistula healing rate of 71.7% [n = 119]
with a median follow-up of 5.5 [1.2–9.8] years.115 Nearly one-third of patients [33.1%]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


achieved complete healing after first surgery, 51.8% [n =86] after the second, and 62.1% [n
= 103] after the third operation.

t
A recent systematic review found nine studies that reported healing, success, or closure

ip
[range 14–81%] across multiple surgical procedures; seven studies reported success rates
ranging from 50–75%.116 However, those studies were of low quality and had limited sample

cr
sizes, various concomitant medical therapies, heterogenous fistula and patient
characteristics, outcomes considered, and definition of outcomes.

us
an
Statement 3.22 ECCO CD Treatment GL [2024]
M

We suggest faecal diversion with a defunctioning ileostomy or colostomy for


treatment of refractory, complex perianal CD [EL4]
e d

Patients with treatment-refractory perianal CD may benefit from faecal diversion [FD] with a
pt

diverting ileostomy or colostomy. Indeed, FD is associated with a high early clinical response
ce

rate and an improved quality of life, although FD often becomes permanent. A systematic
review of 16 retrospective studies with 556 patients with perianal CD found that FD is
associated with early clinical response in 63.8% [95% CI: 54.1–72.5%].117 However, stomas
Ac

were often permanent and only 16.6% [95% CI: 11.8–22.2%] of patients ultimately had
successful ostomy reversal. The rate of proctectomy after failure of temporary diversion was
41.6% [95% CI: 32.6– 51.2%]. Proctitis was associated with increased risk of permanent
diversion.

One study compared FD plus local procedures for perianal CD [n = 13] to local procedure
without FD [n = 26].118 Complete resolution of perianal CD was observed in 11 [85%]
patients with FD versus 5 [19%] patients without FD. Of the FD patients, 6 [46%] had stoma
reversal, of whom 3 [50%] remained disease free, 1 [17%] required successful additional
Manuscript Doi: 10.1093/ecco-jcc/jjae089
local procedures, and 2 [33%, 15% overall] required re-diversion. Thus only 4/13 [31%] of
FD patients ultimately had stoma reversal. Another study of 21 patients showed that
although some patients may achieve complete healing, many do not; initial improvement
was followed by plateau in 7 [33%], temporary improvement in 6 [29%], no effect in 4 [19%],
and healing in 4 [19%] patients.119 In this study, 11 [52%] patients subsequently had
proctocolectomy, 6 [28.6%] had their stoma in situ, and 4 [19%] had stoma reversal. In a

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


large series of 138 patients who had initial FD, a total of 63 [45%] underwent subsequent
total proctocolectomy, 45 [33%] had their stoma without proctectomy, and 30 [22%] had
stoma reversal.120 Independent predictors of lack of stoma reversal included proctitis [OR:

t
7.5; 95% CI: 2.4–33.4], 1–2 seton placements [OR: 3.3; 95% CI: 1.4–8.8], and >2 seton

ip
placements [OR: 6.9; 95% CI: 1.2–132.5]. Biologics were not associated with stoma closure

cr
[p = 0.25].

us
Few studies examined quality of life before and after FD in perianal CD. In a series of 34
patients with FD, compared with similar patients without FD, patients with FD had fewer
perianal CD symptoms [44% vs 79%; p < 0.05], higher Gastrointestinal Quality of Life index
an
scores [68 vs 62 points; p < 0.001], and higher GI symptoms sub-scores [81 vs 67; p <
0.0001] compared with non-diverted patients.121 The most recent meta-analysis evaluating
M

1578 patients managed in the biologic era similarly concluded that FD improved symptoms
and quality of life, while bowel continuity could be successfully restored in a quarter of the
patients.122
e d
pt
ce

Statement 3.23 ECCO CD Treatment GL [2024]

We suggest proctectomy for treatment of refractory, complex perianal CD despite


Ac

defunctioning stoma [EL4]

Proctectomy may be recommended in many patients with perianal CD. However,


proctectomy is associated with a substantial risk of a non-healing perineal wound in the
short-term and a risk of colonic or small-bowel recurrence in the long-term. In a series of 127
patients with perianal CD, proctectomy was required in 32 [25.2%] patients.123 Several
studies discussed independent risk factors for proctectomy, including age at first perianal
fistula [p < 0.02], perianal fistula at the time of CD diagnosis [p < 0.04], >3 fistulae during
Manuscript Doi: 10.1093/ecco-jcc/jjae089
follow-up [p < 0.01], and proctitis [p < 0.0001].124 Other studies also reported malignancy in
the setting of perianal CD as an indication for an oncologic proctectomy.124-126

Proctectomy for perianal CD is typically performed as an abdominoperineal resection [APR]


with a colostomy or as a total proctocolectomy with end-ileostomy in case of extensive
colonic involvement.123-128 In terms of extent of bowel resection in the setting of perianal CD,

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


a single study examined APR with colostomy and reported a clinical recurrence rate of
colonic CD of 22% for an endoscopic colonic recurrence rate of 29%; overall 5% of patients
required completion total colectomy.128 It is important to note that proctocolectomy does not

t
cure CD; a multicentre retrospective study of total proctocolectomy with end-ileostomy

ip
including 193 patients with refractory perianal CD reported a 23% small-bowel recurrence
within 2 years.127 Independent risk factors for recurrence included CD diagnosis at age <18

cr
years [HR: 2.56; 95% CI: 1.40–4.71] and previous small-bowel resection [HR: 2.61; 95% CI:
1.42–4.81].

us
Proctectomy for IBD is often performed as an inter-sphincteric dissection, limiting the size of
an
the perineal incision.123-128 The inter-sphincteric groove may not be identifiable due to
scarring in up to 78% of patients with perianal CD, limiting the ability to perform an inter-
sphincteric dissection and impacting wound healing.126 Indeed, delayed perineal wound
M

healing is often observed after proctectomy in perianal CD.125,129-132 When wounds are left
open to heal by secondary intention, an uncommon practice nowadays, only 58% of perineal
d

wounds of patients with IBD were healed after 6 months of dressing changes.130 Wound
e

irrigation has also been explored in the 1980s and half of perineal CD wounds were healed
at 30 days compared with 87% after APR for cancer in the absence of radiotherapy.132 Male
pt

gender was a risk factor for delayed healing, especially when the drain exited through the
wound instead of laterally. Higher success rates were observed when myocutaneous flaps
ce

were used, although patients are still at risk for subsequent fistulization [20% in a small
study].131 In a large series of 126 patients, 72 [53%] wounds were healed at 12 weeks, while
Ac

delayed healing was observed in 35 [26%] and non-healing in 29 [21%] patients.125


Preoperative perianal sepsis was an independent predictor of a delayed- or non-healing
wound [p = 0.001], suggesting FD prior to proctectomy.129 For non-healing perineal wounds
with metastatic CD, hyperbaric oxygen therapy may be an option.133
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Practice Points

Fistula treatment should start with insertion of a seton followed by medical treatment
[preferably anti-TNF]. In the absence of proctitis, patients should be counselled for surgical
closure.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Perianal fistulae in CD can have a substantial detrimental impact on patient quality of life.
Current biological understanding of perianal fistulizing CD remains inadequate and previous

t
classification systems have not provided clear guidance on therapy in clinical practice. A

ip
new classification presented in Figure 1 identifies four groups of patients.134 Key elements
include stratification according to disease severity and desired outcome. This classification

cr
can guide patients and clinicians in decision making on a ‘treat to patient goal basis’ by a
combined medical and surgical approach.

us
All treatment should start with insertion of a seton to control sepsis and create a patent tract,
an
followed by medical treatment [preferably anti-TNF with high trough level]. After good
response to anti-TNF therapy, seton removal can be considered within 2–8 weeks to aim for
closure with medication only.135 Although clinical closure can be achieved in up to 60% by
M

medication, it should be noted that MRI closure is rare [<10%] with high risk of recurrence
and surgical reintervention.98 MRI closure is more frequently seen after surgical closure
d

under anti-TNF therapy [up to 40%], with no recurrences after long-term follow-up in case of
a completely fibrotic tract on MRI.99 Therefore, in absence of proctitis, amenable patients
e

should be counselled for surgical closure. For patients with an inter-sphincteric or low trans-
pt

sphincteric single fistula tract, fistulotomy can be considered as this procedure will have the
highest success rate.
ce

In case of complex perianal fistulae, AF or LIFT can be offered, depending on fistula


characteristics. Stem cells can be an alternative, especially in patients with multiple internal
Ac

openings or pre-existing complaints of incontinence.

In case of anti-TNF failure and surgically refractory fistulae, more experimental approaches
[such as hyperbaric oxygen therapy or new medical approaches] can be attempted, ideally in
the context of a prospective clinical trial. An algorithm to guide the management of perianal
CD is illustrated in Figure 2.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
4. Surgical management of abdominal Crohn’s disease

4.1 Preoperative optimization

Statement 4.1 ECCO CD Treatment GL [2024]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


We recommend elective bowel resection over emergency surgery in patients with CD
[EL2]

t
A meta-analysis of cohort series including 75’971 CD patients from 15 countries reported a

ip
significantly lower mortality among patients who underwent elective [0.6%; 95% CI: 0.2%-
1.7%] vs emergent surgery [3.6%; 95% CI: 1.8%-6.9%], highlighting the importance of

cr
perioperative optimization and avoidance whenever possible of emergent surgeries136.

Statement 4.2 ECCO CD Treatment GL [2024] us


an
Pre-operative optimization should be initiated, followed by re-assessment of the
patient for surgical intervention [EL3]
M

A recent meta-analysis showed that emergency bowel resection is associated with a higher
d

risk of overall postoperative complications and abdominal septic complications.137 This is


e

consistent with a European Society of Coloproctology prospective snapshot audit in which


pt

emergency surgical intervention was associated with unfavourable postoperative


outcome.138 Another recent multicentre international observational study concluded that
ce

emergency intervention in patients with an abdominal abscess increased the risk of


postoperative complications and abscess recurrence.139 Moreover, patients undergoing
Ac

emergency surgery for CD have a higher rate of stoma formation.140,141 Lastly, laparoscopic
surgery in the emergency setting has a higher conversion rate and involves resection of
longer segments of small bowel, which is a concern in CD due to a lifetime risk of short
bowel.140
The drivers behind these unfavourable outcomes may be patient status and the environment
of care typical of an emergency situation. Emergency resection [within 48 hours of
admission] is performed on tissue characterized by profuse oedema and acute inflammation
in a patient often in an unstable condition by a team that may not be specialized in IBD or
even colorectal surgery. Patients with CD who undergo emergency operation typically have
Manuscript Doi: 10.1093/ecco-jcc/jjae089
a severe form of disease, are malnourished, and are often on steroids, immunomodulators,
biologicals, or combinations thereof with a higher likelihood of undrained abscesses, fistulae,
or both at time of emergency surgery. Drainage of an abscess and relieving obstruction
together with preoperative optimization should be initiated immediately on admission, as
described in recent prospective cohort series142,143 and advised in ECCO topical
reviews.144,145

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Preoperative optimization of an emergency CD patient and transfer of care from the acute to
the specialized/elective setting is key to improving short- and long-term postoperative
outcomes. On the other hand, free bowel perforation is one of the few situations where

t
urgent surgery may be mandatory as bowel perforation is a very rare but serious and

ip
potentially life-threatening complication in CD. The literature is characterized by low-quality,

cr
heterogenous studies based on historical data. A study from Korea estimated the incidence
to be 2.15% in the Korean CD population.

us
There are two important points to consider when CD leads to bowel perforation.
an
1. Bowel-wall thickness: Bowel-wall thickening in CD occurs due to chronic inflammation
and scarring and differs from ischaemic bowel perforation, which occurs when there is a
decreased blood supply to the bowel, potentially resulting in a perforation. Symptoms,
M

diagnostic approach, and treatment may also differ between these conditions.
2. Size of perforation: Bowel perforation in CD can vary in size and presentation. While
d

some cases may involve small or microscopic perforations, others can present as larger
perforations. Timely diagnosis and appropriate treatment can prevent further
e

complications and improve outcomes.


pt

A small-bowel perforation can, in very selected situations and under supervision of an


experienced colorectal surgeon, be managed conservatively. This mandates a very close
ce

clinical follow-up and the capacity to operate immediately should the patient deteriorate.
The early involvement of a multidisciplinary team consisting of an IBD gastroenterologist, an
Ac

IBD surgeon, a radiologist, and a dietician is mandatory in emergency presentation of CD


due to the complexity of the disease and management.

Statement 4.3 ECCO CD Treatment GL [2024]

We recommend control of sepsis prior to abdominal surgery for CD [EL3]


Manuscript Doi: 10.1093/ecco-jcc/jjae089
Statement 4.4 ECCO CD Treatment GL [2024]

We suggest use of intravenous antibiotics and percutaneous image-guided drainage


as the first-line treatment for intra-abdominal abscesses related to CD [EL3]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Statement 4.5 ECCO CD Treatment GL [2024]

We suggest conservative treatment following successful percutaneous image-guided


drainage of an intra-abdominal abscess in carefully selected cases. A low threshold

t
ip
for surgery is recommended in the event of medically refractory cases [EL4].

cr
Penetrating CD complicated by intra-abdominal abscesses [IASC] represents a complex
condition requiring involvement of interventional radiologists, gastroenterologists, and

us
surgeons. An [elective] operative approach appears indicated in most patients, as
conservative management leads to complete abscess resolution in less than 30% of
an
selected cases, while delayed elective surgery is associated with improved postoperative
outcomes, avoidance of a stoma, and abscess recurrence.146,147 148
M

Observational studies indicate that failure to control IASC preoperatively increases the risk of
postoperative complications, anastomotic leaks, postoperative sepsis, and stoma formation,
resulting in an increased length of hospital stay.139,149-151 Percutaneous drainage [PD] under
d

ultrasonographic or CT guidance may be the primary approach for treatment of well-defined


e

abscesses. Successful drainage rates of 74–100%, allowing avoidance of emergency


pt

surgery in 14–85% of patients, were reported.18 PD with antibiotics to control IASC resulted
in better quality of life than surgery alone, provided abscesses were completely
ce

drained.139,149,152 PD and antibiotic therapy should be combined with perioperative


optimization, including nutritional support and stopping or decreasing corticosteroids.
Ac

Despite PD, these patients still present with higher morbidity than those without preoperative
IASC.140
It is worth noting that when performed by specialized high-volume IBD surgeons, early
laparoscopic surgery [<1 week after admission] was safe, feasible, and associated with
similar morbidity rates when compared with delayed surgery [within 3 weeks after initial
admission, including PD in 28% of patients].153 However, steroid treatment before PD and
short waiting interval [<2 weeks] were associated with a higher risk of abscess recurrence,
while anaemia and long waiting interval [>4 weeks] increased the risk of stoma
construction.153 Overall, performing surgery 2–4 weeks after successful PD was associated
Manuscript Doi: 10.1093/ecco-jcc/jjae089
with the lowest risk of postoperative IASC.139 Identifying patients who may be treated without
surgery is challenging and currently relies on clinical judgment rather than on evidence. In
general, medically refractory disease, presence of stenosis, or an enterocutaneous fistula
represent clear indications for surgery.152

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Statement 4.6 ECCO CD Treatment GL [2024]
We recommend endoscopic balloon dilatation as a treatment option for small-bowel
strictures <5 cm in length when technical expertise is available [EL2]

t
ip
In a review of 1463 patients with CD who underwent 3213 endoscopic balloon dilatation

cr
[EBD] procedures, a stricture length <5 cm was mostly amenable to EBD and associated

us
with a surgery-free outcome; every additional centimetre in stricture length increased the
need for surgery by 8% [p = 0.008].154 This is consistent with other reviews.155-157
Inflammation, disease activity, type of stricture, balloon diameter, and duration of inflation did
an
not affect outcomes.154,156
While therapeutic success can be achieved after a single dilation, several dilations may be
M

necessary to resolve obstructive symptoms; however, repeat dilation may reduce quality of
life.158,159 Although accessory endoscopic techniques, including local steroid injection, cutting
procedures [e.g. Argon beaming], and stent implantation have been proposed to improve
d

resolution,154 the evidence is weak. Some retrospective cohort studies suggested that
e

combined therapy with anti-TNF and EBD may prevent intestinal stricture recurrence and
pt

surgery in hospitalized patients with CD.160,161


An unresolved controversy is the dilation efficacy of primary versus anastomotic strictures.
ce

Identification of predictive factors for the long-term success of EBD may assist clinical
decision making and an individualized treatment approach in stricturing CD.162
Ac

In conclusion, short-term therapeutic success of EBD is high in a selected group of patients


when technical expertise is available. However, the impact on long-term quality of life, need
for repeat dilations, and strictureplasty or bowel resection is less clear.
Practice point

Whenever possible, elective surgery is preferable to an emergency procedure in both


fistulizing and obstructive CD. The control of IASC is multidisciplinary and draws from
interventional radiology, infectious disease, gastroenterology, and surgery. Imaging
[sonography, CT, MRI], swift drainage, antibiotics, intensified perioperative therapy, and
specialist care are the mainstays of treatment. PD is mostly a bridge intervention rather than
Manuscript Doi: 10.1093/ecco-jcc/jjae089
a definitive solution; elective surgery performed 2-4 weeks thereafter minimizes
postoperative complications and need for a stoma.

