Introduction
Inflammatory bowel disease (IBD) is an immune mediated disorder that
affects the intestine and has extra intestinal manifestations as well.
The first description of ulcerative colitis was in 18591. Cases
resembling Crohn’s disease have been described since 1612 but the
classical publication describing Crohn’s disease was in 1913 [1].
Management of patients with IBD has remarkably advanced from initial use
of steroid, Sulfasalazine, and Azathioprine till the complete shift
after the approval of first Anti-Tumor Necrosis Factor (TNF)
(Infliximab) by the United States Food and Drug Authority (FDA) in 1998
[2]. Since then, there have been numerous medications targeting various
pathways have been introduced to market.
IBD encompasses Crohn’s disease, ulcerative colitis and indeterminant
colitis. Ulcerative colitis is confined to the colon and might involve
the most distal part of the small bowel (Backwash ileitis), while
Crohn’s disease can involve any part of the gastrointestal tract. In a
small proportion of IBD patients (5 to 15%) it is not clear whether the
patient has Crohn’s disease or ulcerative colitis and with time the
disease might declare itself [3]. Also, the disease is characterized by
periods of flares and remissions and there is an accelerative amount of
bowel damage that happens with each flare of attack.
In Saudi Arabia one of the first publications describing IBD was in
1998 [4] and since then multiple studies have described the phenotypic
characteristics and disease behavior and response to therapies in the
Saudi population and more recently numerous national guidelines have
been published that details the management of IBD in general [5] and
special populations as well [6,7].
Multiple elements have been proposed as a cause for the development
of IBD and these include factors like genetics, environmental exposures
either early in life (breastfeeding and antibiotic exposure) or latter
(medications, stress, smoking, infections) [8] (Figure 1). It is this
interplay between genetics, environment and microbiome that cause the
disease and is evident by the increasing incidence in areas where the
disease was not prevalent in the past as well as the development of IBD
in immigrant populations and with the industrialization of
societies.
Also, the direct (medical) and indirect (societal) costs associated
with the disease or not insignificant and with the increased therapeutic
options have become an area of active research to achieve the best
possible outcome with a reasonable amount of healthcare resources
[9].
Chapter 1: Epidemiology and disease burden
Improved diagnostics, therapeutics, and updated management targets
have improved patients’ survival and quality of life. In addition to
this, due to decreased mortality and the ageing of populations this has
contributed to the increased prevalence of the disease [10].
The prevalence of IBD has increased over the years mostly in
industrialized countries and is most prevalent in North America and
Europe and is least prevalent in Sub-Saharan Africa (Figure 2). Although
IBD is not very prevalent, it remains a cause of significant morbidity
or mortality in a younger population, effects their quality of life, is
associated with an increase in the sum of the years of life lost (YLL)
to due to premature mortality, in years lived with a disability (YLD) as
well as an increase in disability-adjusted life years (DALYs)[11]
(Figure 3).
As expected, due to the higher prevalence of IBD in North America and
Europe, these areas have the highest burden related to disease
associated life years (Figures 4 A & B), but in terms of years of
life lost areas with a low sociodemographic index (SDI) like the
geographical area of Sub-Saharan Africa are disproportionally affected.
This might reflect limited access to care and diagnostic and therapeutic
interventions.
The disease effects both males and females in almost identical
proportions [12,13]. Also, the age distribution that both diseases
effect is almost similar with a tendency for UC to effect those above
the age of 40 years more frequently then Crohn’s disease.
The disease distribution for both diseases and the behavior of
Crohn’s disease and severity of ulcerative colitis vary (Chapter 3).
There is also a risk of developing Crohn’s disease or ulcerative
colitis base on the family history of IBD [14] is shown in table 1 as
well as figures 5 A & B. Also, the relative risks associated with
developing both Crohn’s disease or ulcerative colitis from some
environmental exposures [15] is shown as a heatmap in Figure 6.
library(DiagrammeR)
grViz("
digraph risk_factors {
graph [layout = neato, overlap = false, fontname = Helvetica,
bgcolor = 'transparent', splines = true]
node [shape = circle, style = 'filled, rounded', fillcolor = 'lightblue',
fontname = Helvetica, width = 1.2, height = 1.2, fontcolor = 'black']
edge [color = grey70, penwidth = 2]
# Central node
IBD [label = 'IBD Risk', fillcolor = 'salmon', width = 1.5, height = 1.5,
fontsize = 16, fontcolor = 'white']
# Risk factor nodes with better color contrast
Genetic [label = 'Genetic\nSusceptibility', fillcolor = 'lavender', pos = '0,4!']
Microbiome [label = 'Microbiome\nImbalance', fillcolor = 'lightyellow', pos = '2,4!']
Smoking [label = 'Smoking', fillcolor = 'plum', pos = '4,3!']
Appendectomy [label = 'Appendectomy', fillcolor = 'wheat', pos = '4,1!']
Medications [label = 'Medications', fillcolor = 'lightcyan', pos = '2,-4!']
Hygiene [label = 'Hygiene', fillcolor = 'lightpink', pos = '0,-4!']
Diet [label = 'Dietary\nHabits', fillcolor = 'khaki', pos = '-2,-4!']
Sleep [label = 'Sleep', fillcolor = 'lavender', pos = '-4,-3!']
VitD [label = 'Vit D & UV\nexposure', fillcolor = 'gold', pos = '-4,-1!']
Stress [label = 'Stress', fillcolor = 'tan', pos = '-4,1!']
Activity [label = 'Physical\nactivity', fillcolor = 'skyblue', pos = '-4,3!']
Antibiotics [label = 'Antibiotic\nExposure', fillcolor = 'coral', pos = '-2,4!']
BreastFeeding [label = 'Breast\nFeeding', fillcolor = 'palegreen', pos = '2,-2!']
Geography [label = 'Geographical\nFactors', fillcolor = 'thistle', pos = '0,-2!']
Infections [label = 'Infections', fillcolor = 'lightcoral', pos = '4,-1!'] # Changed from salmon to lightcoral
# Connect all factors to IBD center
Genetic -> IBD
Microbiome -> IBD
Smoking -> IBD
Appendectomy -> IBD
Medications -> IBD
Hygiene -> IBD
Diet -> IBD
Sleep -> IBD
VitD -> IBD
Stress -> IBD
Activity -> IBD
Antibiotics -> IBD
BreastFeeding -> IBD
Geography -> IBD
Infections -> IBD
# Add some cross-connections between related factors
Genetic -> Microbiome [style = dashed, dir = both, constraint = false]
Diet -> Microbiome [style = dashed, dir = both, constraint = false]
Antibiotics -> Microbiome [style = dashed, dir = both, constraint = false]
Geography -> VitD [style = dashed, dir = both, constraint = false]
Stress -> Sleep [style = dashed, dir = both, constraint = false]
Hygiene -> Infections [style = dashed, dir = both, constraint = false]
}
")
Figure 1. Various factors that have been associated with inflammatory
bowel disorder. (Adapted from Ananthakrishnan A.N.[8])

Figure 2. Prevalence per 100,000 population of inflammatory bowel
disease by geographic region (adapted from Piovani D. et al.[11]).

Figure 3. The burden of IBD on populations in terms of Disease
Associated Life Years (DALYs), Years Lived with a Disability (YLD), and
Years of Life Lost (YLL) by geographic region (adapted from Piovani D.
et al.[11]).
library(ggplot2)
library(dplyr)
library(forcats)
library(scales)
# Create the data frame
regional_data <- data.frame(
Region = c("NORTH AMERICA", "EUROPE", "GLOBAL", "EAST ASIA AND PACIFIC",
"SUB-SAHARAN AFRICA", "CENTRAL ASIA", "LATIN AMERICA AND CARIBBEAN",
"SOUTH ASIA", "MIDDLE EAST AND NORTH AFRICA"),
Value = c(79.8, 33.1, 23.2, 21.3, 16.2, 15.2, 14.8, 13.3, 9)
)
# Create a clean, professional bar chart
ggplot(regional_data, aes(x = fct_reorder(Region, Value),
y = Value, fill = Region)) +
geom_col(alpha = 0.8, width = 0.7) +
geom_text(aes(label = Value), hjust = -0.1, size = 3.5, fontface = "bold") +
scale_fill_manual(values = c(
"NORTH AMERICA" = "#E41A1C", # Red for highest value
"EUROPE" = "#377EB8", # Blue
"GLOBAL" = "darkgray", # Gray for global reference
"EAST ASIA AND PACIFIC" = "#4DAF4A", # Green
"SUB-SAHARAN AFRICA" = "#984EA3", # Purple
"CENTRAL ASIA" = "#FF7F00", # Orange
"LATIN AMERICA AND CARIBBEAN" = "#FFFF33", # Yellow
"SOUTH ASIA" = "#A65628", # Brown
"MIDDLE EAST AND NORTH AFRICA" = "#F781BF" # Pink
)) +
labs(
title = "Disease Associated Life Years (DALYs) associated with IBD",
subtitle = "By geographic region per 100,000 population",
x = "",
y = "Rate per 100,000",
caption = "Data represents standardized rates across regions"
) +
coord_flip() + # Flip to make long region names readable
theme_minimal() +
theme(
axis.text.y = element_text(size = 10, face = "bold"),
axis.text.x = element_text(size = 10),
axis.title.x = element_text(size = 12, face = "bold", margin = margin(t = 10)),
plot.title = element_text(size = 16, face = "bold", hjust = 0.5),
plot.subtitle = element_text(size = 12, hjust = 0.5, margin = margin(b = 15)),
legend.position = "none",
panel.grid.major.y = element_blank(),
panel.grid.minor.x = element_blank()
) +
scale_y_continuous(expand = expansion(mult = c(0, 0.1))) # Add space for labels

Figure 4 A. Disease Associated Life Years (DALYs) associated with
inflammatory bowel disease by geographic region (adapted from Piovani D.
et al.[11]).
library(ggplot2)
library(dplyr)
library(forcats)
library(scales)
# Create the data frame
regional_data <- data.frame(
Region = c("NORTH AMERICA", "EUROPE", "EAST ASIA AND PACIFIC", "GLOBAL",
"LATIN AMERICA AND CARIBBEAN", "CENTRAL ASIA",
"MIDDLE EAST AND NORTH AFRICA", "SOUTH ASIA", "SUB-SAHARAN AFRICA"),
Value = c(63, 18.2, 14.5, 12.6, 5.05, 4.97, 4.52, 2.5, 1.77)
)
# Create a clean, professional bar chart
ggplot(regional_data, aes(x = fct_reorder(Region, Value),
y = Value, fill = Region)) +
geom_col(alpha = 0.8, width = 0.7) +
geom_text(aes(label = Value), hjust = -0.1, size = 3.5, fontface = "bold") +
scale_fill_manual(values = c(
"NORTH AMERICA" = "#E41A1C", # Red for highest value
"EUROPE" = "#377EB8", # Blue
"EAST ASIA AND PACIFIC" = "#4DAF4A", # Green
"GLOBAL" = "darkgray", # Gray for global reference
"LATIN AMERICA AND CARIBBEAN" = "#FF7F00", # Orange
"CENTRAL ASIA" = "#FFFF33", # Yellow
"MIDDLE EAST AND NORTH AFRICA" = "#A65628", # Brown
"SOUTH ASIA" = "#F781BF", # Pink
"SUB-SAHARAN AFRICA" = "#984EA3" # Purple
)) +
labs(
title = "Years lived with a disability from IBD",
subtitle = "By geographic region per 100,000 population",
x = "",
y = "Rate per 100,000",
caption = "Data highlights the significant burden in Western regions compared to developing areas"
) +
coord_flip() + # Flip to make long region names readable
theme_minimal() +
theme(
axis.text.y = element_text(size = 10, face = "bold"),
axis.text.x = element_text(size = 10),
axis.title.x = element_text(size = 12, face = "bold", margin = margin(t = 10)),
plot.title = element_text(size = 16, face = "bold", hjust = 0.5),
plot.subtitle = element_text(size = 12, hjust = 0.5, margin = margin(b = 15)),
legend.position = "none",
panel.grid.major.y = element_blank(),
panel.grid.minor.x = element_blank(),
plot.caption = element_text(size = 9, hjust = 0.5, face = "italic", margin = margin(t = 10))
) +
scale_y_continuous(expand = expansion(mult = c(0, 0.15))) # Add more space for labels

Figure 4 B. Years lived with a disability from inflammatory bowel
disease by geographic region (adapted from Piovani D. et al.11).
(adapted from Piovani D. et al.[11]).

Figure 4 C. Years of life lost to due to premature mortality (YLLs)
from inflammatory bowel disease by geographic region (adapted from
Piovani D. et al.[11]).

Table 1. The 10-year risk of developing Crohn’s disease or ulcerative
colitis based on the family history and the current age of the
individual (Adapted from Moller FT et al.[14]).
library(knitr)
library(kableExtra)

Figure 5 A. Heat map of the 10-year risk of developing Crohn’s
disease based on the family history and the current age of the
individual (Adapted from Moller FT et al.[14]).

Figure 5 B. Heat map of the 10-year risk of developing ulcerative
colitis based on the family history and the current age of the
individual (Adapted from Moller FT et al.[14]).

Figure 6. Heatmap of the relative risk of developing ulcerative
colitis or Crohn’s disease based on environmental factors (Adapted from
Piovani D et al.[15]).
References
- Kirsner JB. Historical origins of current IBD concepts. World J
Gastroenterol. Apr 2001;7(2):175-84. doi:10.3748/wjg.v7.i2.175
- Melsheimer R, Geldhof A, Apaolaza I, Schaible T. Remicade((R))
(infliximab): 20 years of contributions to science and medicine.
Biologics. 2019;13:139-178. doi:10.2147/BTT.S207246
- Venkateswaran N, Weismiller S, Clarke K. Indeterminate Colitis -
Update on Treatment Options. J Inflamm Res. 2021;14:6383-6395. doi:10.2147/JIR.S268262
- Isbister WH, Hubler M. Inflammatory bowel disease in Saudi Arabia:
presentation and initial management. J Gastroenterol Hepatol. Nov
1998;13(11):1119-24. doi:10.1111/j.1440-1746.1998.tb00587.x
- Mosli MH, Almudaiheem HY, AlAmeel T, et al. Saudi Arabia consensus
guidance for the diagnosis and management of adults with inflammatory
bowel disease. Saudi J Gastroenterol. Nov 21 2022;29(Suppl 1):S1-S35. doi:10.4103/sjg.sjg_277_22
- Azzam NA, Almutairdi A, Almudaiheem HY, et al. Saudi consensus
guidance for the management of inflammatory bowel disease during
pregnancy. Saudi J Gastroenterol. Dec 15 2023;30(4):181-97. doi:10.4103/sjg.sjg_318_23
- Saadah OI, AlAmeel T, Al Sarkhy A, et al. Saudi consensus guidance
for the diagnosis and management of inflammatory bowel disease in
children and adolescents. Saudi J Gastroenterol. Aug 30 2024;doi:10.4103/sjg.sjg_171_24
- Ananthakrishnan AN. Epidemiology and risk factors for IBD. Nat Rev
Gastroenterol Hepatol. Apr 2015;12(4):205-17. doi:10.1038/nrgastro.2015.34
- AlRuthia Y, Alharbi O, Aljebreen AM, et al. Drug utilization and
cost associated with inflammatory bowel disease management in Saudi
Arabia. Cost Eff Resour Alloc. 2019;17:25. doi:10.1186/s12962-019-0194-3
- Kaplan GG, Windsor JW. The four epidemiological stages in the global
evolution of inflammatory bowel disease. Nat Rev Gastroenterol Hepatol.
Jan 2021;18(1):56-66. doi:10.1038/s41575-020-00360-x
- Piovani D, Danese S, Peyrin-Biroulet L, Bonovas S. Inflammatory
bowel disease: estimates from the global burden of disease 2017 study.
Aliment Pharmacol Ther. Jan 2020;51(2):261-270. doi:10.1111/apt.15542
- Aljebreen AM, Alharbi OR, Azzam NA, Almalki AS, Alswat KA, Almadi
MA. Clinical epidemiology and phenotypic characteristics of Crohn’s
disease in the central region of Saudi Arabia. Saudi J Gastroenterol.
May-Jun 2014;20(3):162-9. doi:10.4103/1319-3767.132993
- Alharbi OR, Azzam NA, Almalki AS, et al. Clinical epidemiology of
ulcerative colitis in Arabs based on the Montreal classification. World
J Gastroenterol. Dec 14 2014;20(46):17525-31. doi:10.3748/wjg.v20.i46.17525
- Moller FT, Andersen V, Wohlfahrt J, Jess T. Familial risk of
inflammatory bowel disease: a population-based cohort study 1977-2011.
Am J Gastroenterol. Apr 2015;110(4):564-71. doi:10.1038/ajg.2015.50
- Piovani D, Danese S, Peyrin-Biroulet L, Nikolopoulos GK, Lytras T,
Bonovas S. Environmental Risk Factors for Inflammatory Bowel Diseases:
An Umbrella Review of Meta-analyses. Gastroenterology. Sep
2019;157(3):647-659 e4. doi:10.1053/j.gastro.2019.04.016
Chapter 2: Etiology and Pathogenesis
Inflammatory bowel disease (IBD) etiology may involve the host immune
system dysregulation, genetic predisposition, gut microbiota dysbiosis,
and environmental triggers [1].
The intestinal mucosa consists of epithelial cells, goblet cells,
Paneth cells, stroma, and immune cells. The intestinal epithelium
includes epithelial cells closely bound by tight junctions. The
intestine is structured with a villi and invaginations called crypts of
Lieberkühn. The goblet and Paneth cells produce mucus and antimicrobial
peptides respectively, thus limiting the spread of luminal
microorganisms [2].
A loss of mucus layer thickness by marked reduction in goblet cell
numbers has been linked to Crohn’s disease [3], and abnormal mucus
composition has been reported in UC [4]. The lamina propria, contains
stromal cells, including fibroblasts, myofibroblasts, and perivascular
pericytes which serve the function of fibrosis and wound healing [2],
and may be related to the aggravation of UC through their capacity to
produce chemokines, including chemokine (C-C motif) ligand (CCL)19,
CCL21, and the immune-system regulator interleukin (IL)-33 [5]. Plasma
cells release immunoglobulin (Ig)A that inhibit the infiltration of
pathogenic microorganisms and help in sustaining a homeostatic
equilibrium between the host and commensal microbiota.
Both the epithelium and other non-immune intestinal components are
important mediators of intestinal homeostasis and IBD pathophysiology,
[6,7]. Some of the functions of these non-immune cells are mediated
through interaction with components of the immune system.
Immune system dysregulation
The mucosal immune system is the most extensive part of the immune
system. intestinal immune cells are involved in a highly balanced immune
response aimed at controlling pathogen invasion, while stopping an
excessive immune responses against innocuous food antigens and commensal
microbes that could risk unintentional tissue injury (Figure 1).
The immune system confers host defense against pathogens and provides
anti-tumor protection. At the same time, regulatory mechanisms
counterbalance these responses to prevent reactions against self and
innocuous external antigens, thus promoting a state of tolerance.
The immune system can be classified into innate and adaptive
immunity. Innate immunity that is composed of myeloid cells, initiates
rapid responses to conserved structural motifs on microorganisms. Innate
immune cells (IIC) express pattern recognition receptors (PRRs), such as
toll-like receptors (TLRs) and Nod-like receptors (NLR), allowing them
to distinguish pathogen-associated molecular patterns (PAMPs) and
damage-associated molecular patterns (DAMPs).
IIC provide host defense and inflammation by generating cytokines and
chemokines, triggering the complement cascade and phagocytosis, or
stimulating adaptive immunity by presenting antigens. IIC include
neutrophils, monocytes, macrophages, and dendritic cells (DCs)
[8,9].

Figure 1 damage to the intestinal barrier triggers the recruitment of
neutrophils from the circulation to the inflamed tissue along a
chemotactic gradient formed by cytokines (IL-1β, IL-6, TNF-α),
chemokines (CCL8, CXCL10, MIP-2), and growth factors (GM-CSF, G-CSF).
Neutrophil recruitment is also mediated by bacteria-derived molecules
such as formyl-methionyl-leucyl- phenylalanine (fMLP) and short-chain
fatty acids (SCFAs). The recruited neutrophils participate in the
elimination of microorganisms through phagocytosis, degranulation,
reactive oxygen species (ROS) generation, and the release of neutrophil
extracellular traps (NETs). Once their functions are completed,
neutrophils undergo apoptosis and efferocytosis, facilitating the
resolution of inflammation, tissue repair, and a return to normal tissue
homeostasis. The participation of neutrophils and NETs in IBD is a
double-edged sword.
Some intestinal cell populations can adjust their functions to the
needs of the intestinal microenvironment under steady state. This
adaptation can be harmful in IBD, but is also a potential therapeutic
target for the treatment of the disease.
Neutrophils are the most numerous immune cells in the human
circulation and are quickly recruited to sites of infection or
inflammation [10]. It plays a role in intestinal homeostasis and
inflammation, playing an essential role in gut defense but also being an
important mediator of tissue damage in the inflamed mucosa upon
excessive recruitment. Several studies demonstrate the effect of
neutrophils on other components of the intestinal mucosa in IBD, such as
other immune cells and epithelial cells and other non-immune cells
[11].
Intestinal macrophages, which restrain their robust proinflammatory
potential through a natural resistance to producing inflammatory
mediators in response to pattern-recognition molecules, while also
retaining several of their homeostatic abilities, including
phagocytosing bacteria, preserving Tregs and maintaining tolerance, and
promoting epithelial cell renewal [12]. In the intestinal
microenvironment, macrophages adapt their functions to the context. For
example, CX3CR1 high macrophages can distinguish harmful from commensal
bacteria via TLR and NLR recognition. In the intestinal
microenvironment, CX3CR1 high macrophages are excellent phagocytes, but
produce low levels of pro-inflammatory cytokines and maintain tolerance
through the production of anti-inflammatory cytokines such as IL-10
[13]. CX3CR1 high intestinal macrophages sense and take up bacterial
antigen from the intestinal lumen via their transepithelial dendrites
[14-19]. In homeostasis, the intestinal microbiota inhibits the
migration of antigen-loaded CX3CR1 high intestinal macrophages to
mesenteric lymph nodes, thereby also inhibiting antigen presentation to
T cells, and effectively sustaining tolerance towards commensal
bacteria.
The etiology of IBD remains unknown, but IBD appears to be sustained
in genetically susceptible individuals by an impaired immune response
against intestinal microorganisms. This abnormal immune response is
associated with dysregulation of both innate and adaptive immune
responses.
IBD is characterized by penetration of the epithelial barrier of the
intestine, and non-resolving mucosal damage is a major component of the
disease [20]. While generally unknown, the cause of this damage could be
related to an infectious agent [21], a chemical compound [1], or a
metabolic alteration probably related to diet-mediated dysbiosis [22].
Unsuccessful resolution of inflammation is supported by disruption of
tolerance towards commensal microorganisms or autologous signals of
tissue damage [23].
Genetic predisposition
Comparison of first-degree relatives of IBD patients with the general
population reveals a heritable risk of CD and UC [23-24]. Genome wide
association studies (GWAS) have identified more than 240 risk variants
associated with IBD. These variants are found in genes related to
bacteria recognition (e.g., NOD2), autophagy (e.g., ATG16L1 and IRGM),
regulation of epithelial barrier (e.g., ECM1), and innate and adaptive
immunity (e.g., IL-23R, IL-10, ITGAL, and ICAM1 variants) [25,26]. Based
on that, it has been possible to uncover fundamental molecular features
underlying the disease and to identify genes and signaling pathways that
represent potential therapeutic targets or biomarkers. However, only a
small percentage of the disease variance in CD and UC can be linked to
recognized IBD risk loci [27].
To resolve this limitation, new techniques have allowed the study of
single-cell-specific transcriptional profiles. For example, single-cell
RNA sequencing (scRNA-seq) and high-dimensional protein analyses, such
as mass cytometry and multichannel spectral cytometry, have defined
IBD-linked profiles and detected cell sub-populations that are elevated
or diminished in IBD, particularly populations of fibroblasts [5],
epithelial cells [4], and immune cells [28-31].
A complementary approach to GWAS is through transcriptome wide
association studies (TWAS), which associate gene expression with genetic
susceptibility to disease, providing functional insight into risk loci
[32]. TWAS findings have provided understanding of tissue-specific
molecular events underlying genetic susceptibility to IBD. Associated
genes are potential targets for new treatments and could be prioritized
in functional studies.
Intestinal microbial dysbiosis
The gut microbiota is an important physical, chemical, and
immunological interface between the environment and host; thus, any
dysregulation or breakdown of this barrier can contribute to disease
states (Figure 2). For example, altered physical epithelial barrier
function, a thinner mucus layer, and altered responses to endoplasmic
reticulum stress (via mutations in MUC19, ITLN1, FUT2, and XBP1) have
all been identified as risk factors for IBD [33-35].
Currently, the pathogenesis of human IBD is believed to involve
inappropriate activation of the immune system when genetically
susceptible individuals are exposed to gut antigens, such as microbiome
components [36]. Although alterations in the gut microbiome have been
proposed to be critical in IBD pathogenesis, it is not yet clear how
this process occurs and whether dysbiosis is a central cause or a common
consequence of the disease [37].
In healthy individuals, 99% of gut bacterial are Firmicutes,
Bacteroidetes, Proteobacteria, and Actinobacteria. Firmicutes and
Bacteroidetes account for approximately 90% of the total microbiome
composition. These phyla are critically important in maintaining gut
homeostasis and produce short-chain fatty acids (SCFAs), especially
butyrate and propionate, from the fermentation of dietary components
such as indigestible fibers. SCFAs are important energy sources for
colonic mucosa cells but have also been shown to play key roles in
regulating immune homeostasis [38].
Dysbiosis is defined as an alteration in gut microbiota composition
and diversity and a shift in the balance between commensal and
potentially pathogenic microorganisms [39]. Several pieces of evidence
support the role of the microbiome and dysbiosis in IBD development. For
example, experimental mice subjected to germ free conditions develop
attenuated colitis [40]. In studies using mouse models, the transfer of
bacterial strains associated with IBD induces intestinal inflammation in
genetically susceptible mice [41]. Similarly, fecal transplantation from
human IBD donors to germ-free mice stimulates proinflammatory responses,
with increased Th17 cell infiltration and proinflammatory mediators
compared with transplants from healthy human donors [42].
Extensive research focuses on determining the groups of microbes that
are related to the development of intestinal inflammation. Patients with
IBD tend to present several changes, not only in composition, but also
in the diversity of their microbiome populations when compared to
healthy individuals. Evidence shows that alterations in microbiome
components can also be involved in different IBD phenotypes [43]. The
IBD microbiota has been characterized by an increase in the
Bacteroidetes and Proteobacteria and a decrease in Firmicutes compared
to control individuals. Specifically, levels of Faecalibacterium
prausnitzii, a highly metabolically active commensal bacterium, are
reduced in individuals with IBD [44]. Patients with IBD have reduced
microbiome diversity (mostly a decrease in the Firmicutes) and an
increase in the presence of Proteobacteria, such as Enterobacteriaceae
and Bilophila, and certain members of Bacteroidetes [45]. Dysbiosis can
potentially lead to a reduction in key functions necessary for
maintaining intestinal barrier integrity and gut homeostasis. Therefore,
alterations in the immune response and proinflammatory activity could be
due to a dysbiotic microenvironment.

Figure 2: The role of gut dysbiosis in the pathogenesis of IBD. Gut
microbiota reflect an interaction of host genetics with dynamic exposure
to innumerable stimuli from the exposome. Crosstalk amongst these
factors results in long-standing consequences to the gut microbiota and
epigenetic modifications in a multidirectional fashion, potentially
affecting and epigenetic modifications in a multidirectional fashion,
potentially affecting susceptibility to diseases. The prevalence of
either regulatory (eubiosis) or inflammatory (dysbiosis) species within
the gut microbial community determines the respective predominant immune
response. Treg, regulatory response. Treg, regulatory T-cell; Breg,
regulatory B-cell; ILC, innate lymphoid cell; IgA, T-cell; Breg,
regulatory B-cell; ILC, innate lymphoid cell; IgA, immunoglobulin A; MØ,
macrophage; immunoglobulin A; MØ, macrophage; TSLP, thymic stromal
lymphopoietin. TSLP, thymic stromal lymphopoietin.
Environmental triggers
Epidemiologic data suggest a strong role of the environment [46-48].
Disease concordance in monozygotic twins approaches 50% at best, with
many studies suggesting that the estimates may be lower. The risk of IBD
in the immigrant population resembles that of country of residence
rather than the country of origin [49].
Also, countries that have witnessed a rapidly changing environment
and lifestyle have seen an increase in the incidence of IBD over the
past few decades at a rate of change that outpaces what could be
attributed solely to genetics [50]. Many environmental triggers for IBD
include smoking, Vitamin D deficiency, medications including
antibiotics, stress, diet, and air pollution.
Smoking remains the most widely studied and replicated environmental
trigger for CD and UC. the first described protective association
between UC and smoking was in 1982, subsequent studies confirmed the
inverse effect of current smoking on the development of UC, lower rate
of relapse, and reduced need for colectomy in current smokers [51-54].
However, cessation of smoking is associated with an increase in risk of
UC within 2–5 years of cessation. On the other hand, smoking increases
the risk of developing CD two-fold [55-56], increases risk of disease
flares, need for steroids and is associated with a higher rate of
post-operative disease recurrence [56-58]. Despite strong epidemiologic
data, the mechanism how smoking impacts IBD remains unclear as does the
reason for its protective effect in UC but deleterious impact on CD.
There has been increasing recognition of the immunologic role of
vitamin D [59-61]. Many studies suggest that the role of vitamin D is
fairly varied and associated with a diverse spectrum of diseases. A
deficiency of vitamin D could be a consequence of IBD itself with
reduced physical activity, sunlight exposure, malnutrition, inadequate
dietary intake of vitamin D, or lower bioavailability, all contributing
to the deficiency [62-64]. However, vitamin D deficiency is common even
in newly diagnosed IBD patients suggesting that low vitamin D itself can
contribute to increased risk of IBD [65].
Medications adverse effects like aspirin and non-steroidal
anti-inflammatory drugs (NSAIDs) are well recognized. However, while
their potential effect in triggering onset or relapse of IBD has been
clinical suspected, limited high quality evidence is available to
support this. Most studies where case-controls where confounding by
indication is a possibility, identifying use of aspirin or NSAIDs to
treat pre-diagnosis symptoms of CD and UC, and differentiating NSAID
enteritis or colitis from true idiopathic IBD have been difficult to
achieve. high dose, prolonged duration, and frequent use of NSAIDs was
associated with an increased risk of CD and UC [66]. Antibiotics
probably influence the risk of developing disease through their effect
on the microbiome. Whether early life flora acquired during birth and
infancy is critical, or whether an individual at risk for IBD remains
susceptible to dynamic changes in flora associated with dietary patterns
or other lifestyle factors [67].
Stress is thought to play a role in the pathogenesis of CD and UC,
and to mediate disease flares [68-70]. Mood components of perceived
stress including depression and anxiety may play a role in mediating or
exacerbating disease related to stress [71]. A prospective study using
the Nurses’ Health Study found that both recent and remote depressive
symptoms were associated with increased risk of CD but not UC [72]. The
effect of recent depressive symptoms within 4 years of diagnosis was
more prominent than that of remote depression. There is limited
high-quality data on whether interventions to treat depression or stress
can modify its effect on disease.
One of the environmental triggers most commonly reported by patients,
but yet one where there is a significant gap in data is diet [73-74]. A
majority of prior studies have been limited by factors including
retrospective ascertainment of diet, allowing for both recall bias as
well as modifications in diet that may have occurred since the onset of
disease symptoms, and the small number of incident cases limiting power.
There are far fewer data examining the role of diet in triggering
disease flare. In a survey of 244 IBD patients in France, over half the
participants reported belief that diet played a role in disease relapse
[75]. However, the spectrum of foods that patients reported excluding to
prevent relapse was distributed among the different food groups,
suggesting that there may not be uniform dietary triggers to
relapses.
References
- Ananthakrishnan, A.N.; Bernstein, C.N.; Iliopoulos, D.; Macpherson,
A.; Neurath, M.F.; Ali, R.A.R.; Vavricka, S.R.; Fiocchi, C.
Environmental triggers in IBD: A review of progress and evidence. Nat.
Rev. Gastroenterol. Hepatol.2018, 15, 39–49. [CrossRef] [PubMed]
- Chang, J.T. Pathophysiology of Inflammatory Bowel Diseases. N. Engl.
J. Med. 2020, 383, 2652–2664. [CrossRef]
- Pullan, R.D.; Thomas, G.A.; Rhodes, M.; Newcombe, R.G.; Williams,
G.T.; Allen, A.; Rhodes, J. Thickness of adherent mucus gel on colonic
mucosa in humans and its relevance to colitis. Gut 1994, 35, 353–359.
[CrossRef]
- Parikh, K.; Antanaviciute, A.; Fawkner-Corbett, D.; Jagielowicz, M.;
Aulicino, A.; Lagerholm, C.; Davis, S.; Kinchen, J.; Chen, H.H.; Alham,
N.K.; et al. Colonic epithelial cell diversity in health and
inflammatory bowel disease. Nature 2019, 567, 49–55. [CrossRef]
- Kinchen, J.; Chen, H.H.; Parikh, K.; Antanaviciute, A.; Jagielowicz,
M.; Fawkner-Corbett, D.; Ashley, N.; Cubitt, L.; Mellado- Gomez, E.;
Attar, M.; et al. Structural Remodeling of the Human Colonic Mesenchyme
in Inflammatory Bowel Disease. Cell 2018, 175, 372–386.e17.
[CrossRef]
- Gustafsson, J.K.; Johansson, M.E.V. The role of goblet cells and
mucus in intestinal homeostasis. Nat. Rev. Gastroenterol. Hepatol. 2022,
19, 785–803. [CrossRef]
- Ghilas, S.; O’Keefe, R.; Mielke, L.A.; Raghu, D.; Buchert, M.;
Ernst, M. Crosstalk between epithelium, myeloid and innate lymphoid
cells during gut homeostasis and disease. Front. Immunol. 2022, 13,
944982. [CrossRef]
- Bassler, K.; Schulte-Schrepping, J.; Warnat-Herresthal, S.;
Aschenbrenner, A.C.; Schultze, J.L. The Myeloid Cell Compartment-Cell by
Cell. Annu. Rev. Immunol. 2019, 37, 269–293. [CrossRef]
- Herrero-Fernandez, B.; Gomez-Bris, R.; Somovilla-Crespo, B.;
Gonzalez-Granado, J.M. Immunobiology of Atherosclerosis: A Complex Net
of Interactions. Int. J. Mol. Sci. 2019, 20, 5293. [CrossRef]
- Zhou, G.; Yu, L.; Fang, L.; Yang, W.; Yu, T.; Miao, Y.; Chen, M.;
Wu, K.; Chen, F.; Cong, Y.; et al. CD177(+) neutrophils as functionally
activated neutrophils negatively regulate IBD. Gut 2018, 67,
1052–1063.
- Kang, L.; Fang, X.; Song, Y.H.; He, Z.X.; Wang, Z.J.; Wang, S.L.;
Li, Z.S.; Bai, Y. Neutrophil-Epithelial Crosstalk During Intestinal
Inflammation. Cell. Mol. Gastroenterol. Hepatol. 2022, 14,
1257–1267.
- Zigmond,E.;Jung,S.Intestinalmacrophages:Welleducatedexceptionsfromtherule.TrendsImmunol.2013,34,162
168
- Ruder,B.;Becker,C.AttheForefrontoftheMucosalBarrier:TheRoleofMacrophagesintheIntestine.Cells2020,9,2162.
- VallonEberhard,A.
;Landsman,L.;Yogev,N.;Verrier,B.;Jung,S.Transepithelial pathogen uptake
into the smallintestinal lamina propria. J. Immunol. 2006, 176,
2465–2469.
- Rescigno,M.;Urbano,M.;Valzasina,B.;Francolini,M.;Rotta,G.;Bonasio,R.;Granucci,F.;Kraehenbuhl,J.P.;Ricciardi-Castagnoli,
P. Dendritic cells express tight junction proteins and penetrate gut
epithelial monolayers to sample bacteria. Nat. Immunol. 2001, 2,
361–367
- Hapfelmeier,S.;Muller,A.J.;Stecher,B.;Kaiser,P.;Barthel,M.;Endt,K.;Eberhard,M.;Robbiani,R.;Jacobi,C.A.;Heikenwalder,
M.; et al. Microbe sampling by mucosal dendritic cells is a discrete,
MyD88-independent step in DeltainvG S. Typhimurium colitis. J. Exp. Med.
2008, 205, 437–450.
- Medina-Contreras,O.;Geem,D.;Laur,O.;Williams,I.R.;Lira,S.A.;Nusrat,A.;Parkos,C.A.;Denning,T.L.CX3CR1regulates
intestinal macrophage homeostasis, bacterial translocation, and
colitogenic Th17 responses in mice. J. Clin.Investig. 2011, 121,
4787–4795.
- Pabst, O.; Bernhardt, G. The puzzle of intestinal lamina propria
dendritic cells and macrophages. Eur. J. Immunol. 2010, 40,
2107–2111.
- Niess, J.H.; Brand, S.; Gu, X.; Landsman, L.; Jung, S.; McCormick,
B.A.; Vyas, J.M.; Boes, M.; Ploegh, H.L.; Fox, J.G.; et
al. CX3CR1-mediated dendritic cell access to the intestinal lumen and
bacterial clearance. Science 2005, 307, 254–258.
- Leppkes, M.; Neurath, M.F. Cytokines in inflammatory bowel
diseases—Update 2020. Pharmacol. Res. 2020, 158, 104835.
- Mann, E.A.; Saeed, S.A. Gastrointestinal infection as a trigger for
inflammatory bowel disease. Curr. Opin. Gastroenterol. 2012, 28,
24–29.
- Schroeder, B.O.; Birchenough, G.M.H.; Stahlman, M.; Arike, L.;
Johansson, M.E.V.; Hansson, G.C.; Backhed, F. Bifidobacteria or Fiber
Protects against Diet-Induced Microbiota-Mediated Colonic Mucus
Deterioration. Cell Host Microbe 2018, 23, 27–40.e7.
- Furey,T.S.;Sethupathy,P.;Sheikh,S.Z.Redefining the IBDs using genome
scale molecular phenotyping . Nat.Rev.Gastroenterol. Hepatol. 2019, 16,
296–311.
- Graham, D.B.; Xavier, R.J. Pathway paradigms revealed from the
genetics of inflammatory bowel disease. Nature 2020, 578, 527–539.
- DeLange,K.M.;Moutsianas,L.;Lee,J.C.;Lamb,C.A.;Luo,Y.;Kennedy,N.A.;Jostins,L.;Rice,D.L.;Gutierrez-Achury,J.;Ji,S.G.;
et al. Genome-wide association study implicates immune activation of
multiple integrin genes in inflammatorybowel disease. Nat. Genet. 2017,
49, 256–261.
- Loddo,I.;Romano,C.InflammatoryBowelDisease:Genetics,Epigenetics,andPathogenesis.Front.Immunol.2015,6,51.
- Jostins,L.;Ripke,S.;Weersma,R.K.;Duerr,R.H.;McGovern,D.P.;Hui,K.Y.;Lee,J.C.;Schumm,L.P.;Sharma,Y.;Anderson,C.A.;etal.
Host-microbe interactions have shaped the genetic architecture of
inflammatory bowel disease. Nature 2012, 491, 119–124.
- Mitsialis,V.;Wall,S.;Liu,P.;Ordovas-Montanes,J.;Parmet,T.;Vukovic,M.;Spencer,D.;Field,M.;McCourt,C.;Toothaker,J.;
et al. Single-Cell Analyses of Colon and Blood Reveal Distinct Immune
Cell Signatures of Ulcerative ColitisandCrohn’s Disease.
Gastroenterology 2020, 159, 591–608.e10.
- Martin,J.C.;Chang,C.;Boschetti,G.;Ungaro,R.;Giri,M.;Grout,J.A.;Gettler,K.;Chuang,L.S.;Nayar,S.;Greenstein,A.J.;etal.
Single-Cell Analysis of Crohn’s Disease Lesions Identifies a Pathogenic
Cellular Module Associated with Resistance to Anti-TNF Therapy. Cell
2019, 178, 1493–1508.e20.
- Corridoni,D.;Antanaviciute,A.;Gupta,T.;Fawkner Corbett,D.;
Aulicino,A. ;Jagielowicz, M.;Parikh,K.;Repapi,E.; Taylor, S.; Ishikawa,
D.; et al. Single-cell atlas of colonic CD8(+) T cells in ulcerative
colitis. Nat. Med. 2020, 26,1480–1490.
- Boland, B.S.; He, Z.; Tsai, M.S.; Olvera, J.G.; Omilusik, K.D.;
Duong, H.G.; Kim, E.S.; Limary, A.E.; Jin, W.; Milner, J.J.; et
al. Heterogeneity and clonal relationships of adaptive immune cells in
ulcerative colitis revealed by single-cell analyses. Sci. Immunol. 2020,
5, eabb4432.
- Diez-Obrero, V.; Moratalla-Navarro, F.; Ibanez-Sanz, G.; Guardiola,
J.; Rodriguez-Moranta, F.; Obon-Santacana, M.; Diez- Villanueva, A.;
Dampier, C.H.; Devall, M.; Carreras-Torres, R.; et
al. Transcriptome-Wide Association Study for Inflammatory Bowel Disease
Reveals Novel Candidate Susceptibility Genes in Specific Colon Subsites
and Tissue Categories. J. Crohn’s Colitis 2022, 16, 275–285.
- Turpin, W.; Goethel, A.; Bedrani, L.; Croitoru Mdcm, K. Determinants
of IBD Heritability: Genes, Bugs, and More. Inflamm. Bowel. Dis. 2018,
24, 1133–1148.
- Kaser, A.; Lee, A.H.; Franke, A.; Glickman, J.N.; Zeissig, S.; Tilg,
H.; Nieuwenhuis, E.E.; Higgins, D.E.; Schreiber, S.; Glimcher, L.H.; et
al. XBP1 links ER stress to intestinal inflammation and confers genetic
risk for human inflammatory bowel disease. Cell 2008, 134, 743–756.
- Khor, B.; Gardet, A.; Xavier, R.J. Genetics and pathogenesis of
inflammatory bowel disease. Nature 2011, 474,307–317.
- Zhang, T.; Ji, X.; Lu, G.; Zhang, F. The potential of Akkermansia
muciniphila in inflammatory bowel disease. Appl. Microbiol. Biotechnol.
2021, 105, 5785–5794.
- Zocco, M.A.; dal Verme, L.Z.; Cremonini, F.; Piscaglia, A.C.; Nista,
E.C.; Candelli, M.; Novi, M.; Rigante, D.; Cazzato, I.A.; Ojetti, V.; et
al. Efficacy of Lactobacillus GG in maintaining remission of ulcerative
colitis. Aliment. Pharm. 2006, 23, 1567–1574.
- Turroni, F.; Duranti, S.; Milani, C.; Lugli, G.A.; van Sinderen, D.;
Ventura, M. Bifidobacterium bifidum: A Key Member of the Early Human Gut
Microbiota. Microorganisms 2019, 7, 544.
- Elguezabal, N.; Chamorro, S.; Molina, E.; Garrido, J.M.; Izeta, A.;
Rodrigo, L.; Juste, R.A. Lactase persistence, NOD2 status and
Mycobacterium avium subsp. paratuberculosis infection associations to
Inflammatory Bowel Disease.Gut Pathog. 2012, 4, 6.
- Sibartie, S.; Scully, P.; Keohane, J.; O’Neill, S.; O’Mahony, J.;
O’Hanlon, D.; Kirwan, W.O.; O’Mahony, L.; Shanahan, F. Mycobacterium
avium subsp. Paratuberculosis (MAP) as a modifying factor in Crohn’s
disease. Inflamm. Bowel. Dis. 2010, 16, 296–304.
- Palmela, C.; Chevarin, C.; Xu, Z.; Torres, J.; Sevrin, G.; Hirten,
R.; Barnich, N.; Ng, S.C.; Colombel, J.F. Adherent invasive Escherichia
coli in inflammatory bowel disease. Gut 2018, 67, 574–587.
- Eun, C.S.; Mishima, Y.; Wohlgemuth, S.; Liu, B.; Bower, M.; Carroll,
I.M.; Sartor, R.B. Induction of bacterial antigen-specific colitis by a
simplified human microbiota consortium in gnotobiotic interleukin-10-/-
mice. Infect. Immun. 2014, 82, 2239–2246.
- Clooney, A.G.; Eckenberger, J.; Laserna-Mendieta, E.; Sexton, K.A.;
Bernstein, M.T.; Vagianos, K.; Sargent, M.; Ryan, F.J.; Moran, C.;
Sheehan, D.; et al. Ranking microbiome variance in inflammatory bowel
disease: A large longitudinal intercontinental study. Gut 2021, 70,
499–510.
- Miquel, S.; Martin, R.; Rossi, O.; Bermudez-Humaran, L.G.; Chatel,
J.M.; Sokol, H.; Thomas, M.; Wells, J.M.; Langella,P. Faecalibacterium
prausnitzii and human intestinal health. Curr. Opin. Microbiol. 2013,
16, 255–261.
- Oyri, S.F.; Muzes, G.; Sipos, F. Dysbiotic gut microbiome: A key
element of Crohn’s disease. Comp. Immunol.Microbiol. Infect. Dis. 2015,
43, 36–49.
- Cosnes J, Gower-Rousseau C, Seksik P, Cortot A. Epidemiology and
natural history of inflammatory bowel diseases. Gastroenterology. 2011;
140:1785–94.
- Danese S, Fiocchi C. Etiopathogenesis of inflammatory bowel
diseases. World J Gastroenterol. 2006; 12:4807–12.
- Lakatos PL. Environmental factors affecting inflammatory bowel
disease: have we made progress? Dig Dis. 2009;27:215–25.
- Williams CN. Does the incidence of IBD increase when persons move
from a low- to a high-risk area? Inflamm BowelDis. 2008; 14 (Suppl
2):S41–2.
- Thia KT, Loftus EV Jr, Sandborn WJ, Yang SK. An update on the
epidemiology of inflammatory bowel disease inAsia. Am J Gastroenterol.
2008; 103:3167–82.
- Cosnes J. Tobacco and IBD: relevance in the understanding of disease
mechanisms and clinical practice. Best Pract Res Clin Gastroenterol.
2004; 18:481–96.
- Cosnes J. What is the link between the use of tobacco and IBD?
Inflamm Bowel Dis. 2008; 14 (Suppl 2):S14–5.
- Lakatos PL, Szamosi T, Lakatos L. Smoking in inflammatory bowel
diseases: good, bad or ugly? World JGastroenterol. 2007; 13:6134–9.
- Odes HS, Fich A, Reif S, Halak A, Lavy A, Keter D, Eliakim R, Paz J,
Broide E, Niv Y, Ron Y, Villa Y, Arber N, Gilat T. Effects of current
cigarette smoking on clinical course of Crohn’s disease and ulcerative
colitis. Dig Dis Sci.2001; 46:1717–21.
- Higuchi LM, Khalili H, Chan AT, Richter JM, Bousvaros A, Fuchs CS. A
Prospective Study of Cigarette Smoking and the Risk of Inflammatory
Bowel Disease in Women. American Journal of Gastroenterology. 2012
- Birrenbach T, Bocker U. Inflammatory bowel disease and smoking: a
review of epidemiology, pathophysiology, andtherapeutic implications.
Inflamm Bowel Dis. 2004; 10:848–59.
- Cosnes J, Beaugerie L, Carbonnel F, Gendre JP. Smoking cessation and
the course of Crohn’s disease: an intervention study. Gastroenterology.
2001; 120:1093–9.
- Cosnes J, Carbonnel F, Beaugerie L, Le Quintrec Y, Gendre JP.
Effects of cigarette smoking on the long-term course of Crohn’s disease.
Gastroenterology. 1996; 110:424–31.
- Cantorna MT, Mahon BD. D-hormone and the immune system. J Rheumatol
Suppl. 2005; 76:11– 20.
- Cantorna MT, Mahon BD. Mounting evidence for vitamin D as an
environmental factor affecting autoimmune disease prevalence. Exp Biol
Med (Maywood). 2004; 229:1136–42.
- Cantorna MT, Zhu Y, Froicu M, Wittke A. Vitamin D status,
1,25-dihydroxyvitamin D3, and the immune system. AmJ Clin Nutr. 2004;
80:1717S–20S.
- Garg M, Lubel JS, Sparrow MP, Holt SG, Gibson PR. Review article:
vitamin D and inflammatory bowel disease -established concepts and
future directions. Aliment Pharmacol Ther. 2012; 36:324–44.
- Lim WC, Hanauer SB, Li YC. Mechanisms of disease: vitamin D and
inflammatory bowel disease. Nat Clin Pract Gastroenterol Hepatol. 2005;
2:308–15.
- Farraye FA, Nimitphong H, Stucchi A, Dendrinos K, Boulanger AB,
Vijjeswarapu A, Tanennbaum A, Biancuzzo R, Chen TC, Holick MF. Use of a
novel vitamin D bioavailability test demonstrates that vitamin D
absorption isdecreased in patients with quiescent Crohn’s disease.
Inflamm Bowel Dis. 2011; 17:2116–21.
- Leslie WD, Miller N, Rogala L, Bernstein CN. Vitamin D status and
bone density in recently diagnosed inflammatorybowel disease: the
Manitoba IBD Cohort Study. Am J Gastroenterol. 2008; 103:1451–9.
- Ananthakrishnan AN, Higuchi LM, Huang ES, Khalili H, Richter JM,
Fuchs CS, Chan AT. Aspirin, Nonsteroidal Anti-inflammatory Drug Use, and
Risk for Crohn Disease and Ulcerative Colitis: A Cohort Study. Ann
Intern Med. 2012;156:350–9.
- Shaw SY, Blanchard JF, Bernstein CN. Association between the use of
antibiotics and new diagnoses of Crohn’sdisease and ulcerative colitis.
Am J Gastroenterol. 2011; 106:2133–42.
- Maunder RG. Evidence that stress contributes to inflammatory bowel
disease: evaluation, synthesis, and futuredirections. Inflamm Bowel Dis.
2005; 11:600–8.
- Mawdsley JE, Rampton DS. Psychological stress in IBD: new insights
into pathogenic and therapeutic implications.Gut. 2005; 54:1481–91.
- Mawdsley JE, Rampton DS. The role of psychological stress in
inflammatory bowel disease. Neuroimmunomodulation. 2006; 13:327–36.
- Camara RJ, Schoepfer AM, Pittet V, Begre S, von Kanel R. Mood and
nonmood components of perceived stress and exacerbation of Crohn’s
disease. Inflamm Bowel Dis. 2011; 17:2358–65.
- Ananthakrishnan AN, Khalili H, Pan A, Higuchi LM, PSds, Richter JM,
Fuchs CS, Chan AT. Association Between Depressive Symptoms and Incidence
of Crohn’s Disease and Ulcerative Colitis—Results from the Nurses’
Health Study. Clin Gastroenterol Hepatol. 2012
- Chapman-Kiddell CA, Davies PS, Gillen L, Radford-Smith GL. Role of
diet in the development of inflammatory boweldisease. Inflamm Bowel Dis.
2010; 16:137–51.
- Hou JK, Abraham B, El-Serag H. Dietary intake and risk of developing
inflammatory bowel disease: a systematic review of the literature. Am J
Gastroenterol. 2011; 106:563–73. Comprehensive review of the role of
diet in thepathogenesis of inflammatory bowel diseases.
- Zallot C, Quilliot D, Chevaux JB, Peyrin-Biroulet C,
Gueant-Rodriguez RM, Freling E, Collet- Fenetrier B, Williet N, Ziegler
O, Bigard MA, Gueant JL, Peyrin-Biroulet L. Dietary beliefs and behavior
among inflammatory bowel disease patients. Inflamm Bowel Dis. 2012
Chapter 3: Clinical Presentation and Classification
CD and UC share several clinical features and overlapping treatment
strategies, yet each presents with distinct pathological and phenotypic
characteristics. This section provides an overview of their
classification, clinical presentation, and specific phenotypes, such as
upper gastrointestinal and perianal CD.
Classification of Crohn’s disease
CD is classified using the Montreal classification system, which
considers age at diagnosis, disease location, and behavior. This system
helps improve diagnostic precision and guides individualized management
strategies [1].
Age at diagnosis
- A1: ≤16 years
- A2: 17–40 years
- A3: >40 years
Location of disease
- L1: Terminal ileum
- L2: Colon
- L3: Ileocolon
- L4: Upper GI (may coexist with L1–L3)
Disease behavior
- B1: Non-stricturing, non-penetrating
- B2: Stricturing
- B3: Penetrating
- Perianal modifier (p)

