You are on page 1of 44

Expert Opinion on Biological Therapy

ISSN: 1471-2598 (Print) 1744-7682 (Online) Journal homepage: https://www.tandfonline.com/loi/iebt20

Choosing the most appropriate biologic therapy


for Crohn’s disease according to concomitant
extra-intestinal manifestations, comorbidities, or
physiologic conditions

Ciro Romano, Sergio Esposito, Roberta Ferrara & Giovanna Cuomo

To cite this article: Ciro Romano, Sergio Esposito, Roberta Ferrara & Giovanna Cuomo (2019):
Choosing the most appropriate biologic therapy for Crohn’s disease according to concomitant extra-
intestinal manifestations, comorbidities, or physiologic conditions, Expert Opinion on Biological
Therapy, DOI: 10.1080/14712598.2020.1689953

To link to this article: https://doi.org/10.1080/14712598.2020.1689953

Accepted author version posted online: 05


Nov 2019.

Submit your article to this journal

Article views: 2

View related articles

View Crossmark data

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=iebt20
1

Publisher: Taylor & Francis & Informa UK Limited, trading as Taylor & Francis Group

Journal: Expert Opinion on Biological Therapy

DOI: 10.1080/14712598.2020.1689953

Choosing the most appropriate biologic therapy for Crohn’s disease

t
according to concomitant extra-intestinal manifestations,

ip
comorbidities, or physiologic conditions

cr
us
Ciro Romano¹.; Sergio Esposito¹.; Roberta Ferrara¹.; Giovanna Cuomo¹
an
M
¹University of Campania Luigi Vanvitelli, Piazza Miraglia-Division of Internal Medicine, Department

of Medical and Surgical Sciences, Naples, Italy


ed
pt
ce
Ac
2

ABSTRACT

t
ip
Introduction: Approximately half of Crohn’s disease (CD) patients suffer from concomitant extra-intestinal

manifestations (EIMs). Moreover, CD patients may suffer from comorbidities or have physiologic

cr
characteristics that may influence the outcome of a biologic treatment. Previous guidelines, published when

us
only TNF-α antagonists were available as biological agents, addressed only management of CD patients with

the most common EIMs. an


Areas covered: Because of the recent introduction of new biologics for the treatment of Crohn’s disease as
M
well as increased awareness about comorbidities potentially able to affect the impact of biological drugs,

here we provide an update on management considering old and new biologics with proven efficacy on both
ed

Crohn’s disease and associated conditions, in order to ideally profile the patient to the best of the current

knowledge.
pt

Expert Opinion: While waiting for identification and validation of widely available and reliable biomarkers
ce

able to predict which biologic may yield the best response in the individual IBD patient (rigorous precision

medicine), the choice of biologic agents in CD patients in order to achieve the best outcome still lies on a
Ac

thorough assessment of patient-related characteristics as well as deep knowledge of the properties and place

in therapy of each biologic drug.

Keywords: Crohn’s disease, biologic therapy, extra-intestinal manifestations, comorbidities, pregnancy,

elderly patients
3

t
ip
cr
ARTICLE HIGHLIGHTS

us
• Real-life Crohn’s disease (CD) patients often suffer from concomitant extra-intestinal manifestations
an
and comorbidities

• The current availability of biologic drugs with different molecular targets requires comprehensive
M
knowledge of their characteristics for optimal management of CD patients with coexisting extra-

intestinal manifestations and comorbidities


ed

• Tailored treatment suggestions for extra-intestinal manifestations and comorbidities simultaneously

occurring in CD patients are provided based on the available evidence


pt

• Tailored treatment suggestions are provided for such physiologic conditions as pregnancy and old

age based on the available evidence


ce
Ac

1. INTRODUCTION

Crohn’s disease (CD) is a chronic inflammatory disease of the gastrointestinal tract, typically involving the

terminal ileum and colon, although all segments of the gastrointestinal tract can be affected. The course of

the disease is progressive, with a relapsing and remitting inflammatory pattern, often leading to such

complications as strictures, fistulas, or abscesses ultimately requiring surgery [1]. Moreover, CD may be

associated with extra-intestinal manifestations (EIMs) of disease, some of which may even predate intestinal

involvement. Indeed, up to 50% of patients present with cutaneous, articular, or ocular EIMs that can
4

precede diagnosis. Among these, erythema nodosum and oligoarticular large joint arthritis are typically

associated with active intestinal disease, while axial spondilarthritis or primary sclerosing cholangitis are

independent of disease activity [2]. Ideally, therapy of CD should warrant sustained remission, so as to stop

the course of the disease, control EIMs, and prevent complications. The introduction of biologic drugs in the

armamentarium of therapies for inflammatory bowel disease (IBD) has improved the outcome of the disease;

however, a substantial proportion of patients may not satisfactorily respond to these therapies thus requiring

t
eventually surgery [3]. In some instances, the characteristics and/or associated comorbidities of CD patients

ip
may have a negative impact on the successful outcome of biologic therapy; in other cases, the choice of a

cr
specific biologic agent should also take into account the associated EIM, since some biologic drugs work

well on CD but not on associated EIMs. Although guidelines for management of EIMs are already available

us
[4], here we provide an update considering the latest evidence and focus on the use of biologic drugs in

specific CD contexts, beyond EIMs, that may favor choice of a biologic agent over another because of a
an
predictable better outcome, so as to ideally profile the patient to the best of the current knowledge.
M

1.1 BIOLOGIC DRUGS CURRENTLY AUTHORIZED FOR CROHN’S DISEASE TREATMENT


ed

Six biologic agents have thus far been granted approval for use in CD. They may be classified according to

structural characteristics, mechanism of action, route and frequency of administration. Half of them share the
pt

same molecular target (i.e., TNF-α). Table 1 summarizes the main characteristics of each biologic agent
ce

currently available for CD treatment. Natalizumab has primary indication for multiple sclerosis; it is also

available for CD in the USA.


Ac

1.2 SPECIFIC CONDITIONS

Real-life CD patients eligible for biologic therapy may present with concomitant EIMs, comorbidities, or

specific physiologic conditions (Table 2) that may raise more than one question as to which biotherapy may

be more appropriate considering the whole clinical picture. Management suggestions for common conditions

are detailed below.


5

2. EXTRA-INTESTINAL MANIFESTATIONS

2.1 Enteropathic spondyloarthritis

Joint involvement is the most common EIM of CD [5,6]. Patients may present with peripheral or

axial involvement, collectively known as enteropathic spondylarthritis. Peripheral joint arthritis

occurs in 10-20% of CD patients, is usually non-erosive and non-deforming, and may present with

t
ip
an oligoarticular or polyarticular pattern. Oligoarticular involvement is characterized by asymmetric

arthritis, usually of large joints of lower limbs (i.e., hips, knees, and ankles), whose activity parallels

cr
that of CD; polyarticular involvement is typically symmetric and also affects small joints, with

us
longstanding pain and swelling, independently of CD activity. Thus, control of oligoarticular arthritis

is usually achieved with therapies aimed at controlling CD, while polyarticular arthritis may require
an
an independent treatment strategy [7].

Axial involvement, suggested by persistent inflammatory back pain, is underlain by sacroiliitis. Five
M
to 20% of CD patients may suffer from axial spondylarthritis. Magnetic resonance imaging is the

diagnostic tool of choice to reveal inflammation of one or both sacroiliac joints, even before sequelae
ed

of sacroiliitis become apparent on conventional radiology. Like polyarticular peripheral arthritis,

axial involvement runs an independent course from that of CD, thus possibly predating CD and
pt

needing exclusive treatment. Finally, axial and peripheral joint involvement may coexist in 3-6% of

CD patients [2,7]. The correct recognition of the pattern of articular involvement, along with
ce

assessment of CD activity, is paramount in order to implement the most appropriate biologic

treatment [8]. Since guidelines for treatment of CD and spondylarthritis are separately available [9-
Ac

11], close collaboration between gastroenterologists and rheumatologists is crucial in order to ensure

optimal management of combined conditions. In general, judicious application of guidelines, based

on recognition of the prevalently active disease and the type of biologic agents suitable for treatment

of both conditions, is usually advised.

Treatment of peripheral arthritis differs depending on the oligoarticular or polyarticular pattern and

coexistence of quiescent or active CD; in general, peripheral arthritis is observed with higher
6

frequency in colitis than in ileitis [12]. The same therapeutic considerations apply to axial

involvement. The basic clinical scenarios are as follows (Figure 1):

- In case of peripheral oligoarthritis, since this type of articular involvement is self-limiting and

parallels intestinal disease activity, if biologic treatment is necessary to control CD activity, either

TNF-α inhibitors or ustekinumab at the doses and administration schedules used in gastroenterology

usually suffice for concomitant control of peripheral oligoarthritis (Table 1). Vedolizumab, despite

t
being highly specific for a gut-selective integrin [13], may also be considered for this type of

ip
arthritis, perhaps better after failure of TNF-α inhibitors or ustekinumab on CD activity control,

cr
since the main goal of biologic therapy in this setting is to dampen intestinal inflammation; in fact,

following control of CD, this type of arthritis usually resolves. Indeed, clinical benefit of

us
vedolizumab therapy on inflammatory arthralgia/arthritis has been documented in patients with

synchronous occurrence of intestinal and articular inflammation [14,15].


an
- In case of peripheral polyarthritis, biologic therapy differs depending on the activity of underlying

CD. As stated above, this type of arthritis is independent of CD activity; thus, it may occur with or
M

without active CD. In case of active CD, both TNF-α inhibitors and ustekinumab may be used,

according to the doses and schedules used in gastroenterology; if CD is quiescent, the same agents
ed

may be administered, following the doses and the schedules used in rheumatology (Table 1).

Vedolizumab in this context may not be the most appropriate choice, since this agent has been
pt

observed to be associated with flares of arthritis in several case series [14-17], despite α4β7 integrin
ce

has also been found on leukocytes infiltrating the synovium [18,19]. However, in some instances,

caution should be exercised in interpreting this cause-effect relationship. In fact, when vedolizumab
Ac

is introduced in therapy following failure of a TNF-α inhibitor to control CD activity, arthritis flares

may be due to loss of the effect of the previous biologic drug on arthritis activity rather than to “true”

vedolizumab-induced arthritis [16].