Primary conservative management of bowel obstruction includes rehydration, nasogastric


decompression, imaging, and consideration of high-dose steroid therapy. Frequent
monitoring and surgical consultation are critical. Surgery can be deferred in most cases but

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


should be considered during follow-up. Definitive non-surgical management may be
successful but must be carefully balanced and discussed with the individual patient.

t
ip
Statement 4.7 ECCO CD Treatment GL [2024]

cr
We recommend preoperative nutritional assessment and identification of nutritional
risk by IBD-dedicated dietitians for patients with CD who need surgery [EL2]

us
an
Statement 4.8 ECCO CD Treatment GL [2024]
When feasible, enteral nutrition should be the strategy of choice for preoperative
optimization in patients with CD [EL3]
M

Malnutrition is common in patients with CD requiring surgery and is a risk factor for adverse
d

postoperative outcomes and complications. Systematic nutritional risk screening [body mass
e

index, unintentional weight loss, reduced dietary intake,


pt

illness severity] together with perioperative nutritional support may mitigate the perioperative
risks associated with malnutrition. An ECCO consensus and topical review on perioperative
ce

dietary therapy in CD concluded than exclusive enteral nutrition [EEN] represented a valid
preoperative optimization strategy for reducing complications and improving nutritional status
Ac

in patients with CD, likely by modulating inflammatory status and improving microbial
composition.145,163-165
The benefits of preoperative EEN have been consistently reported, leading to a marked
reduction of postoperative morbidity [21.9% vs 73.2%; OR: 0.09; 95% CI: 0.06–0.13; p <
0.01], although data on biochemical optimization are still debatable.166-168 Conversely, the
role of parenteral nutrition [PN] in the preoperative optimization strategy is more debated. 169
Importantly, EEN requires dedicated nutritional support and high patient compliance to be
successful.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
The use of PN in the perioperative period should be reserved for patients unable to tolerate
EEN, do not meet their nutritional requirements with EEN, or in which EEN is
contraindicated.170 In a recent prospective multicentric cohort study, preoperative EEN
reduced morbidity for infection and temporary stoma requirement in malnourished patients
with CD 164 . In another recent cohort study, patients receiving preoperative PN had
significantly lower rates of non-infectious complications [OR: 0.07; 95% CI: 0.01–0.80; p =

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


0.03]. A subset of frail patients with severe CD who did not tolerate EEN and required PN
presented a similarly high rate of IASC and primary stoma as when upfront surgery was
elected. Hence, the advantage of providing PN to this subgroup of frail patients is

t
questionable, as these patients may benefit from an early surgical approach followed by

ip
nutritional replacement.171 Therefore, early surgery with postoperative optimization may be

cr
considered in frail, severely ill patients who do not tolerate EEN and accept a diverting
stoma.

us
an
Statement 4.9 ECCO CD Treatment GL [2024]
We recommend that steroids should be tapered whenever possible before surgery to
reduce the risk of complications [EL2]
M

Previous ECCO guidelines have reported that treatment with >20 mg prednisolone daily for
d

>6 weeks increases the risk of postoperative septic complications.11,18,172 Whilst there is no
e

large RCT confirming this position, one large multicentre cohort study and numerous
pt

retrospective cohort studies have identified this risk [summarized in 3 meta-analyses].173-175


Indeed, preoperative steroid use was a significant risk factor for major complications,
ce

including an overall increased risk of postoperative complications [OR: 1.41; 95% CI: 1.07–
1.87] and a specifically increased risk of postoperative IASC [OR: 1.68; 95% CI: 1.24–
2.28].174,176 Patients who received >40 mg perioperative oral steroids had the highest risk of
Ac

overall complications [OR: 2.04; 95% CI: 1.28–3.26]. A meta-analysis confirmed an almost
doubling of total wound infections [OR: 1.70; 95% CI: 1.38–2.09].173 Similar to the results
from the large multicentre cohort study, an increased risk for anastomotic leak was also
observed [OR: 1.51; 95% CI: 1.02–2.25].175
Steroids should be reduced before surgery as part of a preoperative optimization strategy in
combination with nutritional optimization and drainage of sepsis. If this is not possible,
consideration should be given to a staged procedure with a temporary stoma.
Manuscript Doi: 10.1093/ecco-jcc/jjae089

Statement 4.10 ECCO CD Treatment GL [2024]


We recommend against cessation of biologics prior to surgery as current evidence
suggests that preoperative treatment with anti-TNF therapy [EL3], vedolizumab [EL3],
and ustekinumab [EL4] does not increase the risk of postoperative complications in

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


patients with CD undergoing abdominal surgery

Anti-TNF therapy

t
Use of biologicals in patients with CD undergoing surgery remains controversial. Concern

ip
exists over the desired modulation of the immune response and the potential to increase
postoperative complications. Several retrospective studies regarding anti-TNF agents have

cr
been published over the last 20 years. Some suggested an increased incidence of

us
complications in patients receiving anti-TNF agents preoperatively and other studies showed
no difference. Several meta-analyses have also reported varying conclusions.177 Several
prospective studies also reached inconsistent conclusions. This variation probably
an
represents heterogenous populations, different outcomes, and inconsistent definitions of
outcomes. Most evidence is concentrated on infliximab and adalimumab.177 The PUCCINI
M

trial is the largest prospective trial to date and revealed no difference in the rate of any
infection between patients using biological therapy and those not.178 Detectable preoperative
serum concentrations of anti-TNF agents also did not increase the risk of surgical site or
d

overall infection rates.178 Hence, anti-TNF therapy can be continued prior to surgery.
e
pt

Vedolizumab
Although initial retrospective data suggest that VDZ leads to an increased risk of
ce

postoperative infection, subsequent studies showed no increased risk. These data were
confirmed by most, but not all, recent meta-analyses.179-182 The latest of these showed no
significant differences in overall complications [OR: 1.04; 95% CI: 0.48–2.24],180 infectious
Ac

complications [OR: 1.00; 95% CI: 0.37–2.69], or surgical site infections [OR: 1.45; 95% CI:
0.33–6.32] for those receiving VDZ preoperatively. Therefore, VDZ can be continued prior to
surgery.

Ustekinumab
Although one meta-analysis focused on ustekinumab and postoperative complications, the
comparator was patients receiving anti-TNF therapy.183 No difference in complications and
infectious complications were identified. The only cohort study comparing ustekinumab with
Manuscript Doi: 10.1093/ecco-jcc/jjae089
non-biological therapy revealed that preoperative use of ustekinumab is an independent risk
factor for intra-abdominal sepsis [OR: 2.93; 95% CI: 1.16–7.40; p = 0.02].184 Although further
studies are required to confirm the safety of ustekinumab and surgery, current data suggest
that cessation before surgery may not be necessary.

There is no available evidence of the possible impact of preoperative use of CZP,

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


rizankizumab, or JAK inhibitors on postoperative morbidity in patients with CD undergoing
abdominal surgery. The safety of continuing newer biological agents prior to surgery remains
unknown.

t
ip
Practice points

cr
Preoperative optimization is a key element in successful management of complex situations
and chronic disease. Many aspects of optimal perioperative care are generic and common to

of CD [venous thromboembolism prophylaxis, us


all abdominal procedures,185 although some aspects are particularly important in the context
nutrition, iron management, drug
an
management, minimally invasive approaches, and bowel- and sphincter-sparing
techniques].186,187 While high-dose steroids should be tapered to reduce surgical morbidity,
M

current biological therapy can safely be continued perioperatively.


e d

4.1 Surgical techniques


pt

Statement 4.11 ECCO CD Treatment GL [2024]


ce

We recommend a laparoscopic approach as the first line in abdominal surgery for CD


[EL2]
Ac

A Cochrane review of two randomized trials188,189 showed no difference in complications


between laparoscopic and open surgery for CD. A more recent review190 showed a benefit
for patients operated by laparoscopy, with fewer complications and lower rate of incisional
hernia. This review included both randomized trials and observational studies. While this
may potentially introduce some bias, based on strong evidence for the benefits of
laparoscopy, especially in relation to reduced adhesions, the current evidence
strongly supports recommending laparoscopy as the first-line approach. Laparoscopic
resection for recurrent CD is also feasible but is associated with higher risk for conversion.191
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Importantly, in the absence of expertise to perform laparoscopic surgery, emergency
operations should not be delayed.

Statement 4.12 ECCO CD Treatment GL [2024]


We recommend laparoscopic resection as an alternative to infliximab [EL2] or

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


adalimumab [EL4] therapy in patients with limited terminal ileal or ileocecal disease

A randomized, controlled, open-label, multicentre trial assigned 143 patients with non-

t
ip
stricturing CD of the terminal ileum to receive either laparoscopic ileocecal resection [n = 73]
or infliximab [n = 70]. At 12-month follow-up, quality of life and body-image perception were

cr
comparable.192 Patients treated with infliximab had fewer days of sick leave from work.
Serious complications related to treatment occurred in 4 resected patients versus 2 in the

us
anti-TNF group. Crossover among groups was needed in 37% of patients treated with
infliximab and in 26% of those who underwent surgery. Long-term data from the randomized
an
trial revealed no surgical recurrence in the surgery group after 5 years, while 50% in the anti-
TNF group had surgery at 5 years.193 A recent meta-analysis suggests reduced risk of
overall and surgical recurrence and reduced use of postoperative biologic therapy if surgery
M

is performed early.194 Based on these data, early surgery has a benefit in patients with
limited terminal ileal CD and represents a reasonable alternative to escalating medical
d

therapy. Patients should be advised early about a surgical option.


e
pt

Statement 4.13 ECCO CD Treatment GL [2024]


We suggest stapled side-to-side anastomoses in small-bowel or ileocolic resections
ce

for CD [EL3]
Ac

Surgeons place great importance on the technical aspects of their work, which can be
influenced by various factors, including their training, personal experience, available
resources, and the clinical scenario. The choice of the optimal anastomosis technique in
small-bowel and ileocolic resections has been a subject of controversy. In recent years,
there has been a growing body of evidence supporting the use of side-to-side anastomosis,
and this support has been consistent over time.
A significant meta-analysis on 661 patients operated for cancer and CD revealed a
significantly higher anastomotic leak rate in end-to-end anastomoses compared with side-to-
side anastomoses [OR: 4.37; p = 0.02]. This was also observed in the subgroup of ileocolic
Manuscript Doi: 10.1093/ecco-jcc/jjae089
anastomoses [OR: 3.8; p = 0.05].195 Furthermore, overall postoperative complications [OR:
2.64; p <0.001] and hospital stay length were higher [by 2.81 days; p = 0.007] when an end-
to-end anastomosis was performed. A subsequent meta-analysis confirmed the superiority
of side-to-side anastomosis in overall postoperative complications [OR 0.6; p = 0.01].
However, there were no statistically significant differences in leak rates, endoscopic and
symptomatic recurrence, or reoperation for recurrence.196

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


A meta-analysis compared 396 stapled side-to-side anastomoses with 425 hand-sewn end-
to-end anastomoses and found that stapled side-to-side anastomoses outperformed in all
endpoints, namely overall postoperative complications [OR: 0.54; 95% CI: 0.32–0.93],

t
anastomotic leak [OR: 0.45; 95% CI: 0.20–1.00], recurrence [OR: 0.20; 95% CI: 0.07–0.55],

ip
and reoperation for recurrence [OR: 0.18; 95% CI: 0.07–0.45].197

cr
A network meta-analysis of 11 trials and 1113 patients further substantiated the superiority
of stapled side-to-side anastomosis regarding overall complications, clinical recurrence, and

us
reoperation for recurrence. However, the choice of anastomosis technique did not seem to
affect leak rates, surgical-site infections, mortality, or length of hospital stay.198 A more
an
recent systematic review suggested that stapled side-to-side anastomoses may lower the
risk of surgical recurrence in CD, potentially reducing rates of reoperations compared with
hand-sewn end-to-end anastomoses [OR: 0.22; 95% CI: 0.05–0.95].199 In case of
M

emergency bowel resection, a retrospective study involving 92 bowel resections


recommended use of stapled side-to-side anastomoses, which was associated with fewer
endoscopic recurrences than hand-sewn end-to-end anastomoses [OR: 38.12; p = 0.01].200
d

This was corroborated by another retrospective study.201 However, a recent multicentre,


e

retrospective, observational study examining 427 intestinal anastomoses in CD found no


pt

significant difference in postoperative complications.202


Overall, the quality of the studies included in systematic reviews and meta-analyses was
ce

notably limited, with only a minority of patients participating in RCTs and heterogenous
populations studied. Despite this limitation, the prevailing consensus leans toward a
Ac

preference for stapled side-to-side anastomosis, which is associated with lower rates of
postoperative complications and allows for an intracorporeal anastomosis. Furthermore, it
was suggested that the diameter of the anastomosis may be a significant risk factor for
recurrence, as a wider anastomosis is thought to be associated with a reduced likelihood of
clinical and surgical recurrences. Importantly, the width of the anastomosis is determined by
its inlet, more than by the length of a staple line or a suture line. Endoscopic appraisal of an
early recurrence should consider the type of anastomosis healing. Indeed, stapled [everted
mucosa] and hand-sewn [inverted mucosa] have a different healing pattern and healing time,
Manuscript Doi: 10.1093/ecco-jcc/jjae089
which should neither be confused endoscopically with an early recurrence, nor lead to
overtreatment.