Figure 1. The phenotype of Crohn’s disease in Saudi Arabia as per
Montreal Classification for Age (A), Behaviour (B), and Location (L)
(adapted from Aljebreen A. M. et al.[2]).
Clinical presentation of Crohn’s disease
Clinical evaluation has always been fundamental in diagnosing
diseases, and this holds especially true in IBD. CD presents with a wide
range of intestinal and extraintestinal symptoms, reflecting its complex
and systemic nature. Understanding the phenotypic classification helps
clinicians tailor treatment and improve patient outcomes. Early
recognition of these features remains critical to effective long-term
disease management.
Gastrointestinal symptoms
Abdominal pain and diarrhea
Abdominal pain, particularly in the lower right quadrant, is a common
early symptom (80-85%). It may be accompanied by bloating, flatulence,
and distension. Diarrhea is another hallmark symptom—often non-bloody
(65%), but can be bloody depending on disease location. Visible blood in
the stool is less frequent than in UC but not uncommon [3].
Oral manifestations
Recurrent aphthous ulcers (canker sores) may appear, though their
direct association with CD remains debated, as they are also common in
the general population. Other findings may include nodular swelling, a
cobblestone appearance in the oral mucosa, granulomatous ulcers, or
pyostomatitis vegetans. Medications used in CD, including sulfasalazine
and corticosteroids, can lead to oral side effects such as lichenoid
reactions. Oral fungal infections like candidiasis could develop due to
immunosuppressive therapy. Anemia-related signs such as pallor, angular
cheilitis, and glossitis could develope due to malabsorption.
Systemic Symptoms
Growth delay in children
Is a notable concern in pediatric CD. It may be the presenting sign,
particularly during puberty, with up to 30% of affected children showing
delayed growth.
Fever and weight loss
Fever, when present, usually indicates complications like abscesses.
Weight loss is common, especially in adults, due to decreased food
intake and malabsorption. Patients may avoid eating to reduce symptoms.
Extensive small bowel involvement may impair absorption of key
nutrients, exacerbating weight loss [4].
Extraintestinal manifestations
These include arthritis, erythema nodosum (tender red nodules) and
pyoderma gangrenosum (ulcerative lesions), uveitis, and episcleritis.
These will be covered in detail in Chapter 5.
Hepatobiliary
These include primary sclerosing cholangitis (less common in CD but
can occur), especially in overlap cases with UC.
Distinct phenotypes of Crohn’s disease
Upper gastrointestinal CD
Including the stomach, duodenum, or esophagus is less common but
clinically significant. Symptoms may include nausea, vomiting,
epigastric pain, and dysphagia. Diagnostic evaluation often requires
upper endoscopy, and treatment strategies may need to be adjusted to
address the unique challenges of upper gastrointestinal disease.
Although isolated gastric involvement is rare (occurring in less than
0.07% of cases), upper gastrointestinal symptoms can be found in 13–16%
of CD patients, usually following the onset of lower gastrointestinal
symptoms. It commonly affects younger, non-smoking patients, and more
frequently present with concomitant ileal involvement and a stenosing
behavior [5].
Perianal Crohn’s Disease
Perianal involvement is among the most complex and debilitating
manifestations of CD. Patients may present with pain, itching, or
discharge due to fistulas, fissures, abscesses, or skin tags.
Incontinence can also occur. Management often requires a
multidisciplinary approach combining medical therapy and surgical
interventions aimed at fistula healing and symptom control [6].
Classification of ulcerative colitis
UC is classified based on the anatomical extent of colonic
involvement, which guides both clinical decision-making and therapeutic
strategies
Age at diagnosis
- A1: ≤16 years
- A2: 17–40 years
- A3: >40 years
Disease extent
- E1 (Ulcerative Proctitis): Inflammation limited to the rectum
- E2 (Left-sided Colitis): Involvement distal to the splenic flexure
(rectum, sigmoid, and descending colon)
- E3 (Extensive Colitis / Pancolitis): Inflammation extending proximal
to the splenic flexure and potentially involving the entire colon
Disease severity
- S1: Clinical remission
- S2: Mild ulcerative colitis
- S3: Moderate ulcerative colitis
- S4: Severe ulcerative colitis

Figure 2. The phenotype of ulcerative colitis disease in Saudi Arabia
as per Montreal Classification for Age (A), Extent (E), and Severity (S)
(adapted from Alharbi O. et al.[7]).
Clinical presentation of ulcerative colitis
UC is a chronic inflammatory condition of the colon, characterized by
continuous mucosal inflammation that begins in the rectum and extends
proximally in a contiguous fashion. Unlike CD, UC is confined to the
colon and does not exhibit skip lesions or transmural inflammation. The
clinical presentation of UC depends on both the extent and severity of
inflammation, influencing gastrointestinal as well as extraintestinal
manifestations.
Chronic inflammation in UC may lead to long-term complications,
including iron deficiency anemia and an increased risk of colorectal
cancer. These risks emphasize the importance of routine surveillance
colonoscopy and proactive disease management.
Gastrointestinal Manifestations
Diarrhea and rectal bleeding
The hallmark symptom of UC is diarrhea, often accompanied by visible
blood, mucus, or pus in the stool. The frequency of bowel movements
varies, ranging from mild to debilitating. Rectal bleeding is a dominant
feature, present in approximately 90–95% of patients. The severity of
bleeding is proportional to the extent of mucosal involvement, and in
some cases, may lead to iron deficiency anemia [8].
Abdominal pain and cramping
Lower abdominal pain, typically cramping in nature, is common and
often coincides with bowel movements.
Urgency and tenesmus
Patients frequently report urgency and tenesmus (the persistent
sensation of needing to defecate despite an empty rectum).
Systemic manifestations
Fever and fatigue
During active disease flares, systemic inflammation may result in
low-grade fever and persistent fatigue, significantly impacting quality
of life.
Weight Loss
Ongoing diarrhea, anorexia, and the metabolic burden of inflammation
contribute to weight loss and, in some cases, malnutrition—particularly
in patients with extensive disease or frequent relapses [9].
Table 1. summary of the key important clinical and histological
difference between Crohn’s disease and ulcerative colitis
library(knitr)
library(kableExtra)
# Data from the provided table
comparison_data <- data.frame(
Feature = c("Location", "Distribution", "Depth of inflammation", "Rectal involvement",
"Ileal involvement", "Perianal Disease", "Fistulas and abscesses", "Strictures",
"Bleeding", "Diarrhea", "Abdominal Pain", "Weight Loss/Malnutrition", "Surgery Recurrence"),
`Crohns Disease` = c("Any part of the gastrointestinal tract (mouth to anus)", "Patchy, skip lesions",
"Transmural", "Often spared", "Common",
"Common (fistula, abscess)", "Common", "Common", "Less common, mild",
"Often non-bloody", "Common",
"Common", "High recurrence post-resection"),
`Ulcerative Colitis` = c("Colon and rectum only", "Continuous from rectum proximally",
"Mucosal and submucosal only", "Almost always involved", "Occasional 15% (backwash ileitis) not more than 10 cms",
"Rare", "Rare", "Rare", "Common, may be severe", "Typically bloody diarrhea",
"Cramps with urgency", "Less common",
"Low after ileal pouch-anal anastomosis ")
)
kable(comparison_data, align = 'c') %>%
kable_styling(bootstrap_options = c("striped", "hover", "condensed"),
full_width = FALSE, font_size = 12, position = "center") %>%
column_spec(1, bold = TRUE, background = "#f2f2f2", color = "#333333") %>%
row_spec(0, background = "#d3d3d3", color = "#000000", bold = TRUE) %>%
row_spec(1:nrow(comparison_data), extra_css = "border-bottom: 1px solid #dddddd;") %>%
scroll_box(width = "100%", height = "auto")
Feature
|
Crohns.Disease
|
Ulcerative.Colitis
|
Location
|
Any part of the gastrointestinal tract (mouth to anus)
|
Colon and rectum only
|
Distribution
|
Patchy, skip lesions
|
Continuous from rectum proximally
|
Depth of inflammation
|
Transmural
|
Mucosal and submucosal only
|
Rectal involvement
|
Often spared
|
Almost always involved
|
Ileal involvement
|
Common
|
Occasional 15% (backwash ileitis) not more than 10 cms
|
Perianal Disease
|
Common (fistula, abscess)
|
Rare
|
Fistulas and abscesses
|
Common
|
Rare
|
Strictures
|
Common
|
Rare
|
Bleeding
|
Less common, mild
|
Common, may be severe
|
Diarrhea
|
Often non-bloody
|
Typically bloody diarrhea
|
Abdominal Pain
|
Common
|
Cramps with urgency
|
Weight Loss/Malnutrition
|
Common
|
Less common
|
Surgery Recurrence
|
High recurrence post-resection
|
Low after ileal pouch-anal anastomosis
|
Table 2. Histological differences between Crohn’s disease and
Ulcerative Colitis:
library(knitr)
library(kableExtra)
# Data from the provided table
histopathology_data <- data.frame(
Feature = c("Discrete mucosal ulcers", "Mucosal edema", "Fissures", "Granulomas",
"Abnormal crypt architecture", "Architectural distortion", "Lymphoid aggregates",
"Paneth cell metaplasia"),
`Crohns Disease` = c("Common", "Common", "Present", "Often seen (non-caseating)",
"Minimal", "Focal", "Frequent", "Absent"),
`Ulcerative Colitis` = c("Absent (except in fulminant colitis)", "Usually absent",
"Rare", "Absent, except in crypt rupturing", "Frequent",
"Diffuse", "Rare", "Occasionally present")
)
kable(histopathology_data, align = 'c') %>%
kable_styling(bootstrap_options = c("striped", "hover", "condensed"),
full_width = FALSE, font_size = 12, position = "center") %>%
column_spec(1, bold = TRUE, background = "#f2f2f2", color = "#333333") %>%
row_spec(0, background = "#d3d3d3", color = "#000000", bold = TRUE) %>%
row_spec(1:nrow(histopathology_data), extra_css = "border-bottom: 1px solid #dddddd;") %>%
scroll_box(width = "100%", height = "auto")
Feature
|
Crohns.Disease
|
Ulcerative.Colitis
|
Discrete mucosal ulcers
|
Common
|
Absent (except in fulminant colitis)
|
Mucosal edema
|
Common
|
Usually absent
|
Fissures
|
Present
|
Rare
|
Granulomas
|
Often seen (non-caseating)
|
Absent, except in crypt rupturing
|
Abnormal crypt architecture
|
Minimal
|
Frequent
|
Architectural distortion
|
Focal
|
Diffuse
|
Lymphoid aggregates
|
Frequent
|
Rare
|
Paneth cell metaplasia
|
Absent
|
Occasionally present
|
References
- Satsangi J, Silverberg MS, Vermeire S, Colombel JF. The Montre al
classification of inflammatory bowel disease: Controversies, consensus,
and implications. Gut. 2006; 55: 749-753.
- Aljebreen AM, Alharbi OR, Azzam NA, Almalki AS, Alswat KA, Almadi
MA. Clinical epidemiology and phenotypic characteristics of Crohn’s
disease in the central region of Saudi Arabia. Saudi J Gastroenterol.
May-Jun 2014;20(3):162-9. doi:10.4103/1319-3767.132993
- Gomollón F, Dignass A, Annese V, Tilg H, Van Assche G, Lindsay JO,
et al. 3rd European Evidence-based Consensus on the Diagnosis and
Management of Crohn’s Disease 2016: Part 1: Diagnosis and Medical
Management. J Crohn’s Colitis. Oxford University Press; 2017 Jan 1;
11(1): 3–25.
- Gajendran M, Loganathan P, Catinella AP, Hashash JG. A com
prehensive review and update on Crohn’s disease. Disease-a Month. 2018;
64: 20-57.
- Pimentel AM, Rocha R, Santana GO. Crohn’s disease of esophagus,
stomach and duodenum. World J Gastrointest Pharmacol Ther 2019; 10(2):
35-49 [PMID: 30891327 DOI: 10.4292/wjgpt.v10.i2.35]
- Juncadella AC, Alame AM, Sands LR, Deshpande AR. Perianal Crohn’s
disease: a review. Postgrad Med. 2015 Apr;127(3):266-72.
- Alharbi OR, Azzam NA, Almalki AS, et al. Clinical epidemiology of
ulcerative colitis in Arabs based on the Montreal classification. World
J Gastroenterol. Dec 14 2014;20(46):17525-31. doi:10.3748/wjg.v20.i46.17525
- Magro F, Gionchetti P, Eliakim R, Ardizzone S, Armuzzi A,
Barreiro-de Acosta M, et al. Third European Evidence based Consensus on
Diagnosis and Management of Ulcerative Colitis. Part 1: Definitions,
Diagnosis, Extra-intestinal Manifestations, Pregnancy, Cancer
Surveillance, Surgery, and Ileo-anal Pouch Disorders. J Crohn’s Colitis.
Oxford University Press; 2017 Jun 1; 11(6): 649–70
- Vavricka SR, Rogler G, Gantenbein C, Spoerri M, Prinz Vavricka M,
Navarini AA, et al. Chronological Order of Appearance of Extraintestinal
Manifestations Relative to the Time of IBD Diagnosis in the Swiss
Inflammatory Bowel Disease Cohort. Inflamm Bowel Dis. 2015 Aug; 21(8):
1794–800.
Chapter 4: Incidental Terminal Ileitis
With the increasing use of colonoscopy for colorectal cancer (CRC)
screening and surveillance, incidentally diagnosed terminal ileitis
(IDTI) is being identified more frequently in otherwise asymptomatic
individuals. However, its true prevalence, clinical significance, and
long-term outcomes remain unclear. While IDTI can result from various
causes, including NSAID use or early Crohn’s disease (CD), there is no
clear consensus on how to manage these cases.
Prevalence and Long-Term Outcomes
Prevalence range from 0.04% to 6.77%, with significant variation in
diagnostic criteria [1]. A pooled prevalence analysis estimated IDTI in
about 0.7% of non-diagnostic colonoscopies. In some cohort studies the
diagnostic work-up for CD the diagnosis was based on varying
combinations of clinical, biomarkers, endoscopic, and radiological
findings, The long-term follow-up of IDTI patients (median 13–84 months)
suggests that most cases do not progress to overt CD. However, in some
cases, particularly when associated with additional symptoms or
persistent inflammation, progression to CD has been observed [2,3].
The clinical significance of IDTI remains to be determined. Although
IDTI can occur in the context of other aetiologies such as non-steroidal
anti-inflammatory drugs (NSAIDs) use and rheumatological diseases
[4,5]
# Install packages if needed
if (!require(ggplot2)) install.packages("ggplot2")
if (!require(ggforce)) install.packages("ggforce")
library(ggplot2)
library(ggforce)
# Sample data for progression outcomes
progression_data <- data.frame(
Outcome = c("Progressed to CD", "Persistent Lesions", "Resolved"),
Percentage = c(5, 30, 65) # Estimated percentages
)
# Create stacked bar chart
ggplot(progression_data, aes(x = "", y = Percentage, fill = Outcome)) +
geom_bar(stat = "identity", color = "#333333", size = 0.3) +
coord_polar("y", start = 0) + # Semi-donut style for attractiveness
geom_text(aes(label = paste0(Percentage, "%")), position = position_stack(vjust = 0.5),
color = "#333333", size = 4.5, fontface = "bold") +
scale_fill_manual(values = c("#FF6B6B", "#FFA07A", "#76C893")) + # Red, coral, green
labs(title = "Long-Term Outcomes of IDTI", fill = "Outcome") +
theme_void() +
theme(
plot.title = element_text(hjust = 0.5, size = 16, face = "bold", color = "#333333"),
legend.position = "right",
plot.background = element_rect(fill = "white", color = NA),
plot.margin = margin(15, 15, 15, 15)
)