- In case of axial involvement, which is independent of CD activity, the type of therapy again depends

on the underlying CD activity. Since long-term use of nonsteroidal antinflammatory drugs

(NSAIDs), which have been shown to be useful in controlling axial spondylarthritis [11], are

contraindicated in CD patients, resort to biologic therapy is more immediate. In case of quiescent


7

CD, first-line biologics are TNF-α inhibitors, since randomized trials of ustekinumab in patients

with axial spondyloarthritis failed to achieve the key primary and secondary endpoints, despite

preliminary evidence of positive outcomes in this setting [20]. This also means that, following

failure of a first TNF-α inhibitor, a second agent in the same class should be tried. Doses and

administration schedules are per rheumatologic recommendations (Table 1). In case of active CD

and axial disease, TNF-α inhibitors should be administered following the protocols used in

t
gastroenterology. For vedolizumab, considerations are the same as for peripheral polyarthritis, that

ip
is, there is no evidence of clinical benefit on axial disease.

cr
- In case of overlapping features (i.e., concomitant peripheral and axial involvement), considerations

are the same as in case of isolated axial involvement.

us
- If predominant manifestations of spondylarthritis are enthesitis and/or dactylitis, the most effective
an
strategy is use of biologic agents [21]. Sound choices are, again, TNF-α inhibitors or ustekinumab,

at doses and schedules depending on the underlying activity of CD. Vedolizumab is not considered
M
appropriate for concomitant treatment of enthesitis and/or dactylitis.
ed

2.2 Erythema nodosum

Up to 15% of patients may suffer from recurrent episodes of erythema nodosum (EN), the most
pt

common cutaneous EIM of CD. EN is classified as a panniculitis, as it involves the subcutaneous fat,
ce

and appears to be more frequent in patients with isolated colonic involvement [22]. EN presents with

raised, tender, red or violet, 1- to 5-cm subcutaneous nodules, typically located bilaterally on the
Ac

extensor surface of lower limbs, is self-limiting, and resolves without sequelae. Its course usually

depends on the underlying CD activity. Since control of CD activity is usually propedeutic to

achieving remission of EN episodes, any type of available biologic therapy, if necessary for control

of CD activity, can be chosen by the clinician, as appropriate. Real-life data from the latest European

Crohn’s and Colitis Organisation (ECCO) conference have also indicated efficacy of ustekinumab on

EN in case of failure of prior anti-TNF-α treatment, while vedolizumab therapy appeared to be less

effective [23]. Nevertheless, since EN has been found to be frequently associated with peripheral
8

arthritis [22,24], biologic therapy should be implemented primarily considering the biotherapies

effective on joint manifestations (i.e., TNF-α inhibitors or ustekinumab). Other possible associations

are with ocular involvement and pyoderma gangrenosum [22], which should also be taken into

account when deciding the choice of biologic therapy (see next paragraphs for details and figure 2).

Refractory cases usually respond well to TNF-α antagonists [25-27], if not used as first-choice

biologic agents.

t
ip
2.3 Pyoderma gangrenosum

As opposed to EN, pyoderma gangrenosum (PG) is a severe, debilitating EIM of IBD, more

cr
frequently observed in ulcerative colitis than in CD. In the latter case, PG usually follows onset of

us
CD in most of the patients [7]. PG is characterized by single or multiple painful erythematous

papules or pustules, evolving in deeply excavated ulcerations with purulent but sterile material,
an
characterized by high recurrence rates. The lesions usually localize on shins or the peristomal area,

although any skin district can be affected, particularly at sites of previous trauma (e.g., venipuncture
M
or biopsy), suggesting pathergy phenomenon [28]. PG may parallel activity of underlying CD or run

an independent course; its incidence in CD is between 0.1-1.2%. Typical associations are with
ed

pancolitis, permanent stoma, EN, and ocular involvement [28]. Since the disease course is severely

debilitating, early, aggressive treatment is warranted in order to attempt rapid resolution of lesions.
pt

For these reasons, biologic therapy may be an early option when considering treatment strategies.

TNF-α antagonists, the only biotherapies available for IBD treatment at the beginning of the
ce

biologic era, were tried first in the treatment of recalcitrant PG cases, based on a hypothetical shared
Ac

pathomechanism with CD. Indeed, blocking TNF-α resulted in healing of PG in a number of case

reports and small series [25,29-32], making therapies with anti-TNF-α monoclonal antibodies sound

choices for changing the course of the disease. More recently, some authors have documented high

expression of IL-23 in a recalcitrant PG lesion, both at the transcriptional and protein level. This

finding led to a treatment trial with ustekinumab, an inhibitor of both IL-12 and IL-23, which

resulted in complete healing within 14 weeks of treatment [33]. Targeting IL-23 was then confirmed

to be another effective treatment strategy for IBD-associated PG, with clinical benefit also reported
9

for peristomal localization [34-36]. Recent real-life data from the latest ECCO conference also

support use of ustekinumab for inducing remission of PG in patients failing therapy with TNF-α

inhibitors [23]. Ustekinumab efficacy is not surprising, given the prior positive experiences with

anti-TNF-α strategies in PG, since IL-23 appears to act upstream of TNF-α, therefore inhibition of

IL-23 may lead to TNF-α downregulation [37]. Conversely, no efficacy data have emerged for

vedolizumab [23].

t
Finally, in case of multiple associations, a biologic agent known to be possibly effective on all

ip
manifestations should be used (Figure 2).

cr
us
an
2.4 Scleritis and uveitis
M
Scleritis and uveitis represent the most bothersome ocular EIM of CD. In general, eye

manifestations, including also such mild conditions as conjunctivitis and episcleritis, occur in 2-6%
ed

of patients and usually are associated with joint involvement [38]. Management of scleritis and

uveitis requires close collaboration with an ophthalmologist, since ocular involvement may lead to
pt

visual impairment up to loss of vision in untreated patients. Eye manifestations usually follow the

course of CD activity; hence, treatment of the underlying IBD with conventional or biological
ce

therapies, as appropriate, along with the therapy suggested by the ophthalmologist, may suffice for

control of ocular inflammation [7]. Refractory cases naïve to biologic therapy have been successfully
Ac

treated with TNF-α inhibitors [39-42]. Particularly, adalimumab has been shown to be effective in

patients with different noninfectious forms of uveitis despite continued steroid treatment for at least

2 weeks and to lower the risk of uveitic flare or loss of visual acuity upon corticosteroid withdrawal

[43,44]. Ustekinumab may be tried as a rescue therapy in case of failure of TNF-α inhibitors [45-47],

while awaiting for efficacy confirmation from ongoing trials investigating the effect of IL-12/IL-23

blockade in sight-threatening uveitis (ClinicalTrials.gov identifiers: NCT02911116, NCT03847272).


10

2.5. Primary sclerosing cholangitis

Primary sclerosing cholangitis (PSC), a chronic cholestatic disease due to inflammation and

progressive fibrosis of the intrahepatic and extrahepatic bile ducts, is strongly associated with IBD,

since nearly 80% of PSC patients suffer from concomitant IBD [48]. Conversely, less than 10% of

IBD patients have an attendant condition of biliary duct inflammation. Treatment outcomes are

t
ip
disappointing since no medical therapies have been shown to prevent progression in PSC [49]. On

the other hand, the role of biologics in PSC remains uncertain. Although TNF-α has been found to

cr
promote inflammation around bile ducts in PSC through recruitment of T cells [50,51], there is no

us
robust evidence that TNF-α inhibitors may be effective. Indeed, although a transient benefit,

evaluated as a decrease in alkaline phosphatase (ALP) levels, was observed for adalimumab as
an
opposed to infliximab and vedolizumab in the largest study published so far, the authors concluded

that current biologic therapies do not appear to be effective for treatment of PSC [52]. A transient
M
reduction in ALP levels was also seen in 2 out of 3 adalimumab-treated patients in a smaller case

series [53]. In that study, one patient on infliximab had reduced γ-glutamyltransferase levels after 6
ed

and 12 months of therapy, but liver function tests tended to deteriorate thereafter [53]. With regard to

vedolizumab, the rationale for its use in IBD-associated PSC stems from the observation of gut
pt

adhesion molecules also in the PSC liver, which could mediate recruitment of gut-derived, α4β7-

expressing effector T cells from the inflamed bowel to the liver [54]. While this rationale has not yet
ce

translated into clinical benefit [52], there has also been a report of acute cholestasis after
Ac

vedolizumab treatment, progressing to chronic liver injury despite immediate drug withdrawal and

corticosteroid administration [55]. Nevertheless, a recent review suggests that TNF-α antagonists

may benefit subgroups of CD-associated PSC patients by either resolving intestinal inflammation or

by a direct effect on the liver; similarly, vedolizumab may work in subsets of IBD patients with

concomitant PSC [56]. However, biomarkers capable of identifying potential responders are

currently lacking. Thus, while waiting for innovative therapies, able to induce a paradigm shift in the

treatment of PSC, if the clinician’s choice for biologic therapy falls on TNF-α antagonists,
11

adalimumab may be a candidate drug among this class of biologics to treat CD patients with

attendant chronic inflammation of bile ducts, on the basis of the available, albeit scarce, evidence.

2.6. Glomerulonephritis

Parenchymal renal involvement in IBD is rare [57,58]. IgA nephropathy is by far the most common

t
type of glomerulonephritis reported in IBD patients, followed by membranoproliferative and

ip
minimal change glomerulonephritis [57,58]. Inclusion of glomerulonephritis among EIMs of CD is

cr
suggested by the general improvement of renal function following achievement of intestinal

remission [59]. Therefore, in patients with concomitant nephropathy, the primary goal is to treat the

us
underlying bowel inflammation to limit renal damage. This concept holds particularly true for

prevention of amyloidosis, which cannot be considered a classic EIM of CD, but rather a
an
complication occurring when chronic bowel inflammation is not appropriately abated in a timely

fashion [58,59]. The typical clinical presentation of CD-related amyloidosis (∼70% of patients) is
M

renal failure and nephrotic range proteinuria. Once CD patients present with overt amyloidosis and

initiation of biologic therapy is considered, the clinician should opt for TNF-α inhibitors. Indeed,
ed

infliximab has been shown to decrease serum amyloid A circulating levels and proteinuria as well as

to stabilize renal function and to improve survival in several reports [60-65]. However, it is unclear
pt

if reversibility of established damage can be achieved. Of importance, due to their high molecular
ce

weight, TNF-α inhibitors are not affected by glomerular filtration rate; thus, no adjustments in

dosing are needed based on creatinine clearance.


Ac

2.7 Neurologic manifestations

Peripheral neuropathies are the most common neurological manifestations in patients with CD.

These neuropathies are generally independent of intestinal activity and do not respond to IBD-

specific treatment [66,67]. Further complicating matters are reports describing onset of peripheral

neuropathy following therapy with TNF-α antagonists [68-72], which is nonetheless a rare adverse
12

event overall [71]. Conversely, a beneficial effect of these biotherapies on peripheral nerve

involvement complicating CD has been much rarely reported [73]. However, since evidence is

anecdotal, there is no reason to exclude a priori TNF-α inhibitors from the possible therapeutic

choices in CD patients with attendant peripheral neuropathy. Clearly, a vigilant eye must be kept on

the outcome of neurological involvement so as to promptly swap to alternatives to TNF-α

antagonists (i.e., ustekinumab or vedolizumab), in case of unexpected worsening.

t
With regard to central nervous system (CNS) involvement associated with IBD, demyelinating

ip
diseases such as multiple sclerosis (MS) and ischemic optic neuropathy are the most common

cr
entities [67]. TNF-α inhibitors are contraindicated in patients with demyelinating diseases since they

may exacerbate disease or trigger demyelination [74]. Although well-tolerated, ustekinumab did not

us
show efficacy in relapsing-remitting MS in a phase II clinical trial [75]. Natalizumab has been the

first monoclonal antibody approved for MS treatment [76]; in countries where natalizumab is also
an
indicated for treatment of CD, this monoclonal antibody may be the best choice for combined CD

and MS.
M

3. COMORBIDITIES
ed

3.1. Obesity

Obesity is increasing in prevalence among CD patients and may constitute a further challenge in their
pt

management [77]. Indeed, obese patients have been shown to experience earlier loss of response during

biological therapy [78,79] and higher rates of postoperative complications as compared to lean subjects
ce

[80]. With regard to biologic therapy, obesity has been demonstrated to hamper the effects of TNF-α
Ac

inhibitors [78,79]; particularly, studies in obese rheumatoid arthritis patients revealed that infliximab had

the worst performance when compared to other TNF-α antagonists [81], probably due to a lower

distribution volume with respect to other same class biologics [82,83]. However, infliximab treatment in

obese CD patients may be optimized by increasing doses both at the induction phase and during

maintenance according to serum concentrations, suggesting that a personalized treatment strategy may

aid overcoming impaired treatment responses in this setting [84]. Specifically, infliximab trough

concentrations >3.5 μg/ml at week 14 have been associated with sustained response to therapy in CD
13

[82]; moreover, keeping trough concentrations at ≥5 μg/ml during maintenance therapy are considered

sufficient for infliximab to exert its clinical effects. In practice, loss of response on ideal trough

concentrations should prompt a change in biologic therapy (secondary loss of response), while a trough

concentration below the above threshold combined with an unsatisfactory clinical response should

encourage dose escalation [82].