Statement 4.14 ECCO CD Treatment GL [2024]


We suggest that the Kono-S anastomosis can be an alternative surgical approach to

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


other types of anastomoses after ileocecal resection [EL3]

Kono-S anastomosis was first described in 2011 as a new hand-sewn anti-mesenteric

t
ip
functional end-to-end anastomosis designed with the aim to reduce anastomotic CD
recurrence after ileocecal resection.

cr
In the first retrospective study,203 Kono-S anastomosis was associated with a reduction in
both median endoscopic recurrence score [Rutgeerts’ score] and surgical recurrence rate at

us
5 years with no safety issues. These findings were then confirmed by a larger international
multicentre retrospective study including 187 patients, reporting a 10-year surgical
an
recurrence-free rate of 98.6%.204
Performing a Kono-S anastomosis was associated with longer operative time, similar short-
term outcomes, and likely lower endoscopic recurrence rate than side-to-side
M

anastomosis.205 In another two retrospective cohort studies following patients for up to 5


years, Kono-S anastomosis was associated with a lower leak rate than end-to-end
d

anastomosis206 or stapled anastomosis207, which in the authors’ opinion could explain the
lower surgical recurrence rate observed in the long term.
e

More recently, early results from the first RCT208 comparing Kono-S and side-to-side
pt

anastomoses demonstrated a significant reduction in the 6-month endoscopic recurrence


rate and mean Rutgeerts’ score, comparable postoperative outcomes, and a trend toward a
ce

reduced surgical recurrence rate, although this was not statistically significant. This and
other trials are still ongoing with definitive results expected in the near future.
Ac

Several meta-analyses, including the aforementioned RCT and observational studies,


concluded that Kono-S anastomosis was associated with a reduced endoscopic recurrence
rate and comparable short-term outcomes.199,209,210 More limited evidence suggested a
reduction in surgical recurrence and leak rate in Kono-S anastomosis than with conventional
anastomoses. However, the most recent prospective study on Kono-S did not confirm a
reduction in endoscopic recurrence rates and reported similar Rutgeerts’ scores and clinical
recurrence rates between conventional anastomosis and Kono-S.211 Therefore, a definitive
conclusion on the benefit of a Kono-S anastomosis cannot yet be made. Multicentre RCTs
Manuscript Doi: 10.1093/ecco-jcc/jjae089
are currently ongoing across the US and Europe and will probably provide definitive answers
on the role of Kono-S anastomosis.212-214

Statement 4.15 ECCO CD Treatment GL [2024]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


There is insufficient evidence to recommend extensive mesenteric excision in
surgery for ileocecal CD [EL4]

t
Extensive mesenteric excision may reduce the incidence of recurrence after resection by

ip
possibly removing a ‘sump’ of pro-inflammatory substances from the vicinity of the
anastomosis. The current evidence for this is weak. Two systematic reviews addressed

cr
extensive mesenteric excision199,209 but both only included one small historical case-control
study.215 This single case-control study compared 30 patients undergoing extensive

us
mesenteric excision with a surgical recurrence rate of 2.9% at 5 years to a historical control
group of 34 patients who had a 5-year recurrence rate of 40%.215 Several ongoing trials
an
address the possible benefit of a wide mesenteric excision in the context of CD. Such an
excision cannot currently be recommended in routine care.
M

Statement 4.16 ECCO CD Treatment GL [2024]


d

We suggest a temporary stoma formation in patients with CD if they are not


e

sufficiently optimized for surgery [EL4]


pt

The decision to create a stoma [primary anastomosis and protective stoma or no


ce

anastomosis and split stoma] in the context of steroid intake relies mostly on clinical grounds
and experience. There are limited data comparing strategies between primary anastomosis
Ac

or secondary anastomosis in patients with CD treated with steroids.216 However, prolonged


[>6 weeks] and high-dose [≥20 mg prednisolone equivalent] steroid use are associated with
postoperative infectious complications, including anastomotic leakage.149,174,217,218

Statement 4.17 ECCO CD Treatment GL [2024]


We recommend strictureplasty as an alternative treatment option to resection in
small-bowel CD [EL2]
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Location of CD in the ileum, use of biologics before surgery, and non-conventional
strictureplasty [SP] predict early site-specific recurrence after SP.219,220 However, procedure-
specific recurrence rates are available only for some SP techniques.221 The wide range of
recurrence rates after SP [3–25%] reflects the variability of the population case mix and most
importantly of the follow-up length.220 An extended follow-up time [>5 years] is mandatory to
appraise the true outcome of SP.220 Morbidity and postoperative hospital length of stay were

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


similar for bowel resection and SP.221-223 Overall, the results of SP compare well with the
recurrence rate after bowel resection, while preserving bowel length.

t
ip
Statement 4.18 ECCO CD Treatment GL [2024]

cr
We suggest segmental colectomy in selected cases of colonic CD [EL4]

us
When a single colonic segment is affected, a segmental colectomy may be the
recommended course of action. On the other hand, the involvement of multiple colon
an
segments generally indicates [sub]total colectomy. A meta-analysis compared 223 cases of
subtotal or total colectomies with ileorectal anastomosis and 265 cases of segmental
colectomies in CD.224 In this analysis, there were no significant differences in recurrence
M

rates, complications, or need for a permanent stoma. However, it is worth noting that
recurrence occurred on average 4.4 years later in patients who underwent a subtotal or total
d

colectomy [p < 0.001].


A recent meta-analysis included patients who underwent segmental colectomy [n = 500],
e

subtotal colectomy [n = 510], or total proctocolectomy [n = 426]. Complications were more


pt

frequent after segmental colectomy compared with subtotal colectomy [OR: 2.84; 95% CI:
1.16–6.96] and after proctocolectomy compared with subtotal colectomy [OR: 0.19; 95% CI:
ce

0.09–0.38].225 This indicates that subtotal colectomy is generally considered a safer


procedure, although segmental colectomy resulted in fewer patients requiring permanent
Ac

stoma [OR: 0.52; 95% CI: 0.35–0.77]. Subtotal colectomy had higher rates of CD recurrence
[OR: 3.53; 95% CI: 2.45–5.10] and need for repeat surgery [OR: 3.52; 95% CI: 2.27–5.44]
than total proctocolectomy. However, no significant difference in recurrence was observed
between segmental and subtotal colectomy. In rare situations where two distinct colon
segments are affected, it may be worth considering two segmental resections as an
alternative to subtotal colectomy, particularly for patients who have extensive small-bowel
loss. 11
A recent retrospective analysis that included 55 [sub]total colectomies and 30 segmental
colonic resections indicated a trend towards increased postoperative complications after
Manuscript Doi: 10.1093/ecco-jcc/jjae089
segmental colectomy [Clavien-Dindo grade ≥ III] of 13.3% versus 7.3% after [sub]total
colectomy. Additionally, there was a trend toward higher rates of hospital readmissions
[13.3% vs 1.8%] and reinterventions [13.3% vs 3.6%] after segmental resection compared
with [sub]total colectomy.226 Another recent multicentre retrospective study including 687
patients concluded that segmental resection was a safe option compared with total
colectomy with the additional benefit of reducing ostomy formation without increasing the risk

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


of surgical recurrence, particularly in the era of biologics.227 However, the heterogeneity of
the included patients was a limitation of this analysis.
A further retrospective, single-centre study included 200 patients who underwent segmental

t
colectomy. A surgical recurrence rate of 31% was observed. Risk factors of recurrence and

ip
subsequent [sub]total colectomy in multivariate analysis were the presence of three or more

cr
affected sites [HR: 2.47; 95% CI: 1.22–5.00; p = 0.018] and presence of perianal disease
[HR: 3.23; 95% CI: 1.29–8.07; p = 0.006].228

us
In summary, the extent of colonic resection is determined by the clinical presentation
[elective vs emergency surgery] and by the number of colonic segments involved
an
[unisegmental vs pancolitis]. Segmental colectomy is generally favoured whenever feasible
as this does not increase the risk of recurrence, particularly in the modern era of biologics
and when other risk factors for recurrence [such as number of affected locations and
M

presence of perianal disease] are absent.


e d

Statement 4.19 ECCO CD Treatment GL [2024]


pt

We suggest proctocolectomy as a treatment for CD-associated colorectal cancer or


high-grade dysplasia and segmental colectomy followed by endoscopic surveillance
ce

in selected cases [EL3]

Patients with chronic inflammation of the large bowel are at an increased risk of
Ac

development of colorectal cancer [CRC], as described in an European Evidence-based


Consensus: IBD and Malignancies.229 Two meta-analyses of cohort studies have clarified the
increased risk of CRC in patients with IBD.230,231 The pooled standardized incidence ratio
[SIR] for CRC was 1.7 [95% CI: 1.2–2.2] in all patients with IBD and 1.9 in CD [95% CI: 1.4–
2.5]. However, the HR of CRC increased in all age groups [HR: 1.40; 95% CI: 1.27–1.53],
consistent with a recent Scandinavian cohort study.231 There was higher risk with extensive
colitis and younger IBD diagnosis [age <30 years], with a SIR of 6.4 [95% CI: 2.4–17.5] and
Manuscript Doi: 10.1093/ecco-jcc/jjae089
7.2 [95% CI: 2.9–17.8], respectively. Cumulative risks of cancer were 1%, 2%, and 5% after
10, 20, and >20 years disease duration, respectively.
These reports indicate that the risk of CRC is increased in patients with IBD, but not to the
extent previously reported and not in all patients.
In a Danish cohort,232 CRC patients with CD had a lower frequency of Duke’s A- and B-stage
tumours [36% vs 42%] and a higher frequency of Duke’s C- [31% vs 27%] and D-stage

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


tumours [23% vs 21%], whereas the frequency of unknown-stage tumours [10%] resembled
that of non IBD-related CRC. The 5-year adjusted mortality rate ratios for patients with
ulcerative colitis [UC] or CD were increased by 1.14 [95% CI: 1.03–1.27] and 1.26 [95% CI:

t
1.07–1.49], respectively, compared with patients without IBD. In contrast, in an Irish

ip
population-based study, patients with IBD-related CRC were about 7 years younger at

cr
cancer diagnosis than patients with non-IBD CRC but survived about 3 years longer. Older
age, male sex, smoking, and advanced CRC grade and stage were independently

us
associated with shorter survival times. When propensity score matching was used to
analyse outcomes, the survival times of CRC patients with and without IBD were not
significantly different.233 Taken together, these results reveal that patients with IBD tend to
an
develop CRC at younger ages than patients without IBD. However, no effect of IBD on
patient survival has been consistently demonstrated.
M

The risk of CRC in CD increases with longer disease duration, extent of colitis, a familial
history of CRC, coexistent primary sclerosing cholangitis, and the degree and duration of
inflammation. CRC in CD tends to have higher histological grade and more often
d

mucinous/signet-ring histological characteristics.11,229,234-236


e

The previous ECCO-ESCP Consensus on Surgery for CD11 recommended proctocolectomy


pt

in fit patients with preoperative diagnosis of cancer or high-grade dysplasia due to the
multifocal nature of dysplasia in CD colitis and the reported high rate of metachronous colon
ce

cancer after segmental surgical resection.237,238 However, caution is required when


comparing cancer incidence between patients with CD undergoing regular colonoscopies
Ac

and the general population offered cancer screening; lead time bias may overestimate a
possible causal association. Furthermore, the onset of CD is often unclear, while many
cancers are diagnosed concomitantly or immediately after a diagnosis of CD and thus have
a debatable association with CD. Indeed, the incidence of metachronous CRC after
segmental resection is much lower than initially thought239-241 and the prior reported high rate
of metachronous cancer may be attributed to inadequate surgery or even underestimation of
synchronous tumours. Furthermore, most of the available data originate from the early
1970s, when both endoscopic and therapeutic interventions were very different from current
standards.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Therefore, segmental resections and endoscopic surveillance may be proposed in selected
patients after proper consent or in patients who are at high risk for surgery.
Importantly, patients with CRC in CD should be operated according to the principles of
oncological surgery, including adequate lymphadenectomy.242,243 The same principles of
oncological surgery should be considered in the presence of a colonic stenosis and long-
lasting extensive CD colitis can easily be missed upon endoscopic biopsy. Strictureplasty is

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


not recommended in this context.11,237

t
Statement 4.20 ECCO CD Treatment GL [2024]

ip
We suggest a defunctioning stoma for non-acute refractory CD colitis to delay or
avoid the need for colectomy [EL5]

cr
us
The following two options may be discussed in the presence of refractory CD colitis: a
[sub]total colectomy, particularly as a potentially life-saving procedure in fulminant colitis,
and a defunctioning ileostomy to divert the faecal stream and allow for remission, together
an
with intensified medical therapy 244. A diverting ileostomy may delay further procedures,
facilitate perioperative optimization, and allow for a limited resection if required at a later
M

stage [i.e. segmental colectomy]. The clinical scenario in which a diverting stoma is
performed to aid the management of extensive perineal disease is covered elsewhere and is
not the focus of the present statement.
d

The literature preceding the biologic era reported initial remission rates of up to 90%245-248
e

following creation of a defunctioning stoma, which is more than the 50–80% reported in more
pt

recent series.249,250 Lasting restoration of bowel continuity/stoma reversal was effective in up


to two-thirds of patients but was much lower when perianal disease was also present [i.e.
ce

29–42%.]6,7 Surgical complications of defunctioning stoma creation were in the expected


range of 3–10% for stoma prolapse/hernia and <5% for renal failure due to high-output
stoma.250 Further bowel resection was reported in up to half of the patients in recent
Ac

series.249,250 Risk factors for proctocolectomy were severe refractory perianal disease,
requirement for combined medical therapy, and a history of more than one biologic drug. For
these patients, early colectomy and end ileostomy [as opposed to a defunctioning ileostomy]
may be discussed.
The following factors should be taken into account when a proctocolectomy is required and
ileal pouch anal anastomosis is considered. In general, more patients have postoperative
pelvic sepsis and a higher pouch failure rate when compared with patients with IPAA for UC.
Patients also have more bowel movements and daytime incontinence when compared with
Manuscript Doi: 10.1093/ecco-jcc/jjae089
patients with IPAA for UC. It is worth noting that in selected patients with isolated CD colitis
without small-bowel or perianal involvement, outcomes similar to patients with IPAA for UC
can be obtained [no difference in pelvic sepsis, stool frequency, incontinence, score on
quality-of-life surveys, or pouch failure].251-256

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Statement 4.21 ECCO CD Treatment GL [2024]
We recommend CD surgery is performed in high-volume IBD centres [EL3]

t
ip
The data and appreciation of the benefit of centralization of IBD surgery in high-volume
centres is controversial. Nationwide studies suggested lower mortality in high-volume

cr
centres, although patients who are frailer and sicker are over-represented in these
centres.257,258 The definition of a high-volume, expert centre and of referral criteria are

us
particularly controversial. ECCO has defined quality-of-care criteria and standards for the
care of IBD patients, including patient volume, in a position paper.259
an
M

Practice points

When surgery becomes necessary, it is important to thoroughly assess the bowel, ideally
d

preoperatively with MRI enterography. MRI enterography may reveal a distinction between
e

inflammatory strictures [amenable to intensified medical therapy] and fibrotic strictures.