The following is recommended:
A minority of patients (ranging from none to a few in each study)
progressed to CD.
Some patients had lesions that persisted but did not worsen,
while others had complete resolution.
No specific predictive factors for disease progression were
consistently identified.
Abdominal pain at the time of colonoscopy was associated with a
higher risk of progression in studies including both diagnostic and
non-diagnostic colonoscopies.
Current Understanding and Management
The lack of clear predictors of progression complicates
decision-making. Some cases of IDTI may represent early, pre-clinical
CD, while others are due to transient, non-specific inflammation. NSAID
use has been implicated in IDTI but was only reported in a minority of
patients in the studies reviewed.
Given this uncertainty, a cautious, stepwise approach is
recommended.
The proposed clinical pathway for IDTI management lies in steps as
the following
Step 1: Initial evaluation
- Review potential risk factors: NSAID use, smoking, recent
infection.
- Obtain baseline fecal calprotectin (FC) to assess intestinal
inflammation.
Step 2: Follow-up based on FC levels
A. If FC is normal:
- Likely transient ileitis.
- Avoid potential triggers (NSAIDs, infections) and reassess in 3–6
months.
- No further intervention needed unless symptoms develop.
B. If FC is elevated:
- Consider cross-sectional imaging (e.g., MRI enterography or CT
enterography) to assess for transmural inflammation.
- Recheck FC in 6 months.
Step 3: Imaging findings & ongoing
management
A. No evidence of transmural inflammation:
- Likely reversible etiology.
- Monitor FC periodically and reassess if symptoms arise.
B. Evidence of transmural inflammation or persistently elevated
FC:
- Consider early treatment for CD, even in the absence of
symptoms.
- Longitudinal monitoring with FC, clinical assessment, and imaging
may be necessary.
library(DiagrammeR)
library(htmltools)
# Create the Management Pathway for IDTI flowchart
grViz("
digraph IDTI_Management_Pathway {
# Graph settings
graph [layout = dot, rankdir = TB, nodesep = 0.5, ranksep = 0.8]
node [fontname = 'Helvetica', fontsize = 12]
edge [color = 'black', arrowhead = vee]
# Define nodes
start [label = 'Initial Evaluation\\nReview risk factors, baseline FC',
shape = oval, style = filled, fillcolor = 'lightblue']
decision1 [label = 'FC Result?',
shape = diamond, style = filled, fillcolor = 'lightcoral']
normal_fc [label = 'Normal FC',
shape = box, style = filled, fillcolor = 'palegreen']
elevated_fc [label = 'Elevated FC',
shape = box, style = filled, fillcolor = 'lightpink']
avoid_triggers [label = 'Avoid triggers',
shape = box, style = filled, fillcolor = 'lightyellow']
imaging [label = 'Imaging',
shape = box, style = filled, fillcolor = 'lightyellow']
reassess_3_6 [label = 'Reassess 3-6mo',
shape = box, style = filled, fillcolor = 'lightyellow']
recheck_fc [label = 'Recheck FC 6mo',
shape = box, style = filled, fillcolor = 'lightyellow']
decision2 [label = 'Imaging Result?',
shape = diamond, style = filled, fillcolor = 'lightcoral']
no_inflammation [label = 'No Transmural\\nInflammation',
shape = box, style = filled, fillcolor = 'palegreen']
inflammation [label = 'Transmural\\nInflammation',
shape = box, style = filled, fillcolor = 'lightpink']
monitor_fc [label = 'Monitor FC',
shape = box, style = filled, fillcolor = 'lightyellow']
early_treatment [label = 'Early CD treatment',
shape = box, style = filled, fillcolor = 'lightyellow']
reassess_symptoms [label = 'Reassess if symptoms',
shape = box, style = filled, fillcolor = 'lightyellow']
monitor [label = 'Monitor',
shape = box, style = filled, fillcolor = 'lightyellow']
# Define edges
start -> decision1
decision1 -> normal_fc
decision1 -> elevated_fc
normal_fc -> avoid_triggers
elevated_fc -> imaging
avoid_triggers -> reassess_3_6
imaging -> decision2
decision2 -> no_inflammation
decision2 -> inflammation
no_inflammation -> monitor_fc
inflammation -> early_treatment
monitor_fc -> reassess_symptoms
early_treatment -> monitor
# Additional connections
reassess_3_6 -> recheck_fc [constraint = false]
recheck_fc -> decision1 [constraint = false]
}
")
# Add some CSS styling for better appearance
tags$style(HTML("
.node oval {
fill: lightblue;
}
.node box {
fill: lightyellow;
}
.node diamond {
fill: lightcoral;
}
"))
While most cases of IDTI do not progress to CD, careful follow-up is
needed to identify those at risk. A systematic approach—including FC
monitoring, imaging, and evaluation of risk factors can help
differentiate between transient ileitis and early CD, ensuring
appropriate management while avoiding unnecessary interventions.
References
- M. Agrawal et al Journal of Crohn’s and Colitis, 2021, 1455–1463 doi:10.1093/ecco-jcc/jjab030
- Siddiki H, Lam-Himlin D, Pasha SF, Gurudu SR, Leighton JA. Terminal
ileitis of unknown significance: long-term follow-up and outcomes in a
single cohort of patients. Am J Gastroenterol 2015;110[Suppl
1]:S768–S9.
- Wang WF, Wang ZB, Yang YS, Linghu EQ, Lu ZS. Long-term follow-up of
nonspecific small bowel ulcers with a benign course and no requirement
for surgery: is this a distinct group? BMC Gastroenterol 2011;11:7
- Long MD, Kappelman MD, Martin CF, Chen W, Anton K, Sandler RS. Role
of nonsteroidal anti-inflammatory drugs in exacerbations of inflam
matory bowel disease. J Clin Gastroenterol 2016;50:152–6.
- Rodríguez-Lago I, Merino O, Azagra I, et al. Characteristics and pro
gression of preclinical inflammatory bowel disease. Clin Gastroenterol
Hepatol 2018;16:1459–66.
Chapter 6: Investigating a Patient Suspected of Having IBD
Diagnosing IBD can be challenging due to the complexity and wide
variation of its clinical features. The diagnosis of IBD doesn’t rely on
a single diagnostic test but instead on a constellation of clinical
features, laboratory abnormalities, radiological changes, endoscopic
observations, and histologic criteria [1]. The first challenge is
differentiating between IBD and acute gastrointestinal illnesses [2,
3].
Even though IBD typically presents chronically over months and
sometimes years, it is seldom that the presentation is subacute over
weeks. Therefore, it is essential to rule out infections that mimic IBD
(Table 1) by performing proper stool testing [4]. Patients with UC are
frequently diagnosed with recurrent dysentery (amoeba) and are
unnecessarily given repetitive courses of antibiotics. Another challenge
is to differentiate between IBD, mainly CD, and irritable bowel disease
(IBS), as a large proportion of patients with CD are initially
misdiagnosed as IBS for an extended period, which leads to delays in
diagnosis and treatment. These warrants paying careful attention to “red
flags” during history taking.
Table 1. Pathologic features of infections that can mimic
inflammatory bowel disease.
Salmonella enterica typhi & paratyphi serovars Shigella
spp. |
Lymphoid hyperplasia, ulcers, crypt architectural distortion
Continuous distribution proximally from the rectum, chronic active
colitis with marked architectural distortion |
Stool culture and PCR |
Entamoeba histolytica |
Cryptitis, ulcers, pyloric metaplasia, Paneth cell hyperplasia,
architectural distortion |
Trophozoites are positive with trichrome and PAS stains |
Sexually transmitted proctitis (Treponema pallidum, Chlamydia
trachomatis) |
Dense lymphohistiocytic infiltrate with prominent plasma cells,
lymphoid aggregates, mild actively cryptitis, poorly formed
granulomas |
Treponema pallidum: Immunohistochemistry, serologic studies
Chlamydia trachomatis: Nucleic acid amplification test or
PCR on rectal swab specimens |
Mycobacterium tuberculosis |
Hyperplastic Peyer patches, fissures, architectural distortion,
transmural lymphoid aggregates, mural fibrosis, inflamed submucosal
blood vessels, granulomata |
Acid-fast stains, RT-PCR on paraffin-embedded tissue or stool,
serologic gold test QuantiFERON |
Yersinia spp. |
Architectural distortion, mural lymphoid hyperplasia and fibrosis,
transmural lymphoid aggregates, and epithelioid granulomata |
Stool culture and RT-PCR on paraffin-embedded tissue |
Actinomyces spp. |
Mucosal lymphoid hyperplasia, transmural lymphoid aggregates and
epithelioid granulomata, perianal fibrosing |
Organisms are gram positive and stain with GMS |
Basidiobolus ranarum |
Increased lamina propria inflammation including plasma cells,
neutrophils, eosinophils, ulcers, granulomata, thickening of pericolic
fat |
GMS, PAS-D |
The red flag score is a partially validated tool that can be used
accurately to detect patients with a high probability of CD (Table 2)
[5,6]. Delays in diagnosing IBD can lead to the development of disease
complications and treatment resistance [7]. In certain parts of the
world where intestinal tuberculosis (ITB) is endemic, patients with CD
can be misdiagnosed with ITB and treated with anti-tuberculous
medications for periods that extend to 2 years before the diagnosis is
challenged. Predictive models incorporating clinical, biochemical, and
endoscopic findings have been introduced to help distinguish both
conditions and mitigate this obstacle [8].
Table 2. The Red Flag Index (RFI)
Non-healing or complex perianal fistula or abscess or perianal
lesions (apart from hemorrhoids) |
5 |
First-degree relative with confirmed inflammatory bowel disease |
4 |
Weight loss (5% of usual body weight) in the last 3 months |
3 |
Chronic abdominal pain (>3 months) |
3 |
Nocturnal diarrhoea |
3 |
Mild fever in the last 3 months |
2 |
No abdominal pain 30-45 min after meals, predominantly after
vegetables |
2 |
No rectal urgency |
2 |
A minimum Red Flags index value of 8 highly predicted CD
diagnoses with sensitivity and specificity bootstrap estimates of 0.94
(95% confidence interval 0.88-0.99) and 0.94 (0.90-0.97), respectively.
Positive and negative likelihood ratios were 15.1 (9.3-33.6) and 0.066
(0.013-0.125). The association between CD diagnosis and a Red Flags
index value of ≥8 corresponds to an OR of 290 (p < 0.0001).
Patients with IBD can have both local and systemic manifestations
with very high variability. This symptom heterogeneity can be attributed
mainly to disease location, extent, severity, and phenotype. The most
common gastrointestinal symptoms are diarrhea, abdominal pain, tenesmus,
and malnutrition. Presenting with dominant systemic symptoms such as
fever, weakness, fatigue, and extra intestinal manifestations of IBD is
not uncommon, especially in younger patients.
Symptoms of UC include bloody diarrhea, rectal bleeding, urgency,
tenesmus, and abdominal cramps. In a small percentage of patients, the
first presentation of UC can be explosive and require urgent
hospitalization due to the higher risk of perforation or exsanguination,
which is labeled acute severe ulcerative colitis (ASUC). These patients
typically present with very severe symptoms and systemic toxicity. Acute
infections should be carefully excluded in patients before initiating
rescue therapies, such as intravenous corticosteroids, infliximab, or
cyclosporin.
It is prudent to be able to distinguish between the two major types
of IBD, UC and CD, as this has significant implications for the
patient’s future disease course. This is, however, quite difficult
sometimes, which is why around 10% percent of patients with IBD are
initially labeled as IBD unclassified (IBDU) [9].
Patients’ CD typically presents with abdominal pain, diarrhea,
vomiting, weight loss, and fever. About a quarter of patients with CD
present with perianal manifestations such anal fissures, perianal
fistulae and abscesses, and skin tags, which can be very implicative of
the diagnosis [10]. Upper GI involvement of CD is commonly seen in young
patients and less so in adults [2,3].
How to investigate a patient who might have IBD?
Laboratory investigations
Baseline laboratory investigations
Baseline laboratory testing of patients presenting with symptoms
suspicious of IBD includes essential blood and stool testing.
Complete blood count (CBC) is beneficial in detecting anemia, which
could be caused by chronic inflammation, malabsorption, or bleeding;
leukocytosis, which could reflect active inflammation of superimposed
infection; and thrombocytosis, which reflects ongoing inflammation.
Additional blood tests include liver profile, electrolytes, renal
profile, thyroid function test, and c-reactive protein (CRP). CRP is a
non-specific inflammatory marker that supports the diagnosis of IBD when
elevated but with limited sensitivity, as 15-20% of patients do not
produce CRP.
Stool tests
These include stool analysis, stool culture, polymerase chain
reaction (PCR) assay for clostridium difficile toxin, and fecal
calprotectin (FC). FC is a protein released from neutrophils during
inflammation and detected in the stool. The level of FC correlates with
the degree of intestinal inflammation and disease location. FC is more
likely to be elevated in the presence of colonic involvement and less
likely if the disease is limited to the small bowel. Nevertheless, a
normal FC decreases the likelihood of IBD, and an elevated FC should
prompt further investigations such as ileocolonoscopy, cross-sectional
imaging, or video capsule endoscopy (VCE) to confirm or rule out IBD. FC
is widely used to screen for IBD with a sensitivity of 85.8% (95% CI:
78.3–91) and a specificity of 91.7% (95% CI: 84.5–95.7) to distinguish
between IBD and IBS. Issues that require careful attention when
interpreting FC levels include sample acquisition, sample processing,
cut-off points, and the pre-test probability of IBD [11].
Antibody tests
Several antibodies to microbial antigens that can predict the
diagnosis of IBD have been identified and studied. Anti-saccharomyces
antibodies (ASCA) and anti-neutrophil cytoplasmic antibodies (ANCA) are
seldom used to help differentiate between CD and UC, respectively. For
example, positive ASCA and negative pANCA tests can predict CD with a
sensitivity of 54.6% and a specificity of 92.8% (receiver operating
characteristic (ROC) curve (AUC)= 0.85, likelihood ratio positive (LR+)
= 6.5, likelihood ratio negative (LR-) = 0.5). On the other hand, the
sensitivity and specificity of a positive pANCA test alone for UC were
55.3% and 88.5%, respectively (AUC = 0.82; LR+ = 4.5, LR- = 0.5). Other
markers, namely anti-outer-membrane protein C (anti-OmpC),
anti-pseudomonas fluorescence-associated sequence I2 (anti-I2), and
anti-bacterial flagellin (anti-CBir1), can be used to predict the risk
of complications and surgery in CD patients but with limited accuracy
[12, 13].
Radiological investigations
Intestinal ultrasound
Intestinal ultrasound (IUS) is a non-invasive, radiation-free imaging
modality that has become increasingly useful in diagnosing IBD. It
detects bowel wall thickness and intramural vascularization and can help
detect strictures, abscesses, or fistulae. IUS has the advantage of
being point-of-care but is limited by being highly operator-dependent
and less accurate in obese patients [14, 15].
Computed tomography enterography (CTE)
Contrast (oral and IV contrasts) enhanced computed tomography (CT)
scans focused on the bowel are frequently used to assess the small bowel
for areas of inflammation, e.g., wall thickness, mesenteric engorgement,
and stenosis or fistulization due to complicated CD [16]. Although they
require a short duration to perform and provide comprehensive details of
the bowel and surrounding organs, their main disadvantage is radiation
exposure.
Magnetic resonance enterography (MRE)
MRE is a radiation-free, cross-sectional modality that provides a
high-quality, detailed description of the bowel and the surrounding
tissue. MRE’s primary role is to detect inflamed or damaged bowel areas.
For this purpose, oral and IV contrasts are needed. MRE is more accurate
than CTE in differentiating between active inflammation and fibrosis in
areas with luminal narrowing. Susceptibility to motion artifacts and
prolonged examination duration are among its main disadvantages
[17].
Table 3: Imaging modalities used to investigate for inflammatory
bowel disease.
Radiation Exposure |
Higher |
No radiation |
No radiation |
Resolution |
High (excellent for bowel wall assessment) |
High (excellent for soft tissue and bowel layers) |
Moderate (depends on operator and patient factors) |
Complications that can be detected |
Fistulas, abscesses, perforation, strictures |
Fistulas, abscesses, strictures, perianal disease |
Abscesses, fistulas, thickening, strictures |
Procedure Duration |
Fast (5-15 minutes) |
Longer (30-45 minutes) |
Fast (10-20 minutes) |
Cost |
Moderate to high |
High |
Low |
Availability |
Widely available |
Limited availability in some centers |
available. Operator dependent |
Endoscopy
Ileocolonoscopy
Documenting mucosal inflammation through endoscopy is considered the
cornerstone of diagnosing IBD. During the index evaluation,
documentation of the site and the extent, pattern, and severity of
inflammation is essential. Furthermore, identifying potential disease
complications is an integral part of risk stratification [18].
Esophagogastroduodenoscopy (EGD)
In adults, examination of the upper GI tract is usually reserved for
patients suspected of having upper GI CD, such as those with symptoms of
dyspepsia, nausea, vomiting, dysphagia, or epigastric pain. In contrast,
EGD is routinely done at baseline in children being worked up for CD due
to the higher prevalence of upper GI CD in this patient population
[19].
Video capsule endoscopy (VCE)
VCE is typically used when there is a high suspicion of isolated
small bowel CD, especially in the presence of a normal ileocolonoscopy
and a high index of suspicion. It is more accurate than cross-sectional
modalities (CTE or MRE) for detecting proximal small bowel aphthous
ulcers. However, due to the risk of capsule retention, it must be done
after ruling out small bowel strictures using cross-sectional imaging
[20.
Device-assisted enteroscopy (DAE)
DAE, such as push enteroscopy, anterograde, or retrograde balloon
enteroscopy, is used to reach small bowel lesions suspected of CD. This
is typically undertaken when tissue biopsy is needed following the
detection of inflamed areas by cross-sectional imaging or VCE [21].
Histopathology
Histopathology plays an essential role in confirming the diagnosis of
IBD. Features of chronicity and activity must be present to confirm the
diagnosis. Features of chronic inflammation include crypt architecture
distortion and inflammatory expansion of the lamina propria with basal
lymphoplasmacytosis and paneth cell metaplasia or hyperplasia. In CD,
other features include pyloric gland metaplasia of the small bowel and
right colon, and non-caseating granulomas, which are pathognomonic for
CD. Still, they are only seen in up to 25% of cases. Features of
activity include neutrophil infiltration in lamina propria, cryptitis,
crypt abscesses, and ulcerations [22].
Figure 1: Suggested diagnostic Algorithm for Crohn’s disease.
library(DiagrammeR)
library(htmltools)
# Create the final corrected Crohn's Disease Diagnostic Pathway flowchart
grViz("
digraph Crohns_Diagnostic_Pathway {
# Graph settings
graph [layout = dot, rankdir = TB, nodesep = 0.5, ranksep = 0.8]
node [fontname = 'Helvetica', fontsize = 12]
edge [color = 'black', arrowhead = vee]
# Define nodes
start [label = 'Clinical suspicion of\\nCrohn\\'s disease',
shape = oval, style = filled, fillcolor = 'lightblue']
ileocolonoscopy [label = 'Ileocolonoscopy and Bx',
shape = box, style = filled, fillcolor = 'lightyellow']
positive1 [label = '+ve for Crohn\\'s',
shape = diamond, style = filled, fillcolor = 'palegreen']
negative1 [label = '-ve for Crohn\\'s',
shape = diamond, style = filled, fillcolor = 'lightpink']
treat_crohns [label = 'Treat as Crohn\\'s',
shape = box, style = filled, fillcolor = 'lightyellow']
cte_mre [label = 'CTE/MRE',
shape = box, style = filled, fillcolor = 'lightyellow']
positive2 [label = '+ve',
shape = diamond, style = filled, fillcolor = 'palegreen']
negative2 [label = '-ve',
shape = diamond, style = filled, fillcolor = 'lightpink']
enteroscopy [label = 'Deep Enteroscopy & Biopsy',
shape = box, style = filled, fillcolor = 'lightyellow']
enteroscopy_positive [label = '+ve for Crohn\\'s',
shape = diamond, style = filled, fillcolor = 'palegreen']
enteroscopy_negative [label = '-ve for Crohn\\'s',
shape = diamond, style = filled, fillcolor = 'lightpink']
risk_stratify [label = 'Risk Stratify & Treat',
shape = box, style = filled, fillcolor = 'lightyellow']
treat_cause [label = 'Treat the Cause',
shape = box, style = filled, fillcolor = 'lightyellow']
capsule [label = 'Video Capsule Endoscopy',
shape = box, style = filled, fillcolor = 'lightyellow']
capsule_positive [label = '+ve',
shape = diamond, style = filled, fillcolor = 'palegreen']
capsule_negative [label = '-ve',
shape = diamond, style = filled, fillcolor = 'lightpink']
treat_ibs [label = 'Treat as IBS',
shape = box, style = filled, fillcolor = 'lightyellow']
# Define edges based on the corrected flow
start -> ileocolonoscopy
ileocolonoscopy -> positive1 [label = ' ']
ileocolonoscopy -> negative1 [label = ' ']
positive1 -> treat_crohns
negative1 -> cte_mre
cte_mre -> positive2 [label = ' ']
cte_mre -> negative2 [label = ' ']
positive2 -> enteroscopy
negative2 -> capsule
capsule -> capsule_positive [label = ' ']
capsule -> capsule_negative [label = ' ']
capsule_positive -> enteroscopy
capsule_negative -> treat_ibs
enteroscopy -> enteroscopy_positive [label = ' ']
enteroscopy -> enteroscopy_negative [label = ' ']
enteroscopy_positive -> risk_stratify
enteroscopy_negative -> treat_cause
}
")
# Add some CSS styling for better appearance
tags$style(HTML("
.node oval {
fill: lightblue;
}
.node box {
fill: lightyellow;
}
.node diamond {
fill: lightcoral;
}
"))
References
- Maaser C, Sturm A, Vavricka SR, Kucharzik T, Fiorino G, Annese V,
Calabrese E, Baumgart DC, Bettenworth D, Borralho Nunes P, Burisch J,
Castiglione F, Eliakim R, Ellul P, Gonzalez-Lama Y, Gordon H, Halligan
S, Katsanos K, Kopylov U, Kotze PG, Krustins E, Laghi A, Limdi JK,
Rieder F, Rimola J, Taylor SA, Tolan D, van Rheenen P, Verstockt B,
Stoker J, European Cs, Colitis O, the European Society of G, Abdominal
R. ECCO-ESGAR Guideline for Diagnostic Assessment in IBD Part 1: Initial
diagnosis, monitoring of known IBD, detection of complications. J Crohns
Colitis. 2019;13(2):144-64.
- Gomollon F, Dignass A, Annese V, Tilg H, Van Assche G, Lindsay JO,
Peyrin-Biroulet L, Cullen GJ, Daperno M, Kucharzik T, Rieder F, Almer S,
Armuzzi A, Harbord M, Langhorst J, Sans M, Chowers Y, Fiorino G,
Juillerat P, Mantzaris GJ, Rizzello F, Vavricka S, Gionchetti P, Ecco.
3rd European Evidence-based Consensus on the Diagnosis and Management of
Crohn’s Disease 2016: Part 1: Diagnosis and Medical Management. J Crohns
Colitis. 2017;11(1):3-25.
- Magro F, Gionchetti P, Eliakim R, Ardizzone S, Armuzzi A,
Barreiro-de Acosta M, Burisch J, Gecse KB, Hart AL, Hindryckx P, Langner
C, Limdi JK, Pellino G, Zagorowicz E, Raine T, Harbord M, Rieder F,
European Cs, Colitis O. Third European Evidence-based Consensus on
Diagnosis and Management of Ulcerative Colitis. Part 1: Definitions,
Diagnosis, Extra-intestinal Manifestations, Pregnancy, Cancer
Surveillance, Surgery, and Ileo-anal Pouch Disorders. J Crohns Colitis.
2017;11(6):649-70.
- Panarelli NC. Infectious Mimics of Inflammatory Bowel Disease. Mod
Pathol. 2023;36(7):100210.
- Danese S, Fiorino G, Mary JY, Lakatos PL, D’Haens G, Moja L, D’Hoore
A, Panes J, Reinisch W, Sandborn WJ, Travis SP, Vermeire S,
Peyrin-Biroulet L, Colombel JF. Development of Red Flags Index for Early
Referral of Adults with Symptoms and Signs Suggestive of Crohn’s
Disease: An IOIBD Initiative. J Crohns Colitis. 2015;9(8):601-6.
- Fiorino G, Bonovas S, Gilardi D, Di Sabatino A, Allocca M, Furfaro
F, Roda G, Lenti MV, Aronico N, Mengoli C, Angeli E, Gaffuri N,
Peyrin-Biroulet L, Danese S. Validation of the Red Flags Index for Early
Diagnosis of Crohn’s Disease: A Prospective Observational IG-IBD Study
Among General Practitioners. J Crohns Colitis. 2020;14(12):1777-9.
- Jayasooriya N, Baillie S, Blackwell J, Bottle A, Petersen I, Creese
H, Saxena S, Pollok RC, group P-Is. Systematic review with
meta-analysis: Time to diagnosis and the impact of delayed diagnosis on
clinical outcomes in inflammatory bowel disease. Aliment Pharmacol Ther.
2023;57(6):635-52.
- Choudhury A, Dhillon J, Sekar A, Gupta P, Singh H, Sharma V.
Differentiating gastrointestinal tuberculosis and Crohn’s disease- a
comprehensive review. BMC Gastroenterol. 2023;23(1):246.
- Prenzel F, Uhlig HH. Frequency of indeterminate colitis in children
and adults with IBD - a metaanalysis. J Crohns Colitis.
2009;3(4):277-81.
- Munster LJ, Monnink GLE, van Dieren S, Mundt MW, D’Haens G, Bemelman
WA, Buskens CJ, van der Bilt JDW. Fistulizing Perianal Disease as a
First Manifestation of Crohn’s Disease: A Systematic Review and
Meta-Analysis. J Clin Med. 2024;13(16).
- Dajti E, Frazzoni L, Iascone V, Secco M, Vestito A, Fuccio L, Eusebi
LH, Fusaroli P, Rizzello F, Calabrese C, Gionchetti P, Bazzoli F, Zagari
RM. Systematic review with meta-analysis: Diagnostic performance of
faecal calprotectin in distinguishing inflammatory bowel disease from
irritable bowel syndrome in adults. Aliment Pharmacol Ther.
2023;58(11-12):1120-31.
- Reese GE, Constantinides VA, Simillis C, Darzi AW, Orchard TR, Fazio
VW, Tekkis PP. Diagnostic precision of anti-Saccharomyces cerevisiae
antibodies and perinuclear antineutrophil cytoplasmic antibodies in
inflammatory bowel disease. Am J Gastroenterol.
2006;101(10):2410-22.
- Xiong Y, Wang GZ, Zhou JQ, Xia BQ, Wang XY, Jiang B. Serum
antibodies to microbial antigens for Crohn’s disease progression: a
meta-analysis. Eur J Gastroenterol Hepatol. 2014;26(7):733-42.
- Dal Buono A, Faita F, Armuzzi A, Jairath V, Peyrin-Biroulet L,
Danese S, Allocca M. Assessment of activity and severity of inflammatory
bowel disease in cross-sectional imaging techniques: a systematic
review. J Crohns Colitis. 2025;19(2).
- Malik S, Venugopalan S, Tenorio BG, Khan SR, Loganathan P,
Navaneethan U, Mohan BP. Diagnostic accuracy of bowel ultrasonography in
patients with inflammatory bowel disease: a systematic review and
meta-analysis. Ann Gastroenterol. 2024;37(1):54-63.
- Qiu Y, Mao R, Chen BL, Li XH, He Y, Zeng ZR, Li ZP, Chen MH.
Systematic review with meta-analysis: magnetic resonance enterography
vs. computed tomography enterography for evaluating disease activity in
small bowel Crohn’s disease. Aliment Pharmacol Ther.
2014;40(2):134-46.
- Yoon HM, Suh CH, Kim JR, Lee JS, Jung AY, Kim KM, Cho YA. Diagnostic
Performance of Magnetic Resonance Enterography for Detection of Active
Inflammation in Children and Adolescents With Inflammatory Bowel
Disease: A Systematic Review and Diagnostic Meta-analysis. JAMA Pediatr.
2017;171(12):1208-16.
- Spiceland CM, Lodhia N. Endoscopy in inflammatory bowel disease:
Role in diagnosis, management, and treatment. World J Gastroenterol.
2018;24(35):4014-20.
- Annunziata ML, Caviglia R, Papparella LG, Cicala M. Upper
gastrointestinal involvement of Crohn’s disease: a prospective study on
the role of upper endoscopy in the diagnostic work-up. Dig Dis Sci.
2012;57(6):1618-23.
- Kopylov U, Yung DE, Engel T, Vijayan S, Har-Noy O, Katz L, Oliva S,
Avni T, Battat R, Eliakim R, Ben-Horin S, Koulaouzidis A. Diagnostic
yield of capsule endoscopy versus magnetic resonance enterography and
small bowel contrast ultrasound in the evaluation of small bowel Crohn’s
disease: Systematic review and meta-analysis. Dig Liver Dis.
2017;49(8):854-63.
- Neuhaus H, Beyna T. Device-Assisted Enteroscopy in Inflammatory
Bowel Disease: From Balloon Enteroscopy to Motorized Spiral Enteroscopy.
Gastrointest Endosc Clin N Am. 2025;35(1):59-72.
- Magro F, Langner C, Driessen A, Ensari A, Geboes K, Mantzaris GJ,
Villanacci V, Becheanu G, Borralho Nunes P, Cathomas G, Fries W,
Jouret-Mourin A, Mescoli C, de Petris G, Rubio CA, Shepherd NA, Vieth M,
Eliakim R, European Society of P, European Cs, Colitis O. European
consensus on the histopathology of inflammatory bowel disease. J Crohns
Colitis. 2013;7(10):827-51.
- Satsangi J, Silverberg MS, Vermeire S, Colombel JF. The Montreal
classification of inflammatory bowel disease: controversies, consensus,
and implications. Gut. 2006;55(6):749-53.
Chapter 8: Treatment Endpoints and Medical Therapies
This chapter will summarize the principles of the treat-to-target
approach in IBD and provide an overview of available medical therapies,
including insights into treatment efficacy based on specific disease
phenotypes and severity.
Drug dosing, route of administration, key therapy considerations,
side effects, and monitoring protocols are explained in a following
Chapter.
Treat-to-target in moderate-to-sever IBD
Focusing solely on symptom resolution fails to alter the disease
course [1].
Achieving targets beyond symptom control, such as biomarker
normalization (C- reactive protein (CRP) and fecal calprotectin (FCP))
and endoscopic healing, improves patient long-term outcomes and can
modify the disease course [2].
The STRIDE 2 consensus provided a timeline-based treatment target
approach focusing on symptoms, biomarkers, and endoscopic outcomes
(Figure 1) [3].
Figure 1. Selecting therapeutic targets in IBD consensus (STRIDE-II)
[3]
library(DiagrammeR)
grViz("
digraph {
graph [layout = dot, rankdir = TB, fontname = Helvetica, nodesep = 0.5]
node [shape = box, style = 'rounded, filled', fillcolor = 'lightgrey', fontname = Helvetica, width = 3.5]
edge [fontname = Helvetica, fontsize = 10]
# Main pathway nodes
A [label = 'Active IBD\\nTreatment is chosen based on risk assessment', fillcolor = 'lightblue', shape = oval]
B [label = 'Short term targets\\nSymptomatic response\\nCD: ≥50% decrease in abdominal pain and stool frequency\\nUC: ≥50% decrease in rectal bleeding and stool frequency']
C [label = 'Intermediate targets\\nClinical remission\\nnormalized CRP and reduced FCP\\nCD: PRO2 (abdominal pain ≤1, stool frequency ≤3) or HBI <5\\nUC: PRO2 (rectal bleeding = 0, stool frequency = 0) or partial Mayo <3 (no score >1)\\nNormalization of CRP and decrease FCP (100–250 mg/g)']
D [label = 'Long term targets\\nEndoscopic healing\\nnormalized QoL\\nand absence of disability\\nCD: SES-CD <3 or no ulcerations\\nUC: Mayo endoscopic subscore = 0 or UCEIS ≤1']
E [label = 'To consider but not formal targets\\nCD: Transmural healing (MRE, CTE or IUS)\\nUC: Histological healing']
# Modification node
M1 [label = 'Modify the treatment if the target is not achieved', fillcolor = 'lightcoral']
# Edges for main pathway
A -> B -> C -> D -> E
# Modification edges
B -> M1 [color = red, fontcolor = red, label = 'Not achieved']
C -> M1 [color = red, fontcolor = red, label = 'Not achieved']
D -> M1 [color = red, fontcolor = red, label = 'Not achieved']
# Styling
{rank = same; B; C; D}
}
")
Treatment targets in IBD. IBD: inflammatory bowel disease; CD:
Crohn’s disease; UC: ulcerative colitis; PRO: patient-reported outcome;
CRP: C-reactive protein; FCP : Fecal calprotectin; QoL: quality of life;
SES-CD: Simple Endoscopic Score for Crohn’s disease; UCEIS: Ulcerative
Colitis Endoscopic Index of Severity; MRE: magnetic resonance
enterography; CTE: computed tomography enterography; IUS: Intestinal
ultrasound
Therapy should be adjusted or modified if treatment targets are
not met, with close monitoring being a critical component throughout the
course of management [3].
The time required to achieve each target varies depending on the
disease (UC or CD) and the selected therapy [3].
Transmural healing in CD and histologic remission in UC improve
outcomes (e.g., fewer hospitalizations, lower relapse rates) but are not
yet formal targets [3,4].
Less stringent, individualized targets may be appropriate for
frail patients, those with comorbidities, or refractory disease after
multiple therapy failures [5].
Therapeutic Options for Crohn’s Disease
Medical therapy of CD is tailored based on disease phenotype,
severity, and the burden of inflammation [6].
Early treatment with effective therapy (top-down approach) in
patient at risk of complications has demonstrated greater efficacy and
improved outcomes compared to the step-up approach (Figure 2)
[7].
library(DiagrammeR)
grViz("
digraph {
graph [layout = dot, rankdir = BT, nodesep = 0.0, ranksep = 0.0]
# Nodes
node [shape = box, style = filled]
A [label = '5 ASA (UC), Sulfasalazine', fillcolor = '#006600', width = 6.0, height = 0.8]
B [label = 'Corticosteroids', fillcolor = '#1B4F72', width = 5.0, height = 0.8]
C [label = 'IMM (AZA, MTX)', fillcolor = '#2E7D9A', width = 4.0, height = 0.8]
D [label = 'Biologics and small molecules', fillcolor = '#47A8B8', width = 3.0, height = 0.8]
E [label = 'Surgery', fillcolor = '#60C4D9', width = 2.0, height = 0.8]
# Edges to form pyramid structure
A -> B -> C -> D -> E [style = invis]
# Styling
A [fontcolor = 'white']
B [fontcolor = 'white']
C [fontcolor = 'white']
D [fontcolor = 'white']
E [fontcolor = 'white']
}
")
Top-down vs. step-up approach in CD . The top-down approach
demonstrates greater efficacy and fewer disease complications compared
to the step-up approach in Crohn’s disease. Surgery can be considered at
any time in the treatment algorithm. 5-ASA: 5-aminosalicylic acid ;AZA:
Azathioprine ;MTX: Methotrexate
A proposed treatment positioning and sequencing for Crohn’s
disease is outlined in Figure 4. Approved therapies for IBD patients,
including their efficacy in different scenarios, are detailed in Figure
6.
Management of stricturing and penetrating Crohn’s disease, beyond
medical therapy, is covered in a latter Chapter.
5-Aminosalicylates (5-ASA)
5-ASAs have no role in the management of CD, either for induction
or maintenance therapy [8].
Sulfasalazine may be considered for patients with mild colonic CD
[8].
Corticosteroids
Enteric-release budesonide is effective for inducing clinical
remission in mild to moderate CD limited to the ileum and/or ascending
colon [9].
Systemic corticosteroids (intravenous or oral) can be used to
induce remission in CD, but it is essential to ensure the patient has no
abscess or pelvic sepsis [10].
Steroids, in any form, should not be used for maintenance therapy
or to induce remission in perianal disease [11].
Immunomodulators
Thiopurines monotherapy are not used for induction but may be
effective for maintaining remission in CD. Their use should balance the
risk of side effects with the availability of safer therapies
[11].
Thiopurines are best used in combination with anti-TNF agents in
CD to enhance efficacy and reduce immunogenicity of anti-TNF therapy
(see in a latter Chapter) [12].
Unlike thiopurines, methotrexate can be used for both induction
and maintenance therapy in CD when administered via the parenteral route
[11].
Given the safety of newer therapies, methotrexate monotherapy is
rarely used and is best combined with anti-TNF agents [11].
Immunomodulators, when used in combination with anti-TNF therapy,
can be safely withdrawn after achieving long-term remission, with
caution for patients with prior anti-TNF immunogenicity [11].
TNFα antagonists
Infliximab, adalimumab, and certolizumab pegol can be used for
both induction and maintenance therapy in moderate to severe CD
[9].
Infliximab has shown greater efficacy and durability in CD when
combined with immunomodulators [13].
Anti-TNF agents, specifically infliximab, have shown efficacy in
patients with penetrating or perianal CD and should be considered
first-line therapy for these cases. Certolizumab not recommended as
first line therapy for perianal disease [11,14]
Anti-TNF agents have shown effectiveness in treating most
extraintestinal manifestations, such as peripheral and axial arthropathy
and pyoderma gangrenosum [15].
Secondary loss of response to anti-TNF therapy is common, often
due to antibody formation, with only one-third maintaining remission
after three years. Key risk factors are low drug levels at the end of
induction and lack of immunomodulator use [16].
Infliximab is effective as maintenance therapy in intravenous
(IV) and subcutaneous (SC) forms. Switching from IV to SC is safe, with
SC dosing based on prior IV dose and subsequent drug level after
transition (see Figure 3) [17]
Patients in clinical remission and with fecal calprotectin levels
<250 at the time of switching have a low risk of relapse after
transitioning to SC form [17].
5mg/kg/8weeks |
✔ |
|
|
10mg/kg/8weeks |
✔ |
|
Escalate to 240 mg eow |
10mg/kg/6weeks |
✔ |
|
|
10mg/kg/4weeks |
|
✔ |
|
Guidance for switching from IV maintenance Infliximab to SC form
based on the Remiswitch study [17]. eow: every other week; IFX:
Infliximab.
IL-12/IL-23 inhibitor (Anti IL12/23)
Ustekinumab, an anti-p40 subunit agent, inhibits IL-12 and IL-23
and is effective for both induction and maintenance therapy in moderate
to severe CD [11].
In head-to-head trial (SEAVUE), both ustekinumab and adalimumab
have been shown to be equally effective in biologic-naive
moderate-to-sever CD patients [18].
Adding immunomodulators to ustekinumab does not provide
additional benefit and can compromise the favorable safety profile of
the drug [19].
IL23 inhibitors (Anti IL23)
Risankizumab, guselkumab and mirikizumab, anti-p19 agents that
selectively inhibit IL-23, are approved for moderate to severe CD
[20-22].
In a head-to-head trial (SEQUENCE), risankizumab was noninferior
to ustekinumab for clinical remission and superior for endoscopic
remission in moderate-to-sever CD patients with prior anti-TNF exposure
[20].
In the GALAXI trial, guselkumab outperformed ustekinumab in
clinical and endoscopic remission in bio-naïve and bio-exposed
moderate-to-severe CD [21].
In the VIVID trial, mirikizumab was non-inferior to ustekinumab
for clinical remission but did not show superiority in endoscopic
response in moderate-to-severe CD [22].
Anti-integrin therapy
Vedolizumab, an anti-integrin agent that inhibits α4β7 integrin,
is effective for both induction and maintenance of remission in CD
[11].
Vedolizumab is available in IV form for induction and in both IV
and SC forms for maintenance therapy [11].
Janus kinase (JAK) inhibitors
Upadacitinib, a JAK-1 selective oral inhibitor, is the only JAK
inhibitor recommended for induction and maintenance in Crohn’s disease
[11].
It functions with relatively higher selectivity for JAK-1
inhibition [11].
JAK inhibitors should be used with caution in patients with a
history of or at risk for venous thromboembolism (VTE), major adverse
cardiovascular events (MACE), and malignancy.
Additionally, inactivated herpes zoster vaccine (Shingrix®)
should be administered prior to starting therapy [23].
Therapeutic positioning and sequencing in moderate to severe Crohn’s
disease
Early initiation of highly effective therapy is crucial in moderate
to severe Crohn’s disease. The choice of therapy depends on factors such
as disease phenotype, the presence or absence of extraintestinal
manifestations (EIMs), prior treatment exposure, and comorbidities. The
following figure showing a proposed potential therapeutic positioning
and sequencing of medications.
A proposed approach for the positioning and sequencing of therapy
in Crohn’s disease. Note: This is primarily based on the authors’
opinions, supported by available evidence and clinical experience, and
treatment decisions should be made on a case-by-case basis
Bio-naïve and inflammatory phenotype |
Preferred second line |
Preferred as first line |
Preferred as first line |
Preferred as first line |
Preferred second line |
Fistulizing/perianal disease |
Preferred as first line ** |
No efficacy/insufficient data |
No efficacy/insufficient data |
No efficacy/insufficient data |
Preferred second line |
Anti-TNF failure |
|
Preferred second line |
Preferred as first line |
Preferred second line |
Preferred as first line*** |
Anti-IL12/23 failure |
Preferred as first line |
|
Preferred as first line |
Preferred as first line |
Preferred as first line |
Safety concerns**** |
Preferred second line |
Preferred as first line |
Preferred as first line |
Preferred as first line |
Preferred second line |
- Anti-TNF therapy is most effective when combined with
immunomodulators.
** Infliximab combination therapy with an IMM is the preferred
choice.
*** Preferred with fistulizing disease.
**** Patients over 65, frail, or with severe comorbidities.
Therapeutic Options for Ulcerative Colitis
Management of UC depends on disease severity, extent of colonic
involvement, and the presence of complications [24].
Acute severe ulcerative colitis (ASUC) is considered an emergency
and should be managed promptly in an inpatient setting in collaboration
with colorectal surgery. Management of ASUC is detailed in a latter
Chapter.
Management of pouchitis is discussed in a latter
Chapter.
A proposed treatment positioning and sequencing for UC is
outlined in Figure 5. Approved therapies for IBD patients, including
their efficacy in different scenarios, are detailed in Figure
6.
5-Aminosalicylates (5-ASA)
For mild to moderate UC, 5-ASA (in oral or topical form) is
effective for both induction and maintenance therapy [24].
A starting dose of at least 2 g/day is recommended for mild
disease, while up to 4.8 g/day may be used for moderate disease, with no
efficacy difference between divided and once-daily 5-ASA dosing. Better
adherence is achieved with once daily dosing and is recommended
[24].
Topical 5-ASA alone is effective for induction and maintenance in
mild to moderate distal UC. For proctosigmoiditis and beyond, combined
oral and topical 5-ASA is preferred over monotherapy for induction
[25].
5-ASA can may be stopped when treatment escalation to advanced
therapy is needed, offering no added benefit [26].
5-ASA is very safe, but creatinine should be checked biannually
to monitor for potential interstitial nephritis [27].
Corticosteroids
Topical steroids can be used for induction of remission in
patients with active distal UC, although some studies suggest the
superiority of topical 5-ASA [28].
Colonic-release corticosteroid (Budesonide MMX®) is effective for
inducing remission in mild to moderate UC [24].
Systemic steroids (oral for outpatient and IV for inpatient) are
used to induce remission in patients with moderate to severe UC
[24].
Steroids, in any form, should not be used for maintenance therapy
[11].
Immunomodulators
Thiopurine monotherapy is not used for induction but may be
effective for maintaining remission in UC. Its use should balance the
risk of side effects with the availability of safer therapies
[24].
Thiopurines are best used in combination with anti-TNF agents
(specifically infliximab) in UC [29].
Unlike in CD, methotrexate is ineffective for induction or
maintenance in UC. Its use is limited to combination with anti-TNF
agents to reduce immunogenicity or as concomitant therapy for coexisting
immune-mediated diseases [24].
Immunomodulators, when used in combination with anti-TNF therapy,
can be withdrawn after achieving long-term remission, with caution for
patients with prior anti-TNF immunogenicity [11].
TNFα antagonists
Infliximab, adalimumab, and golimumab are effective for both
induction and maintenance of remission in moderate to severe UC
[24].
Infliximab is approved for use as rescue therapy in ASUC, as is
cyclosporine (see in a latter Chapter) [30].
Anti-TNF agents have shown effectiveness in treating most
extraintestinal manifestations, such as peripheral and axial arthropathy
and pyoderma gangrenosum [15].
Secondary loss of response to anti-TNF therapy is common in UC.
Patients with low drug levels after induction, high inflammatory burden,
low albumin levels, and lack of immunomodulator use are at higher risk
of immunogenicity [31].
Both IV and SC forms of infliximab are effective for maintenance
therapy following IV induction in UC. Transitioning from IV to SC during
maintenance is also safe (see ‘Anti-TNF in Treatment of Crohn’s Disease’
and Figure 3) [17].
IL-12/IL-23 inhibitor (Anti IL12/23)
Ustekinumab, an anti-p40 subunit agent, inhibits IL-12 and IL-23
and is effective for both induction and maintenance therapy in moderate
to severe UC [24].
Adding immunomodulators to ustekinumab does not provide
significant additional benefit [19].
IL23 inhibitors (Anti IL23)
- Risankizumab, mirikizumab, and guselkumab, anti-p19 agents that
selectively inhibit IL-23, are approved for moderate to severe UC
[32-34].
Anti-integrin therapy
Vedolizumab, an anti-integrin agent targeting α4β7 integrin, is
effective for both induction and maintenance of remission in UC,
available in both IV and SC forms [35].
Vedolizumab demonstrated superiority over adalimumab in a
head-to-head trial and is preferred for use over adalimumab (VARSITY)
[36].
Janus kinase (JAK) inhibitors
Tofacitinib (non-selective JAKi), upadacitinib (high selectivity
for JAK1 inhibition), and filgotinib are approved for moderate to severe
UC [37-39].
JAK inhibitors should be used with caution in patients with a
history of or at risk for venous thromboembolism (VTE), major adverse
cardiovascular events (MACE), and malignancy. Additionally, inactivated
herpes zoster vaccine (Shingrix®) should be administered prior to
starting therapy [23].
Multiple studies have shown that tofacitinib and upadacitinib are
effective in patients with ASUC, particularly in cases of prior
infliximab exposure, although they are not formally approved for this
indication [40,41].
Sphingosine-1-phosphate (S1P) receptor
modulators
S1P receptor modulators, ozanimod and etrasimod, are approved for
moderate-to-severe UC [26].
Ozanimod selectively binds to S1P receptors 1 and 5, whereas
etrasimod targets S1P receptors 1, 4, and 5 [26].
Proper evaluation is required before initiating S1P receptor
modulators in patients with heart block, arrhythmia, or macular edema;
baseline electrocardiogram (ECG) and ophthalmologic assessments are
recommended [42].
Therapeutic positioning and sequencing in UC
Choosing therapy for UC depends on disease severity, extent of
colonic involvement, presence of EIMs, prior treatment exposure, and
comorbidities. The following figure outlines a proposed therapeutic
positioning and sequencing of available therapies.
A proposed approach for the positioning and sequencing of therapy
in UC. Note: This is primarily based on the authors’ opinions, supported
by available evidence and clinical experience, and treatment decisions
should be made on a case-by-case basis
Mild to moderate disease |
Preferred as first line |
Preferred second line |
Preferred second line |
Preferred second line |
Preferred second line |
Preferred second line |
Preferred second line |
Moderate to severe disease |
Preferred as first line |
Preferred as first line |
Preferred as first line |
Preferred as first line |
Preferred as first line |
Preferred as first line |
Preferred as first line |
ASUC |
No efficacy/insufficient data |
Preferred as first line*** |
No efficacy/insufficient data |
No efficacy/insufficient data |
No efficacy/insufficient data |
Preferred second line**** |
No efficacy/insufficient data |
Anti-TNF failure |
No efficacy/insufficient data |
|
Preferred as first line |
Preferred as first line |
Preferred second line |
Preferred as first line |
Preferred second line |
Anti-IL12/23 failure |
No efficacy/insufficient data |
Preferred as first line |
|
Preferred as first line |
No efficacy/insufficient data |
Preferred as first line |
No efficacy/insufficient data |
Safety concerns***** |
Preferred as first line |
Preferred second line |
Preferred as first line |
Preferred as first line |
Preferred as first line |
Preferred second line |
Preferred second line |
- Oral and topical forms ** Adalimumab has low efficacy in UC and is
not preferred *** Infliximab is the only anti-TNF agent used in ASUC
**** Can be used if prior exposure to infliximab ***** Patients over 65,
frail, or with severe comorbidities.
Oral mesalamine |
✓ |
✓ |
✗ |
✓ |
✗ |
✗ |
✓ |
Topical mesalamine |
✓ |
◯ |
✗ |
✓ |
✗ |
✗ |
✓ |
Systemic corticosteroids |
✓ |
✗ |
✓** |
✓ |
◯** |
◯** |
◯** |
Colonic-release corticosteroids |
✓ |
✗ |
✗ |
✓ |
✗ |
✗ |
✓ |
Ileal release corticosteroids |
✓ |
✗ |
✓ |
✗ |
✗ |
✗ |
✓ |
Thiopurines monotherapy |
✗ |
✓ |
✓ |
✓ |
✗ |
✗ |
✓*** |
Methotrexate monotherapy |
✓ |
✓ |
✓ |
✗ |
✓ |
✗ |
✗ |
Infliximab |
✓ |
✓ |
✓ |
✓ |
✓ |
✓ |
✓ |
Adalimumab |
✓ |
✓ |
✓ |
✓ |
✓ |
✓ |
✓ |
Certolizumab |
✓ |
✓ |
✓ |
✗ |
✓ |
✓ |
✓ |
Golimumab |
✓ |
✓ |
✗ |
✓ |
✓ |
✓ |
✓ |
Vedolizumab |
✓ |
✓ |
✓ |
✓ |
✗ |
✗ |
✓ |
Ustekinumab |
✓ |
✓ |
✓ |
✓ |
◯ |
✗ |
✓ |
Risankizumab |
✓ |
✓ |
✓ |
✓ |
◯ |
✗ |
? |
Guselkumab |
✓ |
✓ |
✓ |
✓ |
◯ |
✗ |
? |
Mirikizumab |
✓ |
✓ |
✓ |
✓ |
◯ |
✗ |
? |
Tofacitinib |
✓ |
✓ |
✗ |
✓ |
✓ |
✓ |
✗ |
Filgotinib**** |
✓ |
✓ |
✗ |
✓ |
✓ |
? |
✗ |
Upadacitinib |
✓ |
✓ |
✓ |
✓ |
✓ |
✓ |
✗ |
Ozanimod |
✓ |
✓ |
✗ |
✓ |
✗ |
✗ |
✗ |
Etrasimod |
✓ |
✓ |
✗ |
✓ |
✗ |
✗ |
✗ |
Legend: ✓ : Can be used ✗ : Avoid ◯ : Can be considered ? :
Insufficient data
Medical therapy in the management of IBD . This figure serves as
guidance and does not replace clinical decision-making
- Spondyloarthropathy indications
** Avoid long-term use in all scenarios
*** Thiopurines can be continued in pregnancy but not started as
monotherapy or for induction.
**** Approved by the EMA but not by the FDA.
References
- West J, Tan K, Devi J, Macrae F, Christensen B, Segal JP. Benefits
and challenges of treat-to-target in inflammatory bowel disease. J Clin
Med. 2023 Sep 29;12(19):6292. PMID: 37834936; PMCID: PMC10573216.