Ustekinumab efficacy may also be limited by excess body weight [85,86]. However, in studies carried

t
out in psoriasis patients, doubling the 45-mg dose in patients weighing > 100 kg was associated with

ip
improved response rates [87,88]. In CD patients, the i.v. induction dose is calculated based on body

cr
weight while the maintenance phase includes only the 90-mg dose. Moreover, dosing intervals may be

reduced to 8 weeks based on clinician judgement of efficacy, as opposed to the 12-week dose intervals

us
typically used in protocols of psoriasis and psoriatic arthritis treatment. Again, therapeutic drug

monitoring may help decide how to best manage administration of ustekinumab in obese patients.
an
Depending on the select outcome (i.e., clinical response at week 8, biological response at week 8, clinical

and biochemical response at week 16, 50% decrease in fecal calprotectin at week 8, etc.) and the time
M

point of drug trough level assessment (week 4 or week 8 from start of therapy), a number of serum drug

thresholds ranging from >2 to >15.9 μg/ml have been established as predictors of good response [89].
ed

Thus, tailoring ustekinumab doses and administration intervals upon clinical judgement and therapeutic

drug monitoring may help achieve treatment goals in obese patients as well. There are no sufficient data
pt

about the impact of body weight on vedolizumab efficacy in CD; however, a retrospective study in
ce

biologic-treated ulcerative colitis patients also identified a high body mass index as a limiting factor for

vedolizumab efficacy [90]. If feasible, measuring vedolizumab levels at week 6 may be useful for
Ac

outcome prediction in IBD patients, since approximately >20 μg/ml have been shown to be associated

with improved clinical responses, including mucosal healing rates during maintenance. However, drug

thresholds predictive of good outcome may vary (from >18 to >35.2 μg/ml) depending on the time of

measurement (week 2, week 6, or week 14 from start of therapy) and the desired outcome (clinical

response at week 6, biochemical remission at week 6, clinical response at week 14, mucosal healing

within 52 weeks, sustained remission, etc.) [89]. Thus, measuring serum levels of vedolizumab may help

adjusting the therapeutic dose before concluding for a lack of response [91].
14

Finally, body weight has also been shown to be the most important factor associated with natalizumab

effective distribution [92]. Indeed, natalizumab efficacy has been demonstrated to be dependent on

saturations of VLA-4 on lymphocytes of more than 70%, and the median saturation of Very Late

Antigen-4 (VLA-4) on lymphocytes declines with increasing body weight [93]. However, since patients

heavier than 75 kg still show lymphocyte VLA-4 saturations greater than 85%, body weight may be an

issue in patients with frank obesity. Nevertheless, since natalizumab is associated with the development

t
of progressive multifocal leukoencephalopathy (PML) and no tests are currently available for reliable

ip
prediction of those at risk of developing PML, increasing natalizumab doses may further predispose

cr
patients to this potentially fatal complication [94]; therefore, it is not an advisable therapeutic choice in

obese patients due to the anticipated need of using higher than usual doses.

us
3.2. Non-alcoholic fatty liver disease (NAFLD)/non-alcoholic steatohepatitis (NASH)
an
According to a recent systematic review with meta-analysis including 5620 patients, the overall pooled

prevalence of NAFLD in IBD patients has been calculated to be 27.5%, with even higher rates in elderly
M

patients, coexisting metabolic comorbidities, methotrexate use, prior surgery, and longer duration of IBD

[95]. Importantly, NASH poses an increased risk of morbidity and mortality due to possible progression
ed

to liver cirrhosis. Unfortunately, there is no established medical treatment for NASH, owing to an

incomplete understanding of its pathogenesis. According to several lines of evidence, such


pt

proinflammatory cytokines as TNF-α and interleukin (IL)-6 may be involved in the pathogenesis of
ce

NASH by promoting liver inflammation, insulin resistance, and hepatocyte apoptosis [96]. However,

anti-TNF-α therapy has not been evaluated for the treatment of NASH. Notwithstanding, anecdotal
Ac

reports highlight the beneficial effects of TNF-α inhibitors on altered liver histology and biochemistry

due to experimentally-induced or naturally occurring NASH in mouse and human hosts, respectively

[97,98]. Thus, based on the scant evidence available and while waiting for confirmatory results in large

ad hoc studies, patients with active CD and concomitant NAFLD/NASH may be considered for biologic

therapy with TNF-α inhibitors; moreover, in case CD is in control but concomitant peripheral arthritis is

bothersome to the NAFLD/NASH patient, biologic therapy may be started at rheumatologic doses just

for control of this EIM, skipping the trial with conventional antirheumatic drugs (e.g., sulphasalazine or
15

methotrexate) because of concerns about possible liver toxicity. Finally, since NAFLD/NASH is

commonly observed in overweight/obese patients (Figure 2), dosage and administration schedules of

TNF-α inhibitors should be adjusted taking into account the role of body weight on efficacy (Figure 2),

as previously detailed.

3.3. Diabetes mellitus

t
ip
Patients with IBD may have an increased risk of developing type 2 diabetes mellitus [99]. Insulin

resistance is the most important pathophysiologic feature of type 2 diabetes mellitus and prediabetic

cr
states. Since TNF-α has been shown to contribute to the pathogenesis of insulin resistance during the

us
course of chronic inflammatory diseases [100], therapies aimed at neutralizing TNF-α should

theoretically result in amelioration of insulin resistance. Indeed, several pieces of evidence point to a
an
beneficial role of TNF-α inhibitors in improving glucose homeostasis, both in experimental models and

human subjects [101-103]. Noteworthy, relapse of diabetes has been observed following discontinuation
M
of infliximab [104], further strengthening the role of TNF-α in modulating insulin resistance. Although

available data supports the concept that targeting inflammation improves insulin sensitivity and
ed

pancreatic β-cell function, thus suggesting preferential use of TNF-α inhibitors for CD patients suffering

from concomitant type 2 diabetes mellitus, it should also be remembered that the positive metabolic
pt

effects registered in most clinical trials carried out so far have been rather modest [105]. Finally, the

higher prevalence of fatty liver disease in diabetic patients may, again, suggest early implementation of
ce

biologic therapy in this subset of patients (i.e., CD and type 2 diabetes and fatty liver disease) if

peripheral arthritis also coexists, in order to avoid possible adverse effects of conventional antirheumatic
Ac

drugs used for relief of joint inflammation on the liver (Figure 2).

On a purely theoretical basis, as experimental and clinical data are still insufficient, the combination of

type 1 diabetes mellitus and CD would be best approached by using ustekinumab. In type 1 diabetes

mellitus, proinflammatory pathways are mainly orchestrated by Th1 and Th17 cells, T cell subsets

secreting interferon-γ (IFN-γ) and IL-17, respectively [106]. IL-12 and IL-23 are crucial cytokines for
16

sustaining Th1 and Th17 cell activity. Ustekinumab, a dual inhibitor of IL-12 and IL-23, has been shown

to be effective in dampening IFN-γ- and IL-17-driven inflammation [106].

In both type 1 and type 2 diabetes mellitus, it is wise to start biologic therapy when glucose homeostasis

is under control, since decompensated diabetes may represent an additional risk for serious infections

during biologic therapy.

t
ip
3.4. Cardiovascular disease

TNF-α inhibitors have long been known to worsen heart failure [107]. Conversely, there are no reports

cr
of exacerbation of heart failure using biologics with a different molecular target. Therefore,

us
ustekinumab, vedolizumab or natalizumab may be used in place of TNF-α inhibitors, as appropriate, in

patients with CD and concomitant heart failure. TNF-α antagonists should only be used if there are no
an
other reasonable options, and then, perhaps, only in compensated heart failure.

Conversely, anti-TNF-α treatment has been shown to reduce cardiovascular risk by preventing
M

progression of subclinical atherosclerosis and arterial stiffness in patients with different types of chronic

inflammatory diseases [108,109]. Indeed, a chronic inflammatory state is deemed to be responsible for
ed

accelerated atherosclerosis in these patients [110]; however, whether this effect is specific to TNF-α

inhibitors or relates to better control of chronic inflammation, regardless of the therapeutic strategy
pt

implemented, is currently unknown. Therefore, early control of chronic inflammation may be beneficial
ce

for reducing cardiovascular risk and this should be borne in mind when deciding whether to scale up

therapy, particularly in case of other concomitant cardiovascular risk factors (i.e., smoking, hypertension,
Ac

dyslipidemia, etc.).

3.5. Psoriasis

Common genetic and inflammatory pathways have been implicated in psoriasis and IBD [111], therefore

it is not surprising that the two conditions are frequently associated [112]. Patients with CD and

comorbid psoriasis may be effectively treated with TNF-α inhibitors or ustekinumab [113-118], whereas

vedolizumab and natalizumab are not considered effective on skin manifestations. It should not be
17

overlooked that paradoxical psoriasis may occur during treatment with biological agents, particularly

TNF-α inhibitors [119]. Female gender and history of extra-intestinal manifestations have been

suggested to predispose to development of paradoxical psoriatic lesions. Stopping TNF-α inhibitor

therapy and swapping to ustekinumab has been shown to result in complete remission in nearly all cases,

while maintaining or attaining intestinal remission [120].

t
ip
3.6. History of infections

cr
A recent systematic review and meta-analysis including a total of > 5000 IBD patients from different

trials found no increased infection risk in CD patients treated with TNF-α inhibitors, as opposed to

us
ulcerative colitis patients who were shown to be at increased infection risk vs. placebo with the same

biotherapies [121]. Anti-integrin therapy was found to be safe as well. Nevertheless, in patients with a
an
history of frequent serious infections (i.e., requiring hospitalization), ustekinumab or vedolizumab may

be prudentially considered as first choices in accordance with the patient’s characteristics.