Systematically assessing the bowel during surgery may identify further strictures. To
pt

maximize bowel preservation, the IBD surgeon should be familiar with the different kinds of
strictureplasties, including non-conventional strictureplasties. Nonetheless, strictureplasty of
ce

the colon is not recommended.260


Ac

The anastomotic technique of choice is not firmly established, although a stapled side-to-
side anastomosis is suggested in small-bowel or ileocolic resections. While segmental
colectomy is advisable when a single colon segment is involved, an oncologic
proctocolectomy is recommended when colonic dysplasia or a neoplasia is identified.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
4.2 Postoperative management

Statement 4.22 ECCO CD Treatment GL [2024]


We recommend endoscopic surveillance within 6–12 months after surgical resection
in CD [EL2]

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


A systematic review that included one unblinded RCT and four retrospective cohort studies
revealed a lower recurrence rate in the endoscopy-based management group than in the

t
control group.261 Similarly, another systematic review concluded that mucosal changes can

ip
be observed in up to 73% of cases within 1 year after surgical resection when patients
undergo endoscopic monitoring.262

cr
In a study that randomized 174 patients in a 2:1 ratio, some underwent colonoscopy at 6

us
months with active therapy while others did not undergo colonoscopy and received standard
care. At the 18-month timepoint, clinical recurrence was lower [37.7% vs 46.1%; RR 0.82;
95% CI: 0.56–1.18] in the colonoscopy group and endoscopic recurrence was higher in the
an
group that received standard care compared with those under active surveillance [67% vs
49%; p = 0.03].263
M

Another systematic review that included 26 prospective studies reported the presence of
mucosal lesions in up to 70% of cases with a median endoscopic follow-up of 12 months.
Notably, more than 50% of these lesions were located at the anastomotic site. Interestingly,
d

despite receiving medical treatment, 41% of patients exhibited significant lesions.264 These
e

findings are consistent with similar results presented by other studies.265,266 Endoscopic
pt

monitoring within 6–12 months following surgical resection allows for identification of patients
who may experience disease recurrence, even with ongoing medical therapy, enabling
ce

proactive intervention.
Ac

Statement 4.23 ECCO CD Treatment GL [2024]


We suggest postoperative prophylactic medical therapy after ileocolic resection in
patients with CD at high risk of recurrence [EL3]

Prophylaxis for postoperative recurrence is recommended in patients at high risk for


recurrence. Thiopurines appear to be more effective than placebo in preventing
postoperative recurrence according to different studies.267 Infliximab was more effective than
placebo in preventing endoscopic, but not clinical, recurrence in the prospective PREVENT
Manuscript Doi: 10.1093/ecco-jcc/jjae089
268
trial. Overall, anti-TNF agents are the most effective therapy in preventing postoperative
endoscopic recurrence.269 More recent evidence from observational studies described the
efficacy of biologics with different mechanisms of action [ustekinumab and VDZ] in
prevention of recurrence.270 A prospective study presented in abstract form demonstrated
that VDZ was more efficacious than placebo in preventing endoscopic recurrence. Patients
treated with VDZ had a 77.8% chance of having a lower Rutgeerts’ score than patients with

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


placebo 6 months after an ileocolic resection [p < 0.0001].271 A retrospective multicentre
study from Spain analysed postoperative recurrence rates in 40 patients treated with
ustekinumab and 25 treated with VDZ [all had previous exposure to anti-TNF]. The

t
cumulative probability of clinical postoperative recurrence at 12 months after surgery was

ip
32% and 30% for ustekinumab and VDZ, respectively. The rate of endoscopic recurrence

cr
was 42% for ustekinumab and 40% for VDZ.272 High-risk patients include those that smoke,
have penetrating disease, or present with an IASC, fistula, or both.273,274

us
an
Statement 4.24 ECCO CD Treatment GL [2024]
We recommend extended thromboembolism prophylaxis following hospital discharge
after CD surgery [EL2]
M

Although thromboprophylaxis is well documented in patients who have surgery after CRC,
d

there is limited evidence in IBD. A recent systematic review suggested that postoperative
DVT risk was similar in IBD to that of patients with advanced CRC. The risk was highest in
e

those who had a subtotal colectomy or a proctectomy. The dosage of low molecular weight
pt

heparin was also assessed in a single-centre study, suggesting that a dose of 4000 IU/day
of low molecular weight heparin was insufficient for IBD patients.275 A minimal duration of
ce

thromboprophylaxis of 2 weeks postoperatively was suggested.276


Ac

5. Conclusion

There are many options and crossroads in decision making for surgery in CD. Some
approaches have been tested over time and were described in these surgical guidelines.
Although sufficient training, technical expertise, and an adequate caseload to achieve and
maintain subspecialization in IBD surgery are important, the key to success in managing CD
is a multidisciplinary team, as no specialist alone can solve the CD equation. The present
Manuscript Doi: 10.1093/ecco-jcc/jjae089
guidelines have been written with this interdisciplinary approach in mind and summarize the
currently available knowledge. The degree of certainty in some aspects of surgery for CD is
closer to eminence than evidence, thus paving the way for further research and better
answers. Consideration of patient lifestyle preference is integral to shared decision making
and key to achieve best standard of care. Revealing gaps in evidence is the first step, as
research focused on clinical needs and gaps in the current evidence will inform guideline

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


updates. Meanwhile, dynamic integration of gains in knowledge into the ECCO e-Guide will
allow for rapid dissemination. Guidelines provide guidance to the clinician, who adapt expert
knowledge, generic evidence and patient lifestyle preference to individualize care. It is hoped

t
that the present work will contribute to optimizing care for patients with CD.

ip
cr
us
an
M
e d
pt
ce
Ac
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Funding

This project was initiated, funded, and supported by the ECCO.

Acknowledgements

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


We gratefully thank Dr Paul Freudenberger for the literature search and full-text retrieval;
Torsten Karge for support on informatics and on the web Guidelines platform; and the ECCO
Office for logistical and coordination support.

t
ip
cr
We gratefully thank the EFCCA patient representatives Bastien Corsat, Xavier Donnet,
Evelyn Gross, Antonio Valdivia, Janek Kapper, and Lucie Lastikova who proactively

us
collaborated in the development of these Guidelines.
an
We thank Ibrahim Ethem Gecim for his work in the abstract screening process.
M

We would like to thank and acknowledge the ECCO National Representatives and additional
d

reviewers, who acted as external reviewers and provided suggestions on the


e

recommendations and supporting text to this document: Pascal Juillerat, Allesandra Soriano,
Mark Samaan, Tiago Cúrdia Gonçalves, Edoardo Savarino, Federica Furfaro, Davide
pt

Giuseppe Ribaldone, Gulustan Babayeva-Sadiqova, Aurelien Amiot, Gianmichele Meucci,


Iago Rodríguez Lago, Mathieu Uzzan, Gerassimos Mantzaris, Beatriz Gros Alcalde, Vito
ce

Annese, Eduard Brunet Mas, Maria Jose Garcia, Eirini Zach, John Marshall, Carla Felice,
Maha Maher, Paul Pollack, Andreas Blesl, Negreanu Lucian, Ferdinando D'Amico, Dimitrios
Ac

Karagiannis, Patrick Allen, Oliver Bachmann, Imerio Angriman, Anna Kagramanova,


Dahham Alsoud, Natália Queiroz, Usha Chauhan, Petra Golovics, Chen Sarbagili, Lorenzo
Bertani, Ulf Helwig, Clas-Göran af Björkesten, Ante Bogut, Anthony Buisson, Ignacio
Catalan-Serra, Aslı Çifcibaşı Örmeci, Marco Daperno, Mihai Mircea Diculescu, Dana
Duricová, Piotr Eder, Magdalena Gawron-Kiszka, Ayal Hirsch, Ondrej Hradsky, Hendrik
Laja, Sara Onali, Samuel Raimundo Fernandes, Christian Philipp Selinger, Helena Tavares
de Sousa, Svetlana Turcan, Sophie Vieujean, and Yamile Zabana.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Authors’ contribution

Michel Adamina, Pär Myrelid, Hannah Gordon, and Tim Raine coordinated the project; Silvia
Minozzi provided expert methodology advice, trained the working group members, and
performed the analysis of data; Uri Kopylov, Bram Verstockt, Maria Chaparro, Christianne
Buskens, and Janindra Warusavitarne coordinated the working groups; all the Authors listed

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


contributed to the identification of relevant data, data interpretation, and drafted and
discussed the final recommendations; all Authors participated in the final Consensus; Michel
Adamina and Pär Myrelid drafted this manuscript.

t
ip
Conflict of interests

cr
ECCO has diligently maintained a disclosure policy of potential conflicts of interest [CoI]. The

us
conflict-of-interest declaration is based on a form used by the International Committee of
Medical Journal Editors [ICMJE]. The CoI statement is not only stored at the ECCO Office
an
and the editorial office of JCC but is also open to public scrutiny on the ECCO website
[https://www.ecco-ibd.eu/about-ecco/ecco-disclosures.html], providing a comprehensive
overview of potential conflicts of interest of authors.
M
d

Disclaimer
e

The ECCO Guidelines are targeted at health care professionals only and are based on an
pt

international consensus process.


ce

This process includes intensive literature research as explained in the methodology section
Ac

and may not reflect subsequent scientific developments, if any, until the next Guidelines
update is prepared. Readers of the Guidelines acknowledge that research about medical
and health issues is constantly evolving and diagnoses, treatments, and dose schedules for
medications are being revised continually. Therefore, the European Crohn´s and Colitis
Organisation (ECCO) encourages all readers to also consult the most up-to-date published
product information and data sheets provided by the manufacturers as well as the most
recent codes of conduct and safety regulations.

Any treatment decisions are to be made at the sole discretion and within the exclusive
responsibility of the individual clinician and should not be based exclusively on the content of
Manuscript Doi: 10.1093/ecco-jcc/jjae089
the ECCO Guidelines. The European Crohn´s and Colitis Organisation (ECCO) and/or any
of its staff members and/or any consensus contributor may not be held liable for any
information published in good faith in the ECCO Consensus Guidelines. ECCO makes no
representations or warranties, express or implied, as to the accuracy or completeness of the
whole or any part of the Guidelines. ECCO does not accept, and expressly disclaims,
responsibility for any liability, loss or risk that may be claimed or incurred as a consequence

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


of the use or application of the whole or any part of the Guidelines.

t
ip
When the Guidelines mention trade names, commercial products or organizations, this does
not constitute any endorsement by ECCO and/or any consensus contributor.

cr
us
an
M
e d
pt
ce
Ac
Manuscript Doi: 10.1093/ecco-jcc/jjae089
References

1. Ng SC, Shi HY, Hamidi N, et al. Worldwide incidence and prevalence of inflammatory
bowel disease in the 21st century: A systematic review of population-based studies.
Lancet 2018;390:2769-78.
2. Frolkis AD, Dykeman J, Negron ME, et al. Risk of surgery for inflammatory bowel

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


diseases has decreased over time: A systematic review and meta-analysis of
population-based studies. Gastroenterology 2013;145:996-1006.
3. Adamina M, Gerasimidis K, Sigall-Boneh R, et al. Perioperative dietary therapy in
inflammatory bowel disease. J Crohns Colitis 2019.

t
4. Bouguen G, Peyrin-Biroulet L. Surgery for adult crohn's disease: What is the actual

ip
risk? Gut 2011;60:1178-81.
5. Burke JP, Velupillai Y, O'Connell PR, Coffey JC. National trends in intestinal resection

cr
for crohn's disease in the post-biologic era. Int J Colorectal Dis 2013;28:1401-6.
6. Bemelman WA, collaborators SE. Evolving role of ibd surgery. J Crohns Colitis

us
2018;12:1005-7.
7. Lightner AL, Shen B. Perioperative use of immunosuppressive medications in patients
with crohn's disease in the new "biological era". Gastroenterol Rep (Oxf) 2017;5:165-
77.
an
8. Reinglas J, Gonczi L, Kurt Z, Bessissow T, Lakatos PL. Positioning of old and new
biologicals and small molecules in the treatment of inflammatory bowel diseases.
World J Gastroenterol 2018;24:3567-82.
M

9. Reinglas J, Restellini S, Gonczi L, et al. Harmonization of quality of care in an ibd


center impacts disease outcomes: Importance of structure, process indicators and
rapid access clinic. Dig Liver Dis 2019;51:340-5.
d

10. Koltun WA. Better together: Improved care of the ibd patient using the multi-
disciplinary ibd center. Expert Rev Gastroenterol Hepatol 2017;11:491-3.
e

11. Bemelman WA, Warusavitarne J, Sampietro GM, et al. Ecco-escp consensus on


pt

surgery for crohn's disease. J Crohns Colitis 2018;12:1-16.


12. Harbord M, Annese V, Vavricka SR, et al. The first european evidence-based
consensus on extra-intestinal manifestations in inflammatory bowel disease. J Crohns
ce

Colitis 2016;10:239-54.
13. Kemp K, Dibley L, Chauhan U, et al. Second n-ecco consensus statements on the
european nursing roles in caring for patients with crohn's disease or ulcerative colitis.
Ac

J Crohns Colitis 2018;12:760-76.