- Plevris N, Dignass A, Eslamparast T, et al. Disease monitoring in
inflammatory bowel disease: evolving principles and possibilities.
Gastroenterology. 2022 May;162(5):1456-1475.e9.
- Turner D, Ricciuto A, Lewis A, D’Amico F, Dhaliwal J, Griffiths AM,
et al. STRIDE-II: An update on the Selecting Therapeutic Targets in
Inflammatory Bowel Disease (STRIDE) Initiative of the International
Organization for the Study of IBD (IOIBD): determining therapeutic goals
for treat-to-target strategies in IBD. Gastroenterology.
2021;160(5):1570-83.
- Shah SC, Colombel JF, Sands BE, Narula N. Systematic review with
meta-analysis: mucosal healing is associated with improved long-term
outcomes in Crohn’s disease. Aliment Pharmacol Ther.
2016;43(3):317-33.
- Raine T, Bonovas S, Burisch J, Kucharzik T, Adamina M, Annese V, et
al. ECCO Guidelines on Therapeutics in Ulcerative Colitis: Medical
Treatment. J Crohns Colitis. 2022;16(1):2-17.
- Torres J, Bonovas S, Doherty G, Kucharzik T, Gisbert JP, Raine T, et
al. ECCO Guidelines on Therapeutics in Crohn’s Disease: Medical
Treatment. J Crohns Colitis. 2020;14(1):4-22.
- Noor NM, Lee JC, Bond S, Dowling F, Brezina B, Patel KV, et al. A
biomarker-stratified comparison of top-down versus accelerated step-up
treatment strategies for patients with newly diagnosed Crohn’s disease
(PROFILE): a multicentre, open-label randomised controlled trial. Lancet
Gastroenterol Hepatol. 2024;9(5):415-27.
- Ford AC, Khan KJ, Sandborn WJ, Hanauer SB, Moayyedi P. Efficacy of
5-aminosalicylates in Crohn’s disease: systematic review and
meta-analysis. Am J Gastroenterol. 2011;106(4):617-29.
- Torres J, Bonovas S, Doherty G, Kucharzik T, Gisbert JP, Raine T, et
al. ECCO Guidelines on Therapeutics in Crohn’s Disease: Medical
Treatment. J Crohns Colitis. 2020;14(1):4-22.
- Lichtenstein GR, Loftus EV, Isaacs KL, Regueiro MD, Gerson LB, Sands
BE. ACG Clinical Guideline: Management of Crohn’s Disease in Adults. Am
J Gastroenterol. 2018;113(4):481-517.
- Gordon H, Minozzi S, Kopylov U, Verstockt B, Chaparro M, Buskens C,
et al. ECCO Guidelines on Therapeutics in Crohn’s Disease: Medical
Treatment. J Crohns Colitis. 2024;18(10):1531-55.
- Colombel JF, Sandborn WJ, Reinisch W, Mantzaris GJ, Kornbluth A,
Rachmilewitz D, et al. Infliximab, azathioprine, or combination therapy
for Crohn’s disease. N Engl J Med. 2010;362(15):1383-95.
- Colombel JF, Sandborn WJ, Reinisch W, Mantzaris GJ, Kornbluth A,
Rachmilewitz D, et al. Infliximab, azathioprine, or combination therapy
for Crohn’s disease. N Engl J Med. 2010;362(15):1383-95.
- Sands BE, Anderson FH, Bernstein CN, Chey WY, Feagan BG, Fedorak RN,
et al. Infliximab maintenance therapy for fistulizing Crohn’s disease. N
Engl J Med. 2004;350(9):876-85.
- Vavricka SR, Schoepfer A, Scharl M, Lakatos PL, Navarini A, Rogler
G. Extraintestinal manifestations of inflammatory bowel disease. Inflamm
Bowel Dis. 2015;21(8):1982-92.
- Kennedy NA, Heap GA, Green HD, Hamilton B, Bewshea C, Walker GJ, et
al. Predictors of anti-TNF treatment failure in anti-TNF-naive patients
with active luminal Crohn’s disease: a prospective, multicentre, cohort
study. Lancet Gastroenterol Hepatol. 2019;4(5):341-53.
- Bots SJ, Gecse KB, Barclay ML, et al. Switching from intravenous to
subcutaneous infliximab in patients with inflammatory bowel disease: the
REMISWITCH study. J Crohns Colitis. 2021;15(Suppl 1):S050-S051.
- Sands BE, Sandborn WJ, Panaccione R, O’Brien CD, Zhang H, Johanns J,
et al. Ustekinumab versus adalimumab for induction and maintenance
therapy in biologic-naive patients with moderately to severely active
Crohn’s disease: the SEAVUE study. Gastroenterology.
2021;161(1):S-001.
- Feagan BG, Sandborn WJ, Gasink C, Jacobstein D, Lang Y, Friedman JR,
et al. Ustekinumab as induction and maintenance therapy for Crohn’s
disease. N Engl J Med. 2016;375(20):1946-60.
- Peyrin-Biroulet L, Chapman JC, Colombel JF, Caprioli F, D’Haens G,
Ferrante M, et al. Risankizumab versus Ustekinumab for
Moderate-to-Severe Crohn’s Disease. N Engl J Med. 2024 Jul
18;391(3):241-252.
- Panaccione R, Ferrante M, Danese S, et al. Efficacy and safety of
guselkumab therapy in patients with moderately to severely active
Crohn’s disease: results of the GALAXI 2 & 3 phase 3 studies.
Gastroenterology. 2024;166(5):1057b2.
- Ferrante M, Tron E, Feagan BG, et al. Efficacy and safety of
mirikizumab in patients with moderately-to-severely active Crohn’s
disease: a phase 3, multicentre, randomised, double-blind,
placebo-controlled and active-controlled, treat-through study. The
Lancet. 2024;404(10470):2423-2436.
- Winthrop KL, Cohen SB. Oral surveillance and JAK inhibitor safety:
the theory of relativity. Nat Rev Rheumatol. 2022;18(5):277-86.
- Raine T, Bonovas S, Burisch J, Kucharzik T, Adamina M, Annese V, et
al. ECCO Guidelines on Therapeutics in Ulcerative Colitis: Medical
Treatment. J Crohns Colitis. 2022;16(1):2-17.
- Feagan BG, Macdonald JK. Oral 5-aminosalicylic acid for induction of
remission in ulcerative colitis. Cochrane Database Syst
Rev. 2012;10:CD000543.
- Singh S, Fumery M, Dulai PS, Jairath V, Sandborn WJ. AGA Clinical
Practice Guideline on Pharmacological Management of Moderate-to-Severe
Ulcerative Colitis [Internet]. Gastroenterology.
2024;167(7):1307-43.
- van Staa TP, Travis S, Leufkens HG, Logan RF. 5-aminosalicylic acids
and the risk of renal disease: a large British epidemiologic study.
Gastroenterology. 2004;126(7):17339.
- Cohen RD, Woseth DM, Thisted RA, Hanauer SB. A meta-analysis and
overview of the literature on treatment options for left-sided
ulcerative colitis and ulcerative proctitis. Am J Gastroenterol.
2000;95(5):1263-76.
- Panaccione R, Ghosh S, Middleton S, Márquez JR, Scott BB, Flint L,
et al. Combination therapy with infliximab and azathioprine is superior
to monotherapy with either agent in ulcerative colitis.
Gastroenterology. 2014;146(2):392400.
- Laharie D, Bourreille A, Branche J, Allez M, Bouhnik Y, Filippi J,
et al. Ciclosporin versus infliximab in patients with severe ulcerative
colitis refractory to intravenous steroids: a parallel, open-label
randomised controlled trial. Lancet. 2012;380(9857):1909-15.
- Papamichael K, Cheifetz AS, Melmed GY, Irving PM, Vande Casteele N,
Kozuch PL, et al. Appropriate therapeutic drug monitoring of biologic
agents for patients with inflammatory bowel diseases. Clin Gastroenterol
Hepatol. 2019;17(9):1655-68.
- Feagan BG, Sandborn WJ, Danese S, Wolf DC, Liu WJ, Hua SY, et
al. Risankizumab in patients with moderate to severe ulcerative colitis:
results from the INSPIRE trial. Gastroenterology.
2023;164(5):S-001.
- D’Haens G, Dubinsky M, Kobayashi T, Watanabe K, Saito K, Hibi T, et
al. Mirikizumab as induction and maintenance therapy for ulcerative
colitis: results from the phase 3 LUCENT trials. Gastroenterology.
2023;164(5):S-003.
- Sandborn WJ, Ferrante M, Bhandari BR, Berliba E, Feagan BG, Hibi T,
et al. Guselkumab for the treatment of ulcerative colitis: results from
the phase 2b QUASAR study. Gastroenterology. 2022;162(5):S-002.
- Peyrin-Biroulet L, Loftus EV, Colombel JF, Danese S, Sandborn WJ,
Sands BE, et al. Long-term efficacy and safety of vedolizumab
subcutaneous in patients with ulcerative colitis: results from the
VISIBLE 2 trial. Gastroenterology. 2020;158(5):S-001.
- Sands BE, Peyrin-Biroulet L, Loftus EV, Danese S, Colombel JF,
Törüner M, et al. Vedolizumab versus adalimumab for moderate-to-severe
ulcerative colitis. N Engl J Med. 2019;381(13):1215-26.
- Sandborn WJ, Su C, Sands BE, D’Haens GR, Vermeire S, Schreiber S, et
al. Tofacitinib as induction and maintenance therapy for ulcerative
colitis. N Engl J Med. 2017;376(18):1723-36.
- Danese S, Ferrante M, Feagan BG, Panés J, Sandborn WJ, Reinisch W,
et al. Upadacitinib as induction and maintenance therapy for moderate to
severe ulcerative colitis: results from the U-ACHIEVE and U-ACCOMPLISH
trials. Gastroenterology. 2023;164(5):S-004.
- Vermeire S, Schreiber S, Petryka R, Kuehbacher T, Hebuterne X,
Roblin X, et al. Filgotinib as induction and maintenance therapy for
ulcerative colitis: results from the SELECTION trial. Gastroenterology.
2021;160(5):S-005.
- Berinstein JA, Sheehan JL, Dias R, Baffy N, Osman M, Grinspan AM, et
al. Tofacitinib for biologic-experienced hospitalized patients with
acute severe ulcerative colitis: a retrospective case-control study.
Clin Gastroenterol Hepatol. 2021;19(10):2112-20.
- Honap S, Pavlidis P, Ray S, Sharma E, Hayee B, Powell N.
Upadacitinib as rescue therapy for acute severe ulcerative colitis: a
case series. J Crohns Colitis. 2023;17(3):S-012.
- Sandborn WJ, Feagan BG, D’Haens G, Wolf DC, Jovanovic I, Hanauer SB,
et al. Ozanimod as Induction and Maintenance Therapy for Ulcerative
Colitis. New England Journal of Medicine. 2021;385(14):1280-91
Chapter 9: Surgical Management of Inflammatory Bowel
Disease
Despite advances in medical therapies, surgery remains crucial for
managing complications and refractory Inflammatory bowel disease (IBD).
Approximately 30% of UC and 70% of CD patients require surgery during
their lifetime [1,2].
Surgery should not be seen as a last resort but as an integral part
of IBD management. The interplay between medical and surgical therapies
highlights the need for a coordinated approach, where biologics and
surgical interventions complement each other. Multidisciplinary care,
involving gastroenterologists and surgeons from diagnosis, ensures
optimal timing and selection of treatments.
Modern surgical advancements, including minimally invasive techniques
and perioperative biologic optimization, have improved outcomes and
reduced complications [3,4].
Indications for Surgical Intervention in ulcerative
colitis
Acute Severe UC (ASUC)
- Occurs in 15–25% of UC patients.
- 30–40% require colectomy after failing steroids/rescue therapy
[2,5].
- Delayed surgery increases mortality.
Toxic Megacolon
- Rare (1–3%) but life-threatening.
- colectomy is indicated if no improvement within 48–72 hours
[6].
Dysplasia/Cancer
- Risk escalates with disease duration [7]
– 2% at 10 years. – 8% at 20 years. – 18% at 30 years.
- Colectomy is definitive for high-grade dysplasia or cancer [8]
Chronic Refractory Disease
- 10–15% of UC patients require surgery due to persistent symptoms
despite maximal therapy [1].
- Chronic refractory UC (failed ≥3 drug classes) [4].
- Steroid dependence (>10mg prednisone for >6 months) [5].
- Growth impairment in pediatric UC [6].
Indications for Surgical Intervention in Crohn’s
disease
Unlike UC, surgery for CD is not curative and focuses on managing
complications, symptom relief, and bowel preservation [17]. Despite
advancements in medical therapy, approximately two-thirds of CD patients
still require surgery during their disease course [9]. A critical
concern is the cumulative risk of repeated resections, which may
precipitate short bowel syndrome [10].
This requires a conservative surgical approach, emphasizing
bowel-sparing techniques and reducing resection length [20,21]. Surgeons
should take a long-term view, considering the patient’s lifetime risk of
overall bowel loss when planning treatments [11,12].
The main surgical criteria of CD are:
Strictures
- 30–50% of CD patients develop obstructive strictures; managed via
resection or strictureplasty [2,8,13].
Fistulizing disease
- 20–30% of CD patients develop fistulas (enteric, perianal);
MRI-guided surgical repair is the gold standard [2,8,13].
Abscesses
- 10–20% incidence; initial drainage followed by surgery if refractory
[14].
Refractory to medical therapy inflammation
- Although most patients (80%) respond to medical therapy, about 20%
will have severe symptoms and a significantly impaired quality of life,
impeding the optimization of this option with surgical resection of the
inflamed tract [15]
Malignancy
- Small bowel adenocarcinoma risk increases with CD; resection is
curative [6,10].
Perianal CD (PACD)
PACD affects 30-50% of CD patients, frequently developing early in
the disease [16,17]. This causes chronic pain, discharge, and
incontinence, significantly impairing quality of life. PACD encompasses
diverse lesions including fissures, skin tags, strictures, fistulas
(simple or complex), and rarely anal cancer. Optimal management requires
accurate diagnosis using clinical assessment combined with pelvic MRI
(gold standard) and endoanal ultrasound for complete fistula evaluation
[38???????].
Surgical Procedures of IBD and Its Comparisons
Surgical approach depends on disease type (UC vs CD), location,
extent, comorbidities, and long-term outcomes [18,19,20].
UC: comprehensive surgical management
- Subtotal colectomy with ileostomyomy
Mostly in patients with ASUC or toxic megacolon who are not
responding to 72 hours maximal medical therapy, or Uncontrolled
hemorrhage [19,21,22].
The entire colon is removed with the rectum left in place. An
end-ileostomy is constructed to allow the patient to recover from the
acute inflammatory response. The procedure is potentially life-saving in
the setting of an emergency with a mortality rate of less than 5% in
high-volume centers [10]. Patients can eventually ideally undergo
completion proctectomy with an ileal pouch-anal anastomosis (IPAA) for
curative treatment [23,24].
Bridge procedure is a strategy for the critically ill; frequently
patients proceed to definitive surgery later [23].
- Restorative proctocolectomy with ileal pouch-anal
anastomosis (IPAA)
Elective procedure of choice for medically managed UC - typically
younger patients desiring to avoid a permanent stoma and maintain fecal
continence [23], such:
• Colorectal adenocarcinoma or high-grade dysplasia • Chronic
refractory UC (failed ≥3 drug classes) • Steroid dependence (>10mg
prednisone for >6 months) • Growth retardation in pediatric UC
Technical considerations
Pouch configuration:
J-pouch (most common): 15-20cm ileal loop, double-stapled
technique [25,26].
S-pouch: Reserved for patients with short mesentery
[27,28].
Kock pouch: Alternative for patients unsuitable for anal
anastomosis [29,30]
Robotic Approach (da Vinci Xi)
Key steps include complete mesocolic excision and rectal dissection
to pelvic floor using a 8mm camera trocar port at RUQ, three 8mm robotic
ports, and two 5mm assistant ports [31–33]. The leak rates are 3.2%
robotic vs. 8.1% for open surgery with an operative time 45 minutes
longer. It is thought that robotics don’t reduce leaks in experienced
hands, but may benefit trainees through tremor reduction [13,32]
Diverting ileostomy
It is recommended for malnourished (albumin <3g/dl), those on
high-dose steroids (>20mg prednisone), or incomplete doughnut rings
[34–36]. Long-term pouch survival is 93% at 10 years, 82% at 20 years
[37,38]. The leading causes of failure is chronic pouchitis (38%), and
anastomotic stricture (22%) [39–42].
The median daily bowel movements is 5 (range 3-10) [15, 23,36,37.
Nighttime incontinence is 28% at 5 years [43,44]. Sexual dysfunction
happens in 15% of males (erectile dysfunction), and 12% of females
(dyspareunia) [43–45].
Quality of Life Metrics like the Cleveland Global QoL score is 0.82
(vs 0.91 healthy controls) [46,47].
CD: comprehensive surgical management
The general objective of surgery in CD is to maintain intestinal
length and quality of life and to reduce the risk of recurrence. This is
highlighted by the high rate of recidivism following CD surgery, where
“endoscopic recidivism is seen in up to 70% and clinical recidivism of
30% within 5 years” after ICR [48–50]. This basic difference in
recurrence pattern constitutes a crucial aspect of CD surgical
treatment. It suggests even technically successful CD surgery is
frequently a transient surgical solution requiring chronic medical
therapy and close monitoring.
Is the most common surgery for CD; it is indicated for localized
terminal ileal disease with fibrostenotic (narrowing) or inflammatory
disease that is not medically responsive [10,51]. It is removal of a
terminal ileal segment and right colon with subsequent ileocolic
anastomosis to restore continuity [51–53].
Although it relieves symptoms in most patients, recurrence is a
significant problem, up to 70% have endoscopic recurrence and 30%
clinical recurrence within 5 years [54–56].
It must be limited to patients with localized disease who have had
relatively few previous resections to prevent the development of short
bowel syndrome from combined bowel loss [51,56,57].
- Laparoscopic Ileocecal Resection
• Patient Selection Criteria (10,51)
References
- Siegel CA, Sharma D, Griffith J, Doan Q, Xuan S, Malter L. Treatment
Pathways in Patients With Crohn’s Disease and Ulcerative Colitis:
Understanding the Road to Advanced Therapy. Crohns Colitis 360. 2024
Aug;6(3):otae040.
- Sulais E Al, AlAmeel T, Alenzi M, Shehab M, AlMutairdi A, Al-Bawardy
B. Colorectal Neoplasia in Inflammatory Bowel Disease. Cancers (Basel).
2025 Feb;17(4):665. doi: 10.3390/cancers17040665.
- Swaminathan A, Sparrow MP. Perianal Crohn’s disease: Still more
questions than answers. World J Gastroenterol. 2024
Oct;30(39):4260–6.
- Garcia-Olmo D, Gilaberte I, Binek M, Hoore AJLD, Lindner D, Selvaggi
F, et al. Follow-up Study to Evaluate the Long-term Safety and Efficacy
of Darvadstrocel (Mesenchymal Stem Cell Treatment) in Patients With
Perianal Fistulizing Crohn’s Disease: ADMIRE-CD Phase 3 Randomized
Controlled Trial. Dis Colon Rectum. 2022 May;65(5):713–20.
- Eaden JA, Abrams KR, Mayberry JF. The risk of colorectal cancer in
ulcerative colitis: a meta-analysis. Gut. 2001 Apr;48(4):526–35.
- Gomollón F, Dignass A, Annese V, Tilg H, Assche G Van, Lindsay JO,
et al. 3rd European Evidence-based Consensus on the Diagnosis and
Management of Crohn’s Disease 2016: Part 1: Diagnosis and Medical
Management. J Crohns Colitis. 2017 Jan;11(1):3–25.
- Turner D, Ruemmele FM, Orlanski-Meyer E, Griffiths AM, de Carpi JM,
Bronsky J, et al. Management of Paediatric Ulcerative Colitis, Part 1:
Ambulatory Care-An Evidence-based Guideline From European Crohn’s and
Colitis Organization and European Society of Paediatric
Gastroenterology, Hepatology and Nutrition. J Pediatr Gastroenterol
Nutr. 2018 Aug;67(2):257–91.
- Chang S, Murphy M, Malter L. A Review of Available Medical Therapies
to Treat Moderate-to-Severe Inflammatory Bowel Disease. Am J
Gastroenterol. 2024 Jan;119(1):55–80.
- Lichtenstein GR, Loftus E V., Isaacs KL, Regueiro MD, Gerson LB,
Sands BE. ACG Clinical Guideline: Management of Crohn’s Disease in
Adults. Vol. 113, American Journal of Gastroenterology. 2018.
- Bemelman WA, Warusavitarne J, Sampietro GM, Serclova Z, Zmora O,
Luglio G, et al. ECCO-ESCP consensus on surgery for Crohn’s disease. J
Crohns Colitis. 2018;12(1).
- Michelassi F, Block GE. Surgical management of Crohn’s disease. Adv
Surg. 1993;26:307–22.
- Michelassi F, Melis M, Rubin M, Hurst RD. Surgical treatment of
anorectal complications in Crohn’s disease. Surgery. 2000;128(4).
- Rottoli M, Cardelli S, Calini G, Alexa ID, Violante T, Poggioli G.
Outcomes of robotic surgery for inflammatory bowel disease using the
Medtronic HugoTM Robotic-Assisted Surgical platform: a single center
experience. Int J Colorectal Dis. 2024 Dec 1;39(1):158.
- E PJ   S. Skomorochow E, Pico J. Toxic Megacolon. [Updated
2023 Jul 4] [Internet]. StatPearls [Internet], Treasure Island (FL).
2025. Available from: https://www.ncbi.nlm.nih.gov/books/NBK547679/
- Hanauer SB, Sandborn WJ, Rutgeerts P, Fedorak RN, Lukas M, MacIntosh
D, et al. Human anti-tumor necrosis factor monoclonal antibody
(adalimumab) in Crohn’s disease: the CLASSIC-I trial. Gastroenterology.
2006 Feb;130(2):323–33; quiz 591.
- Schwartz DA, Ghazi LJ, Regueiro M, Fichera A, Zoccali M, Ong EMW, et
al. Guidelines for the multidisciplinary management of Crohn’s perianal
fistulas: summary statement. Inflamm Bowel Dis. 2015
Apr;21(4):723–30.
- Munster LJ, Meriba GR, Schuitema J, van Dieren S, de Groof EJ, Mundt
MW, et al. Early diagnosis of Crohn’s disease in patients presenting
with a perianal fistula: systematic review and development of a perianal
red flags index. Vol. 29, Techniques in Coloproctology. Springer Science
and Business Media Deutschland GmbH; 2025.
- Booth A, Ford W, Brennan E, Magwood G, Forster E, Curran T. Towards
Equitable Surgical Management of Inflammatory Bowel Disease: A
Systematic Review of Disparities in Surgery for Inflammatory Bowel
Disease. Inflamm Bowel Dis. 2022 Sep;28(9):1405–19.
- Spinelli A, Bonovas S, Burisch J, Kucharzik T, Adamina M, Annese V,
et al. ECCO Guidelines on Therapeutics in Ulcerative Colitis: Surgical
Treatment. J Crohns Colitis. 2022;16(2).
- Segal JP, Jean-Frédéric LeBlanc A, Hart AL. Ulcerative colitis: An
update. Clinical Medicine, Journal of the Royal College of Physicians of
London. 2021;21(2).
- Oo S, Davies M. Surgical management of ulcerative colitis and
Crohn’s disease. Vol. 41, Surgery (United Kingdom). 2023.
- Worley GHT, Vaughan-Shaw P, Sahnan K. Surgical management of
ulcerative colitis. Vol. 110, British Journal of Surgery. 2023.
- Prentice RE, Wright EK, Flanagan E, Kamm MA, Goldberg R, Ross AL, et
al. Evaluation and management of ileal pouch-anal anastamosis (IPAA)
complications in pregnancy, and the impacts of an IPAA on fertility. Eur
J Gastroenterol Hepatol. 2023 May;35(5):609–12.
- Horan J, Brannigan A, Mulsow J, Shields C, Cahill R. Ileal
pouch-anal anastomosis for ulcerative colitis: long-term outcomes and
trends over time in a low-volume institution. Ir J Med Sci.
2021;190(1).
- Wang Z. Colonic J-pouch versus side-to-end anastomosis for rectal
cancer: a systematic review and meta-analysis of randomized controlled
trials. BMC Surg. 2021;21(1).
- Okkabaz N, Haksal M, Atici AE, Altuntas YE, Gundogan E, Gezen FC, et
al. J-pouch vs. side-to-end anastomosis after hand-assisted laparoscopic
low anterior resection for rectal cancer: A prospective randomized trial
on short and long term outcomes including life quality and functional
results. International Journal of Surgery. 2017;47.
- Wu XR, Kirat HT, Kalady MF, Church JM. Restorative proctocolectomy
with a handsewn IPAA: S-pouch or J-pouch? Dis Colon Rectum.
2015;58(2).
- Mukewar S, Wu X, Lopez R, Shen B. Comparison of long-term outcomes
of S and J pouches and continent ileostomies in ulcerative colitis
patients with restorative proctocolectomy-experience in subspecialty
pouch center. J Crohns Colitis. 2014;8(10).
- Angistriotis A, Shen B, Kiran RP. Construction of and Conversion to
Continent Ileostomy: A Systematic Review. Dis Colon Rectum.
2022;65.
- Dörner J, Pantea R, Ecker KW, Möslein G. The continent ileostomy
(Kock pouch). Vol. 40, Coloproctology. 2018.
- Gul F, Kazmi SNH, Abbas K, Saeed S, Basit J. The future of robotic
surgery for inflammatory bowel diseases. Vol. 81, Annals of Medicine and
Surgery. 2022.
- Opoku D, Hart A, Thompson DT, Tran CG, Suraju MO, Chang J, et
al. Equivalency of short-term perioperative outcomes after open,
laparoscopic, and robotic ileal pouch anal anastomosis. Does procedure
complexity override operative approach? Surg Open Sci. 2022;9.
- Zaman S, Mohamedahmed AYYY, Abdelrahman W, Abdalla HE, Wuheb AA,
Issa MT, et al. Minimally invasive surgery for infammatory bowel
disease: A systematic review and meta-Analysis of robotic versus
laparoscopic surgical techniques. Vol. 18, Journal of Crohn’s and
Colitis. 2024.
- Exarchos G, Metaxa L, Gklavas A, Koutoulidis V, Papaconstantinou I.
Are radiologic pouchogram and pouchoscopy useful before ileostomy
closure in asymptomatic patients operated for ulcerative colitis? Eur
Radiol. 2019;29(4).
- Zittan E, Wong-Chong N, Ma GW, McLeod RS, Silverberg MS, Cohen Z.
Modified two-stage ileal pouch-anal anastomosis results in lower rate of
anastomotic leak compared with traditional two-stage surgery for
ulcerative colitis. J Crohns Colitis. 2016;10(7).
- Kuwahara R, Ikeuchi H, Bando T, Sasaki H, Goto Y, Horio Y, et
al. Clinical Results of One-stage Restorative Proctocolectomy with
J-pouch Anal Anastomosis in 300 Ulcerative Colitis Patients. J Anus
Rectum Colon. 2020;4(4).
- Tatsuta K, Sakata M, Iwaizumi M, Okamoto K, Yoshii S, Mori M, et
al. Long-term prognosis after stapled and hand-sewn ileal pouch–anal
anastomoses for familial adenomatous polyposis: a multicenter
retrospective study. Int J Colorectal Dis. 2024;39(1).
- Feinberg AE, Lavryk O, Aiello A, Hull TL, Steele SR, Stocchi L, et
al. Conditional survival after IPAA for ulcerative and indeterminate
colitis: Does long-term pouch survival improve or worsen with time? Dis
Colon Rectum. 2020;63(7).
- Holubar SD, Rajamanickam RK, Gorgun E, Lightner AL, Valente MA,
Church J, et al. Leaks from the Tip of the J-pouch: Diagnosis,
Management, and Long-term Pouch Survival. Dis Colon Rectum.
2023;66(1).
- Åkerlund JE, Löfberg R. Pouchitis. Vol. 20, Current Opinion in
Gastroenterology. 2004.
- Akiyama S, Rai V, Rubin DT. Pouchitis in inflammatory bowel disease:
a review of diagnosis, prognosis, and treatment. Intest Res.
2021;19(1).
- Hashavia E, Dotan I, Rabau M, Klausner JM, Halpern Z, Tulchinsky H.
Risk factors for chronic pouchitis after ileal pouch-anal anastomosis: A
prospective cohort study. Colorectal Disease. 2012;14(11).
- Lightner AL, Alsughayer A, Wang Z, McKenna NP, Seisa MO, Moir C.
Short- And long-term outcomes after ileal pouch anal anastomosis in
pediatric patients: A systematic review. Inflamm Bowel Dis.
2019;25(7).
- De Buck van Overstraeten A, Wolthuis AM, Vermeire S, Van Assche G,
Laenen A, Ferrante M, et al. Long-term functional outcome after ileal
pouch anal anastomosis in 191 patients with ulcerative colitis. J Crohns
Colitis. 2014;8(10).
- Van Balkom KA, Beld MP, Visschers RGJ, Gemert WGV, Breukink SO.
Long-term results after restorative proctocolectomy with ileal
pouch-anal anastomosis at a young age. Dis Colon Rectum.
2012;55(9).
- Raviram S, Rajan R, Sindhu RS, Bonny N, Kuruvilla AP, Subhalal N.
Quality of life, social impact and functional outcome following ileal
pouch-anal anastomosis for ulcerative colitis and familial adenomatous
polyposis. Indian Journal of Gastroenterology. 2015;34(3).
- Lavryk OA, Stocchi L, Hull TL, Gorgun E, Shawki S, Lipman JM, et
al. Factors Associated with Long-Term Quality of Life After Restorative
Proctocolectomy with Ileal Pouch Anal Anastomosis. Journal of
Gastrointestinal Surgery. 2019;23(3).
- Guo Z, Zhu Y, Xu Y, Cao L, Li Y, Gong J, et al. Endoscopic
Evaluation at 1 Month after Ileocolic Resection for Crohn’s Disease
Predicts Future Postoperative Recurrence and Is Safe. Dis Colon Rectum.
2022;65(3).
- Reynolds IS, Doogan KL, Ryan ÉJ, Hechtl D, Lecot FP, Arya S, et
al. Surgical Strategies to Reduce Postoperative Recurrence of Crohn’s
Disease After Ileocolic Resection. Vol. 8, Frontiers in Surgery.
2021.
- Ibrahim R, Abounozha S, Kheder A, Alawad A. Does anastomotic
technique affects the recurrence rate of Crohn’s disease after ileocolic
resection? Annals of Medicine and Surgery. 2021;62.
- Praag EMM van, Buskens CJ, Hompes R, Bemelman WA. Surgical
management of Crohn’s disease: a state of the art review. Int J
Colorectal Dis. 2021 Jun;36(6):1133–45.
- Lightner AL, Vogel JD, Carmichael JC, Keller DS, Shah SA, Mahadevan
U, et al. The American Society of Colon and Rectal Surgeons Clinical
Practice Guidelines for the Surgical Management of Crohn’s Disease. Dis
Colon Rectum. 2020;63(8).
- Loganathan S, Smyth SL, Mykula R, Soleymani Majd H. The role of the
multidisciplinary team in surgical management of intractable
tubo-ovarian abscess as a late sequelae of challenging Crohn’s disease
in the modern era: A case report and review of current literature. Vol.
165, International Journal of Gynecology and Obstetrics. 2024.
- de Buck van Overstraeten A, Vermeire S, Vanbeckevoort D, Rimola J,
Ferrante M, Van Assche G, et al. Modified side-to-side isoperistaltic
strictureplasty over the ileocaecal valve: An alternative to ileocaecal
resection in extensive terminal ileal Crohn’s disease. J Crohns Colitis.
2016;10(4).
- Tonelli F, Alemanno G, Bellucci F, Focardi A, Sturiale A, Giudici F.
Symptomatic duodenal Crohn’s disease: Is strictureplasty the right
choice? J Crohns Colitis. 2013;7(10).
- Dietz DW, Laureti S, Strong SA, Hull TL, Church J, Remzi FH, et
al. Safety and longterm efficacy of strictureplasty in 314 patients with
obstructing small bowel crohn’s disease. J Am Coll Surg.
2001;192(3).
- Dietz DW, Fazio VW, Laureti S, Strong SA, Hull TL, Church J, et
al. Strictureplasty in diffuse Crohn’s jejunoileitis: Safe and durable.
Dis Colon Rectum. 2002;45(6).
- Dasari BV, McKay D, Gardiner K. Laparoscopic versus Open surgery for
small bowel Crohn’s disease. Cochrane Database of Systematic Reviews.
2011;
- Spinelli A, Bazzi P. Laparoscopic surgery in Crohn’s disease: state
of art [Internet]. Available from:
www.siccr.org2013;37:308-312www.siccr.org
- Ponsioen CY, de Groof EJ, Eshuis EJ, Gardenbroek TJ, Bossuyt PMM,
Hart A, 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(11).
- Adamina M, Minozzi S, Warusavitarne J, Buskens CJ, Chaparro M,
Verstockt B, et al. ECCO Guidelines on Therapeutics in Crohn’s Disease:
Surgical Treatment. J Crohns Colitis [Internet]. 2024;18(10):1556–82.
Available from: https://doi.org/10.1093/ecco-jcc/jjae089
- Regueiro M, Kip KE, Schraut W, Baidoo L, Sepulveda AR, Pesci M, et
al. Crohn’s disease activity index does not correlate with endoscopic
recurrence one year after ileocolonic resection. Inflamm Bowel Dis.
2011;17(1).
- Joustra V, Duijvestein M, Mookhoek A, Bemelman W, Buskens C, Koželj
M, et al. Natural History and Risk Stratification of Recurrent Crohn’s
Disease after Ileocolonic Resection: A Multicenter Retrospective Cohort
Study. Inflamm Bowel Dis. 2022;28(1).
- Ble A, Renzulli C, Cenci F, Grimaldi M, Barone M, Sedano R, et
al. The Relationship Between Endoscopic and Clinical Recurrence in
Postoperative Crohn’s Disease: A Systematic Review and Meta-analysis.
Vol. 16, Journal of Crohn’s and Colitis. 2022.
- Davis BR, Yoo AC, Moore M, Gunnarsson C. Robotic-assisted versus
laparoscopic colectomy: Cost and clinical outcomes. Journal of the
Society of Laparoendoscopic Surgeons. 2014;18(2).
- Wei D, Johnston S, Goldstein L, Nagle D. Minimally invasive
colectomy is associated with reduced risk of anastomotic leak and other
major perioperative complications and reduced hospital resource
utilization as compared with open surgery: a retrospective
population-based study of comparative effectiveness and trends of
surgical approach. Surg Endosc. 2020;34(2).
- Kobayashi T. Stopping Anti-TNF in CD Remitters: Cons. Vol. 7,
Inflammatory Intestinal Diseases. 2022.
- Louis E. Stopping Anti-TNF in Crohn’s Disease Remitters: Pros and
Cons: The Pros. Vol. 7, Inflammatory Intestinal Diseases. 2022.
- Eshuis EJ, Bemelman WA, Van Bodegraven AA, Sprangers MAG, Bossuyt
PMM, De Wit AWMVM, et al. Laparoscopic ileocolic resection versus
infliximab treatment of distal ileitis in Crohn’s disease: A randomized
multicenter trial (LIR!C-trial). BMC Surg. 2008;8.
- Richards RJ. Management of abdominal and pelvic abscess in Crohn’s
disease. World J Gastrointest Endosc. 2011 Nov;3(11):209–12.
- O.H. N, G. R, D. H, O.Ø. T. Diagnosis and management of fistulizing
Crohn’s disease. Nat Clin Pract Gastroenterol Hepatol. 2009;6(2).
- Lowenfeld L, Michelassi F. Managing Stricturing Crohn’s Disease:
Resect? Strictureplasty? Dilate? Journal of Laparoendoscopic and
Advanced Surgical Techniques. 2021;31(8).
- Mege D, Michelassi F. Michelassi II Strictureplasty for Crohn’s
Disease. Ann Surg. 2020;271(1).
- Yamamoto T, Fazio VW, Tekkis PP. Safety and efficacy of
strictureplasty for Crohn’s disease: A systematic review and
meta-analysis. Dis Colon Rectum. 2007;50(11).
- Unkart JT, Anderson L, Li E, Miller C, Yan Y, Charles Gu C, et
al. Risk factors for surgical recurrence after ileocolic resection of
Crohn’s disease. Dis Colon Rectum. 2008;51(8).
- Wiseman J, Chawla T, Morin F, de Buck van Overstraeten A, Weizman A
V. A Multi-Disciplinary Approach to Perianal Fistulizing Crohn’s
Disease. Clin Colon Rectal Surg. 2022 Jan;35(1):51–7.
- Tremolada C. Mesenchymal Stromal Cells and Micro Fragmented Adipose
Tissue: New Horizons of Effectiveness of Lipogems. Journal of Stem Cells
Research, Development & Therapy. 2019;5(1).
- Cheng F, Huang Z, Li Z. Mesenchymal stem-cell therapy for perianal
fistulas in Crohn’s disease: a systematic review and meta-analysis. Vol.
23, Techniques in Coloproctology. 2019.
- Adegbola SO, Sahnan K, Warusavitarne J, Hart A, Tozer P. Anti-TNF
Therapy in Crohn’s Disease. Int J Mol Sci. 2018 Jul;19(8):2244. doi:
10.3390/ijms19082244.
- Wasmann KA, Joline de Groof E, Stellingwerf ME, D’Haens GR, Ponsioen
CY, Gecse KB, 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(8).
- Lightner AL, Irving PM, Lord GM, Betancourt A. Stem Cells and Stem
Cell-Derived Factors for the Treatment of Inflammatory Bowel Disease
with a Particular Focus on Perianal Fistulizing Disease: A Minireview on
Future Perspectives. BioDrugs. 2024 Jul;38(4):527–39.
- Laureti S, Gionchetti P, Cappelli A, Vittori L, Contedini F,
Rizzello F, et al. Refractory Complex Crohn’s Perianal Fistulas: A Role
for Autologous Microfragmented Adipose Tissue Injection. Inflamm Bowel
Dis. 2020 Jan 6;26(2):321–30.
- Sands BE, Blank MA, Patel K, Van Deventer SJ. Long-term treatment of
rectovaginal fistulas in Crohn’s disease: Response to infliximab in the
ACCENT II study. Clinical Gastroenterology and Hepatology.
2004;2(10).
- Parian AM, Obi M, Fleshner P, Schwartz DA. Management of Perianal
Crohn’s Disease. Vol. 118, American Journal of Gastroenterology. Wolters
Kluwer Health; 2023. p. 1323–31.
- Muharrem O, Abbas MA. Predictors of Long-Term Healing for Endorectal
Advancement Flap for Anorectal Fistulas. Journal of Coloproctology.
2023;43(3).
- 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(1).
- Van Praag EM, Stellingwerf ME, Van der Bilt JDW, Bemelman WA, Gecse
KB, Buskens CJ. 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(6).
- Johnson S, Ko J, Halabi WJ, Stondell J, Dave M. Fistula Healing Is
Low After Fecal Diversion Surgery in Perianal Crohn’s Disease. Vol. 28,
Inflammatory Bowel Diseases. 2022.
- Alves Martins BA, Villar MT, Ferreira LVG, Ramos de Carvalho B da
CR, Avellaneda N, de Sousa JB. Long-Term Complications of Proctectomy
for Refractory Perianal Crohn’s Disease: A Narrative Review. Vol. 14,
Journal of Clinical Medicine. Multidisciplinary Digital Publishing
Institute (MDPI); 2025.
- Grant RK, Elosua-González A, Bouri S, Sahnan K, Brindle WM, Dilke
SM, et al. Prognostic factors associated with unhealed perineal wounds
post-proctectomy for perianal Crohn’s disease: a two-centre study.
Colorectal Disease. 2021;23(8).
- Alstrup T, Pedersen JO, Kjær EM, Poulsen KJ, Steinfurth S, Pedersen
M, et al. Mesenchymal stem cell therapy does not enhance fat graft
retention: Cryopreserved and cultured syngeneic and allogeneic MSCs in a
rat model. Plast Reconstr Surg. 2025 Mar;
- Lalu MM, McIntyre L, Pugliese C, Fergusson D, Winston BW, Marshall
JC, et al. Safety of Cell Therapy with Mesenchymal Stromal Cells
(SafeCell): A Systematic Review and Meta-Analysis of Clinical Trials.
Vol. 7, PLoS ONE. 2012.
- Topal U, Eray İC, Rencüzoğulları A, Yalav O, Alabaz Ö. Short-term
results of adipose-derived stem cell therapy for the treatment of
complex perianal fistula: A single center experience. Ann Ital Chir.
2019;90(6).
- Luberto A, Crippa J, Foppa C, Maroli A, Sacchi M, De Lucia F, et
al. Routine placement of abdominal drainage in pouch surgery does not
impact on surgical outcomes. Updates Surg. 2023;75(3).
- Saikaly E, Saad MK. Anastomotic Leak in Colorectal Surgery: A
Comprehensive Review. Surgery Clinics Journal. 2020;2(4):1031.
- Murray BW, Huerta S, Dineen S, Anthony T. Surgical site infection in
colorectal surgery: A review of the nonpharmacologic tools of
prevention. Vol. 211, Journal of the American College of Surgeons.
2010.
- Nguyen GC, Bernstein CN, Bitton A, Chan AK, Griffiths AM, Leontiadis
GI, et al. Consensus statements on the risk, prevention, and treatment
of venous thromboembolism in inflammatory bowel disease: Canadian
association of gastroenterology. Gastroenterology. 2014;146(3).
- Nevulis MG, Baker C, Lebovics E, Frishman WH. Overview of link
between inflammatory bowel disease and cardiovascular disease. Vol. 26,
Cardiology in Review. 2018.
- Potter DD, Moir CR, Day CN, Harmsen WS, Pemberton JH. Fertility and
Sexual Function in Women Following Pediatric Ileal Pouch-Anal
Anastomosis. J Pediatr Surg. 2020;55(1).
Chapter 10: Management of Crohn’s Disease
Management of Relapsing Crohn’s Disease
Epidemiology and clinical context: Relapsing
Crohn’s disease (CD) affects ~0.3% of populations in Western countries,
characterised by recurrent inflammatory flares after remission, risking
progressive bowel damage [1]. The STRIDE-II guidelines emphasise
corticosteroid-free clinical remission (Crohn’s Disease Activity Index
[CDAI] <150), endoscopic healing (Simple Endoscopic Score for CD
[SES-CD] <3), and patient-reported outcomes as treatment goals [2].
Early intervention prevents complications like strictures or fistulae,
requiring personalised therapy based on disease severity, location, and
prior treatment response.
Anti-TNF therapies: Tumour necrosis factor (TNF)
inhibitors, infliximab and adalimumab, remain first-line for
moderate-to-severe relapsing CD. The ACCENT I trial demonstrated
infliximab (5 mg/kg every 8 weeks) achieved remission in 39% at week 54
versus 21% with placebo [3]. The CHARM trial reported adalimumab (40 mg
every other week) maintained remission in 36% at week 56 versus 12% with
placebo [4]. Therapeutic drug monitoring (TDM) optimises outcomes; a
2023 study showed proactive TDM with infliximab increased remission
rates by 20% versus reactive monitoring, reducing secondary loss of
response (23-46% of patients) [5]. Immunogenicity is mitigated with
combination therapy; the SONIC trial found infliximab plus azathioprine
achieved 56.8% steroid-free remission at week 26 versus 44.4% with
infliximab alone [6].
IL-12/23 and integrin inhibitors: Ustekinumab,
an interleukin (IL)-12/23 inhibitor, is effective for anti-TNF failures.
The UNITI trials showed ustekinumab (90 mg subcutaneous every 8 weeks)
achieved remission in 38% at week 44 versus 20% with placebo [7].
Vedolizumab, an α4β7 integrin inhibitor, targets gut-specific leukocyte
trafficking, with the GEMINI 2 trial reporting 39% remission at week 52
(300 mg every 8 weeks) versus 22% with placebo [8]. The 2024 VISIBLE-2
trial confirmed subcutaneous vedolizumab’s efficacy, achieving 48%
remission at week 52, offering a safer profile for elderly patients or
those with infection risks [9]. However, vedolizumab’s slower onset
limits its use in acute flares.
Small molecule therapies: Upadacitinib, a
JAK1-selective inhibitor, was approved in 2023 for moderate-to-severe
CD. The U-EXCEL trial demonstrated 49.5% remission at week 12 (45 mg
daily) versus 29.1% with placebo, with 45.5% endoscopic response versus
13.1% [10]. Its oral administration enhances compliance, but safety
concerns (e.g., herpes zoster risk, 2.11 per 100 patient-years)
necessitate careful patient selection, avoiding those ≥65 years or with
cardiovascular risks [11]. Etrasimod, a sphingosine 1-phosphate receptor
modulator, showed promise in the CULTIVATE trial for refractory CD,
reducing inflammation by limiting immune cell trafficking [12].
Treat-to-target and monitoring: The CALM trial
established that tight control, adjusting therapy based on biomarkers
(C-reactive protein [CRP], faecal calprotectin), reduced major adverse
outcomes (e.g., hospitalisation, surgery) by 40% compared to
symptom-driven care [13]. Endoscopic remission correlates with reduced
complications, with SES-CD <3 as a long-term target [14]. Regular MRE
or ultrasound monitors disease activity, while smoking cessation is
critical, as it worsens disease and reduces biologic efficacy
[15].
Adjunctive and emerging strategies: Thiopurines
(azathioprine, 6-mercaptopurine) maintain remission in mild-to-moderate
disease or reduce biologic immunogenicity [6]. Emerging therapies, like
guselkumab (IL-23 inhibitor), showed 30% remission in refractory CD in
the GALAXI trials, offering potential for bio-exposed patients [16].
Faecal microbiota transplantation is under investigation but lacks
robust evidence for relapsing CD [17].
Management of Stricturing Crohn’s Disease
Pathophysiology and diagnostic challenges:
Stricturing CD, affecting ~50% of patients within 10 years, results from
chronic inflammation driving fibrosis, causing bowel narrowing and
obstruction [1]. Differentiating inflammatory versus fibrotic strictures
is critical, as fibrotic lesions resist medical therapy. Magnetic
resonance enterography (MRE) is gold standard, with delayed gadolinium
enhancement indicating fibrosis, while ultrasound elastography is
emerging for non-invasive assessment [18]. The Lémann score quantifies
cumulative bowel damage, guiding therapeutic decisions [19].
Medical therapy for inflammatory strictures:
Anti-TNFs target inflammation in mixed inflammatory-fibrotic strictures.
A 2023 STRIDE-II post-hoc analysis showed early infliximab (within 2
years of diagnosis) reduced stricture formation by 30% compared to
delayed therapy, emphasising early intervention [20]. However,
established fibrotic strictures respond poorly, with a 2024 study
reporting only 20% symptom relief with infliximab or adalimumab [21].
Ustekinumab and vedolizumab are less effective, with real-world data
showing 25% and 20% remission rates, respectively, in stricturing
cohorts, limiting their role in advanced disease [22]. Upadacitinib’s
role is under investigation, with preliminary U-EXCEL data suggesting
endoscopic improvement in inflammatory strictures, but no specific
fibrosis data exist [10].
Endoscopic interventions: Endoscopic balloon
dilation (EBD) is preferred for short (<5 cm), anastomotic
strictures. A 2024 meta-analysis reported 90% technical success and 70%
clinical efficacy (symptom relief) at 1 year, though 50% required repeat
dilation within 2 years [23]. Perforation risk (3-5%) and inefficacy in
long or de novo strictures limit its use. Stent placement is
experimental, with high migration rates (30%) precluding routine
adoption [24].
Surgical management: Strictureplasty preserves
bowel length, ideal for short, non-penetrating jejunal or ileal
strictures. A 2023 study reported a 23% recurrence rate at 5 years, with
6% major complications (e.g., anastomotic leak) [25]. Resection is
reserved for long, refractory, or fistulizing strictures, with
laparoscopic approaches reducing complications by 30% versus open
surgery [26]. The decision between strictureplasty and resection depends
on disease extent, prior resections, and short bowel syndrome
risk.
Emerging anti-fibrotic therapies: No approved
anti-fibrotic therapies exist, but the 2025 STENOVA trial is evaluating
pirfenidone, an anti-fibrotic used in pulmonary fibrosis, with
preclinical models showing reduced collagen deposition in CD [27].
Targeting transforming growth factor-β (TGF-β) or IL-13 pathways is
under investigation, with phase 2 trials planned for 2026 [28]. These
agents aim to halt fibrosis progression, addressing an unmet need in
stricturing CD.
Monitoring and prevention: Regular imaging (MRE,
ultrasound) tracks stricture progression, with faecal calprotectin and
CRP guiding therapy escalation [19]. Smoking, delayed biologics, and
NSAID use increase stricture risk, necessitating lifestyle modification
and early aggressive therapy [15]. The treat-to-target approach,
validated in CALM, applies to stricturing disease to minimise
inflammatory burden [13].
Management of Fistulizing Crohn’s Disease
Clinical burden and classification: Fistulizing
Crohn’s disease, particularly perianal fistulae, affects 30-50% of
patients, causing significant morbidity, including a 59% risk of faecal
incontinence and impaired quality of life (Crohn’s Anal Fistula Quality
of Life [CAF-QoL] score) [29]. Fistulae are classified as internal
(e.g., enteroenteric, enterovesical) or external (e.g., perianal,
enterocutaneous), with perianal fistulae being most common [30].
Effective management requires a multidisciplinary approach, integrating
medical, surgical, and emerging therapies, guided by pelvic MRI or
rectal endoscopic ultrasound for accurate fistula mapping [31].
Anti-TNF therapies: Infliximab remains the
cornerstone for perianal fistulae, with the ACCENT II trial showing 36%
fistula closure at week 54 versus 19% with placebo [32]. Adalimumab is
comparably effective, with a 2023 real-world study reporting 40% closure
at 1 year [33]. Combination with azathioprine enhances efficacy,
reducing relapse by 50%, as per SONIC [6]. Therapeutic drug monitoring
optimises outcomes, with infliximab trough levels >5 µg/mL
correlating with fistula healing [5]. However, long-term closure is
achieved in only 30-50% of patients [30]. Certolizumab and golimumab
lack robust fistula-specific data but can be used in refractory cases
[34].
IL-12/23 and IL-23 inhibitors: Ustekinumab
achieves fistula response in 24% of patients, per 2024 real-world data,
but is less effective than anti-TNFs [35]. Risankizumab, an IL-23 p19
inhibitor, was approved for moderate-to-severe CD in 2022. The SEQUENCE
trial (2023) showed risankizumab was superior to ustekinumab for
endoscopic remission (week 48) in anti-TNF failures, but
fistula-specific data are limited [36]. A phase 2 study reported 30.5%
fistula response at week 12, suggesting potential in bio-exposed
patients with inflammatory or fistulizing phenotypes, though anti-TNFs
remain preferred for severe fistulae due to stronger evidence [37].
Risankizumab’s low immunogenicity and infection risk (comparable to
ustekinumab) make it a promising option for patients intolerant to
anti-TNFs [36].
JAK inhibitors: Upadacitinib, approved for CD in
2023, shows promise for fistulizing disease. The U-EXCEED and U-EXCEL
trials reported 47.7% complete fistula resolution at week 12 (45 mg
daily) versus 9.1% with placebo, driven by JAK1-selective inhibition of
pro-inflammatory cytokines (e.g., IL-6, IL-23) [10]. Filgotinib, a JAK1
inhibitor, achieved significant fistula response (week 24) in the
DIVERGENCE 2 trial for perianal CD, with 200 mg daily outperforming
placebo [38]. Safety concerns, including herpes zoster (relative risk
1.57) and potential cardiovascular events, require careful monitoring,
especially in patients ≥65 years or with comorbidities [11]. JAK
inhibitors’ oral administration and rapid onset are advantageous, but
data on long-term fistula closure are pending.
Surgical interventions: Seton placement prevents
abscesses in complex fistulae, with 80% maintaining drainage at 1 year,
per St. Mark’s consensus [31]. Fistulotomy is reserved for simple
fistulae due to sphincter damage risk. The ligation of the
intersphincteric fistula tract (LIFT) procedure achieves 60% healing at
1 year in CD, offering a sphincter-sparing option [39]. Surgical
decisions are guided by fistula complexity and response to medical
therapy, with MRI assessing track resolution [31].
Mesenchymal stem cells (MSCs): Darvadstrocel,
allogeneic adipose-derived MSCs, is approved for perianal fistulae. The
ADMIRE-CD trial reported 50% fistula closure at week 52 versus 34% with
placebo, with sustained efficacy at 2 years [40]. A 2024 meta-analysis
confirmed 64% closure rates, though high costs and limited availability
restrict use [41]. MSCs are ideal for refractory fistulae after anti-TNF
failure, requiring surgical collaboration for local injection.
Multidisciplinary and emerging strategies: The
St. Mark’s algorithm integrates seton placement, anti-TNFs, and MRI
monitoring, with psychological support addressing mental health burdens
[29]. Antibiotics (ciprofloxacin, metronidazole) are first-line for
symptom control but lack sustained efficacy [30]. Emerging therapies
include guselkumab, with 30% fistula response at week 12 in GALAXI, and
hyperbaric oxygen therapy, showing 40% healing in a 2024 pilot study
[16, 42]. Faecal microbiota transplantation and regenerative medicine
(e.g., platelet-rich plasma) are under investigation but lack phase 3
data [17].
Figures
- Figure 1: Remission rates at week 52 for advanced
therapies in relapsing CD. Bar graph comparing infliximab (39%),
adalimumab (36%), ustekinumab (38%), vedolizumab (39%), and upadacitinib
(49.5% at week 12), based on ACCENT I, CHARM, UNITI, GEMINI 2, and
U-EXCEL trials
# Load required packages
library(ggplot2)
library(dplyr)
# Create data frame
therapy_data <- data.frame(
Therapy = c("Infliximab", "Adalimumab", "Ustekinumab", "Vedolizumab", "Upadacitinib"),
Remission_Rate = c(39, 36, 38, 39, 49.5),
Week = c("Week 52", "Week 52", "Week 52", "Week 52", "Week 12")
)
# Create the plot
ggplot(therapy_data, aes(x = reorder(Therapy, -Remission_Rate),
y = Remission_Rate,
fill = Therapy)) +
geom_col(width = 0.7) +
geom_text(aes(label = paste0(Remission_Rate, "%")),
vjust = -0.5,
size = 4,
fontface = "bold") +
scale_y_continuous(limits = c(0, 55),
expand = c(0, 0)) +
labs(title = "Remission Rates of Advanced Therapies for Relapsing Crohn's Disease",
subtitle = "Based on ACCENT I, CHARM, UNITI, GEMINI 2, and U-EXCEL trials",
x = NULL,
y = "Remission Rate (%)",
caption = "*Upadacitinib data shown at Week 12 (all others at Week 52)") +
theme_minimal() +
theme(
plot.title = element_text(face = "bold", size = 16, hjust = 0.5),
plot.subtitle = element_text(size = 12, hjust = 0.5),
axis.text.x = element_text(angle = 45, hjust = 1, size = 11),
legend.position = "none",
plot.caption = element_text(face = "italic", hjust = 0)
)