M
ed

4. PHYSIOLOGIC CONDITIONS

4.1 Pregnancy and child-bearing age


pt

Although IBD increases the risk of pregnancy complications irrespective of disease activity, adverse
ce

pregnancy outcomes occur more frequently in case of active disease [122]. Thus, CD occurring during

child-bearing age raises concerns about the impact of the disease and its treatment on fertility, maternal
Ac

and fetal health during pregnancy, and breastfeeding safety. Ideally, women should conceive while CD is

quiescent. With regard to biologic therapy, collective evidence from hundreds of pregnancies in chronic

inflammatory diseases have suggested that exposure to TNF-α inhibitors at the time of conception or

during the first trimester is not associated with an increased risk of adverse pregnancy and fetal outcomes

[123]. However, monoclonal antibodies do cross the placenta during the second and third trimester and

have been shown to be functional in the fetus [123]. Nevertheless, data from the TREAT registry,

summarizing a 13-year experience with infliximab given before or during pregnancy, showed that the
18

clinical condition of infants born to women with gestational infliximab exposure was similar to those

without exposure, even though a lower live birth rate was recorded among infliximab-exposed women

[124]. However, these patients had more severe CD and were more likely to have been exposed to

immunosuppressive drugs [124]. Likewise, the recent European multicenter TEDDY study did not find

an increased short-term and long-term risk of severe infections in children of mothers with IBD, who had

been exposed to TNF-α inhibitors in utero with respect to children never exposed to such drugs [125].

t
Still, among TNF-α inhibitors, is now available a modified monoclonal antibody lacking the Fc

ip
fragment, namely, certolizumab pegol, which has become first choice, where available, for biologic

cr
therapy during pregnancy. In fact, certolizumab pegol is unable to bind the neonatal Fc receptor and thus

does not cross the placental barrier, as opposed to other therapeutic monoclonal antibodies [126,127].

us
Consistently, pregnancy outcomes after exposure to certolizumab pegol do not suggest harmful effects

on fetal/child health, such as teratogenicity or increased risk of fetal death [128,129]; moreover,
an
certolizumab pegol treatment can be continued during breast feeding as no or minimal (but clinically

insignificant) transfer occurs into maternal milk [130]. In a wider perspective, apart from pregnancy, it
M

may be judicious to start treatment of sexually-active CD patients in child-bearing age with certolizumab

pegol, since this approach will not need adjustments even in case of planned or unexpected pregnancies.
ed

Besides, most of the EIMs of CD are amenable to treatment with certolizumab pegol. Certolizumab

pegol, however, is licensed for treatment of CD only in the USA and Switzerland.
pt

The data for ustekinumab in pregnancy are limited to case studies and registry data [131-133].
ce

Experience in the field of rheumatology suggests that ustekinumab does not result in an increased risk of

miscarriage or congenital malformation [134]. However, controlled data are lacking and continuing
Ac

ustekinumab throughout pregnancy is a matter of debate [135]. Likewise, limited data are available about

outcomes of pregnancies exposed to vedolizumab [136-140]. No significant safety concerns have been

thus far identified, although vedolizumab-exposed pregnancies have been found to be associated with

more complications in both mothers and infants [139,140]; however, when excluding from the analysis

patients with active disease, no significant difference was observed with respect to pregnancies exposed

to TNF-α antagonists or to conventional therapy, suggesting that complications may more likely arise

because of active disease in pregnancy rather than as a drug adverse effect [139]. Vedolizumab may
19

nonetheless be considered during pregnancy only if strictly necessary, with benefits to the mother

outweighing the risks to the mother/unborn child, as complications cannot be completely excluded [140].

Larger prospective studies are awaited to clarify this issue.

4.2. Elderly patients

The widely accepted definition of elderly-onset IBD is disease occurrence at an age of 60 years or older

t
ip
[141]. About 25-35% of the IBD population falls within the age range of elderly people, with about 15%

having true late-onset IBD and about 20% transitioning into older age having been diagnosed with the

cr
disease at a younger age [142,143]. Infections and related serious complications are more common in

us
elderly IBD patients [144], and elderly IBD patients treated with TNF-α inhibitors have an increased risk

of severe infection compared with younger patients [145]. Moreover, the frequent coexistence of heart
an
failure in the elderly population makes TNF-α inhibitors unsuitable for treatment of CD (Figure 2). By

contrast, ustekinumab and vedolizumab appear to have a better safety profile and may thus be
M
prudentially chosen as primary biotherapies [146-148].
ed

5. CONCLUSIONS
pt

Choosing the right drug for the right patient at the right time is one of the biggest unmet needs in current IBD

care. While waiting for further studies to address these issues, a thorough assessment of patient
ce

characteristics is necessary to choose the biologic agent likely to work best in each specific case.
Ac

6. EXPERT OPINION

TNF-α inhibitors have been the mainstay of treatment for IBD since their introduction in the market as first

available biotherapies and still have a prominent role in the treatment of CD. Subsequently, a number of

further biologic agents with a different mechanism of action (i.e., targeting different molecules involved in

inflammation or leukocyte migration) have obtained market authorization and have been shown to be

suitable options for treatment of CD patients. Since real-life patients with CD often suffer from associated
20

comorbidities or EIMs, which are usually excluded from or unaccounted for in clinical trials, the choice of

biologic therapy in the everyday clinical practice should take into account all possible patient-related

variables capable to affect treatment effectiveness.

In the next future, innovative therapies are likely to receive market authorization for treatment of CD. As the

pathophysiologic mechanisms of CD and associated EIMs and comorbidities are progressively unraveled,

finely tuned targeted therapy may become available capable to control the whole spectrum of disease.

t
Meanwhile, due to the association with PML, the availability of alternative safer biologic agents, and the

ip
introduction of newer drugs, natalizumab is less likely to be used in CD patients failing conventional medical

cr
therapy in the next future. Conversely, the use of other biologic agents is likely to be refined as more clinical

data become available with respect to specific subsets of real-life CD patients.

us
Innovative therapies are now mainly represented by so-called small oral molecules, which act intracellularly

through unique mechanisms of action. Specifically, their mechanism of action involves inhibition of pro-
an
inflammatory cytokine synthesis through intracellular Janus kinase (JAK) signaling inhibition, inhibition of

the TGF-β pathway, or interference with lymphocyte trafficking via sphingosine 1-phosphate (S1P) receptor
M

modulation [149]; in addition, some other small molecules with different molecular targets are in the pipeline

[150]. There is, however, weak evidence of substantial efficacy in CD to date, although some of these
ed

molecules have demonstrated beneficial effects in patients with UC (as in the case of tofacitinib, a

preferential inhibitor of JAK1 and JAK3) [151]. Conversely, the phase 2 study of filgotinib, a selective JAK1
pt

inhibitor, has demonstrated more robust clinical efficacy in moderately to severely active CD [152]. The
ce

phase 3 study, including biologic-naïve and biologic-experienced CD patients with moderately to severely

active disease, is ongoing and is expected to be completed by November 2021 (ClinicalTrials.gov identifier:
Ac

NCT02914561). Clearly, because of the limited experience accumulated thus far, the role of small molecules

in patients with concomitant EIMs and/or comorbidities has yet to be determined. Nevertheless, it is

predictable that EIMs may benefit from novel treatments for CD provided they are demonstrated to be

superior in efficacy than currently available drugs, as most EIMs follow the course of the underlying CD

activity.

Real-life CD patients may suffer from concomitant combinations of EIMs or comorbidities which sometimes

may not be optimally managed with the same biologic drug, e.g, the biologic drug may be effective on CD
21

and beneficial on the coexisting comorbidity but may not be suitable for the associated EIM(s) or vice versa.

Therefore, the idea of combining biologic therapies with different molecular targets may be entertained.

Indeed, a number of case reports or series have been published reporting on increased efficacy of combined

biologic therapy in IBD with no concomitant increase in adverse events, mainly involving the combination

of a TNF-α inhibitor with vedolizumab or ustekinumab [153-157]. Moreover, one short-term controlled trial

assessing the safety and tolerability of concurrent natalizumab in 79 active CD patients despite infliximab

t
therapy concluded that combined biologic therapy was well tolerated with no increase in adverse events,

ip
while even recording a trend toward improved disease activity [158]. A recent systematic review analyzing

cr
the limited available literature to date on combining biologics across a number of immune-mediated

conditions concluded for a lack of consistent superior efficacy with respect to biologic monotherapy while

us
still raising safety concerns, particularly with regard to serious, life-threatening infections [159]. An open

label prospective study is currently ongoing to investigate the effect of triple combination therapy including
an
vedolizumab, adalimumab, and oral methotrexate on endoscopic remission in patients with newly diagnosed

moderately to severely active CD stratified at higher risk for complications (Clinical Trials.gov
M

identifier: NCT02764762), which should provide more reliable data on the effectiveness and safety of the

combinatorial approach, at least in the short-term. Indeed, long-term safety issues, including cancer
ed

susceptibility, will not be resolved in the next future; thus, a prudent approach may still be the wisest choice,

by selecting a single biotherapy following thorough assessment of patient characteristics in order to


pt

maximize treatment outcomes.


ce

Ideally, to implement a personalized medicine in CD, biomarkers able to predict treatment response to the

biologic drugs currently available would be desirable. Work is still in progress and is at the current time far
Ac

from becoming reality, since sophisticated instrumentations, available only in research centers, are needed

for identification, characterization, and interpretation of candidate biomarkers, which need then to be

validated in clinical studies. Fields of intense investigations include proteomics, molecular imaging, and

genetic analysis, which will hopefully provide clinicians with reliable biomarkers of response prediction to a

given biologic drug or small molecule, in terms of control of both CD activity and associated EIMs [160-

162]. Meanwhile, the choice of biologic agents in CD patients in order to achieve the best outcome still lies

on a thorough assessment of patient-related characteristics as well as deep knowledge of the properties and
22

place in therapy of each biologic drug. By critically tempering all the currently available evidence on patient

characteristics and biologic agent properties, the clinician may nonetheless opt for the best therapeutic option

for the individual CD patient (Figure 1).

Funding

This paper was not funded

Declaration of Interest

t
ip
The authors have no relevant affiliations or financial involvement with any organization or entity with a

cr
financial interest in or financial conflict with the subject matter or materials discussed in the manuscript.

This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants

us
or patents received or pending, or royalties. an
Reviewer Disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose
M
ed
pt
ce

REFERENCES
Ac

* = of interest, ** = of considerable interest

1. Torres J, Mehandru S, Colombel J-F, Peyrin-Biroulet L. Crohn’s disease. Lancet. 2017;389:1741-

1755.

2. Colìa R, Corrado A, Cantatore FP. (2016) Rheumatologic and extraintestinal manifestations of

inflammatory bowel diseases. Ann Med. 2016;48:577-585.

3. Chang MI, Cohen BL, Greenstein AJ. A review of the impact of biologics on surgical complications

in Crohn's disease. Inflamm Bowel Dis. 2015;21:1472-1477.


23

4. Harbord M, Annese V, Vavricka SR, et al. The first European evidence-based consensus on extra-

intestinal manifestations in inflammatory bowel disease. J Crohns Colitis. 2016;10:239-254.