14. Maaser C, Sturm A, Vavricka SR, et al. Ecco-esgar guideline for diagnostic assessment
in ibd part 1: Initial diagnosis, monitoring of known ibd, detection of complications. J
Crohns Colitis 2019;13:144-64.
15. Sturm A, Maaser C, Calabrese E, et al. Ecco-esgar guideline for diagnostic assessment
in ibd part 2: Ibd scores and general principles and technical aspects. J Crohns Colitis
2019;13:273-84.
16. Sturm A, Maaser C, Mendall M, et al. European crohn's and colitis organisation
topical review on ibd in the elderly. J Crohns Colitis 2017;11:263-73.
17. Gordon H. Ecco guidelines on therapeutics in crohn's disease:
Medical treatment. J Crohns Colitis 2024.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
18. Adamina M, Bonovas S, Raine T, et al. Ecco guidelines on therapeutics in crohn's
disease: Surgical treatment. J Crohns Colitis 2020;14:155-68.
19. Torres J, Bonovas S, Doherty G, et al. Ecco guidelines on therapeutics in crohn's
disease: Medical treatment. J Crohns Colitis 2020;14:4-22.
20. J. H, I. C, P. G, et al. Explanation of the 2011 oxford centre for evidence-based
medicine (ocebm) levels of evidence (background document).
https://www.cebm.net/index.aspx?o=5653 Accessed 09.11.2019, 2011.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


21. Panes J, Rimola J. Perianal fistulizing crohn's disease: Pathogenesis, diagnosis and
therapy. Nat Rev Gastroenterol Hepatol 2017;14:652-64.
22. Thia KT, Mahadevan U, Feagan BG, et al. Ciprofloxacin or metronidazole for the
treatment of perianal fistulas in patients with crohn's disease: A randomized, double-

t
blind, placebo-controlled pilot study. Inflamm Bowel Dis 2009;15:17-24.

ip
23. Khan KJ, Ullman TA, Ford AC, et al. Antibiotic therapy in inflammatory bowel disease:
A systematic review and meta-analysis. Am J Gastroenterol 2011;106:661-73.

cr
24. Dewint P, Hansen BE, Verhey E, et al. Adalimumab combined with ciprofloxacin is
superior to adalimumab monotherapy in perianal fistula closure in crohn's disease: A
randomised, double-blind, placebo controlled trial (adafi). Gut 2014;63:292-9.

us
25. Klein M, Binder HJ, Mitchell M, Aaronson R, Spiro H. Treatment of crohn's disease
with azathioprine: A controlled evaluation. Gastroenterology 1974;66:916-22.
26. Rhodes J, Bainton D, Beck P, Campbell H. Controlled trial of azathioprine in crohn's
an
disease. Lancet 1971;2:1273-6.
27. Willoughby JM, Beckett J, Kumar PJ, Dawson AM. Controlled trial of azathioprine in
crohn's disease. Lancet 1971;2:944-7.
M

28. Willoughby JM, Kumar P, Beckett J, Dawson AM. A double-blind trial of azathioprine
in crohn's disease. Gut 1971;12:864.
29. Chande N, Patton PH, Tsoulis DJ, Thomas BS, MacDonald JK. Azathioprine or 6-
d

mercaptopurine for maintenance of remission in crohn's disease. Cochrane Database


Syst Rev 2015;2015:CD000067.
e

30. Present DH, Korelitz BI, Wisch N, et al. Treatment of crohn's disease with 6-
mercaptopurine. A long-term, randomized, double-blind study. N Engl J Med
pt

1980;302:981-7.
31. Present DH, Rutgeerts P, Targan S, et al. Infliximab for the treatment of fistulas in
ce

patients with crohn's disease. N Engl J Med 1999;340:1398-405.


32. Sands BE, Anderson FH, Bernstein CN, et al. Infliximab maintenance therapy for
fistulizing crohn's disease. N Engl J Med 2004;350:876-85.
Ac

33. Gordon M, Sinopoulou V, Akobeng AK, et al. Infliximab for medical induction of
remission in crohn's disease. Cochrane Database Syst Rev 2023;11:CD012623.
34. Davidov Y, Ungar B, Bar-Yoseph H, et al. Association of induction infliximab levels
with clinical response in perianal crohn's disease. J Crohns Colitis 2017;11:549-55.
35. De Gregorio M, Lee T, Krishnaprasad K, et al. Higher anti-tumor necrosis factor-alpha
levels correlate with improved radiologic outcomes in crohn's perianal fistulas. Clin
Gastroenterol Hepatol 2022;20:1306-14.
36. Papamichael K, Vande Casteele N, Jeyarajah J, et al. Higher postinduction infliximab
concentrations are associated with improved clinical outcomes in fistulizing crohn's
disease: An accent-ii post hoc analysis. Am J Gastroenterol 2021;116:1007-14.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
37. Strik AS, Lowenberg M, Buskens CJ, et al. Higher anti-tnf serum levels are associated
with perianal fistula closure in crohn's disease patients. Scand J Gastroenterol
2019;54:453-8.
38. Yarur AJ, Kanagala V, Stein DJ, et al. Higher infliximab trough levels are associated
with perianal fistula healing in patients with crohn's disease. Aliment Pharmacol Ther
2017;45:933-40.
39. Colombel JF, Schwartz DA, Sandborn WJ, et al. Adalimumab for the treatment of

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


fistulas in patients with crohn's disease. Gut 2009;58:940-8.
40. Colombel JF, Sandborn WJ, Rutgeerts P, et al. Adalimumab for maintenance of clinical
response and remission in patients with crohn's disease: The charm trial.
Gastroenterology 2007;132:52-65.

t
41. Echarri A, Castro J, Barreiro M, et al. Evaluation of adalimumab therapy in

ip
multidisciplinary strategy for perianal crohn's disease patients with infliximab failure.
J Crohns Colitis 2010;4:654-60.

cr
42. Lichtiger S, Binion DG, Wolf DC, et al. The choice trial: Adalimumab demonstrates
safety, fistula healing, improved quality of life and increased work productivity in
patients with crohn's disease who failed prior infliximab therapy. Aliment Pharmacol

us
Ther 2010;32:1228-39.
43. Panaccione R, Colombel JF, Sandborn WJ, et al. Adalimumab maintains remission of
crohn's disease after up to 4 years of treatment: Data from charm and adhere.
an
Aliment Pharmacol Ther 2013;38:1236-47.
44. Castano-Milla C, Chaparro M, Saro C, et al. Effectiveness of adalimumab in perianal
fistulas in crohn's disease patients naive to anti-tnf therapy. J Clin Gastroenterol
M

2015;49:34-40.
45. Plevris N, Jenkinson PW, Arnott ID, Jones GR, Lees CW. Higher anti-tumor necrosis
factor levels are associated with perianal fistula healing and fistula closure in crohn's
d

disease. Eur J Gastroenterol Hepatol 2020;32:32-7.


46. Sandborn WJ, Feagan BG, Stoinov S, et al. Certolizumab pegol for the treatment of
e

crohn's disease. N Engl J Med 2007;357:228-38.


47. Schreiber S, Lawrance IC, Thomsen OO, et al. Randomised clinical trial: Certolizumab
pt

pegol for fistulas in crohn's disease - subgroup results from a placebo-controlled


study. Aliment Pharmacol Ther 2011;33:185-93.
ce

48. Feagan BG, Schwartz D, Danese S, et al. Efficacy of vedolizumab in fistulising crohn's
disease: Exploratory analyses of data from gemini 2. J Crohns Colitis 2018;12:621-6.
49. Lee MJ, Parker CE, Taylor SR, et al. Efficacy of medical therapies for fistulizing crohn's
Ac

disease: Systematic review and meta-analysis. Clin Gastroenterol Hepatol


2018;16:1879-92.
50. Schwartz DA, Peyrin-Biroulet L, Lasch K, Adsul S, Danese S. Efficacy and safety of 2
vedolizumab intravenous regimens for perianal fistulizing crohn's disease: Enterprise
study. Clin Gastroenterol Hepatol 2022;20:1059-67 e9.
51. Sands B, Gasink C, Jacobstein D, et al. Fistula healing in pivotal studies of
ustekinumab in crohn’s disease. Gastroenterology 2017;152:S185.
52. Godoy Brewer GM, Salem G, Afzal MA, et al. Ustekinumab is effective for perianal
fistulising crohn's disease: A real-world experience and systematic review with meta-
analysis. BMJ Open Gastroenterol 2021;8.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
53. Colombel JF, Irving P, Rieder F, et al. Efficacy and safety of upadacitinib for the
treatment of fistulas and fissures in patients with crohn’s disease. Journal of Crohn's
and Colitis 2023;17.
54. Davies M, Harris D, Lohana P, et al. The surgical management of fistula-in-ano in a
specialist colorectal unit. Int J Colorectal Dis 2008;23:833-8.
55. Herissay A, Siproudhis L, Le Balc'h E, et al. Combined strategies following surgical
drainage for perianal fistulizing crohn's disease: Failure rates and prognostic factors.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Colorectal Dis 2021;23:159-68.
56. Morrison JG, Gathright JB, Jr., Ray JE, et al. Surgical management of anorectal fistulas
in crohn's disease. Dis Colon Rectum 1989;32:492-6.
57. Papaconstantinou I, Kontis E, Koutoulidis V, Mantzaris G, Vassiliou I. Surgical

t
management of fistula-in-ano among patients with crohn's disease: Analysis of

ip
outcomes after fistulotomy or seton placement-single-center experience.
Scandinavian journal of surgery : SJS : official organ for the Finnish Surgical Society

cr
and the Scandinavian Surgical Society 2017;106:211-5.
58. Park MY, Yoon YS, Kim HE, et al. Surgical options for perianal fistula in patients with
crohn's disease: A comparison of seton placement, fistulotomy, and stem cell

us
therapy. Asian J Surg 2021;44:1383-8.
59. van der Hagen SJ, Baeten CG, Soeters PB, van Gemert WG. Long-term outcome
following mucosal advancement flap for high perianal fistulas and fistulotomy for low
an
perianal fistulas: Recurrent perianal fistulas: Failure of treatment or recurrent patient
disease? Int J Colorectal Dis 2006;21:784-90.
60. Williams JG, Rothenberger DA, Nemer FD, Goldberg SM. Fistula-in-ano in crohn's
M

disease. Results of aggressive surgical treatment. Dis Colon Rectum 1991;34:378-84.


61. Takesue Y, Ohge H, Yokoyama T, et al. Long-term results of seton drainage on
complex anal fistulae in patients with crohn's disease. J Gastroenterol 2002;37:912-5.
d

62. Merten J, Eichelmann AK, Mennigen R, et al. Minor sphincter sparing surgery for
successful closure of perianal fistulas in patients with crohn's disease. J Clin Med
e

2021;10.
63. Rozalen V, Pares D, Sanchez E, et al. Advancement flap technique for anal fistula in
pt

patients with crohn's disease: A systematic review of the literature. Cir Esp
2017;95:558-65.
ce

64. Stellingwerf ME, van Praag EM, Tozer PJ, Bemelman WA, Buskens CJ. Systematic
review and meta-analysis of endorectal advancement flap and ligation of the
intersphincteric fistula tract for cryptoglandular and crohn's high perianal fistulas. BJS
Ac

Open 2019;3:231-41.
65. Bessi G, Siproudhis L, Merlini l'Heritier A, et al. Advancement flap procedure in crohn
and non-crohn perineal fistulas: A simple surgical approach. Colorectal Dis
2019;21:66-72.
66. Roper MT, Trinidad SM, Ramamoorthy SL, et al. Endorectal advancement flaps for
perianal fistulae in crohn's disease: Careful patient selection leads to optimal
outcomes. J Gastrointest Surg 2019;23:2277-84.
67. Seifarth C, Lehmann KS, Holmer C, Pozios I. Healing of rectal advancement flaps for
anal fistulas in patients with and without crohn's disease: A retrospective cohort
analysis. BMC Surg 2021;21:283.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
68. van Praag EM, Stellingwerf ME, van der Bilt JDW, et al. Ligation of the
intersphincteric fistula tract and endorectal advancement flap for high perianal
fistulas in crohn's disease: A retrospective cohort study. J Crohns Colitis 2020;14:757-
63.
69. Emile SH, Khan SM, Adejumo A, Koroye O. Ligation of intersphincteric fistula tract
(lift) in treatment of anal fistula: An updated systematic review, meta-analysis, and
meta-regression of the predictors of failure. Surgery 2020;167:484-92.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


70. Meima-van Praag EM, van Rijn KL, Monraats MA, Buskens CJ, Stoker J. Magnetic
resonance imaging after ligation of the intersphincteric fistula tract for high perianal
fistulas in crohn's disease: A retrospective cohort study. Colorectal Dis 2021;23:169-
77.

t
71. Wood T, Truong A, Mujukian A, Zaghiyan K, Fleshner P. Increasing experience with the

ip
lift procedure in crohn's disease patients with complex anal fistula. Tech Coloproctol
2022;26:205-12.

cr
72. Grimaud JC, Munoz-Bongrand N, Siproudhis L, et al. Fibrin glue is effective healing
perianal fistulas in patients with crohn's disease. Gastroenterology 2010;138:2275-
81, 81 e1.

us
73. Vidon M, Munoz-Bongrand N, Lambert J, et al. Long-term efficacy of fibrin glue
injection for perianal fistulas in patients with crohn's disease. Colorectal Dis
2021;23:894-900.
an
74. Senejoux A, Siproudhis L, Abramowitz L, et al. Fistula plug in fistulising ano-perineal
crohn's disease: A randomised controlled trial. J Crohns Colitis 2016;10:141-8.
75. Nasseri Y, Cassella L, Berns M, Zaghiyan K, Cohen J. The anal fistula plug in crohn's
M

disease patients with fistula-in-ano: A systematic review. Colorectal Dis 2016;18:351-


6.
76. Adamina M, Ross T, Guenin MO, et al. Anal fistula plug: A prospective evaluation of
d

success, continence and quality of life in the treatment of complex fistulae.


Colorectal Dis 2014;16:547-54.
e

77. Garg P, Song J, Bhatia A, Kalia H, Menon GR. The efficacy of anal fistula plug in fistula-
in-ano: A systematic review. Colorectal Dis 2010;12:965-70.
pt

78. Chase TJG, Quddus A, Selvakumar D, Cunha P, Cuming T. Vaaft for complex anal
fistula: A useful tool, however, cure is unlikely. Tech Coloproctol 2021;25:1115-21.
ce

79. Schwandner O. Video-assisted anal fistula treatment (vaaft) combined with


advancement flap repair in crohn's disease. Tech Coloproctol 2013;17:221-5.
80. Adegbola SO, Sahnan K, Tozer PJ, et al. Symptom amelioration in crohn's perianal
Ac

fistulas using video-assisted anal fistula treatment (vaaft). J Crohns Colitis


2018;12:1067-72.
81. Mennigen R, Laukotter M, Senninger N, Rijcken E. The otsc((r)) proctology clip system
for the closure of refractory anal fistulas. Tech Coloproctol 2015;19:241-6.
82. Prosst RL, Joos AK. Short-term outcomes of a novel endoscopic clipping device for
closure of the internal opening in 100 anorectal fistulas. Tech Coloproctol
2016;20:753-8.
83. de Groof EJ, Sahami S, Lucas C, et al. Treatment of perianal fistula in crohn's disease:
A systematic review and meta-analysis comparing seton drainage and anti-tumour
necrosis factor treatment. Colorectal Dis 2016;18:667-75.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
84. Feroz SH, Ahmed A, Muralidharan A, Thirunavukarasu P. Comparison of the efficacy
of the various treatment modalities in the management of perianal crohn's fistula: A
review. Cureus 2020;12:e11882.
85. Wasmann KA, de Groof EJ, Stellingwerf ME, et al. Treatment of perianal fistulas in
crohn's disease, seton versus anti-tnf versus surgical closure following anti-tnf *pisa+:
A randomised controlled trial. J Crohns Colitis 2020;14:1049-56.
86. McNamara DA, Brophy S, Hyland JM. Perianal crohn's disease and infliximab therapy.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Surgeon 2004;2:258-63.
87. Tandon P, Rhee GG, Schwartz D, McCurdy JD. Strategies to optimize anti-tumor
necrosis factor therapy for perianal fistulizing crohn's disease: A systematic review.
Dig Dis Sci 2019;64:3066-77.

t
88. Yassin NA, Askari A, Warusavitarne J, et al. Systematic review: The combined surgical

ip
and medical treatment of fistulising perianal crohn's disease. Aliment Pharmacol Ther
2014;40:741-9.

cr
89. Bouguen G, Siproudhis L, Gizard E, et al. Long-term outcome of perianal fistulizing
crohn's disease treated with infliximab. Clin Gastroenterol Hepatol 2013;11:975-81
e1-4.

us
90. Regueiro M, Mardini H. Treatment of perianal fistulizing crohn's disease with
infliximab alone or as an adjunct to exam under anesthesia with seton placement.
Inflamm Bowel Dis 2003;9:98-103.
an
91. Gaertner WB, Decanini A, Mellgren A, et al. Does infliximab infusion impact results of
operative treatment for crohn's perianal fistulas? Dis Colon Rectum 2007;50:1754-60.
92. Sciaudone G, Di Stazio C, Limongelli P, et al. Treatment of complex perianal fistulas in
M

crohn disease: Infliximab, surgery or combined approach. Can J Surg 2010;53:299-


304.
93. Sebastian S, Black C, Pugliese D, et al. The role of multimodal treatment in crohn's
d

disease patients with perianal fistula: A multicentre retrospective cohort study.