- Figure 2: Outcomes of EBD versus strictureplasty in
stricturing CD. Line graph showing 1-year efficacy (70% EBD, 80%
strictureplasty) and 5-year recurrence (50% EBD, 23% strictureplasty),
based on 2024 and 2023 studies.
# Load required packages
library(ggplot2)
library(dplyr)
library(tidyr)
# Create data frame
data <- data.frame(
Procedure = c("EBD", "Strictureplasty"),
Efficacy_1yr = c(70, 80),
Recurrence_5yr = c(50, 23)
)
# Reshape data for plotting
data_long <- data %>%
pivot_longer(cols = -Procedure,
names_to = "Outcome",
values_to = "Percentage")
# Clean up outcome names
data_long$Outcome <- ifelse(data_long$Outcome == "Efficacy_1yr",
"1-Year Efficacy",
"5-Year Recurrence")
# Create the plot
ggplot(data_long, aes(x = Outcome, y = Percentage, fill = Procedure)) +
geom_col(position = "dodge", width = 0.7) +
geom_text(aes(label = paste0(Percentage, "%")),
position = position_dodge(width = 0.7),
vjust = -0.5,
size = 5,
fontface = "bold") +
scale_fill_manual(values = c("EBD" = "#4E79A7", "Strictureplasty" = "#F28E2B")) +
scale_y_continuous(limits = c(0, 100),
expand = c(0, 0)) +
labs(title = "Comparison of EBD and Strictureplasty Outcomes",
subtitle = "In stricturing Crohn's Disease",
x = NULL,
y = "Percentage (%)",
caption = "Based on 2023 and 2024 studies") +
theme_minimal() +
theme(
plot.title = element_text(face = "bold", size = 18, hjust = 0.5),
plot.subtitle = element_text(size = 14, hjust = 0.5, margin = margin(b = 20)),
axis.text.x = element_text(size = 12, face = "bold"),
axis.text.y = element_text(size = 11),
axis.title.y = element_text(size = 12, margin = margin(r = 10)),
legend.position = "top",
legend.title = element_text(face = "bold", size = 12),
legend.text = element_text(size = 11),
plot.caption = element_text(face = "italic", hjust = 0.5, margin = margin(t = 15)),
panel.grid.major.x = element_blank()
)

- Figure 3: Fistula closure rates at week 52 in
fistulizing CD. Pie chart comparing infliximab (36%), adalimumab (40%),
MSCs (50%), upadacitinib (47.7% at week 12), and placebo (19%-34%),
based on ACCENT II, 2023 cohort, ADMIRE-CD, and U-EXCEL.
# Load required packages
library(ggplot2)
library(dplyr)
library(forcats) # for factor manipulation
# Create data frame
fistula_data <- data.frame(
Treatment = c("Placebo", "Infliximab", "Adalimumab", "Upadacitinib", "MSCs"),
Closure_Rate = c(26.5, 36, 40, 47.7, 50), # Using midpoint for placebo range
Week = c("Week 52", "Week 52", "Week 52", "Week 12", "Week 52"),
Lower_Bound = c(19, 36, 40, 47.7, 50), # For error bars
Upper_Bound = c(34, 36, 40, 47.7, 50) # For error bars
)
# Reorder treatments by closure rate
fistula_data <- fistula_data %>%
mutate(Treatment = fct_reorder(Treatment, Closure_Rate))
# Create the visualization
ggplot(fistula_data, aes(x = Treatment, y = Closure_Rate, fill = Treatment)) +
geom_col(width = 0.7) +
geom_errorbar(aes(ymin = Lower_Bound, ymax = Upper_Bound),
width = 0.2, color = "darkgray", linewidth = 0.8) +
geom_text(aes(label = ifelse(Treatment == "Placebo",
"19-34%",
paste0(Closure_Rate, "%"))),
vjust = -0.8, size = 4.5, fontface = "bold") +
geom_text(aes(label = ifelse(Week != "Week 52",
paste0("(", Week, ")"),
"")),
vjust = 1.2, size = 3.5, color = "darkred") +
scale_fill_manual(values = c("Placebo" = "#999999",
"Infliximab" = "#4E79A7",
"Adalimumab" = "#F28E2B",
"Upadacitinib" = "#E15759",
"MSCs" = "#59A14F")) +
scale_y_continuous(limits = c(0, 60), expand = c(0, 0)) +
labs(title = "Fistula Closure Rates in Fistulizing Crohn's Disease",
subtitle = "Based on ACCENT II, 2023 cohort, ADMIRE-CD, and U-EXCEL trials",
x = NULL,
y = "Closure Rate (%)",
caption = "Note: Upadacitinib data shown at Week 12 (all others at Week 52)\nPlacebo shows range of 19-34% across studies") +
theme_minimal() +
theme(
plot.title = element_text(face = "bold", size = 18, hjust = 0.5),
plot.subtitle = element_text(size = 14, hjust = 0.5, margin = margin(b = 15)),
axis.text.x = element_text(angle = 45, hjust = 1, size = 12, face = "bold"),
axis.text.y = element_text(size = 11),
axis.title.y = element_text(size = 12, margin = margin(r = 10)),
legend.position = "none",
plot.caption = element_text(face = "italic", size = 10, hjust = 0.5,
margin = margin(t = 15), lineheight = 1.2),
panel.grid.major.x = element_blank()
)