5. Salvarani C, Vlachonikolis IG, van der Heijde DM, Fornaciari G, Macchioni P, Beltrami M, et al.

Musculoskeletal manifestations in a population-based cohort of inflammatory bowel disease

patients. Scand J Gastroenterol. 2001;36:1307-1313.

6. Stolwijk C, Pierik M, Landew_e R, Masclee A, van Tubergen A. Prevalence of self-reported

t
spondyloarthritis features in a cohort of patients with inflammatory bowel disease. Can J

ip
Gastroenterol. 2013;27:199-205.

cr
7. Ott C, Schölmerich J. Extraintestinal manifestations and complications in IBD. Nat Rev

Gastroenterol Hepatol. 2013;10:585-595.

us
8. Olivieri I, Cantini F, Castiglione F, Felice C, Gionchetti P, Orlando A, et al. Italian Expert Panel on

the management of patients with coexisting spondyloarthritis and inflammatory bowel disease.
an
Autoimmun Rev. 2014;13:822-830.

*first published suggestions for combined therapy of spondyloarthritis and inflammatory bowel
M

disease

9. Gomollón F, Dignass A, Annese V, Tilg H, Van Assche G, Lindsay JO, et al. 3rd European
ed

evidence-based consensus on the diagnosis and management of Crohn's disease 2016: Part 1:

Diagnosis and medical management. J Crohns Colitis. 2017;11:3-25.


pt

**updated guidelines for diagnosis and management of Crohn's disease


ce

10. Gionchetti P, Dignass A, Danese S, Magro Dias FJ, Rogler G, Lakatos PL, et al. 3rd European

evidence-based consensus on the diagnosis and management of Crohn's disease 2016: Part 2:
Ac

Surgical management and special situations. J Crohns Colitis. 2017;11:135-149.

**updated guidelines for diagnosis and management of Crohn's disease

11. van der Heijde D, Ramiro S, Landewé R, Baraliakos X, Van den Bosch F, Sepriano A, et al. 2016

update of the ASAS-EULAR management recommendations for axial spondyloarthritis. Ann

Rheum Dis. 2017;76:978-991.

12. Salvarani C, Fries W. Clinical features and epidemiology of spondyloarthritides associated with

inflammatory bowel disease. World J Gastroenterol. 2009;15:2449-2455.


24

13. Argollo M, Fiorino G, Peyrin-Biroulet L, Danese S. Vedolizumab for the treatment of Crohn’s

disease. Expert Rev Clin Immunol. 2018;14:179-189.

14. Tadbiri S, Peyrin-Biroulet L, Serrero M, Filippi J, Pariente B, Roblin X, et al. Impact of

vedolizumab therapy on extra-intestinal manifestations in patients with inflammatory bowel disease:

a multicentre cohort study nested in the OBSERV-IBD cohort. Aliment Pharmacol Ther.

2018;47:485-493.

t
*a study reporting on the outcome of extra-intestinal manifestations in patients with inflammatory

ip
bowel disease treated with vedolizumab

cr
15. Macaluso FS, Orlando R, Fries W, et al. The real-world effectiveness of vedolizumab on intestinal

us
and articular outcomes in inflammatory bowel diseases. Dig Liver Dis. 2018;50:675-681.

*a further study reporting on the outcome of extra-intestinal manifestations in patients with


an
inflammatory bowel disease treated with vedolizumab
M
16. Varkas G, Thevissen K, De Brabanter G, Van Praet L, Czul-Gurdian F, Cypers H, et al. An

induction or flare of arthritis and/or sacroiliitis by vedolizumab in inflammatory bowel disease: a

case series. Ann Rheum Dis. 2017;76:878-881.


ed

17. Wendling D, Sondag M, Verhoeven F, Vuitton L, Koch S, Prati C. Arthritis occurrence or

reactivation under Vedolizumab treatment for inflammatory bowel disease. A four cases report.
pt

Joint Bone Spine. 2018;85:255-256.


ce

18. Salmi M, Jalkanen S. Endothelial ligands and homing of mucosal leukocytes in extraintestinal

manifestations of IBD. Inflamm Bowel Dis. 1998;4:149-56.


Ac

19. Salmi M, Jalkanen S. Human leukocyte subpopulations from inflamed gut bind to joint vasculature

using distinct sets of adhesion molecules. J Immunol. 2001;166:4650-4657.

20. Tahir H. Therapies in ankylosing spondylitis - from clinical trials to clinical practice. Rheumatology

(Oxford). 2018;57(suppl_6):vi23-vi28.

21. Coates LC, Kavanaugh A, Mease PJ, Soriano ER, Laura Acosta-Felquer M, Armstrong AW, et al.

Group for Research and Assessment of Psoriasis and Psoriatic Arthritis 2015 treatment

recommendations for psoriatic arthritis. Arthritis Rheumatol. 2016;68:1060-1071.


25

22. Farhi D, Cosnes J, Zizi N, Chosidow O, Seksik P, Beaugerie L, Aractingi S, Khosrotehrani K.

Significance of erythema nodosum and pyoderma gangrenosum in inflammatory bowel diseases: a

cohort study of 2402 patients. Medicine (Baltimore). 2008;87:281-293.

23. Phillips FM, Verstockt B, Sebastian S, et al. Inflammatory cutaneous lesions in inflammatory bowel

disease treated with vedolizumab or ustekinumab: an ECCO CONFER multi-centre case series. J

Crohns Colitis 2019;13(Suppl_1):S292-S293.

t
24. Vavricka SR, Brun L, Ballabeni P, Pittet V, Prinz Vavricka BM, Zeitz J, Rogler G, Schoepfer AM.

ip
Frequency and risk factors for extraintestinal manifestations in the Swiss inflammatory bowel

cr
disease cohort. Am J Gastroenterol. 2011;106:110-119.

25. Vavricka SR, Gubler M, Gantenbein C, Spoerri M, Fröhlich F, Seibold F et al. Anti-TNF treatment

us
for extraintestinal manifestations of inflammatory bowel disease in the Swiss IBD Cohort Study.

Inflamm Bowel Dis. 2017;23:1174-1181.


an
26. Löfberg R, Louis EV, Reinisch W, Robinson AM, Kron M, Camez A et al. Adalimumab produces

clinical remission and reduces extraintestinal manifestations in Crohn’s disease: results from CARE.
M

Inflamm Bowel Dis. 2012;18:1-9.

27. Ortego-Centeno N, Callejas-Rubio JL, Sanchez-Cano D, Caballero-Morales T. Refractory chronic


ed

erythema nodosum successfully treated with adalimumab. J Eur Acad Dermatol Venereol.

2007;21:408-410.
pt

28. Greuter T, Navarini A, Vavricka SR. Skin manifestations of inflammatory bowel disease. Clinic
ce

Rev Allerg Immunol. 2017;53:413-427.

29. Fonder MA, Cummins DL, Ehst BD, Anhalt GJ, Meyerle JH. Adalimumab therapy for recalcitrant
Ac

pyoderma gangrenosum. J Burns Wounds. 2006;5:e8.

30. Brooklyn TN, Dunnill MG, Shetty A, Bowden JJ, Williams JD, Griffiths CE et al. Infliximab for the

treatment of pyoderma gangrenosum: a randomised, double blind, placebo controlled trial. Gut.

2006;55:505-509.

**the only randomized, double blind, placebo-controlled trial of infliximab in the treatment of

pyoderma gangrenosum
26

31. Zampeli VA, Lippert U, Nikolakis G, Makrantonaki E, Tzellos TG, Krause U, Zouboulis CC.

Disseminated refractory pyoderma gangraenosum during an ulcerative colitis flare. Treatment with

infliximab. J Dermatol Case Rep. 2015;9:62-66.

32. Hurabielle C, Schneider P, Baudry C, Bagot M, Allez M, Viguier M. Certolizumab pegol - A new

therapeutic option for refractory disseminated pyoderma gangrenosum associated with Crohn's

disease. J Dermatolog Treat. 2016;27:67-69.

t
33. Guenova E, Teske A, Fehrenbacher B, Hoerber S, Adamczyk A, Schaller M, Hoetzenecker W,

ip
Biedermann T. Interleukin 23 expression in pyoderma gangrenosum and targeted therapy with

cr
ustekinumab. Arch Dermatol. 2011;147:1203-1205.

**demonstration of the role of IL-23 in pyoderma gangrenosum-associated inflammation

us
34. Goldminz AM, Botto NC, Gottlieb AB. Severely recalcitrant pyoderma gangrenosum successfully

treated with ustekinumab. J Am Acad Dermatol. 2012;67:e237-238.


an
35. Fahmy M, Ramamoorthy S, Hata T, Sandborn WJ. Ustekinumab for peristomal pyoderma

gangrenosum. Am J Gastroenterol. 2012;107:794-795.


M

36. Greb JE, Gottlieb AB, Goldminz AM. High-dose ustekinumab for the treatment of severe,

recalcitrant pyoderma gangrenosum. Dermatol Ther. 2016;29:482-483.


ed

37. Moschen AR, Tilg H, Raine T. IL-12, IL-23 and IL-17 in IBD: immunobiology and therapeutic

targeting. Nat Rev Gastroenterol Hepatol. 2019;16:185-196.


pt

38. Bernstein CN, Blanchard JF, Rawsthorne P, Yu N. The prevalence of extraintestinal diseases in
ce

inflammatory bowel disease: a population based study. Am J Gastroenterol. 2001;96:1116 -1122.

39. Fries W, Giofré MR, Catanoso M, Lo Gullo R. Treatment of acute uveitis associated with Crohn's
Ac

disease and sacroileitis with infliximab. Am J Gastroenterol. 2002;97:499-500.

40. Murphy CC, Ayliffe WH, Booth A, Makanjuola D, Andrews PA, Jayne D. Tumor necrosis factor

alpha blockade with infliximab for refractory uveitis and scleritis. Ophthalmology. 2004;111:352-

356.

41. Barrie A, Regueiro M. Biologic therapy in the management of extraintestinal manifestations of

inflammatory bowel disease. Inflamm Bowel Dis. 2007;13:1424-1429.


27

42. Ally MR, Veerappan GR, Koff JM. Treatment of recurrent Crohn's uveitis with infliximab. Am J

Gastroenterol. 2008;103:2150-2151.

43. Jaffe GJ, Dick AD, Brézin AP, et al. Adalimumab in patients with active noninfectious uveitis. N

Engl J Med. 2016;375:932-943.

44. Nguyen QD, Merrill PT, Jaffe GJ, et al. Adalimumab for prevention of uveitic flare in patients with

inactive non-infectious uveitis controlled by corticosteroids (VISUAL II): a multicentre, double-

t
masked, randomised, placebo-controlled phase 3 trial. Lancet. 2016;388:1183-1192.

ip
45. Mugheddu C, Atzori L, Del Piano M, Lappi A, Pau M, Murgia S, Zucca I, Rongioletti F. Successful

cr
ustekinumab treatment of noninfectious uveitis and concomitant severe psoriatic arthritis and plaque

psoriasis. Dermatol Ther. 2017;30(5).

us
46. Pepple KL, Lin P. Targeting interleukin-23 in the treatment of noninfectious uveitis.

Ophthalmology. 2018;125:1977-1983.
an
47. Arepalli S, Rosenbaum JT. The use of biologics for uveitis associated with spondyloarthritis. Curr

Opin Rheumatol. 2019;31:349-354.