Aliment Pharmacol Ther 2018;48:941-50.
e

94. van der Hagen SJ, Baeten CG, Soeters PB, et al. Anti-tnf-alpha (infliximab) used as
induction treatment in case of active proctitis in a multistep strategy followed by
pt

definitive surgery of complex anal fistulas in crohn's disease: A preliminary report.


Dis Colon Rectum 2005;48:758-67.
ce

95. Kaminski JP, Zaghiyan K, Fleshner P. Increasing experience of ligation of the


intersphincteric fistula tract for patients with crohn's disease: What have we learned?
Colorectal Dis 2017;19:750-5.
Ac

96. Laland M, Francois M, D'Amico F, et al. Identification of the optimal medical and
surgical management for patients with perianal fistulising crohn's disease. Colorectal
Dis 2023;25:75-82.
97. Abramowitz L, Brochard C, Pigot F, et al. Surgical closure, mainly with glue injection
and anti-tumour necrosis factor alpha, in fistulizing perianal crohn's disease: A
multicentre randomized controlled trial. Colorectal Dis 2022;24:210-9.
98. Meima-van Praag EM, van Rijn KL, Wasmann K, et al. Short-term anti-tnf therapy
with surgical closure versus anti-tnf therapy in the treatment of perianal fistulas in
crohn's disease (pisa-ii): A patient preference randomised trial. Lancet Gastroenterol
Hepatol 2022;7:617-26.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
99. Meima-van Praag EM, Becker MAJ, van Rijn KL, et al. Short-term anti-tnf therapy with
surgical closure versus anti-tnf therapy alone for crohn's perianal fistulas (pisa-ii):
Long-term outcomes of an international, multicentre patient preference, randomised
controlled trial. EClinicalMedicine 2023;61:102045.
100. Bislenghi G, Wolthuis A, Van Assche G, et al. Cx601 (darvadstrocel) for the treatment
of perianal fistulizing crohn's disease. Expert Opin Biol Ther 2019;19:607-16.
101. Panes J, Garcia-Olmo D, Van Assche G, et al. Long-term efficacy and safety of stem

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


cell therapy (cx601) for complex perianal fistulas in patients with crohn's disease.
Gastroenterology 2018;154:1334-42 e4.
102. Garcia-Olmo D, Gilaberte I, Binek M, et al. Follow-up study to evaluate the long-term
safety and efficacy of darvadstrocel (mesenchymal stem cell treatment) in patients

t
with perianal fistulizing crohn's disease: Admire-cd phase 3 randomized controlled

ip
trial. Dis Colon Rectum 2022;65:713-20.
103. Lightner AL, Wang Z, Zubair AC, Dozois EJ. A systematic review and meta-analysis of

cr
mesenchymal stem cell injections for the treatment of perianal crohn's disease:
Progress made and future directions. Dis Colon Rectum 2018;61:629-40.
104. Takeda. Study to assess efficacy and safety of cx601, adult allogeneic expanded

us
adipose-derived stem cells (easc) for the treatment of complex perianal fistula(s) in
participants with crohn's disease (admire-cd-ii). ClinicalTrials.Gov, 2017.
105. Cheng F, Huang Z, Li Z. Efficacy and safety of mesenchymal stem cells in treatment of
an
complex perianal fistulas: A meta-analysis. Stem Cells Int 2020;2020:8816737.
106. Dietz AB, Dozois EJ, Fletcher JG, et al. Autologous mesenchymal stem cells, applied in
a bioabsorbable matrix, for treatment of perianal fistulas in patients with crohn's
M

disease. Gastroenterology 2017;153:59-62 e2.


107. Dige A, Hougaard HT, Agnholt J, et al. Efficacy of injection of freshly collected
autologous adipose tissue into perianal fistulas in patients with crohn's disease.
d

Gastroenterology 2019;156:2208-16 e1.


108. Dozois EJ, Lightner AL, Dietz AB, et al. Durable response in patients with refractory
e

fistulizing perianal crohn's disease using autologous mesenchymal stem cells on a


dissolvable matrix: Results from the phase i stem cell on matrix plug trial. Dis Colon
pt

Rectum 2023;66:243-52.
109. Herreros MD, Garcia-Olmo D, Guadalajara H, et al. Stem cell therapy: A
ce

compassionate use program in perianal fistula. Stem Cells Int 2019;2019:6132340.


110. Park MY, Yoon YS, Lee JL, et al. Comparative perianal fistula closure rates following
autologous adipose tissue-derived stem cell transplantation or treatment with anti-
Ac

tumor necrosis factor agents after seton placement in patients with crohn's disease:
A retrospective observational study. Stem Cell Res Ther 2021;12:401.
111. Wainstein C, Quera R, Fluxa D, et al. Stem cell therapy in refractory perineal crohn's
disease: Long-term follow-up. Colorectal Dis 2018.
112. Laureti S, Gionchetti P, Cappelli A, et al. Refractory complex crohn's perianal fistulas:
A role for autologous microfragmented adipose tissue injection. Inflamm Bowel Dis
2020;26:321-30.
113. Sands BE, Blank MA, Patel K, van Deventer SJ, Study AI. Long-term treatment of
rectovaginal fistulas in crohn's disease: Response to infliximab in the accent ii study.
Clin Gastroenterol Hepatol 2004;2:912-20.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
114. Le Baut G, Peyrin-Biroulet L, Bouguen G, et al. Anti-tnf therapy for genital fistulas in
female patients with crohn's disease: A nationwide study from the groupe d'etude
therapeutique des affections inflammatoires du tube digestif (getaid). Aliment
Pharmacol Ther 2018;48:831-8.
115. Otero-Pineiro AM, Jia X, Pedersen KE, et al. Surgical intervention is effective for the
treatment of crohn's-related rectovaginal fistulas: Experience from a tertiary
inflammatory bowel disease practice. J Crohns Colitis 2023;17:396-403.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


116. Iglay K, Bennett D, Kappelman MD, et al. A systematic review of the patient burden of
crohn's disease-related rectovaginal and anovaginal fistulas. BMC Gastroenterol
2022;22:36.
117. Singh S, Ding NS, Mathis KL, et al. Systematic review with meta-analysis: Faecal

t
diversion for management of perianal crohn's disease. Aliment Pharmacol Ther

ip
2015;42:783-92.
118. Rehg KL, Sanchez JE, Krieger BR, Marcet JE. Fecal diversion in perirectal fistulizing

cr
crohn's disease is an underutilized and potentially temporary means of successful
treatment. Am Surg 2009;75:715-8.
119. Hong MK, Craig Lynch A, Bell S, et al. Faecal diversion in the management of perianal

us
crohn's disease. Colorectal Dis 2011;13:171-6.
120. Gu J, Valente MA, Remzi FH, Stocchi L. Factors affecting the fate of faecal diversion in
patients with perianal crohn's disease. Colorectal Dis 2015;17:66-72.
an
121. Kasparek MS, Glatzle J, Temeltcheva T, et al. Long-term quality of life in patients with
crohn's disease and perianal fistulas: Influence of fecal diversion. Dis Colon Rectum
2007;50:2067-74.
M

122. Jew M, Meserve J, Eisenstein S, et al. Temporary faecal diversion for refractory
perianal and/or distal colonic crohn's disease in the biologic era: An updated
systematic review with meta-analysis. J Crohns Colitis 2023.
d

123. McKee RF, Keenan RA. Perianal crohn's disease--is it all bad news? Dis Colon Rectum
1996;39:136-42.
e

124. Regimbeau JM, Panis Y, Marteau P, Benoist S, Valleur P. Surgical treatment of


anoperineal crohn's disease: Can abdominoperineal resection be predicted? J Am
pt

Coll Surg 1999;189:171-6.


125. Schlegel N, Kim M, Reibetanz J, et al. Sphincter-sparing intersphincteric rectal
ce

resection as an alternative to proctectomy in long-standing fistulizing and stenotic


crohn's proctitis? Int J Colorectal Dis 2015;30:655-63.
126. Williams JG, Hughes LE. Abdominoperineal resection for severe perianal crohn's
Ac

disease. Dis Colon Rectum 1990;33:402-7.


127. Hollis RH, Smith N, Sapci I, et al. Small bowel crohn's disease recurrence is common
after total proctocolectomy for crohn's colitis. Dis Colon Rectum 2022;65:390-8.
128. Lightner AL, Steele SR, Delaney CP, et al. Colonic disease recurrence following
proctectomy with end colostomy for anorectal crohn's disease. Colorectal Dis
2021;23:2425-35.
129. Li W, Stocchi L, Elagili F, Kiran RP, Strong SA. Healing of the perineal wound after
proctectomy in crohn's disease patients: Only preoperative perineal sepsis predicts
poor outcome. Tech Coloproctol 2017;21:715-20.
130. McGarity WC, Mallory C, Walker J. Perineal wound healing after abdominoperineal
resection. Am Surg 1976;42:206-11.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
131. Schaden D, Schauer G, Haas F, Berger A. Myocutaneous flaps and proctocolectomy in
severe perianal crohn's disease--a single stage procedure. Int J Colorectal Dis
2007;22:1453-7.
132. Waits JO, Dozois RR, Kelly KA. Primary closure and continuous irrigation of the
perineal wound after proctectomy. Mayo Clin Proc 1982;57:185-8.
133. Lansdorp CA, Buskens CJ, Gecse KB, D'Haens GR, Van Hulst RA. Wound healing of
metastatic perineal crohn's disease using hyperbaric oxygen therapy: A case series.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


United European Gastroenterol J 2020;8:820-7.
134. Geldof J, Iqbal N, LeBlanc JF, et al. Classifying perianal fistulising crohn's disease: An
expert consensus to guide decision-making in daily practice and clinical trials. Lancet
Gastroenterol Hepatol 2022;7:576-84.

t
135. de Groof EJ, Cabral VN, Buskens CJ, et al. Systematic review of evidence and

ip
consensus on perianal fistula: An analysis of national and international guidelines.
Colorectal Dis 2016;18:O119-34.

cr
136. Singh S, Al-Darmaki A, Frolkis AD, et al. Postoperative mortality among patients with
inflammatory bowel diseases: A systematic review and meta-analysis of population-
based studies. Gastroenterology 2015;149:928-37.

us
137. Udholm LS, Rasmussen SL, Madsbøll TK, Omairi M, El-Hussuna A. A systemic review
and metaanalysis of postoperative outcomes in urgent and elective bowel resection
in patients with crohn's disease. Int J Colorectal Dis 2021;36:253-63.
an
138. Risk factors for unfavourable postoperative outcome in patients with crohn's disease
undergoing right hemicolectomy or ileocaecal resection an international audit by
escp and s-ecco. Colorectal Dis 2017.
M

139. El-Hussuna A, Karer MLM, Uldall Nielsen NN, et al. Postoperative complications and
waiting time for surgical intervention after radiologically guided drainage of intra-
abdominal abscess in patients with crohn's disease. BJS Open 2021;5.
d

140. Celentano V, Giglio MC, Pellino G, et al. High complication rate in crohn's disease
surgery following percutaneous drainage of intra-abdominal abscess: A multicentre
e

study. Int J Colorectal Dis 2022;37:1421-8.


141. Sakurai Kimura CM, Scanavini Neto A, Queiroz NSF, et al. Abdominal surgery in
pt

crohn's disease: Risk factors for complications. Inflamm Intest Dis 2021;6:18-24.
142. Fahim M, Dijksman LM, Derksen WJM, et al. Prospective multicentre study of a new
ce

bowel obstruction treatment in colorectal surgery: Reduced morbidity and mortality.


Eur J Surg Oncol 2021;47:2414-20.
143. Liu R-Q, Qiao S-H, Wang K-H, et al. Prospective evaluation of intestinal
Ac

decompression in treatment of acute bowel obstruction from crohn's disease.


Gastroenterology report 2019;7:263-71.
144. Sebastian S, Segal JP, Hedin C, et al. Ecco topical review: Roadmap to optimal peri-
operative care in ibd. J Crohns Colitis 2023;17:153-69.
145. Adamina M, Gerasimidis K, Sigall-Boneh R, et al. Perioperative dietary therapy in
inflammatory bowel disease. J Crohns Colitis 2020;14:431-44.
146. Clancy C, Boland T, Deasy J, McNamara D, Burke JP. A meta-analysis of percutaneous
drainage versus surgery as the initial treatment of crohn's disease-related intra-
abdominal abscess. J Crohns Colitis 2016;10:202-8.
147. El-Hussuna A, Steenholdt C, Merrild Karer ML, et al. Watchful waiting after
radiological guided drainage of intra-abdominal abscess in patients with crohn’s
Manuscript Doi: 10.1093/ecco-jcc/jjae089
disease might be associated with increased rates of stoma construction. Crohn's &
Colitis 360 2023;5.
148. Waked B, Holvoet T, Geldof J, et al. Conservative management of spontaneous intra-
abdominal abscess in crohn's disease: Outcome and prognostic factors. J Dig Dis
2021;22:263-70.
149. Huang W, Tang Y, Nong L, Sun Y. Risk factors for postoperative intra-abdominal septic
complications after surgery in crohn's disease: A meta-analysis of observational

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


studies. J Crohns Colitis 2015;9:293-301.
150. da Luz Moreira A, Stocchi L, Tan E, Tekkis PP, Fazio VW. Outcomes of crohn's disease
presenting with abdominopelvic abscess. Dis Colon Rectum 2009;52:906-12.
151. Luong TV, Grandt SD, Negoi I, Palubinskas S, El-Hussuna A. Preoperative factors

t
associated with prolonged postoperative in-hospital length of stay in patients with

ip
crohn's disease undergoing intestinal resection or strictureplasty. Int J Colorectal Dis
2019;34:1925-31.

cr
152. He X, Lin X, Lian L, et al. Preoperative percutaneous drainage of spontaneous intra-
abdominal abscess in patients with crohn's disease: A meta-analysis. J Clin
Gastroenterol 2015;49:e82-90.

us
153. Peyser DK, Carmichael H, Dean A, et al. Early versus delayed ileocolic resection for
complicated crohn's disease: Is "cooling off" necessary? Surg Endosc 2022;36:4290-8.
154. Bettenworth D, Gustavsson A, Atreja A, et al. A pooled analysis of efficacy, safety, and
an
long-term outcome of endoscopic balloon dilation therapy for patients with
stricturing crohn's disease. Inflamm Bowel Dis 2017;23:133-42.
155. Hassan C, Zullo A, De Francesco V, et al. Systematic review: Endoscopic dilatation in
M

crohn's disease. Alimentary pharmacology & therapeutics 2007;26:1457-64.