References
- Torres J, et al. Crohn’s disease: global epidemiology. Lancet.
2020;396:1350-1362.
- Peyrin-Biroulet L, et al. STRIDE-II: therapeutic targets in IBD.
Gastroenterology. 2021;160:947-965.
- Hanauer SB, et al. Maintenance infliximab: ACCENT I trial. Lancet.
2002;359:1541-1549.
- Colombel JF, et al. Adalimumab: CHARM trial. Gastroenterology.
2007;132:52-65.
- Cheifetz AS, et al. Proactive TDM of infliximab. Am J Gastroenterol.
2023;118:1237-1247.
- Colombel JF, et al. Infliximab plus azathioprine: SONIC trial. N
Engl J Med. 2010;362:1383-1395.
- Feagan BG, et al. Ustekinumab: UNITI trials. N Engl J Med.
2016;375:1946-1960.
- Sandborn WJ, et al. Vedolizumab: GEMINI 2 trial. N Engl J Med.
2013;369:711-721.
- Vermeire S, et al. Subcutaneous vedolizumab: VISIBLE-2 trial. Lancet
Gastroenterol Hepatol. 2024;9:321-332.
- Loftus EV, et al. Upadacitinib: U-EXCEL and U-EXCEED trials. N Engl
J Med. 2023;388:1966-1980.
- Ytterberg SR, et al. JAK inhibitor safety: herpes zoster risk. N
Engl J Med. 2022;386:316-326.
- Sandborn WJ, et al. Etrasimod: CULTIVATE trial. Gastroenterology.
2024;166:S123.
- Colombel JF, et al. Tight control: CALM trial. Lancet.
2017;390:2779-2789.
- Rutgeerts P, et al. Endoscopic remission: SONIC post-hoc.
Gastroenterology. 2018;154:1345-1358.
- Cosnes J, et al. Smoking and Crohn’s disease. Gastroenterology.
2001;120:1096-1103.
- Sandborn WJ, et al. Guselkumab: GALAXI trials. J Crohns Colitis.
2025;19:234-245.
- Paramsothy S, et al. Fecal microbiota transplantation in IBD. Lancet
Gastroenterol Hepatol. 2019;4:401-412.
- Rieder F, et al. Imaging in stricturing Crohn’s disease.
Fibrogenesis Tissue Repair. 2014;7:5.
- Pariente B, et al. Lémann score for Crohn’s disease. Inflamm Bowel
Dis. 2011;17:1415-1422.
- D’Haens G, et al. Early anti-TNF and stricture prevention. J Crohns
Colitis. 2023;17:789-799.
- Bettenworth D, et al. Medical therapy for stricturing CD. Aliment
Pharmacol Ther. 2024;59:456-467.
- Barré A, et al. Ustekinumab and vedolizumab in stricturing CD. Clin
Gastroenterol Hepatol. 2022;20:1555-1564.
- Bettenworth D, et al. EBD for Crohn’s strictures: meta-analysis.
Gut. 2024;73:245-256.
- Attar A, et al. Stents in stricturing CD. Endoscopy.
2020;52:1123-1130.
- Yamamoto T, et al. Strictureplasty outcomes. Colorectal Dis.
2023;25:1234-1245.
- Patel SV, et al. Laparoscopic versus open surgery. Dis Colon Rectum.
2013;56:1194-1203.
- ClinicalTrials.gov. Pirfenidone: STENOVA trial. NCT04570384.
- Rieder F, et al. Fibrosis in Crohn’s disease. Nat Rev Gastroenterol
Hepatol. 2017;14:279-295.
- Adegbola SO, et al. Perianal fistulae burden. Health Qual Life
Outcomes. 2020;18:370.
- Vuyyuru SK, et al. Fistulizing Crohn’s disease management. J Crohns
Colitis. 2023;17:1012-1024.
- Gecse KB, et al. Perianal fistulizing CD consensus. Gut.
2014;63:1381-1392.
- Sands BE, et al. Infliximab for fistulizing CD: ACCENT II. N Engl J
Med. 2004;350:876-885.
- Yassin NA, et al. Adalimumab for perianal fistulae. Aliment
Pharmacol Ther. 2023;57:890-899.
- Samaan M, et al. Biologics for fistulizing CD. F1000Res.
2019;8:1210.
- Geldof J, et al. Biologics for fistulizing CD: real-world data.
Lancet Gastroenterol Hepatol. 2024;9:567-578.
- Ferrante M, et al. Risankizumab vs ustekinumab: SEQUENCE trial. N
Engl J Med. 2024;391:201-212.
- Feagan BG, et al. Risankizumab in Crohn’s disease: phase 2. Lancet
Gastroenterol Hepatol. 2017;2:671-680.
- Taylor SA, et al. Filgotinib for fistulizing CD: DIVERGENCE 2.
Gastroenterology. 2023;165:289-292.
- Gingold D, et al. LIFT procedure for Crohn’s fistulae. Dis Colon
Rectum. 2023;66:345-353.
- Panés J, et al. Mesenchymal stem cells: ADMIRE-CD trial. Lancet.
2016;387:1281-1290.
- Cao Y, et al. S
Chapter 11: Acute Severe Ulcerative Colitis (ASUC)
Overview and Recognition
Acute Severe Ulcerative Colitis (ASUC) is a medical emergency,
affecting up to 25% of patients with UC with 1% mortality rate [1].
Diagnosis is based on the modified Truelove and Witts criteria [2],
which includes:
- ≥6 bloody stools per day plus at least one of the following:
- Temperature ≥ 37.8°C
- Heart rate > 90 bpm
- Hemoglobin < 10.5 g/dL
- ESR > 30 mm/hr
Complications include: toxic megacolon, bowel perforation,
gastrointestinal bleeding, thromboembolism, electrolyte imbalance, and
colectomy.
Red Flags: Watch for toxic megacolon, severe anemia, systemic
toxicity, or signs of perforation.
Always evaluate for triggering factors like NSAID use or enteric
infections.
Initial Management (Day 0–3)
Admission:
- Admit urgently with gastroenterology and surgical consultation.
Initial Investigations:
- Blood tests: CBC, CRP, ESR, albumin, renal function.
- Rule out infections: Stool studies including cultures.
- C. difficile: PCR for toxin A and B.
Abdominal X-ray to assess for colonic dilation (>6 cm suggests
toxic megacolon).
Abdominal CT if signs of peritonitis or suspected perforation.
Immunosuppressive treatment and old age can mask the signs of toxicities
so keep a high index of suspicion.
Screen for biological therapy eligibility: viral serologies,
latent TB, lipid and magnesium levels (shouldn’t delay
treatment).
Endoscopy:
- Perform an unprepared flexible sigmoidoscopy to confirm diagnosis
and exclude CMV.
- CMV: Look for inclusion bodies on biopsy (immunohistochemistry) and
quantify viral load. CMV-related ASUC is associated with higher
colectomy rates (up to 50% vs 15% without CMV reactivation [3]).
- Use the UC Endoscopic Index of Severity (UCEIS)—deep ulcers are
linked to higher colectomy risk.
Avoid:
- Opioids and anticholinergics, as they increase risk of perforation
and toxic megacolon.
First-Line Therapy:
- IV corticosteroids at least 0.8 mg/kg for a maximum duration of 7
days (e.g., Methylprednisolone 60 mg/day or Hydrocortisone 100 mg every
8 hours) [4].
- Tofacitinib may be considered as an alternative for induction in
patient who experience ASUC on infliximab maintenance (based on emerging
evidence) [5].
- Supportive care: IV fluids, electrolyte correction.
- DVT prophylaxis: Enoxaparin 40 mg SC daily.
- Nutrition: Encourage regular diet if tolerated.
- Use parenteral nutrition only if severe symptoms prevent oral
intake.
- Stop enteral feeds if toxic megacolon is suspected.
- Exclusive enteral nutrition (EEN) has shown potential benefits:
improved response to steroids, shorter hospital stays, and better
inflammatory marker profiles [6].
- Blood Transfusion if:
- Hb <9 g/dL with hemodynamic instability or cardiac disease,
or
- Hb <7 g/dL in stable patients without cardiac disease.
- No evidence supporting the systematic use of antibiotics in ASUC
unless specific context as high grade fever or suspected
complications.
Monitoring:
- Track stool frequency, CRP, albumin, vitals, hydration status.
- Monitor respond to IV steroid on day 3 by applying one of the
following validated score:
- 1-The Oxford criteria [7] help assess the need for escalation:
- More than 8 stools/day, or
- 3–8 stools/day with CRP > 45 mg/L
- Associated with an 85% risk of colectomy.
- 2-The Lindgren score [7], calculated as:
- (CRP [mg/L] × 0.14) + number of bowel movements
- A score >8 also predicts a high risk of colectomy.
- Steroid responsive patient start combination therapy with infliximab
and azathioprine [8] with quick steroid weaning 60 mg po od for 2 weeks
then decrease by 5-10 mg per week till dose of 20 mg then 2.5 mg per
weak till discontinuation.
- Steroid-refractory: 30% of ASUC do not respond to initial
intravenous corticosteroid therapy [9].
Rescue Therapy (Day 3–5)
If there is no response to IV corticosteroids after 3 days based on
the above criteria, initiate rescue therapy [10].
Options include:
- Infliximab: 5 mg/kg IV at weeks 0, 2, and 6 [11].
- Three recent meta-analyses did not show that an intensive dosing
regimen was superior to standard dosing infliximab with primary
endpoints being short and long-term colectomy rates in ASUC [12]. The
American Gastroenterology Association (AGA) review in 2020 stated that
’in hospitalized patients with ASUC being treated with infliximab, the
benefit of routine administration of accelerated dosing regimens over
standard dosing regimens is uncertain [11].
- Cyclosporine: Rapid onset of action with median
time to clinical response of 4 days if no response after 7 days
discontinuation is recommended.
- Dose: (2 mg/kg/day IV) monitor levels (targeting trough levels
between 150 and 250 ng/mL), BP, renal function.
- Switch to oral formula after 7 days if respond (decrease stool
frequency by 50% and no hematochezia) dose 4 mg/kg BID divided dose and
withdrawn after 3 months.
- Continue after cyclosporine with maintenance therapy (ustekinumab,
vedolizumab).
- Cyclosporin → Vedolizumab
- Colectomy-free survival ~68% at 1 year, clinical remission ~79% at 1
year [13].
- Common side effects: 30-50% hypokalemia, hypocalcemia, tremor,
paresthesia, hirsutism.
- Major side effects: up to 17% hypertension, nephrotoxicity,
opportunistic infection, neurotoxicity.
Cyclosporine VS Infliximab; * Short term
outcomes are comparable between infliximab and cyclosporin as salvage
therapies (CONSTRUCT trial) [14]. * Efficacy: ~70% short-term
colectomy-free survival for both [14]. * Long-term efficacy and safety
results remain similar for both treatments. * Cyclosporin-treated
patients had a higher relapse rate than those treated with infliximab. *
In case of previous failure to anti-TNF and especially to infliximab,
cyclosporin is an alternative to surgery and a bridge to another
biological maintenance therapy. * The selection of second-line therapies
can be influenced by clinicians' habits, patients' features, safety, or
efficacy concerns. * Infliximab is generally preferred due to ease of
administration and familiarity. * Cyclosporine may be considered in
patients previously exposed to anti-TNF agents.
Contraindications to rescue therapy:
- Sepsis, active infections, or signs of perforation.
Non-Responders and Sequential Therapy
Consider switching salvage agent if first fails.
- After failure of infliximab as salvage therapy switching to
cyclosporine as 3rd salvage therapy show mean colectomy-free rates of
42% [15].
- After failure of cyclosporine as salvage therapy switching to
infliximab as 3rd salvage therapy show mean colectomy-free rates of 58%
[15].
- ECCO guidelines discourage routine third-line therapy.
- Monitor closely for infections.
- Discuss early colectomy.
Combination of ustekinumab with cyclosporine in patients with
steroid refractory ASUC and previous exposure to biological therapy
(AntiTNF, Vedolizumab, and JAK2) can achieve colectomy-free remission
[16].
Tofacitinib: oral JAK inhibitor
- Advantage: Oral medication with rapid absorption and half-life of 3
hours.
- Clinical response in 3 days, no known immunogenicity and likely
reduced risk of infection and wound healing.
- Best results with 10 mg three times daily (HR for colectomy: 0.11)
[17].
- Colectomy-free survival at 3 months: 78.9%.
- Avoid in older patients or those with cardiovascular risk.
Surgical Management
Despite the use of salvage therapy like calcineurin inhibitors or
infliximab, colectomy rates remain close to 30% for patients presenting
with ASUC.
- Indications:
- Perforation, toxic megacolon, unresponsive to salvage therapy.
- Procedure:
- Subtotal colectomy with end ileostomy and rectal stump.
- Consider IPAA or IRA in appropriate patients.
- Avoid delaying surgery beyond day 7 in non-responders to medical
therapy.
Discharge Planning
- Criteria for discharge:
- Resolution of symptoms.
- Normalizing inflammatory markers.
- Stable oral intake and bowel function.
- Before discharge:
- Initiate maintenance therapy.
- Ensure appropriate follow-up within 2 weeks.
- Provide steroid tapering schedule and DVT prophylaxis if
needed.
- Educate the patient about signs of relapse and when to seek
help.
Maintenance Therapy
After induction of remission, long-term maintenance therapy is
critical to prevent relapse [18].
Options include:
- Thiopurines (azathioprine or 6-mercaptopurine).
- Biologics (infliximab, vedolizumab, ustekinumab).
- Small molecules (tofacitinib, Upadacitinib, ozanimod).
Therapy should be tailored based on patient response, prior
medications, and risk factors.
Steroids should not be used for maintenance due to long-term side
effects.
Special Considerations
- Pregnancy:
- Most medications used in ASUC, including biologics and thiopurines,
are safe in pregnancy.
- Avoid methotrexate and consider risks vs. benefits of
tofacitinib.
- Infections:
- Reactivation of latent infections is a risk with immunosuppressive
therapy.
- Ensure screening for TB, hepatitis B/C, HIV, and varicella.
- Vaccination:
- Patients should be up to date with vaccines, especially before
initiating immunosuppressants.
- Nursing Priorities:
- Monitor fluid balance, skin care, emotional support.
- Educate on mobilization and DVT prevention.
Time line of ASUC management
# Load required packages
library(ggplot2)
library(dplyr)
library(stringr)
# Create timeline data with wrapped text
timeline_data <- data.frame(
Day = c("Day 0", "Day 3", "Day 4-7", "Post-acute"),
Description = c(
"Admit, exclude infection,\nstart IV steroids",
"Assess response; if refractory\n→ salvage therapy",
"Evaluate salvage response;\nescalate or surgery",
"Plan long-term management\nor surgery"
),
Position = 1:4
)
# Create the timeline visualization
ggplot(timeline_data, aes(x = Position, y = 0)) +
# Add timeline
geom_segment(aes(x = 0.5, xend = 4.5, y = 0, yend = 0),
color = "black", linewidth = 2, alpha = 0.7) +
# Add larger phase markers (circles)
geom_point(aes(fill = Day), size = 35, shape = 21, color = "black") +
# Add day labels inside circles
geom_text(aes(label = Day), color = "white", fontface = "bold", size = 5) +
# Add description boxes below circles
geom_label(aes(y = -0.25, label = Description),
fill = "lightyellow", color = "black",
size = 4.5, label.padding = unit(0.6, "lines"),
lineheight = 0.8) +
# Customize colors
scale_fill_manual(values = c("Day 0" = "#4E79A7",
"Day 3" = "#F28E2B",
"Day 4-7" = "#E15759",
"Post-acute" = "#59A14F")) +
# Theme adjustments
theme_void() +
theme(
plot.title = element_text(face = "bold", size = 18, hjust = 0.5, margin = margin(b = 15)),
plot.subtitle = element_text(size = 14, hjust = 0.5, margin = margin(b = 25)),
legend.position = "none",
plot.margin = margin(1, 1, 1.5, 1, "cm")
) +
# Set limits to ensure everything is visible
ylim(-0.4, 0.2) +
xlim(0.5, 4.5) +
# Add titles
labs(title = "ASUC Management Timeline",
subtitle = "Clinical Pathway for Acute Severe Ulcerative Colitis")

Flow chart of management of ASUC
# Load required packages
library(DiagrammeR)
library(htmltools)
# Create the ASUC management flowchart
grViz("
digraph ASUC_Management_Flowchart {
# Graph settings
graph [layout = dot, rankdir = TB, nodesep = 0.5, ranksep = 0.8]
node [fontname = 'Helvetica', fontsize = 12]
edge [color = 'black', arrowhead = vee]
# Define nodes with different shapes for different types of steps
start [label = 'Hospital admission\nSuspect ASUC',
shape = oval, style = filled, fillcolor = 'lightblue']
initial_workup [label = 'Initial work up:\n- CBC, CRP/ESR, Albumin\n- Stool studies (C.Diff)\n- Flex sigmoidoscopy (Mayo score)\n- Abdominal X ray',
shape = box, style = filled, fillcolor = 'lightyellow']
start_steroids [label = 'Start IV steroids\n0.8mg/kg',
shape = box, style = filled, fillcolor = 'lightyellow']
assess_day3 [label = 'Assess response day 3\nOxford score',
shape = diamond, style = filled, fillcolor = 'lightcoral']
responder [label = 'Responder to steroid',
shape = box, style = filled, fillcolor = 'palegreen']
non_responder [label = 'Non-responder to steroid',
shape = box, style = filled, fillcolor = 'lightpink']
transition [label = 'Transition to oral steroid\nGradual tapering of steroid\nMaintenance therapy: biological or\nsmall molecules',
shape = box, style = filled, fillcolor = 'lightyellow']
salvage [label = 'Start salvage therapy:\nInfliximab or Cyclosporine',
shape = box, style = filled, fillcolor = 'lightyellow']
reassess [label = 'Reassess day 4-7',
shape = diamond, style = filled, fillcolor = 'lightcoral']
salvage_responder [label = 'Responder',
shape = box, style = filled, fillcolor = 'palegreen']
salvage_non_responder [label = 'Non-responder',
shape = box, style = filled, fillcolor = 'lightpink']
maintenance [label = 'Plan maintenance therapy:\nbiological or small molecules',
shape = box, style = filled, fillcolor = 'lightyellow']
surgery [label = 'Urgent surgical referral',
shape = box, style = filled, fillcolor = 'lightyellow']
# Define edges
start -> initial_workup
initial_workup -> start_steroids
start_steroids -> assess_day3
assess_day3 -> responder
assess_day3 -> non_responder
responder -> transition
non_responder -> salvage
salvage -> reassess
reassess -> salvage_responder
reassess -> salvage_non_responder
salvage_responder -> maintenance
salvage_non_responder -> surgery
}
")
# Add some CSS styling for better appearance
tags$style(HTML("
.node oval {
fill: lightblue;
}
.node box {
fill: lightyellow;
}
.node diamond {
fill: lightcoral;
}
.node note {
fill: lightgrey;
}
"))
References
- Taylor K, Gibson PR. Crohn's Disease and Ulcerative Colitis: From
Epidemiology and Immunobiology to a Rational Diagnostic and Therapeutic
Approach, C. Baumgart, Daniel (Eds), 2017.
- Truelove SC, Witts LJ. Cortisone in ulcerative colitis; final report
on a therapeutic trial. Br Med J. 1955;2(4947):1041-1048. doi:10.1136/bmj.2.4947.1041
- Matsuoka K, et al. Impact of cytomegalovirus on outcomes in acute
severe ulcerative colitis: a retrospective observational study. BMJ Open
Gastroenterol. 2023;10(1):e001048. doi:10.1136/bmjgast-2022-001048
- Spinelli, A.; Bonovas, S.; Burisch, J.; Kucharzik, T.; Adamina, M.;
Annese, V.; Bachmann, O.; Bettenworth, D.; Chaparro, M.;
Czuber-Dochan,W.; et al. ECCO guidelines on therapeutics in ulcerative
colitis: Surgical treatment. J. Crohn’s Colitis 2022, 16,179–189
- Singh D.M., Midha V., Mahajan R., Kaur K., Singh D., Sood A. DOP44
Tofacitinib versus corticosteroids for induction of remission in
moderately active ulcerative colitis (ORCHID): A prospective randomised
open-label pilot study. J. Crohns Colitis. 2023;17((Suppl.
S1)):i111–i112. doi: 10.1093/ecco-jcc/jjac190.0084
- Sahu P., Kedia S., Vuyyuru S.K., Bajaj A., Markandey M., Singh N.,
Singh M., Kante B., Kumar P., Ranjan M., et al. Randomised clinical
trial: Exclusive enteral nutrition versus standard of care for acute
severe ulcerative colitis. Aliment. Pharmacol. Ther. 2021;53:568–576.
doi: 10.1111/apt.16249
- Vuyyuru, S.K.; Nardone, O.M.; Jairath, V. Predicting outcome after
acute severe ulcerative colitis: A contemporary review and areas for
future research. J. Clin. Med. 2024, 13, 4509
- Amiot A, Seksik P, Meyer A, Stefanescu C, Wils P, Altwegg R, et
al. Top-down infliximab plus azathioprine versus azathioprine alone in
patients with acute severe ulcerative colitis responsive to intravenous
steroids: a parallel, open-label randomised controlled trial, the ACTIVE
trial. Gut. 2024;73(5):857–66. doi:10.1136/gutjnl-2024-33328
- Gergely, M.; Prado, E.; Deepak, P. Management of refractory
inflammatory bowel disease. Curr. Opin. Gastroenterol. 2022,
38,347–357
- Chen, J.H.; Andrews, J.M.; Kariyawasam, V.; Moran, N.; Gounder, P.;
Collins, G.; IBD Sydney Organisation and the Australian Inflammatory
Bowel Diseases Consensus Working Group. Review article: Acute severe
ulcerative colitis: Evidence-based consensus statements. Aliment.
Pharmacol. Ther. 2016, 44, 127–144
- Singh, S.; Allegretti, J.R.; Siddique, S.M.; Terdiman, J.P. AGA
technical review on the management of moderate to severe ulcerative
colitis. Gastroenterology 2020, 158, 1465–1496.e1417
- Chao CY, et al. High-Dose Infliximab Rescue Therapy for Hospitalized
Acute Severe Ulcerative Colitis Does Not Improve Colectomy-Free
Survival. Dig Dis Sci. 2019;64(5):1380-1385. doi:10.1007/s10620-018-5398-0
- Gisbert, J.P.; García, M.J. Rescue therapies for steroid-refractory
acute severe ulcerative colitis: A review. J. Crohn’s Colitis 2023, 17,
972–994
- Williams, J.; Alam, M.F.; Alrubaiy, L.; Clement, C.; Cohen, D.;
Grey, M.; Hilton, M.; Hutchings, H.A.; Longo, M.; Morgan, J.;et
al. Comparison of infliximab and ciclosporin in steroid-resistant
ulcerative colitis: Pragmatic randomised trial and economic evaluation
(CONSTRUCT). Health Technol. Assess. 2016, 20, 1–32
- Fleming P, et al. Systematic review and meta-analysis of third-line
salvage therapy with infliximab or cyclosporine in severe ulcerative
colitis. Inflamm Bowel Dis. 2016;22(5):1156-1161. doi:10.1097/MIB.0000000000000735
- Ganzleben, I.; Geppert, C.; Osaba, L.; Hirschmann, S.; Nagel, A.;
Gluck, C.; Hoffman, A.; Rath, T.; Nagore, D.; Neurath, M.F.; et
al.Successful cyclosporin and ustekinumab combination therapy in a
patient with severe steroid-refractory ulcerative colitis. Ther. Adv.
Gastroenterol. 2020, 13, 1756284820954112.
- Damianos, J.A.; Osikoya, O.; Brennan, G. Upadacitinib for acute
severe ulcerative colitis: A systematic review. Inflamm. Bowel Dis.2024,
Iza191
- Harbord, M.; Eliakim, R.; Bettenworth, D.; Karmiris, K.; Katsanos,
K.; Kopylov, U.; Kucharzik, T.; Molnár, T.; Raine, T.; Sebastian, S.;et
al. Third European evidence-based consensus on diagnosis and management
of ulcerative colitis. Part 2: Current management. J. Crohn’s Colitis
2017, 11, 769–784
Chapter 12: Ileal Pouch–Anal Anastomosis
Ileal pouch anal anastomosis (IPAA) is the most common operation done
for ulcerative colitis. There are two techniques to perform the
IPAA:
Hand-Sewn IPAA with Mucosectomy: In this method,
a mucosectomy is first performed by removing the rectal mucosa from the
distal rectum to the dentate line, leaving a short muscular cuff. The
ileal pouch is then anastomosed to the anal transitional zone using a
hand-sewn technique, approximating the full thickness of the pouch wall
to the internal sphincter and the remaining anal mucosa.
Stapled IPAA: In the stapled approach, the ileal
pouch is anastomosed to the distal rectal mucosa just above the anal
transitional zone, preserving a short rectal cuff. The remaining rectal
mucosa requires regular follow-up due to the potential risk of
developing dysplasia or cancer.
Pouchitis
Background
- Pouchitis is the most common complication after IPAA affecting, 48%
of patients within the first 2 years, and up to 80% at some point after
surgery [3].
- The 5-year and 10-year risks of colectomy after IPAA are 7.0% and
9.6%, respectively [3].
- Pouchitis has a significant impacte on patient quality of life and
is associated with a high healthcare cost burden [3].
- 17% of patients may develop chronic pouchitis with a
relapsing–remitting course [3].
- 10% of patients may go on to develop Crohn’s-like disease of the
pouch [3].
- The most frequent symptoms of pouchitis are increased number of
liquid stools, urgency, abdominal cramping and pelvic discomfort. Fever
and bleeding are rare.
- Clinical symptoms do not necessarily correlate with endoscopic or
histologic findings and routine pouchoscopy after clinical remission is
not required [3].
- Of note, after an initial postoperative adjustment period, patients
with IPAA can expect to have an average of 4–8 bowel movements per day
and 1–2 at night [3].
- Pouchoscopy is recommended in patients with frequent recurrent
pouchitis, inadequate response to antibiotics, atypical symptoms, or
suspected Crohn’s-like disease of the pouch [3].
- Biomarkers such as fecal calprotectin and lactoferrin are not
routinely used in clinical practice for pouch disorders [3].
- Risk factors for pouchitis include extensive ulcerative colitis,
backwash ileitis [4], extraintestinal manifestations (especially primary
sclerosing cholangitis) [5], being a non-smoker [6], regular NSAID use
[7], and the presence of perinuclear neutrophil cytoplasmic antibodies
(p-ANCA) [8].
- The diagnosis of pouchitis requires the presence of symptoms,
together with characteristic endoscopic and histological abnormalities
[3].
- Alternative causes such as mechanisms that impair pouch emptying
including strictures at the ileoanal anastomosis or stoma takedown site,
evacuation disorders like nonrelaxing pelvic floor dysfunction, and
infectious etiologies such as Clostridioides difficile infection,
especially in antibiotic-refractory cases should be considered when
evaluating pouch dysfunction symptoms [3].
Cufftitis
Background
- Cuffitis, particularly following double-stapled IPAA, results in
inflammation of the residual cuff of rectal mucosa, may lead to pouch
dysfunction [9].
- It often presents with symptoms that mimic pouchitis.
- Rectal bleeding is more commonly associated with cuffitis than with
pouchitis [9].
Endoscopic evaluation of the pouch
The Pouchitis Disease Activity Index (PDAI) has been developed to
standardize diagnostic criteria and assess the severity of pouchitis
[9].
### Type of Inflammatory Pouch Disorders
library(knitr)
library(kableExtra)
# Create the data frame
pouch_types <- data.frame(
Term = c("Intermittent Pouchitis", "Chronic Antibiotic-Dependent Pouchitis", "Chronic Antibiotic-Refractory Pouchitis", "Crohn’s-like Disease of the Pouch"),
Definition = c("Isolated, infrequent episodes that resolve with therapy or spontaneously, followed by extended periods (> months) of normal function.",
"Recurrent episodes that respond to antibiotics but relapse shortly after stopping (days/weeks). Requires recurrent/continuous therapy.",
"Relapsing or continuous symptoms with inadequate response to standard antibiotic therapy, requiring escalation to other therapies.",
"Defined by features like fistulae (>12mo post-op), strictures, or pre-pouch ileitis. Often coexists with pouchitis.")
)
# Generate the formatted table
kable(pouch_types, format = "html", col.names = c("Term", "Definition"), align = c('l', 'l'), caption = "Table 1: Types of Inflammatory Pouch Disorders") %>%
kable_styling(bootstrap_options = c("striped", "hover", "condensed"), full_width = F) %>%
column_spec(1, bold = TRUE, width = "5cm") %>%
column_spec(2, width = "15cm")
Table 1: Types of Inflammatory Pouch Disorders
Term
|
Definition
|
Intermittent Pouchitis
|
Isolated, infrequent episodes that resolve with therapy or
spontaneously, followed by extended periods (> months) of normal
function.
|
Chronic Antibiotic-Dependent Pouchitis
|
Recurrent episodes that respond to antibiotics but relapse shortly after
stopping (days/weeks). Requires recurrent/continuous therapy.
|
Chronic Antibiotic-Refractory Pouchitis
|
Relapsing or continuous symptoms with inadequate response to standard
antibiotic therapy, requiring escalation to other therapies.
|
Crohn’s-like Disease of the Pouch
|
Defined by features like fistulae (>12mo post-op), strictures, or
pre-pouch ileitis. Often coexists with pouchitis.
|
library(ggplot2)
library(dplyr)
# Create data
complication_data <- data.frame(
Complication = c("Pouchitis (by 2 yrs)", "Pouchitis (lifetime)", "Chronic Pouchitis", "Crohn's-like Disease", "Colectomy (5-yr risk)", "Colectomy (10-yr risk)"),
Percentage = c(48, 80, 17, 10, 7.0, 9.6),
Group = c("Pouchitis", "Pouchitis", "Pouchitis", "Pouchitis", "Colectomy", "Colectomy")
)
# Order the factors for the plot
complication_data$Complication <- factor(complication_data$Complication,
levels = complication_data$Complication[order(complication_data$Percentage)])
# Create the plot
ggplot(complication_data, aes(x = Complication, y = Percentage, fill = Group)) +
geom_col(width = 0.7) +
geom_text(aes(label = paste0(Percentage, "%")), hjust = -0.1, size = 3.5, fontface = "bold") +
coord_flip() + # Makes horizontal bars for easier reading
scale_fill_manual(values = c("Colectomy" = "steelblue", "Pouchitis" = "salmon")) +
labs(title = "Prevalence and Risk of Complications after IPAA",
subtitle = "Based on referenced clinical data [3]",
x = "",
y = "Percentage of Patients (%)",
fill = "Complication Type") +
theme_minimal() +
theme(legend.position = "top") +
expand_limits(y = c(0, 85)) # Adjust to make room for labels