M

48. Broome U, Bergquist A. Primary sclerosing cholangitis, inflammatory bowel disease, and colon

cancer. Semin Liver Dis. 2006;26:31-41.


ed

49. Venkatesh PG, Navaneethan U, Shen B. Hepatobiliary disorders and complications of inflammatory

bowel disease. J Dig Dis. 2011;12:245-256.


pt

50. Bo X, Broome U, Remberger M, Sumitran-Holgersson S. Tumour necrosis factor α impairs function


ce

of liver derived T lymphocytes and natural killer cells in patients with primary sclerosing

cholangitis. Gut. 2001;49:131-141.


Ac

51. Liaskou E, Jeffery LE, Trivedi PJ, Reynolds GM, Suresh S, Bruns T, et al. Loss of CD28 expression

by liver-infiltrating T cells contributes to pathogenesis of primary sclerosing cholangitis.

Gastroenterology. 2014;147: 221-232.e7.

52. Tse CS, Loftus EV Jr, Raffals LE, Gossard AA, Lightner AL. Effects of vedolizumab, adalimumab

and infliximab on biliary inflammation in individuals with primary sclerosing cholangitis and

inflammatory bowel disease. Aliment Pharmacol Ther. 2018;48:190-195.


28

*one of the few studies reporting outcome of primary sclerosing cholangitis following biologic

therapy for IBD

53. Franceschet I, Cazzagon N, Del Ross T, D'Inca R, Buja A, Floreani A. Primary sclerosing

cholangitis associated with inflammatory bowel disease: an observational study in a Southern

Europe population focusing on new therapeutic options. Eur J Gastroenterol Hepatol. 2016;28:508-

513.

t
54. Halilbasic E, Fuchs C, Hofer H, Paumgartner G, Trauner M. Therapy of primary sclerosing

ip
cholangitis - today and tomorrow. Dig Dis. 2015;Suppl 2:149-163.

cr
55. Stine JG, Wang J, Behm BW. Chronic cholestatic liver injury attributable to vedolizumab. J Clin

Transl Hepatol. 2016;4:277-280.

us
56. Lynch KD, Keshav S, Chapman RW. The use of biologics in patients with inflammatory bowel

disease and primary sclerosing cholangitis. Curr Hepatol Rep. 2019;18:115-126.


an
57. Vegh Z, Macsai E, Lakatos L, Lakatos PL. The incidence of glomerulonephritis in a population-

based inception cohort of patients with inflammatory bowel disease. Dig Liver Dis. 2017;49:718-
M

719.

58. Ambruzs JM, Larsen CP. Renal manifestations of inflammatory bowel disease. Rheum Dis Clin
ed

North Am. 2018;44:699-714.

59. Oikonomou K, Kapsoritakis A, Eleftheriadis T, Stefanidis I, Potamianos S. Renal manifestations


pt

and complications of inflammatory bowel disease. Inflamm Bowel Dis. 2011;17:1034-1045.


ce

60. Park YK, Han DS, Eun CS. Systemic amyloidosis with Crohn’s disease treated with infliximab.

Inflamm Bowel Dis. 2008;14:431-432.


Ac

61. Said Y, Debbeche R, Hamzaoui L, et al. Infiximab for treatment of systemic amyloidosis associated

with Crohn’s disease. J Crohns Colitis. 2011;5:171-172.

62. Iizuka M, Sagara S, Etou T. Efficacy of scheduled infliximab maintenance therapy on systemic

amyloidosis associated with Crohn’s disease. Inflamm Bowel Dis. 2011;17:E67-68.

63. Pukitis A, Zake T, Groma V, et al. Effect of infliximab induction therapy on secondary systemic

amyloidosis associated with Crohn’s disease: case report and review of the literature. J

Gastrointestin Liver Dis. 2013;22:333-336.


29

64. Tosca Cuquerella J, Bosca-Watts MM, Anton Ausejo R, et al. Amyloidosis in inflammatory bowel

disease: a systematic review of epidemiology, clinical features, and treatment. J Crohns Colitis.

2016;10:1245-1253.

65. Esatoglu SN, Hatemi G, Ugurlu S, Gokturk A, Tascilar K, Ozdogan H. Long-term follow-up of

secondary amyloidosis patients treated with tumor necrosis factor inhibitor therapy: A STROBE-

compliant observational study. Medicine (Baltimore). 2017;96:e7859.

t
66. Singh S, Kumar N, Loftus EV Jr, Kane SV. Neurologic complications in patients with inflammatory

ip
bowel disease: increasing relevance in the era of biologics. Inflamm Bowel Dis. 2013;19:864-872.

cr
67. Zois CD, Katsanos KH, Kosmidou M, Tsianos EV. Neurologic manifestations in inflammatory

bowel diseases: current knowledge and novel insights. J Crohns Colitis. 2010;4:115-124.

us
68. Stübgen JP. Tumor necrosis factor-alpha antagonists and neuropathy. Muscle Nerve. 2008;37:281-

292
an
69. Burger DC, Florin TH. Peripheral neuropathy with infliximab therapy in inflammatory bowel

disease. Inflamm Bowel Dis. 2009;15:1772.


M

70. Cesarini M, Angelucci E, Foglietta T, Vernia P. Guillain-Barrè syndrome after treatment with

human anti-tumor necrosis factorα (adalimumab) in a Crohn's disease patient: case report and
ed

literature review. J Crohns Colitis. 2011;5:619-622.

71. Tsouni P, Bill O, Truffert A, Liaudat C, Ochsner F, Steck AJ, Kuntzer T. Anti-TNF alpha
pt

medications and neuropathy. J Peripher Nerv Syst. 2015;20:397-402.


ce

72. Yagita M, Hamano T, Hatachi S, Fujita M. Peripheral neuropathies during biologic therapies. Mod

Rheumatol. 2016;26:288-293.
Ac

73. Rodinò S, Saccà N, D'Amico T, Fragomeni A, Giglio A. Severe polyneuropathy complicating active

Crohn's disease: rapid response to Infliximab. Gut. 2003;52:1070.

74. Kemanetzoglou E, Andreadou E. CNS demyelination with TNF-α blockers. Curr Neurol Neurosci

Rep. 2017;17:36.

75. Segal BM, Constantinescu CS, Raychaudhuri A, Kim L, Fidelus-Gort R, Kasper LH; Ustekinumab

MS Investigators. Repeated subcutaneous injections of IL12/23 p40 neutralising antibody,


30

ustekinumab, in patients with relapsing-remitting multiple sclerosis: a phase II, double-blind,

placebo-controlled, randomised, dose-ranging study. Lancet Neurol. 2008;7:796-804.

76. Gensicke H, Leppert D, Yaldizli Ö, Lindberg RL, Mehling M, Kappos L, Kuhle J. Monoclonal

antibodies and recombinant immunoglobulins for the treatment of multiple sclerosis. CNS Drugs.

2012;26:11-37.

77. Moran GW, Dubeau MF, Kaplan GG, Panaccione R, Ghosh S. The increasing weight of Crohn’s

t
disease subjects in clinical trials: a hypothesis-generatings time-trend analysis. Inflamm Bowel Dis.

ip
2013;19:2949-2956.

cr
78. Harper JW, Sinanan MN, Zisman TL. Increased body mass index is associated with earlier time to

loss of response to infliximab in patients with inflammatory bowel disease. Inflamm Bowel Dis.

us
2013;19:2118-2124.

79. Shan J, Zhang J. Impact of obesity on the efficacy of different biologic agents in inflammatory
an
diseases: A systematic review and meta-analysis. Joint Bone Spine. 2019;86:173-183.

80. Ding Z, Wu XR, Remer EM, Lian L, Stocchi L, Li Y, McCullough A, Remzi FH, Shen B.
M

Association between high visceral fat area and postoperative complications in patients with Crohn’s

disease following primary surgery. Colorectal Dis. 2016;18:163-172.


ed

81. Gremese E, Carletto A, Padovan M, Atzeni F, Raffeiner B, Giardina AR, et al. Obesity and

reduction of the response rate to anti-tumor necrosis factor α in rheumatoid arthritis: an approach to
pt

a personalized medicine. Arthritis Care Res. 2013;65:94-100.


ce

82. Hemperly A, Vande Casteele N. Clinical pharmacokinetics and pharmacodynamics of infliximab in

the treatment of inflammatory bowel disease. Clin Pharmacokinet. 2018;57:929-942.


Ac

83. Vande Casteele N, Baert F, Bian S, Dreesen E, Compernolle G, Van Assche G, Ferrante M,

Vermeire S, Gils A. Subcutaneous absorption contributes to observed interindividual variability in

adalimumab serum concentrations in Crohn's disease: a prospective multicentre study. J Crohns

Colitis. 2019. pii: jjz050.

84. Guerbau L, Gerard R, Duveau N, Staumont-Sallé D, Branche J, Maunoury V, Cattan S, Wils P,

Boualit M, Libier L, Cotteau-Leroy A, Desreumaux P, Nachury M, Pariente B. Patients with


31

Crohn's disease with high body mass index present more frequent and rapid loss of response to

infliximab. Inflamm Bowel Dis. 2017;23:1853-1859.

85. Puig L. Obesity and psoriasis: body weight and body mass index influence the response to

biological treatment. J Eur Acad Dermatol Venereol. 2011;25:1007-1011.

86. Puig L, Ruiz-Salas V. Long-term efficacy, safety and drug survival of ustekinumab in a Spanish

cohort of patients with moderate to severe plaque psoriasis. Dermatology. 2015;230:46-54.

t
87. Lebwohl M, Yeilding N, Szapary P, Wang Y, Li S, Zhu Y, Reich K, Langley RG, Papp KA. Impact

ip
of weight on the efficacy and safety of ustekinumab in patients with moderate to severe psoriasis:

cr
rationale for dosing recommendations. J Am Acad Dermatol. 2010;63:571-579.

88. Del Alcázar E, Ferran M, López-Ferrer A, Notario J, Vidal D, Riera J, Aparicio G, Gallardo F,

us
Vilarrasa E, Alsina M, Puig L, Ferrándiz C, Carrascosa JM. Effectiveness and safety of

ustekinumab 90 mg in patients weighing 100 kg or less: a retrospective, observational, multicenter


an
study. J Dermatolog Treat. 2019:1-5.

89. Sparrow MP, Papamichael K, Ward MG, Riviere P, Laharie D, Paul S, Roblin X. Therapeutic drug
M

monitoring of biologics during induction to prevent primary non-response. J Crohns Colitis. 2019,

doi: 10.1093/ecco-jcc/jjz162.
ed

90. Kurnool S, Nguyen NH, Proudfoot J, Dulai PS, Boland BS, Vande Casteele N, Evans E, Grunvald

EL, Zarrinpar A, Sandborn WJ, Singh S. High body mass index is associated with increased risk of
pt

treatment failure and surgery in biologic-treated patients with ulcerative colitis. Aliment Pharmacol
ce

Ther. 2018;47:1472-1479.

91. Ward MG, Sparrow MP, Roblin X. Therapeutic drug monitoring of vedolizumab in inflammatory
Ac

bowel disease: current data and future directions. Therap Adv Gastroenterol.