156. Morar PS, Faiz O, Warusavitarne J, et al. Systematic review with meta-analysis:
Endoscopic balloon dilatation for crohn's disease strictures. Aliment Pharmacol Ther
d

2015;42:1137-48.
157. Navaneethan U, Lourdusamy V, Njei B, Shen B. Endoscopic balloon dilation in the
e

management of strictures in crohn's disease: A systematic review and meta-analysis


of non-randomized trials. Surg Endosc 2016;30:5434-43.
pt

158. Andújar X, Loras C, González B, et al. Efficacy and safety of endoscopic balloon
dilation in inflammatory bowel disease: Results of the large multicenter study of the
ce

eneida registry. Surg Endosc 2020;34:1112-22.


159. Bamba S, Sakemi R, Fujii T, et al. A nationwide, multi-center, retrospective study of
symptomatic small bowel stricture in patients with crohn's disease. J Gastroenterol
Ac

2020;55:615-26.
160. Uda A, Kuwabara H, Shimizu S, Iwakiri R, Fushimi K. Optimal use of biologics with
endoscopic balloon dilatation for repeated intestinal strictures in crohn's disease.
JGH Open 2020;4:532-40.
161. Lu C, Baraty B, Lee Robertson H, et al. Systematic review: Medical therapy for
fibrostenosing crohn's disease. Aliment Pharmacol Ther 2020;51:1233-46.
162. Bettenworth D, Bokemeyer A, Kou L, et al. Systematic review with meta-analysis:
Efficacy of balloon-assisted enteroscopy for dilation of small bowel crohn's disease
strictures. Aliment Pharmacol Ther 2020;52:1104-16.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
163. Costa-Santos MP, Palmela C, Torres J, et al. Preoperative enteral nutrition in adults
with complicated crohn's disease: Effect on disease outcomes and gut microbiota.
Nutrition 2020;70s:100009.
164. Ge X, Tang S, Yang X, et al. The role of exclusive enteral nutrition in the preoperative
optimization of laparoscopic surgery for patients with crohn's disease: A cohort
study. Int J Surg 2019;65:39-44.
165. Rocha A, Bessa I, Lago P, et al. Preoperative enteral nutrition and surgical outcomes

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


in adults with crohn's disease: A systematic review. GE Port J Gastroenterol
2019;26:184-95.
166. Gordon-Dixon A, Gore-Rodney J, Hampal R, et al. The role of exclusive enteral
nutrition in the pre-operative optimisation of adult patients with crohn's disease. A

t
systematic review. Clin Nutr ESPEN 2021;46:99-105.

ip
167. Jiang T, Jiang Y, Jin Q, et al. Role of perioperative nutritional status and enteral
nutrition in predicting and preventing post-operative complications in patients with

cr
crohn's disease. Front Nutr 2022;9:1085037.
168. Meade S, Patel KV, Luber RP, et al. A retrospective cohort study: Pre-operative oral
enteral nutritional optimisation for crohn's disease in a uk tertiary ibd centre. Aliment

us
Pharmacol Ther 2022;56:646-63.
169. Brennan GT, Ha I, Hogan C, et al. Does preoperative enteral or parenteral nutrition
reduce postoperative complications in crohn's disease patients: A meta-analysis. Eur J
an
Gastroenterol Hepatol 2018;30:997-1002.
170. Traiki TAB, Alshammari SA, Aljomah NA, et al. The impact of preoperative total
parenteral nutrition on the surgical complications of crohn's disease: A retrospective
M

cohort study. Saudi J Gastroenterol 2023;29:158-63.


171. Abdalla S, Benoist S, Maggiori L, et al. Impact of preoperative enteral nutritional
support on postoperative outcome in patients with crohn's disease complicated by
d

malnutrition: Results of a subgroup analysis of the nationwide cohort registry from


the getaid chirurgie group. Colorectal Dis 2021;23:1451-62.
e

172. Gionchetti P, Dignass A, Danese S, et al. 3rd european evidence-based consensus on


the diagnosis and management of crohn's disease 2016: Part 2: Surgical management
pt

and special situations. J Crohns Colitis 2017;11:135-49.


173. Law CC, Bell C, Koh D, et al. Risk of postoperative infectious complications from
ce

medical therapies in inflammatory bowel disease. Cochrane Database Syst Rev


2020;10:Cd013256.
174. Subramanian V, Saxena S, Kang JY, Pollok RC. Preoperative steroid use and risk of
Ac

postoperative complications in patients with inflammatory bowel disease undergoing


abdominal surgery. Am J Gastroenterol 2008;103:2373-81.
175. Abou Khalil M, Abou-Khalil J, Motter J, et al. Immunosuppressed patients with
crohn's disease are at increased risk of postoperative complications: Results from the
acs-nsqip database. J Gastrointest Surg 2019;23:1188-97.
176. Freund MR, Emile SH, Horesh N, et al. Redo ileocolic resection for recurrent
crohn&#x2019;s disease: A review and meta-analysis of surgical outcomes. Surgery
2022;172:1614-21.
177. Quaresma AB, Yamamoto T, Kotze PG. Biologics and surgical outcomes in crohn's
disease: Is there a direct relationship? Therap Adv Gastroenterol
2020;13:1756284820931738.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
178. Cohen BL, Fleshner P, Kane SV, et al. Prospective cohort study to investigate the
safety of preoperative tumor necrosis factor inhibitor exposure in patients with
inflammatory bowel disease undergoing intra-abdominal surgery. Gastroenterology
2022;163:204-21.
179. Law CCY, Narula A, Lightner AL, et al. Systematic review and meta-analysis:
Preoperative vedolizumab treatment and postoperative complications in patients
with inflammatory bowel disease. J Crohns Colitis 2018;12:538-45.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


180. Moosvi Z, Duong JT, Bechtold ML, Nguyen DL. Systematic review and meta-analysis:
Preoperative vedolizumab and postoperative complications in patients with ibd.
South Med J 2021;114:98-105.
181. Yung DE, Horesh N, Lightner AL, et al. Systematic review and meta-analysis:

t
Vedolizumab and postoperative complications in inflammatory bowel disease.

ip
Inflammatory Bowel Diseases 2018;24:2327-38.
182. Guo D, Jiang K, Hong J, et al. Association between vedolizumab and postoperative

cr
complications in ibd: A systematic review and meta-analysis. Int J Colorectal Dis
2021;36:2081-92.
183. Garg R, Mohan BP, Ponnada S, et al. Postoperative outcomes after preoperative

us
ustekinumab exposure in patients with crohn's disease: A systematic review and
meta-analysis. Annals of gastroenterology 2021;34:691-8.
184. Lightner AL, McKenna NP, Tse CS, et al. Postoperative outcomes in ustekinumab-
an
treated patients undergoing abdominal operations for crohn's disease. J Crohns
Colitis 2018;12:402-7.
185. Adamina M, Gie O, Demartines N, Ris F. Contemporary perioperative care strategies.
M

Br J Surg 2013;100:38-54.
186. Zangenberg MS, Horesh N, Kopylov U, El-Hussuna A. Preoperative optimization of
patients with inflammatory bowel disease undergoing gastrointestinal surgery: A
d

systematic review. Int J Colorectal Dis 2017;32:1663-76.


187. Barnes EL, Lightner AL, Regueiro M. Peri-operative and post-operative management
e

of patients with crohn's disease and ulcerative colitis. Clin Gastroenterol Hepatol
2019.
pt

188. Dasari BV, McKay D, Gardiner K. Laparoscopic versus open surgery for small bowel
crohn's disease. Cochrane Database Syst Rev 2011:CD006956.
ce

189. Tan JJ, Tjandra JJ. Laparoscopic surgery for crohn's disease: A meta-analysis. Dis Colon
Rectum 2007;50:576-85.
190. Patel SV, Patel SV, Ramagopalan SV, Ott MC. Laparoscopic surgery for crohn's disease:
Ac

A meta-analysis of perioperative complications and long term outcomes compared


with open surgery. BMC Surg 2013;13:14.
191. Shigeta K, Okabayashi K, Hasegawa H, et al. Meta-analysis of laparoscopic surgery for
recurrent crohn's disease. Surg Today 2016;46:970-8.
192. Ponsioen CY, de Groof EJ, Eshuis EJ, et al. Laparoscopic ileocaecal resection versus
infliximab for terminal ileitis in crohn's disease: A randomised controlled, open-label,
multicentre trial. Lancet Gastroenterol Hepatol 2017;2:785-92.
193. Stevens TW, Haasnoot ML, D'Haens GR, et al. Laparoscopic ileocaecal resection
versus infliximab for terminal ileitis in crohn's disease: Retrospective long-term
follow-up of the lir!C trial. Lancet Gastroenterol Hepatol 2020;5:900-7.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
194. Ryan É J, Orsi G, Boland MR, et al. Meta-analysis of early bowel resection versus
initial medical therapy in patient's with ileocolonic crohn's disease. Int J Colorectal
Dis 2020;35:501-12.
195. Simillis C, Purkayastha S, Yamamoto T, et al. A meta-analysis comparing conventional
end-to-end anastomosis vs. Other anastomotic configurations after resection in
crohn's disease. Dis Colon Rectum 2007;50:1674-87.
196. Guo Z, Li Y, Zhu W, et al. Comparing outcomes between side-to-side anastomosis and

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


other anastomotic configurations after intestinal resection for patients with crohn's
disease: A meta-analysis. World J Surg 2013;37:893-901.
197. He X, Chen Z, Huang J, et al. Stapled side-to-side anastomosis might be better than
handsewn end-to-end anastomosis in ileocolic resection for crohn's disease: A meta-

t
analysis. Dig Dis Sci 2014;59:1544-51.

ip
198. Feng JS, Li JY, Yang Z, et al. Stapled side-to-side anastomosis might be benefit in
intestinal resection for crohn's disease: A systematic review and network meta-

cr
analysis. Medicine (Baltimore) 2018;97:e0315.
199. Kellil T, Chaouch MA, Guedich A, et al. Surgical features to reduce anastomotic
recurrence of crohn's disease that requires reoperation: A systematic review. Surg

us
Today 2022;52:542-9.
200. Thin LWY, Picardo S, Sooben S, et al. Ileocolonic end-to-end anastomoses in crohn's
disease increase the risk of early post-operative endoscopic recurrence in those
an
undergoing an emergency resection. J Gastrointest Surg 2021;25:241-51.
201. Zhou J, Li Y, Gong J, Zhu W. Frequency and risk factors of surgical recurrence of
crohn's disease after primary bowel resection. Turk J Gastroenterol 2018;29:655-63.
M

202. Celentano V, Pellino G, Spinelli A, et al. Anastomosis configuration and technique


following ileocaecal resection for crohn's disease: A multicentre study. Updates Surg
2021;73:149-56.
d

203. Kono T, Ashida T, Ebisawa Y, et al. A new antimesenteric functional end-to-end


handsewn anastomosis: Surgical prevention of anastomotic recurrence in crohn's
e

disease. Dis Colon Rectum 2011;54:586-92.


204. Kono T, Fichera A, Maeda K, et al. Kono-s anastomosis for surgical prophylaxis of
pt

anastomotic recurrence in crohn's disease: An international multicenter study. J


Gastrointest Surg 2016;20:783-90.
ce

205. Horisberger K, Birrer DL, Rickenbacher A, Turina M. Experiences with the kono-s
anastomosis in crohn's disease of the terminal ileum-a cohort study. Langenbecks
Arch Surg 2021;406:1173-80.
Ac

206. Shimada N, Ohge H, Kono T, et al. Surgical recurrence at anastomotic site after bowel
resection in crohn's disease: Comparison of kono-s and end-to-end anastomosis. J
Gastrointest Surg 2019;23:312-9.
207. Fichera A, Mangrola AM, Olortegui KS, et al. Long-term outcomes of the kono-s
anastomosis: A multicenter study. Dis Colon Rectum 2024;67:406-13.
208. Luglio G, Rispo A, Imperatore N, et al. Surgical prevention of anastomotic recurrence
by excluding mesentery in crohn's disease: The supreme-cd study - a randomized
clinical trial. Ann Surg 2020;272:210-7.
209. Alshantti A, Hind D, Hancock L, Brown SR. The role of kono-s anastomosis and
mesenteric resection in reducing recurrence after surgery for crohn's disease: A
systematic review. Colorectal Dis 2021;23:7-17.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
210. Ng CH, Chin YH, Lin SY, et al. Kono-s anastomosis for crohn's disease: A systemic
review, meta-analysis, and meta-regression. Surg Today 2021;51:493-501.
211. Tyrode G, Lakkis Z, Vernerey D, et al. Kono-s anastomosis is not superior to
conventional anastomosis for the reduction of postoperative endoscopic recurrence
in crohn’s disease. Inflammatory Bowel Diseases 2023.
212. (AMC-UvA) AMC-UvA. Comparing hand-sewn (end-to-end or kono-s) to stapled
anastomosis in ileocolic resection for crohn's disease (end2end). 2023 edn.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


ClinicalTrials.gov, 2023.
213. University WMCoC. Study of the kono-s anastomosis versus the side-to-side
functional end anastomosis. ClinicalTrials.Gov, 2014.
214. Haanappel AEG, Bellato V, Buskens CJ, et al. Optimising surgical anastomosis in

t
ileocolic resection for crohn's disease with respect to recurrence and functionality:

ip
Two international parallel randomized controlled trials comparing handsewn (end-to-
end or kono-s) to stapled anastomosis (hand2end and the end2end studies). BMC

cr
Surg 2024;24:71.
215. Coffey CJ, Kiernan MG, Sahebally SM, et al. Inclusion of the mesentery in ileocolic
resection for crohn's disease is associated with reduced surgical recurrence. J Crohns

us
Colitis 2018;12:1139-50.
216. Myrelid P, Soderholm JD, Olaison G, Sjodahl R, Andersson P. Split stoma in resectional
surgery of high-risk patients with ileocolonic crohn's disease. Colorectal Dis
an
2012;14:188-93.
217. Aberra FN, Lewis JD, Hass D, et al. Corticosteroids and immunomodulators:
Postoperative infectious complication risk in inflammatory bowel disease patients.
M

Gastroenterology 2003;125:320-7.
218. Rizzo G, Armuzzi A, Pugliese D, et al. Anti-tnf-alpha therapies do not increase early
postoperative complications in patients with inflammatory bowel disease. An italian
d

single-center experience. Int J Colorectal Dis 2011;26:1435-44.