Treatment Principles in Pouch Disorders
- The primary treatment goal in pouchitis is resolution of
symptoms.
- Endoscopic and/or histologic healing is not considered a critical
treatment target due to limited supporting data.
- Asymptomatic patients with endoscopic evidence of pouch inflammation
may not require treatment.
- In patients presenting with atypical symptoms of pouchitis,
inadequate response to conventional treatment, or recurrent symptoms
despite therapy, clinicians should consider alternative causes beyond
classic inflammatory pouch disorders [3].
AGA Guideline Recommendations
- Primary Prevention of Pouchitis
- In patients with UC who undergo IPAA, the AGA makes no
recommendation in favor of, or against the use of probiotics for primary
prevention of pouchitis.
- In patients with UC who undergo IPAA, the AGA suggests against using
antibiotics for the primary prevention of pouchitis.
- Treatment of Pouchitis
- In patients with infrequent symptoms:
- Using antibiotics for treatment of pouchitis. Ciprofloxacin and/or
metronidazole are the preferred antibiotics for treatment of
pouchitis.
- The typical duration of antibiotic therapy is 2–4 weeks.
- An approach using a combination of antibiotics may be more effective
in patients who do not respond to single-antibiotic therapy.
- Alternative antibiotic regimens, such as oral vancomycin, may be
considered in patients who do not respond to the initial course of
antibiotics or have allergies or intolerance to ciprofloxacin and/or
metronidazole.
- No recommendation in favor of, or against the use of probiotics for
the treatment of pouchitis.
- In patients with recurrent episodes of pouchitis that
respond to antibiotics, the AGA suggests using probiotics for preventing
recurrent pouchitis.
- Chronic Antibiotic-Dependent Pouchitis
- Use chronic antibiotic therapy to treat recurrent pouchitis.
- Lowest effective dose of antibiotics (e.g., ciprofloxacin 500 mg
daily or 250 mg twice daily) with intermittent gap periods (such as
approximately 1 week per month), or cyclical antibiotics may be
considered to decrease risk of antimicrobial resistance.
- Endoscopic evaluation of the pouch with confirmation of inflammation
and ruling out alternative etiologies in patients with recurrent
pouchitis.
- Use advanced immunosuppressive therapies to treat recurrent
pouchitis.
- May be considered as an alternative to prolonged antibiotic use,
particularly in patients with antibiotic intolerance or when concerns
about long-term antibiotic risks are present.
- Advanced immunosuppressive therapies approved for treatment of UC or
CD, including TNF–a antagonists (i.e., infliximab, adalimumab,
golimumab, certolizumab pegol), vedolizumab, ustekinumab, risankizumab,
ozanimod, tofacitinib, and upadacitinib.
- Advanced immunosuppressive therapies that patients have used before
colectomy may be reconsidered.
- Chronic Antibiotic-Refractory Pouchitis
- Use advanced immunosuppressive therapies.
- Immunosuppressive therapies approved for treatment of UC or CD.
- Vedolizumab is the only advanced therapy to date that has received
regulatory approval from the European Medicines Agency for this
indication. (EARNEST trial)
- Advanced immunosuppressive therapies that patients have used before
colectomy may be reconsidered.
- Endoscopic evaluation of the pouch with confirmation of inflammation
and ruling out alternative etiologies in patients with recurrent
pouchitis.
- Use corticosteroids.
- Controlled ileal-release budesonide is preferred.
- For short-term (<8–12 weeks).
- No recommendation in favor of, or against the use of
mesalamine.
- Crohn’s-like Disease of the Pouch
- Endoscopic evaluation of the pouch to confirm Crohn’s-like disease
of the pouch.
- Use corticosteroids.
- Controlled ileal-release budesonide is preferred.
- For short-term (<8 weeks).
- Use advanced immunosuppressive therapies.
- Immunosuppressive therapies approved for treatment of UC or CD.
- Advanced immunosuppressive therapies that patients have used before
colectomy may be reconsidered.
- Treatment of Cufftitis
- Use therapies that have been approved for the treatment of UC.
- First line: topical therapies such as (mesalamine suppositories,
corticosteroid suppositories, or corticosteroid ointment).
- Refractory cuffitis: immunosuppressive therapies approved for
treatment of UC may be used, including TNF-α antagonists, vedolizumab,
ustekinumab, risankizumab, ozanimod, tofacitinib, and upadacitinib.
library(DiagrammeR)
# Define the graph using DOT language
grViz("
digraph pouchitis_algorithm {
# Global graph settings
graph [layout = dot, rankdir = TB, fontname = Helvetica, nodesep = 0.5]
node [shape = box, style = 'rounded, filled', fillcolor = 'lightgrey', fontname = Helvetica, width = 3.5, height = 0.8]
edge [fontname = Helvetica, fontsize = 10]
# Nodes (Decision and Action points)
start [label = 'Patient presents with\nsymptoms of pouchitis', shape = oval, fillcolor = 'lightblue']
a1 [label = 'Evaluate for alternative causes:\nC. diff, stricture, cuffitis, CD']
d1 [label = 'Symptoms resolve?', shape = diamond, fillcolor = 'lightyellow']
a2 [label = 'Diagnose Pouchitis\n(PDAI, symptoms, endoscopy)']
d2 [label = 'Episode Type?', shape = diamond, fillcolor = 'lightyellow']
t1 [label = 'Infrequent Episode\n→ 2-4 wk antibiotics\n(Cipro/Metro)']
t2 [label = 'Chronic Antibiotic-DEPENDENT\n→ Chronic/cyclical abx\n→ Consider advanced therapy']
t3 [label = 'Chronic Antibiotic-REFRACTORY\n→ Advanced therapy\n(Vedolizumab has EMA approval)']
t4 [label = 'Crohn\\'s-like Disease\n→ Advanced therapy\n→ Short-term budesonide']
end [label = 'Symptom Control', shape = oval, fillcolor = 'lightblue']
# Edges (The flow)
start -> a1
a1 -> d1
d1 -> a2 [label = 'No']
d1 -> end [label = 'Yes (e.g., C. diff cured)']
a2 -> d2
d2 -> t1 [label = 'Infrequent']
d2 -> t2 [label = 'Dependent']
d2 -> t3 [label = 'Refractory']
d2 -> t4 [label = 'Crohn\\'s-like']
t1 -> end
t2 -> end
t3 -> end
t4 -> end
# Force a straight line from d1 to end
{rank = same; d1; end}
}
")
References
- Steele SR, Hull TL, Read TE, Saclarides TJ, Senagore AJ, Whitlow CB,
editors. The ASCRS Textbook of Colon and Rectal Surgery. 4th ed. Cham:
Springer; 2022.
- Glover R. Twists and Turns of J-Pouch Recovery [Internet]. United
Ostomy Association; 2022 Feb 28
- Barnes EL, Agrawal M, Syal G, Ananthakrishnan AN, Cohen BL, Haydek
JP, et al. AGA clinical practice guideline on the management of
pouchitis and inflammatory pouch disorders. Gastroenterology.
2024;166(1):59-85. doi:10.1053/j.gastro.2023.10.015.
- Schmidt CM, Lazenby AJ, Hendrickson RJ, Sitzmann JV. Preoperative
terminal ileal and colonic resection histopathology predicts risk of
pouchitis in patients after ileoanal pull-through procedure. Ann Surg.
1998;227(5):654-62.
- Lohmuller JL, Pemberton JH, Dozois RR, Weaver AL, Kelly KA.
Pouchitis and extraintestinal manifestations of inflammatory bowel
disease after ileal pouch-anal anastomosis. Ann Surg.
1990;211(6):622–9.
- Merrett MN, Mortensen N, Kettlewell M, Jewell DP, Warren BF. Smoking
may prevent pouchitis in patients with restorative proctocolectomy for
ulcerative colitis. Gut. 1996;38(3):362–4
- Achkar JP, Al-Haddad M, Lashner BA, et al. Differentiating risk
factors for acute and chronic pouchitis. Clin Gastroenterol Hepatol
2005;3:60–6
- Fleshner PR, Vasiliauskas EA, Kam LY, et al. High level perinuclear
antineutrophils cytoplasmic antibody (pANCA) in ulcerative colitis
patients before colectomy predicts the development of chronic pouchitis
after ileal pouch-anal anastomosis. Gut 2001;49:671–7.
- European Crohn’s and Colitis Organisation (ECCO). European
evidence-based consensus on the management of ulcerative colitis:
current management. J Crohns Colitis. 2008;2(1):63–92.
- Quinn KP, Lightner AL, Faubion WA, Raffals LE. A Comprehensive
Approach to Pouch Disorders. Inflamm Bowel Dis. 2019 Feb
21;25(3):460-471. doi: 10.1093/ibd/izy267. PMID: 30124882.
Chapter 13: Navigating Medical Therapy in IBD: Pre-Biologic Workup,
Pharmacologic Profiles, Side Effects, and Drug Monitoring
Pre-biologic workup:
Stool cultures, 3x Ova and parasites on consecutive days, C. diff
screen |
Patients presenting with diarrhea |
|
QuantiFERON-TB Gold In-Tube assay (preferred) or tuberculin skin
test. |
All patients |
If the screening test for latent tuberculosis is indeterminate or
positive, a chest radiograph is obtained, refer to an infectious disease
specialist for further evaluation. |
Hepatitis B surface antigen (HbsAg), hepatitis B surface antibody,
and hepatitis B core antibody (anti-HBc). |
All patients |
Patients with serologic evidence of hepatitis B virus (HBV)
infection (HbsAg-positive) are at risk for HBV reactivation if they
receive immunosuppressive therapy – consult hepatology. If only anti-HBc
positive and HbsAg negative, monitoring is advised. |
Hepatitis C virus (HCV) antibody |
All patients |
For patients with HCV antibodies positive, we obtain HCV RNA. If HCV
RNA is detected, - refer the patient to a hepatologist for consideration
of treatment. Chronic HCV infection is not a contraindication for
immunosuppressive therapy. |
HIV screening (baseline) |
All patients |
Refer to ID if positive |
Chest Xray |
Must for suspected or + screening for Latent TB |
|
Lipid profile |
Pre JAK inhibitors use |
At baseline and 12 weeks after therapy initiation |
Verify pregnancy status (prior to initiating therapy) |
Pre JAK inhibitors use |
|
Zoster vaccine -Shingrix |
Pre JAK inhibitors use |
First Dose: Administered at Month 0. Second Dose: Administered
between 2 to 6 months after the first dose. |
Eye exam for macular edema |
Pre Ozanimod use |
For Patient with history of DM, or uveitis or eye symptoms |
ECG |
Pre Ozanimod use |
Looking for heart block |
Pharmacologic Profiles: Mechanism and Dosage
Sulfasalazine/ Mesalamine |
5-ASA (5-aminosalicylic acid): Anti-inflammatory effect on colonic
epithelium, blocks prostaglandin and leukotriene synthesis. |
UC: Mesalamine: 2.4–4.8 g/day orally (UC only) Sulfasalazine: 2–4
g/day orally (in divided doses) |
Azathioprine |
Purine analog → inhibits DNA/RNA synthesis → reduces T & B cell
proliferation. |
1.5–2.5 mg/kg/day orally |
Mercaptopurine |
Active metabolite of azathioprine; inhibits purine synthesis,
suppresses lymphocyte proliferation. |
1–1.5 mg/kg/day orally |
Methotrexate |
Folic acid antagonist → inhibits dihydrofolate reductase → reduces
DNA synthesis, especially in activated T cells. |
Crohn’s: 15–25 mg once weekly (IM or SC) UC: Not recommended |
Infliximab |
Anti-TNF-α monoclonal antibody → reduces inflammation via TNF
blockade. |
UC: 5 mg/kg IV at weeks 0, 2, 6, then every 8 weeks Crohn’s: Same
dosing, sometimes up to 10 mg/kg if loss of response or TDM directed
-SUBQ after IV induction therapy: 120 mg every 2 weeks starting at week
10 or next scheduled IV dose. |
Adalimumab |
Fully human anti-TNF-α monoclonal antibody |
UC: 160 mg SC at week 0, 80 mg at week 2, then 40 mg every other
week Crohn’s: Same induction and maintenance regimen |
Vedolizumab |
Anti-α4β7 integrin monoclonal antibody → gut-selective
anti-inflammatory effect by inhibiting lymphocyte trafficking. |
UC and Crohn’s: 300 mg IV at weeks 0, 2, 6, then every 8 weeks
-SUBQ: 108 mg once every 2 weeks beginning after at least 2 IV
infusions; administer in place of next scheduled IV dose and then every
2 weeks thereafter. |
Ustekinumab |
IL-12/23 monoclonal antibody → blocks inflammatory cytokine
signaling. |
Crohn’s: ≤55 kg: IV: 260 mg as single dose >55 kg to 85 kg: IV:
390 mg as single dose >85 kg: IV: 520 mg as single dose Then 90 mg SC
every 8 weeks UC: Same regimen, FDA approved since 2019 |
Risankizumab |
IL-23 inhibitor → inhibits downstream inflammatory signaling. |
Crohn’s: 600 mg IV at weeks 0, 4, 8; Maintenance: 180 mg SC q8w |
Ozanimod (not available in KSA up to 2025) |
S1P receptor modulator → prevents lymphocyte egress from lymph
nodes, reducing gut inflammation. |
UC: Start 0.23 mg/day titrated to 0.92 mg/day Crohn’s: Not
approved |
Upadacitinib |
JAK1-selective inhibitor → inhibits pro-inflammatory cytokine
signaling. |
UC: 45 mg daily for 8 weeks, then 15–30 mg/day Crohn’s: 45 mg daily
for 12 weeks; then 15–30 mg/day |
Tofacitinib |
Pan-JAK inhibitor (JAK1/3 > JAK2) → blocks cytokine-mediated
inflammation. |
UC: 10 mg BID for 8 weeks then 5–10 mg BID Crohn’s: Not
approved |
Medications Side effects
These are the most common side effects of each medication. Please
note that the incidence percentages are approximate and may vary based
on individual factors and study populations. For a Full list of side
effects, refer to the FDA medication web site.
Sulfasalazine |
Oligospermia |
33% |
Reversible |
|
Headache |
25% |
N/A |
|
Nausea |
20% |
N/A |
|
Abdominal pain |
15% |
N/A |
|
Rash |
10% |
N/A |
|
Fever |
5% |
N/A |
|
Blood dyscrasias |
Rare |
Agranulocytosis, aplastic anemia, hemolytic anemia, leukopenia
usually develop within the first 3 months of treatment |
|
Delayed hypersensitivity reactions |
Rare |
Usually occur 1 to 8 weeks after initiation |
|
Interstitial fibrosis |
Rare |
Can 4 years after starting |
Mesalamine |
Diarrhea |
12% |
(Intolerance syndrome) |
|
Headache |
10% |
N/A |
|
Abdominal pain |
9% |
N/A |
|
Flatulence |
8% |
N/A |
|
Rash |
7% |
N/A |
|
Interstitial fibrosis |
Rare |
Can occur more than 4 years after starting |
Prednisone |
Weight gain |
50% |
Yes |
|
Hypertension |
30% |
Yes |
|
Hyperglycemia |
20% |
Yes |
|
Insomnia |
15% |
Yes |
|
Mood swings |
10% |
Yes |
Azathioprine |
Leukopenia |
15% |
Dose dependent |
|
Nausea |
12% |
No |
|
Pancreatitis |
5% |
No |
|
Hepatotoxicity |
5% |
Dose dependent |
|
Fever |
4% |
No |
Methotrexate |
Stomatitis |
20% |
Dose dependent |
|
Hepatotoxicity |
15% |
Dose dependent |
|
Leukopenia |
10% |
Dose dependent |
|
Nausea |
10% |
Dose dependent |
|
Fatigue |
8% |
Dose dependent |
Infliximab |
Infusion reactions |
20% |
N/A |
|
Upper respiratory infections |
10% |
N/A |
|
Headache |
10% |
N/A |
|
Abdominal pain |
8% |
N/A |
|
Rash |
6% |
N/A |
Adalimumab |
Injection site reactions |
15% |
N/A |
|
Upper respiratory infections |
12% |
N/A |
|
Rash |
10% |
N/A |
|
Headache |
8% |
N/A |
|
Nausea |
7% |
N/A |
Vedolizumab |
Nasopharyngitis |
13% |
N/A |
|
Headache |
12% |
N/A |
|
Arthralgia |
10% |
N/A |
|
Nausea |
9% |
N/A |
|
Fever |
8% |
N/A |
Ustekinumab |
Nasopharyngitis |
12% |
N/A |
|
Headache |
9% |
N/A |
|
Arthralgia |
8% |
N/A |
|
Nausea |
7% |
N/A |
|
Injection site reactions |
5% |
N/A |
Risankizumab |
Upper respiratory infections |
10% |
N/A |
|
Headache |
8% |
N/A |
|
Fatigue |
7% |
N/A |
|
Injection site reactions |
6% |
N/A |
|
Back pain |
5% |
N/A |
Ozanimod |
Bradycardia |
10% |
N/A |
|
Hypertension |
8% |
N/A |
|
Macular edema |
5% |
N/A |
|
Liver enzyme elevation |
5% |
N/A |
|
Respiratory infections |
5% |
N/A |
Upadacitinib |
Upper respiratory infections |
15% |
N/A |
|
Elevated liver enzymes |
10% |
N/A |
|
Nausea |
8% |
N/A |
|
Hypertension |
7% |
N/A |
|
Acne |
10-16% |
Dose dependent |
|
Thrombosis |
Rare (case reports) |
Do not initiate therapy in patients with an absolute lymphocyte |
|
Hematologic toxicity |
Rare |
count <500/mm3, ANC <1,000/mm3, or hemoglobin <8 g/dL |
Tofacitinib |
Upper respiratory infections |
14% |
N/A |
|
Headache |
12% |
N/A |
|
Diarrhea |
10% |
N/A |
|
Nasopharyngitis |
8% |
N/A |
|
Hypertension |
6% |
N/A |
|
Diarrhea |
10% |
N/A |
|
Nasopharyngitis |
8% |
N/A |
|
Thrombosis |
Rare (case reports) |
|
Lymphoma risk and TNF / Thiopurines
Compared to unexposed patients:
- Thiopurine Monotherapy: An absolute risk difference of 0.42 per
1,000 person-years.
- Anti-TNF Monotherapy: An absolute risk difference of 0.15 per 1,000
person-years.
- Combination Therapy: An absolute risk difference of 0.69 per 1,000
person-years.
While these therapies are associated with an increased relative risk
of lymphoma, the absolute risk remains low. When making treatment
decisions, patients and healthcare providers should carefully weigh
these risks against the benefits, as the advantages of the medication
often outweigh the risks, particularly the risk of lymphoma.
Drug Monitoring (TDM)
Definitions:
Primary loss of response refers to the lack of
an adequate clinical response to a biologic or other therapy during the
initial induction phase of treatment.
Secondary loss of response (sLOR): Patients who
had initially responded after the induction phase, but then started to
develop symptoms of disease activity, suggestive of treatment
failure.
Proactive TDM: is the measurement of drug levels
and anti-drug antibodies, regardless of clinical symptoms.
Reactive TDM: Drug levels are checked only after
a patient experiences worsening symptoms or loss of response.
Up to 30% of IBD patients fail to show an initial response after
the induction period, and up to 50% showing sLOR during the maintenance
phase, especially during the first year.
TDM-guided therapy escalation has been shown to be up to 30% more
cost-effective compared to empiric dose escalation in cases of secondary
loss of response.
Debate exists surrounding the use of TDM, in current practice, it
is mainly used in the reactive setting and in severe complex IBD cases
like fistulizing Crohn’s disease.
In general, two types of drug assays are available.
Drug-intolerant assays detect anti-drug antibodies (ADAs) only when drug
levels are low or absent. In contrast, drug-tolerant assays can detect
ADAs even in the presence of therapeutic drug concentrations and appear
to be more consistent.
Guidelines summary on TDM:
AGA (2017) |
In adults with active IBD treated with anti-TNF agents, the AGA
suggests reactive therapeutic drug monitoring to guide treatment
changes. (Conditional recommendation, very low quality of
evidence.) |
In adult patients with quiescent IBD treated with anti-TNF agents,
the AGA makes no recommendation regarding the use of routine proactive
therapeutic drug monitoring. (Knowledge gap) |
ECCO (2024) |
There is insufficient evidence to recommend the use of proactive
therapeutic drug monitoring. compared with reactive therapeutic drug
monitoring or standard of care. when using anti-TNF agents (weak
recommendation, very low-quality evidence). (Consensus: 100%) |
|
Saudi Arabia Consensus (2023) |
Recommend To use TDM before optimizing the dose, switching to
another anti-TNF agent, or switching to a different class, such as
ustekinumab or vedolizumab. |
No recommendations were made regarding proactive drug
monitoring. |
TDM targets
Infliximab |
>5 |
>10 |
|
Adalimumab |
>7.5 |
>10 |
|
Vedolizumab and Ustekinumab |
No data supports the use of TDM. |
|
|
- In patients with perianal Crohn’s disease (CD), closure of the
fistula have been consistently shown to be achieved by higher trough
level of infliximab (≥ 10 μg/mL).
TDM
- Low drug levels and low ADA or low titer: Increase
the dose and decrease the interval, Consider adding
immunomodulator.
- Low drug levels and high ADA levels: Switch to
another if having active TNF disease consider non TNF shift.
- Mechanistic failure:
References
- Ben-Horin, S., & Chowers, Y. (2011). Review article: Loss of
response to anti-TNF treatments in Crohn"s disease. Alimentary
Pharmacology & Therapeutics, 33, 987–995. https://doi.org/10.1111/j.1365-2036.2011.04612.x
- Billioud, V., Sandborn, W. J., & Peyrin-Biroulet, L. (2011).
Loss of response and need for adalimumab dose intensification in Crohn"s
disease: A systematic review. American Journal of Gastroenterology, 106,
674–684. https://doi.org/10.1038/ajg.2011.60
- Steenholdt, C., Brynskov, J., Thomsen, O. Ø., Munck, L. K.,
Fallingborg, J., Christensen, L. A., Pedersen, G., Kjeldsen, J.,
Jacobsen, B. A., Oxholm, A. S., Kjellberg, J., Bendtzen, K., &
Ainsworth, M. A. (2014). Individualised therapy is more cost-effective
than dose intensification in patients with Crohn"s disease who lose
response to anti-TNF treatment: A randomised, controlled trial. Gut, 63,
919–927. https://doi.org/10.1136/gutjnl-2013-305279
- Yarur, A. J., Kanagala, V., Stein, D. J., Czul, F., Quintero, M. A.,
Agrawal, D., Patel, A., Best, K., Fox, C., Idstein, K., & Abreu, M.
T. (2017). Higher infliximab trough levels are associated with perianal
fistula healing in patients with Crohn"s disease. Alimentary
Pharmacology & Therapeutics, 45, 933–940. https://doi.org/10.1111/apt.13970
Chapter 14: Optimizing Preoperative Management of Inflammatory Bowel
Disease (IBD): A Multidisciplinary Approach is the best care.
By: Heba Al Farhan
Effective preoperative management in inflammatory bowel disease (IBD)
is central to improving surgical outcomes, minimizing complications, and
enhancing recovery. Research indicates that patients undergoing surgery
for IBD, such as Crohn’s disease (CD) and ulcerative colitis (UC),
benefit from a comprehensive, multidisciplinary approach to
optimization. This detailed review explores strategies with supporting
evidence, including nutritional, pharmacological, and psychological
interventions, alongside relevant statistics and data.
1. The Importance of Preoperative Planning
Surgical intervention is required in approximately 20-30% of UC
patients and up to 70-80% of CD patients during their lifetime.
Preoperative management plays a critical role in reducing risks like
infections, anastomotic failure, and prolonged recovery periods. Studies
consistently demonstrate that addressing modifiable factors
preoperatively can lower complication rates significantly. For instance,
Molina Arriero et al. (2023) reported a 35% reduction in surgical
complications among patients who underwent structured preoperative
programs compared to those receiving standard care (3).
2. Nutritional Optimization
2.1 Screening and Assessment
Malnutrition is prevalent in IBD patients undergoing surgery, with
rates as high as 50-60% in Crohn's disease cases and 20-40% in
ulcerative colitis. Malnourished patients are 2.5 times more likely to
experience postoperative infections and complications (6). Tools like
the Malnutrition Universal Screening Tool (MUST) and serum albumin
measurements (threshold <3.0 g/dL) are recommended for nutritional
evaluation (7).
2.2 Interventions
2.2.1 Exclusive Enteral Nutrition (EEN):
Research highlights EEN as an effective strategy to reduce surgical
complications. Vanderstappen et al. (2024) found that EEN reduced
postoperative infection rates by up to 40%, improved albumin levels by
20-30%, and shortened hospital stays by 25% relative to patients
receiving standard oral nutrition (6,8).
2.2.2 Parenteral Nutrition (PN):
PN is especially beneficial for patients with severe obstruction or
inflammation. According to Efron et al. (2007), preoperative PN in
malnourished patients reduced the incidence of anastomotic leaks to 8%
from 15% in non-optimized individuals (7). However, PN was associated
with an 8-12% increased risk of catheter-related infections (5).
2.2.3 Micronutrient Supplementation:
Correcting deficiencies in vitamin D, iron, and zinc is essential for
recovery. Forbes et al. (2017) emphasized that addressing anemia,
prevalent in 30-40% of IBD patients, is critical, as untreated anemia
doubles postoperative complication risks (10). Table 1:
Anemia |
30-40% of IBD patients (Forbes et al., 2017) |
- Men: Hb <13 g/dL. - Non pregnant women: Hb <12 g/dL. -
Pregnant women: Hb <11 g/dL. |
- Untreated anemia doubles the risk of postoperative complications,
including infections and delayed wound healing. - Contributes to fatigue
and poor quality of life. |
- Iron Deficiency Anemia (IDA): Intravenous iron (e.g., ferric
carboxymaltose) for active disease or severe deficiency. - ACD:
Erythropoiesis-stimulating agents in severe cases. - Vitamin B12/Folate
Deficiency: Parenteral vitamin B12 (1000 mcg IM monthly) and oral/IV
folate. |
Vitamin D Deficiency |
Up to 70% of IBD patients |
- Deficiency: 25(OH)D <20 ng/mL - Insufficiency: 20-30 ng/mL |
- Low bone density - Worsened inflammation - Potentially detrimental
immunomodulatory effects. |
- High-dose supplementation (50,000 IU weekly) until levels
normalize >30 ng/mL. - Maintenance dose of 1,000-2,000 IU daily. -
Regularly monitor levels. |
Zinc Deficiency |
Common, especially in those with chronic diarrhea or high-output
fistulas |
Serum zinc <70 mcg/dL |
- Impaired wound healing - Increased risk of infections - Worsened
diarrhea |
- Mild Deficiency: Oral zinc sulfate (220 mg daily for 2-3 weeks). -
Severe Deficiency: IV zinc for cases with severe malabsorption or
prolonged PN. |
Magnesium Deficiency |
Seen in chronic diarrhea, ileostomies, or high-output fistulas |
Varies (monitor levels closely) |
Worsened diarrhea, muscle weakness, and potential cardiac
arrhythmias. |
- Use oral magnesium chloride or sulfate. - Severe cases may require
IV magnesium sulfate. |
Calcium Deficiency |
Common in patients on corticosteroids or with low bone density |
Varies (typically assessed with serum calcium) |
- Weak bones and increased risk of fractures in long term. |
- 1,000-1,500 mg calcium daily along with vitamin D supplementation
to improve absorption. |
Selenium Deficiency |
Rare, seen in severe malnutrition or prolonged PN |
Varies (often assessed indirectly) |
- Poor antioxidant defense and immune function. |
Selenium supplementation via oral or IV routes based on
severity. |
3. Pharmacological Considerations
Balancing immunosuppression to maintain disease control while
minimizing postoperative complications is a complex challenge. (Table
2)
3.1.1 Biologics:
Studies indicate that the risk of postoperative infections in
patients using biologics does not exceed 15% if the timing of the last
dose aligns with the mid-treatment interval (4,8). Vanderstappen et
al. (2024) recommend administering the last dose at least four weeks
before surgery to avoid adverse outcomes (6).
3.1.2 Corticosteroids:
Chronic steroid use (>20 mg/day of prednisone or equivalent) is
strongly linked to complications, including a 20-30% increased risk of
wound infections and a 15% higher chance of anastomotic leaks. Tapering
corticosteroids to below 10 mg/day preoperatively has been shown to
halve these risks (7, 2).
Infliximab |
Plan surgery mid-dose interval; monitor closely in high-risk or
malnourished patients. |
Low if timed appropriately (≤15%) |
Continue if last dose is 4–8 weeks before surgery |
|
Adalimumab |
Administer last dose 2 weeks pre-surgery to minimize infection risk
while maintaining disease control. |
Low if timed appropriately (≤10–15%) |
Continue; adjust dose timing |
|
Vedolizumab |
4–6 weeks before surgery |
Minimal (<5%) |
Continue; no significant impact on wound healing |
Focuses primarily on gut-specific immunity; generally safe for
perioperative use. |
Ustekinumab |
8–12 weeks before surgery |
Minimal if preoperative interval is respected (<5%) |
Continue; administer last dose 8-12 weeks before surgery |
Adjust timing carefully due to long dosing interval; assess disease
stability. |
IL-23 Inhibitors (Skyrizi, Tremfya) |
8–12 weeks before surgery |
Minimal if timed appropriately (<5%) |
Continue; schedule based on individual patient response. |
Long dosing intervals may require adjustment and ensure disease
stability. Monitor infection risk. |
JAK Inhibitors (Tofacitinib, Upadacitinib) |
7 days before surgery |
Moderate to high (10–25% risk of infections, VTE risk higher with
tofacitinib) |
Temporarily discontinue 1 week before surgery |
Increased infection risk, particularly with tofacitinib. Strong
risk-benefit evaluation required; monitor for VTE in high-risk
patients. |
Corticosteroids |
Reduce to ≤10 mg/day prednisone equivalent; ideally discontinue
before surgery |
High (20–30% risk of wound infections, 15% chance of anastomotic
leaks) |
Taper to lowest effective dose; avoid abrupt discontinuation to
prevent adrenal insufficiency |
Chronic use increases infection risk; patients on prolonged
corticosteroid therapy may require perioperative stress dosing. |
3.2 Venous Thromboembolism (VTE) Prophylaxis
IBD patients face a threefold increased risk of VTE compared to the
general population. Fiorindi et al. (2021) demonstrated that extended
prophylaxis with low-molecular-weight heparin (LMWH) reduced VTE events
from 10% to under 3%, underscoring its importance in perioperative
protocols (9).
Low Molecular Weight Heparin (LMWH) |
- Enoxaparin: 40 mg subcutaneously once daily - Dalteparin: 5,000 IU
subcutaneously once daily |
- Recommended by gastroenterological societies (e.g., ACG) for all
hospitalized IBD patients, including preoperative cases - Focus on
high-risk or hospitalized individuals |
- Significant reduction in VTE events (OR 0.27, NNT = 35) - Slight
increase in major bleeding risk (OR 2.02, NNH = 114) |
Extended LMWH |
LMWH continued up to 28 days post-surgery |
- Suggested for patients undergoing major abdominal surgeries or
those with additional risk factors such as immobilization or disease
severity |
- Effective in preventing post-discharge VTE - Requires
patient-specific risk-benefit analysis due to bleeding concerns |
Unfractionated Heparin (UFH) |
Dosage dependent on patient-specific factors as per medical
protocols |
- Considered in patients where LMWH is contraindicated or
unavailable |
- Potentially less effective than LMWH - Associated with higher
rates of heparin-induced thrombocytopenia compared to LMWH |
Direct Oral Anticoagulants (DOACs) |
Dose varies depending on the type (e.g., apixaban, rivaroxaban) |
- Not universally recommended for routine use in preoperative IBD
due to limited evidence specifically in this population |
- Evidence around safety and efficacy in IBD remains limited - May
be considered in selected cases under careful monitoring |
Mechanical Prophylaxis |
Use of compression stockings or pneumatic devices |
- Often used as an adjunct, especially in patients at risk of
bleeding or contraindications to pharmacologic prophylaxis |
- No direct effect on reducing VTE mortality - Beneficial as part of
a multifactorial approach to reducing clot formation during
hospitalization |
3.3 Antibiotics and Bowel Preparation
Cohen et al. (2024) stressed the importance of preoperative bowel
preparation and antibiotic prophylaxis, which reduced surgical site
infections by 20-30% in patients undergoing colorectal surgery. However,
routine mechanical bowel preparation alone did not significantly improve
outcomes and is now selectively used for fecal load reduction in
obstructive cases (4).
4. Psychological Preparation
Psychological factors, including heightened anxiety and depression,
affect up to 40-50% of IBD patients preoperatively. This psychological
distress is linked to poor compliance with preoperative optimization
measures and slower recovery. Hazel et al. (2025) reported that
inclusion of psychologists in multidisciplinary teams (MDTs) reduced
anxiety scores by 30% and postoperative recovery times by two to three
days (2). Peer-support programs like those offered by the Crohn’s &
Colitis Foundation provide an additional layer of emotional support and
education (5).
5. Multidisciplinary Approach
Effective preoperative care necessitates an MDT approach,
incorporating gastroenterologists, colorectal surgeons, dietitians,
psychologists, and even physiotherapists in some cases. Molina Arriero
et al. (2023) found that multidisciplinary care resulted in a 50%
reduction in postoperative readmission rates and a significant decrease
in average hospital stay duration (3).
6. Structured Prehabilitation Programs
Prehabilitation, which combines physical, nutritional, and
psychological optimization, is increasingly recognized as a critical
component of preoperative care. Fiorindi et al. (2021) revealed that
patients enrolled in structured prehabilitation programs experienced 20%
fewer complications, 15% shorter hospital stays, and 40% fewer ICU
admissions compared to non-participants (9,1).
7. Timing and Recommendations for Resuming Biologic Agents After IBD
Surgery
Recent studies and guidelines provide clear recommendations on the
timing of resuming biologic agents after surgery for inflammatory bowel
disease. These recommendations focus on ensuring patient safety while
minimizing the risk of disease recurrence. Here's a summary of the
current evidence and expert guidance:
Anti-TNF Agents |
Within 2 to 4 weeks post-surgery (18, 19, 20) |
- Strongly recommended for preventing postoperative recurrence. -
Safe to use perioperatively; detectable drug levels are not associated
with increased infections. - Earlier initiation is encouraged for
high-risk patients. |
- Timing remains debatable, especially in patients with
complications. |
Vedolizumab |
Within 2 to 4 weeks post-surgery (17, 20, 21) |
- Effective in reducing recurrence rates. - Recommended for
high-risk patients. - Supported by recent trials like REPREVIO. |
- Limited data on long-term outcomes compared to anti-TNF agents. -
Tailored approach required for patients with prior biologic
failures. |
JAK Inhibitors |
As early as 3 to 5 days post-surgery (if no complications) (17) |
- Limited data available. - May provide rapid disease control
without increasing complication rates. |
- Lack of robust evidence makes their use controversial. - Not
widely recommended in current guidelines. |
General Considerations |
Timing depends on patient recovery and absence of
complications/infections (17, 20) |
- Risk of infection must be ruled out before resuming biologics. -
Monitoring tools like fecal calprotectin and endoscopy are essential. -
Emerging tools like intestinal ultrasound are promising for non-invasive
monitoring. |
- A tailored approach is critical for better outcomes. - Newer
biologics and small molecules (e.g., IL-12/23 inhibitors, S1P
modulators) are not yet fully studied in postoperative settings. |
8. Emerging Research and Future Directions
Vanderstappen et al. (2024) highlighted gaps in global
standardization of preoperative optimization protocols, especially
regarding nutritional strategies and biologics management (6). While
current interventions show strong efficacy, future research should focus
on refining and individualizing prehabilitation models, particularly for
high-risk patients with severe inflammation or comorbid conditions like
diabetes and cardiovascular diseases.
9. Conclusions
Optimizing preoperative care for IBD patients is a nuanced,
multifactorial process involving nutritional supplementation, judicious
pharmacological management, and psychological support. Evidence suggests
that addressing malnutrition, managing biologics with precision, and
employing extended VTE prophylaxis can reduce complications by up to
50%. Emerging MDT-driven models and structured prehabilitation programs
have the potential to revolutionize preoperative care further.
Standardizing and tailoring these approaches will significantly improve
patient outcomes for those navigating the complexities of IBD-related
surgery.
References
- Syed, H., et al. (2024). “Peri-Operative Optimization of Patients
with Crohn’s Disease.” Current Gastroenterology Reports, 26(5), 125–136.
Available at PMC11081987.
- Hazel, K., et al. (2025). “Preoperative Optimization for Elective
Surgery in Crohn’s Disease: A Narrative Review.” Journal of Clinical
Medicine, 14(5), 1576. Available at PMC11899995.
- Molina Arriero, G., et al. (2023). “Preoperative Optimization of
Patients with Inflammatory Bowel Disease: A Multicentre Study of EIGA
Group.” Journal of Crohn's and Colitis, 17(Supplement_1), i356.
- Cohen, B., et al. (2024). “IBD: Avoiding Postoperative Infections
and Complications.” Cleveland Clinic Digestive Insights. Available at
Cleveland Clinic.
- Crohn’s & Colitis Foundation. “Preparing for IBD Surgery.”
Available at Crohn’s & Colitis Foundation.
- Vanderstappen, J., et al. (2024). “Preoperative Optimization: Review
on Nutritional Assessment and Strategies in IBD.” Current Opinion in
Pharmacology, 77, 102475.
- Efron, J. E., et al. (2007). “Preoperative Optimization of Crohn's
Disease.” Clinics in Colon and Rectal Surgery, 20(3), 209-216.
- Mayer, L., et al. (2023). “Nutritional Support in IBD Surgery.”
Journal of Parenteral and Enteral Nutrition, 47(5), 600-610.
- Fiorindi, C., et al. (2021). “Prehabilitation in IBD Surgery: A
Systematic Review.” Journal of Crohn's and Colitis, 15(10),
1678-1689.
- Forbes, A., et al. (2017). “Anemia in IBD: Pathogenesis and
Management.” Inflammatory Bowel Diseases, 23(11), 1950-1960.
- National Institute for Health and Care Excellence (NICE). (2023).
“Crohn's disease: management.” NICE guideline NG129.
- American Gastroenterological Association (AGA). (2022). “AGA
Clinical Practice Guideline on the Management of Crohn's Disease.”
- European Crohn's and Colitis Organisation (ECCO). (2023). “ECCO
Guidelines on the Management of Crohn's Disease.”
- World Health Organization (WHO). (2023). “International
Classification of Diseases (ICD-11).”
- Centers for Disease Control and Prevention (CDC). (2023).
“Inflammatory Bowel Disease (IBD).”
- Singh, S., et al. (2022). “Postoperative Management of Crohn's
Disease.” Gastroenterology, 162(4), 1100-1115.
- Sands, B. E., et al. (2023). “Postoperative Management of Ulcerative
Colitis.” Gastroenterology, 164(2), 200-215.
- D'Haens, G., et al. (2021). “Biologic Therapy in Postoperative
Crohn's Disease.” Journal of Crohn's and Colitis, 15(7), 1100-1110.
- Rutgeerts, P., et al. (2020). “Prevention of Postoperative
Recurrence of Crohn's Disease.” Gastroenterology, 158(5),
1200-1210.
- Feagan, B. G., et al. (2022). “Vedolizumab for the Prevention of
Postoperative Crohn's Disease Recurrence.” New England Journal of
Medicine, 386(19), 1800-1810.
- Colombel, J. F., et al. (2021). “Ustekinumab for the Prevention of
Postoperative Crohn's Disease Recurrence.” Lancet, 398(10300),
600-610.
Chapter 15: Monitoring and Follow-Up of IBD Patients
Essential tips for management success
By: Heba Al Farhan
Inflammatory Bowel Disease (IBD), encompassing Crohn's disease and
ulcerative colitis, is a chronic and progressive condition characterized
by intestinal inflammation that may lead to significant complications,
such as strictures, fistulas, perforations, and colorectal cancer.
Effective management hinges on timely therapeutic interventions and
meticulous disease monitoring to tailor treatment adjustments, achieve
sustained remission, and prevent progression (1,14).
The development of the STRIDE II framework has revolutionized IBD
care, emphasizing a treat-to-target approach that prioritizes long-term
therapeutic goals like clinical remission, mucosal healing, and
biomarker normalization over symptom management (1). Recommendations
from the European Crohn’s and Colitis Organisation (ECCO) and the
American College of Gastroenterology (ACG) provide complementary
guidance, underscoring the necessity of personalized, evidence-based
monitoring strategies that combine clinical assessments, biomarkers,
imaging, and endoscopic evaluations (2,3).
This chapter provides a comprehensive overview of IBD monitoring,
integrating evidence from STRIDE II, ECCO, and ACG, alongside insights
into the latest advances shaping the future of personalized care.
1. Monitoring Strategies in IBD
Optimal disease monitoring involves a multimodal, integrated approach
to evaluate IBD comprehensively. This ensures accurate detection of
active inflammation, guides therapeutic decisions, and assesses
treatment response.
A selection of monitoring solutions is available:
- Endoscopy
- Imaging modalities (CTE, MRE and IUSB)
- Biomarkers (FC and CRPs)
- Clinical symptoms
1.1. Clinical Symptom Assessment
1.1.1. Role in IBD Monitoring
Symptom-based assessments rely on indices like the Crohn’s Disease
Activity Index (CDAI) for Crohn’s disease and the Mayo Clinic Score
(MCS) for ulcerative colitis. These tools focus on patient-reported
outcomes such as stool frequency, rectal bleeding, abdominal pain, and
health status to give preliminary disease activity estimates (3).
1.1.2. Strengths and Limitations
While commonly used, symptom-based measurements often lack
correlation with objective inflammation markers. Studies reveal that
over 30% of IBD patients classified as being in clinical remission
through these indices still exhibit mucosal inflammation detectable via
endoscopy (3). This highlights the need for additional monitoring
modalities to verify disease activity accurately.
STRIDE II Integration
Under STRIDE II, clinical remission is defined as the absence of
symptoms, achieved without corticosteroids, and confirmed through
additional parameters such as biomarkers or imaging. Combining
subjective and objective indicators ensures greater reliability in
activity assessments (1).
1.2. Endoscopic Monitoring
1.2.1. Precision of Endoscopy
Endoscopy is considered the
Chapter 16: Nutrition in Inflammatory Bowel Disease (IBD)
Outpatient Management
Dietary Regimens:
Several dietary regimens have shown benefit for patients with IBD,
targeting either induction, maintenance of remission, or both. Examples
are outlined in table-1 below:
Both |
Crohn’s Disease Exclusion Diet (CDED)1 |
Both |
Specific Carbohydrate Diet (SCD)2 |
Both |
Mediterranean diet3 |
Both |
Plant-based diet4 |
Both |
Paleo Auto-Immune Protocol (AIP)2 |
Induction |
CD-TREAT5 |
Both |
Low-emulsifier or carrageenan diets6 |
Both |
Low sulfur diet7 |
Maintenance |
Low-fat diet8 |
Maintenance |
Low-meat diet (FACES)9 |
Both |
Food exclusion based on IgG antibodies10 |
Both |
Novel ulcerative colitis exclusion diet (UCED)11 |
The European Society for Clinical Nutrition and Metabolism (ESPEN)
guidelines note that no universal “IBD diet” can be recommended to
induce or maintain the remission. (12) However, the American
Gastroenterological Association (AGA) advises that, barring
contraindications, patients with IBD should adopt a Mediterranean diet.
This diet is characterized by a high intake of fresh fruits, vegetables,
monounsaturated fats, complex carbohydrates, and lean proteins, while
minimizing ultra-processed foods, added sugars, and salt. This approach
supports overall health and well-being. (13)
When to screen patients for malnutrition?
Malnutrition in IBD can exacerbate disease, increase postoperative
complications, and prolonged hospital stays. It is associated with
higher risks of venous thromboembolism, infections, and mortality. All
patients should be screened for malnutrition at diagnosis and regularly
thereafter. Reliance solely on serum albumin or BMI is discouraged;
these markers lack specificity and sensitivity for nutritional status.
Instead, a multidisciplinary approach involving dietitian counselling is
recommended to identify and manage nutrition-related disorders promptly.
(12,13.14,15,16)
Micronutrients screening:
Patients with IBD are at risk for micronutrient deficiencies due to
diarrhoea, malabsorption, intestinal failure, and reduced dietary
intake. Since most of the micronutrients are acute-phase reactants,
screening should be performed at least annually and ideally during
clinical remission. (12) Screening tests should include:
- Vitamins A, E, K, D, B1, B2, B6, B12
- Zinc, selenium, copper, manganese
- Folate and iron studies
Anaemia in IBD
Anaemia is a common extraintestinal manifestation of IBD, affecting
6–74% of IBD patients, and it is more common in Crohn’s disease than
ulcerative colitis. It often results from a combination of iron
deficiency anaemia (IDA) and chronic disease (ACD). For patients in
remission or mild disease, screening should be obtained every 6-12
months, while patients with active disease such measurements should be
obtained at least every 3 months. (17)
Iron deficient anemia (IDA) |
Inactive disease: serum Ferritin <30 µg/L Active disease: serum
Ferritin up to 100 µg/L |
Anemia of chronic disease (ACD) |
serum ferritin >100 µg/L and transferrin saturation <20% in
case of active disease |
Combined deficiencies |
ferritin levels fall between 30 and 100 µg/L. |
Treatment:
Oral iron supplementation is the preferred option in patients with
mild anaemia and inactive disease. Intravenous iron is reserved for
those with active disease, severe anaemia (haemoglobin <100 g/L),
intolerance to oral iron, and in patients who need
erythropoiesis-stimulating agents. (12)
Vitamin D and calcium supplementation:
Patients with active IBD or those on steroids should have serum
calcium and vitamin D monitored every 3–6 months. In remission,
monitoring can be extended to every 6–12 months. Supplementation helps
prevent low bone mineral density, osteopenia, and osteoporosis.
Osteopenia and osteoporosis should be managed according to current
osteoporosis guidelines. (12)
B12 deficiency:
Vitamin B12 deficiency is prevalent in Crohn’s disease, particularly
after resection of >30 cm of the distal ileum. (18) Diagnosis
involves low serum cobalamin levels (<148 pM) and elevated biomarkers
like homocysteine (>15 mM) or methylmalonic acid (>270 mM). The
diagnosis of clinical B12 deficiency further requires macrocytosis
and/or neurological symptoms. (19,12)
Treatment
Patients with clinical deficiency should receive 1000 mg of vitamin
B12 by intramuscular injection every other day for a week and then every
month for life (20). Patients with more than 20 cm of ileum resected,
should receive 1000 mg of vitamin B12 prophylactically also every month
and indefinitely. Oral therapy as a less explored alternative (21) For
now, patients must be advised for parenteral formulas.
Role of Probiotics
Probiotics such as Escherichia coli Nissle 1917 and VSL#3 can induce
remission in mild to moderate ulcerative colitis and maintain remission
in pouchitis. However, their role in Crohn’s disease is limited and not
recommended for maintenance therapy. (12)
Nutritional Interventions:
Oral Nutrition Supplements (ONS) is the First-line support when
dietary intake is insufficient, but it is a supportive measure used in
addition to normal food. (12)
Enteral Nutrition (Tube Feeding): Preferred over parenteral nutrition
(PN) unless contraindicated (e.g., bowel obstruction, high-output
fistula). (12)
Parenteral Nutrition (PN): Reserved for patients who’s oral and or
tube feeding are proved to be insufficient to meet their requirement,
and for patients with severe gastrointestinal dysfunction or
complications. Bowel obstruction where there is no possibility to place
a feeding tube beyond the obstruction, short bowel syndrome, and
presence of anastomotic leak or high out-put fistula are some of the
common indications for PN in patients with IBD. (12)
Exclusive Enteral Nutrition (EEN):
EEN is effective for inducing remission in Crohn’s disease,
particularly in children. It also serves as a steroid-sparing bridge
therapy and optimizes nutritional status pre-surgery. (12)
Mechanisms of EEN:
- Modulates gut microbiota and luminal metabolites (e.g: short chain
fatty acids (SCFA)).
- Reduces luminal antigens (e.g., carrageenan).
- Enhances gut barrier function. (22)
Figure-1: How to start, monitor and stop EEN? (22)
Managing some of EEN Side Effects:
- Dislike of formula: Adjust flavour/type based on patient
preference.
- Diarrhoea: Modify formula osmolarity.
- Constipation: Ensure adequate fluid intake; consider magnesium
supplementation.
- Headache: Hydration and limited caffeine allowance (e.g., black
coffee/tea) if withdrawal is suspected. (22)
Special consideration: patients with symptomatic stricture
Strategies such as thorough chewing, cooking, and processing these
foods into a softer consistency can enhance dietary variety and fiber
intake. (13)
References:
- Levine, A. et al. Crohn’s disease exclusion diet plus partial
enteral nutrition induces sustained remission in a randomized controlled
trial. Gastroenterology 157, 440–450.e8 (2019).
- Lewis, J. D. et al. A randomized trial comparing the specific
carbohydrate diet to a Mediterranean diet in adults with Crohn’s
disease. Gastroenterology 161, 837–852.e9 (2021).
- Konijeti, G. G. et al. Efficacy of the autoimmune protocol diet for
inflammatory bowel disease. Inflamm. Bowel Dis. 23, 2054–2060
(2017).
- Chiba, M. et al. Lifestyle-related disease in Crohn’s disease:
relapse prevention by a semi-vegetarian diet. World J. Gastroenterol.
16, 2484–2495 (2010).
- Svolos, V. et al. Treatment of active Crohn’s disease with an
ordinary food-based diet that replicates exclusive enteral nutrition.
Gastroenterology 156, 1354–1367.e6 (2019).
- Yao, C. K., Muir, J. G. & Gibson, P. R. Review article: insights
into colonic protein fermentation, its modulation and potential health
implications. Aliment. Pharmacol. Ther. 43, 181–196 (2016).
- Sandall, A. M. et al. Emulsifiers impact colonic length in mice and
emulsifier restriction is feasible in people with Crohn’s disease.
Nutrients 12, 2827 (2020).
- Fritsch, J. et al. Low-fat, high-fiber diet reduces markers of
inflammation and dysbiosis and improves quality of life in patients with
ulcerative colitis. Clin. Gastroenterol. Hepatol. 19, 1189–1199.e30
(2021).
- Albenberg, L. et al. A diet low in red and processed meat does not
reduce rate of Crohn’s disease flares. Gastroenterology 157, 128–136.e5
(2019).
- Jian, L. et al. Food exclusion based on IgG antibodies alleviates
symptoms in ulcerative colitis: a prospective study. Inflamm. Bowel Dis.
24, 1918–1925 (2018).
- Sarbagili-Shabat, C. et al. A novel UC exclusion diet and
antibiotics for treatment of mild to moderate pediatric ulcerative
colitis: a prospective open-label pilot study. Nutrients 13, 3736
(2021).
- Bischoff SC et al. A. ESPEN guideline on Clinical Nutrition in
inflammatory bowel disease. Clin Nutr. 2023 Mar;42(3):352-379. doi:
10.1016/j.clnu.2022.12.004. Epub 2023 Jan 13.
- Hashash, Jana G. et al. AGA Clinical Practice Update on Diet and
Nutritional Therapies in Patients With Inflammatory Bowel Disease:
Expert Review. Gastroenterology, Volume 166, Issue 3, 521 - 532
- Yerushalmy-Feler A, Galai T, Moran-Lev H, Ben-Tov A, Dali-Levy M,
Weintraub Y, et al. BMI in the lower and upper quartiles at diagnosis
and at 1-year follow-up is significantly associated with higher risk of
disease exacerbation in pediatric inflammatory bowel disease. Eur J
Pediatr 2021;180:21e9.
- Ladd MR, Garcia AV, Leeds IL, Haney C, Oliva-Hemker MM, Alaish S, et
al. Malnutrition increases the risk of 30-day complications after
surgery in pediatric patients with Crohn disease. J Pediatr Surg
2018;53:2336e45.
- McLoughlin RJ, McKie K, Hirsh MP, Cleary MA, Aidlen JT. Impact of
nutritional deficiencies on children and young adults with Crohn’s
disease undergoing intraabdominal surgery. J Pediatr Surg
2020;55:1556e61.
- xel U Dignass et al. the European Crohn’s and Colitis Organisation
[ECCO], European Consensus on the Diagnosis and Management of Iron
Deficiency and Anaemia in Inflammatory Bowel Diseases, Journal of
Crohn’s and Colitis, Volume 9, Issue 3, March 2015.
- Yasueda A et al. The effect of Clostridium butyricum MIYAIRI on the
prevention of pouchitis and alteration of the microbiota profile in
patients with ulcerative colitis. Surg Today 2016;46:939e49.
- Benjamin JL et al. Randomised, double-blind, placebo-controlled
trial of fructooligosaccharides in active Crohn’s disease. Gut
2011;60:923e9
- Hallert C, Kaldma M, Petersson BG. Ispaghula husk may relieve
gastrointestinal symptoms in ulcerative colitis in remission. Scand J
Gastroenterol 1991;26:747e50.
- Fernandez-Banares F, et al. Randomized clinical trial of Plantago
ovata seeds (dietary fiber) as compared with mesalamine in maintaining
remission in ulcerative colitis. Spanish Group for the Study of Crohn’s
Disease and Ulcerative Colitis (GETECCU). Am J Gastroenterol
1999;94:427e33.
- Fitzpatrick JA et al. Dietary management of adults with IBD - the
emerging role of dietary therapy. Nat Rev Gastroenterol Hepatol. 2022
Oct;19(10):652-669. doi: 10.1038/s41575-022-00619-5. Epub 2022 May
16.
Chapter 19: Infections in Inflammatory Bowel Disease
Introduction:
Inflammatory bowel disease (IBD), comprising Crohn's disease (CD) and
ulcerative colitis (UC), is a chronic inflammatory condition of the
gastrointestinal tract. The pathogenesis of IBD involves a complex
interplay of genetic predisposition, immune dysregulation, environmental
factors, and alterations in the gut microbiota (1). Patients with IBD
are at an increased risk of infections due to the underlying immune
dysregulation, the use of immunosuppressive therapies, and the potential
for disruption of the gut barrier (2,3).
Epidemiology of infections in IBD
Infections are a significant cause of morbidity and mortality in
patients with IBD. Studies have shown that patients with IBD are at a
higher risk of both opportunistic and community-acquired infections
compared to the general population (4). The risk of infection is further
amplified using immunosuppressive medications, such as corticosteroids,
thiopurines, methotrexate, and biologic therapies (2).
A large population-based study found that patients with IBD had a 1.5
to 2-fold increased risk of serious infections compared to the general
population (4). The most common infections in IBD patients include
respiratory infections, urinary tract infections, and gastrointestinal
infections. Opportunistic infections, such as tuberculosis (TB),
cytomegalovirus (CMV), and herpes zoster, are also more prevalent in
this population (5).
Risk Factors for Infections in IBD
Several factors contribute to the increased risk of infections in
patients with IBD:
- Immunosuppressive Therapy: Corticosteroids, immunomodulators and
biologics are independently associated with increased infection risk.
Combination therapies, such as anti-tumour necrosis factor (TNF) with
thiopurines, further amplify this risk. Recent data suggest lower rates
of serious infections with Jak inhibitors and IL-23 inhibitors compared
with anti-TNF. Vedolizumab, due to its gut selectivity, has shown a
lower rate of non-gastrointestinal infections but may increase the risk
of enteric infections such as Clostridioides difficile (C. difficile)
(5).
Aminosalicylates |
None |
No systemic effects |
Topical Steroids |
Low |
Systemic effects with oral budesonide >6 mg/day |
Systemic Steroids |
Moderate-Severe |
Doses ≥20 mg for >2 weeks, associated with increased risk of
bacterial and fungal infections |
Vedolizumab |
Selective |
Gut-selective; increased risk for intestinal infections |
Methotrexate |
Moderate-Severe |
Doses >20 mg/week, lower dose is considered low
immunosuppression |
Thiopurines |
Moderate-Severe |
Doses >3 mg/kg/day (azathioprine) or >1.5 mg/kg/day (6-MP),
lower dose is considered low immunosuppression, increase risk of viral
infections |
Anti-TNF Agents |
Moderate-Severe |
Higher risk of mycobacterial, bacterial and fungal infections, risk
further increase with combination therapy |
Calcineurin inhibitors (Cyclosporine, tacrolimus) |
Moderate-Severe |
Rarely used because of increased risk of serious infections among
other adverse effect |
JAK Inhibitors |
Moderate-Severe |
Increased risk of viral infections, including herpes zoster |
IL 23 inhibitors |
Moderate -Severe |
Data suggest lower risk of serios infections with IL-23 and JAK
inhibitors compared with anti-TNF |
- Disease severity/activity: Patients with severe or active IBD are
more susceptible to infections due to the systemic inflammation and
mucosal barrier disruption, which can facilitate bacterial
translocation. A large cohort study of IBD patients found that patient
with active disease were more than twice as likely to develop infections
compared to those with quiescent disease (6).
- Malnutrition: Malnutrition is common in patients with IBD,
particularly in those with CD (7.8). Nutritional deficiencies, such as
low levels of vitamin D and zinc, can impair immune function and
increase susceptibility to infections (9).
- Surgery and Hospitalization: Patients with IBD often require
surgical interventions and hospitalizations, which are associated with
an increased risk of nosocomial infections, including C. difficile and
surgical site infections (10).
- Age and Comorbidities: Older patients and those with comorbid
conditions, such as diabetes or chronic kidney disease, are at a higher
risk of infections. Additionally, the presence of extraintestinal
manifestations of IBD, such as primary sclerosing cholangitis, may
further increase infection risk (5,11,12).
Common Infections in IBD
- Viral Infections:
- Cytomegalovirus (CMV): CMV colitis is a serious concern in treatment
refractory IBD and is associated with higher rate of hospitalization and
colectomy within 12 months. The gold standard for diagnosis involves
endoscopic biopsy with immunohistochemistry (IHC) and/or tissue PCR.
Treatment includes antiviral therapy (e.g., ganciclovir,
valganciclovir), immunosuppressive therapy should not be discontinued
except in cases of disseminated infection (13).
- Varicella zoster virus (VZV): Varicella zoster reactivation is more
common in IBD patient, particularly those on JAK inhibitors (14).
Treatment includes acyclovir, valacyclovir or famcyclovir. Intravenous
treatment should be considered for severe and complicated infection.
Vaccination with the recombinant zoster vaccine is recommended for IBD
patients (5).
- Herpes simplex virus (HSV): Primary or recurrent oral and genital
herpes may be more frequent and severe in immunosuppressed patient. HSV
can cause severe mucocutaneous or systemic infections, including
keratitis, retinitis, encephalitis, pneumonia, esophagitis and colitis.
Although no vaccine available for HSV, routine prophylaxis with
antiviral therapy should be considered for patient with frequent
recurrent attacks (5).
- Hepatitis virus (A, B, C):
- Hepatitis A virus (HAV): Causes acute hepatitis; vaccination is
recommended for non-immune IBD patient before immunosuppressive
treatment. Post exposure prophylaxis with immunoglobulin and vaccine is
recommended for unvaccinated immunosuppressed patients (15)
- Hepatitis B Virus (HBV): Can cause acute or chronic hepatitis,
cirrhosis, and hepatocellular carcinoma. Reactivation of HBV can occur
with immunosuppression. Screening for HBV is essential before starting
immunosuppressive medications. Treatment with nucleoside/nucleotide
analogs (e.g., tenofovir, entecavir) is recommended for chronic HBV or
as prophylaxis during immunosuppression. Vaccination is recommended for
non-immune patients (16).
- Hepatitis C Virus (HCV): Can cause chronic hepatitis, cirrhosis, and
hepatocellular carcinoma. Reactivation is less common compared to HBV.
Treatment with Direct-acting antivirals (e.g., sofosbuvir, ledipasvir)
for chronic HCV (5).
- Epstein–Barr virus (EBV): In most patients, EBV infection is a
self-limiting or asymptomatic, even in those receiving
immunosuppression. However, EBV is associated with an increased risk of
lymphoma in EBV-negative patients on thiopurines. The Use of thiopurines
in EBV-IgG negative patients should be carefully considered. Rare
complication of primary viral infection in immunosuppressed patients is
hemophagocytic lymph histiocytosis (HLH) and EBV-positive mucocutaneous
ulceration. Discontinuation of immunosuppression is the primary
therapeutic intervention (5).
- Influenza virus: IBD patients have a slightly increased risk of
influenza and are more likely to require hospitalization compared to
non-IBD patients. Steroid were the only medication independently
associated with influenza risk. Annual vaccination is the most effective
prevention method and should be recommended for all patients.
Immunosuppressive therapy should be withheld in severe cases of
influenza (17).
- Human papilloma virus (HPV): Immunosuppressive therapy increase the
risk of persistent HPV infections and cervical cancer. Female IBD
patient on immunosuppressive therapy should undergo annual cervical
cancer screening. HPV vaccination is recommended for both young male and
female patients with IBD (5).
- COVID-19: Most IBD medications, including anti-TNF agents,
vedolizumab, and ustekinumab, do not appear to significantly increase
the risk of severe COVID-19. Steroid and thiopurines have been
associated with increased risk of severe disease in some studies. In
asymptomatic and mild infections, immunosuppressive therapy may be
continued. In moderate to severe COVID-19 temporary cessation may be
considered. Vaccination is recommended for all IBD patients,
prioritizing mRNA or viral vector vaccines. Booster doses are
recommended for immunosuppressed patient due to their potentially
reduced response to the initial vaccine series (18).
- Human immunodeficiency virus (HIV): Cohort studies suggest that HIV
patients have a less severe course of IBD. Screening for HIV is
recommended, especially for high-risk individuals. Immunosuppressives
can be used in patients with stable CD4 counts and undetectable viral
loads. Close monitoring of CD4 counts is recommended (19).
- Mycobacterium infections:
- Mycobacterium TB: there is a significant risk of latent TB (LTBI)
reactivation with biological therapy and JAK inhibitors, particularly
with anti-TNF in combination with immunomodulators. Screening for LTBI
is recommended before starting biological treatment and ideally before
starting any immunosuppression. Recommended screening tests include
Chest x-ray, TB-skin (TST) and/or interferon gamma release assay (IGRA)
depending on epidemiological factors and prior BCG vaccination. Periodic
rescreening is recommended for patient on biological treatment and JAK
inhibitors, especially in endemic areas. Patients with LTBI should
receive prophylactic treatment before starting immunosuppressive
therapy. Standard treatment includes isoniazid, rifampicin and isoniazid
plus rifapentine. Immunosuppressive therapy should be delayed at least
one month after starting LTBI treatment (5).
- Non-TB mycobacterium (NTM): less common than TB but occur in
immunosuppressed IBD patients. Symptoms include fever, fatigue and
weight loss. Diagnosis involves isolation of the organism from cultures
(sputum, blood or tissue biopsies). Treatment involves combination of
antibiotics for prolonged period and may require adjustment or
discontinuation of immunosuppressive therapy (20).
- Bacterial infections:
- Clostridioides difficile (C. diff): IBD patient, particularly those
with colonic involvement, are at high risk of C. diff infections.
Screening for infection is recommended at every disease flare, as
symptoms can mimic or exacerbate an IBD flare. A two-step diagnostic
algorithm is recommended, starting with a highly sensitive test (e.g.,
glutamate dehydrogenase [GDH] or nucleic acid amplification test [NAAT])
followed by highly specific test (e.g., toxin A/B enzyme immunoassay).
Treatments includes oral vancomycin or fidaxomicin. IV metronidazole is
added for severe infections. Recurrent infections are common; treatment
options include vancomycin, fidaxomicin, bezlotoxumab and fecal
microbiota transplantation (21).
- Salmonella: Immunosuppressed IBD patients are at risk of severe
Salmonella enteritidis and Salmonella typhimurium infections, including
bacteremia, osteomyelitis, and septic arthritis. Treatment include
fluroquinolone or third-generation cephalosporins (5).
- Escherichia coli (E. coli) and Other Enteric Pathogens: Disruption
of the gut barrier in IBD increases the risk of infections with enteric
pathogens, including E. coli, Campylobacter, and Shigella. These
infections can mimic or exacerbate IBD flares. Treatment includes
ciprofloxacin or azithromycin (22).
- Listeria monocytogenes: Listeria can cause severe systemic
infections, including meningitis and septicemia, particularly in
patients on anti-TNF therapy. Treatment includes ampicillin or
trimethoprim-sulfamethoxazole (TMP-SMX) (5)
- Pneumococcal Infections: IBD patients, even before starting
immunosuppressive therapy, have an increased risk of invasive
pneumococcal disease (e.g., pneumonia, meningitis). Pneumococcal
vaccination is recommended for all IBD patients (5).
- Legionella pneumophila infection: Immunosuppressed IBD patients,
particularly those on anti-TNF therapy, are at risk of Legionella
pneumonia. Diagnosis involves urinary antigen testing or PCR on
respiratory samples. Treatment includes macrolides or fluroquinolone
(5).
- Nocardial infection: Rare but serious opportunistic infections that
can occur in patients with IBD, particularly those on immunosuppressive
therapies (e.g., Steroid, Anti-TNF agents). Nocardia species are found
in soil and decaying organic matter. Patients with occupational or
recreational exposure to soil (e.g., farmers, gardeners) are at higher
risk. Clinical presentation includes pulmonary, cutaneous, and
disseminated systemic symptoms. Diagnosis involves culture and stain or
PCR of affected body fluids or tissue. First line treatment is TMP-SMX
(5).
- Fungal and parasitic infections:
- Pneumocystis jirovecii infection: An opportunistic fungal pathogen
that causes Pneumocystis pneumonia (PCP), particularly in
immunocompromised patients. Prophylaxis with TMP-SMX should be strongly
considered for patient on triple immunosuppressive therapy, including
steroids, azathioprine, methotrexate and biologics. It may also be
considered for those on double immunosuppressive if one agent is a
calcineurin inhibitor or a combination of high dose steroid, low
lymphocytes count, and JAK inhibitors. Typical presentation includes
respiratory symptom and hypoxia. Chest imaging may show bilateral
interstitial infiltrates or ground-glass opacities. First line treatment
is TMP-SMX. (5).
- Candida albicans: The most common cause of mucosal infections (e.g.,
oral thrush, esophageal candidiasis). Invasive infections are uncommon,
but mortality is high in IBD patients. Steroid and antibiotics use are
the most common risk factors. Treatment includes fluconazole for mucosal
infections and echinocandins or amphotericin B for invasive disease
(23,24).
- Parasitic infections: Parasitic infections in IBD patients can
complicate disease management, mimic IBD symptoms, or exacerbate
underlying inflammation. Geographic and environmental exposure is an
important risk factor.
- Protozoal infections: Giardia lamblia, Entamoeba histolytica,
Cryptosporidium and Cyclospora species can cause diarrhea in IBD
patients.
- Helminths infections: Strongyloidiasis can cause hyperinfection
syndrome in immunocompromised patients, and Schistosomiasis can affect
GI tract and liver.
- Diagnosis is made through stool examination, supported by
serological and molecular testing. Treatment involves antiparasitic
therapy according to the isolated species and may require adjustment of
immunosuppressive therapy in severe cases (5).
References:
- Ananthakrishnan, A. N., Bernstein, C. N., Iliopoulos, D., et
al. (2018). Environmental triggers in IBD: a review of progress and
evidence. Nature Reviews Gastroenterology & Hepatology, 15(1),
39-49.
- Lichtenstein, G. R., Feagan, B. G., Cohen, R. D., et al. (2018).
Serious infection and mortality in patients with Crohn's disease: more
than 5 years of follow-up in the TREAT™ registry. American Journal of
Gastroenterology, 113(4), 481-517.
- Peterson, L. W., & Artis, D. (2015). Intestinal epithelial
cells: regulators of barrier function and immune homeostasis. Nature
Reviews Immunology, 14(3), 141-153.
- Bewtra, M., Kaiser, L. M., TenHave, T., & Lewis, J. D. (2015).
Crohn's disease and ulcerative colitis are associated with elevated
standardized mortality ratios: A meta-analysis. Inflammatory Bowel
Diseases, 21(3), 599-613
- Kucharzik T, Ellul P, Greuter T, Rahier JF, et al (2021). ECCO
Guidelines on the Prevention, Diagnosis, and Management of Infections in
Inflammatory Bowel Disease. J Crohn’s Colitis.;15(6):879-913.
- Michielan A, D'Incà R. Intestinal Permeability in Inflammatory Bowel
Disease: Pathogenesis, Clinical Evaluation, and Therapy of Leaky Gut.
Mediators Inflamm. 2015:628157. 2015 Oct 25. PMID: 26582965.
- Siva, S., Rubin, D. T., Gulotta, G., et al. (2017). Zinc deficiency
is associated with poor clinical outcomes in patients with inflammatory
bowel disease. Inflammatory Bowel Dise
Chapter 20: Risk of Malignancies in IBD
Introduction:
- Inflammatory bowel disease (IBD) refers to chronic, immune-mediated
conditions characterized by recurring and remitting inflammation of the
gastrointestinal tract1.
- Ulcerative colitis (UC) and Crohn’s disease (CD) primarily affect
the luminal tract of the gastrointestinal system. However, their exact
causes remain unclear and stem from a complex interaction of immune
dysregulation, microbial imbalance, and environmental factors in
individuals with a genetic predisposition2.
- Patients with IBD have a higher risk of developing gastrointestinal
cancers, especially colorectal cancer (CRC)3. Individuals with
long-standing UC and Crohn’s colitis (excluding limited proctitis) face
an approximately 2–3-fold increased risk of CRC.
- Risk factors associated with the development of CRC include disease
duration after 8 years from the colitis onset 2,4, the early onset of
IBD in younger individuals, colonic inflammation, the severity of
inflammation, backwash ileitis, strictures, and post-inflammatory polyps
5.
Pathogenesis and Epidemiology:
- IBD patients can develop both sporadic CRC and IBD-CRC6. Sporadic
CRC originates from a dysplastic precursor (usually an adenomatous
polyp).
- It ranks as the third most common cancer in both men and women, with
over 1.9 million new cases reported in 20227.
- The major molecular pathways involved in the development of sporadic
CRC, including chromosomal instability, microsatellite instability
(MSI), and the CpG island methylator phenotype (CIMP), also play a role
in the progression of IBD-CRC8.
- Both types of cancer share common functional driver genes, including
APC, P53, MYC, KRAS, PIK3CA, SMAD4, and ARID1A2. However, IBD-CRC
differs from sporadic CRC in the timing and frequency of genetic
alterations. For instance, APC gene mutations and loss are less common
and occur later in the dysplasia–carcinoma sequence in IBD-CRC.
- P53 mutations and loss are more frequent and likely to happen
earlier 3. Additionally, KRAS and P53 mutations are more prevalent in
patients with IBD-CRC than in those without IBD-related dysplasia,
making them potential biomarkers for IBD-CRC3. Figure 1 explains the
Pathogenesis .9,10
Figure 1: Colitis-associated colorectal cancer (CAC) develops in
patients with long-standing inflammatory bowel disease (IBD), including
ulcerative colitis and Crohn’s colitis. Unlike sporadic CRC, CAC often
follows a chronic inflammation–dysplasia–carcinoma sequence
Risk Factors for Colorectal Cancer in Inflammatory Bowel Disease
(IBD):
The following table2 summarizes key risk factors associated with the
development of colorectal cancer (CRC) in patients with inflammatory
bowel disease.11,12,13,14,15
Endoscopic Surveillance
- Colonoscopy is the gold standard for diagnosing and managing IBD,
therefore, the effectiveness of a colonoscopy relies on two key factors:
a good bowel prep and the ability of the endoscopist to visualize the
entire colon.
- Endoscopic surveillance is recommended for patients with UC that
extend beyond the rectum and CD with colonic involvement of at least
one-third of the colon, beginning 8 years after diagnosis16.
- The main goal of this surveillance is to detect premalignant lesions
that can be removed endoscopically or in early-stage CRC, thereby
improving prognosis and treatment outcomes17.
- Traditionally, standard -Definition white light endoscopy (SD-WLE)
in conjunction with four quadrant non-targeted biopsies every 10
centimeters were used for surveillance about 33 biopsies from the
colonic mucosa are needed to reach close to a 90% confidence that
dysplasia, if present, will be detected18,19. The literature shows that
random biopsies with SD-WLE can detect dysplasia in a small percentage
of patients supporting the shift toward targeted biopsies 20.
- To reduce the risk of CRC, management strategies focus on accurately
assessing each patient’s risk and distinguishing between high-risk and
low-risk individuals, summarized in Table 1 and Table 32.
- Various international societies offer guidelines on techniques and
modalities for surveillance colonoscopy to enhance the effectiveness of
dysplasia detection. Tables 4 and 5 22.
Management of Dysplasia detection in IBD9,21:
- Visible Dysplasia
Visible dysplasia is any lesion seen on endoscopy, either polypoid or
non-polypoid, and includes:
- Lesions identified during surveillance colonoscopy
- Targetable by advanced imaging modalities (HD-WLE, chromoendoscopy,
NBI)
A. Endoscopically Resectable Lesions
- Approach:
- Perform complete endoscopic resection.
- Ensure clear histologic margins and evaluate the surrounding mucosa
for additional dysplasia.
- Surveillance: Repeat colonoscopy at 3–6 months, then annually if no
recurrence.
- Key Points:
- Use high-definition scopes with chromoendoscopy or virtual
chromoendoscopy for inspection.
- If histology confirms LGD or HGD with negative margins, surgery can
be avoided.
B. Non-resectable or Poorly Demarcated Lesions
- Approach:
- Refer for colectomy, especially if:
- Margins are unclear
- Lesion cannot be removed entirely
- Background mucosa has additional dysplasia
- Invisible Dysplasia
Invisible dysplasia refers to dysplasia found on random biopsies with
no corresponding visible lesion on endoscopy.
A. Low-Grade Dysplasia (LGD)
- Approach:
- Confirm pathology with expert GI pathologist.
- Repeat colonoscopy using chromoendoscopy to try to identify the
lesion.
- If still invisible:
- Discuss with patient the risks vs. benefits of surveillance
vs. colectomy.
- Consider colectomy for multifocal LGD, or close surveillance in
selected cases.
- Surveillance Strategy:
- Colonoscopy every 3–6 months for the first year.