2018;11:1756284818772786.

92. Li H, Shi FH, Huang SY, Zhang SG, Chen ML. A review on clinical pharmacokinetics,

pharmacodynamics, and pharmacogenomics of natalizumab: a humanized anti-α4 integrin

monoclonal antibody. Curr Drug Metab. 2018;19:1213-1223.

93. Khatri BO, Man S, Giovannoni G, Koo AP, Lee JC, Tucky B, et al. Effect of plasma exchange in

accelerating natalizumab clearance and restoring leukocyte function. Neurology. 2009;72:402-409.


32

94. Nelson SM, Nguyen TM, McDonald JW, MacDonald JK. Natalizumab for induction of remission in

Crohn's disease. Cochrane Database Syst Rev. 2018;8:CD006097.

95. Zou ZY, Shen B, Fan JG. Systematic review with meta-analysis: epidemiology of nonalcoholic fatty

liver disease in patients with inflammatory bowel disease. Inflamm Bowel Dis. 2019. pii: izz043.

doi: 10.1093/ibd/izz043.

96. Liu W, Baker RD, Bhatia T, Zhu L, Baker SS. Pathogenesis of nonalcoholic steatohepatitis. Cell

t
Mol Life Sci. 2016;73:1969-87.

ip
97. Koca SS, Bahcecioglu IH, Poyrazoglu OK, Ozercan IH, Sahin K, Ustundag B. The treatment with

cr
antibody of TNF-alpha reduces the inflammation, necrosis and fibrosis in the non-alcoholic

steatohepatitis induced by methionine- and choline-deficient diet. Inflammation. 2008;31:91-98.

us
98. Schramm C, Schneider A, Marx A, Lohse AW. Adalimumab could suppress the activity of non

alcoholic steatohepatitis (NASH). Z Gastroenterol. 2008;46:1369-1371.


an
99. Jess T, Jensen BW, Andersson M, Villumsen M, Allin KH. Inflammatory bowel disease increases

risk of type 2 diabetes in a nationwide cohort study. Clin Gastroenterol Hepatol. 2019. pii: S1542-
M

3565(19)30841-9. doi: 10.1016/j.cgh.2019.07.052.

100. Wieser V, Moschen AR, Tilg H. Inflammation, cytokines and insulin resistance: a clinical
ed

perspective. Arch Immunol Ther Exp. 2013;61:119-125.

101. Araújo EP, De Souza CT, Ueno M, Cintra DE, Bertolo MB, Carvalheira JB, Saad MJ,
pt

Velloso LA. Infliximab restores glucose homeostasis in an animal model of diet-induced obesity
ce

and diabetes. Endocrinology. 2007;148:5991-5997.

102. Yazdani-Biuki B, Stelzl H, Brezinschek HP, Hermann J, Mueller T, Krippl P, Graninger W,


Ac

Wascher TC. Improvement of insulin sensitivity in insulin resistant subjects during prolonged

treatment with the anti-TNF-alpha antibody infliximab. Eur J Clin Invest. 2004;34:641-642.

103. Gupta-Ganguli M, Cox K, Means B, Gerling I, Solomon SS. Does therapy with anti-TNF-

alpha improve glucose tolerance and control in patients with type 2 diabetes? Diabetes Care.

2011;34:e121.
33

104. Yazdani-Biuki B, Mueller T, Brezinschek HP, Hermann J, Graninger W, Wascher TC.

Relapse of diabetes after interruption of chronic administration of anti-tumor necrosis factor-alpha

antibody infliximab: a case observation. Diabetes Care. 2006;29:1712-1713.

*an interesting observation supporting the role for TNF-α inhibitors in improving diabetes

105. Esser N, Paquot N, Scheen AJ. Anti-inflammatory agents to treat or prevent type 2 diabetes,

metabolic syndrome and cardiovascular disease. Expert Opin Investig Drugs. 2015;24:283-307.

t
106. Marwaha AK, Tan S, Dutz JP. Targeting the IL-17/IFN-γ axis as a potential new clinical

ip
therapy for type 1 diabetes. Clin Immunol. 2014;154:84-89.

cr
107. Chung ES, Packer M, Lo KH, Fasanmade AA, Willerson JT; Anti-TNF Therapy Against

Congestive Heart Failure Investigators. Randomized, double-blind, placebo-controlled, pilot trial of

us
infliximab, a chimeric monoclonal antibody to tumor necrosis factor-alpha, in patients with

moderate-to-severe heart failure: results of the anti-TNF Therapy Against Congestive Heart Failure
an
(ATTACH) trial. Circulation. 2003;107:3133-3140.

108. Tam LS, Kitas GD, González-Gay MA. Can suppression of inflammation by anti-TNF
M

prevent progression of subclinical atherosclerosis in inflammatory arthritis? Rheumatology

(Oxford). 2014;53:1108-1119.
ed

109. van Sijl AM, van Eijk IC, Peters MJ, Serné EH, van der Horst-Bruinsma IE, Smulders YM,

Nurmohamed MT. Tumour necrosis factor blocking agents and progression of subclinical
pt

atherosclerosis in patients with ankylosing spondylitis Ann Rheum Dis. 2015;74:119-123.


ce

110. Sanjadi M, Rezvanie Sichanie Z, Totonchi H, Karami J, Rezaei R, Aslani S. Atherosclerosis

and autoimmunity: a growing relationship. Int J Rheum Dis. 2018;21:908-921.


Ac

111. Ellinghaus D, Ellinghaus E, Nair RP, Stuart PE, Esko T, Metspalu A, et al. Combined

analysis of genome-wide association studies for Crohn disease and psoriasis identifies seven shared

susceptibility loci. Am J Hum Genet. 2012;90:636-647.

112. Halling ML, Kjeldsen J, Knudsen T, Nielsen J, Hansen LK. Patients with inflammatory

bowel disease have increased risk of autoimmune and inflammatory diseases. World J

Gastroenterol. 2017;23:6137-6146.
34

113. Mansouri B, Patel M, Menter A. Biological therapies for psoriasis. Expert Opin Biol Ther.

2013;13:1715-30.

114. Conrad C, Gilliet M. Psoriasis: from pathogenesis to targeted therapies. Clin Rev Allergy

Immunol. 2018;54:102-113.

115. Shah VV, Lee EB, Reddy S, Lin EJ, Wu JJ. Comparison of guidelines for the use of TNF

inhibitors for psoriasis in the United States, Canada, Europe and the United Kingdom: a critical

t
appraisal and comprehensive review. J Dermatolog Treat. 2018;29:586-592.

ip
116. Frieder J, Kivelevitch D, Fiore CT, Saad S, Menter A. The impact of biologic agents on

cr
health-related quality of life outcomes in patients with psoriasis. Expert Rev Clin Immunol.

2018;14:1-19.

us
117. Thibodaux RJ, Triche MW, Espinoza LR. Ustekinumab for the treatment of psoriasis and

psoriatic arthritis: a drug evaluation and literature review. Expert Opin Biol Ther. 2018;18:821-827.
an
118. Singh JA, Guyatt G, Ogdie A, Gladman DD, Deal C, Deodhar A, et al. Special article: 2018

American College of Rheumatology/National Psoriasis Foundation guideline for the treatment of


M

psoriatic arthritis. Arthritis Rheumatol. 2019;71:5-32.

119. Barthel C, Biedermann L, Frei P, Vavricka SR, Kündig T, Fried M, Rogler G, Scharl M.
ed

Induction or exacerbation of psoriasis in patients with Crohn's disease under treatment with anti-

TNF antibodies. Digestion. 2014;89:209-15.


pt

120. Santos S, Gamelas V, Carvalho D, Bernardes C, Saiote J, Ramos J. Therapeutic strategies in


ce

the approach of paradoxical psoriasis in IBD: experience of a centre. J Crohns Colitis.

2019;13(Suppl_1):S403-S404.
Ac

121. Shah ED, Farida JP, Siegel CA, Chong K, Melmed GY. Risk for overall infection with anti-

TNF and anti-integrin agents used in IBD: a systematic review and meta-analysis. Inflamm Bowel

Dis. 2017;23:570-577.

122. McConnell RA, Mahadevan U. Pregnancy and the patient with inflammatory bowel disease:

fertility, treatment, delivery, and complications. Gastroenterol Clin North Am. 2016;45:285-301.

123. Hyrich KL, Verstappen SM. Biologic therapies and pregnancy: the story so far.

Rheumatology (Oxford). 2014;53:1377-1385.


35

124. Lichtenstein GR, Feagan BG, Mahadevan U, et al. Pregnancy outcomes reported during the

13-year TREAT registry: a descriptive report. Am J Gastroenterol. 2018;113:1678-1688.

125. Chaparro M, Verreth A, Lobaton T, et al. Long-term safety of in utero exposure to anti-

TNFα drugs for the treatment of inflammatory bowel disease: results from the multicenter European

TEDDY study. Am J Gastroenterol. 2018;113:396-403.

126. Porter C, Armstrong-Fisher S, Kopotsha T, Smith B, Baker T, Kevorkian L, Nesbitt A.

t
Certolizumab pegol does not bind the neonatal Fc receptor (FcRn): consequences for FcRn-

ip
mediated in vitro transcytosis and ex vivo human placental transfer. J Reprod Immunol.

cr
2016;116:7-12.

**demonstration of the molecular mechanism determining impaired placental transfer of

us
certolizumab

127. Mariette X, Förger F, Abraham B, Flynn AD, Moltó A, Flipo RM, et al. Lack of placental
an
transfer of certolizumab pegol during pregnancy: results from CRIB, a prospective, postmarketing,

pharmacokinetic study. Ann Rheum Dis. 2018;77:228-233.


M

**a study confirming the safety of certolizumab pegol in pregnancy

128. Clowse ME, Wolf DC, Förger F, Cush JJ, Golembesky A, Shaughnessy L, et al. Pregnancy
ed

outcomes in subjects exposed to certolizumab pegol. J Rheumatol. 2015;42:2270-2278.

129. Clowse MEB, Scheuerle AE, Chambers C, Afzali A, Kimball AB, Cush JJ, et al. Pregnancy
pt

outcomes after exposure to certolizumab pegol: updated results from a pharmacovigilance safety
ce

database. Arthritis Rheumatol. 2018;70:1399-1407.

130. Clowse ME, Förger F, Hwang C, Thorp J, Dolhain RJ, van Tubergen A, et al. Minimal to no
Ac

transfer of certolizumab pegol into breast milk: results from CRADLE, a prospective,

postmarketing, multicentre, pharmacokinetic study. Ann Rheum Dis. 2017;76:1890-1896.

**a study confirming the safety of certolizumab pegol in breastfeeding

131. Cortes X, Borrás-Blasco J, Antequera B, et al. Ustekinumab therapy for Crohn's disease

during pregnancy: a case report and review of the literature. J Clin Pharm Ther. 2017;42:234-236.
36

132. Rowan CR, Cullen G, Mulcahy HE, et al. Ustekinumab drug levels in maternal and cord

blood in a woman with Crohn's disease treated until 33 weeks of gestation. J Crohns Colitis.