219. Rottoli M, Vallicelli C, Ghignone F, et al. Predictors of early recurrence after
e

strictureplasty for crohn's disease of the small bowel during the years of biologics.
Digestive and liver disease : official journal of the Italian Society of Gastroenterology
pt

and the Italian Association for the Study of the Liver 2019;51 5:663-8.
220. Rottoli M, Tanzanu M, Manzo CA, et al. Strictureplasty for crohn's disease of the small
ce

bowel in the biologic era: Long-term outcomes and risk factors for recurrence. Tech
Coloproctol 2020;24:711-20.
221. Bislenghi G, Sucameli F, Fieuws S, et al. Non-conventional versus conventional
Ac

strictureplasties for crohn's disease. A systematic review and meta-analysis of


treatment outcomes. J Crohns Colitis 2022;16:319-30.
222. Reese GE, Purkayastha S, Tilney HS, et al. Strictureplasty vs resection in small bowel
crohn's disease: An evaluation of short-term outcomes and recurrence. Colorectal Dis
2007;9:686-94.
223. Butt WT, Ryan É J, Boland MR, et al. Strictureplasty versus bowel resection for the
surgical management of fibrostenotic crohn's disease: A systematic review and meta-
analysis. Int J Colorectal Dis 2020;35:705-17.
224. Tekkis PP, Purkayastha S, Lanitis S, et al. A comparison of segmental vs subtotal/total
colectomy for colonic crohn's disease: A meta-analysis. Colorectal disease : the
Manuscript Doi: 10.1093/ecco-jcc/jjae089
official journal of the Association of Coloproctology of Great Britain and Ireland
2006;8:82-90.
225. Angriman I, Pirozzolo G, Bardini R, et al. A systematic review of segmental vs subtotal
colectomy and subtotal colectomy vs total proctocolectomy for colonic crohn's
disease. Colorectal disease : the official journal of the Association of Coloproctology
of Great Britain and Ireland 2017;19:e279-e87.
226. Collaborative SCsoCsds. Surgical treatment of colonic crohn ’ s disease : A national

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


snapshot study. Langenbeck's Archives of Surgery 2020;406:1165-72.
227. Pellino G, Rottoli M, Mineccia M, et al. Segmental versus total colectomy for crohn's
disease in the biologic era: Results from the scotch international, multicentric study.
Journal of Crohn's and Colitis 2022;16:1853-61.

t
228. Scaringi S, Di Bella A, Boni L, et al. New perspectives on the long-term outcome of

ip
segmental colectomy for crohn’s colitis: An observational study on 200 patients. Int J
Colorectal Dis 2018;33:479-85.

cr
229. Annese V, Beaugerie L, Egan L, et al. European evidence-based consensus:
Inflammatory bowel disease and malignancies. J Crohns Colitis 2015;9:945-65.
230. Pedersen N, Duricova D, Elkjaer M, et al. Risk of extra-intestinal cancer in

us
inflammatory bowel disease: Meta-analysis of population-based cohort studies. Am J
Gastroenterol 2010;105:1480-7.
231. Lutgens MW, van Oijen MG, van der Heijden GJ, et al. Declining risk of colorectal
an
cancer in inflammatory bowel disease: An updated meta-analysis of population-
based cohort studies. Inflamm Bowel Dis 2013;19:789-99.
232. Ording AG, Horváth-Puhó E, Erichsen R, et al. Five-year mortality in colorectal cancer
M

patients with ulcerative colitis or crohn's disease: A nationwide population-based


cohort study. Inflammatory Bowel Diseases 2013;19:800–5.
233. Ali RA, Dooley C, Comber H, Newell J, Egan LJ. Clinical features, treatment, and
d

survival of patients with colorectal cancer with or without inflammatory bowel


disease. Clin Gastroenterol Hepatol 2011;9:584-9.e1-2.
e

234. Mescoli C, Albertoni L, D'Incá R, Rugge M. Dysplasia in inflammatory bowel diseases.


Dig Liver Dis 2013;45:186-94.
pt

235. Neri B, Mancone R, Savino L, et al. Mucinous and signet-ring cell colonic
adenocarcinoma in inflammatory bowel disease: A case-control study. Cancers
ce

(Basel) 2023;15.
236. Kiran RP, Nisar PJ, Goldblum JR, et al. Dysplasia associated with crohn's colitis:
Segmental colectomy or more extended resection? Ann Surg 2012;256:221-6.
Ac

237. Maser EA, Sachar DB, Kruse D, et al. High rates of metachronous colon cancer or
dysplasia after segmental resection or subtotal colectomy in crohn's colitis. Inflamm
Bowel Dis 2013;19:1827-32.
238. Olén O, Erichsen R, Sachs MC, et al. Colorectal cancer in crohn's disease: A
scandinavian population-based cohort study. Lancet Gastroenterol Hepatol
2020;5:475-84.
239. Sensi B, Khan J, Warusavitarne J, et al. Long-term oncological outcome of segmental
versus extended colectomy for colorectal cancer in crohn's disease: Results from an
international multicentre study. J Crohns Colitis 2022;16:954-62.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
240. Bogach J, Pond G, Eskicioglu C, Simunovic M, Seow H. Extent of surgical resection in
inflammatory bowel disease associated colorectal cancer: A population-based study.
Journal of Gastrointestinal Surgery 2021;25:2610-8.
241. Connelly TM, Koltun WA. The surgical treatment of inflammatory bowel disease-
associated dysplasia. Expert Rev Gastroenterol Hepatol 2013;7:307-21; quiz 22.
242. Van Cutsem E, Nordlinger B, Cervantes A. Advanced colorectal cancer: Esmo clinical
practice guidelines for treatment. Ann Oncol 2010;21 Suppl 5:v93-7.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


243. Lovasz BD, Lakatos L, Golovics PA, et al. Risk of colorectal cancer in crohn's disease
patients with colonic involvement and stenosing disease in a population-based
cohort from hungary. J Gastrointestin Liver Dis 2013;22:265-8.
244. Uzzan M, Stefanescu C, Maggiori L, et al. Case series: Does a combination of anti-tnf

t
antibodies and transient ileal fecal stream diversion in severe crohn's colitis with

ip
perianal fistula prevent definitive stoma? Am J Gastroenterol 2013;108:1666-8.
245. Burman JH, Thompson H, Cooke WT, Williams JA. The effects of diversion of intestinal

cr
contents on the progress of crohn's disease of the large bowel. Gut 1971;12:11-5.
246. Edwards CM, George BD, Jewell DP, et al. Role of a defunctioning stoma in the
management of large bowel crohn's disease. The British journal of surgery

us
2000;87:1063-6.
247. Harper PH, Truelove SC, Lee EC, Kettlewell MG, Jewell DP. Split ileostomy and
ileocolostomy for crohn's disease of the colon and ulcerative colitis: A 20 year survey.
an
Gut 1983;24:106-13.
248. Spivak J, Landers CJ, Vasiliauskas EA, et al. Antibodies to i2 predict clinical response to
fecal diversion in crohn's disease. Inflammatory bowel diseases 2006;12:1122-30.
M

249. Bafford AC, Latushko A, Hansraj N, Jambaulikar G, Ghazi LJ. The use of temporary
fecal diversion in colonic and perianal crohn's disease does not improve outcomes.
Digestive diseases and sciences 2017;62:2079-86.
d

250. Mennigen R, Heptner B, Senninger N, Rijcken E. Temporary fecal diversion in the


management of colorectal and perianal crohn's disease. Gastroenterology research
e

and practice 2015;2015:286315-.


251. Bemelman WA, Warusavitarne J, Sampietro GM, et al. Ecco-escp consensus on
pt

surgery for crohn's disease. Journal of Crohn's & colitis 2018;12:1-16.


252. Chang S, Shen B, Remzi F. When not to pouch: Important considerations for patient
ce

selection for ileal pouch-anal anastomosis. Gastroenterology & hepatology


2017;13:466-75.
253. Fazio VW, Kiran RP, Remzi FH, et al. Ileal pouch anal anastomosis: Analysis of
Ac

outcome and quality of life in 3707 patients. Ann Surg 2013;257:679-85.


254. Le Q, Melmed G, Dubinsky M, et al. Surgical outcome of ileal pouch-anal anastomosis
when used intentionally for well-defined crohn's disease. Inflammatory bowel
diseases 2013;19:30-6.
255. Manilich E, Remzi FH, Fazio VW, Church JM, Kiran RP. Prognostic modeling of
preoperative risk factors of pouch failure. Dis Colon Rectum 2012;55:393-9.
256. Reese GE, Lovegrove RE, Tilney HS, et al. The effect of crohn's disease on outcomes
after restorative proctocolectomy. Diseases of the colon and rectum 2007;50:239-50.
257. Ananthakrishnan AN, McGinley EL, Binion DG. Does it matter where you are
hospitalized for inflammatory bowel disease? A nationwide analysis of hospital
volume. Am J Gastroenterol 2008;103:2789-98.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
258. Williams H, Alabbadi S, Khaitov S, Egorova N, Greenstein A. Association of hospital
volume with postoperative outcomes in crohn's disease. Colorectal Disease
2023;25:688-94.
259. Fiorino G, Lytras T, Younge L, et al. Quality of care standards in inflammatory bowel
diseases: A european crohn's and colitis organisation *ecco+ position paper. Journal of
Crohn's & colitis 2020;14:1037-48.
260. !!! INVALID CITATION !!! 12.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


261. Candia R B-SGAMHHCNGC. Colonoscopy-guided therapy for the prevention of post-
operative recurrence of crohn’s disease (review). Cochrane Library 2020.
262. De Cruz P, Kamm MA, Prideaux L, Allen PB, Desmond PV. Postoperative recurrent
luminal crohn's disease: A systematic review. Inflammatory Bowel Diseases

t
2012;18:758-77.

ip
263. De Cruz P, Kamm MA, Hamilton AL, et al. Crohn's disease management after
intestinal resection: A randomised trial. The Lancet 2015;385:1406-17.

cr
264. Schlussel AT, Cherng NB, Alavi K. Current trends and challenges in the postoperative
medical management of crohn's disease: A systematic review: Elsevier Inc.; 2017.
265. De Cruz P, Hamilton AL, Burrell KJ, et al. Endoscopic prediction of crohn's disease

us
postoperative recurrence. Inflammatory Bowel Diseases 2022;28:680-8.
266. Yamamoto T, Watanabe T. Strategies for the prevention of postoperative recurrence
of crohn's disease. Colorectal Disease 2013;15:1471-80.
an
267. Gjuladin-Hellon T, Gordon M, Iheozor-Ejiofor Z, Akobeng AK. Oral 5-aminosalicylic
acid for maintenance of surgically-induced remission in crohn's disease. Cochrane
Database Syst Rev 2019;6:Cd008414.
M

268. Regueiro M, Feagan BG, Zou B, et al. Infliximab reduces endoscopic, but not clinical,
recurrence of crohn's disease after ileocolonic resection. Gastroenterology
2016;150:1568-78.
d

269. Zhao Y, Ma T, Chen YF, et al. Biologics for the prevention of postoperative crohn's
disease recurrence: A systematic review and meta-analysis. Clin Res Hepatol
e

Gastroenterol 2015;39:637-49.
270. Ferrante M, Pouillon L, Mañosa M, et al. Results of the eighth scientific workshop of
pt

ecco: Prevention and treatment of postoperative recurrence in patients with crohn’s


disease undergoing an ileocolonic resection with ileocolonic anastomosis. Journal of
ce

Crohn's and Colitis 2023;17:1707-22.


271. D'Haens G, Taxonera C, Lopez-Sanroman A, et al. Op14 prevention of postoperative
recurrence of crohn's disease with vedolizumab: First results of the prospective
Ac

placebo-controlled randomised trial reprevio. Journal of Crohn's and Colitis


2023;17:i19-i.
272. Mañosa M, Fernández-Clotet A, Nos P, et al. Ustekinumab and vedolizumab for the
prevention of postoperative recurrence of crohn's disease: Results from the eneida
registry. Dig Liver Dis 2023;55:46-52.
273. Ozgur I, Kulle CB, Buyuk M, et al. What are the predictors for recurrence of crohn's
disease after surgery? Medicine 2021;100:e25340.
274. Lee KE, Cantrell S, Shen B, Faye AS. Post-operative prevention and monitoring of
crohn’s disease recurrence. Gastroenterology Report 2022;10.
Manuscript Doi: 10.1093/ecco-jcc/jjae089
275. Scarpa M, Pilon F, Pengo V, et al. Deep venous thrombosis after surgery for
inflammatory bowel disease: Is standard dose low molecular weight heparin
prophylaxis enough? World journal of surgery 2010;34:1629-36.
276. McKechnie T, Wang J, Springer JE, et al. Extended thromboprophylaxis following
colorectal surgery in patients with inflammatory bowel disease: A comprehensive
systematic clinical review. Colorectal Disease 2020;22:663-78.

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


t
ip
cr
us
an
M
e d
pt
ce
Ac
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Figures

Figure 1. Classification of perianal fistulising Crohn’s disease

At any moment throughout its disease course, perianal fistulising Crohn’s disease can be

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


classified into one of four classes134

(reprinted with permission from Elsevier, License Number 5760781248280).

t
ip
cr
Class 2a: repair

us
Symptomatic fistulae suitable
for combined medical and Class 2c-i: early and Class 3: severe disease Class 4a: repair
surgical closure or repair rapidly progressive with exhausted Symptomatic sinuses
(including seton removal) disease perineum and or wounds suitable
and patient goal is fistula Early and rapidly adverse features for combined medical
Class 1: minimal disease
progressive disease Severely symptomatic and
Minimal symptoms and
an
destructive to the disease (despite patient goal
anorectal disease burden,
perineum or to quality defunctioning),
requiring minimal
Class 2: chronic of life (or both), such with irreversible
intervention over time
symptomatic that early intervention perineal
fistulae destruction, or
These patients will align
Perianal fistulising Defunctioni Proctecto Class 4: perineal
M

with one of three groups,


Crohn's disease ng my symptoms after
according to their goals, as
proctectomy
well as their symptoms
and impact on quality of Class 2c-ii: gradually Class 4b: symptom
debilitating disease control Chronic
Gradually debilitating symptoms related to
d

symptomatic fistulae sinuses or wounds that


Class 2b: symptom unsuitable for surgical affect quality of life
control Chronic repair, and that are
e

symptoms related to which cause severe


fistulae (pain and symptoms, limiting or patient goal
discharge) that affect quality of life so is symptom control
pt

quality of life. Fistulae are markedly that


currently unsuitable for
ce
Ac
Manuscript Doi: 10.1093/ecco-jcc/jjae089
Figure 2. Treatment algorithm for Class 2A CD fistulae aiming for repair.

Perianal fistula abscess

Downloaded from https://academic.oup.com/ecco-jcc/advance-article/doi/10.1093/ecco-jcc/jjae089/7693896 by guest on 20 July 2024


Seton drainage I&D + antibiotics

t
Start anti-TNF

ip
cr
No proctitis Proctitis

Single internal
opening us
Multiple internal
openings
an
Superficial Transsphincteric
M

[intersphincteric] tract
tract
e d

Fistulotomy Surgical closure Stem cells Seton removal


under anti-TNF under medical
pt

treatment
[AF or LIFT]
ce
Ac

Fistula recurrence

Class 2a [repair]: Class 2b [symptom control]

- Consider approach outside - Medical treatment


guideline *VAAFT/OVESCO+
- Chronic seton
- Experimental therapies

You might also like