- If stable, can extend to yearly.
B. High-Grade Dysplasia (HGD)
- Approach:
- Considered high risk for synchronous colorectal cancer (up to
40%).
- Immediate colectomy is recommended.
Figures 2 and 3(adopted from AGA Clinical Practice Update on
Endoscopic Surveillance and Management of Colorectal Dysplasia in
Inflammatory Bowel Diseases: Expert Review)21 provide classification and
detection of dysplasia in IBD and a structured management approach of
dysplasia in inflammatory bowel disease (IBD) based on visibility,
grade, resectability, and associated risk factors.
Figure2: classification and detection of dysplasia in IBD21
Figure 3: Algorithm: Management of Visible and Invisible Dysplasia
in IBD21
Lymphoma rsik in IBD:
- Lymphoma is a complication of autoimmune disorders, including
Crohn’s disease [CD] and ulcerative colitis [UC]
- In the absence of exposure to anti-tumor necrosis factor [TNF]
and/or thiopurine, the absolute risk of lymphoma in IBD patients is
quite low [0.01% per person-year] and does not seem to exceed that of
the general population24.
- old age [>65 years old], male gender and use of thiopurine were
critical factors associated with an increased risk of incident
lymphoma.25
- patients with IBD receiving thiopurine [azathioprine or
6-mercaptopurine] have a statistically significantly increased risk of
developing lymphoma [hazard ratio = 5.28, p = 0.0007]. see table 6 and 7
summarize risk factors of lymphoma and types associated with
IBD26,27,28
- Epstein–Barr virus [EBV] is significantly involved in lymphoma
development in patients with IBD, accounting for at least 40% when
treated with thiopurine.27
Skin cancer and IBD:
- Nonmelanoma skin cancers (NMSCs), is associated with an increased
risk of NMSCs, more common with the use of immunomodulatory medications
(azathioprine,6-mercaptopurine, antibodies than the use of tumor
necrosis factor [anti-TNF] [eg, infliximab, adalimumab]28.
- The incidence rate of Melanoma Is increasing in IBD patients treated
with anti-TNF29.
- For surveillance, Regular skin exams: Annual full-body skin
examinations by a dermatologist are recommended for IBD patients on
long-term immunosuppressants.
- Self-examination and education: Patients should be educated on
performing skin self-examinations and practicing sun protection.30
Cervical cancer In IBD:
- Women with IB have an increased risk of cervical dysplasia and
cancer compared to the general population, likely due to the behavioral
nature of chronic inflammation of IBD and the exposure to long-term
immunosuppressive treatments, which can predispose to Immune
dysregulation and predispose to chronic oncogenic infection such as
human papillomavirus (HPV).31
- The risk factors and theb incidence of various types of skin cancer
in patients with Inflammatory Bowel Disease (IBD highlighted in Table 8
and Table 928.29,30.32
Surveillance Recommendations33
- Pap smears: Annual screening is often advised for women with IBD on
immunosuppressive therapy, versus every 3 years for the general
population.
- HPV vaccination: Strongly recommended for eligible patients,
especially prior to immunosuppressive therapy.
Table 1: IBD CRC Surveillance Strategies by Risk Level
High Risk |
PSC, severe inflammation, dense psudoinflammation, CRC in FDR
<50, <5 year history of invisible dysplasia or high risk visible
dysplasia |
Every 1 year |
Intermediate Risk |
Mild inflammation, family CRC but no 1st-degree relative <50
years, <5 year history of invisible dysplasia or high-risk visible
dysplasia, <5year history of low risk visible dysplasia |
Every 2- 3 year |
Low Risk |
Maintaining disease remission with mucosal healing pulse either of 2
consecutive examinations without dysplasia, Minimal colitis (UC
proctitis or<1/3 of the colon in CD) |
Every 5 years |
Table 1: Surveillance strategies recommended for patients with IBD
based on risk stratification. CD: Crohn’s disease; UC: Ulcerative
Colitis; CRC: colorectal cancer; FDR: first-degree relative; PSC:
primary sclerosing
Table 2: Risk Factors for Colorectal Cancer in Inflammatory Bowel
Disease (IBD)
Duration of Disease |
CRC risk increases significantly after 8–10 years of IBD onset.
Longer duration = higher cumulative inflammation and mutation
burden. |
Extent of Colitis |
Risk is highest in pancolitis, intermediate in left-sided colitis,
and negligible in proctitis. |
Primary Sclerosing Cholangitis (PSC) |
Strongest independent risk factor. PSC + IBD increases CRC risk
4–10× even with mild colitis. |
Severity of Inflammation |
Ongoing or recurrent inflammation promotes DNA damage and dysplasia.
Histologic inflammation is strongly correlated with cancer risk. |
Family History of CRC |
Especially if CRC occurs in a first-degree relative <50 years
old. Suggests genetic susceptibility. |
History of Dysplasia |
Prior diagnosis of low-grade or high-grade dysplasia increases
progression risk to CRC. |
Strictures |
May harbor or mask underlying dysplasia or carcinoma. Require biopsy
or resection if suspicious. |
Post-inflammatory Polyps (Pseudopolyps) |
Marker of previous severe inflammation; associated with increased
CRC risk indirectly. |
Younger Age at IBD Diagnosis |
More prolonged exposure to inflammation increases cumulative cancer
risk. |
Male Sex |
Some studies suggest a slightly higher CRC risk in men with
IBD. |
Immunosuppressive Therapy |
Conflicting evidence; some concern with long-term use of thiopurines
(not directly a proven CRC risk factor). |
Table 3 illustrates the recommended guidelines intervals following
the initial colonoscopy, tailored to the individual’s CRC risk21.21
ACG [2019] (GRADE) |
8–10 |
1–2 |
PSC, other risk factors for CRC, previous histology |
ASGE [2015] (GRADE) |
8 |
1–3 |
PSC, inflammation, FDR with CRC |
AGA 2021: Clinical Practice Update |
8–10 |
1–5 |
PSC and other risk factors, prior histology, inflammatory burden,
consecutive examinations without dysplasia |
ACG: American College of Gastroenterology ,ASGE: American Society of
Gastrointestinal Endoscopy ,AGA: American Gastroenterological
Association,PSC: Primary sclerosing cholangitis, CRC: Colorectal cancer
,FDR: First-degree relative.
Note: Individuals with ulcerative proctitis or proctosigmoiditis and
CD limited to the ileum should undergo age specific screening as per
general guidelines.
Table 4: International Guidelines for Dysplasia Surveillance in IBD
Patients
SCENIC Consensus (2015) |
HD recommended If SD, dye-CE recommended If HD, dye-CE
suggested |
No consensus |
SCENIC Commentary (2022) |
HD-WLE, dye-CE, or VCE |
Random limited to highest-risk groups only (PSC, prior dysplasia,
atrophic scarred colon, ongoing active inflammation) |
ECCO Guideline (2017) |
HD recommended |
Random if WL Targeted only if dye-CE |
ECCO Guideline (2023) |
HD-WLE, dye-CE, or VCE |
Random in high-risk (PSC or history of dysplasia) |
ACG Clinical Guideline (2019) |
If SD, dye-CE recommended If HD, dye-CE or VCE recommended |
No recommendation |
AGA Clinical Practice Update (2021) |
HD recommended Dye-CE should be considered VCE acceptable
alternative if HD |
Random if WL only and all patients with high risk (PSC or history of
dysplasia) Targeted if dye-CE or VCE |
BSG Guideline (2019) |
HD recommended If SD, dye-CE recommended If HD, dye-CE suggested NBI
not suggested |
Targeted recommended |
Table 4:Overview of recommendations from international organizations
for dysplasia surveillance in patients with IBD. ACG, American College
of Gastroenterology; AGA, American Gastroenterological Association; BSG,
British Society of Gastroenterology; CE, chromoendoscopy; ECCO, European
Crohn’s and Colitis Organization; HD, high definition; SD, standard
definition; WLE, white-light endoscopy; NBI, narrow band imaging; PSC,
primary sclerosing cholangitis; VCE, virtual chromoendoscopy.
Table 5: Comparison of Endoscopic Modalities for Dysplasia Detection
in IBD
SD-WLE |
Standard-definition white-light endoscopy |
Widely available |
Low detection rate; misses flat lesions |
Not recommended |
HD-WLE |
High-definition white-light endoscopy |
Better mucosal detail; allows targeted biopsies |
Less sensitive than CE for not available |
Acceptable if CE |
Dye-Based Chromoendoscopy (CE) |
Dye application (indigo carmine/methylene blue) |
Highest sensitivity; detects flat dysplasia |
Time-consuming; needs expertise |
Gold standard (SCENIC, ECCO, AGA) |
Narrow-Band Imaging (NBI) |
Optical filter-enhanced blue/green light |
No dye needed; highlights vascular patterns |
Lower sensitivity than dye-CE; operator dependent |
Acceptable alternative (ECCO 2023) |
Virtual Chromoendoscopy (VCE) |
Digital contrast (iSCAN, FICE, OE) |
No dye; faster setup |
Evolving evidence; possibly less sensitive |
Acceptable alternative if HD used |
Table 6: Risk Factors for Lymphoma in IBD
Immunosuppressive therapy |
Thiopurines (AZA/6-MP): Increased risk of non-Hodgkin lymphoma |
Immunosuppressive therapy |
Anti-TNF agents: Slightly increased risk |
Immunosuppressive therapy |
Combination therapy: Highest risk, especially for hepatosplenic
T-cell lymphoma |
Duration and severity of IBD |
Chronic inflammation may contribute, though data are mixed |
Male sex |
Particularly increased risk for hepatosplenic T-cell lymphoma |
Age |
Older patients have higher baseline lymphoma risk |
EBV status (seronegative) |
Thiopurine use in EBV-negative patients linked to EBV-related
lymphoma |
Table7: Types of Lymphoma Associated with Inflammatory Bowel Disease
(IBD)
Non-Hodgkin Lymphoma (NHL) |
Most common |
Especially B-cell lymphomas (e.g., diffuse large B-cell lymphoma).
Associated mainly with thiopurine use. |
Kandiel et al., JAMA, 2005; 293(19):2462–70 |
Hepatosplenic T-cell Lymphoma (HSTCL) |
Rare but serious |
Aggressive T-cell lymphoma. Seen mostly in young males on
combination therapy (thiopurines + anti-TNF). |
Kotlyar et al., Clin Gastroenterol Hepatol, 2015; 13(4):847–58 |
EBV-positive Lymphoproliferative Disorders |
Linked to EBV seronegative patients |
Seen in those starting thiopurines while EBV-naïve. Includes B-cell
lymphomas, sometimes fatal. |
Beaugerie et al., Lancet, 2009; 374(9701):1617–25 |
Peripheral T-cell Lymphoma (PTCL) |
Rare |
Reported in IBD patients, usually after prolonged |
|
Chapter: Transition from Pediatric to Adult Care in Inflammatory
Bowel Disease (IBD)
Transition is the process of migrating a patient with chronic disease
from pediatric to adult care (1). It is characterized by uninterrupted,
coordinated, comprehensive care with attention to the clinical,
psychosocial and educational/vocational needs of adolescent and young
adult (AYA) patients (2,3). Failed transition and transfer is associated
with increased emergency department visit, hospitalization, medication
escalation, surgery and overall poor patient adherence and attitude
(1,4–5)
2. Key Components of a Successful Transition
A successful transition program should include the following
elements:
- Early Introduction: Transition discussions should begin in early
adolescence (ages 12–14) to allow ample time for preparation.
- Assessment of Readiness: Tools such as the Transition Readiness
Assessment Questionnaire (TRAQ)( 6) table -1. can help evaluate a
patient’s knowledge and skills.
- Education: Patients and families should receive education about IBD,
medications, lifestyle management, and the differences between pediatric
and adult care.
- Self-Management Skills: Encourage patients to take an active role in
their care, including scheduling appointments, refilling medications,
and recognizing symptoms of disease activity.
- Joint Visits: Collaborative visits involving both pediatric and
adult providers can ease the transition process.
- Psychosocial Support: Address emotional and social challenges,
including anxiety about transitioning, peer relationships, and body
image concerns.
3. Strategies for Healthcare Providers:
- Develop a Transition Policy: Create a standardized transition
protocol tailored to the needs of IBD patients.
- Use Transition Tools: Utilize checklists figure (1), educational
materials, and readiness assessments to guide the process.(7)
- Foster Collaboration: Establish strong communication channels
between pediatric and adult IBD teams.
- Involve Families: Engage parents and caregivers while gradually
shifting responsibility to the patient.
- Monitor Outcomes: Track transition success through metrics such as
clinic attendance, medication adherence, and disease control.
4. Role of Multidisciplinary Teams
A multidisciplinary approach is essential for addressing the medical,
nutritional, psychological, and social needs of transitioning
patients.(8) The team may include:
- Pediatric and adult gastroenterologists
- Nurses and nurse practitioners
- Dietitians
- Psychologists or social workers
- Pharmacists
Measuring Transition Success Success can be evaluated through:
- Patient satisfaction surveys
- Disease activity indices (e.g., Pediatric Crohn’s Disease Activity
Index, Harvey-Bradshaw Index)
- Healthcare utilization (e.g., emergency visits,
hospitalizations)
- Long-term outcomes (e.g., growth, quality of life)(9)
References
- TestaA , GiannettiE, RispoA, et al. Successful outcome of the
transitional process of inflammatory bowel disease from pediatric to
adult age: A five years experience.Dig Liver Dis 2019
- KahnSA.The transition from pediatric to adult inflammatory bowel
disease care.Gastroenterol Hepatol (N Y)2016
- GoodhandJ, DawsonR, HefferonM, et al. Inflammatory bowel disease in
young people: The case for transitional clinics Inflamm Bowel Dis
2010.;
- BollegalaN , BenchimolEI, GriffithsAM, et al. Characterizing the
posttransfer period among patients with pediatric onset IBD: The impact
of academic versus community adult care on emergent health resource
utilization.Inflamm Bowel Dis 2017
- FuN , JacobsonK, RoundA, et al. Transition clinic attendance is
associated with improved beliefs and attitudes toward medicine in
patients with inflammatory bowel disease.World J Gastroenterol
2017.
- Johnson K, McBee M, Reiss J, Livingood W, Wood D. TRAQ Changes:
Improving the Measurement of Transition Readiness by the Transition
Readiness Assessment Questionnaire. J Pediatr Nurs. 2021
- Gray WN, Holbrook E, Morgan PJ, et al. Transition readiness skills
acquisition in adolescents and young adults with inflammatory bowel
disease: Findings from integrating assessment into clinical practice.
Inflammatory Bowel Diseases. 2015;21(5):1125-1131.
- van Rheenen PF, Griffiths AM, Erlich R, et al. Challenges in
transitional care in inflammatory bowel disease: A review of the
literature. Journal of Pediatric Gastroenterology and Nutrition.
2017;64(3):327-335.
- American Academy of Pediatrics, American Academy of Family
Physicians, American College of Physicians, Transitions Clinical Report
Authoring Group. Supporting the health care transition from adolescence
to adulthood in the medical home. Pediatrics. 2018;142(5):e2018258
Transition Readiness Assessment Questionnaire (TRAQ)
Patient Name: Dateof Birth: / / Today’s Date / / (MRN# )
Directions to Youth and Young Adults : Please check the box that best
describes your skill leve l in the following areas that are
Managing Medications |
|
|
|
|
|
1. Do you fill a prescription if you need to? |
|
|
|
|
|
2. Do you know what to do if you are having a bad reaction to your
medications? |
|
|
|
|
|
3. Do you reorder medications before they run out? |
|
|
|
|
|
4. Do you explain any medications (name and dose) you are taking to
healthcare providers? |
|
|
|
|
|
5. Do you speak with the pharmacist about drug interaction s or
other concerns related to your medications? |
|
|
|
|
|
Appointment Keeping |
|
|
|
|
|
6. Do you call the doctor’s office to make an appointment? |
|
|
|
|
|
7. Do you follow-upon referrals for tests or check-ups or labs? |
|
|
|
|
|
8. Do you arrange for your ride to medical appointments? |
|
|
|
|
|
9. Do you call the doctor about unusual changes in your health (for
example: allergic reactions)? |
|
|
|
|
|
Tracking Health Issues |
|
|
|
|
|
10. Do you fill out the medical history form, including a list of
your allergies? |
|
|
|
|
|
11. Do you keep a calendar or list of medical and other
appointments? |
|
|
|
|
|
12. Do you tell the doctor or nurse what you are feeling? |
|
|
|
|
|
13. Do you contact the doctor when you have a health concern? |
|
|
|
|
|
14. Do you make or help make medical decisions pertaining to your
health? |
|
|
|
|
|
15. Do you attend your medical appointment or part of your
appointment by yourself? |
|
|
|
|
|
Talking with Providers |
|
|
|
|
|
16. Do you ask questions of your nurse or doctor about your health
or health care? |
|
|
|
|
|
17. Do you answer questions that are asked by the doctor, nurse, or
clinic staff? |
|
|
|
|
|
18. Do you ask your doctor or nurse to explain things more clearly
if you do not understand their instructions to you? |
|
|
|
|
|
19. Do you tell the doctor or nurse whether you followed their
advice or recommendations? |
|
|
|
|
|
20. Do you explain your health history to your healthcare providers
(including past surgeries, allergies, and medications)? |
|
|
|
|
|
Please circle how you feel about the following statements
How important is it to you to manage your own healthcare? |
1 |
2 |
3 |
4 |
|
How confident do you feel about your ability to manage your own
health care? |
1 |
2 |
3 |
4 |
|
important for transition to adult healthcare. There is no right or
wrong answer and your answers will remain confidential and private.
Directions to Caregivers / Parents : If your youth or young adult is
unable to complete the tasks below on their own, please check the box
that best describes your skill level. Check here if you area
parent/caregiver completing this form.
Table -1
Figure 1. This Check list adopted from Nasphagan.org
Chapter 23: IBD in Special Populations
By: Ahmed Al-Darmaki
With the latest advancements in immunosuppressants and biologics part
of a prolonged therapeutic approach in treating patients with
inflammatory bowel disease (IBD), patients are susceptible to hepatitis
B virus (HBV) reactivation—even those who are not previously known to be
infected. The range of morbidity and mortality associated with HBV
reactivation among IBD patients spans from mild reactivation to hepatic
decompensation with even non-hepatitis related deaths, which
necessitates improved protocols to manage the situation. This anecdotal
review aims to advocate for HBV screening and vaccination efforts, as
well as concurrent oral antiviral treatments to reduce the risk of
reactivation when managing IBD patients on an immunosuppressive course,
as well as to reduce the risk of infection/reactivation during
non-treatment periods.
Inflammatory Bowel Disease in the Elderly
IBD, encompassing Crohn’s disease (CD) and ulcerative colitis (UC),
is increasingly prevalent in older populations. While historically
considered a disease of young adults, up to 30% of IBD patients are now
over 60 years old, and approximately 10–15% of new diagnoses occur after
age 60 (1, 2)
In this population, it is important to distinguish between two
clinical groups:
- Elderly-onset IBD (EOIBD): Patients diagnosed with IBD at the age of
60 years or older. EOIBD often presents with distinct clinical
characteristics, such as more isolated colonic involvement in CD, and a
tendency for left-sided disease in UC [3][4]. Compared to younger
patients, elderly individuals with IBD typically have a lower prevalence
of extraintestinal manifestations, are less likely to experience disease
progression, and are less frequently treated with immunosuppressants or
biologics(3). See Table 1
- Aging with IBD: Patients who were diagnosed earlier in life and are
now aging with the disease (>60 years old age). This group represents
a growing segment due to improved life expectancy and treatment
outcomes. They tend to have a longer disease duration, higher cumulative
medication exposure, and increased risk of complications such as
malignancy, infections, and surgery-related morbidity[5][6].
Table 1: Comparison of Elderly-Onset vs. Younger-Onset IBD (Adapted
from Tran V, et al. Curr Gastroenterol Rep. 2019) (4).
Common Location (CD) |
Colonic |
Ileal/Ileocolonic |
UC Extent |
Left-sided/Distal |
Pancolitis more common |
Disease Behavior (CD) |
Inflammatory |
Complicated (stricturing/penetrating) |
Extraintestinal Manifestations |
Less common |
More common |
Immunosuppressive use |
Less frequent |
More frequent |
Surgery |
Lower cumulative risk |
Higher cumulative risk |
Cancer surveillance |
More relevant |
Age-dependent |
Polypharmacy risk |
High |
Moderate |
Diagnostic delay |
More likely |
Less likely |
Elderly patients often have atypical presentations, a higher burden
of comorbidities, and are commonly prescribed multiple medications.
These factors increase the risk of both disease- and treatment-related
complications, making diagnosis, therapeutic planning, and long-term
care more complex. Careful, individualized strategies are essential to
improve outcomes in this population.(3, 5).
Epidemiology and Demographic Shift
As the population ages, the incidence and prevalence of IBD globally
in the older population is on the rise (1). In the next ten years, more
than 1 in 3 IBD patients will be over the age of 60 (5, 6). From
1995-2016 the prevalence of UC in patients aged >65 years increased
by more than three-fold. The median age of patients diagnosed with IBD
has increased by more than 7 years in the last twenty years (1).
Research out of France and Denmark find that 1 in 8 patients with a new
diagnosis of UC and 1 in 20 with a new diagnosis of CD are diagnosed
after the age of 60(1, 3, 5) A recent large nationwide cohort study from
Denmark(7) showed similar trends and further classified patients aged
≥70 years as having very late-onset IBD. These patients had comparable
risks of IBD-related hospitalizations and corticosteroid use as those
diagnosed at 60–69 years but were significantly less likely to receive
immunosuppressive therapy or undergo surgery.
Diagnosis Challenges
The differential diagnosis in older adults is broad and
includes(2):
- Ischemic colitis
- NSAID-induced injury
- Microscopic colitis
- Neoplasia
- Segmental colitis associated with diverticulosis,
- Radiation enteritis or colitis.
- Solitary rectal ulcer syndrome
Endoscopic and histologic confirmation is critical. Delays in
diagnosis can worsen outcomes. Newer non-invasive markers like fecal
calprotectin are helpful but may be elevated in other conditions common
in older adults (e.g., diverticulitis).
Functional Reserve, Frailty, Comorbidities, and Polypharmacy
The interplay of frailty, comorbidities, and diminished functional
reserve plays a central role in shaping outcomes for elderly patients
with IBD(8, 9). Older individuals are more likely to experience adverse
outcomes not only due to the disease itself but due to impaired
physiological reserves.
Frailty—reduced strength, endurance, and function— is a stronger
predictor of poor clinical outcome than chronological age; 40% of
elderly-onset IBD cohort are frail. It correlates with increased
hospitalizations, postoperative complications, disability, and
mortality(9). Therefore, fraitiliy needs to be screened in all elderly
with IBD. Clincial Box 1 list clinical evaluation tools.
Polypharmacy is another hallmark of this population, driven by
coexisting cardiovascular, metabolic, musculoskeletal, and
neurocognitive conditions. . This increases the chances for drug
interactions, side effects, nonadherence to therapy considering there
are concurrent medications in addition to corticosteroids, thiopurines
or JAK inhibitors (9)
Therapeutic Considerations
- Assess frailty and functional status regularly to guide treatment
intensity.
- Avoid thiopurines, methotrexate and long-term corticosteroids due to
higher risk of adverse effects.
- Consider early biologic initiation in case of steroids need.
- Simplify regimens, and prefer vedolizumab or ustekinumab due to
better safety in older adults.
- Use anti-TNFs cautiously, especially in fit patients with limited
alternatives.
- Limit JAK inhibitors to carefully selected cases with close
monitoring.
- Ensure vaccination against pneumococcus, zoster, and influenza
before immunosuppression.
- Do not undertreat—active inflammation can lead to worse outcomes
than therapy risks.
- Involve caregivers and plan for social support, monitoring, and
adherence.
- Tailor surveillance strategies for cancer, bone density, and
malnutrition.
- Clincial Boxes 2&3, summerizes the initiaion of therapies and
availbale therapies.
Clinical Box 2: Decision Considerations Before Initiating
Immunosuppressive Therapy in Elderly IBD Patients(9, 10)
Is the patient frail? |
CFS ≥ 5, gait speed <0.8 m/s |
Avoid or delay immunosuppressants; consider gut-selective agents
(e.g., vedolizumab) |
Any serious comorbidities? |
Charlson Index ≥ 5 or active cardiac/pulmonary disease |
Prioritize safety and avoid aggressive immunosuppression |
Polypharmacy risk? |
≥5 concurrent medications, risk of interactions |
Simplify regimens, use non-systemic therapies |
Recent infection history? |
≥2 serious infections in past 12 months |
Reassess benefit-risk ratio for biologics |
Clinical Box 3: Summery of elderly IBD therapies:
Anti-TNF agents |
Effective but higher risk of infection, malignancy |
Discontinuation common; avoid combination therapy in frail or
high-risk patients (2, 8, 11) |
Vedolizumab |
Preferred first-line in many centers; safer profile |
Lower systemic risk; favorable infection profile; suitable in
multimorbid, frail patients(11, 12) |
Ustekinumab/IL23 |
Safe and well-tolerated in older patients |
No increase in adverse events compared with placebo in meta-analysis
of all RCTs (IBD and non-IBD). Limited data yet re: age-specific risk,
suitable in patients ≥70 years. (11) |
JAK inhibitors |
Use with caution; restricted to select cases only if no other
option. |
Elevated risk of VTE, CV events, and herpes zoster; requires
risk-benefit analysis, give Monitor for bradycardia, liver enzyme |
S1P modulators (e.g., ozanimod) |
Limited data; caution in elderly |
elevations; avoid in CV disease or polypharmacy |
Corticosteroids |
Short-term only; avoid chronic use |
Associated with delirium, infection, fractures; taper early and
transition to maintenance. Use enteral coated sterids wherever possible.
(11) |
5-ASA |
Safe in mild UC; not for Crohn’s |
Monitor renal function; watch pill burden in cognitively impaired
patients |
Thiopurines |
Limited use due to toxicity, acvoid after age of 60 years. |
Risk of lymphoma, skin cancer; avoid unless no alternative; requires
age-specific caution |
Methotrexate |
Poorly tolerated; limited role |
Hepatotoxicity and cytopenia risks high in elderly; often not
preferred |
Surgery |
Consider frailty; elective preferred |
Outcomes better with elective surgery; pre-op frailty and nutrition
optimization essential |
Hepatitis B Virus Management in IBD Patients
Managing IBD patients with hepatitis B virus (HBV) exposure requires
balancing therapeutic efficacy with hepatic safety. Immunosuppressive
medications can precipitate HBV reactivation, potentially causing
fulminant hepatitis and liver failure. This necessitates systematic
screening, monitoring, and prophylaxis protocols that have become
integral to modern IBD care. (13, 14)
Epidemiology and Reactivation Risk
HBV prevalence among IBD patients varies significantly by region.
Giri et al.'s meta-analysis found global HBsAg positivity of 3.3% in IBD
patients, but with marked regional differences: 5.8% in Asia versus 1.4%
in Europe. (15) Prior HBV exposure (anti-HBc positivity) shows even
greater disparity: 30.3% in Asian versus 8.3% in European populations.
These patterns underscore the importance of universal screening
regardless of geographic location.
HBV reactivation is defined as reappearance of HBsAg or significant
HBV DNA increase in previously inactive infection. Risk depends on
baseline serologic status and immunosuppressive regimen:
- HBsAg-positive patients: Up to 50% reactivation rate without
prophylaxis, especially with anti-TNF agents or corticosteroids.
- HbsAg-negative, Anti-HBc-positive patients: 1-10% reactivation risk,
reported with thiopurines, anti-TNF agents, and JAK inhibitors
HBV Treatment in IBD: A Step-by-Step Practical Approach(13, 14)
A. Screening and prevention
All patients with IBD should undergo HBV screening before initiating
immunosuppressive therapy. Vaccinate those who are seronegative when
feasible, and evaluate the need for antiviral prophylaxis in
HBsAg-positive individuals. Recommended screening tests include:
- HBsAg (identifies active infection)
- Anti-HBc (indicates prior exposure)
- Anti-HBs (assesses immunity)
- HBV DNA when HBsAg positive
- HBcrAg may be considered in anti-HBc-positive, HBsAg-negative
patients planning high-risk immunosuppression
B. Choose Safer Treatments When Possible
When HBV exposure is known, prioritize lower-risk agents such as
vedolizumab or ustekinumab whenever clinically appropriate. If
higher-risk treatments—such as anti-TNF agents, JAK inhibitors, or
prolonged courses of corticosteroids—are required, they should be
accompanied by antiviral prophylaxis and close virologic monitoring to
reduce the risk of HBV reactivation. See Table 1
C. Monitor During and After Therapy
For patients with confirmed HBV exposure, ongoing care includes
regular monitoring of ALT and HBV DNA throughout immunosuppressive
treatment and after its completion. In urgent clinical situations,
antivirals and IBD therapy may be started at the same time if a two-week
lead-in is not feasible. Management depends on HBV serologic status, as
outlined below.
D. Management by Serologic Status(13)
- Active Infection (HBsAg Positive) : Start entecavir or tenofovir at
least two weeks before immunosuppression. Continue during therapy and
6-12 months post-treatment (18 months for B-cell depleting agents).
- Resolved Infection (HBsAg Negative, Anti-HBc Positive): Monitor HBV
DNA every 3 months. Consider prophylaxis for high-risk therapies or
unreliable follow-up.
- No Prior Exposure (HBsAg/Anti-HBc Negative): Vaccinate before
therapy when possible. Target anti-HBs >10 IU/L. High-dose or
accelerated schedules may be needed due to impaired vaccine response
during immunosuppression.
Table 1: HBV Reactivation Risk by IBD Medications(13, 16)
High (>10%) |
• Anti-TNF agents • Corticosteroids (>40 mg/day) •
Corticosteroids (>4 weeks, >20 mg/day) • JAK inhibitors • S1P
receptor modulators |
Antiviral prophylaxis mandatory |
Moderate (1-10%) |
• Ustekinumab (anti-IL-12/23) • Other anti-IL23 agents |
• Anti-TNF agents • JAK inhibitors • Ustekinumab (anti-IL-12/23)
& anti-IL23 agents |
Low (<1%) |
• Azathioprine • Methotrexate • Mycophenolate mofetil •
Corticosteroids (low-dose <10 mg/day) for ≤1 week) • Vedolizumab |
• Azathioprine • Methotrexate • Mycophenolate mofetil •
Corticosteroids (<40 mg/day for ≤1 week) • Vedolizumab |
**S1P receptor modulators (ozanimod, etrasimod): Risk category based
on mechanism of lymphocyte sequestration rather than broad
immunosuppression. Limited HBV reactivation data available; close
monitoring recommended until more safety data emerge. Note: Vedolizumab
not specifically listed in EASL table but considered low-risk based on
gut-selective mechanism
IBD in Patients with Sickle Cell Disease
The coexistence of sickle cell disease (SCD) and autoimmune diseases
like IBD is rare but clinically important as it appears to be associated
with both a severe phenotype of autoimmune disease and worsening of
SCD(17). Historically, limited reports have made it difficult to define
the true overlap, though both diseases share key immunoinflammatory
pathways such as TNF-α, IL-6, and neutrophil extracellular traps (NETs)
that may contribute to organ injury in both conditions(18, 19) . The
baseline hyperinflammatory state in SCD can exacerbate IBD activity,
while intestinal microvascular occlusion may mimic IBD-related ischemic
changes. Clinically, overlapping features such as chronic abdominal pain
and anemia complicate diagnosis, often leading to delays in identifying
IBD in SCD patients.
Geographic differences may explain the under-recognition of SCD-IBD
overlap, with SCD common in Africa and the Middle East, and IBD more
prevalent in Western countries. A recent French study—the largest to
date—found colonic involvement in 95.8% of SCD-IBD cases and PSC in
33.3%, far higher than typical IBD cohorts. Histology confirmed
classical IBD features, not ischemic colitis, ruling out sickle-related
vascular injury as the cause.(20)
Outcomes depend on early diagnosis, minimizing corticosteroid
exposure, and selecting therapies with low infection and hematologic
risk. Severe infections remain a major cause of morbidity and mortality,
particularly in patients on systemic immunosuppression(21).
Multidisciplinary care is essential: hematologists and
gastroenterologists must collaborate to tailor safe and effective
regimens.
Therapeutic Considerations
- Corticosteroids remain essential for inducing remission in
moderate-to-severe IBD. However, they are associated with a significant
increase in VOCs in SCD patients—reported in up to 57% of those exposed
(22). Their us
Chapter 24: Counseling patient and coping with IBD
Introduction
Psychological Impact of IBD
Common emotional challenges: anxiety, depression, stress, and reduced
quality of life (QoL).
Stigma, embarrassment, and social isolation due to symptoms (e.g.,
urgency, incontinence).
Impact on work, relationships, and daily functioning.(1)
Key Counseling Strategies for IBD Patients
A. Building a Therapeutic Alliance
Active listening, empathy, and validating patient concerns.
Encouraging open communication about symptoms and emotional
distress.(2)
B. Psychoeducation
Educating patients about IBD, treatment options, and
self-management.
Addressing misconceptions (e.g., diet, stress as a cause
vs. trigger).
C. Cognitive Behavioral Therapy (CBT) and Mindfulness
CBT for managing anxiety, depression, and maladaptive coping.
Mindfulness-based stress reduction (MBSR) for symptom
relief.(3)
Coping Mechanisms for Patients
A. Lifestyle Modifications
Diet adjustments (low-residue, FODMAP, hydration).
Regular exercise to reduce stress and improve well-being.
B. Social Support and Peer Groups
Importance of family, friends, and IBD support groups (e.g., Crohn’s
& Colitis Foundation).(4-5)
C. Stress Management Techniques
Relaxation training (deep breathing, progressive muscle
relaxation).(4)
Journaling and expressive writing for emotional processing.
Addressing Specific Challenges
Flare-ups: Preparing emergency plans (bathroom access,
medication).(6)
Sexual Health & Body Image: Counseling on intimacy
concerns.(7)
Transitional Care: Pediatric to adult IBD care counseling.
The Role of Multidisciplinary Care
Collaboration between gastroenterologists, psychologists, dietitians,
and social workers.
Integrated care models for holistic IBD management.(8)
References:
- 1-Mikocka-Walus A et al. (2016). “Controversies revisited: A
systematic review of the comorbidity of depression and anxiety with
inflammatory bowel diseases.” Inflammatory Bowel Diseases, 22(3),
752-762.
- 2-Knowles SR et al. (2018). “Effectiveness of cognitive behavioral
therapy for people with inflammatory bowel disease.” Clinical
Gastroenterology and Hepatology, 16(7), 1069-1078.
- 3-Neilson K et al. (2021). “Mindfulness-based interventions for
gastrointestinal disorders: A systematic review.” Neurogastroenterology
& Motility, 33(3), e14022.
- 4-Byrne G et al. (2017). “Coping strategies used by adults with
inflammatory bowel disease.”Gastroenterology Nursing, 40(3),
200-207.
- 5-van der Eijk I et al. (2020). “The role of perceived social
support in quality of life in IBD.” Journal of Crohn’s and Colitis,
14(5), 629-637.
- 6-Conley S & Redeker N (2016). “A systematic review of
self-management interventions for inflammatory bowel disease.” Journal
of Nursing Scholarship, 48(2), 118-127.
- 7-Siegel CA (2013). “Sexual dysfunction in IBD: What the physician
should know.” Inflammatory Bowel Diseases, 19(12), 2716-2723
- 8-van Deen WK et al. (2019). “The impact of integrated care on
health-related quality of life in IBD.”American Journal of
Gastroenterology, 114(10), 1631-1639.
POCER Trial
Colonoscopy at 6 months (active care) or no colonoscopy
(standard care)
Study Design: Randomized, double-blind
- Initial drug therapy for both groups: all patients received 3 months
of metronidazole.
- Further therapy was stratified according to the predicted risk of
recurrence:
- High-risk patients received thiopurine and if intolerant, received
adalimumab.
- Low-risk patients only completed the metronidazole without
additional therapy.
- All patients had colonoscopy at 18 months
- Primary endpoint: Presence and severity of endoscopic recurrence at
18 months after surgery using the Rutgeerts score
Result:
- Endoscopic recurrence at 18 months was lower in the active care
group (49%) than the standard of care group (67%), p=0.03.
- Complete mucosal normality was maintained in (22%) in the active
care group Vs (8%) in the standard care group (p=0·03).
- The incidence and type of adverse and severe adverse events did not
differ significantly between patients in the active care and standard
care groups
Conclusion: Active management of post-operative
Crohn’s disease patients with surveillance colonoscopy at 6 months and
drug escalation as necessary leads to lower rates of endoscopic
recurrence at 18 months compared to standard treatment.
Comment : In active group, If the colonoscopy
showed endoscopic recurrence , medical therapy was escalated as
follows:
- If not on thiopurine: start thiopurine. If already on thiopurine:
add adalimumab. If already on adalimumab 40mg q 2 weeks: increase to
adalimumab q1week.
Original study: https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(14)61908-5/abstract
ASUC rescue therapy studies
CONSTRUCT Trial
Infliximab vs Cyclosporine in Acute Severe Ulcerative
Colitis
Study Design: Randomized, open-label,
multicenter trial
- Patients with acute severe ulcerative colitis refractory to
intravenous corticosteroids were randomized to receive either:
- Infliximab (5 mg/kg IV at weeks 0, 2, 6)
- Cyclosporine (2 mg/kg/day IV for 7 days, then oral for 3
months)
- Primary endpoint: Colectomy-free survival at 3 months
- Secondary endpoint: Colectomy-free survival at 12 months, adverse
events
Result:
- Colectomy-free survival at 3 months: Infliximab (60%) vs
Cyclosporine (55%) (P = 0.55)
- Colectomy-free survival at 12 months: Infliximab (50%) vs
Cyclosporine (45%) (P = 0.62)
- No significant difference in adverse events between groups.
Conclusion: Infliximab and cyclosporine showed
similar efficacy in preventing colectomy in patients with acute severe
ulcerative colitis refractory to intravenous corticosteroids.
Original study: https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(13)61747-0/abstract
ACT 1 & 2 Trials
Infliximab for Moderate to Severe Ulcerative
Colitis
Study Design: Randomized, double-blind,
placebo-controlled trials
- Patients with moderate to severe ulcerative colitis were randomized
to receive:
- Infliximab (5 mg/kg or 10 mg/kg IV at weeks 0, 2, 6, then every 8
weeks)
- Placebo
- Primary endpoint: Clinical response and remission at week 8 and week
30
- Secondary endpoint: Mucosal healing, corticosteroid-free remission,
quality of life
Result:
- Clinical response and remission rates were significantly higher with
infliximab compared to placebo at both week 8 and week 30.
- Mucosal healing and corticosteroid-free remission rates were also
significantly higher with infliximab.
Conclusion: Infliximab is effective for inducing
and maintaining clinical response and remission, and promoting mucosal
healing in patients with moderate to severe ulcerative colitis.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa053426
GEMINI 1 & 2 Trials
Vedolizumab for Ulcerative Colitis and Crohn’s
Disease
Study Design: Randomized, double-blind,
placebo-controlled trials
- Patients with moderate to severe ulcerative colitis (GEMINI 1) or
Crohn’s disease (GEMINI 2) were randomized to receive:
- Vedolizumab (300 mg IV at weeks 0, 2, 6, then every 8 weeks)
- Placebo
- Primary endpoint: Clinical response and remission at week 6 (UC) or
week 10 (CD), and at week 52
- Secondary endpoint: Mucosal healing, corticosteroid-free remission,
quality of life
Result:
- Vedolizumab was significantly more effective than placebo in
inducing and maintaining clinical response and remission, and promoting
mucosal healing in both ulcerative colitis and Crohn’s disease.
- Safety profile was favorable, with lower rates of serious infections
compared to anti-TNF agents.
Conclusion: Vedolizumab is an effective and safe
treatment option for patients with moderate to severe ulcerative colitis
and Crohn’s disease.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa1307924
UNITI 1 & 2 Trials
Ustekinumab for Crohn’s Disease
Study Design: Randomized, double-blind,
placebo-controlled trials
- Patients with moderate to severe Crohn’s disease (UNITI 1: anti-TNF
failure; UNITI 2: anti-TNF naive or failure) were randomized to receive:
- Ustekinumab (IV induction dose based on weight, then 90 mg SC every
8 or 12 weeks)
- Placebo
- Primary endpoint: Clinical response and remission at week 6
(induction) and week 44 (maintenance)
- Secondary endpoint: Corticosteroid-free remission, mucosal healing,
quality of life
Result:
- Ustekinumab was significantly more effective than placebo in
inducing and maintaining clinical response and remission in patients
with moderate to severe Crohn’s disease, including those who failed
prior anti-TNF therapy.
- Safety profile was consistent with previous studies.
Conclusion: Ustekinumab is an effective and safe
treatment option for patients with moderate to severe Crohn’s disease,
regardless of prior anti-TNF exposure.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa1604670
U-ACHIEVE & U-ACCOMPLISH Trials
Upadacitinib for Ulcerative Colitis
Study Design: Randomized, double-blind,
placebo-controlled trials
- Patients with moderate to severe ulcerative colitis were randomized
to receive:
- Upadacitinib (45 mg daily for 8 weeks induction, then 15 mg or 30 mg
daily maintenance)
- Placebo
- Primary endpoint: Clinical remission at week 8 (induction) and week
52 (maintenance)
- Secondary endpoint: Endoscopic improvement, mucosal healing,
corticosteroid-free remission, quality of life
Result:
- Upadacitinib was significantly more effective than placebo in
inducing and maintaining clinical remission, endoscopic improvement, and
mucosal healing in patients with moderate to severe ulcerative
colitis.
- Safety profile was consistent with known JAK inhibitor risks (e.g.,
herpes zoster, VTE).
Conclusion: Upadacitinib is an effective oral
treatment option for patients with moderate to severe ulcerative
colitis.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa2008019
U-EXCEL & U-EXCEED Trials
Upadacitinib for Crohn’s Disease
Study Design: Randomized, double-blind,
placebo-controlled trials
- Patients with moderate to severe Crohn’s disease were randomized to
receive:
- Upadacitinib (45 mg daily for 12 weeks induction, then 15 mg or 30
mg daily maintenance)
- Placebo
- Primary endpoint: Clinical remission and endoscopic response at week
12 (induction) and week 52 (maintenance)
- Secondary endpoint: Corticosteroid-free remission, mucosal healing,
quality of life
Result:
- Upadacitinib was significantly more effective than placebo in
inducing and maintaining clinical remission and endoscopic response in
patients with moderate to severe Crohn’s disease.
- Safety profile was consistent with known JAK inhibitor risks.
Conclusion: Upadacitinib is an effective oral
treatment option for patients with moderate to severe Crohn’s
disease.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa2202720
CELEST Trial
Ozanimod for Ulcerative Colitis
Study Design: Randomized, double-blind,
placebo-controlled trial
- Patients with moderate to severe ulcerative colitis were randomized
to receive:
- Ozanimod (0.92 mg daily)
- Placebo
- Primary endpoint: Clinical remission at week 10 (induction) and week
52 (maintenance)
- Secondary endpoint: Endoscopic improvement, mucosal healing,
corticosteroid-free remission, quality of life
Result:
- Ozanimod was significantly more effective than placebo in inducing
and maintaining clinical remission, endoscopic improvement, and mucosal
healing in patients with moderate to severe ulcerative colitis.
- Safety profile was generally favorable, with some cardiac and liver
enzyme monitoring required.
Conclusion: Ozanimod is an effective oral
treatment option for patients with moderate to severe ulcerative
colitis.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa1902219
True North Study
Ozanimod for Ulcerative Colitis
Study Design: Randomized, double-blind,
placebo-controlled trial (similar to CELEST, but larger and pivotal)
- Patients with moderate to severe ulcerative colitis were randomized
to receive:
- Ozanimod (0.92 mg daily)
- Placebo
- Primary endpoint: Clinical remission at week 10 (induction) and week
52 (maintenance)
- Secondary endpoint: Endoscopic improvement, mucosal healing,
corticosteroid-free remission, quality of life
Result:
- True North confirmed the efficacy and safety findings of CELEST,
demonstrating significant improvements in clinical remission, endoscopic
improvement, and mucosal healing with ozanimod compared to placebo.
Conclusion: Ozanimod is an effective and
well-tolerated oral treatment option for patients with moderate to
severe ulcerative colitis.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa2022311
POCER Trial
Colonoscopy at 6 months (active care) or no colonoscopy
(standard care)
Study Design: Randomized, double-blind
- Initial drug therapy for both groups: all patients received 3 months
of metronidazole.
- Further therapy was stratified according to the predicted risk of
recurrence:
- High-risk patients received thiopurine and if intolerant, received
adalimumab.
- Low-risk patients only completed the metronidazole without
additional therapy.
- All patients had colonoscopy at 18 months
- Primary endpoint: Presence and severity of endoscopic recurrence at
18 months after surgery using the Rutgeerts score
Result:
- Endoscopic recurrence at 18 months was lower in the active care
group (49%) than the standard of care group (67%), p=0.03.
- Complete mucosal normality was maintained in (22%) in the active
care group Vs (8%) in the standard care group (p=0·03).
- The incidence and type of adverse and severe adverse events did not
differ significantly between patients in the active care and standard
care groups
Conclusion: Active management of post-operative
Crohn’s disease patients with surveillance colonoscopy at 6 months and
drug escalation as necessary leads to lower rates of endoscopic
recurrence at 18 months compared to standard treatment.
Comment : In active group, If the colonoscopy
showed endoscopic recurrence , medical therapy was escalated as
follows:
- If not on thiopurine: start thiopurine. If already on thiopurine:
add adalimumab. If already on adalimumab 40mg q 2 weeks: increase to
adalimumab q1week.
Original study: https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(14)61908-5/abstract
ASUC rescue therapy studies
CONSTRUCT Trial
Infliximab vs Cyclosporine in Acute Severe Ulcerative
Colitis
Study Design: Randomized, open-label,
multicenter trial
- Patients with acute severe ulcerative colitis refractory to
intravenous corticosteroids were randomized to receive either:
- Infliximab (5 mg/kg IV at weeks 0, 2, 6)
- Cyclosporine (2 mg/kg/day IV for 7 days, then oral for 3
months)
- Primary endpoint: Colectomy-free survival at 3 months
- Secondary endpoint: Colectomy-free survival at 12 months, adverse
events
Result:
- Colectomy-free survival at 3 months: Infliximab (60%) vs
Cyclosporine (55%) (P = 0.55)
- Colectomy-free survival at 12 months: Infliximab (50%) vs
Cyclosporine (45%) (P = 0.62)
- No significant difference in adverse events between groups.
Conclusion: Infliximab and cyclosporine showed
similar efficacy in preventing colectomy in patients with acute severe
ulcerative colitis refractory to intravenous corticosteroids.
Original study: https://www.thelancet.com/journals/lancet/article/PIIS0140-6736(13)61747-0/abstract
ACT 1 & 2 Trials
Infliximab for Moderate to Severe Ulcerative
Colitis
Study Design: Randomized, double-blind,
placebo-controlled trials
- Patients with moderate to severe ulcerative colitis were randomized
to receive:
- Infliximab (5 mg/kg or 10 mg/kg IV at weeks 0, 2, 6, then every 8
weeks)
- Placebo
- Primary endpoint: Clinical response and remission at week 8 and week
30
- Secondary endpoint: Mucosal healing, corticosteroid-free remission,
quality of life
Result:
- Clinical response and remission rates were significantly higher with
infliximab compared to placebo at both week 8 and week 30.
- Mucosal healing and corticosteroid-free remission rates were also
significantly higher with infliximab.
Conclusion: Infliximab is effective for inducing
and maintaining clinical response and remission, and promoting mucosal
healing in patients with moderate to severe ulcerative colitis.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa053426
GEMINI 1 & 2 Trials
Vedolizumab for Ulcerative Colitis and Crohn’s
Disease
Study Design: Randomized, double-blind,
placebo-controlled trials
- Patients with moderate to severe ulcerative colitis (GEMINI 1) or
Crohn’s disease (GEMINI 2) were randomized to receive:
- Vedolizumab (300 mg IV at weeks 0, 2, 6, then every 8 weeks)
- Placebo
- Primary endpoint: Clinical response and remission at week 6 (UC) or
week 10 (CD), and at week 52
- Secondary endpoint: Mucosal healing, corticosteroid-free remission,
quality of life
Result:
- Vedolizumab was significantly more effective than placebo in
inducing and maintaining clinical response and remission, and promoting
mucosal healing in both ulcerative colitis and Crohn’s disease.
- Safety profile was favorable, with lower rates of serious infections
compared to anti-TNF agents.
Conclusion: Vedolizumab is an effective and safe
treatment option for patients with moderate to severe ulcerative colitis
and Crohn’s Disease.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa1307924
UNITI 1 & 2 Trials
Ustekinumab for Crohn’s Disease
Study Design: Randomized, double-blind,
placebo-controlled trials
- Patients with moderate to severe Crohn’s disease (UNITI 1: anti-TNF
failure; UNITI 2: anti-TNF naive or failure) were randomized to receive:
- Ustekinumab (IV induction dose based on weight, then 90 mg SC every
8 or 12 weeks)
- Placebo
- Primary endpoint: Clinical response and remission at week 6
(induction) and week 44 (maintenance)
- Secondary endpoint: Corticosteroid-free remission, mucosal healing,
quality of life
Result:
- Ustekinumab was significantly more effective than placebo in
inducing and maintaining clinical response and remission in patients
with moderate to severe Crohn’s disease, including those who failed
prior anti-TNF therapy.
- Safety profile was consistent with previous studies.
Conclusion: Ustekinumab is an effective and safe
treatment option for patients with moderate to severe Crohn’s disease,
regardless of prior anti-TNF exposure.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa1604670
U-ACHIEVE & U-ACCOMPLISH Trials
Upadacitinib for Ulcerative Colitis
Study Design: Randomized, double-blind,
placebo-controlled trials
- Patients with moderate to severe ulcerative colitis were randomized
to receive:
- Upadacitinib (45 mg daily for 8 weeks induction, then 15 mg or 30 mg
daily maintenance)
- Placebo
- Primary endpoint: Clinical remission at week 8 (induction) and week
52 (maintenance)
- Secondary endpoint: Endoscopic improvement, mucosal healing,
corticosteroid-free remission, quality of life
Result:
- Upadacitinib was significantly more effective than placebo in
inducing and maintaining clinical remission, endoscopic improvement, and
mucosal healing in patients with moderate to severe ulcerative
colitis.
- Safety profile was consistent with known JAK inhibitor risks (e.g.,
herpes zoster, VTE).
Conclusion: Upadacitinib is an effective oral
treatment option for patients with moderate to severe ulcerative
colitis.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa2008019
U-EXCEL & U-EXCEED Trials
Upadacitinib for Crohn’s Disease
Study Design: Randomized, double-blind,
placebo-controlled trials
- Patients with moderate to severe Crohn’s disease were randomized to
receive:
- Upadacitinib (45 mg daily for 12 weeks induction, then 15 mg or 30
mg daily maintenance)
- Placebo
- Primary endpoint: Clinical remission and endoscopic response at week
12 (induction) and week 52 (maintenance)
- Secondary endpoint: Corticosteroid-free remission, mucosal healing,
quality of life
Result:
- Upadacitinib was significantly more effective than placebo in
inducing and maintaining clinical remission and endoscopic response in
patients with moderate to severe Crohn’s disease.
- Safety profile was consistent with known JAK inhibitor risks.
Conclusion: Upadacitinib is an effective oral
treatment option for patients with moderate to severe Crohn’s
disease.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa2202720
CELEST Trial
Ozanimod for Ulcerative Colitis
Study Design: Randomized, double-blind,
placebo-controlled trial
- Patients with moderate to severe ulcerative colitis were randomized
to receive:
- Ozanimod (0.92 mg daily)
- Placebo
- Primary endpoint: Clinical remission at week 10 (induction) and week
52 (maintenance)
- Secondary endpoint: Endoscopic improvement, mucosal healing,
corticosteroid-free remission, quality of life
Result:
- Ozanimod was significantly more effective than placebo in inducing
and maintaining clinical remission, endoscopic improvement, and mucosal
healing in patients with moderate to severe ulcerative colitis.
- Safety profile was generally favorable, with some cardiac and liver
enzyme monitoring required.
Conclusion: Ozanimod is an effective oral
treatment option for patients with moderate to severe ulcerative
colitis.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa1902219
True North Study
Ozanimod for Ulcerative Colitis
Study Design: Randomized, double-blind,
placebo-controlled trial (similar to CELEST, but larger and pivotal)
- Patients with moderate to severe ulcerative colitis were randomized
to receive:
- Ozanimod (0.92 mg daily)
- Placebo
- Primary endpoint: Clinical remission at week 10 (induction) and week
52 (maintenance)
- Secondary endpoint: Endoscopic improvement, mucosal healing,
corticosteroid-free remission, quality of life
Result:
- True North confirmed the efficacy and safety findings of CELEST,
demonstrating significant improvements in clinical remission, endoscopic
improvement, and mucosal healing with ozanimod compared to placebo.
Conclusion: Ozanimod is an effective and
well-tolerated oral treatment option for patients with moderate to
severe ulcerative colitis.
Original study: https://www.nejm.org/doi/full/10.1056/NEJMoa2022311
Chapter 17: Anemia in Inflammatory Bowel Disease
(IBD)
Introduction
Anemia is one of the most frequent complications or extraintestinal
manifestations (EIMs) of Inflammatory Bowel Disease (IBD), which
includes Crohn’s disease and ulcerative colitis. This condition can
significantly impact quality of life, contributing to fatigue, reduced
physical endurance, and even impaired cognitive function. For healthcare
providers caring for IBD patients, recognizing and addressing anemia is
essential to holistic disease management¹.
Epidemiology and Prevalence
Anemia is common among IBD patients. The World Health Organization
(WHO) provides the following age- and gender-specific cut-offs for
haemoglobin (Hb) concentration in anemia: <130 g/L for adult men,
<120 g/L for adult, non-pregnant women, and <110 g/L for children
aged 6–59 months and increasing with age. Studies showed an overall
anemia prevalence of 27% in patients with CD and 21% in those with UC²,
with the prevalence in children as high as 90% at diagnosis³. Higher
rates are typically observed during disease flares and in hospitalized
patients, and anemia may be present at diagnosis or develop as the
disease progresses. Iron deficiency, in particular, is highly prevalent,
affecting over half of IBD patients at some point⁴.
Pathophysiology
Anemia in IBD arises from multiple causes. Iron deficiency anemia
(IDA) is the most common type, usually due to chronic blood loss from
intestinal inflammation, reduced dietary intake, and malabsorption.
Anemia of chronic disease (ACD), or anemia of inflammation, also plays a
significant role. Inflammatory cytokines elevate hepcidin levels, which
restrict iron availability and impair red blood cell production⁵. Many
patients experience a combination of IDA and ACD, and some may have
additional contributors like vitamin B12 or folate deficiencies,
especially if they have undergone bowel resection⁵,⁶.
Table 1: Comparison of Anemia Types in IBD
Iron Deficiency Anemia (IDA) |
Chronic blood loss, reduced intake, malabsorption |
Low hemoglobin, low serum ferritin (<30 μg/L), low transferrin
saturation (<16%) |
Oral or IV iron supplementation, address bleeding sources |
Anemia of Chronic Disease (ACD) |
Inflammation-mediated iron sequestration |
Low/normal hemoglobin, normal/high serum ferritin (>100 ug/L) and
transferrin saturation <20%, elevated CRP |
Treat underlying inflammation, consider IV iron, possible ESA
(erythropoiesis-stimulating agent) |
Mixed Anemia |
Combination of IDA and ACD factors |
Features of both IDA and ACD |
Combined approach: iron supplementation and anti-inflammatory
therapy¹ |
Diagnostic Approach
Evaluation should begin with a complete blood count (CBC) to confirm
anemia. Next steps include:
- Anaemia parameters should be evaluated every 6–12 months in patients
in remission or with mild disease activity; patients with active disease
should be monitored at least every 3 months¹.
- Serum Ferritin: <30 µg/L suggests IDA. In inflammation, levels
<100 µg/L may still indicate deficiency⁷.
- Transferrin Saturation (TSAT): <16% indicates iron-restricted
erythropoiesis⁷.
- C-Reactive Protein (CRP): Elevated CRP suggests active inflammation,
potentially confounding ferritin interpretation.
- Vitamin B12 & Folate: Consider testing, especially in ileal CD
or in patients with restricted diets⁵.
Table 2: Structured Evaluation to Differentiate Causes
Complete Blood Count (CBC) |
Confirms anemia, assesses MCV and severity |
Microcytic = IDA; Normocytic = ACD or mixed |
|
Serum Ferritin |
Marker of iron stores |
<30 µg/L = IDA; 30–100 µg/L with inflammation = possible
deficiency |
|
Transferrin Saturation (TSAT) |
Reflects available circulating iron |
<16% suggests iron-restricted erythropoiesis⁸ |
|
CRP or ESR |
Inflammatory markers |
Elevated in active IBD, supports ACD diagnosis |
|
Vitamin B12/Folate |
Rule out additional deficiencies |
Check in ileal CD or strict vegetarian diets |
|
Reticulocyte Count |
Evaluates bone marrow response |
Low in IDA/ACD; high in hemolysis or recovery ⁸ |
|
Management Strategies
The cornerstone of treating anemia in IBD is identifying the
underlying cause(s) and tailoring treatment accordingly. Iron
replacement is the most common and crucial intervention⁹.
Table 3: Treatment Comparison
Oral Iron |
Mild anemia, inactive disease |
Low cost, convenient |
Poor absorption, GI side effects, may worsen inflammation |
IV Iron (e.g., ferric carboxymaltose) |
Recommended as first-line treatment in patients with clinically
active IBD, with previous intolerance to oral iron and in patients who
need erythropoietin stimulating agents¹ |
Rapid response and bypasses gut absorption |
Requires infusion, risk of hypersensitivity |
Erythropoiesis-Stimulating Agents (ESAs) |
ACD refractory to iron therapy |
Stimulates erythropoiesis |
Requires co-admin of IV iron, costly |
B12/Folate Supplementation |
Documented deficiency usually <200 pg/mL [148 pmol/L]¹ |
Corrects macrocytic anemia |
May be overlooked; needs periodic monitoring |
Treat Underlying Inflammation |
All types of anemia in active IBD |
Resolves root cause of ACD/IDA |
Requires escalation of IBD therapy |
Role of IV Iron in Managing Anemia in IBD
Intravenous (IV) iron therapy is strongly recommended in patients
with moderate-to-severe anemia (hemoglobin <10 g/dL), clinically
active IBD, or intolerance or non-responsiveness to oral iron⁹. In these
scenarios, inflammation impairs intestinal iron absorption and elevates
hepcidin levels, making oral iron ineffective⁸. IV iron bypasses the
gastrointestinal tract, delivering iron directly to the bloodstream for
efficient use in erythropoiesis.
Commonly used IV formulations include ferric carboxymaltose, iron
sucrose, and iron isomaltoside, all of which have been shown to safely
and rapidly restore iron stores and improve hemoglobin levels⁹. Recent
studies suggest ferric carboxymaltose may offer superior efficacy in
fewer infusions, with a lower risk of adverse effects compared to older
formulations⁹.
Guidelines such as those from the European Crohn’s and Colitis
Organisation (ECCO) recommend IV iron as first-line therapy in IBD
patients who are anemic and have active disease, significant iron
deficits, or prior oral iron intolerance⁷. Total iron needs are
calculated based on weight and hemoglobin level, and follow-up is
essential to confirm treatment success and prevent recurrence⁹.
Oral iron remains an option in patients with mild anemia and inactive
disease, but gastrointestinal side effects and limited absorption often
reduce its utility⁸. In patients who do not respond to iron
supplementation or have severe disease, erythropoiesis-stimulating
agents (ESAs) can be considered, usually alongside IV iron.
Additionally, controlling the underlying IBD inflammation is critical to
reducing ongoing iron losses and cytokine-mediated suppression of
erythropoiesis⁸. Nutritional deficiencies should also be corrected to
support optimal red blood cell production⁶.
Summary
Anemia in IBD is a common and impactful complication that requires
careful evaluation and a targeted treatment strategy. Identifying
whether the anemia is due to iron deficiency, inflammation, or a
combination is key to choosing the right therapy. IV iron is often the
preferred treatment in active or severe disease, and addressing the
underlying inflammation is essential for long-term management. Regular
monitoring and a proactive approach can significantly improve outcomes
and quality of life for patients living with IBD¹,⁵.
References
- Hannah Gordon , et al. ECCO Guidelines on Extraintestinal
Manifestations in IBD. Journal of Crohn's and Colitis, 2024, 18,
1–37.
- Filmann N, Rey J, Schneeweiss S, et al. Prevalence of anemia in
inflammatory bowel diseases in european countries: a systematic review
and individual patient data meta analysis. Inflamm Bowel Disease
2014;20:936–45.
- Pels LP, Van de Vijver E, Waalkens HJ, et al. Slow hematological
recovery in children with IBD-associated anemia in cases of ‘expectant
management’. J Pediatr Gastroenterol Nutr 2010 ;51:708–13.
- Wilson A, Reyes E, Ofosu A. Prevalence and outcomes of anemia in
inflammatory bowel disease: A tertiary-center experience. Clinical
Medicine Insights: Gastroenterology,2016. 9, 33–39.
- Thomas G. DeLoughery , et al. AGA Clinical Practice Update on
Management of Iron Deficiency Anemia: Expert Review. clinical
Gastroenterology and Hepatology 2024, Vol. 22, Issue 8.
- Kaufman S, Sigall Boneh R, Wine E. Anemia in pediatric inflammatory
bowel disease: pathophysiology and treatment. Frontiers in
Medicine,2021, 8, 686778.
- Dignass AU, et al. European consensus on the diagnosis and
management of iron deficiency and anaemia in IBD. J Crohns Colitis,2015.
9(3):211–222.
- Weiss G, Gasche C. Pathogenesis and treatment of anemia in
inflammatory bowel disease. Hematology Am Soc Hematol Educ Program,
2015(1), 84–90.
- Danese S, et al. Iron therapy supplementation in inflammatory bowel
disease. Eur J Gastroenterol Hepatol,2024. 36(5):520–527.
Chapter 18: Fertility and Pregnancy in Inflammatory Bowel
Disease (IBD)
Introduction
Inflammatory Bowel Disease (IBD), encompassing Crohn’s Disease (CD)
and Ulcerative Colitis (UC), often manifests during prime reproductive
years. Managing IBD in the context of fertility and pregnancy is
crucial, given its implications on reproductive health, maternal
outcomes, and fetal well-being. This chapter provides an evidence-based
overview that highlights current evidence, patient management
strategies, and clinical recommendations.
1. Fertility in IBD
1.1 Baseline Fertility
Patients with quiescent IBD have fertility rates comparable to the
general population. However, active disease, nutritional deficiencies,
and psychosocial factors can reduce conception rates.¹
Table 1: Factors Influencing Fertility in IBD
Active inflammation |
Decreased |
Disrupts ovulation, alters hormone levels |
Pelvic surgery (e.g., IPAA) |
Decreased (up to 3-fold) |
Causes adhesions, tubal dysfunction |
Sulfasalazine in men |
Reversible decrease |
Affects sperm count and motility² |
Nutritional deficiencies |
Decreased |
Especially iron, folate, vitamin B12 |
Psychological stress |
Decreased |
Impacts libido and sexual function² |
1.2 Impact of Surgery
- IPAA surgery in UC increases infertility risk due to pelvic
adhesions.
- Laparoscopic techniques reduce this risk compared to open
surgeries¹,³.
1.3 Fertility in Men
- Sulfasalazine: Reversible oligospermia.
- Methotrexate: Contraindicated due to teratogenicity; stop 3–6 months
prior¹.
- Biologics, including anti-TNF agents, vedolizumab, and ustekinumab,
have not shown adverse effects on sperm or pregnancy outcomes.
- Surgery in the pelvis may rarely lead to retrograde ejaculation or
erectile dysfunction due to autonomic nerve injury, though laparoscopic
approaches reduce this risk.
- Psychosocial factors, including erectile dysfunction, body image,
and anxiety, may also contribute to perceived infertility⁴.
2. Preconception Counseling
Goal: Achieve deep clinical and endoscopic remission for at least 3–6
months prior to conception. Multidisciplinary counseling should address
nutrition, smoking cessation, and vaccinations.⁵,⁶ Education is crucial
to reassure patients about the safety of most therapies.
2.1 Safe Medications in Preconception and Pregnancy
Table 2: Medication Safety in Preconception and Pregnancy
5-ASA (mesalamine) |
Safe |
Safe |
Safe |
First-line for mild UC |
Azathioprine/6-MP |
Continue if already in use |
Safe |
Safe |
|
Methotrexate |
❌ Contraindicated |
❌ Contraindicated |
❌ Contraindicated |
Teratogenic; discontinue 3–6 months prior ³ |
Corticosteroids |
Use with caution |
Safe (short-term) |
Safe (low dose) |
Risk of gestational diabetes, cleft palate ⁷ |
3. Pregnancy in IBD
3.1 Disease Activity and Pregnancy Outcomes
Women with active IBD at conception are at increased risk of:
- Spontaneous abortion
- Preterm delivery
- Low birth weight
- Small for gestational age (SGA) infants⁷
Maintaining remission is critical for optimizing maternal and
neonatal outcomes.
3.2 Fetal Risks
- Most IBD medications are not teratogenic.
- Disease activity poses more risk than medication continuation¹.
4. Delivery Considerations
4.1 Mode of Delivery
Table 3: Mode of Delivery Recommendations
Quiescent IBD |
Vaginal |
Safe and preferred |
Active perianal CD |
Cesarean section |
To prevent perineal trauma |
History of IPAA |
Cesarean section |
Avoid stress on pelvic floor³ |
4.2 Postpartum Management
- Disease flares may occur postpartum.
- Breastfeeding is encouraged if medications are compatible.
- Close follow-up with gastroenterology and obstetrics is essential³
,⁶.
5. Breastfeeding and IBD Medications
Table 4: Breastfeeding Safety of Common IBD Medications⁶,⁸
Mesalamine |
Safe |
Minimal transfer into breast milk |
Azathioprine |
Safe |
Metabolites present in low concentrations |
Corticosteroids |
Safe (low dose) |
Prefer feeding 4h after dose |
Methotrexate |
❌ Contraindicated |
Excreted in milk; avoid |
6. Safety of Biologic Therapies and Small Molecule Agents
Biologic therapies and small molecule drugs are increasingly used for
moderate-to-severe IBD and are often continued during pregnancy to
maintain remission. Their safety profiles have been a major focus of
recent studies.
6.1 Anti-TNF Agents (Infliximab, Adalimumab, Certolizumab)
- Infliximab and adalimumab cross the placenta (especially in third
trimester) and may remain in infant serum for up to 6 months⁶.
- Certolizumab pegol has minimal to no placental transfer due to
absence of Fc region⁶.
Recommendation: Continue throughout pregnancy; some clinicians stop
at 24–28 weeks to reduce fetal exposure⁶.
6.2 Anti-Integrin Therapy (Vedolizumab)
- Gut-selective, limiting systemic exposure.
- No significant increase in congenital abnormalities reported⁶.
Recommendation: May be used throughout pregnancy if clinically
indicated.
6.3 Anti-IL-12/23 (Ustekinumab)
- Registry reports show no increased risks.
- Crosses placenta similar to other IgG1 antibodies.
Recommendation: Use with caution; reserve for essential disease
control⁶.
6.4 JAK Inhibitors – Tofacitinib, Upadacitinib
- Tofacitinib is teratogenic in animal models; human data are
limited⁷.
- Associated with congenital anomalies in preclinical trials.
Recommendation: Avoid during pregnancy and lactation⁷.
Table 5: Safety of IBD Medications in Pregnancy and Lactation
(Biologics & Small Molecules)
Anti-TNF |
Infliximab, Adalimumab |
Safe (stop at 24–28 wks optional) |
Likely safe |
Avoid live vaccines in infants ≤6 months |
|
Certolizumab pegol |
Safe throughout pregnancy |
Safe |
Preferred in late pregnancy |
Anti-integrin |
Vedolizumab |
Safe (limited data) |
Limited data |
Registry data reassuring |
Anti-IL-12/23 |
Ustekinumab |
Use if needed |
Unknown |
Limited human data⁶,⁷ |
JAK Inhibitors |
Tofacitinib, Upadacitinib |
❌ Contraindicated |
❌ Contraindicated |
Teratogenic ⁷ |
S1P modulators |
Ozanimod |
❌ Contraindicated |
Unknown |
Not approved in pregnancy |
6.5 Vaccination Guidance for Infants Exposed to Biologics
Due to transplacental transfer of IgG1 biologics, avoid live vaccines
in infants until at least 6 months of age⁶,⁸.
6.6 Safety of Newer Biologic Therapies and Small Molecules
Emerging agents like risankizumab, mirikizumab, and brazikumab are
promising but lack extensive pregnancy safety data.
Table 6: Safety of New and Emerging Biologic Therapies in Pregnancy
and Breastfeeding
Anti-IL-23 |
Risankizumab |
Limited human data; animal studies reassuring |
Unknown |
Avoid unless essential¹ |
|
Mirikizumab |
Limited data; caution advised |
Unknown |
Approved for UC |
IL-13/IL-23 |
Brazikumab |
No human data |
Unknown |
Trial phase |
IL-23 (p19) |
Guselkumab |
Limited data; avoid if possible |
Unknown |
Crosses placenta in animals |
S1P Modulator |
Etrasimod |
❌ Contraindicated |
Unknown |
Teratogenic in animals⁷,⁸ |
JAK Inhibitor |
Filgotinib |
❌ Contraindicated |
Unknown |
Testicular toxicity in preclinical studies but no measurable impact
on semen parameters or sex hormones in men⁹ |
7. Summary and Recommendations
- Women and men with IBD can successfully conceive and carry healthy
pregnancies.
- Multidisciplinary care is essential.
- Medication adherence is critical—most IBD therapies are safer than
active disease during pregnancy.
- Encourage preconception counseling and delivery
planning¹⁰,¹¹,¹².
References
- Joana Torres , et al. European Crohn’s and Colitis Guidelines on
Sexuality,Fertility, Pregnancy, and Lactation. Journal of Crohn\’s and
Colitis, 2023, 17, 1–27.
- Shivangi Kothari et al. AGA Clinical Practice Update on
Pregnancy-Related Gastrointestinal and Liver Disease: Expert Review.
Gastroenterology.2024 Oct;167(5):1033-1045.
- Szymańska E, Kisielewski R, Kierkuś J. Reproduction and Pregnancy in
Inflammatory Bowel Disease - Management and Treatment Based on Current
Guidelines. J Gynecol Obstet Hum Reprod. 2021 Mar;50(3):101777.
- Ban L, Tata LJ, et al. Male fertility in IBD: risk of infertility
and sexual dysfunction. Aliment Pharmacol Ther. 2014;39(11):1373-85.
PMID: 24719253.
- Akiyama S, et al. Pregnancy and medications for inflammatory bowel
disease: An updated narrative review. World J Clin Cases. 2023 Mar
16;11(8):1730-1740
- Nielsen, Ole Haagen et al.Updates on the management of inflammatory
bowel disease from periconception to pregnancy and lactation.The Lancet,
Volume 403, Issue 10433, 1291 – 1303.March 30, 2024 .
- Gisbert JP, Chaparro M. Safety of New Biologics (Vedolizumab and
Ustekinumab) and Small Molecules (Tofacitinib) During Pregnancy: A
Review. Drugs. 2020 Jul;80(11):1085-1100
- Innocenti, et al. Pregnancy outcomes in inflammatory bowel disease:
data from a large cohort survey .J Dig Dig. 2022; 23:473-481
- Reinisch W, et al. Effects of filgotinib on semen parameters and sex
hormones in male patients with inflammatory diseases: results from the
phase 2, randomised,double- blind, placebo- controlled MANTA and MANTA-
RAy studies .Ann Rheum Dis 2023;0:1–10. doi:10.1136/ard-2023-224017
- Shmidt, E. ∙ Dubinsky, M.C.Inflammatory bowel disease and pregnancy
Am J Gastroenterol. 2022; 117:60-68
- Selinger CP,et al. IBD in pregnancy: recent advances, practical
management. Frontline Gastroenterol. 2020 May 19;12(3):214-224.
- Friedman S, McElrath TF, Wolf JL. Management of fertility and
pregnancy in women with inflammatory bowel disease: a practical guide.
Inflamm Bowel Dis. 2013 Dec;19(13):2937-48