2018;12:376-378.

133. Wieringa JW, Driessen GJ, Van Der Woude CJ. Pregnant women with inflammatory bowel

disease: the effects of biologicals on pregnancy, outcome of infants, and the developing immune

system. Expert Rev Gastroenterol Hepatol. 2018;12:811-818.

t
134. Götestam Skorpen C , Hoeltzenbein M , Tincani A , et al. The EULAR points to consider for

ip
use of antirheumatic drugs before pregnancy, and during pregnancy and lactation. Ann Rheum Dis.

cr
2016;75:795-810.

135. AlAmeel T, Al Sulais E. Ustekinumab and pregnancy: continue or cease before you

us
conceive? Gastroenterology. 2019 doi:10.1053/j.gastro.2019.05.077

136. Julsgaard M , Kjeldsen J , Baumgart DC. Vedolizumab safety in pregnancy and newborn
an
outcomes. Gut. 2017;66:1866-1867.

137. Mahadevan U, Vermeire S, Lasch K, et al. Vedolizumab exposure in pregnancy: outcomes


M

from clinical studies in inflammatory bowel disease. Aliment Pharmacol Ther. 2017;45:941-950.

138. Bar-Gil Shitrit A, Ben Yaʼacov A, Livovsky DM, et al. Exposure to vedolizumab in IBD
ed

pregnant women appears of low risk for mother and neonate: a first prospective comparison study.

Am J Gastroenterol. 2019;114:1172-1175.
pt

139. Moens A, van der Woude C, M Julsgaard M, et al. Pregnancy outcomes in IBD patients
ce

treated with vedolizumab, anti-TNF, or conventional therapy. J Crohns Colitis.

2019;13(Suppl_1):S041-S042.
Ac

140. Moens A, van Hoeve K, Humblet E, et al. Outcome of pregnancies in female patients with

inflammatory bowel diseases treated with vedolizumab. J Crohns Colitis. 2019;13:12-18.

141. Sturm A, Maaser C, Mendall M, Karagiannis D, Karatzas P, Ipenburg N, et al. European

Crohn's and Colitis Organisation topical review on IBD in the elderly. J Crohns Colitis.

2017;11:263-273.
37

142. Jeuring SF, van den Heuvel TR, Zeegers MP, et al. Epidemiology and longterm outcome of

inflammatory bowel disease diagnosed at elderly age – an increasing distinct entity? Inflamm

Bowel Dis. 2016;22:1425-1434.

143. Nguyen GC, Sheng L, Benchimol EI. Health care utilization in elderly onset inflammatory

bowel disease: A population-based study. Inflamm Bowel Dis. 2015;21:777-782.

144. Ananthakrishnan AN, McGinley EL. Infection-related hospitalizations are associated with

t
increased mortality in patients with inflammatory bowel disease. J Crohns Colitis. 2013;7:107-112.

ip
145. Cottone M, Kohn A, Daperno M, et al. Advanced age is an independent risk factor for severe

cr
infections and mortality in patients given anti-tumor necrosis factor therapy for inflammatory bowel

disease. Clin Gastroenterol Hepatol. 2011;9:30-35.

us
146. Hayashi M, Umezawa Y, Fukuchi O, Ito T, Saeki H, Nakagawa H. Efficacy and safety of

ustekinumab treatment in elderly patients with psoriasis. J Dermatol. 2014;41:974-980.


an
147. Pudipeddi A, Kariyawasam V, Haifer C, Baraty B, Paramsothy S, Leong RW. Safety of

drugs used for the treatment of Crohn's disease. Expert Opin Drug Saf. 2019;18:357-367.
M

148. McLean LP, Cross RK. Integrin antagonists as potential therapeutic options for the treatment

of Crohn's disease. Expert Opin Investig Drugs. 2016;25:263-273.


ed

149. Boland BS, Vermeire S. Janus kinase antagonists and other novel small molecules for the

treatment of Crohn's disease. Gastroenterol Clin North Am. 2017;46:627-644.


pt

150. Olivera P, Danese S, Peyrin-Biroulet L. Next generation of small molecules in inflammatory


ce

bowel disease. Gut. 2016;66:199-209.

151. Sandborn WJ, Su C, Sands BE, D’Haens GR, Vermeire S, Schreiber S, et al. Tofacitinib as
Ac

induction and maintenance therapy for ulcerative colitis. N Engl J Med. 2017;376:1723-1736.

152. Vermeire S, Schreiber S, Petryka R, Kuehbacher T, Hebuterne X, Roblin X, et al. Clinical

remission in patients with moderate-to-severe Crohn’s disease treated with filgotinib (the FITZROY

study): results from a phase 2, double-blind, randomised, placebo-controlled trial. Lancet.

2017;389:266-275.

153. Hirten R, Longman RS, Bosworth BP, Steinlauf A, Scherl E. Vedolizumab and infliximab

combination therapy in the treatment of Crohn’s disease. Am J Gastroenterol. 2015;110:1737-1738.


38

154. Yzet C, Dupas JL, Fumery M. Ustekinumab and anti-TNF combination therapy in patients

with inflammatory bowel disease. Am J Gastroenterol. 2016;111:748-749.

155. Fischer S, Rath T, Geppert CI, et al. Long-term combination therapy with anti-TNF plus

vedolizumab induces and maintains remission in therapy-refractory ulcerative colitis. Am J

Gastroenterol. 2017;112:1621-1623.

156. Bethge J, Meffert S, Ellrichmann M, Conrad C, Nikolaus S, Schreiber S. Combination

t
therapy with vedolizumab and etanercept in a patient with pouchitis and spondylarthritis. BMJ Open

ip
Gastroenterol. 2017;4:e000127.

cr
157. Mao EJ, Lewin S, Terdiman JP, Beck K. Safety of dual biological therapy in Crohn's

disease: a case series of vedolizumab in combination with other biologics. BMJ Open Gastroenterol.

us
2018;5:e000243.

158. Sands BE, Kozarek R, Spainhour J, et al. Safety and tolerability of concurrent natalizumab
an
treatment for patients with Crohn's disease not in remission while receiving infliximab. Inflamm

Bowel Dis. 2007;13:2-11.


M

159. Hirten RP, Iacucci M, Shah S, Ghosh S, Colombel JF. Combining biologics in inflammatory

bowel disease and other immune mediated inflammatory disorders. Clin Gastroenterol Hepatol.
ed

2018;16:1374-1384.

160. Gisbert JP, Chaparro M. Clinical usefulness of proteomics in inflammatory bowel disease: a
pt

comprehensive review. J Crohns Colitis. 2019;13:374-384.


ce

161. Atreya R, Neumann H, Neufert C, et al. In vivo imaging using fluorescent antibodies to

tumor necrosis factor predicts therapeutic response in Crohn's disease. Nat Med. 2014;20:313-318.
Ac

162. Reinglas J, Gonczi L, Kurt Z, Bessissow T, Lakatos PL. Positioning of old and new

biologicals and small molecules in the treatment of inflammatory bowel diseases. World J

Gastroenterol. 2018;24:3567-3582.

FIGURE LEGENDS
39

Figure 1. Summary of suggested therapeutic choices in Crohn’s disease patients eligible to biologic therapy,

according to concomitant extra-intestinal manifestations (A), comorbidities (B), or physiologic

characteristics (C). DM, diabetes mellitus; NAFLD, non-alcoholic fatty liver disease; NASH, non-alcoholic

steatohepatitis; PSC, primary sclerosing cholangitis. See text for details.

Figure 2. Examples of suggested therapeutic choices when multiple EIMs or comorbidities coexist. NAFLD,

non-alcoholic fatty liver disease; NASH, non-alcoholic steatohepatitis. See text for details.

t
ip
cr
us
an
M
ed
pt
ce
Ac
40

Table 1. Main characteristics of the biologic agents used for Crohn’s disease treatment

Name Structure Molecular Route of Frequency of Schedule used in Crohn’s Schedul


target administration administration disease
Adalimumab fully human TNF-α s.c. biweekly 160 mg at week 0, 80 mg at 40 mg every
monoclonal week 2 (induction phase)
IgG1 and then 40 mg every 2
antibody weeks
Certolizumab PEGylated TNF-α s.c. biweekly 400 mg at weeks 0, 2 and 4 400 mg
pegol Fab of a (induction phase) and then (induction p
humanized 400 mg every 4 weeks e
monoclonal
antibody

t
Infliximab chimeric TNF-α i.v. every 8 weeks during 5 mg/kg at weeks 0, 2 and 6 3-5 mg/k

ip
human- maintenance (induction phase) and then (induction p
mouse IgG1 treatment (for every 8 weeks

cr
monoclonal induction, three
antibody infusions at T0, T2,
and T6 weeks)
Natalizumab humanized α4 i.v. every 4 weeks 300 mg every 4 weeks

us
IgG4 integrin
monoclonal
antibody
Ustekinumab fully human IL-12 and i.v. and s.c. every 8-12 weeks 260-520 mg i.v. based on
an 45 or 90 m
monoclonal IL-23 during maintenance body weight respectiv
IgG1 treatment (for (approximatively 6 mg/kg) (induction p
antibody induction, one at week 0, followed by 90
weight-based mg s.c. at week 8 (induction
M
infusional dose at T0) phase) and then 90 mg s.c.
every 8-12 weeks
Vedolizumab humanized α4β7 i.v. every 8 weeks during 300 mg at weeks 0, 2 and 6
IgG1 integrin maintenance (induction phase) and then
ed

monoclonal treatment (for 300 mg every 8 weeks


antibody induction, three
infusions at T0, T2,
and T6 weeks)
pt

Fab, antigen-binding fragment; IL, interleukin; i.v., intravenous; s.c., subcutaneous; TNF, tumor necrosis
factor
ce
Ac
41

Table 2. Examples of extra-intestinal manifestations and comorbidities possibly coexisting with Crohn’s
disease at the time of consideration of biologic therapy. Included are also physiologic conditions that should
be taken into account before starting any type of biotherapy.

EXTRA-INTESTINAL MANIFESTATIONS COMORBIDITIES PHYSIOLOGIC CONDITIONS


JOINTS METABOLIC CHILD-BEARING AGE and
• peripheral oligoarthritis • obesity PREGNANCY
• peripheral polyarthritis • NAFLD/NASH
• axial spondilarthritis • diabetes mellitus
• mixed peripheral and axial

t
spondylarthritis

ip
cr
SKIN CARDIOVASCULAR OLD AGE
• erythema nodosum • atherosclerosis
• pyoderma gangrenosum • heart failure

us
EYE DERMATOLOGIC
• scleritis • psoriasis
• uveitis
LIVER IMMUNOLOGIC
an
• primary sclerosing cholangitis • history of infections
KIDNEY
• glomerulonephritis
M
(amyloidosis)
NERVOUS SYSTEM
• peripheral neuropathies
• multiple sclerosis
ed

NAFLD, non-alcoholic fatty liver disease; NASH, non-alcoholic steatohepatitis


pt
ce
Ac
Fig 1
Ac
ce
pt
ed
M
an
us
cr
ip
t
42
2
fig 2
Ac
ce
pt
ed
M
an
us
cr
ip
t
43
3

You might also like